Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Curr Opin Oncol ; 36(3): 164-168, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38573205

RESUMO

PURPOSE OF REVIEW: Patients with biochemically recurrent prostate cancer (BCR) after unsuccessful curative therapies frequently have an indolent and asymptomatic disease course for years. There are no prospective data showing that treating BCR improves overall survival despite new imaging strategies and emerging therapeutic data. Managing BCR requires a unique perspective in oncology that balances toxicities and disease kinetics. RECENT FINDINGS: Prostate specific membrane antigen (PSMA) imaging is now widely available and can define subclinical disease in patients with BCR who otherwise have negative CT and bone scans, but limited data exists showing that treating PSMA-positive disease has long term impact. A phase 3 trial demonstrated that the androgen receptor pathway inhibitor enzalutamide either alone or with androgen deprivation therapy (ADT) was superior in delaying metastasis, relative to ADT alone. Survival benefits from this study remain unknown. SUMMARY: BCR is a heterogeneous population where overtreatment may present greater risk to patients than a disease course that is often indolent. Management of BCR should be individualized based on disease kinetics. Given the unique biology of BCR, future therapeutic research should emphasize an approach that alters disease trajectory without accompanying side effects and should explore options beyond ADT-based strategies.


Assuntos
Antagonistas de Androgênios , Neoplasias da Próstata , Masculino , Humanos , Antagonistas de Androgênios/uso terapêutico , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/tratamento farmacológico , Progressão da Doença , Antagonistas de Receptores de Andrógenos , Tomografia por Emissão de Pósitrons
2.
Cancer ; 130(9): 1629-1641, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38161319

RESUMO

BACKGROUND: Patients with localized, unfavorable intermediate-risk and high-risk prostate cancer have an increased risk of relapse after radical prostatectomy (RP). The authors previously reported on part 1 of this phase 2 trial testing neoadjuvant apalutamide, abiraterone, prednisone, plus leuprolide (AAPL) or abiraterone, prednisone, and leuprolide (APL) for 6 months followed by RP. The results demonstrated favorable pathologic responses (tumor <5 mm) in 20.3% of patients (n = 24 of 118). Herein, the authors report the results of part 2. METHODS: For part 2, patients were randomized 1:1 to receive either AAPL for 12 months (arm 2A) or observation (arm 2B), stratified by neoadjuvant therapy and pathologic tumor classification. The primary end point was 3-year biochemical progression-free survival. Secondary end points included safety and testosterone recovery (>200 ng/dL). RESULTS: Overall, 82 of 118 patients (69%) enrolled in part 1 were randomized to part 2. A higher proportion of patients who were not randomized to adjuvant therapy had a favorable prostatectomy pathologic response (32.3% in nonrandomized patients compared with 17.1% in randomized patients). In the intent-to-treat analysis, the 3-year biochemical progression-free survival rate was 81% for arm 2A and 72% for arm 2B (hazard ratio, 0.81; 90% confidence interval, 0.43-1.49). Of the randomized patients, 81% had testosterone recovery in the AAPL group compared with 95% in the observation group, with a median time to recovery of <12 months in both arms. CONCLUSIONS: In this study, because 30% of patients declined adjuvant treatment, part B was underpowered to detect differences between arms. Future perioperative studies should be biomarker-directed and include strategies for investigator and patient engagement to ensure compliance with protocol procedures.


Assuntos
Antígeno Prostático Específico , Neoplasias da Próstata , Masculino , Humanos , Leuprolida/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/cirurgia , Neoplasias da Próstata/diagnóstico , Antagonistas de Androgênios/efeitos adversos , Androgênios , Prednisona , Resultado do Tratamento , Recidiva Local de Neoplasia/cirurgia , Prostatectomia/métodos , Testosterona
3.
bioRxiv ; 2023 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-37577497

RESUMO

Xp11 translocation renal cell carcinoma (tRCC) is a female-predominant kidney cancer driven by translocations between the TFE3 gene on chromosome Xp11.2 and partner genes located on either chrX or on autosomes. The rearrangement processes that underlie TFE3 fusions, and whether they are linked to the female sex bias of this cancer, are largely unexplored. Moreover, whether oncogenic TFE3 fusions arise from both the active and inactive X chromosomes in females remains unknown. Here we address these questions by haplotype-specific analyses of whole-genome sequences of 29 tRCC samples from 15 patients and by re-analysis of 145 published tRCC whole-exome sequences. We show that TFE3 fusions universally arise as reciprocal translocations with minimal DNA loss or insertion at paired break ends. Strikingly, we observe a near exact 2:1 female:male ratio in TFE3 fusions arising via X:autosomal translocation (but not via X inversion), which accounts for the female predominance of tRCC. This 2:1 ratio is at least partially attributable to oncogenic fusions involving the inactive X chromosome and is accompanied by partial re-activation of silenced chrX genes on the rearranged chromosome. Our results highlight how somatic alterations involving the X chromosome place unique constraints on tumor initiation and exemplify how genetic rearrangements of the sex chromosomes can underlie cancer sex differences.

4.
Urology ; 173: 147-148, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36958911
5.
Urol Oncol ; 41(4): 204.e1-204.e6, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36754665

RESUMO

PURPOSE: Treatment toxicity from surgery radical prostatectomy (RP) or radiation therapy (RT) has been well studied in patients with localized prostate cancer. However, little is known about lingering toxicities in patients who develop metastatic recurrence. We aimed to compare the prevalence of local treatment-related side effects in patients with metastatic recurrence and those in remission, and to explore to what extent medical oncologists address this morbidity. METHODS: This was a single site, cross-sectional study evaluating patient-reported outcomes using the Expanded Prostate Cancer Index Clinical Practice (EPIC-CP) instrument, which measures Health-Related Quality of Life (HRQoL) across urinary, bowel, sexual, and hormonal domains, with higher scores reflecting increased symptom burden. The primary endpoint was differences in overall and domain-specific EPIC-CP scores between the metastatic and localized cohorts, with secondary endpoints evaluating provider interventions for symptom alleviation. RESULTS: Median total EPIC-CP scores were higher in the metastatic cohort (18.0, IQR 13.0-24.0) compared to the localized cohort (10.0, 6.0-15.0) (P < 0.001). This difference was mostly driven by worsening symptoms in the sexual (8.0, 8.0-9.0 vs. 6.0, 3.0-8.0) (P < 0.001) and hormonal domains (2.0, 1.0-6.0 vs. 0.0, 0.0-2.0) (P < 0.001), although there were also differences in the urinary irritation/obstruction (3.0, 0.0-3.0 vs. 1.0, 0.0-2.0) (P < 0.001) and bowel domains (1.0, 0.0-3.0 vs. 0.0, 0.0-0.0) (P < 0.001). There was a trend towards higher scores in patients that had received RT as primary treatment. Medical oncologists rarely changed management in response to local symptoms. CONCLUSION: Prostate cancer patients with metastatic recurrence suffer from a higher burden of localized treatment-related symptoms compared with patients in remission, with primary RT associated with more prevalent toxicity than radical prostatectomy. There is an unmet need for more intensive management of local symptoms. Further studies should focus on factors that portend long term worse morbidity.


Assuntos
Neoplasias da Próstata , Qualidade de Vida , Masculino , Humanos , Prevalência , Estudos Transversais , Neoplasias da Próstata/radioterapia , Neoplasias da Próstata/cirurgia , Prostatectomia/efeitos adversos , Morbidade
6.
Cancer Res ; 83(2): 219-238, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36413141

RESUMO

Abiraterone is a standard treatment for metastatic castrate-resistant prostate cancer (mCRPC) that slows disease progression by abrogating androgen synthesis and antagonizing the androgen receptor (AR). Here we report that inhibitors of the mitotic regulator polo-like kinase-1 (Plk1), including the clinically active third-generation Plk1 inhibitor onvansertib, synergizes with abiraterone in vitro and in vivo to kill a subset of cancer cells from a wide variety of tumor types in an androgen-independent manner. Gene-expression analysis identified an AR-independent synergy-specific gene set signature upregulated upon abiraterone treatment that is dominated by pathways related to mitosis and the mitotic spindle. Abiraterone treatment alone caused defects in mitotic spindle orientation, failure of complete chromosome condensation, and improper cell division independently of its effects on AR signaling. These effects, although mild following abiraterone monotherapy, resulted in profound sensitization to the antimitotic effects of Plk1 inhibition, leading to spindle assembly checkpoint-dependent mitotic cancer cell death and entosis. In a murine patient-derived xenograft model of abiraterone-resistant metastatic castration-resistant prostate cancer (mCRPC), combined onvansertib and abiraterone resulted in enhanced mitotic arrest and dramatic inhibition of tumor cell growth compared with either agent alone. Overall, this work establishes a mechanistic basis for the phase II clinical trial (NCT03414034) testing combined onvansertib and abiraterone in mCRPC patients and indicates this combination may have broad utility for cancer treatment. SIGNIFICANCE: Abiraterone treatment induces mitotic defects that sensitize cancer cells to Plk1 inhibition, revealing an AR-independent mechanism for this synergistic combination that is applicable to a variety of cancer types.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Receptores Androgênicos , Masculino , Humanos , Animais , Camundongos , Receptores Androgênicos/metabolismo , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/metabolismo , Androgênios , Mitose
7.
Expert Rev Anticancer Ther ; 22(7): 695-702, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35698870

RESUMO

INTRODUCTION: Tivozanib is a selective vascular endothelial growth factor receptor (VEGFR)-inhibitor designed to, more specifically, bind to the VEGF receptor with fewer off-target interactions with other tyrosine kinase receptors in the treatment of advanced renal cell carcinoma (RCC). AREAS COVERED: Both preclinical and early clinical studies have suggested tivozanib could be a more potent VEGFR inhibitor with less off-target toxicities for patients. After a complicated clinical development process, the drug was approved by the FDA for third- and fourth-line use in relapsed, refractory renal cell carcinoma (RCC) in March of 2021 based on the results of the TIVO-3 trial. However, questions remain regarding the proper incorporation of tivozanib in the current treatment landscape of RCC. EXPERT OPINION: Here, we review the existing literature surrounding tivozanib and comment on its optimal use in current and future clinical practice. We suggest that tivozanib may be considered in relapsed, refractory RCC in the later-line treatment setting following progression on both immune checkpoint inhibitors (ICIs) and nonselective VEGFR-TKIs. We anticipate the application of tivozanib in RCC will continue to evolve as trials exploring tivozanib in combination with ICIs may move this drug earlier in the future treatment landscape of RCC.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Recidiva Local de Neoplasia , Compostos de Fenilureia , Quinolinas , Inibidores da Angiogênese/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/patologia , Aprovação de Drogas , Humanos , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/patologia , Recidiva Local de Neoplasia/tratamento farmacológico , Compostos de Fenilureia/uso terapêutico , Quinolinas/uso terapêutico , Receptores de Fatores de Crescimento do Endotélio Vascular , Estados Unidos , Fator A de Crescimento do Endotélio Vascular
8.
Prostate ; 82(8): 883-893, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35254710

RESUMO

BACKGROUND: Men of African ancestry (AA) with prostate cancer suffer from worse outcomes. However, a recent analysis of patients treated with the dendritic cell vaccine sipuleucel-T for prostate cancer suggested that AA patients could have improved outcomes relative to whites. METHODS: We conducted a focused literature review of Medline-indexed articles and clinical trials listed on clinicaltrials.gov. RESULTS: We identify several studies pointing to enrichment of inflammatory cellular infiltrates and cytokine signaling among AA patients with prostate cancer. We outline potential genomic and transcriptomic alterations that may contribute to immunogenicity. Last, we investigate differences in host immunity and vaccine responsiveness that may be enhanced in AA patients. CONCLUSIONS: AA patients with prostate cancer may be enriched for an immunogenic phenotype. Dedicated studies are needed to better understand the immune mechanisms that contribute to existing cancer disparities and test immune-based therapies in this population.


Assuntos
Negro ou Afro-Americano , Neoplasias da Próstata , Negro ou Afro-Americano/genética , População Negra/genética , Humanos , Masculino , Neoplasias da Próstata/terapia , Transcriptoma , População Branca
9.
Cancer Res ; 82(8): 1518-1533, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35131873

RESUMO

Wnt signaling driven by genomic alterations in genes including APC and CTNNB, which encodes ß-catenin, have been implicated in prostate cancer development and progression to metastatic castration-resistant prostate cancer (mCRPC). However, nongenomic drivers and downstream effectors of Wnt signaling in prostate cancer and the therapeutic potential of targeting this pathway in prostate cancer have not been fully established. Here we analyzed Wnt/ß-catenin signaling in prostate cancer and identified effectors distinct from those found in other tissues, including aryl hydrocarbon receptor and RUNX1, which are linked to stem cell maintenance, and ROR1, a noncanonical Wnt5a coreceptor. Wnt/ß-catenin signaling-mediated increases in ROR1 enhanced noncanonical responses to Wnt5a. Regarding upstream drivers, APC genomic loss, but not its epigenetic downregulation commonly observed in prostate cancer, was strongly associated with Wnt/ß-catenin pathway activation in clinical samples. Tumor cell upregulation of the Wnt transporter Wntless (WLS) was strongly associated with Wnt/ß-catenin pathway activity in primary prostate cancer but also associated with both canonical and noncanonical Wnt signaling in mCRPC. IHC confirmed tumor cell WLS expression in primary prostate cancer and mCRPC, and patient-derived prostate cancer xenografts expressing WLS were responsive to treatment with Wnt synthesis inhibitor ETC-1922159. These findings reveal that Wnt/ß-catenin signaling in prostate cancer drives stem cell maintenance and invasion and primes for noncanonical Wnt signaling through ROR1. They further show that autocrine Wnt production is a nongenomic driver of canonical and noncanonical Wnt signaling in prostate cancer, which can be targeted with Wnt synthesis inhibitors to suppress tumor growth. SIGNIFICANCE: This work provides fundamental insights into Wnt signaling and prostate cancer cell biology and indicates that a subset of prostate cancer driven by autocrine Wnt signaling is sensitive to Wnt synthesis inhibitors.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase , Via de Sinalização Wnt , Comunicação Autócrina , Humanos , Masculino , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
10.
Artigo em Inglês | MEDLINE | ID: mdl-34568716

RESUMO

Metastatic prostate cancer is initially sensitive to androgen receptor inhibition, but eventually becomes castration-resistant prostate cancer (mCRPC). Early use of more intensive therapies targeting androgen receptor and other oncogenic drivers in treatment-naïve primary prostate cancer (PC) may be more effective than that in advanced mCRPC. However, analysis of primary tumors may not reveal targetable metastatic drivers that are subclonal in the primary tumor or acquired at metastatic sites. METHODS: PC samples spanning one patient's clinical course: diagnostic biopsies, pre- or post-enzalutamide metastatic biopsies, and rapid autopsy samples including a patient-derived xenograft (PDX) were analyzed by targeted exome sequencing followed by phylogenetic analysis. RESULTS: Left- and right-lobe primary PC tumors appeared to diverge, with the right acquiring additional shared mutations and striking differences in copy number alterations that later appeared in metastatic samples during the treatment course and at autopsy, whereas the left base tumor maintained a quiet copy number alteration landscape and partitioned into a dead-end node. RB1 loss, a common finding in advanced castration-resistant disease, was identified throughout mCRPC samples, but not in the primary tumor. Significantly, a truncal EGFR-activating mutation (R108K) was identified in the primary tumor and was also found to be maintained in the mCRPC samples and in a PDX model. Furthermore, the PDX model remained sensitive to the EGFR inhibitor erlotinib, despite the presence of both RB1 and BRCA2 losses. CONCLUSION: These findings indicate that truncal alterations identified in primary PC can drive advanced mCRPC, even in the presence of additional strong oncogenic drivers (ie, RB1 and BRCA2 loss), and suggest that earlier detection and targeting of these truncal alterations may be effective at halting disease progression.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Biópsia , Carcinogênese , Humanos , Masculino , Filogenia , Neoplasias de Próstata Resistentes à Castração/genética
11.
Clin Cancer Res ; 27(17): 4836-4847, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34168052

RESUMO

PURPOSE: A subset of primary prostate cancer expresses programmed death-ligand 1 (PD-L1), but whether they have a unique tumor immune microenvironment or genomic features is unclear. EXPERIMENTAL DESIGN: We selected PD-L1-positive high-grade and/or high-risk primary prostate cancer, characterized tumor-infiltrating lymphocytes with multiplex immunofluorescence, and identified genomic alterations in immunogenic and nonimmunogenic tumor foci. RESULTS: One quarter of aggressive localized prostate cancer cases (29/115) had tumor PD-L1 expression more than 5%. This correlated with increased density of CD8+ T cells, a large fraction coexpressing PD-1, versus absent PD-1 expression on sparse CD8 T cells in unselected cases. Most CD8+PD-1+ cells did not express terminal exhaustion markers (TIM3 or LAG3), while a subset expressed TCF1. Consistent with these CD8+PD-1+TCF1+ cells being progenitors, they were found in antigen-presenting cell niches in close proximity to MHC-II+ cells. CD8 T-cell density in immunogenic prostate cancer and renal cell carcinoma (RCC) was nearly identical. Shallow RB1 and BRCA2 losses, and deep deletions of CHD1, were prevalent, the latter being strongly associated with a dendritic cell gene set in The Cancer Genome Atlas. Tumor mutation burden was variable; neither high microsatellite instability nor CDK12 alterations were present. CONCLUSIONS: A subset of localized prostate cancer is immunogenic, manifested by PD-L1 expression and CD8+ T-cell content comparable with RCC. The CD8+ T cells include effector cells and exhausted progenitor cells, which may be expanded by immune checkpoint inhibitors (ICI). Genomic losses of RB1, BRCA2, and CHD1 may be drivers of this phenotype. These findings indicate that immunotherapies may be effective in biomarker-selected subpopulations of patients with localized prostate cancer.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias da Próstata , Antígeno B7-H1/genética , Genes Supressores de Tumor , Humanos , Linfócitos do Interstício Tumoral , Masculino , Fenótipo , Neoplasias da Próstata/genética , Microambiente Tumoral/genética
12.
Nat Cancer ; 2(4): 444-456, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33899001

RESUMO

Prostate cancers are considered to be immunologically 'cold' tumors given the very few patients who respond to checkpoint inhibitor (CPI) therapy. Recently, enrichment of interferon-stimulated genes (ISGs) predicted a favorable response to CPI across various disease sites. The enhancer of zeste homolog-2 (EZH2) is overexpressed in prostate cancer and known to negatively regulate ISGs. In the present study, we demonstrate that EZH2 inhibition in prostate cancer models activates a double-stranded RNA-STING-ISG stress response upregulating genes involved in antigen presentation, Th1 chemokine signaling and interferon response, including programmed cell death protein 1 (PD-L1) that is dependent on STING activation. EZH2 inhibition substantially increased intratumoral trafficking of activated CD8+ T cells and increased M1 tumor-associated macrophages, overall reversing resistance to PD-1 CPI. Our study identifies EZH2 as a potent inhibitor of antitumor immunity and responsiveness to CPI. These data suggest EZH2 inhibition as a therapeutic direction to enhance prostate cancer response to PD-1 CPI.


Assuntos
Receptor de Morte Celular Programada 1 , Neoplasias da Próstata , Linfócitos T CD8-Positivos , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Humanos , Interferons/farmacologia , Masculino , Neoplasias da Próstata/tratamento farmacológico , RNA de Cadeia Dupla
13.
J Urol ; 206(1): 80-87, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33683939

RESUMO

PURPOSE: This multicenter randomized phase 2 trial investigates the impact of intense androgen deprivation on radical prostatectomy pathologic response and radiographic and tissue biomarkers in localized prostate cancer (NCT02903368). MATERIALS AND METHODS: Eligible patients had a Gleason score ≥4+3=7, prostate specific antigen >20 ng/mL or T3 disease and lymph nodes <20 mm. In Part 1, patients were randomized 1:1 to apalutamide, abiraterone acetate, prednisone and leuprolide (AAPL) or abiraterone, prednisone, leuprolide (APL) for 6 cycles (1 cycle=28 days) followed by radical prostatectomy. Surgical specimens underwent central review. The primary end point was the rate of pathologic complete response or minimum residual disease (minimum residual disease, tumor ≤5 mm). Secondary end points included prostate specific antigen response, positive margin rate and safety. Magnetic resonance imaging and tissue biomarkers of pathologic outcomes were explored. RESULTS: The study enrolled 118 patients at 4 sites. Median age was 61 years and 94% of patients had high-risk disease. The combined pathologic complete response or minimum residual disease rate was 22% in the AAPL arm and 20% in the APL arm (difference: 1.5%; 1-sided 95% CI -11%, 14%; 1-sided p=0.4). No new safety signals were observed. There was low concordance and correlation between posttherapy magnetic resonance imaging assessed and pathologically assessed tumor volume. PTEN-loss, ERG positivity and presence of intraductal carcinoma were associated with extensive residual tumor. CONCLUSIONS: Intense neoadjuvant hormone therapy in high-risk prostate cancer resulted in favorable pathologic responses (tumor <5 mm) in 21% of patients. Pathologic responses were similar between treatment arms. Part 2 of this study will investigate the impact of adjuvant hormone therapy on biochemical recurrence.


Assuntos
Acetato de Abiraterona/uso terapêutico , Antineoplásicos Hormonais/uso terapêutico , Antineoplásicos/uso terapêutico , Leuprolida/uso terapêutico , Prednisona/uso terapêutico , Prostatectomia , Neoplasias da Próstata/cirurgia , Tioidantoínas/uso terapêutico , Idoso , Terapia Combinada , Quimioterapia Combinada , Humanos , Masculino , Pessoa de Meia-Idade , Período Pré-Operatório , Neoplasias da Próstata/patologia , Medição de Risco , Resultado do Tratamento
14.
Clin Cancer Res ; 27(1): 320-329, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33037017

RESUMO

PURPOSE: The role of immune-oncologic mechanisms of racial disparities in prostate cancer remains understudied. Limited research exists to evaluate the molecular underpinnings of immune differences in African American men (AAM) and European American men (EAM) prostate tumor microenvironment (TME). EXPERIMENTAL DESIGN: A total of 1,173 radiation-naïve radical prostatectomy samples with whole transcriptome data from the Decipher GRID registry were used. Transcriptomic expressions of 1,260 immune-specific genes were selected to assess immune-oncologic differences between AAM and EAM prostate tumors. Race-specific differential expression of genes was assessed using a rank test, and intergene correlational matrix and gene set enrichment was used for pathway analysis. RESULTS: AAM prostate tumors have significant enrichment of major immune-oncologic pathways, including proinflammatory cytokines, IFNα, IFNγ, TNFα signaling, ILs, and epithelial-mesenchymal transition. AAM TME has higher total immune content score (ICSHIGH) compared with 0 (37.8% vs. 21.9%, P = 0.003). AAM tumors also have lower DNA damage repair and are genomically radiosensitive as compared with EAM. IFITM3 (IFN-inducible transmembrane protein 3) was one of the major proinflammatory genes overexpressed in AAM that predicted increased risk of biochemical recurrence selectively for AAM in both discovery [HRAAM = 2.30; 95% confidence interval (CI), 1.21-4.34; P = 0.01] and validation (HRAAM = 2.42; 95% CI, 1.52-3.86; P = 0.0001) but not in EAM. CONCLUSIONS: Prostate tumors of AAM manifest a unique immune repertoire and have significant enrichment of proinflammatory immune pathways that are associated with poorer outcomes. Observed immune-oncologic differences can aid in a genomically adaptive approach to treating prostate cancer in AAM.


Assuntos
Negro ou Afro-Americano/genética , Regulação Neoplásica da Expressão Gênica/imunologia , Recidiva Local de Neoplasia/imunologia , Neoplasias da Próstata/genética , Microambiente Tumoral/imunologia , Negro ou Afro-Americano/estatística & dados numéricos , Idoso , Conjuntos de Dados como Assunto , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/imunologia , Seguimentos , Genômica/estatística & dados numéricos , Disparidades nos Níveis de Saúde , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/prevenção & controle , Próstata/imunologia , Próstata/patologia , Prostatectomia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/terapia , Medição de Risco/estatística & dados numéricos , Microambiente Tumoral/genética , População Branca/genética , População Branca/estatística & dados numéricos
15.
Urology ; 147: 119-126, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33137348

RESUMO

Pembrolizumab was recently approved for treatment of cancers with high tumor mutational burden (TMB). We conduct a focused literature review of TMB as a predictive biomarker. TMB quantifies the sum of nonsynonymous coding mutations (typically single nucleotide substitutions and short insertion-deletions) per megabase of sequenced DNA. As a proxy for expression of immunogenic neoantigens, TMB may be an effective predictive biomarker for response to immune checkpoint inhibitors. However, like other biomarkers in this setting, TMB has many limitations; the effect of this FDA approval in the current management of genitourinary cancers is likely limited to select situations.


Assuntos
Biomarcadores Tumorais/genética , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias Urogenitais/tratamento farmacológico , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Mutação , Prognóstico , Intervalo Livre de Progressão , Neoplasias Urogenitais/genética , Neoplasias Urogenitais/imunologia , Neoplasias Urogenitais/mortalidade
16.
Urology ; 145: 118-119, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33167168
17.
Urol Clin North Am ; 47(4): 457-467, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33008496

RESUMO

Biochemically recurrent prostate cancer represents a stage of prostate cancer where conventional (continued on next page) computed tomography and technetium Tc 99m bone scan imaging are unable to detect disease after curative intervention despite rising prostate-specific antigen. There is no clear standard of care and no systemic therapy has been shown to improve survival. Immunotherapy-based treatments potentially are attractive options relative to androgen deprivation therapy due to the generally more favorable side-effect profile. Biochemically recurrent prostate cancer patients have a low tumor burden and likely lymph node-based disease, which may make them more likely to respond to immunotherapy.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Imunoterapia/métodos , Recidiva Local de Neoplasia/terapia , Neoplasias da Próstata/patologia , Idoso , Terapia Combinada , Intervalo Livre de Doença , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/diagnóstico por imagem , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/patologia , Seleção de Pacientes , Prognóstico , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/terapia , Medição de Risco , Papel (figurativo) , Análise de Sobrevida , Tomografia Computadorizada por Raios X/métodos , Resultado do Tratamento
19.
Artigo em Inglês | MEDLINE | ID: mdl-31528835

RESUMO

PURPOSE: Despite decreased screening-based detection of clinically insignificant tumors, most diagnosed prostate cancers are still indolent, indicating a need for better strategies for detection of clinically significant disease before treatment. We hypothesized that patients with detectable circulating tumor DNA (ctDNA) were more likely to harbor aggressive disease. METHODS: We applied ultra-low-pass whole-genome sequencing to profile cell-free DNA from 112 patients diagnosed with localized prostate cancer and performed targeted resequencing of plasma DNA for somatic mutations previously identified in matched solid tumor in nine cases. We also performed similar analyses of data from patients with metastatic prostate cancer. RESULTS: In all cases of localized prostate cancer, even in clinically high-risk patients who subsequently had recurrent disease, ultra-low-pass whole-genome sequencing and targeted resequencing did not detect ctDNA in plasma acquired before surgery or before recurrence. In contrast, using both approaches, ctDNA was detected in patients with metastatic prostate cancer. CONCLUSION: Our findings demonstrate clear differences between localized and advanced prostate cancer with respect to the dissemination and detectability of ctDNA. Because allele-specific alterations in ctDNA are below the threshold for detection in localized prostate cancer, other approaches to identify cell-free nucleic acids of tumor origin may demonstrate better specificity for aggressive disease.

20.
Urology ; 131: 182-183, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31451158
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA