Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Angiogenesis ; 25(1): 47-55, 2022 02.
Article in English | MEDLINE | ID: mdl-34028627

ABSTRACT

Hypertension is a common toxicity induced by bevacizumab and other antiangiogenic drugs. There are no biomarkers to predict the risk of bevacizumab-induced hypertension. This study aimed to identify plasma proteins related to the function of the vasculature to predict the risk of severe bevacizumab-induced hypertension. Using pretreated plasma samples from 398 bevacizumab-treated patients in two clinical trials (CALGB 80303 and 90401), the levels of 17 proteins were measured via ELISA. The association between proteins and grade 3 bevacizumab-induced hypertension was performed by calculating the odds ratio (OR) from logistic regression adjusting for age, sex, and clinical trial. Using the optimal cut-point of each protein, sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) for hypertension were estimated. Five proteins showed no difference in levels between clinical trials and were used for analyses. Lower levels of angiopoietin-2 (p = 0.0013, OR 3.41, 95% CI 1.67-7.55), VEGF-A (p = 0.0008, OR 4.25, 95% CI 1.93-10.72), and VCAM-1 (p = 0.0067, OR 2.68, 95% CI 1.34-5.63) were associated with an increased risk of grade 3 hypertension. The multivariable model suggests independent effects of angiopoietin-2 (p = 0.0111, OR 2.71, 95% CI 1.29-6.10), VEGF-A (p = 0.0051, OR 3.66, 95% CI 1.54-9.73), and VCAM-1 (p = 0.0308, OR 2.27, 95% CI 1.10-4.92). The presence of low levels of 2-3 proteins had an OR of 10.06 (95% CI 3.92-34.18, p = 1.80 × 10-5) for the risk of hypertension, with sensitivity of 89.7%, specificity of 53.5%, PPV of 17.3%, and NPV of 97.9%. This is the first study providing evidence of plasma proteins with potential value to predict patients at risk of developing bevacizumab-induced hypertension.Clinical trial registration: ClinicalTrials.gov Identifier: NCT00088894 (CALGB 80303); and NCT00110214 (CALGB 90401).


Subject(s)
Hypertension , Pharmaceutical Preparations , Angiogenesis Inhibitors/adverse effects , Angiopoietin-2 , Bevacizumab/adverse effects , Humans , Hypertension/chemically induced , Vascular Cell Adhesion Molecule-1 , Vascular Endothelial Growth Factor A
2.
Br J Cancer ; 126(2): 265-274, 2022 02.
Article in English | MEDLINE | ID: mdl-34616010

ABSTRACT

BACKGROUND: Hypertension and proteinuria are common bevacizumab-induced toxicities. No validated biomarkers are available for identifying patients at risk of these toxicities. METHODS: A genome-wide association study (GWAS) meta-analysis was performed in 1039 bevacizumab-treated patients of European ancestry in four clinical trials (CALGB 40502, 40503, 80303, 90401). Grade ≥2 hypertension and proteinuria were recorded (CTCAE v.3.0). Single-nucleotide polymorphism (SNP)-toxicity associations were determined using a cause-specific Cox model adjusting for age and sex. RESULTS: The most significant SNP associated with hypertension with concordant effect in three out of the four studies (p-value <0.05 for each study) was rs6770663 (A > G) in KCNAB1, with the G allele increasing the risk of hypertension (p-value = 4.16 × 10-6). The effect of the G allele was replicated in ECOG-ACRIN E5103 in 582 patients (p-value = 0.005). The meta-analysis of all five studies for rs6770663 led to p-value = 7.73 × 10-8, close to genome-wide significance. The most significant SNP associated with proteinuria was rs339947 (C > A, between DNAH5 and TRIO), with the A allele increasing the risk of proteinuria (p-value = 1.58 × 10-7). CONCLUSIONS: The results from the largest study of bevacizumab toxicity provide new markers of drug safety for further evaluations. SNP in KCNAB1 validated in an independent dataset provides evidence toward its clinical applicability to predict bevacizumab-induced hypertension. ClinicalTrials.gov Identifier: NCT00785291 (CALGB 40502); NCT00601900 (CALGB 40503); NCT00088894 (CALGB 80303) and NCT00110214 (CALGB 90401).


Subject(s)
Bevacizumab/adverse effects , Genome-Wide Association Study/methods , Hypertension/pathology , Kv1.3 Potassium Channel/genetics , Neoplasms/drug therapy , Polymorphism, Single Nucleotide , Proteinuria/pathology , Aged , Angiogenesis Inhibitors/adverse effects , Female , Humans , Hypertension/chemically induced , Hypertension/genetics , Male , Middle Aged , Neoplasms/pathology , Proteinuria/chemically induced , Proteinuria/genetics
3.
BMC Cancer ; 21(1): 655, 2021 Jun 02.
Article in English | MEDLINE | ID: mdl-34078304

ABSTRACT

BACKGROUND: The neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), and circulating tumor cells (CTCs) have been associated with survival in castration-resistant prostate cancer (CRPC). However, no study has examined the prognostic value of NLR and PLR in the context of CTCs. METHODS: Baseline CTCs from mCRPC patients were enumerated using the CellSearch System. Baseline NLR and PLR values were calculated using the data from routine complete blood counts. The associations of CTC, NLR, and PLR values, individually and jointly, with progression-free survival (PFS) and overall survival (OS), were evaluated using Kaplan-Meier analysis, as well as univariate and multivariate Cox models. RESULTS: CTCs were detected in 37 (58.7%) of 63 mCRPC patients, and among them, 16 (25.4%) had ≥5 CTCs. The presence of CTCs was significantly associated with a 4.02-fold increased risk for progression and a 3.72-fold increased risk of death during a median follow-up of 17.6 months. OS was shorter among patients with high levels of NLR or PLR than those with low levels (log-rank P = 0.023 and 0.077). Neither NLR nor PLR was individually associated with PFS. Among the 37 patients with detectable CTCs, those with a high NLR had significantly shorter OS (log-rank P = 0.024); however, among the 26 patients without CTCs, the OS difference between high- and low-NLR groups was not statistically significant. Compared to the patients with CTCs and low NLR, those with CTCs and high levels of NLR had a 3.79-fold risk of death (P = 0.036). This association remained significant after adjusting for covariates (P = 0.031). Combination analyses of CTC and PLR did not yield significant results. CONCLUSION: Among patients with detectable CTCs, the use of NLR could further classify patients into different risk groups, suggesting a complementary role for NLR in CTC-based prognostic stratification in mCRPC.


Subject(s)
Lymphocytes/immunology , Neoplastic Cells, Circulating/pathology , Neutrophils/immunology , Prostatic Neoplasms, Castration-Resistant/mortality , Aged , Aged, 80 and over , Blood Platelets , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Lymphocyte Count , Male , Middle Aged , Platelet Count , Prognosis , Progression-Free Survival , Prospective Studies , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/diagnosis , Prostatic Neoplasms, Castration-Resistant/pathology , Retrospective Studies , Risk Assessment/statistics & numerical data
5.
Oncologist ; 21(11): 1296-1297e, 2016 11.
Article in English | MEDLINE | ID: mdl-28178640

ABSTRACT

LESSONS LEARNED: Patients with metastatic castration-resistant prostate cancer did not tolerate the combination of alisertib with abiraterone and prednisone.There was no clear signal indicating that adding alisertib might be beneficial for those patients progressing on abiraterone. BACKGROUND: We hypothesized that Aurora A kinase (AK) contributes to castrate resistance in prostate cancer (PCa) and that inhibiting AK with alisertib can resensitize PCa cells to androgen receptor (AR) inhibitor abiraterone. METHODS: This was a phase I/II trial to determine the safety and efficacy of alisertib when given in combination with abiraterone plus prednisone (AP). Metastatic castration-resistant prostate cancer (mCRPC) patients were treated with dose escalation (alisertib at 30, 40, and 50 mg orally b.i.d., days 1-7 every 21 days) per standard 3+3 design. RESULTS: Nine of 43 planned subjects were enrolled. The maximum tolerated dose (MTD) was not reached, and the dose-limiting toxicities (DLTs) included neutropenic fever (1 of 9), neutropenia (1 of 9), fatigue with memory impairment (1 of 9), and diarrhea/mucositis (1 of 9). No prostate-specific antigen (PSA) decrease or circulating tumor cell (CTC) changes were observed during the study. Pharmacodynamically, adding alisertib did not affect total testosterone or dehydroepiandrosterone (DHEA) levels. There was some change in neuroendocrine markers after therapy. Mean duration on study was 2.5 months. The trial was terminated early. CONCLUSION: A tolerable dose of alisertib in combination with AP in mCRPC was not established in this study. There was no clear signal indicating that alisertib might be beneficial for patients with mCRPC progressing on abiraterone.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aurora Kinase A/antagonists & inhibitors , Azepines/administration & dosage , Prostatic Neoplasms, Castration-Resistant/drug therapy , Pyrimidines/administration & dosage , Aged , Aged, 80 and over , Androstenes/administration & dosage , Androstenes/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Azepines/adverse effects , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis , Prednisone/administration & dosage , Prednisone/adverse effects , Pyrimidines/adverse effects , Testosterone/blood
6.
Oncology (Williston Park) ; 30(2): 187-95, 199, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26888794

ABSTRACT

In 2008, the Prostate Cancer Clinical Trials Working Group 2 (PCWG2) developed consensus guidelines for clinical trial design and conduct that redefined trial endpoints, with a dual-objective paradigm: to (1) controlling, relieving, or eliminating disease manifestations at the start of treatment; and (2) preventing or delaying further disease manifestations. Clinical and translational research in prostate cancer has expanded our current-day understanding of the mechanisms of its pathogenesis, as well as the different clinicopathologic and molecular subtypes of the disease, and has improved the therapeutic armamentarium for the management of metastatic castration-resistant prostate cancer (CRPC). These new advances led to the development of the updated PCWG3 guidelines in 2015. In this review, we analyze our evolving understanding of the biology of CRPC, acquired resistance mechanisms, and emerging therapeutic targets in light of the updated PCWG3 guidelines. We present a joint perspective from the medical oncology and urologic disciplines on the ongoing efforts to advance clinical trial performance in order to discover new therapies for this fatal disease.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Immunotherapy/methods , Prostatic Neoplasms, Castration-Resistant/therapy , Radiopharmaceuticals/therapeutic use , Antineoplastic Agents, Hormonal/adverse effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Consensus , Drug Resistance, Neoplasm , Humans , Male , Molecular Targeted Therapy , Patient Selection , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Radiopharmaceuticals/adverse effects , Risk Factors , Signal Transduction/drug effects , Treatment Outcome
7.
JCO Precis Oncol ; 8: e2300552, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38452310

ABSTRACT

PURPOSE: Germline genetic testing (GT) is important for prostate cancer (PCA) management, clinical trial eligibility, and hereditary cancer risk. However, GT is underutilized and there is a shortage of genetic counselors. To address these gaps, a patient-driven, pretest genetic education webtool was designed and studied compared with traditional genetic counseling (GC) to inform strategies for expanding access to genetic services. METHODS: Technology-enhanced acceleration of germline evaluation for therapy (TARGET) was a multicenter, noninferiority, randomized trial (ClinicalTrials.gov identifier: NCT04447703) comparing a nine-module patient-driven genetic education webtool versus pretest GC. Participants completed surveys measuring decisional conflict, satisfaction, and attitudes toward GT at baseline, after pretest education/counseling, and after GT result disclosure. The primary end point was noninferiority in reducing decisional conflict between webtool and GC using the validated Decisional Conflict Scale. Mixed-effects regression modeling was used to compare decisional conflict between groups. Participants opting for GT received a 51-gene panel, with results delivered to participants and their providers. RESULTS: The analytic data set includes primary outcome data from 315 participants (GC [n = 162] and webtool [n = 153]). Mean difference in decisional conflict score changes between groups was -0.04 (one-sided 95% CI, -∞ to 2.54; P = .01), suggesting the patient-driven webtool was noninferior to GC. Overall, 145 (89.5%) GC and 120 (78.4%) in the webtool arm underwent GT, with pathogenic variants in 15.8% (8.7% in PCA genes). Satisfaction did not differ significantly between arms; knowledge of cancer genetics was higher but attitudes toward GT were less favorable in the webtool arm. CONCLUSION: The results of the TARGET study support the use of patient-driven digital webtools for expanding access to pretest genetic education for PCA GT. Further studies to optimize patient experience and evaluate them in diverse patient populations are warranted.


Subject(s)
Genetic Counseling , Prostatic Neoplasms , Humans , Male , Genetic Counseling/methods , Genetic Testing , Germ Cells , Health Knowledge, Attitudes, Practice , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy
8.
Cancer ; 119(20): 3636-43, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23913744

ABSTRACT

BACKGROUND: Preliminary data suggest a potential decreased benefit of docetaxel in patients with metastatic, castration-resistant prostate cancer (mCRPC) who previously received abiraterone acetate, a novel androgen synthesis inhibitor (ASI). Cancer and Leukemia Group B (CALGB) trial 90401 (Alliance), a phase 3 trial in patients with mCRPC who received docetaxel-based chemotherapy, offered the opportunity to evaluate effect of prior ketoconazole, an earlier generation ASI, on clinical outcomes after docetaxel. METHODS: In CALGB trial 90401, 1050 men with chemotherapy-naive mCRPC were randomized to receive treatment with docetaxel and prednisone that included either bevacizumab or placebo. In total, 1005 men (96%) had data available regarding prior ketoconazole therapy. The observed effects of prior ketoconazole on overall survival (OS), progression-free survival (PFS), prostate-specific antigen (PSA) decline, and the objective response rate (ORR) were assessed using proportional hazards and Poisson regression methods adjusted for validated prognostic factors and treatment arm. RESULTS: Baseline characteristics between patients who did (N=277) and did not (N=728) receive prior ketoconazole therapy were similar. There were no statistically significant differences between patients who did and those who did not receive prior ketoconazole therapy with respect to OS (median OS, 21.1 months vs 22.3 months, respectively; stratified log-rank P=.635), PFS (median PFS, 8.1 months vs 8.6 months, respectively; stratified log-rank P=.342), the proportion achieving a decline ≥ 50% in PSA (61% vs 66%, respectively; relative risk, 1.09; adjusted P=.129), or ORR (39% vs 43%, respectively; relative risk, 1.11; adjusted P=.366). CONCLUSIONS: As measured by OS, PFS, PSA, and the ORR, there was no evidence that prior treatment with ketoconazole had an impact on the clinical outcomes of patients with mCRPC who received subsequent docetaxel-based therapy. The current results highlight the need for prospective studies to assess for potential cross-resistance with novel ASIs and to define the optimal sequence of therapy in mCRPC.


Subject(s)
Androgen Antagonists/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/drug therapy , Liver Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antifungal Agents/pharmacology , Bevacizumab , Bone Neoplasms/mortality , Bone Neoplasms/secondary , Chemotherapy, Adjuvant , Disease Progression , Docetaxel , Drug Therapy, Combination , Follow-Up Studies , Humans , International Agencies , Ketoconazole/pharmacology , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Orchiectomy , Prednisone/pharmacology , Prognosis , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Retrospective Studies , Survival Rate , Taxoids/administration & dosage
9.
BMC Urol ; 13: 37, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23890537

ABSTRACT

BACKGROUND: Imprinting is an important epigenetic regulator of gene expression that is often disrupted in cancer. While loss of imprinting (LOI) has been reported for two genes in prostate cancer (IGF2 and TFPI2), disease-related changes in methylation across all imprinted gene regions has not been investigated. METHODS: Using an Illumina Infinium Methylation Assay, we analyzed methylation of 396 CpG sites in the promoter regions of 56 genes in a pooled sample of 12 pairs of prostate tumor and adjacent normal tissue. Selected LOI identified from the array was validated using the Sequenom EpiTYPER assay for individual samples and further confirmed by expression data from publicly available datasets. RESULTS: Methylation significantly increased in 52 sites and significantly decreased in 17 sites across 28 unique genes (P < 0.05), and the strongest evidence for loss of imprinting was demonstrated in tumor suppressor genes DLK1, PLAGL1, SLC22A18, TP73, and WT1. Differential expression of these five genes in prostate tumor versus normal tissue using array data from a publicly available database were consistent with the observed LOI patterns, and WT1 hypermethylation was confirmed using quantitative DNA methylation analysis. CONCLUSIONS: Together, these findings suggest a more widespread dysregulation of genetic imprinting in prostate cancer than previously reported and warrant further investigation.


Subject(s)
DNA Methylation/genetics , Gene Expression Regulation, Neoplastic/genetics , Genomic Imprinting/genetics , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis/methods , Prostatic Neoplasms/genetics , Base Sequence , Humans , Male , Molecular Sequence Data , Promoter Regions, Genetic/genetics
10.
Ther Adv Urol ; 15: 17562872231182219, 2023.
Article in English | MEDLINE | ID: mdl-37359737

ABSTRACT

Prostate cancer is the most common cancer among men and the second leading cause of cancer-related deaths in men in the United States. The treatment paradigm for prostate cancer has evolved with the emergence of a variety of novel therapies which have improved survival; however, treatment-related toxicities are abundant and durable responses remain rare. Immune checkpoint inhibitors have shown modest activity in a small subset of patients with prostate cancer and have not had an impact on most men with advanced disease. The discovery of prostate-specific membrane antigen (PSMA) and the understanding of its specificity to prostate cancer has identified it as an ideal tumor-associated antigen and has revived the enthusiasm for immunotherapeutics in prostate cancer. T-cell immunotherapy in the form of bispecific T-cell engagers (BiTEs) and chimeric antigen receptor (CAR) T-cell therapy have shown exceptional success in treating various hematologic malignancies, and are now being tested in patients with prostate cancer with drug design centered on various target ligands including not just PSMA, but others as well including six-transmembrane epithelial antigen of the prostate 1 (STEAP1) and prostate stem cell antigen (PSCA). This summative review will focus on the data surrounding PSMA-targeting T-cell therapies. Early clinical studies with both classes of T-cell redirecting therapies have demonstrated antitumor activity; however, there are multiple challenges with this class of agents, including dose-limiting toxicity, 'on-target, off-tumor' immune-related toxicity, and difficulty in maintaining sustained immune responses within a complex and overtly immunosuppressive tumor microenvironment. Reflecting on experiences from recent trials has been key toward understanding mechanisms of immune escape and limitations in developing these drugs in prostate cancer. Newer generation BiTE and CAR T-cell constructs, either alone or as part of combination therapy, are currently under investigation with modifications in drug design to overcome these barriers. Ongoing innovation in drug development will likely foster successful implementation of T-cell immunotherapy bringing transformational change to the treatment of prostate cancer.


New therapies utilizing T-cell immunotherapy for patients with metastatic prostate cancer There are ongoing developments in therapeutic strategies for the treatment of patients with metastatic castrate-resistant prostate cancer. Many of these developments involve the activation of the immune system to target neoplastic prostate cells and tumors. Conventional immunotherapy modalities such as checkpoint inhibitors did not provide robust response in clinical study to warrant a change to the prostate cancer treatment paradigm. However, we are now seeing various agents in the form of bispecific antibodies and chimeric antigen receptor's which influence T-cell activity and are leading to interesting and promising pre-clinical and clinical results. This review article highlights the biologic rationale for employment of T-cell redirecting therapies for the treatment of prostate cancer, and reviews much of the exciting data emerging within the field.

11.
Cancer Discov ; 13(12): 2584-2609, 2023 12 12.
Article in English | MEDLINE | ID: mdl-37676710

ABSTRACT

Signaling rewiring allows tumors to survive therapy. Here we show that the decrease of the master regulator microphthalmia transcription factor (MITF) in lethal prostate cancer unleashes eukaryotic initiation factor 3B (eIF3B)-dependent translation reprogramming of key mRNAs conferring resistance to androgen deprivation therapy (ADT) and promoting immune evasion. Mechanistically, MITF represses through direct promoter binding eIF3B, which in turn regulates the translation of specific mRNAs. Genome-wide eIF3B enhanced cross-linking immunoprecipitation sequencing (eCLIP-seq) showed specialized binding to a UC-rich motif present in subsets of 5' untranslated regions. Indeed, translation of the androgen receptor and major histocompatibility complex I (MHC-I) through this motif is sensitive to eIF3B amount. Notably, pharmacologic targeting of eIF3B-dependent translation in preclinical models sensitizes prostate cancer to ADT and anti-PD-1 therapy. These findings uncover a hidden connection between transcriptional and translational rewiring promoting therapy-refractory lethal prostate cancer and provide a druggable mechanism that may transcend into effective combined therapeutic strategies. SIGNIFICANCE: Our study shows that specialized eIF3B-dependent translation of specific mRNAs released upon downregulation of the master transcription factor MITF confers castration resistance and immune evasion in lethal prostate cancer. Pharmacologic targeting of this mechanism delays castration resistance and increases immune-checkpoint efficacy. This article is featured in Selected Articles from This Issue, p. 2489.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Male , Humans , Transcription Factors , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Immune Evasion , Receptors, Androgen/genetics , Castration , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology
12.
J Clin Oncol ; 41(15): 2736-2746, 2023 05 20.
Article in English | MEDLINE | ID: mdl-37040594

ABSTRACT

PURPOSE: We have previously developed and externally validated a prognostic model of overall survival (OS) in men with metastatic, castration-resistant prostate cancer (mCRPC) treated with docetaxel. We sought to externally validate this model in a broader group of men with docetaxel-naïve mCRPC and in specific subgroups (White, Black, Asian patients, different age groups, and specific treatments) and to classify patients into validated two and three prognostic risk groupings on the basis of the model. METHODS: Data from 8,083 docetaxel-naïve mCRPC men randomly assigned on seven phase III trials were used to validate the prognostic model of OS. We assessed the predictive performance of the model by computing the time-dependent area under the receiver operating characteristic curve (tAUC) and validated the two-risk (low and high) and three-risk prognostic groups (low, intermediate, and high). RESULTS: The tAUC was 0.74 (95% CI, 0.73 to 0.75), and when adjusting for the first-line androgen receptor (AR) inhibitor trial status, the tAUC was 0.75 (95% CI, 0.74 to 0.76). Similar results were observed by the different racial, age, and treatment subgroups. In patients enrolled on first-line AR inhibitor trials, the median OS (months) in the low-, intermediate-, and high-prognostic risk groups were 43.3 (95% CI, 40.7 to 45.8), 27.7 (95% CI, 25.8 to 31.3), and 15.4 (95% CI, 14.0 to 17.9), respectively. Compared with the low-risk prognostic group, the hazard ratios for the high- and intermediate-risk groups were 4.3 (95% CI, 3.6 to 5.1; P < .0001) and 1.9 (95% CI, 1.7 to 2.1; P < .0001). CONCLUSION: This prognostic model for OS in docetaxel-naïve men with mCRPC has been validated using data from seven trials and yields similar results overall and across race, age, and different treatment classes. The prognostic risk groups are robust and can be used to identify groups of patients for enrichment designs and for stratification in randomized clinical trials.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Male , Humans , Prognosis , Docetaxel/therapeutic use , Prostatic Neoplasms, Castration-Resistant/pathology , Proportional Hazards Models , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Treatment Outcome
13.
Eur Urol Focus ; 8(3): 670-673, 2022 05.
Article in English | MEDLINE | ID: mdl-35803854

ABSTRACT

In the expanding precision medicine landscape, along with improvements in and the availability of testing, the use of genetics in the evaluation and treatment of patients has increased significantly. Multiple urologic cancers in different organ systems associated with an inherited gene mutation have been described. As these mutations can impact screening and treatment decisions for patients and their families, it is important for providers to be familiar with the current guidelines for germline testing. Here we summarize the current guidelines regarding germline testing for patients with suspected urologic tumor syndromes. PATIENT SUMMARY: Several cancers of the genitourinary tract can be associated with inherited genetic mutations. Knowledge of when to test for these mutations has implications for both treatment and screening of patients and their family members at risk of genitourinary cancers.


Subject(s)
Genetic Testing , Urologic Neoplasms , Germ Cells , Germ-Line Mutation/genetics , Humans , Syndrome , Urologic Neoplasms/diagnosis , Urologic Neoplasms/genetics
14.
PLoS One ; 17(8): e0273826, 2022.
Article in English | MEDLINE | ID: mdl-36037225

ABSTRACT

INTRODUCTION: Abiraterone acetate and enzalutamide are commonly employed in prostate cancer therapy in an interchangeable manner. These drugs are highly efficacious in androgen antagonism to improve patient outcomes, but they also carry noteworthy risk of adverse effects. Common toxicities vary amongst the two drugs and may have differential interactions with patient co-morbidities, but these patterns are unclear as co-morbidities typically serve as exclusion criteria in clinical trials. Hence, there is no existing guidance on how clinicians may tailor treatment based on patient-specific factors. Analysis of differential patient outcomes between these two drugs can inform future systematic reviews, new clinical studies, and clinical decision making. METHOD AND ANALYSIS: The framework for this methodology was informed by the Joanna Briggs Institute methodology for scoping reviews. Title and abstract screening will be performed by two independent researchers to create an initial study inventory. This will be followed by full-text screening for study inclusion. Population-based studies describing patient outcomes, common toxicities, and associations with patient co-morbidities following abiraterone or enzalutamide therapy will be included. After data is extracted, it will be summarized for presentation. ETHICS AND DISSEMINATION: The findings of this scoping review will be published in a peer-reviewed journal. The results will be used to inform future studies on patient-specific factors informing treatment choice between abiraterone and enzalutamide for castration-resistant prostate cancer. All data are from published openly accessible sources, and therefore, no ethical clearance is necessary. The protocol is also registered at https://doi.org/10.6084/m9.figshare.19149227.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Abiraterone Acetate , Androstenes/adverse effects , Benzamides , Humans , Male , Nitriles/therapeutic use , Phenylthiohydantoin/adverse effects , Prostatic Neoplasms, Castration-Resistant/drug therapy , Review Literature as Topic , Treatment Outcome
15.
Cancers (Basel) ; 14(15)2022 Aug 03.
Article in English | MEDLINE | ID: mdl-35954437

ABSTRACT

Abiraterone acetate (AA) and enzalutamide (ENZ) are commonly used for metastatic prostate cancer. It is unclear how their outcomes and toxicities vary with patient-specific factors because clinical trials typically exclude patients with significant comorbidities. This study aims to fill this knowledge gap and facilitate informed treatment decision making. A registered protocol utilizing PRISMA scoping review methodology was utilized to identify real-world studies. Of 433 non-duplicated publications, 23 were selected by three independent reviewers. ENZ offered a faster and more frequent biochemical response (30-50% vs. 70-75%), slowed progression (HR 0.66; 95% CI 0.50-0.88), and improved overall survival versus AA. ENZ was associated with more fatigue and neurological adverse effects. Conversely, AA increased risk of cardiovascular- (HR 1.82; 95% CI 1.09-3.05) and heart failure-related (HR 2.88; 95% CI 1.09-7.63) hospitalizations. Ultimately, AA was associated with increased length of hospital stay, emergency department visits, and hospitalizations (HR 1.26; 95% CI 1.04-1.53). Accordingly, total costs were higher for AA, although pharmacy costs alone were higher for ENZ. Existing data suggest that AA and ENZ have important differences in outcomes including toxicities, response, disease progression, and survival. Additionally, adherence, healthcare utilization, and costs differ. Further investigation is warranted to inform treatment decisions which optimize patient outcomes.

16.
Can J Urol ; 18(4): 5767-77, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21854708

ABSTRACT

INTRODUCTION: Castrate-resistant prostate cancer (CRPC) is the most clinically advanced form of prostate cancer. Prior to 2004, treatment options for patients with CRPC were limited to palliative care with mitoxantrone. However, two phase III trials in 2004 showed improved survival with docetaxel compared with mitoxantrone in patients with metastatic CRPC. Docetaxel remains the current standard chemotherapy for CRPC. MATERIALS AND METHODS: A literature review was conducted to ascertain agents recently approved or in development for CRPC as well as several treatment algorithms being developed in this patient population. RESULTS: Recently, the US Food and Drug Administration (FDA) approved chemotherapy agents including cabazitaxel, a novel taxane, for the treatment of patients with metastatic CRPC who were previously treated with docetaxel-based chemotherapy. The immunotherapy sipuleucel-T and the androgen biosynthesis inhibitor, abiraterone, have also just been approved. A number of other novel agents are in clinical development for the management of CRPC. CONCLUSIONS: Options for the management of CRPC are rapidly expanding. Based on improved survival with docetaxel in patients with CRPC, chemotherapy is being investigated for use earlier in the prostate cancer disease spectrum. Studies are actively evaluating chemotherapy in the neoadjuvant and adjuvant settings for patients with high risk, localized prostate cancer. A multidisciplinary approach towards patients with difficult-to-manage prostate cancer in which urologists familiarize themselves with these newer systemic agents and refer appropriate patients to oncologists may be beneficial.


Subject(s)
Antineoplastic Agents/therapeutic use , Orchiectomy , Prostatic Neoplasms/drug therapy , Androstenes , Androstenols/therapeutic use , Chemotherapy, Adjuvant , Docetaxel , Humans , Male , Taxoids/therapeutic use , Tissue Extracts/therapeutic use
17.
Urology ; 155: 199-209, 2021 09.
Article in English | MEDLINE | ID: mdl-33373704

ABSTRACT

Androgen deprivation therapy, alone or in combination with androgen signaling inhibitors, is a treatment option for patients with advanced prostate cancer (PC). When making treatment decisions, health care providers must consider the long-term effects of treatment on the patient's overall health and well-being. Herein, we review the effects of these treatments on the musculoskeletal and cardiovascular systems, cognition, and fall risk, and provide management approaches for each. We also include an algorithm to help health care providers implement best clinical practices and interdisciplinary care for preserving the overall well-being of PC patients.


Subject(s)
Androgen Antagonists/adverse effects , Antineoplastic Agents/adverse effects , Prostatic Neoplasms/drug therapy , Androgen Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Humans , Male , Neoplasm Staging , Practice Guidelines as Topic , Prostatic Neoplasms/pathology
18.
Eur Urol Oncol ; 4(2): 297-300, 2021 04.
Article in English | MEDLINE | ID: mdl-32061548

ABSTRACT

Although predominantly urothelial, some bladder cancer and upper tract urothelial cancer (BC/UTUC) harbor histologic variants. Small cell BC (SCBC) variants comprised ˜5% of The Cancer Genome Atlas BC cohort, with a poor prognosis. We describe genomic profiles of BC/UTUC with small cell/neuroendocrine features identified in the Foundation Medicine database from June 2012 to September 2018. Of 3368 BC/UTUC samples, 3.92% (132) harbored small cell/neuroendocrine features by immunohistochemistry. Mutations were noted in: TP53 (92%), RB1 (75%), combined TP53/RB1 (72%), and TERT promoter (68%). Of the samples, 6.5% had TMB ≥ 10 mutations/Mb. RNA expression profiling of 24 pure SCBC and 51 urothelial BC (UBC) muscle-invasive samples evaluated from a separate cohort revealed a large number of differentially expressed genes with suppression of several inflammatory pathways in SCBC compared with UBC. This largest reported SCBC dataset to date confirms enrichment of signatures in SCBC similar to small cell lung cancer and describes unique gene expression compared with UBC. These findings may explain aggressive SCBC phenotype. PATIENT SUMMARY: Small cell bladder cancer (SCBC) is an aggressive subtype that microscopically resembles aggressive small cell lung cancer (SCLC). This study confirms that SCBC shares DNA changes similar to SCLC and that SCBC expresses many genes that urothelial bladder cancer does not, possibly explaining aggressive SCBC activity.


Subject(s)
Urinary Bladder Neoplasms , Biomarkers, Tumor , Genomics , Humans , Mutation , Transcriptome , Tumor Suppressor Protein p53/genetics , Urinary Bladder Neoplasms/genetics
19.
Eur Urol ; 77(2): 158-166, 2020 02.
Article in English | MEDLINE | ID: mdl-31420248

ABSTRACT

BACKGROUND: Elderly patients (≥65yr) with advanced prostate cancer and cardiovascular disease (CVD) conditions are often excluded from clinical trials of abiraterone acetate (AA) or enzalutamide (ENZ). Consequently, little is known about the effects of these medications on these vulnerable patients. OBJECTIVE: To assess the short-term outcomes of AA and ENZ in patients with pre-existing CVDs. DESIGN, SETTING, AND PARTICIPANTS: A population-based retrospective study. The Surveillance, Epidemiology, and End Results-Medicare-linked database was used to identify prostate cancer patients using AA or ENZ. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: The primary endpoint was 6-mo all-cause mortality, analyzed using modified Poisson regression modeling of relative risk (RR) adjusted for confounders and comorbidities. RESULTS AND LIMITATIONS: Among eligible patients (2845 with AA and 1031 with ENZ), 67% had at least one pre-existing CVD. Compared with those without pre-existing CVDs, having one to two pre-existing CVDs was associated with 16% higher 6-mo mortality (RR=1.16, 95% confidence interval [CI]: 1.00-1.36), and the risk increased further among those having three or more CVDs (RR=1.56, 95% CI: 1.29-1.88). Most of the differences in survival of patients with pre-existing CVD condition occurred within the first 6mo of treatment. CONCLUSIONS: After treatment with AA or ENZ, elderly prostate cancer patients with pre-existing CVDs experienced higher short-term mortality than otherwise similar patients without CVDs. Mortality associated with CVDs did not depend on having received AA versus ENZ. PATIENT SUMMARY: Patients with pre-existing cardiovascular diseases (CVDs) experienced higher short-term mortality after abiraterone acetate or enzalutamide than those without pre-existing CVDs. It is recommended that a multidisciplinary team, including a cardiologist, evaluate patients having pre-existing CVDs in the process of making treatment decisions and monitoring potential side effects.


Subject(s)
Abiraterone Acetate/administration & dosage , Androgen Antagonists/administration & dosage , Antineoplastic Agents/administration & dosage , Hospitalization/statistics & numerical data , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/mortality , Administration, Oral , Aged , Aged, 80 and over , Benzamides , Cardiovascular Diseases/complications , Humans , Male , Neoplasm Staging , Nitriles , Phenylthiohydantoin/administration & dosage , Prostatic Neoplasms/complications , Prostatic Neoplasms/pathology , Retrospective Studies , Risk Assessment , Time Factors , Treatment Outcome
20.
JNCI Cancer Spectr ; 4(2): pkaa003, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32368717

ABSTRACT

There are few data regarding disparities in overall survival (OS) between Asian and white men with metastatic castration-resistant prostate cancer (mCRPC). We compared OS of Asian and white mCRPC men treated in phase III clinical trials with docetaxel and prednisone (DP) or a DP-containing regimen. Individual participant data from 8820 men with mCRPC randomly assigned on nine phase III trials to receive DP or a DP-containing regimen were combined. Men enrolled in these trials had a diagnosis of prostate adenocarcinoma. The median overall survival was 18.8 months (95% confidence interval [CI] = 17.4 to 22.1 months) and 21.2 months (95% CI = 20.8 to 21.7 months) for Asian and white men, respectively. The pooled hazard ratio for death for Asian men compared with white men, adjusted for baseline prognostic factors, was 0.95 (95% CI = 0.84 to 1.09), indicating that Asian men were not at increased risk of death. This large analysis showed that Asian men did not have shorter OS duration than white men treated with docetaxel.

SELECTION OF CITATIONS
SEARCH DETAIL