scispace - formally typeset
Search or ask a question
Journal ArticleDOI

PCSD1, a new patient-derived model of bone metastatic prostate cancer, is castrate-resistant in the bone-niche

TL;DR: PCSD1 is a new primary prostate cancer bone metastasis-derived xenograft model to study bone metastatic disease and for pre-clinical drug development of novel therapies for inhibiting therapy resistant prostate cancer growth in the bone-niche.
Abstract: Prostate cancer bone metastasis occurs in 50-90% of men with advanced disease for which there is no cure. Bone metastasis leads to debilitating fractures and severe bone pain. It is associated with therapy resistance and rapid decline. Androgen deprivation therapy (ADT) is standard of care for advanced prostate cancer, however, bone metastatic prostate cancer (PCa) often becomes resistant to ADT. There are few pre-clinical models to understand the interaction between the bone microenvironment and prostate cancer. Here we report the castrate resistant growth in the bone niche of PCSD1, a patient-derived intra-femoral xenograft model of prostate bone metastatic cancer treated with the anti-androgen, bicalutamide. PCSD1 bone-niche model was derived from a human prostate cancer femoral metastasis resected during hemiarthroplasty and serially transplanted into Rag2 −/− ;γ c −/− mice intra-femorally (IF) or sub-cutaneously (SC). At 5 weeks post-transplantation mice received bicalutamide or vehicle control for 18 days. Tumor growth of PCSD1 was measured with calipers. PSA expression in PCSD1 xenograft tumors was determined using quantitative RT-PCR and immunohistochemistry. Expression of AR and PSMA, were also determined with qPCR. PCSD1 xenograft tumor growth capacity was 24 fold greater in the bone (intra-femoral, IF) than in the soft tissue (sub-cutaneous, SC) microenvironment. Treatment with the anti-androgen, bicalutamide, inhibited tumor growth in the sub-cutaneous transplantation site. However, bicalutamide was ineffective in suppressing PCSD1 tumor growth in the bone-niche. Nevertheless, bicalutamide treatment of intra-femoral tumors significantly reduced PSA expression (p < =0.008) and increased AR (p < =0.032) relative to control. PCSD1 tumors were castrate resistant when growing in the bone-niche compared to soft tissue. Bicalutamide had little effect on reducing tumor burden in the bone yet still decreased tumor PSA expression and increased AR expression, thus, this model closely recapitulated castrate-resistant, human prostate cancer bone metastatic disease. PCSD1 is a new primary prostate cancer bone metastasis-derived xenograft model to study bone metastatic disease and for pre-clinical drug development of novel therapies for inhibiting therapy resistant prostate cancer growth in the bone-niche.

Content maybe subject to copyright    Report

Citations
More filters
Journal ArticleDOI
TL;DR: The interaction of PCSD1 cells with different bone region-specific microenvironments specified the type of bone lesion and this approach can be used to determine if different bone regions support more therapy resistant tumor growth, thus, requiring novel treatments.

6 citations


Cites background or methods from "PCSD1, a new patient-derived model ..."

  • ...We showed that PCSD1 tumor cells were resistant to anti-androgen therapy specifically in the bone-niche [30]....

    [...]

  • ...Therefore, the PCSD1 intra-femoral xenograft model can be used to determine quantitatively the effects of current treatments such as enzalutamide and/or docetaxel on survival of prostate cancer cells in the spongy versus cortical boneniches and determine which of these niches may perhaps preferentially support prostate cancer stem cells or castrate-resistant prostate cancer cells [30]....

    [...]

Journal ArticleDOI
TL;DR: A new mechanism of castration resistance is shown in which enzalutamide induced dormancy and novel basal-luminal-like cells in bone metastatic prostate cancer organoids, which can be used to develop therapies targeting dormant APDT-resistant cells and host factors required for SARS-CoV-2 viral entry.
Abstract: Advanced prostate cancer (PCa) patients with bone metastases are treated with androgen pathway directed therapy (APDT). However, this treatment invariably fails and the cancer becomes castration resistant. To elucidate resistance mechanisms and to provide a more predictive pre-clinical research platform reflecting tumor heterogeneity, we established organoids from a patient-derived xenograft (PDX) model of bone metastatic prostate cancer, PCSD1. APDT-resistant PDX-derived organoids (PDOs) emerged when cultured without androgen or with the anti-androgen, enzalutamide. Transcriptomics revealed up-regulation of neurogenic and steroidogenic genes and down-regulation of DNA repair, cell cycle, circadian pathways and the severe acute respiratory syndrome (SARS)-CoV-2 host viral entry factors, ACE2 and TMPRSS2. Time course analysis of the cell cycle in live cells revealed that enzalutamide induced a gradual transition into a reversible dormant state as shown here for the first time at the single cell level in the context of multi-cellular, 3D living organoids using the Fucci2BL fluorescent live cell cycle tracker system. We show here a new mechanism of castration resistance in which enzalutamide induced dormancy and novel basal-luminal-like cells in bone metastatic prostate cancer organoids. These PDX organoids can be used to develop therapies targeting dormant APDT-resistant cells and host factors required for SARS-CoV-2 viral entry.

6 citations

Journal ArticleDOI
TL;DR: The role of immune cells in bone cancer pain remains understudied and here it is sought to recapitulate this symptomology in a murine model.
Abstract: Background Over 70% to 85% of men with advanced prostate cancer (PCa) develop bone metastases characterized by severe bone pain and increased likelihood of bone fracture. These clinical features result in decreased quality of life and act as a predictor of higher mortality. Mechanistically, the skeletal pathologies such as osteolytic lesions and abnormal osteoblastic activity drive these symptoms. The role of immune cells in bone cancer pain remains understudied, here we sought to recapitulate this symptomology in a murine model. Methods The prostate cancer bone metastasis-induced pain model (CIBP) was established by transplanting a mouse prostate cancer cell line into the femur of immunocompetent mice. Pain development, gait dynamics, and the changes in emotional activities like depression and anxiety were evaluated. Animal tissues including femurs, dorsal root ganglion (DRG), and spinal cord were collected at killing and microcomputed tomography (μCT), histology/immunohistochemistry, and quantitative immunofluorescent analysis were performed. Results Mice receiving prostate cancer cells showed a significantly lower threshold for paw withdrawal responses induced by mechanical stimulation compared with their control counterparts. Zero maze and DigiGait analyses indicated reduced and aberrant movement associated emotional activity compared with sham control at 8-weeks postinjection. The μCT analysis showed osteolytic and osteoblastic changes and a 50% reduction of the trabecular volumes within the prostate cancer group. Neurologically we demonstrated, increased calcitonin gene-related peptide (CGRP) and neuronal p75NTR immune-reactivities in both the projected terminals of the superficial dorsal horn and partial afferent neurons in DRG at L2 to L4 level in tumor-bearing mice. Furthermore, our data show elevated nerve growth factor (NGF) and TrkA immunoreactivities in the same segment of the superficial dorsal horn that were, however, not colocalized with CGRP and p75NTR . Conclusions This study describes a novel immunocompetent model of CIBP and demonstrates the contribution of NGF and p75NTR to chronic pain in bone metastasis.

5 citations

Journal ArticleDOI
TL;DR: The Animal Models and Co-clinical Trials (AMCT) Working Group as discussed by the authors was established by the National Cancer Institute (NCI) to establish best practices for co-clinical imaging and to optimize translational quantitative imaging methodologies.
Abstract: The availability of high-fidelity animal models for oncology research has grown enormously in recent years, enabling preclinical studies relevant to prevention, diagnosis, and treatment of cancer to be undertaken. This has led to increased opportunities to conduct co-clinical trials, which are studies on patients that are carried out parallel to or sequentially with animal models of cancer that mirror the biology of the patients’ tumors. Patient-derived xenografts (PDX) and genetically engineered mouse models (GEMM) are considered to be the models that best represent human disease and have high translational value. Notably, one element of co-clinical trials that still needs significant optimization is quantitative imaging. The National Cancer Institute has organized a Co-Clinical Imaging Resource Program (CIRP) network to establish best practices for co-clinical imaging and to optimize translational quantitative imaging methodologies. This overview describes the ten co-clinical trials of investigators from eleven institutions who are currently supported by the CIRP initiative and are members of the Animal Models and Co-clinical Trials (AMCT) Working Group. Each team describes their corresponding clinical trial, type of cancer targeted, rationale for choice of animal models, therapy, and imaging modalities. The strengths and weaknesses of the co-clinical trial design and the challenges encountered are considered. The rich research resources generated by the members of the AMCT Working Group will benefit the broad research community and improve the quality and translational impact of imaging in co-clinical trials.

2 citations

Journal ArticleDOI
TL;DR: Though PDTX do more faithfully recapitulate human cancers than cancer cell lines, more there is a growing interest on PDTX relates to the pressing need of cancer models that faithfully mimic primary human neoplasms.
Abstract: Over the past few decades, the scientific and translational progress in the arena of oncology has fostered the development of many “Precision Medicine” programs across the world. Next‐generation sequencing such as whole genome sequencing and global transcriptomics and more recently single‐cell sequencing have elucidated just a fraction of the complexity and heterogeneity of tumor pathobiology, in tumorigenic events, natural evolution, and response to the selective pressure of conventional and target therapies. The poor efficacy of the current drug discovery programs and the enormous costs sustained to perform obsolete clinical trials have further complicated matters. These new perspectives have led to the formulation of a number of questions, whose solutions require new models and innovative approaches—one of which is patient‐derived tumor xenografts (PDTX). Innovative mouse models will play a critical role, and the National Cancer Institutes have taken note of this with their announcement that they will establish a repository of clinically annotated PDTX for clinic‐therapeutic purposes. Patient‐derived tumor xenografts will become the main models for NCI‐sponsored drug‐screening programs, eventually replacing NCI‐60 human cancer cell lines. Similar initiatives have arisen elsewhere and will hopefully merge into integrated networks to power basic science and clinical oncology. Such a growing interest on PDTX relates to the pressing need of cancer models that faithfully mimic primary human neoplasms. This stems from the notion that individual cancers cannot be fully reproduced by classical in vitro and in vivo tumor models. In the last 25 years, industries and academic centers have screened more than 100 000 compounds. However, only 1 in 10 000 reaches the clinical arena and ultimately is commercialised. There is a general agreement that although many issues contribute to the current inefficient drug discovery pipelines, deficiencies in in vivo models add substantially to the low rate of success. Though PDTX do more faithfully recapitulate human cancers than cancer cell lines, more

1 citations


Cites background from "PCSD1, a new patient-derived model ..."

  • ...In particular, highly aggressive carcinomas or colorectal and lung neoplasms have been proven to engraft efficiently, although the generation of PDTX from selective neoplasms remains largely challenging (ER/PR positive breast cancers) or virtually impossible (prostate cancers).(14-19) In the case of melanoma, Quitana et al(20) have originally demonstrated that PDTX can be established by a single cancer cell, and in the case of small‐cell lung cancers(21) and prostate(22) cancers, blood‐circulating tumor cells can be expanded in immunocompromised mice....

    [...]

References
More filters
Journal ArticleDOI
TL;DR: The magnitude of the decline in cancer death rates from 1991 to 2010 varies substantially by age, race, and sex, ranging from no decline among white women aged 80 years and older to a 55% decline among black men aged 40 years to 49 years.
Abstract: Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States in the current year and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data were collected by the National Cancer Institute, the Centers for Disease Control and Prevention, and the North American Association of Central Cancer Registries and mortality data were collected by the National Center for Health Statistics. A total of 1,665,540 new cancer cases and 585,720 cancer deaths are projected to occur in the United States in 2014. During the most recent 5 years for which there are data (2006-2010), delay-adjusted cancer incidence rates declined slightly in men (by 0.6% per year) and were stable in women, while cancer death rates decreased by 1.8% per year in men and by 1.4% per year in women. The combined cancer death rate (deaths per 100,000 population) has been continuously declining for 2 decades, from a peak of 215.1 in 1991 to 171.8 in 2010. This 20% decline translates to the avoidance of approximately 1,340,400 cancer deaths (952,700 among men and 387,700 among women) during this time period. The magnitude of the decline in cancer death rates from 1991 to 2010 varies substantially by age, race, and sex, ranging from no decline among white women aged 80 years and older to a 55% decline among black men aged 40 years to 49 years. Notably, black men experienced the largest drop within every 10-year age group. Further progress can be accelerated by applying existing cancer control knowledge across all segments of the population.

10,829 citations

Journal ArticleDOI
TL;DR: The inhibition of androgen biosynthesis by abiraterone acetate prolonged overall survival among patients with metastatic castration-resistant prostate cancer who previously received chemotherapy.
Abstract: BACKGROUND Biosynthesis of extragonadal androgen may contribute to the progression of castration-resistant prostate cancer. We evaluated whether abiraterone acetate, an inhibitor of androgen biosynthesis, prolongs overall survival among patients with metastatic castration-resistant prostate cancer who have received chemotherapy. METHODS We randomly assigned, in a 2:1 ratio, 1195 patients who had previously received docetaxel to receive 5 mg of prednisone twice daily with either 1000 mg of abiraterone acetate (797 patients) or placebo (398 patients). The primary end point was overall survival. The secondary end points included time to prostate-specific antigen (PSA) progression (elevation in the PSA level according to prespecified criteria), progression-free survival according to radiologic findings based on prespecified criteria, and the PSA response rate. RESULTS After a median follow-up of 12.8 months, overall survival was longer in the abiraterone acetate–prednisone group than in the placebo–prednisone group (14.8 months vs. 10.9 months; hazard ratio, 0.65; 95% confidence interval, 0.54 to 0.77; P<0.001). Data were unblinded at the interim analysis, since these results exceeded the preplanned criteria for study termination. All secondary end points, including time to PSA progression (10.2 vs. 6.6 months; P<0.001), progression-free survival (5.6 months vs. 3.6 months; P<0.001), and PSA response rate (29% vs. 6%, P<0.001), favored the treatment group. Mineralocorticoid-related adverse events, including fluid retention, hypertension, and hypokalemia, were more frequently reported in the abiraterone acetate–prednisone group than in the placebo–prednisone group. CONCLUSIONS The inhibition of androgen biosynthesis by abiraterone acetate prolonged overall survival among patients with metastatic castration-resistant prostate cancer who previously received chemotherapy. (Funded by Cougar Biotechnology; COU-AA-301 ClinicalTrials.gov number, NCT00638690.)

3,875 citations


"PCSD1, a new patient-derived model ..." refers background in this paper

  • ...Both have been shown to improve survival [8-11], however, eventually, resistance develops to these as well, perhaps through mutations in the androgen receptor [12]....

    [...]

Journal ArticleDOI
TL;DR: Enzalutamide significantly prolonged the survival of men with metastatic castration-resistant prostate cancer after chemotherapy, and was shown with respect to all secondary end points.
Abstract: Background Enzalutamide (formerly called MDV3100) targets multiple steps in the androgen-receptor–signaling pathway, the major driver of prostate-cancer growth. We aimed to evaluate whether enzalutamide prolongs survival in men with castration-resistant prostate cancer after chemotherapy. Methods In our phase 3, double-blind, placebo-controlled trial, we stratified 1199 men with castration-resistant prostate cancer after chemotherapy according to the Eastern Cooperative Oncology Group performance-status score and pain intensity. We randomly assigned them, in a 2:1 ratio, to receive oral enzalutamide at a dose of 160 mg per day (800 patients) or placebo (399 patients). The primary end point was overall survival. Results The study was stopped after a planned interim analysis at the time of 520 deaths. The median overall survival was 18.4 months (95% confidence interval [CI], 17.3 to not yet reached) in the enzalutamide group versus 13.6 months (95% CI, 11.3 to 15.8) in the placebo group (hazard ratio for de...

3,866 citations

Journal ArticleDOI
08 May 2009-Science
TL;DR: The diarylthiohydantoins RD162 and MDV3100 are characterized, two compounds optimized from a screen for nonsteroidal antiandrogens that retain activity in the setting of increased androgen receptor expression that appear to be promising candidates for treatment of advanced prostate cancer.
Abstract: Metastatic prostate cancer is treated with drugs that antagonize androgen action, but most patients progress to a more aggressive form of the disease called castration-resistant prostate cancer, driven by elevated expression of the androgen receptor. Here we characterize the diarylthiohydantoins RD162 and MDV3100, two compounds optimized from a screen for nonsteroidal antiandrogens that retain activity in the setting of increased androgen receptor expression. Both compounds bind to the androgen receptor with greater relative affinity than the clinically used antiandrogen bicalutamide, reduce the efficiency of its nuclear translocation, and impair both DNA binding to androgen response elements and recruitment of coactivators. RD162 and MDV3100 are orally available and induce tumor regression in mouse models of castration-resistant human prostate cancer. Of the first 30 patients treated with MDV3100 in a Phase I/II clinical trial, 13 of 30 (43%) showed sustained declines (by >50%) in serum concentrations of prostate-specific antigen, a biomarker of prostate cancer. These compounds thus appear to be promising candidates for treatment of advanced prostate cancer.

2,046 citations


"PCSD1, a new patient-derived model ..." refers background in this paper

  • ...Bicalutamide, one form of androgen deprivation, acts as a competitive inhibitor of androgens by binding the androgen receptor (AR), impairing DNA binding to Androgen Response Elements (ARE), and impairing recruitment of co-activators necessary for testosterone or DHT to impart their proliferative effect on responsive cells [5]....

    [...]

Journal ArticleDOI
TL;DR: Combined pharmacologic inhibition of PI3K and AR signaling caused near-complete prostate cancer regressions in a Pten-deficient murine prostate cancer model and in human prostate cancer xenografts, indicating that both pathways coordinately support survival.

1,083 citations


"PCSD1, a new patient-derived model ..." refers background in this paper

  • ...Signaling cross-talk as a mechanism of castrate-resistance in prostate cancer that alters steroid hormone receptor activity and transcriptional regulation of gene expression has been demonstrated for AR in as well as other steroid hormone receptors such as GR [19-23]....

    [...]

Related Papers (5)