The prognostic significance of loss of the androgen receptor and neuroendocrine differentiation in prostate biopsy specimens among castration-resistant prostate cancer patients

  • Authors:
    • Akira Komiya
    • Kenji Yasuda
    • Akihiko Watanabe
    • Yasuyoshi Fujiuchi
    • Toyonori Tsuzuki
    • Hideki Fuse
  • View Affiliations

  • Published online on: January 14, 2013     https://doi.org/10.3892/mco.2013.69
  • Pages: 257-262
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Prostate cancer (PCa) is a leading cause of mortality, and despite good response to androgen ablation this response is eventually lost. In the present study, androgen receptor (AR) expression and neuroendocrine differentiation (NED) were evaluated in hormone-sensitive (HSPC) and castration-resistant prostate cancers (CRPC). Prostate tissues were obtained from 20 HSPC patients at diagnosis and 28 CRPC patients at castration‑resistant progression. AR, chromogranin A (CGA) and neuron-specific enolase (NSE) were evaluated by immunohistochemical staining (IHS) in representative positive cores for PCa. IHS intensity was graded as negative, 0; positive, 1+ and strongly positive, 2+. The proportion of the 1+ and 2+ areas in PCa cells was determined. PCa was considered to be in NED if ≥50% of the tumor cells were 1+ or 2+ for CGA or NSE. The observed IHS intensity (0/1+/2+) for AR, CGA and NSE was 0/4/16, 5/11/4 and 11/4/5 in HSPC patients and 9/3/16, 5/8/15 and 8/4/16 in CRPC patients, respectively. AR expression was positive in all the HSPC and 19/28 CRPC patients (P=0.0049). NED was observed in 9/20 HSPC and 20/28 CRPC patients (P=0.0649). NED was significantly associated with a negative AR expression in CRPC patients (P=0.0292). Multivariate analysis revealed that age, AR expression and strong NED were independent parameters for prognosis following castration-resistant progression. In conclusion, prostate biopsy following castration‑resistant progression was necessary. AR was lost in a subset of CRPC. NED was observed more frequently in CRPC vs. HSPC and was associated with a worse prognosis.

Introduction

Prostate cancer (PCa) is a leading cause of mortality in Western countries. In Japan, PCa incidence and mortality rates are on the increase, despite efforts to screen and diagnose patients early and despite extensive efforts to treat this disease (1,2). There are a number of modalities for localized PCa. However, androgen deprivation therapy remains the standard method used to treat patients with advanced PCa. During hormone therapy, PCa responds well to androgen ablation initially. However, this response is gradually lost and the disease becomes castration-resistant prostate cancer (CRPC), defined as disease progression, even in the presence of castration levels of circulating androgens (3,4). The androgen receptor (AR) remains functional and is important in the mechanisms of CRPC (5), even at this stage.

Evidence for persistent hormone dependence in CRPC facilitated the development of novel anti-androgens capable of blocking testosterone synthesis by the testes as well as adrenal glands and prostate tumor tissue. Abiraterone acetate is an oral, selective and irreversible inhibitor of CYP17, a critical enzyme in androgen biosynthesis, which blocks non-gonadal androgen production. Abiraterone at a dose of 1000 mg/day in combination with prednisone at a dose of 10 mg/day was approved by the Food and Drug Administration (FDA) in April, 2011, and by the European Medicines Agency (EMA) in July, 2011 for the treatment of metastatic CRPC. The approvals were based on the results of the COU-AA-301 trial conducted in patients who received previous docetaxel chemotherapy, which demonstrated a survival benefit for the experimental arm versus the placebo (6). New anti-androgens with improved binding properties have also been developed. One of these agents is enzalutamide, an oral AR antagonist small molecule that binds to ARs with a higher affinity compared to bicalutamide and blocks AR nuclear translocation, co-activator recruitment and DNA binding without agonist activity when AR is overexpressed (7). Results of the AFFIRM trial showed that enzalutamide was approved by the FDA in August, 2012 (8). These novel compounds were reported to have survival benefits. However, the CRPC is likely to eventually progress to a phase of disease that is resistant to these types of therapy.

One of the mechanisms of androgen-independent (AI) progression of PCa is neuroendocrine differentiation (NED). Neuroendocrine (NE) cells originally exist in the normal prostate acini and duct and they regulate prostatic growth, differentiation and secretion. The lack of AR expression is indicative of the AI activity of NE cells. Clusters of malignant NE cells are found among adenocarcinoma cells in most PC, sharing a common clonal origin. The prostate NE cells affect the target cells in an AI manner. Therefore, the NE pathway is thought to be one of the most significant mechanisms for AIPC or CRPC (9). NE cells are detected by immunohistochemical staining (IHS) using antibodies against neuropeptides, of which CGA and NSE are the most thoroughly investigated and are believed to be the best markers for prostatic NED (9). Using CGA and NSE as NE markers, Tanaka et al (10) reported that lesions predominantly composed of a neuroendocrine cell tumor were found in 4 of 20 autopsy cases (Japanese patients with PCa). However, AR is expressed in almost all types of cancer of the prostate, before and after androgen ablation therapy (11). Specific downregulation of the AR with anti-AR antibodies or siRNAs results in AI prostate cancer (or CRPC), cell growth inhibition and a reduction in PSA expression (12,13). In addition, recent clinical data, such as abiraterone acetate and enzalutamide, support the AR function in CRPC.

The aims of this study were to compare the AR expression and NED between hormone-sensitive PCa (HSPC) and CRPC patients using IHS and to analyze the diagnostic and prognostic significance of this expression in CRPC.

Materials and methods

Patients and PCa tissues

PCa tissues were retrospectively obtained using prostate needle biopsies from 20 HSPC patients at initial diagnosis. Consecutive CRPC specimens were obtained using prostate needle biopsies in 24 patients and transurethral resections (TUR) in 4 patients (CRPC biopsy). Cores (≥10) were obtained using Transrectal Ultrasound-Guided Prostate (TRUS) biopsies. CRPC was defined as three increases in the PSA level at least 1 month apart, or evidence of a new clinical disease while the patient was receiving androgen deprivation therapy and the testosterone levels were at castrate levels (14). Patient characteristics are shown in Table I. The median follow-up following CRPC biopsies was 15.6 months (range, 1–57). From the formalin-fixed paraffin-embedded tissue samples, multiple 4-μm sections were prepared using a microtome and transferred to glass slides (Fisherbrand Superfrost Plus; Fisher Scientific, Pittsburg, PA, USA). Chromogranin A (CGA), neuron-specific enolase (NSE) and AR were immunohistochemically stained in the representative positive cores or TUR specimens for PCa.

Table I

Patient characteristics.

Table I

Patient characteristics.

HSPC
CRPC
VariablesMeanSDMeanSD
Age (years)74.16.273.37.1
iPSA (ng/ml)11102141.3412.4750.3
rPSA (ng/ml)77.3175.0
Gleason score7.91.5
Time to CRPC (months)n.a.33.551
No.
  M+1025
  M−103

[i] HSPC, hormone-sensitive prostate cancer; CRPC, castration-resistant prostate cancer; SD, standard deviation; iPSA, initial prostate-specific antigen; rPSA, PSA at castration-resistant progression; M, metastasis; n.a., not available.

Antibodies

Antibodies for the IHS were: Anti-CGA, a rabbit polyclonal antibody (A0430; dilution, 1:800) (Dako, Carpinteria, CA, USA); anti-NSE antibody, a mouse monoclonal antibody (M0873; dilution, 1:200) (Dako) and anti-AR antibody, a mouse monoclonal antibody (BSB6076; dilution, 1:50) (BioSB).

IHS

The glass slides were incubated with the primary antibodies at an optimized titer and were diluted using Universal Blocking Reagent (BioGenex, Fremont, CA, USA) for 30 min, then washed three times with KN buffer (KN-09001; Pathology Institute, Toyama, Japan). The sections were then incubated with a horse-radish peroxidase dextran polymer-labeled goat anti-mouse antibody or goat anti-rabbit antibody (ChemMate EnVision kit; dilution 1:100) (Dako) for 30 min at room temperature (RT). The sections were then washed three times in KN buffer and incubated at RT with 3,3′-diaminobenzidine (DAB; Dako) for 10 min. The sections were rinsed several times with distilled water and were counterstained with Mayer’s hematoxylin, dehydrated, cleared and mounted with resinous mounting medium (15).

The staining intensity was graded as negative, 0; positive, 1+ and strongly positive, 2+ (Fig. 1). The proportion of 1+ and 2+ areas in the PCa cells was determined. These analyses were carried out by an experienced pathologist (T.T.). PCa was regarded as NED if ≥50% of the tumor cells were positive for CGA and/or NSE. In addition, PCa was regarded as strong NED if ≥50% of the tumor cells were 2+ for CGA and/or NSE.

Statistical analysis

Statistical analysis was performed using the Chi-square test to compare the two groups. The cause-specific survival was estimated for each study group using the Kaplan-Meier method and the differences were assessed using the Wilcoxon test. For the multivariate analysis, the Cox proportional hazard model was used to identify independent variables to predict cause-specific survival following CRPC biopsy. P<0.05 was considered to indicate a statistically significanct difference. We used the JMP software version 8.0.1 (SAS Institute, Tokyo, Japan) for the statistical analyses. The study was approved by the institutional review board of the University of Toyama.

Results

Immunohistological staining intensity

Distribution of the staining intensity is shown in Table II. CRPC exhibited more cases with strong NE intensity. The AR expression was positive in all the HSPC. However, 9/28 (32.1%) CRPC patients were negative for AR, which was more frequent compared to HSPC patients (P=0.0049, Table III). NED was positive in 9/20 HSPC (45.0%) and in 20/28 CRPC (71.4%). CRPC showed a tendency to have more NED compared to HSPC (P=0.0649; Table III).

Table II

Distribution of IHS intensity.

Table II

Distribution of IHS intensity.

HSPC
CRPC
IHS intensityCGA (No.)NSE (No.)AR (No.)CGA (No.)NSE (No.)AR (No.)
05110589
1+1144843
2+4516151616

[i] IHS, immunohistochemical staining; 0, negative; 1+, positive; 2+, strongly positive; HSPC, hormone-sensitive prostate cancer; CRPC, castration-resistant prostate cancer; CGA, chromogranin A; NSE, neuron-specific enolase; AR, androgen receptor.

Table III

AR expression and NED in HSPC and CRPC.

Table III

AR expression and NED in HSPC and CRPC.

HSPC (No.)CRPC (No.)P-value (Chi-square test)
AR expression
  Negative090.0049
  Positive2019
NED
  Negative1180.0649
  Positive920

[i] AR, androgen receptor; NED, neuroendocrine differentiation; HSPC, hormone-sensitive prostate cancer; CRPC, castration-resistant prostate cancer.

In CRPC, 11/19 patients (57.9%) were NED-positive when the AR was positive. However, NED was positive in the CRPC with a negative AR expression (n=9). Therefore, NED was significantly associated with a negative AR expression in CRPC (P=0.0292, Table IV).

Table IV

Association between NED and androgen receptor expression in CRPC.

Table IV

Association between NED and androgen receptor expression in CRPC.

NED
AR expressionNegative (No.)Positive (No.)P-value (Chi-square test)
Negative090.0292
Positive811

[i] AR, androgen receptor; NED, neuroendocrine differentiation.

Survival after CRPC biopsy

Table V shows the treatment modalities in CRPC patients before and after CRPC biopsy. Prior to CRPC biopsy, the main treatments were hormonal manipulations, whereas cytotoxic agents were mainly used following CRPC biopsy (Table V).

Table V

Treatments before and after CRPC biopsy.

Table V

Treatments before and after CRPC biopsy.

TreatmentsBefore CRPC biopsy (No. of patients)After CRPC biopsy (No. of patients)
Radiation37
Radical prostatectomy20
Alternative anti-androgen136
Estramustine monophosphate520
Cisplatin02
Low-dose prednisone12
Low-dose dexamethasone25
Ethinylestradiol20
Paclitaxel011
Docetaxel016
Etoposide12
Angiotensin II receptor blockers02

[i] CRPC, castration-resistant prostate cancer.

The cause-specific survival was available in 27 patients with CRPC. Fig. 2 shows survival curves based on the AR expression in CRPC. Nine CRPC patients were negative for the AR at CRPC biopsy. AR expression was not associated with prostate cancer-specific survival in this univariate analysis. By contrast, strong NED was associated with a poorer prognosis following CRPC biopsy (P=0.0117; Fig. 3). A multivariate analysis demonstrated that age, AR expression and a strong NED were independent characteristics for prognosis following CRPC biopsy (Table VI).

Table VI

A multivariate analysis of the prognostic features after CRPC biopsy.

Table VI

A multivariate analysis of the prognostic features after CRPC biopsy.

CharacteristicsCI Lower to upper limitsChi-squareP-value (Prob > ChiSq)Hazard ratio
Age at CRPC biopsya (years)0.00233 to 0.255893.991410.04573b1.13021
iPSAa (ng/ml)−0.00051 to 0.000101.020920.312300.99987
rPSAa (ng/ml)−0.02385 to 0.010260.365360.545540.99509
M at CRPC biopsy +/−−2.67880 to 0.066443.356160.066958.30273
AR +/−−2.68520 to −0.096164.598820.03199b12.21495
Strong NED +/−0.42617 to 3.212257.588270.00587b26.75226

a Continuous;

b statistically significant.CRPC, castration-resistant prostate cancer; PSA, prostate-specific antigen; iPSA, initial PSA; rPSA, PSA at castration-resistant progression; M, metastasis; AR, androgen receptor; NED, neuroendocrine differentiation; CI, confidence interval.

Discussion

In the present study, the AR was positive for the HSPC. However, 9/28 patients (32.1%) with CRPC were negative for the AR. Therefore, a significant number of CRPC cases did not have the AR in the primary prostate. The information for AR expression in CRPC is limited. Generally, it has been reported that the AR is expressed in nearly all types of cancers of the prostate, before and after androgen ablation therapy (11). The high levels of AR expression and the development of hypersensitive receptors have been recognized as a feature associated with the development of CRPC (13,16). Hobisch et al (17) showed that metastases from PCa expressed the AR following endocrine therapy. From a Rapid Autopsy Program, Shah et al (18) indicated that the majority of patients continued to express substantial quantities of AR despite having undergone a long-term androgen ablation. The patients demonstrated marked differences in AR expression between different tissue sites (2- to 50-fold) and several patients demonstrated a high amount of AR staining, although they were no longer responding to androgen-deprivation therapy. In the Japanese studies, Takeda et al (19) demonstrated that patients with metastatic PCa with ≥48% AR-positive cells had a significantly better outcome, in terms of progression-free and cause-specific survival compared to patients with <48% AR content. Masai et al (20) have shown that 33% of cancer cells were positive for the AR in regrowing prostates from 8 relapsed cancers after endocrine therapy. The AR-positive cells were more frequently found in untreated PCa. However, the frequency of AR loss was not shown in the Japanese series. Matei et al (21) previously reported that the AR was positive in neoplastic cells in 20/47 patients (53%) with CRPC. Therefore, 47% of CRPC cases lost AR expression, which is higher compared to our series. These results indicate that loss of the AR may be identified in a subset of CRPC and should alert physicians to the use of androgen- or AR-targeted therapies without conducting an evaluation of AR expression. It is important to analyze expression profiles prior to targeted therapies. For example, the expression of the estrogen and progesterone receptors and HER2 are frequently evaluated in breast cancer tissues to estimate the response to treatment. Trastuzumab is used for HER2+ breast cancer therapy (15).

NED was found in 9/20 HSPC (45%) and 20/28 CRPC cases (71%) in the present study. Its frequency was slightly higher in CRPC. Strong NED was found in 9/28 CRPC (32.1%). NED was determined using IHS or serum NE marker concentrations (9). Using IHS, the NE cells were identified in ∼10–100% of the untreated PCa tissues. This large discrepancy in prevalence between studies is explained in part by the lack of quantitative and consistent tissue-imaging techniques. McWilliam et al (22) found NED in 52% of PCa tissues using IHS for CGA and NSE. They also demonstrated a significant correlation between NED and worsening tumor differentiation, the presence of bone metastasis and poor patient survival. Kokubo et al (23) showed that 22% of stage D2 PCa overexpressed CGA by IHS and that CGA positivity was correlated with a shorter time to recurrence after hormone therapy. Kamiya et al (24) demonstrated that the cause-specific survival was significantly poorer after hormone therapy in stage D2 PCa with strongly positive staining for independent CGA and combined CGA with NSE. In the setting of CRPC, Mucci et al (25) reported that NE expression was heterogeneous and observed in 4/12 autopsy cases (33%) when the tumor sites per case were considered among rapid autopsies from men with hormone refractory PCa. Tanaka et al (10) reported that lesions predominantly composed of a neuroendocrine cell tumor were found in 4/20 autopsy cases (20%) of Japanese PCa patients. Therefore, our finding is consistent with findings of previous studies. However, information is limited in terms of the survival significance of NED in CRPC. Tanaka et al (10) also demonstrated that the survival was brief after relapse, although the duration of control by employing endocrine therapy varied in the neuroendocrine cell tumors found at autopsy. An evaluation of NED was performed only within the primary prostate in this study. The state of NED in metastases was unknown unlike the autopsy series. However, NED at CRPC biopsy was a strong and independent prognostic factor in the Cox proportional hazard model. NED was associated with a negative AR expression in this study, which is consistent with previous findings (9) and the significance of NED in the mechanisms of CRPC was confirmed. In addition, these results support the importance of performing a CRPC biopsy. In the present study, we found two neuroendocrine cancers by CRPC biopsy and patients underwent etoposide and cisplatin (EP) therapy (26,27) instead of taxane-based chemotherapy.

There are, however, limitations to this study. The study design was retrospective. The results may be different if performed in a prospective manner. The study sample size was small; however, this number of CRPC specimens is believed to be the best in Japan. The results may have been different if the study was conducted on a larger and different population. Quantification of the staining intensity was visually performed, which is a traditional method for IHS. This may be better analyzed by multiple pathologists or by using computer-aided methods to avoid inter-observer or inter-institutional variations (28). If NED was otherwise determined, the results could also have been affected. The CRPC biopsy findings were obtained only from the prostate. Therefore, the expression profiles in metastatic cites were not evaluated, unlike the autopsy series.

In conclusion, the AR was lost in a subset of CRPC, in which AR-targeted therapy might not be effective. NED was observed more frequently in CRPC vs. HSPC. NED was associated with a loss of the AR and worse prognosis in CRPC. Thus, CRPC biopsy may be useful to better characterize the diseases.

References

1 

Suzuki H, Komiya A, Kamiya N, Imamoto T, Kawamura K, Miura J, Suzuki N, Nakatsu H, Hata A and Ichikawa T: Development of a nomogram to predict probability of positive initial prostate biopsy among Japanese patients. Urology. 67:131–136. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Yano M, Imamoto T, Suzuki H, Fukasawa S, Kojima S, Komiya A, Naya Y and Ichikawa T: The clinical potential of pretreatment serum testosterone level to improve the efficiency of prostate cancer screening. Eur Urol. 51:375–380. 2007. View Article : Google Scholar

3 

Scher HI, Halabi S, Tannock I, Morris M, Sternberg CN, Carducci MA, Eisenberger MA, Higano C, Bubley GJ, Dreicer R, Petrylak D, Kantoff P, Basch E, Kelly WK, Figg WD, Small EJ, Beer TM, Wilding G, Martin A and Hussain M; Prostate Cancer Clinical Trials Working Group: Design and end points of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the Prostate Cancer Clinical Trials Working Group. J Clin Oncol. 26:1148–1159. 2008. View Article : Google Scholar

4 

Scher HI, Buchanan G, Gerald W, Butler LM and Tilley WD: Targeting the androgen receptor: improving outcomes for castration-resistant prostate cancer. Endocr Relat Cancer. 11:459–476. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Attar RM, Takimoto CH and Gottardis MM: Castration-resistant prostate cancer: locking-up the molecular escape routes. Clin Cancer Res. 15:3251–3255. 2009. View Article : Google Scholar : PubMed/NCBI

6 

De Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, Chi KN, Jones RJ, Goodman OB Jr, Saad F, Staffurth JN, Mainwaring P, Harland S, Flaig TW, Hutson TE, Cheng T, Patterson H, Hainsworth JD, Ryan CJ, Sternberg CN, Ellard SL, Fléchon A, Saleh M, Scholz M, Efstathiou E, Zivi A, Bianchini D, Loriot Y, Chieffo N, Kheoh T, Haqq CM and Scher HI; COU-AA-301 Investigators: Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med. 364:1995–2005. 2011.

7 

Tran C, Ouk S, Clegg NJ, Chen Y, Watson PA, Arora V, Wongvipat J, Smith-Jones PM, Yoo D, Kwon A, Wasielewska T, Welsbie D, Chen CD, Higano CS, Beer TM, Hung DT, Scher HI, Jung ME and Sawyers CL: Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science. 324:787–790. 2009. View Article : Google Scholar

8 

Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller MD, de Wit R, Mulders P, Chi KN, Shore ND, Armstrong AJ, Flaig TW, Fléchon A, Mainwaring P, Fleming M, Hainsworth JD, Hirmand M, Selby B, Seely L and de Bono JS; AFFIRM Investigators: Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 367:1187–1197. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Komiya A, Suzuki H, Imamoto T, Kamiya N, Nihei N, Naya Y, Ichikawa T and Fuse H: Neuroendocrine differentiation in the progression of prostate cancer. Int J Urol. 16:37–44. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Tanaka M, Suzuki Y, Takaoka K, Suzuki N, Murakami S, Matsuzaki O and Shimazaki J: Progression of prostate cancer to neuroendocrine cell tumor. Int J Urol. 8:431–436. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Grossmann ME, Huang H and Tindall DJ: Androgen receptor signaling in androgen-refractory prostate cancer. J Natl Cancer Inst. 93:1687–1697. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Zegarra-Moro OL, Schmidt LJ, Huang H and Tindall DJ: Disruption of androgen receptor function inhibits proliferation of androgen-refractory prostate cancer cells. Cancer Res. 62:1008–1013. 2002.PubMed/NCBI

13 

Chen CD, Welsbie DS, Tran C, Baek SH, Chen R, Vessella R, Rosenfeld MG and Sawyers CL: Molecular determinants of resistance to antiandrogen therapy. Nat Med. 10:33–39. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Crook JM, O’Callaghan CJ, Duncan G, Dearnaley DP, Higano CS, Horwitz EM, Frymire E, Malone S, Chin J, Nabid A, Warde P, Corbett T, Angyalfi S, Goldenberg SL, Gospodarowicz MK, Saad F, Logue JP, Hall E, Schellhammer PF, Ding K and Klotz L: Intermittent androgen suppression for rising PSA level after radiotherapy. N Engl J Med. 367:895–903. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Nagata T, Shimada Y, Sekine S, Hori R, Matsui K, Okumura T, Sawada S, Fukuoka J and Tsukada K: Prognostic significance of NANOG and KLF4 for breast cancer. Breast Cancer. Apr 17–2012.(Epub ahead of print).

16 

Bonkhoff H and Berges R: From pathogenesis to prevention of castration resistant prostate cancer. Prostate. 70:100–112. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Hobisch A, Culig Z, Radmayr C, Bartsch G, Klocker H and Hittmair A: Distant metastases from prostatic carcinoma express androgen receptor protein. Cancer Res. 55:3068–3072. 1995.PubMed/NCBI

18 

Shah RB, Mehra R, Chinnaiyan AM, Shen R, Ghosh D, Zhou M, Macvicar GR, Varambally S, Harwood J, Bismar TA, Kim R, Rubin MA and Pienta KJ: Androgen-independent prostate cancer is a heterogeneous group of diseases: lessons from a rapid autopsy program. Cancer Res. 64:9209–9216. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Takeda H, Akakura K, Masai M, Akimoto S, Yatani R and Shimazaki J: Androgen receptor content of prostate carcinoma cells estimated by immunohistochemistry is related to prognosis of patients with stage D2 prostate carcinoma. Cancer. 77:934–940. 1996. View Article : Google Scholar : PubMed/NCBI

20 

Masai M, Sumiya H, Akimoto S, Yatani R, Chang CS, Liao SS and Shimazaki J: Immunohistochemical study of androgen receptor in benign hyperplastic and cancerous human prostates. Prostate. 17:293–300. 1990. View Article : Google Scholar : PubMed/NCBI

21 

Matei DV, Renne G, Pimentel M, Sandri MT, Zorzino L, Botteri E, De Cicco C, Musi G, Brescia A, Mazzoleni F, Tringali V, Detti S and de Cobelli O: Neuroendocrine differentiation in castration-resistant prostate cancer: a systematic diagnostic attempt. Clin Genitourin Cancer. 10:164–173. 2012. View Article : Google Scholar : PubMed/NCBI

22 

McWilliam LJ, Manson C and George NJ: Neuroendocrine differentiation and prognosis in prostatic adenocarcinoma. Br J Urol. 80:287–290. 1997. View Article : Google Scholar : PubMed/NCBI

23 

Kokubo H, Yamada Y, Nishio Y, Fukatsu H, Honda N, Nakagawa A, Saga S, Tsuzuki T and Hara K: Immunohistochemical study of chromogranin A in Stage D2 prostate cancer. Urology. 66:135–140. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Kamiya N, Suzuki H, Kawamura K, Imamoto T, Naya Y, Tochigi N, Kakuta Y, Yamaguchi K, Ishikura H and Ichikawa T: Neuroendocrine differentiation in stage D2 prostate cancers. Int J Urol. 15:423–428. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Mucci NR, Akdas G, Manely S and Rubin MA: Neuroendocrine expression in metastatic prostate cancer: evaluation of high throughput tissue microarrays to detect heterogeneous protein expression. Hum Pathol. 31:406–414. 2000. View Article : Google Scholar

26 

Amato RJ, Logothetis CJ, Hallinan R, Ro JY, Sella A and Dexeus FH: Chemotherapy for small cell carcinoma of prostatic origin. J Urol. 147:935–937. 1992.PubMed/NCBI

27 

Komiya A, Yasuda K, Nozaki T, Fujiuchi Y, Hayashi S and Fuse H: Small cell carcinoma of the prostate after high-dose-rate brachytherapy for low-risk prostatic adenocarcinoma. Oncol Lett. 5:53–56. 2013.PubMed/NCBI

28 

Rizzardi AE, Johnson AT, Vogel RI, Pambuccian SE, Henriksen J, Skubitz AP, Metzger GJ and Schmechel SC: Quantitative comparison of immunohistochemical staining measured by digital image analysis versus pathologist visual scoring. Diagn Pathol. 7:422012. View Article : Google Scholar

Related Articles

Journal Cover

March-April 2013
Volume 1 Issue 2

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Komiya A, Yasuda K, Watanabe A, Fujiuchi Y, Tsuzuki T and Fuse H: The prognostic significance of loss of the androgen receptor and neuroendocrine differentiation in prostate biopsy specimens among castration-resistant prostate cancer patients. Mol Clin Oncol 1: 257-262, 2013
APA
Komiya, A., Yasuda, K., Watanabe, A., Fujiuchi, Y., Tsuzuki, T., & Fuse, H. (2013). The prognostic significance of loss of the androgen receptor and neuroendocrine differentiation in prostate biopsy specimens among castration-resistant prostate cancer patients. Molecular and Clinical Oncology, 1, 257-262. https://doi.org/10.3892/mco.2013.69
MLA
Komiya, A., Yasuda, K., Watanabe, A., Fujiuchi, Y., Tsuzuki, T., Fuse, H."The prognostic significance of loss of the androgen receptor and neuroendocrine differentiation in prostate biopsy specimens among castration-resistant prostate cancer patients". Molecular and Clinical Oncology 1.2 (2013): 257-262.
Chicago
Komiya, A., Yasuda, K., Watanabe, A., Fujiuchi, Y., Tsuzuki, T., Fuse, H."The prognostic significance of loss of the androgen receptor and neuroendocrine differentiation in prostate biopsy specimens among castration-resistant prostate cancer patients". Molecular and Clinical Oncology 1, no. 2 (2013): 257-262. https://doi.org/10.3892/mco.2013.69