15 research outputs found

    The interplay of growth differentiation factor 15 (GDF15) expression and M2 macrophages during prostate carcinogenesis

    Get PDF
    M2 (tumor-supportive) macrophages may upregulate growth differentiation factor 15 (GDF15), which is highly expressed in prostate tumors, but the combined utility of these markers as prognostic biomarkers are unclear. We retrospectively studied 90 prostate cancer cases that underwent radical prostatectomy as their primary treatment and were followed for biochemical recurrence (BCR). These cases also had a benign prostate biopsy at least 1 year or more before their prostate cancer surgery. Using computer algorithms to analyze digitalized immunohistochemically stained slides, GDF15 expression and the presence of M2 macrophages based on the relative density of CD204- and CD68-positive macrophages were measured in prostate: (i) benign biopsy, (ii) cancer and (iii) tumor-adjacent benign (TAB) tissue. Both M2 macrophages (P = 0.0004) and GDF15 (P \u3c 0.0001) showed significant inter-region expression differences. Based on a Cox proportional hazards model, GDF15 expression was not associated with BCR but, in men where GDF15 expression differences between cancer and TAB were highest, the risk of BCR was significantly reduced (hazard ratio = 0.26; 95% confidence interval = 0.09-0.94). In addition, cases with high levels of M2 macrophages in prostate cancer had almost a 5-fold increased risk of BCR (P = 0.01). Expression of GDF15 in prostate TAB was associated with M2 macrophage levels in both prostate cancer and TAB and appeared to moderate M2-macrophage-associated BCR risk. In summary, the relationship of GDF15 expression and CD204-positive M2 macrophage levels is different in a prostate tumor environment compared with an earlier benign biopsy and, collectively, these markers may predict aggressive disease

    Racial differences in the systemic inflammatory response to prostate cancer

    Get PDF
    Systemic inflammation may increase risk for prostate cancer progression, but the role it plays in prostate cancer susceptibility is unknown. From a cohort of over 10,000 men who had either a prostate biopsy or transurethral resection that yielded a benign finding, we analyzed 517 incident prostate cancer cases identified during follow-up and 373 controls with one or more white blood cell tests during a follow-up period between one and 18 years. Multilevel, multivariable longitudinal models were fit to two measures of systemic inflammation, neutrophil-to-lymphocyte ratio (NLR) and monocyte-to-lymphocyte ratio (MLR), to determine NLR and MLR trajectories associated with increased risk for prostate cancer. For both measures, we found no significant differences in the trajectories by case/control status, however in modeling NLR trajectories there was a significant interaction between race (white or Black and case-control status. In race specific models, NLR and MLR values were consistently higher over time among white controls than white cases while case-control differences in NLR and MLR trajectories were not apparent among Black men. When cases were classified as aggressive as compared to non-aggressive, the case-control differences in NLR and MLR values over time among white men were most apparent for non-aggressive cases. For NLR among white men, significant case-control differences were observed for the entire duration of observation for men who had inflammation in their initial prostate specimen. It is possible that, among white men, monitoring of NLR and MLR trajectories after an initial negative biopsy may be useful in monitoring prostate cancer risk

    Growth and differentiation factor 15 and NF-κB expression in benign prostatic biopsies and risk of subsequent prostate cancer detection

    Get PDF
    Growth and differentiation factor 15 (GDF-15), also known as macrophage inhibitory cytokine 1 (MIC-1), may act as both a tumor suppressor and promotor and, by regulating NF-κB and macrophage signaling, promote early prostate carcinogenesis. To determine whether expression of these two inflammation-related proteins affect prostate cancer susceptibility, dual immunostaining of benign prostate biopsies for GDF-15 and NF-κB was done in a study of 503 case-control pairs matched on date, age, and race, nested within a historical cohort of 10,478 men. GDF-15 and NF-κB expression levels were positively correlated (r = 0.39; p \u3c 0.0001), and both were significantly lower in African American (AA) compared with White men. In adjusted models that included both markers, the odds ratio (OR) for NF-κB expression was statistically significant, OR =0.87; p = 0.03; 95% confidence interval (CI) =0.77-0.99, while GDF-15 expression was associated with a nominally increased risk, OR =1.06; p = 0.27; 95% CI =0.96-1.17. When modeling expression levels by quartiles, the highest quartile of NF-κB expression was associated with almost a fifty percent reduction in prostate cancer risk (OR =0.51; p = 0.03; 95% CI =0.29-0.92). In stratified models, NF-κB had the strongest negative association with prostate cancer in non-aggressive cases (p = 0.03), older men (p = 0.03), and in case-control pairs with longer follow-up (p = 0.02). Risk associated with GDF-15 expression was best fit using nonlinear regression modeling where both first (p = 0.02) and second (p = 0.03) order GDF-15 risk terms were associated with significantly increased risk. This modeling approach also revealed significantly increased risk associated with GDF-15 expression for subsamples defined by AA race, aggressive disease, younger age, and in case-control pairs with longer follow-up. Therefore, although positively correlated in benign prostatic biopsies, NF-κB and GDF-15 expression appear to exert opposite effects on risk of prostate tumor development

    Racial Disparities in Expression of GDF15 and NFκB in Prostate Cancer and Benign Prostatic Epithelium

    Get PDF
    Prostate cancer (PC) outcomes are more adverse for African-American (AA) than white (W) men. Growth differentiation factor 15 (GDF15, PDF, NAG-1) is a stress-induced anti-inflammatory cytokine with immunosuppressive and tumor growth-promoting functions. GDF15 inversely regulates NFκB, a transcription factor enabling pro-inflammatory gene expression and becomes constitutively activated in androgen-independent PC. Tissue microarrays (TMAs), prepared from prostatectomy tissue at three institutions, comprised 688 cases (364 W and 324 AA). Each case included ≥3 tumor punches plus ≥3 non-neoplastic punches. TMAs were stained separately for GDF15 and NFκB and evaluated by two pathologists, using the 0-3+ scale. PC, compared to benign epithelium, had elevated mean GDF15 expression (1.93 vs. 0.99) and also, NFκB (1.18 vs. 0.96, both P<0.0001). Only in AA men did PC show gradewise or stagewise altered expression of these markers. In AA men, GDF15 expression fell as stage rose in PC (P=0.007) and also in benign epithelium (P =0.003). In W men, GDF15 expression in benign epithelium fell as stage (P=0.01) and grade (P=0.01) rose. NFκB expression was higher in AA than W men only in high-grade PC (P =0.01). NFκB expression rose with increasing tumor grade only in AA men (P =0.027) and in the benign prostate component only in W men (P=0.007). Benign and tumor NFκB expression did not vary with stage. PC showed significant alterations in GDF15 and NFκB expression in accord with cancer aggressiveness in AA men only: stagewise decrease in GDF15, and gradewide increase in NFκB.  Findings suggest a disparity for immune response by race in prostate carcinogenesis

    Convolutional Neural Network Quantification of Gleason Pattern 4 and Association with Biochemical Recurrence in Intermediate Grade Prostate Tumors

    No full text
    Differential classification of prostate cancer (CaP) grade group (GG) 2 and 3 tumors remains challenging, likely due to the subjective quantification of percentage of Gleason pattern 4 (%GP4). Artificial intelligence assessment of %GP4 may improve its accuracy and reproducibility and provide information for prognosis prediction. To investigate this potential, a convolutional neural network (CNN) model was trained to objectively identify and quantify Gleason pattern (GP) 3 and 4 areas, estimate %GP4, and assess whether CNN-assessed %GP4 is associated with biochemical recurrence (BCR) risk in intermediate risk GG 2 and 3 tumors. The study was conducted in a radical prostatectomy cohort (1999-2012) of African American men from the Henry Ford Health System (Detroit, Michigan). A CNN model that could discriminate four tissue types (stroma, benign glands, GP3 glands, and GP4 glands) was developed using histopathologic images containing GG 1 (n=45) and 4 (n=20) tumor foci. The CNN model was applied to GG 2 (n=153) and 3 (n=62) for %GP4 estimation, and Cox proportional hazard modeling was used to assess the association of %GP4 and BCR, accounting for other clinicopathologic features including GG. The CNN model achieved an overall accuracy of 86% in distinguishing the four tissue types. Further, CNN-assessed %GP4 was significantly higher in GG 3 compared with GG 2 tumors (p=7.2*10(-11)). %GP4 was associated with an increased risk of BCR (adjusted HR=1.09 per 10% increase in %GP4, p=0.010) in GG 2 and 3 tumors. Within GG 2 tumors specifically, %GP4 was more strongly associated with BCR (adjusted HR=1.12, p=0.006). Our findings demonstrate the feasibility of CNN-assessed %GP4 estimation, which is associated with BCR risk. This objective approach could be added to the standard pathological assessment for patients with GG 2 and 3 tumors and act as a surrogate for specialist genitourinary pathologist evaluation when such consultation is not available

    Breast and prostate cancers harbor common somatic copy number alterations that consistently differ by race and are associated with survival

    No full text
    BACKGROUND: Pan-cancer studies of somatic copy number alterations (SCNAs) have demonstrated common SCNA patterns across cancer types, but despite demonstrable differences in aggressiveness of some cancers by race, pan-cancer SCNA variation by race has not been explored. This study investigated a) racial differences in SCNAs in both breast and prostate cancer, b) the degree to which they are shared across cancers, and c) the impact of these shared, race-differentiated SCNAs on cancer survival. METHODS: Utilizing data from The Cancer Genome Atlas (TCGA), SCNAs were identified using GISTIC 2.0, and in each tumor type, differences in SCNA magnitude between African Americans (AA) and European Americans (EA) were tested using linear regression. Unsupervised hierarchical clustering of the copy number of genes residing in race-differentiated SCNAs shared between tumor types was used to identify SCNA-defined patient groups, and Cox proportional hazards regression was used to test for association between those groups and overall/progression-free survival (PFS). RESULTS: We identified SCNAs that differed by race in breast (n = 58 SCNAs; permutation p \u3c 10(- 4)) and prostate tumors (n = 78 SCNAs; permutation p = 0.006). Six race-differentiated SCNAs common to breast and prostate found at chromosomes 5q11.2-q14.1, 5q15-q21.1, 8q21.11-q21.13, 8q21.3-q24.3, 11q22.3, and 13q12.3-q21.3 had consistent differences by race across both tumor types, and all six were of higher magnitude in AAs, with the chromosome 8q regions being the only amplifications. Higher magnitude copy number differences in AAs were also identified at two of these race-differentiated SCNAs in two additional hormonally-driven tumor types: endometrial (8q21.3-q24.3 and 13q12.3-q21.3) and ovarian (13q12.3-q21.3) cancers. Race differentiated SCNA-defined patient groups were significantly associated with survival differences in both cancer types, and these groups also differentiated within triple negative breast cancers based on PFS. While the frequency of the SCNA-defined patient groups differed by race, their effects on survival did not. CONCLUSIONS: This study identified race-differentiated SCNAs shared by two related cancers. The association of SCNA-defined patient groups with survival demonstrates the clinical significance of combinations of these race-differentiated genomic aberrations, and the higher frequency of these alterations in AA relative to EA patients may explain racial disparities in risk of aggressive breast and prostate cancer

    Breast and prostate cancers harbor common somatic copy number alterations that consistently differ by race-ethnicity

    No full text
    Pan-cancer studies of somatic copy number alterations (SCNAs) have demonstrated shared SCNAs across cancer types, but whether these shared SCNAs vary by raceethnicity has not been explored. Utilizing data from The Cancer Genome Atlas (TCGA), we identified SCNAs in breast and prostate tumors, two cancers with racially-disparate outcomes, and then tested for differences in SCNA magnitude by self-reported African American and European American race-ethnicity, as well as by regional chromosomal African ancestry within African Americans. GISTIC2 was applied to high density SNP array data to map SCNA regions in 712 European and 174 African American female breast tumors and 267 European and 42 African American prostate tumors derived from the TCGA dataset. For each tumor, SCNA magnitude was quantified by the area under the logarithm-base 2 copy number curve, and the germline ancestral origin of SCNAs was inferred using RFMix. A linear model was used to assess the association between SCNA magnitude and race-ethnicity (or regional African ancestry) while adjusting age-at-diagnosis and tumor severity. Race-differentiated SCNAs common to breast and prostate were found at chromosomes 5q11-21, 6q12-14, 6q16-22, 8q21-24, 11q22, 13q12-21, and 16q21-24, with 8q21-24 being the only amplification. African American breast and prostate tumors had higher magnitude alterations in the regions on 5q11-21, 8q21-24, 11q22, and 13q12-21, and among African Americans, this higher magnitude at 8q21- 24 and 13q12-21 was consistent with increasing regional African ancestry. Within these regions with higher magnitude SCNAs in African Americans, expression analysis revealed 18 cancer genes, including RB1 and PVT1, differentially expressed by race-ethnicity in both tumors types that were consistent with the observed SCNA differences. While differences in SCNAs by race-ethnicity have been studied in single cancers, this is the first study to identify race-differentiated SCNAs shared by two hormonally-driven cancers and to explore the potential of germline genetic ancestry as a mechanism leading to this differentiation. The differentially expressed genes within SCNAs common to both tumor types could provide further insight into the racially disparate outcomes in breast and prostate cancers

    Breast and prostate cancers harbor common somatic copy number alterations that consistently differ by race-ethnicity

    No full text
    Pan-cancer studies of somatic copy number alterations (SCNAs) have demonstrated shared SCNAs across cancer types, but whether these shared SCNAs vary by raceethnicity has not been explored. Utilizing data from The Cancer Genome Atlas (TCGA), we identified SCNAs in breast and prostate tumors, two cancers with racially-disparate outcomes, and then tested for differences in SCNA magnitude by self-reported African American and European American race-ethnicity, as well as by regional chromosomal African ancestry within African Americans. GISTIC2 was applied to high density SNP array data to map SCNA regions in 712 European and 174 African American female breast tumors and 267 European and 42 African American prostate tumors derived from the TCGA dataset. For each tumor, SCNA magnitude was quantified by the area under the logarithm-base 2 copy number curve, and the germline ancestral origin of SCNAs was inferred using RFMix. A linear model was used to assess the association between SCNA magnitude and race-ethnicity (or regional African ancestry) while adjusting age-at-diagnosis and tumor severity. Race-differentiated SCNAs common to breast and prostate were found at chromosomes 5q11-21, 6q12-14, 6q16-22, 8q21-24, 11q22, 13q12-21, and 16q21-24, with 8q21-24 being the only amplification. African American breast and prostate tumors had higher magnitude alterations in the regions on 5q11-21, 8q21-24, 11q22, and 13q12-21, and among African Americans, this higher magnitude at 8q21- 24 and 13q12-21 was consistent with increasing regional African ancestry. Within these regions with higher magnitude SCNAs in African Americans, expression analysis revealed 18 cancer genes, including RB1 and PVT1, differentially expressed by race-ethnicity in both tumors types that were consistent with the observed SCNA differences. While differences in SCNAs by race-ethnicity have been studied in single cancers, this is the first study to identify race-differentiated SCNAs shared by two hormonally-driven cancers and to explore the potential of germline genetic ancestry as a mechanism leading to this differentiation. The differentially expressed genes within SCNAs common to both tumor types could provide further insight into the racially disparate outcomes in breast and prostate cancers

    Breast and Prostate Cancers Harbor Common Somatic Copy Number Alterations that Consistently Differ by Race-Ethnicity

    No full text
    Pan-cancer studies of somatic copy number alterations (SCNAs) have demonstrated shared SCNAs across cancer types, but whether these shared SCNAs vary by raceethnicity has not been explored. Utilizing data from The Cancer Genome Atlas (TCGA), we identified SCNAs in breast and prostate tumors, two cancers with racially-disparate outcomes, and then tested for differences in SCNA magnitude by self-reported African American and European American race-ethnicity, as well as by regional chromosomal African ancestry within African Americans. GISTIC2 was applied to high density SNP array data to map SCNA regions in 712 European and 174 African American female breast tumors and 267 European and 42 African American prostate tumors derived from the TCGA dataset. For each tumor, SCNA magnitude was quantified by the area under the logarithm-base 2 copy number curve, and the germline ancestral origin of SCNAs was inferred using RFMix. A linear model was used to assess the association between SCNA magnitude and race-ethnicity (or regional African ancestry) while adjusting age-at-diagnosis and tumor severity. Race-differentiated SCNAs common to breast and prostate were found at chromosomes 5q11-21, 6q12-14, 6q16-22, 8q21-24, 11q22, 13q12-21, and 16q21-24, with 8q21-24 being the only amplification. African American breast and prostate tumors had higher magnitude alterations in the regions on 5q11-21, 8q21-24, 11q22, and 13q12-21, and among African Americans, this higher magnitude at 8q21- 24 and 13q12-21 was consistent with increasing regional African ancestry. Within these regions with higher magnitude SCNAs in African Americans, expression analysis revealed 18 cancer genes, including RB1 and PVT1, differentially expressed by race-ethnicity in both tumors types that were consistent with the observed SCNA differences. While differences in SCNAs by race-ethnicity have been studied in single cancers, this is the first study to identify race-differentiated SCNAs shared by two hormonally-driven cancers and to explore the potential of germline genetic ancestry as a mechanism leading to this differentiation. The differentially expressed genes within SCNAs common to both tumor types could provide further insight into the racially disparate outcomes in breast and prostate cancers

    Clear cell renal cell carcinoma with focal psammomatous calcifications: a rare occurrence mimicking translocation carcinoma

    No full text
    AIMS: Renal cell carcinoma (RCC) with clear cells and psammoma-like calcifications would often raise suspicion for MITF family translocation RCC. However, we have rarely encountered tumours consistent with clear cell RCC that contain focal psammomatous calcifications. METHODS AND RESULTS: We identified clear cell RCCs with psammomatous calcifications from multiple institutions and performed immunohistochemistry and fluorescence and RNA in-situ hybridisation (FISH and RNA ISH). Twenty-one tumours were identified: 12 men, nine women, aged 45-83 years. Tumour size was 2.3-14.0 cm (median = 6.75 cm). Nucleolar grade was 3 (n = 14), 2 (n = 4) or 4 (n = 3). In addition to clear cell pattern, morphology included eosinophilic (n = 12), syncytial giant cell (n = 4), rhabdoid (n = 2), branched glandular (n = 1), early spindle cell (n = 1) and poorly differentiated components (n = 1). Labelling for CA9 was usually 80-100% of the tumour cells (n = 17 of 21), but was sometimes decreased in areas of eosinophilic cells (n = 4). All (19 of 19) were positive for CD10. Most (19 of 20) were positive for AMACR (variable staining = 20-100%). Staining was negative for keratin 7, although four showed rare positive cells (four of 20). Results were negative for cathepsin K (none of 19), melan A (none of 17), HMB45 (none of 17), TFE3 (none of 5), TRIM63 RNA ISH (none of 13), and TFE3 (none of 19) and TFEB rearrangements (none of 12). Seven of 19 (37%) showed chromosome 3p deletion. One (one of 19) showed trisomy 7 and 17 without papillary features. CONCLUSIONS: Psammomatous calcifications in RCC with a clear cell pattern suggests a diagnosis of MITF family translocation RCC; however, psammomatous calcifications can rarely be found in true clear cell RCC
    corecore