Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Nature ; 606(7915): 791-796, 2022 06.
Article in English | MEDLINE | ID: mdl-35322234

ABSTRACT

Immune checkpoint blockade has revolutionized the field of oncology, inducing durable anti-tumour immunity in solid tumours. In patients with advanced prostate cancer, immunotherapy treatments have largely failed1-5. Androgen deprivation therapy is classically administered in these patients to inhibit tumour cell growth, and we postulated that this therapy also affects tumour-associated T cells. Here we demonstrate that androgen receptor (AR) blockade sensitizes tumour-bearing hosts to effective checkpoint blockade by directly enhancing CD8 T cell function. Inhibition of AR activity in CD8 T cells prevented T cell exhaustion and improved responsiveness to PD-1 targeted therapy via increased IFNγ expression. AR bound directly to Ifng and eviction of AR with a small molecule significantly increased cytokine production in CD8 T cells. Together, our findings establish that T cell intrinsic AR activity represses IFNγ expression and represents a novel mechanism of immunotherapy resistance.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy , Prostatic Neoplasms , Receptors, Androgen , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Interferon-gamma , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Treatment Failure
2.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35046029

ABSTRACT

Cells are continuously exposed to potentially dangerous compounds. Progressive accumulation of damage is suspected to contribute to neurodegenerative diseases and aging, but the molecular identity of the damage remains largely unknown. Here we report that PARK7, an enzyme mutated in hereditary Parkinson's disease, prevents damage of proteins and metabolites caused by a metabolite of glycolysis. We found that the glycolytic metabolite 1,3-bisphosphoglycerate (1,3-BPG) spontaneously forms a novel reactive intermediate that avidly reacts with amino groups. PARK7 acts by destroying this intermediate, thereby preventing the formation of proteins and metabolites with glycerate and phosphoglycerate modifications on amino groups. As a consequence, inactivation of PARK7 (or its orthologs) in human cell lines, mouse brain, and Drosophila melanogaster leads to the accumulation of these damaged compounds, most of which have not been described before. Our work demonstrates that PARK7 function represents a highly conserved strategy to prevent damage in cells that metabolize carbohydrates. This represents a fundamental link between metabolism and a type of cellular damage that might contribute to the development of Parkinson's disease.


Subject(s)
Glucose/metabolism , Protein Deglycase DJ-1/genetics , Protein Deglycase DJ-1/metabolism , Animals , Biomarkers , Carbohydrate Metabolism , Chromatography, Liquid , Drosophila melanogaster , Gene Knockdown Techniques , Glyceric Acids/metabolism , Glycolysis , Humans , Mass Spectrometry , Metabolic Networks and Pathways , Metabolome , Metabolomics/methods , Mice , Parkinson Disease/etiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Deglycase DJ-1/chemistry
3.
J Biol Chem ; 299(9): 105095, 2023 09.
Article in English | MEDLINE | ID: mdl-37507022

ABSTRACT

Many transcripts are targeted by nonsense-mediated decay (NMD), leading to their degradation and the inhibition of their translation. We found that the protein SUZ domain-containing protein 1 (SZRD1) interacts with the key NMD factor up-frameshift 1. When recruited to NMD-sensitive reporter gene transcripts, SZRD1 increased protein production, at least in part, by relieving translational inhibition. The conserved SUZ domain in SZRD1 was required for this effect. The SUZ domain is present in only three other human proteins besides SZRD1: R3H domain-containing protein 1 and 2 (R3HDM1, R3HDM2) and cAMP-regulated phosphoprotein 21 (ARPP21). We found that ARPP21, similarly to SZRD1, can increase protein production from NMD-sensitive reporter transcripts in an SUZ domain-dependent manner. This indicated that the SUZ domain-containing proteins could prevent translational inhibition of transcripts targeted by NMD. Consistent with the idea that SZRD1 mainly prevents translational inhibition, we did not observe a systematic decrease in the abundance of NMD targets when we knocked down SZRD1. Surprisingly, knockdown of SZRD1 in two different cell lines led to reduced levels of the NMD component UPF3B, which was accompanied by increased levels in a subset of NMD targets. This suggests that SZRD1 is required to maintain normal UPF3B levels and indicates that the effect of SZRD1 on NMD targets is not limited to a relief from translational inhibition. Overall, our study reveals that human SUZ domain-containing proteins play a complex role in regulating protein output from transcripts targeted by NMD.


Subject(s)
Nonsense Mediated mRNA Decay , RNA-Binding Proteins , Humans , Cell Line , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Protein Domains , HeLa Cells , HEK293 Cells
4.
Br J Cancer ; 130(1): 73-81, 2024 01.
Article in English | MEDLINE | ID: mdl-37951974

ABSTRACT

BACKGROUND: Apalutamide plus androgen-deprivation therapy (ADT) improved outcomes in metastatic castration-sensitive prostate cancer (mCSPC) and non-metastatic castration-resistant PC (nmCRPC) in the Phase 3 randomised TITAN and SPARTAN studies, respectively, and maintained health-related quality of life (HRQoL). Apalutamide treatment effect by patient age requires assessment. METHODS: Post-hoc analysis assessed patients receiving 240 mg/day apalutamide (525 TITAN and 806 SPARTAN) or placebo (527 TITAN and 401 SPARTAN) with ongoing ADT, stratified by age groups. Prostate-specific antigen declines, radiographic progression-free survival, metastasis-free survival, overall survival (OS), HRQoL and safety were assessed using descriptive statistics, Kaplan-Meier method, Cox proportional-hazards model and mixed-effects model for repeated measures. RESULTS: Hazard ratios (95% confidence intervals) generally favoured apalutamide plus ADT versus ADT alone across all endpoints regardless of age; e.g., OS values were 0.57 (0.40-0.80), 0.70 (0.54-0.91) and 0.74 (0.40-1.39) (TITAN) and 0.39 (0.19-0.78), 0.89 (0.69-1.16) and 0.81 (0.58-1.15) (SPARTAN) in patients aged <65, 65-79 and ≥80 years. Regardless of age, apalutamide also maintained HRQoL and was tolerated well with a potential trend in rates of adverse events increasing with age. Limitations include post-hoc nature and variability in sample size of age groups. CONCLUSIONS: Apalutamide plus ADT was an effective and well-tolerated option maintaining HRQoL in patients with mCSPC and nmCRPC regardless of age. CLINICAL TRIAL REGISTRATION: TITAN (NCT02489318); SPARTAN (NCT01946204).


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Male , Humans , Aged , Prostatic Neoplasms, Castration-Resistant/pathology , Androgen Antagonists/therapeutic use , Quality of Life , Thiohydantoins/adverse effects
5.
N Engl J Med ; 378(15): 1408-1418, 2018 Apr 12.
Article in English | MEDLINE | ID: mdl-29420164

ABSTRACT

BACKGROUND: Apalutamide, a competitive inhibitor of the androgen receptor, is under development for the treatment of prostate cancer. We evaluated the efficacy of apalutamide in men with nonmetastatic castration-resistant prostate cancer who were at high risk for the development of metastasis. METHODS: We conducted a double-blind, placebo-controlled, phase 3 trial involving men with nonmetastatic castration-resistant prostate cancer and a prostate-specific antigen doubling time of 10 months or less. Patients were randomly assigned, in a 2:1 ratio, to receive apalutamide (240 mg per day) or placebo. All the patients continued to receive androgen-deprivation therapy. The primary end point was metastasis-free survival, which was defined as the time from randomization to the first detection of distant metastasis on imaging or death. RESULTS: A total of 1207 men underwent randomization (806 to the apalutamide group and 401 to the placebo group). In the planned primary analysis, which was performed after 378 events had occurred, median metastasis-free survival was 40.5 months in the apalutamide group as compared with 16.2 months in the placebo group (hazard ratio for metastasis or death, 0.28; 95% confidence interval [CI], 0.23 to 0.35; P<0.001). Time to symptomatic progression was significantly longer with apalutamide than with placebo (hazard ratio, 0.45; 95% CI, 0.32 to 0.63; P<0.001). The rate of adverse events leading to discontinuation of the trial regimen was 10.6% in the apalutamide group and 7.0% in the placebo group. The following adverse events occurred at a higher rate with apalutamide than with placebo: rash (23.8% vs. 5.5%), hypothyroidism (8.1% vs. 2.0%), and fracture (11.7% vs. 6.5%). CONCLUSIONS: Among men with nonmetastatic castration-resistant prostate cancer, metastasis-free survival and time to symptomatic progression were significantly longer with apalutamide than with placebo. (Funded by Janssen Research and Development; SPARTAN ClinicalTrials.gov number, NCT01946204 .).


Subject(s)
Adenocarcinoma/drug therapy , Androgen Antagonists/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Thiohydantoins/therapeutic use , Adenocarcinoma/secondary , Aged , Aged, 80 and over , Androgen Antagonists/adverse effects , Disease Progression , Disease-Free Survival , Double-Blind Method , Exanthema/chemically induced , Humans , Male , Middle Aged , Neoplasm Metastasis/prevention & control , Proportional Hazards Models , Prostate-Specific Antigen , Prostatic Neoplasms, Castration-Resistant/pathology , Thiohydantoins/adverse effects
6.
Future Oncol ; 17(23): 3017-3026, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34044584

ABSTRACT

Current treatment options for men with metastatic castration-resistant prostate cancer (mCRPC) are noncurative, and median survival upon development of mCRPC is approximately 3 years. The novel hormonal agent enzalutamide has an established role in the mCRPC treatment paradigm, and emerging evidence suggests potential synergism with enzalutamide and the PD-1 inhibitor pembrolizumab in men with mCRPC. Here, we describe the design and rationale for the multicenter, randomized, double-blind, Phase III KEYNOTE-641 study, which will be conducted to compare the efficacy and safety of pembrolizumab plus enzalutamide with that of enzalutamide plus placebo in mCRPC. Clinical trial registration: NCT03834493 (ClinicalTrials.gov).


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzamides/administration & dosage , Nitriles/administration & dosage , Phenylthiohydantoin/administration & dosage , Prostatic Neoplasms, Castration-Resistant/drug therapy , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzamides/adverse effects , Clinical Trials, Phase III as Topic , Double-Blind Method , Humans , Male , Middle Aged , Multicenter Studies as Topic , Nitriles/adverse effects , Phenylthiohydantoin/adverse effects , Placebos/administration & dosage , Placebos/adverse effects , Progression-Free Survival , Prostatic Neoplasms, Castration-Resistant/mortality , Prostatic Neoplasms, Castration-Resistant/pathology , Randomized Controlled Trials as Topic
7.
Acta Orthop ; 92(2): 208-214, 2021 04.
Article in English | MEDLINE | ID: mdl-33228428

ABSTRACT

Background and purpose - Focus on prevention, surveillance, and treatment of infection after total hip arthroplasty (THA) in the last decade has resulted in new knowledge and guidelines. Previous publications have suggested an increased incidence of surgical revisions due to infection after THA. We assessed whether there have been changes in the risk of revision due to deep infection after primary THAs reported to the Norwegian Arthroplasty Register (NAR) over the period 2005-2019.Patients and methods - Primary THAs reported to the NAR from January 1, 2005 to December 31, 2019 were included. Adjusted Cox regression analyses with the first revision due to deep infection after primary THA were performed. We investigated changes in the risk of revision as a function of time of primary THA. Time was stratified into 5-year periods. We studied the whole population of THAs, and the subgroups: all-cemented, all-uncemented, reverse hybrid (cemented cup), and hybrid THAs (cemented stem). In addition, we investigated factors that were associated with the risk of revision, and changes in the time span from primary THA to revision.Results - Of the 108,854 primary THAs that met the inclusion criteria, 1,365 (1.3%) were revised due to deep infection. The risk of revision due to infection, at any time after primary surgery, increased through the period studied. Compared with THAs implanted in 2005-2009, the relative risk of revision due to infection was 1.4 (95% CI 1.2-1.7) for 2010-2014, and 1.6 (1.1-1.9) for 2015-2019. We found an increased risk for all types of implant fixation. Compared to 2005-2009, for all THAs, the risk of revision due to infection 0-30 days postoperatively was 2.2 (1.8-2.8) for 2010-2014 and 2.3 (1.8-2.9) for 2015-2019, 31-90 days postoperatively 1.0 (0.7-1.6) for 2010-2014 and 1.6 (1.0-2.5) for 2015-2019, and finally 91 days-1 year postoperatively 1.1 (0.7-1.8) for 2010-2014 and 1.6 (1.0-2.6) for 2015-2019. From 1 to 5 years postoperatively, the risk of revision due to infection was similar to 2005-2009 for both the subsequent time periodsInterpretation - The risk of revision due to deep infection after THA increased throughout the period 2005-2019, but appears to have levelled out after 2010. The increase was mainly due to an increased risk of early revisions, and may partly have been caused by a change of practice rather than a change in the incidence of infection.


Subject(s)
Arthroplasty, Replacement, Hip , Prosthesis-Related Infections/epidemiology , Prosthesis-Related Infections/surgery , Reoperation , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Norway/epidemiology , Postoperative Complications/epidemiology , Postoperative Complications/prevention & control , Postoperative Complications/surgery , Prosthesis-Related Infections/prevention & control , Registries , Risk Factors , Time Factors
8.
Oncology (Williston Park) ; 34(3)2020 Mar 19.
Article in English | MEDLINE | ID: mdl-32212131

ABSTRACT

Indications for checkpoint inhibitors (CPIs) are growing rapidly within the field of oncology; however, they continue to have heterogeneous outcomes in different cancers. Other than mismatch repair deficiency, there are no consistent tests to determine a tumor's susceptibility. By exploring factors beyond the cancer cell, researchers have learned that the efficacy of CPIs may be governed by a myriad of variable host factors, including the tumor microenvironment (TME) and gut microbiome (GMB). The GMB serves as one of the primary organs of immune defense and has well-established local and systemic effects on the host immune system. Recent investigations suggest that the GMB also affects the TME. This review article discusses the concepts of a TME and a GMB and their effects on responses to CPIs. It also reviews recent research investigating these 3 topics, and how it can be applied to using CPIs in prostate cancer. By highlighting this important pathophysiologic process, we hope to provide insight into a possible explanation for differences in interindividual response to CPIs, discuss a potential method for transferring treatment efficacy between patients, and propose a method for expanding the use of CPIs to prostate cancer.


Subject(s)
Gastrointestinal Microbiome , Immune Checkpoint Inhibitors/therapeutic use , Prostatic Neoplasms/microbiology , Prostatic Neoplasms/therapy , Clinical Trials, Phase III as Topic , Humans , Male , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Tumor Microenvironment/immunology
9.
Biochem J ; 476(4): 629-643, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30670572

ABSTRACT

Repair of a certain type of oxidative DNA damage leads to the release of phosphoglycolate, which is an inhibitor of triose phosphate isomerase and is predicted to indirectly inhibit phosphoglycerate mutase activity. Thus, we hypothesized that phosphoglycolate might play a role in a metabolic DNA damage response. Here, we determined how phosphoglycolate is formed in cells, elucidated its effects on cellular metabolism and tested whether DNA damage repair might release sufficient phosphoglycolate to provoke metabolic effects. Phosphoglycolate concentrations were below 5 µM in wild-type U2OS and HCT116 cells and remained unchanged when we inactivated phosphoglycolate phosphatase (PGP), the enzyme that is believed to dephosphorylate phosphoglycolate. Treatment of PGP knockout cell lines with glycolate caused an up to 500-fold increase in phosphoglycolate concentrations, which resulted largely from a side activity of pyruvate kinase. This increase was much higher than in glycolate-treated wild-type cells and was accompanied by metabolite changes consistent with an inhibition of phosphoglycerate mutase, most likely due to the removal of the priming phosphorylation of this enzyme. Surprisingly, we found that phosphoglycolate also inhibits succinate dehydrogenase with a Ki value of <10 µM. Thus, phosphoglycolate can lead to profound metabolic disturbances. In contrast, phosphoglycolate concentrations were not significantly changed when we treated PGP knockout cells with Bleomycin or ionizing radiation, which are known to lead to the release of phosphoglycolate by causing DNA damage. Thus, phosphoglycolate concentrations due to DNA damage are too low to cause major metabolic changes in HCT116 and U2OS cells.


Subject(s)
DNA, Neoplasm , Glycolates , Neoplasm Proteins , Neoplasms , Phosphoric Monoester Hydrolases , Succinate Dehydrogenase , DNA Damage , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Glycolates/metabolism , Glycolates/pharmacology , HCT116 Cells , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism
10.
Lancet Oncol ; 19(10): 1404-1416, 2018 10.
Article in English | MEDLINE | ID: mdl-30213449

ABSTRACT

BACKGROUND: In the SPARTAN trial, addition of apalutamide to androgen deprivation therapy, as compared with placebo plus androgen deprivation therapy, significantly improved metastasis-free survival in men with non-metastatic castration-resistant prostate cancer who were at high risk for development of metastases. We aimed to investigate the effects of apalutamide versus placebo added to androgen deprivation therapy on health-related quality of life (HRQOL). METHODS: SPARTAN is a multicentre, international, randomised, phase 3 trial. Participants were aged 18 years or older, with non-metastatic castration-resistant prostate cancer, a prostate-specific antigen doubling time of 10 months or less, and a prostate-specific antigen concentration of 2 ng/mL or more in serum. Patients were randomly assigned (2:1) to 240 mg oral apalutamide per day plus androgen deprivation therapy, or matched oral placebo plus androgen deprivation therapy, using an interactive voice randomisation system. Permuted block randomisation was used according to the three baseline stratification factors: prostate-specific antigen doubling time (>6 months vs ≤6 months), use of bone-sparing drugs (yes vs no), and presence of local-regional nodal disease (N0 vs N1). Each treatment cycle was 28 days. The primary endpoint was metastasis-free survival. The trial was unblinded in July, 2017. In this prespecified exploratory analysis we assessed HRQOL using the Functional Assessment of Cancer Therapy-Prostate (FACT-P) and EQ-5D-3L questionnaires, which we collected at baseline, day 1 of cycle 1 (before dose), day 1 of treatment cycles 1-6, day 1 of every two cycles from cycles 7 to 13, and day 1 of every four cycles thereafter. This study is registered with ClinicalTrials.gov, number NCT01946204. FINDINGS: Between Oct 14, 2013, and Dec 15, 2016, we randomly assigned 1207 patients to receive apalutamide (n=806) or placebo (n=401). The clinical cutoff date, as for the primary analysis, was May 19, 2017. Median follow-up for overall survival was 20·3 months (IQR 14·8-26·6). FACT-P total and subscale scores were associated with a preservation of HRQOL from baseline to cycle 29 in the apalutamide group; there were similar results for EQ-5D-3L. At baseline, the mean for FACT-P total score in both the apalutamide and placebo groups were consistent with the FACT-P general population norm for US adult men. Group mean patient-reported outcome scores over time show that HRQOL was maintained from baseline after initiation of apalutamide treatment and was similar over time among patients receiving apalutamide versus placebo. Least-squares mean change from baseline shows that HRQOL deterioration was more apparent in the placebo group. INTERPRETATION: In asymptomatic men with high-risk non-metastatic castration-resistant prostate cancer, HRQOL was maintained after initiation of apalutamide treatment. Considered with findings from SPARTAN, patients who received apalutamide had longer metastasis-free survival and longer time to symptomatic progression than did those who received placebo, while preserving HRQOL. FUNDING: Janssen Research & Development.


Subject(s)
Androgen Receptor Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Quality of Life , Thiohydantoins/therapeutic use , Aged , Aged, 80 and over , Androgen Receptor Antagonists/adverse effects , Antineoplastic Agents/adverse effects , Humans , Kallikreins/blood , Male , Neoplasm Metastasis , Patient Reported Outcome Measures , Progression-Free Survival , Prostate-Specific Antigen/blood , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/psychology , Thiohydantoins/adverse effects , Time Factors
11.
Nat Chem Biol ; 12(8): 601-7, 2016 08.
Article in English | MEDLINE | ID: mdl-27294321

ABSTRACT

Metabolic enzymes are very specific. However, most of them show weak side activities toward compounds that are structurally related to their physiological substrates, thereby producing side products that may be toxic. In some cases, 'metabolite repair enzymes' eliminating side products have been identified. We show that mammalian glyceraldehyde 3-phosphate dehydrogenase and pyruvate kinase, two core glycolytic enzymes, produce 4-phosphoerythronate and 2-phospho-L-lactate, respectively. 4-Phosphoerythronate strongly inhibits an enzyme of the pentose phosphate pathway, whereas 2-phospho-L-lactate inhibits the enzyme producing the glycolytic activator fructose 2,6-bisphosphate. We discovered that a single, widely conserved enzyme, known as phosphoglycolate phosphatase (PGP) in mammals, dephosphorylates both 4-phosphoerythronate and 2-phospho-L-lactate, thereby preventing a block in the pentose phosphate pathway and glycolysis. Its yeast ortholog, Pho13, similarly dephosphorylates 4-phosphoerythronate and 2-phosphoglycolate, a side product of pyruvate kinase. Our work illustrates how metabolite repair enzymes can make up for the limited specificity of metabolic enzymes and permit high flux in central metabolic pathways.


Subject(s)
Glycolates/metabolism , Glycolysis , Lactates/metabolism , Phosphoric Monoester Hydrolases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Sugar Acids/metabolism , Glycolates/chemistry , Glycolates/toxicity , Glycolysis/drug effects , HCT116 Cells , Humans , Lactates/chemistry , Lactates/toxicity , Pentose Phosphate Pathway/drug effects , Phosphoric Monoester Hydrolases/deficiency , Phosphorylation , Pyruvate Kinase/metabolism , Saccharomyces cerevisiae/enzymology , Substrate Specificity , Sugar Acids/chemistry , Sugar Acids/toxicity
12.
Oncology (Williston Park) ; 32(12): 599-602, 604-6, 2018 12 17.
Article in English | MEDLINE | ID: mdl-30632129

ABSTRACT

Androgen deprivation therapy (ADT) comes in several forms, such as surgical castration or medical castration using gonadotropin-releasing hormone (GnRH) agonist or GnRH antagonist therapy. ADT is a critical treatment for high-risk and metastatic prostate cancer. There are important differences between GnRH agonists and antagonists. Here we review the mechanism of action between GnRH agonists and antagonists and the studies that led to the approval of degarelix. We also comment on the potential risks and benefits of degarelix, particularly when it comes to cardiovascular health. Finally, we describe an oral GnRH antagonist, which is not currently used in prostate cancer, but is included for completeness.


Subject(s)
Androgen Antagonists/therapeutic use , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Antineoplastic Agents, Hormonal/therapeutic use , Humans , Male , Oligopeptides/therapeutic use , Phenylurea Compounds/therapeutic use , Pyrimidinones/therapeutic use
13.
Anticancer Drugs ; 28(1): 120-126, 2017 01.
Article in English | MEDLINE | ID: mdl-27669423

ABSTRACT

The aims of this study were to report the clinical outcomes in a cohort of men with high-risk prostate cancer treated with neoadjuvant docetaxel and mitoxantrone 10 years after treatment, identify pretreatment clinical parameters that may be predictors of recurrence, and describe tumor-infiltrating leukocytes present in radical prostatectomy specimens. We conducted a phase I/II study of neoadjuvant docetaxel and mitoxantrone before radical prostatectomy in high-risk localized prostate cancer to determine the feasibility of this combination and predictors of prostate cancer recurrence after cytotoxic chemotherapy. After 10 years of follow-up, 34 (63%) of 54 participants experience a recurrence. In univariate analysis, prostate-specific antigen (PSA) density (P=0.01), pathological stage (P=0.03), lymph node status (P<0.0001), seminal vesicle invasion (P=0.003), and tissue vascular endothelial growth factor (VEGF) expression (P=0.016) were significantly associated with recurrence. In multivariate analysis, only lymph node status, PSA density, and VEGF expression were significant predictors of disease recurrence. We used a tissue microarray for the first 50 participants to characterize the tumor-infiltrating lymphocytes and evaluate them for association with recurrence. We measured CD3, CD4, CD8, FoxP3, CD20, CD15, CD68, and CD163 by immunohistochemistry in both tumor and normal prostate specimens, but did not find an association between immunophenotype and recurrence. There was a significantly different density of CD68 and CD163 cells between normal and tumor tissue. Lymph node status, PSA density, and tissue VEGF expression predict recurrence after chemotherapy for high-risk prostate cancer. Additional studies are needed to determine the potential benefit of chemotherapy in the neoadjuvant setting.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/surgery , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/surgery , Chemotherapy, Adjuvant , Cohort Studies , Docetaxel , Follow-Up Studies , Humans , Male , Middle Aged , Mitoxantrone/administration & dosage , Neoadjuvant Therapy , Prostatectomy , Risk Factors , Taxoids/administration & dosage
14.
BJU Int ; 119(5): 709-716, 2017 05.
Article in English | MEDLINE | ID: mdl-27480340

ABSTRACT

OBJECTIVE: To evaluate the biological effects of selective cyclooxygenase-2 inhibition on prostate tissue in patients undergoing radical prostatectomy (RP). PATIENTS AND METHODS: Patients with localised prostate cancer were randomised to receive either celecoxib 400 mg twice daily or placebo for 4 weeks before RP. Specimens were analysed for levels of apoptosis, prostaglandins, and androgen receptor (AR). Effects on serum prostate-specific antigen (PSA) and postoperative opioid use were also measured. RESULTS: In all, 28 of 44 anticipated patients enrolled and completed treatment. One patient in the celecoxib arm had a myocardial infarction postoperatively. For this reason, and safety concerns in other studies, enrolment was halted. The apoptosis index (AI) in tumour cells was 0.29% [95% confidence interval (CI) 0.11-0.47%] vs 0.39% (95% CI 0.00-0.84%) in the celecoxib and placebo arms, respectively (P = 0.68). The AI in benign cells was 0.18% (95% CI 0.03-0.32%) vs 0.13% (95% CI 0.00-0.28%) in the celecoxib and placebo arms, respectively (P = 0.67). Prostaglandin E2 and AR levels were similar in cancerous and benign tissues when comparing the two arms. The median baseline PSA level was 6.0 and 6.2 ng/mL for the celecoxib and placebo groups, respectively, and did not significantly change after celecoxib treatment. There was no difference in postoperative opiate usage between arms. CONCLUSION: Celecoxib had no effect on apoptosis, prostaglandins or AR levels in cancerous or benign prostate tissues. These findings coupled with drug safety concerns should serve to limit interest in these selective drugs as chemopreventive agents.


Subject(s)
Celecoxib/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Prostatectomy , Prostatic Neoplasms/prevention & control , Prostatic Neoplasms/surgery , Aged , Aged, 80 and over , Apoptosis/drug effects , Celecoxib/pharmacology , Chemoprevention , Cyclooxygenase 2 Inhibitors/pharmacology , Double-Blind Method , Humans , Male , Middle Aged , Preoperative Care
15.
Cancer ; 122(16): 2524-33, 2016 Aug 15.
Article in English | MEDLINE | ID: mdl-27192055

ABSTRACT

BACKGROUND: This study was designed to determine the safety, tolerability, and pharmacokinetics (PK) of alisertib (MLN8237) in combination with docetaxel and to identify a recommended dose for the combination. METHODS: Adults with metastatic cancer were treated on 21-day cycles with alisertib (10, 20, 30, or 40 mg) twice daily on days 1 to 7 or days 1 to 5 and with docetaxel (75 or 60 mg/m(2) ) on day 1. The primary objectives were to assess the safety and tolerability of the combination and to determine the recommended phase 2 dose (RP2D) for future studies. Secondary objectives included an efficacy assessment and PK analyses of docetaxel and alisertib. RESULTS: Forty-one patients participated. Eight dose levels were explored with various doses of alisertib and docetaxel. The dose-limiting toxicities were neutropenic fever, neutropenia without fever, stomatitis, and urinary tract infection. The RP2D of this combination was 20 mg of alisertib twice daily on days 1 to 7 and intravenous docetaxel at 75 mg/m(2) on day 1 in 21-day cycles. Eight of the 28 patients (29%) who were efficacy-evaluable had objective responses. These included 1 complete response in a patient with bladder cancer, 6 partial responses in patients with castration-resistant prostate cancer, and 1 partial response in a patient with angiosarcoma. Concomitant administration of alisertib did not produce any clinically meaningful change in docetaxel PK. CONCLUSIONS: Alisertib at 20 mg twice daily on days 1 to 7 with intravenous docetaxel at 75 mg/m(2) on day 1 in a 21-day cycle was well tolerated, and the combination demonstrated antitumor activity. Cancer 2016;122:2524-33. © 2016 American Cancer Society.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Aurora Kinase A/antagonists & inhibitors , Azepines/administration & dosage , Azepines/pharmacokinetics , Biomarkers , Docetaxel , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Multimodal Imaging , Neoplasm Grading , Neoplasm Staging , Neoplasms/diagnosis , Neoplasms/mortality , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Taxoids/administration & dosage , Taxoids/pharmacokinetics , Treatment Outcome
16.
Oncology (Williston Park) ; 30(4): 336-44, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27085332

ABSTRACT

Patients with nonmetastatic castration-resistant prostate cancer (CRPC) are a heterogeneous population with regard to risk of progression to metastatic disease. Treatment selection for nonmetastatic CRPC must balance the risk of disease progression with the side effects of available therapies. Several medication classes are available for use in treating these patients, and other newer agents are under active clinical trial investigation. Here we review current therapies for nonmetastatic CRPC and discuss the recently completed and ongoing trials for this emerging disease state.


Subject(s)
Prostatic Neoplasms, Castration-Resistant/drug therapy , Androgen Antagonists/therapeutic use , Clinical Trials as Topic , Humans , Male , Prostate-Specific Antigen/blood
17.
Invest New Drugs ; 33(2): 480-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25431127

ABSTRACT

INTRODUCTION: Diets high in cruciferous vegetables are associated with lower risk of incidence of prostate cancer, including aggressive forms of this disease. Human intervention studies with cruciferous vegetable-rich diets also demonstrate modulation of gene expression in important pathways in prostate cells. PURPOSE: Sulforaphane is a constituent of these foods postulated to harbor the anti-neoplastic activity based on multiple tumor models. Our own work demonstrates that sulforaphane inhibits AR signaling in prostate cancer cells. Here, we report results from the first clinical trial of sulforaphane-rich extracts in men with prostate cancer. METHODS: We treated 20 patients who had recurrent prostate cancer with 200 µmoles/day of sulforaphane-rich extracts for a maximum period of 20 weeks and determined the proportion of patients with ≥50% PSA declines, the primary endpoint. Only one subject experienced a ≥50% PSA decline. Thus, the primary endpoint was not achieved. Seven patients experienced smaller PSA declines (<50%). There was also a significant lengthening of the on-treatment PSA doubling time (PSADT) compared with the pre-treatment PSADT [6.1 months pre-treatment vs. 9.6 months on-treatment (p = 0.044)]. Finally, treatment with sulforaphane-rich extracts was safe with no Grade 3 adverse events. CONCLUSIONS: Treatment with 200 µmoles/day of sulforaphane-rich extracts did not lead to ≥50% PSA declines in the majority of patients. However, because of the safety of treatment and the effects on PSADT modulation, further studies, including those with higher doses, may be warranted to clarify the role of sulforaphane as a prevention agent or treatment agent.


Subject(s)
Brassica , Isothiocyanates/chemistry , Plant Extracts/pharmacology , Prostatic Neoplasms/drug therapy , Area Under Curve , Chromatography, Liquid , Dose-Response Relationship, Drug , Glutathione Transferase/genetics , Half-Life , Humans , Male , Metabolic Clearance Rate , Neoplasm Recurrence, Local , Plant Extracts/pharmacokinetics , Prostate-Specific Antigen , Sulfoxides , Tandem Mass Spectrometry
18.
Psychooncology ; 24(12): 1686-93, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25753507

ABSTRACT

OBJECTIVE: Prostate cancer patients are at increased risk of depression yet there is no standard intervention to address this. The purpose of this meta-analysis is to examine the efficacy of interventions in reducing depressive symptoms in men with prostate cancer. METHODS: Searches for studies were conducted in four databases and by hand. Randomized controlled trials of any intervention relative to control for depression in prostate cancer patients at any stage of their cancer treatment were included. RESULTS: We identified 11 studies that randomized men with prostate cancer to either an intervention meant to improve some aspect of quality of life or control and reported depressive symptoms scores before and after the intervention or control condition. Two of these were not used in our meta-analysis either for concerns about quality or for lack of depression scores. The interventions identified in the remaining nine articles were exercise (four), information (three), psychotherapy or peer support (three), massage therapy (one), and medication (one). Several publications included more than one type of intervention. A meta-analysis of all studies showed that an intervention of some types significantly improved depressive symptom scores relative to the control condition (improvement in depression score by -0.86 unit (95% CI: -1.42, -0.31)). Isolating the peer support/psychotherapy studies also showed significant improvement (improvement in depression score by -1.09 unit (95% CI: -2.05, -0.13)). CONCLUSION: Treatments to improve depressive symptoms in men with prostate cancer may be effective, with the best evidence supporting the use of peer support/psychotherapy.


Subject(s)
Depression/prevention & control , Prostatic Neoplasms/psychology , Humans , Male , Peer Group , Prostatic Neoplasms/therapy , Psychotherapy , Randomized Controlled Trials as Topic , Social Support
19.
Oncology (Williston Park) ; 29(6): 416-23, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26091674

ABSTRACT

Skeletal-related events contribute substantially to morbidity, mortality and cost in men with metastatic castration-resistant prostate cancer (mCRPC). There are five agents available for treatment in mCRPC that reduce skeletal-related events. Here we discuss the efficacy and safety of zoledronic acid, denosumab, enzalutamide, abiraterone, and radium-223. We include data on and a discussion of duration of treatment with zoledronic acid and denosumab, the only two of these agents that do not have a clinically proven anticancer effect. Finally, we review the available data regarding the cost of denosumab compared with that of zoledronic acid.


Subject(s)
Bone Neoplasms/prevention & control , Fractures, Spontaneous/prevention & control , Pain/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Spinal Cord Compression/prevention & control , Androgen Receptor Antagonists/therapeutic use , Androstenes/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Benzamides , Bone Density Conservation Agents/therapeutic use , Bone Neoplasms/secondary , Cost-Benefit Analysis , Denosumab , Diphosphonates/therapeutic use , Enzyme Inhibitors/therapeutic use , Humans , Imidazoles/therapeutic use , Male , Nitriles , Pain/etiology , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Quality of Life , Radioisotopes/therapeutic use , Radium/therapeutic use , Zoledronic Acid
20.
Res Sq ; 2024 May 30.
Article in English | MEDLINE | ID: mdl-38854112

ABSTRACT

Background: Androgen deprivation therapy (ADT) increases the risk of frailty, falls, and, poor physical functioning in prostate cancer survivors. Detection of frailty is limited to self-report instruments and performance measures, so unbiased tools are needed. We investigated relationships between an unbiased measure - daily life mobility - and ADT history, frailty, falls, and functioning in ADT-treated prostate cancer survivors. Methods: ADT-treated prostate cancer survivors (N=99) were recruited from an exercise clinical trial, an academic medical center, and the community. Participants completed performance measures and surveys to assess frailty, fall history, and physical functioning, then wore instrumented socks to continuously monitor daily life mobility. We performed a principal component analysis on daily life mobility metrics and used regression analyses to investigate relationships between domains of daily life mobility and frailty, fall history, and physical functioning. Results: Daily life mobility metrics clustered into four domains: Gait Pace, Rhythm, Activity, and Balance. Worse scores on Rhythm and Activity were associated with increased odds of frailty (OR 1.59, 95% CI: 1.04, 2.49 and OR 1.81, 95% CI: 1.19, 2.83, respectively). A worse score on Rhythm was associated with increased odds of ≥1 falls in the previous year (OR 1.60, 95% CI: 1.05, 2.47). Worse scores on Gait Pace, Rhythm, and Activity were associated with worse physical functioning. Mobility metrics were similar between current and past users of ADT. Conclusions: Continuous passive monitoring of daily life mobility may identify prostate cancer survivors who have or are developing risk for frailty, falls, and declines in physical functioning.

SELECTION OF CITATIONS
SEARCH DETAIL