ArticlePDF Available

Prostatic Hyperplasia: An Unknown Feature of Acromegaly

Authors:

Abstract and Figures

This study was designed to investigate whether GH and insulin-like growth factor I (IGF-I) excess could lead to the development of benign prostatic hyperplasia and/or prostatic carcinoma. Prostatic diameters and volume as well as the occurrence of prostatic diseases were studied by ultrasonography in 10 untreated acromegalic patients less than 40 yr of age and 10 age- and body mass index-matched healthy males. Serum GH, IGF-I, PRL, testosterone, dihydrotestosterone, prostate-specific antigen, and prostatic acid phosphatase levels were assessed. All patients had secondary hypogonadism, as diagnosed by low testosterone levels, and 4 of 10 patients had hyperprolactinemia. After 1 yr of treatment with octreotide (0.3-0.6 mg/day), ultrasound scan and hormone parameters were repeated. The 4 hyperprolactinemic acromegalics were treated with octreotide and cabergoline (1-2 mg/week) to suppress PRL levels. Symptoms due to prostatic, seminal vesicle, and/or urethral disorders or obstruction were experienced by neither acromegalics nor controls. Digital rectal examination revealed no occurrence of prostatic nodules or other abnormalities. Compared to healthy subjects, a remarkable increase in transversal prostatic diameter and volume was observed in acromegalics. In healthy subjects, prostate volume ranged from 15.1-21.8 mL, whereas in acromegalics it ranged from 21.8-41.8 mL. Similarly, an increased median lobe was observed. In fact, the transitional zone diameter was just detectable in 5 of 10 controls, whereas it was measurable in all acromegalics (18 +/- 1.2 vs. 2.8 +/- 0.3 mm; P < 0.001). The prevalence of periurethral calcifications was more than doubled in acromegalics (50%) compared to that in controls (20%). Treatment with octreotide for 1 yr produced normalization of circulating GH and IGF-I levels in 7 of 10 patients. In these 7 patients, ultrasound evaluation showed a significant reduction of the antero-posterior diameter (26.1 +/- 1 vs. 28.9 +/- 1.6 mm; P < 0.01), the transversal diameter (44.9 +/- 2 vs. 48 +/- 2 mm; P < 0.01), and the cranio-caudal diameter (36.5 +/- 1 vs. 41.3 +/- 1.5 mm; P < 0.001), whereas the transitional zone diameter was unchanged (16.4 +/- 1.5 vs. 17.4 +/- 1.7 mm). As a consequence, a significant decrease in prostate volume was recorded (22.1 +/- 1.1 vs. 29.8 +/- 2.5 mL; P < 0.001). Prostate volume increased in 2 of the 3 patients who did not achieve normalization of GH and IGF-I after octreotide treatment. Finally, after treatment, serum testosterone levels were significantly increased (from 1.5 +/- 0.3 to 3.5 +/- 0.3 microg/L), whereas dihydrotestosterone, dehydroepiandrosterone sulfate, delta4-androstenedione, 17beta-estradiol, prostate-specific antigen, and prostatic acid phosphatase were unchanged. Serum PRL levels were suppressed after cabergoline treatment in all 4 hyperprolactinemic patients throughout the study period. In conclusion, prostate enlargement occurs in young acromegalics with a higher than expected prevalence of micro- and macrocalcifications. This suggests that a careful prostate screening should be included in the work-up and follow-up of acromegalic males.
Content may be subject to copyright.
Prostatic Hyperplasia: An Unknown Feature
of Acromegaly
ANNAMARIA COLAO, PAOLO MARZULLO, DIEGO FERONE, STEFANO SPIEZIA,
GAETANA CERBONE, VALERIA MARINO
`, ANTONELLA DI SARNO,
BARTOLOMEO MEROLA, AND GAETANO LOMBARDI
Department of Clinical and Molecular Endocrinology and Oncology, University Federico II (A.C., P.M.,
D.F., G.C., V.M., A.D.S., B.M., G.L.), and Emergency Unit, Ospedale Incurabili (S.S.), 80131 Naples,
Italy
ABSTRACT
This study was designed to investigate whether GH and insulin-
like growth factor I (IGF-I) excess could lead to the development of
benign prostatic hyperplasia and/or prostatic carcinoma. Prostatic
diameters and volume as well as the occurrence of prostatic diseases
were studied by ultrasonography in 10 untreated acromegalic pa-
tients less than 40 yr of age and 10 age- and body mass index-matched
healthy males. Serum GH, IGF-I, PRL, testosterone, dihydrotestos-
terone, prostate-specific antigen, and prostatic acid phosphatase lev-
els were assessed. All patients had secondary hypogonadism, as di-
agnosed by low testosterone levels, and 4 of 10 patients had
hyperprolactinemia. After 1 yr of treatment with octreotide (0.3–0.6
mg/day), ultrasound scan and hormone parameters were repeated.
The 4 hyperprolactinemic acromegalics were treated with octreotide
and cabergoline (1–2 mg/week) to suppress PRL levels.
Symptoms due to prostatic, seminal vesicle, and/or urethral dis-
orders or obstruction were experienced by neither acromegalics nor
controls. Digital rectal examination revealed no occurrence of pros-
tatic nodules or other abnormalities. Compared to healthy subjects,
a remarkable increase in transversal prostatic diameter and volume
was observed in acromegalics. In healthy subjects, prostate volume
ranged from 15.1–21.8 mL, whereas in acromegalics it ranged from
21.841.8 mL. Similarly, an increased median lobe was observed. In
fact, the transitional zone diameter was just detectable in 5 of 10
controls, whereas it was measurable in all acromegalics (18 61.2 vs.
2.8 60.3 mm; P,0.001). The prevalence of periurethral calcifications
was more than doubled in acromegalics (50%) compared to that in
controls (20%). Treatment with octreotide for 1 yr produced normal-
ization of circulating GH and IGF-I levels in 7 of 10 patients. In these
7 patients, ultrasound evaluation showed a significant reduction of
the antero-posterior diameter (26.1 61vs. 28.9 61.6 mm; P,0.01),
the transversal diameter (44.9 62vs. 48 62 mm; P,0.01), and the
cranio-caudal diameter (36.5 61vs. 41.3 61.5 mm; P,0.001),
whereas the transitional zone diameter was unchanged (16.4 61.5 vs.
17.4 61.7 mm). As a consequence, a significant decrease in prostate
volume was recorded (22.1 61.1 vs. 29.8 62.5 mL; P,0.001).
Prostate volume increased in 2 of the 3 patients who did not achieve
normalization of GH and IGF-I after octreotide treatment. Finally,
after treatment, serum testosterone levels were significantly in-
creased (from 1.5 60.3 to 3.5 60.3
m
g/L), whereas dihydrotestos-
terone, dehydroepiandrosterone sulfate, D
4
-androstenedione, 17
b
-es-
tradiol, prostate-specific antigen, and prostatic acid phosphatase
were unchanged. Serum PRL levels were suppressed after cabergo-
line treatment in all 4 hyperprolactinemic patients throughout the
study period.
In conclusion, prostate enlargement occurs in young acromegalics
with a higher than expected prevalence of micro- and macrocalcifi-
cations. This suggests that a careful prostate screening should be
included in the work-up and follow-up of acromegalic males. (J Clin
Endocrinol Metab 83: 775–779, 1998)
ACROMEGALY, a pituitary disorder caused in most
cases by a GH-secreting adenoma, is characterized by
progressive skeleton abnormalities and visceromegaly,
which lead to a stepwise disfigurement of the patient (1–3).
Whereas thyroid, heart, liver, and bone changes, strictly GH
and insulin-like growth factor I (IGF-I) targeted, have been
widely investigated, no study has been performed until now
to assess the prevalence of prostate hyperplasia in conditions
of chronic excess of GH and IGF-I. In humans, prostate en-
largement starts approximately at the age of 40 yr and rises
from 23% to 88% by the ninth decade. Prostate cancer rep-
resents one of the most common malignancies in adult men
(4).
This study was designed to investigate whether GH and
IGF-I excess could lead to the development of benign pros-
tatic hyperplasia and/or prostatic carcinoma in acromegalic
patients. Prostatic diameters and volume as well as the oc-
currence of prostatic diseases were studied by ultrasonog-
raphy in 10 untreated acromegalic patients less than 40 yr of
age. Serum dihydrotestosterone (DHT) and prostate-specific
antigen (PSA) levels were assessed to evaluate the partici-
pation of this regulatory factors in prostatic growth. After 1
yr of treatment with octreotide (OCT), ultrasound scan and
hormone measurements were repeated to evaluate volume
changes after suppression of GH/IGF-I levels had been
achieved.
Subjects and Methods
Patients
Ten acromegalic patients, less than 40 yr of age (range, 26–39 yr),
and 10 age- and body mass index-matched healthy males entered this
study after their informed consent had been obtained. Acromegaly
was diagnosed on the basis of clinical features, elevated GH serum
levels (36.9 66.9
m
g/L) not suppressible below 2
m
g/L by oral
glucose administration, and elevated IGF-I plasma levels (682.4 6
67.4
m
g/L). Four patients presented with coexistent hyperprolactine-
mia (from 55.5–250
m
g/L; Table 1), whereas all patients suffered from
Received September 10, 1997. Revision received November 21, 1997.
Accepted December 2, 1997.
Address all correspondence and requests for reprints to: Annamaria
Colao, M.D., Ph.D., Department of Clinical and Molecular Endocrinol-
ogy and Oncology, University Federico II, Via S. Pansini 5, 80131 Naples,
Italy.
0021-972X/98/$03.00/0 Vol. 83, No. 3
Journal of Clinical Endocrinology and Metabolism Printed in U.S.A.
Copyright © 1998 by The Endocrine Society
775
hypogonadism and showed reduced serum concentrations of FSH,
LH (data not shown), and testosterone (1.5 60.3
m
g/L; Table 1).
Computed tomography and/or magnetic resonance imaging docu-
mented the presence of macroadenoma in 8 patients and microad-
enoma in 2 patients. At study entry, 4 acromegalic patients and 4
control subjects were smokers; none of the study subjects was a high
alcohol consumer, and all had normal diet intake. None of the patients
who were included in the study had previously received any andro-
gen replacement therapy. The patients’ profiles at study entry are
shown in Tables 1 and 2.
Study design
Circulating GH, IGF-I, PRL, FSH, LH, 17
b
-estradiol, testosterone,
DHT, D
4
-androstenedione (D
4
), dehydroepiandrosterone sulfate
(DHEA-S), PSA, and prostatic acid phosphatase (PAP) were assessed at
least twice at study entry and quarterly during treatment with OCT.
Ultrasound examination was performed at study entry and after treat-
ment with OCT. All patients were treated with OCT (Sandostatina,
Novartis, Milan, Italy) for 1 yr. OCT was initially administered at a daily
dose of 0.15 mg in six patients and 0.3 mg in four patients, according to
patients’ compliance during the acute test (0.1 mg, sc), as previously
reported (5). Subsequently, the dose of 0.3 mg/day was maintained
throughout the follow-up in six patients, whereas it was increased up
to 0.6 mg daily in four patients to obtain GH/IGF-I suppression, im-
provement of clinical signs and symptoms and/or tumor shrinkage. In
the four hyperprolactinemic acromegalics, a combined treatment with
OCT plus cabergoline (Dostinex, Pharmacia and Upjohn, Milan, Italy) at
a dose of 1–2 mg/week was given to suppress serum PRL levels. At
study entry, plasma IGF-I levels were assayed twice in a single sample,
whereas serum GH was calculated as the mean of a 6-h blood sampling
(0800–1400 h, 30-min sampling). During treatment, the final GH level
was calculated as the average value from at least three blood samples
collected at 15-min intervals 2 h after OCT administration. At this time
point, plasma IGF-I concentrations were assayed as single sampling.
Hormone normalization after OCT treatment was considered when
basal and oral glucose tolerance test-suppressed GH values were below
TABLE 2. Ultrasonographic evaluation of prostate parameters in acromegalics and controls at study entry
Subject no.,
age (yr)
Body mass
index
Antero-posterior
diameter (mm)
Transversal
diameter (mm)
Cranio-caudal
diameter (mm)
Transitional
zone (mm) Vol (mL) Ultrasonographic and
power doppler findings
Acromegalic patients
1. 26 25.3 24 51.5 36.4 15 23.4 Single 8-mm MC in RL
2. 28 24.8 31.4 42.1 39.8 20 27.3 Periurethral mC
3. 29 26.3 23.1 43 42.3 19 21.8 Periurethral mC
4. 30 25.5 24.6 52.8 41.4 12 27.9 Normal
5. 31 23.9 32.2 41.5 39.4 19 27.3 Normal
6. 32 24.7 28 42 36.2 14 22.1 Periurethral diffused mC
7. 35 27.0 32.8 40.1 47.8 23.1 32.7 Periurethral MC
8. 37 26.6 34.9 53.6 43 22.2 41.8 Normal
9. 38 27.5 32.1 47.2 41.9 20.5 33 Normal
10. 39 29.4 26 49.2 42.1 15.3 28 Single uthricular cyst
Mean 6SEM 26.1 60.5 28.9 61.3 46.3 61.6
a
41 61.1 18 61.2
a
28.5 61.9
a
Healthy subjects
1. 26 23.3 24.1 34.2 35.4 ND 15.1 Normal
2. 28 25.1 28.1 36.1 41.1 3 21.6 Normal
3. 28 25.7 25.2 40.4 39.1 ND 20.7 Periurethral mC
4. 31 24.2 28.1 34.6 34.9 2 17.6 Normal
5. 31 22.1 28.2 38.9 38.2 ND 21.8 Normal
6. 32 25.1 25.4 36.4 43.9 4 19.2 Normal
7. 35 26.4 24.8 39.1 37.3 3 18.8 Normal
8. 36 27.1 25.6 34.9 40.9 3 19 mC at LL
9. 38 27.3 27.4 35.3 38.1 ND 19.1 Normal
10. 39 27.9 26.1 35 39.4 2 18.7 Normal
Mean 6SEM 25.4 60.6 26.3 60.5 36.5 60.7 38.8 60.9 2.8 60.3 18.2 60.6
MC, Macrocalcification; mC, microcalcification; RL, right lobe; LL, left lobe.
a
P,0.001 vs. controls.
TABLE 1. Hormone profile of the 10 patients at study entry
Patient no.,
age (yr)
Disease
duration
(yr)
Serum GH
(
m
g/L)
Plasma IGF-I
(
m
g/L)
Serum PRL
(
m
g/L)
Serum T
(
m
g/L)
Serum DHT
(nmol/L)
Serum PSA
(
m
g/L)
Serum PAP
(IU/L)
1. 26 6 30 850 12.2 2.2 1.0 1.8 0.8
2. 28 5 65 1120 55.5 1.1 1.0 2.1 1.1
3. 29 7 44 600 250 0.9 0.3 3.5 1.2
4. 30 4 16.5 670 4.4 1.8 0.8 4.8 0.1
5. 31 10 71.2 864 95.2 1.9 0.9 4.1 1.6
6. 32 15 18.9 600 84 0.1 0.1 0.9 1.8
7. 35 15 55 625 10.1 0.1 0.2 1.2 1.6
8. 37 7 16.7 725 3.5 2.1 0.8 3.1 1.5
9. 38 15 9.5 600 10 3.7 0.8 2.0 1.5
10. 39 10 42 570 12.1 1.3 1.0 2.2 1.6
Mean 6SEM 9.4 61.4 36.9 66.9 722.4 655.3 53.7 624.4 1.5 60.3 0.7 60.1 2.6 60.4 1.3 60.2
Normal ranges: GH, less than 5
m
g/L; IGF-I, 110 –502 and 100– 494
m
g/L for patients aged 20 –30 and 31–40 yr, respectively; PRL, 5–15
m
g/L;
testosterone, 3.5–9
m
g/L; DHT, 0.4–1.6 nmol/L; PAP, 0–2.6 U/L; PSA, 0–10
m
g/L.
776 COLAO ET AL. JCE&M1998
Vol 83 No 3
5 and 2
m
g/L, respectively, and IGF-I values were within the normal
ranges.
Hormonal assessment
GH, PRL, testosterone, and 17
b
-estradiol were measured by RIA;
IGF-I, FSH, LH, PSA, and DHT were determined by immunoradiometric
assay; PAP was measured by autoanalyzer. The normal ranges were:
GH, 0–5
m
g/L; IGF-I, 110–502 and 100494
m
g/L, respectively for
patients aged 20–30 and 31–40 yr; PRL, 5–15
m
g/L; FSH and LH, 5–18
mU/mL; testosterone, 3.5–9
m
g/L; DHT, 0.4–1.6 nmol/L; D
4
, 1–3.5
m
g/L; DHEA-S, 60–560
m
g/L; 17
b
-estradiol, 20–70
m
g/L; PAP, 0–2.6
U/L; and PSA, 0–10
m
g/L. All assessments were age adjusted.
Ultrasound examination
All of the patients received a preliminary enema with 120 mL sodium
acid phosphate (Clismalax, Sofar, Milan, Italy) not later than 1 h before
the examination to favor rectal cleanliness. Before ultrasonography,
patients underwent a preliminary digital rectal exploration. Prostate
ultrasonography was carried out with ATL Apogee 800 (Advanced
Technology Laboratories, Bothell, WA) by means of a 9.0-megahertz
end-fire transrectal transducer (2-cm external diameter) and a Power
Echo Color Doppler Advanced Technology module that displays the
total integrated Doppler power in color, to obtain angiographic micro-
maps (6). The transducer, preliminarily covered with ultrasound trans-
mission gel (Acquasonic, Parker Laboratory, Newark, NJ) and a dis-
posable rubber sheat, was lubricated and gradually inserted about 3 cm
into the rectum, then directed toward the anterior rectal wall. The fol-
lowing prostate diameters and features were evaluated in B-mode: an-
tero-posterior, transversal, cranio-caudal, and that including the tran-
sitional zone (TZD); morphology of gland boundary; occurrence of
microcalcifications (#3 mm) and/or macrocalcifications (.3 mm); de-
tection and sizing of intraprostatic nodules; evaluation of seminal ves-
icles; and occurrence of local inflammatory events. Reconstruction by a
standard ellipsoid formula (0.52 3length 3height 3width) allowed the
measurement of total prostate volume. All scans were performed by a
single examiner (S.S.). In agreement with previous findings (7), normal
prostate volume was considered as less than 30 mL.
Statistical analysis
Data are expressed as the mean 6sem. ANOVA, Student’s ttest for
paired data, and linear correlation analysis were applied where appro-
priate. Statistical significance was set at 5%.
Results
Patients with acromegaly showed significantly decreased
serum testosterone levels (Table 1), whereas FSH, LH,
DHEA-S, 17
b
-estradiol (data not shown), DHT, PSA, and
PAP (Table 1) were in the normal range. All patients had
secondary hypogonadism, as diagnosed by low testosterone
levels, and 4 of 10 patients had hyperprolactinemia (no. 2, 3,
5 and 6; Table 1).
Ultrasonographic findings at study entry
Symptoms due to prostatic, seminal vesicle, and/or ure-
thral disorders or obstruction were not experienced by either
acromegalics or controls. Digital rectal examination revealed
no occurrence of prostatic nodules or other abnormalities.
Compared to healthy subjects, a remarkable increase in
transversal diameter and volume of the prostate gland was
observed in acromegalics (Table 2). In healthy subjects, pros-
tate volume ranged from 15.1–21.8 mL, whereas in acrome-
galics, it ranged from 21.8 41.8 mL. In 3 patients (no. 7–9;
Table 2), prostate volume was greater than 30 mL, which was
considered a normal threshold value (7). Similarly, an in-
creased median lobe was observed. In fact, TZD was just
detectable in 5 of 10 controls, whereas it was measurable in
all acromegalics (18 61.2 vs. 2.8 60.3 mm; P,0.001). The
prevalence of periurethral calcifications was more than dou-
bled in acromegalics (50%) compared to that in controls (20%;
Table 2). Among the 10 acromegalics, calcifications in the
periurethral zone were detected in 4 patients (no. 2, 3, 6, and
7), and calcifications within the lobes were found in 1 patient
(no. 1), whereas in another patient (no. 10) a diffuse hypere-
chogenity of prostatic tissue with a single uthricolar cyst was
detected (Table 2). No sign of vesicle inflammation was
shown. No significant difference in prostatic volume (24.6 6
1.5 vs. 31.1 62.6 mL) or serum testosterone levels (1 60.4
vs. 1.9 60.5
m
g/L) was found between hyperprolactinemic
and normoprolactinemic acromegalics. A significant corre-
lation was found only between prostate volume and patient
age (Table 3).
Effect of long term OCT treatment on prostate parameters
and hormone levels
Treatment with OCT for 1 yr induced normalization of
circulating GH and IGF-I levels in 7 of 10 patients (Fig. 1). In
these 7 patients, ultrasound evaluation showed a significant
reduction of antero-posterior diameter (26.1 61vs. 28.9 61.6
mm; P,0.01), transversal diameter (44.9 62vs. 48 62 mm;
P,0.01), and cranio-caudal diameter (36.5 61vs. 41.3 61.5
mm; P,0.001), whereas TZD was unchanged (16.4 61.5 vs.
17.4 61.7 mm). As a consequence, a significant decrease in
prostate volume was recorded (22.1 61.1 vs. 29.8 62.5 mL;
P,0.001). The individual data of the 10 patients are shown
in Fig. 1. Prostate volume increased in 2 (no. 3, from 21.8 to
29.9 mL; no. 5, from 27.3 to 33.5 mL; Tables 1 and 2) of the
3 patients who did not achieve normalization of GH and
TABLE 3. Results of the linear correlation analysis between
prostate volume and clinical and hormone parameters in
acromegalic patients
rP
Age 0.6 0.04
Disease duration 0.09 0.8
Serum GH ;0.2 0.5
Plasma IGF-I ;0.054 0.8
Serum PRL ;0.5 0.09
Serum testosterone 0.3 0.3
Serum DHT 0.14 0.7
FIG. 1. Serum GH profile (left) and prostate volume measured by
ultrasonography (right) in the 10 acromegalics before and after 1 yr
of octreotide treatment.
PROSTATIC DISORDERS IN ACROMEGALY 777
IGF-I after OCT treatment (Fig. 1). In the remaining patient
(no. 2), prostate volume decreased from 27.3 to 20.5 mL
despite evidence that GH decreased from 65 to 11.3
m
g/L, but
was not normalized. In 1 patient (no. 4) of the 7 who nor-
malized GH and IGF-I levels after OCT treatment, a hypo-
echoic nodular zone was detected within the left lobe without
a distinct boundary and with an irregular intralesional echoic
pattern (Fig. 2). The Power Echo Color Doppler evaluation
revealed high intra- and perilesional vascular flow. Fine nee-
dle biopsy revealed nodular hyperplasia. After OCT treat-
ment, calcifications were still detected in the periurethral
zone in 3 patients (no. 2, 5, and 6), whereas in 1 patient they
disappeared. Calcifications within the lobes were visualized
in 2 other patients (no. 4 and 8). In 2 patients, 1 presenting
with single macrocalcification within the right lobe (no. 1)
and 1 with uthricular cyst before therapy (no. 10), the ex-
amination performed 1 yr after OCT treatment showed mi-
crocalcifications in the former and no further detection of the
cyst in the latter patient. Lastly, after OCT treatment, serum
testosterone levels were significantly increased (from 1.5 6
0.3 to 3.5 60.3
m
g/L), whereas DHT, DHEA-S, D
4
, and
17
b
-estradiol levels were unchanged. In the 4 patients with
hyperprolactinemia, serum PRL levels were suppressed
throughout the study (data not shown).
Discussion
In acromegaly, prolonged hypersecretion of GH and IGF-I
constantly causes enlargement of most internal body organs
(1, 2). Thyroid, heart, liver, and bone seem to be strictly GH
and IGF-I targeted, but recent reports suggest a proliferating
effect of GH/IGF-I on colonic mucosa, as demonstrated by
an increased prevalence of colonic polyps detected at pan-
colonoscopy (8, 9). To date, no study has been performed to
assess the prevalence of benign prostate hyperplasia and/or
prostate cancer in acromegaly. In humans, prostate enlarge-
ment seems to begin at the age of 40 yr (10) and develops
almost exclusively in the transitional and periurethral zones
(11). Although evidence for direct or indirect regulatory ef-
fects of androgens on prostatic cell growth and differentia-
tion have been provided (10, 12), their action alone appears
to be insufficient to explain prostatic diseases. Acromegalic
patients can be considered as a peculiar study model, as they
often present low testosterone levels. In the current study,
prostatic enlargement, particularly that affecting the median
lobe, was recorded in all patients despite the evident hy-
pogonadism. In addition, an increased incidence of calcifi-
cations, suggestive of regressive events within the gland, was
observed in the patients (50%) compared to that in healthy
subjects (20%). To prevent volume changes associated with
aging, this study was performed in patients under 40 yr of
age, which is supposed to be the age limit for developing
benign prostatic hyperplasia (10). The normal prostate vol-
ume was reported as 30 mL in a normal population of 181
men, aged 40–79 yr. (7). No data are available in normal men
less than 40 yr of age. Taking into account the threshold of
30 mL, 3 of 10 acromegalic patients had clear-cut prostate
hypertrophy. However, in our series of healthy subjects,
prostate volume ranged from 15.1–21.8 mL, with a mean
value of 18.2 60.6 mL. In comparison with that in age-
matched controls, prostate volume was greater than ex-
pected in all patients but 1. Thus, it could be argued that the
prostate is a primary target tissue of GH and IGF-I. In none
of the patients were PSA levels, digital rectal exploration, or
transrectal ultrasound (TRUS) able to detect the occurrence
of prostatic cancer. This could be due both to the relatively
young age of the patients and to a protective effect that
reduced levels of androgens might exert. In fact, testosterone
acts on prostate growth directly and indirectly, through the
conversion to DHT by 5
a
-reductase (12). A physiological
decrease in testosterone in the elderly seems to be followed
by accumulation of DHT within the gland, because of re-
duced catabolism and enhanced intracellular binding (12).
Therefore, different results of prostatic ultrasound evalua-
tion in older patients could not be excluded. Although the
involvement of several growth factors, such as PRL, epider-
mal growth factor, fibroblast growth factor, and transform-
ing growth factor-
a
and -
b
, has been demonstrated both in
vitro and in vivo (13, 14), little is known about the direct
involvement of GH in prostate development. However, GH
receptors have been demonstrated in the rat prostate (15),
and increased messenger ribonucleic acid transcription for
androgen receptors has been detected after GH and PRL
administration in immature rat prostate (16).
The possibility that prostatic enlargement was actually
due to the chronic excess of GH and IGF-I was supported by
the significant decrease in prostate volume obtained after 1
yr of treatment with OCT in all patients who achieved GH/
IGF-I suppression. As further support, in two of three acro-
megalics who did not achieve hormone suppression after
OCT treatment, prostate volume was increased. As far as the
prevalence of micro- and macrocalcifications was concerned,
the 1-yr treatment with OCT led to the disappearance of
microcalcifications and uthricular cyst in two patients, but
caused the occurrence of microcalcifications in two other
patients. On the basis of the detection of somatostatin re-
ceptors, primarily subtypes 1 and 2, in stromal cells of benign
and malignant prostate (17–19), it is arguable that chronic
OCT administration could regulate the GH/IGFs/IGF-bind-
ing protein paracrine-autocrine pathways within the gland.
OCT could act on prostate size with different mechanisms.
First, it can induce a decrease in prostate dimension by a
direct antiproliferative effect (20) and indirectly by suppress-
ing circulating levels of GH/IGF-I. Second, it may prevent
FIG. 2. Prostate ultrasonographic imaging in patient 4 before (left)
and after (right) 1 yr of octreotide treatment at a dose of 0.3 mg/day.
On the left, the transversal diameter measured at study entry is
shown without any distinct nodular lesion. On the right, a clear-cut
nodule (0.97 31.03 cm) was found after treatment. At cytology per-
formed on a specimen collected by fine needle biopsy, the nodule was
diagnosed as simple hyperplasia.
778 COLAO ET AL. JCE&M1998
Vol 83 No 3
prostate enlargement by inducing apoptotic processes of the
mesenchymal tissue and by modifying the hemodynamics of
local blood circulation (21). The positive effect of OCT treat-
ment on prostate volume and morphology was observed
despite a significant increase in testosterone levels and a
significant improvement of spermatogenic activity (data not
shown).
In conclusion, prostate enlargement occurs in young ac-
romegalics with a higher than expected prevalence of micro-
and macrocalcifications. These findings seem to be related to
the GH/IGF-I excess, as they occur in the presence of evident
hypogonadism. This suggests that a careful prostate screen-
ing, supported by transrectal ultrasound evaluation, should
be included in the work-up of acromegalic males. Androgen
replacement should be carefully monitored to avoid adding
to prostate growth. Long term treatment with OCT can re-
verse prostate enlargement. The occurrence of micro- and
macrocalcifications and even prostate nodules during OCT
treatment indicates that monitoring of prostate size is also
advisable in the follow-up of acromegalic patients.
References
1. Nabarro JDN. 1987 Acromegaly. Clin Endocrinol (Oxf). 26:481–512.
2. Melmed S. 1990 Acromegaly. N Engl J Med. 322:966–977.
3. Colao A, Merola B, Ferone D, Lombardi G. 1997 Extensive experience: ac-
romegaly. J Clin Endocrinol Metab. 82:2777–2781.
4. Woolf SH. 1995 Screening for prostate cancer with prostate-specific antigen.
An examination of the evidence. N Engl J Med. 333:1401–1405.
5. Colao A, Ferone D, Lastoria S, et al. 1996 Prediction of efficacy of octreotide
therapy in patients with acromegaly. J Clin Endocrinol Metab. 81:2356–2362.
6. Rubin JM, Bude RO, Carson PL, Bree RL, Adler RS. 1994 Power Doppler US:
a potentially useful alternative to mean frequency-based Color Doppler US.
Radiology. 190:853–856.
7. Collins GN, Raab GM, Hehir M, King B, Garraway WM. 1995 Reproduc-
ibility and observer variability of transrectal ultrasound measurements of
prostatic volume. Ultrasound Med Biol. 21:1101–1105.
8. Ezzat S, Carey S, Melmed S. 1991 Colon polyps in acromegaly. J Clin Endo-
crinol Metab. 72:245–249.
9. Colao A, Balzano A, Ferone D, et al. 1997 Increased prevalence of colonic
polyps and altered lymphocyte subset pattern in the colonic lamina propria in
acromegaly. Clin Endocrinol (Oxf). 47:23–28.
10. Wilson JD. 1980 The pathogenesis of benign prostatic hyperplasia. Am J Med.
68:745–756.
11. McNeal JE. 1988 Normal histology of the prostate. Am J Surg Pathol.
12:619–633.
12. Cuhna GR, Donjacour AA, Cooke PS, et al. 1987 The endocrinology and
developmental biology of the prostate. Endocr Rev. 8:338–362.
13. Guenette RS, Tenniswood M. 1994 The role of growth factors in the sup-
pression of active cell death in the prostate: an hypothesis. Biochem Cell Biol.
71:553–559.
14. Byrne RL, Leung H, Neal DE. 1996 Peptide growth factors in the prostate as
mediators of stromal epithelial interactions. Br J Urol. 77:627–633.
15. Reiter E, Kecha O, Hennuy B, et al. 1995 Growth hormone directly affects the
function of the different lobes of the rat prostate. Endocrinology.
136:3338–3345.
16. Reiter E, Bonnet B, Sente B, et al. 1992 Growth hormone and prolactin
stimulate androgen receptor, insulin-like growth factor-I (IGF-I) and IGF-I
receptor levels in the prostate of immature rats. Mol Cell Endocrinol. 88:77– 87.
17. Reubi JC, Waser B, Schaer JC, Markwalder R. 1995 Somatostatin receptors in
human prostate and prostate cancer. J Clin Endocrinol Metab. 80:2806–2814.
18. Tatoud R, Degeorges A, Prevost G, et al. 1995 Somatostatin receptor in
prostate tissue and derived cell cultures, and the in vitro growth inhibitory
effect of BIM-23014 analog. Mol Cell Endocrinol. 113:195–204.
19. Sinisi AA, Bellastella A, Prezioso D, et al. 1997 Different expression of
somatostatin receptor subtypes in cultered epithelial cells from human normal
prostate and prostate cancer. J Clin Endocrinol Metab. 82:2566–2569.
20. Hofland LJ, van Koetsveld PM, Wouters N, Waaijers M, Reubi J-C, Lamberts
SWJ. 1992 Dissociation of antiproliferative and antihormonal effects of the
somatostatin analog octreotide on 7315b pituitary tumor cells. Endocrinology.
131:571–577.
21. Reubi JC, Schaer JC, Laissue JA, Waser B. 1996 Somatostatin receptors and
their subtypes in human tumors and in peritumoral vessels. Metab Clin Exp.
45:39–41.
PROSTATIC DISORDERS IN ACROMEGALY 779
... The role played by the GH/IGF-I axis on prostate growth has also been suggested by our recent observations in acro-megalic and GH-deficient patients. In fact, chronically elevated GH, IGF-I, and IGFBP-3 levels were shown to determine prostate overgrowth and structural changes, such as nodules, cysts, and calcifications, in a large proportion of acromegalic patients (13,14), whereas in long-standing GH deficiency associated with hypogonadism, a decrease in prostate size was found (13,15). Prostate hyperplasia was also found in young acromegalic patients, who were not expected to have age-dependent prostate diseases (13,14). ...
... In fact, chronically elevated GH, IGF-I, and IGFBP-3 levels were shown to determine prostate overgrowth and structural changes, such as nodules, cysts, and calcifications, in a large proportion of acromegalic patients (13,14), whereas in long-standing GH deficiency associated with hypogonadism, a decrease in prostate size was found (13,15). Prostate hyperplasia was also found in young acromegalic patients, who were not expected to have age-dependent prostate diseases (13,14). The evidence that prostate disorders occur in presence of hypogonadism and that prostate size decreases after 1 yr of treatment with octreotide in a small group of young patients (14), further supports the hypothesis that chronic GH/IGF-I excess causes prostate hyperplasia. ...
... Prostate hyperplasia was also found in young acromegalic patients, who were not expected to have age-dependent prostate diseases (13,14). The evidence that prostate disorders occur in presence of hypogonadism and that prostate size decreases after 1 yr of treatment with octreotide in a small group of young patients (14), further supports the hypothesis that chronic GH/IGF-I excess causes prostate hyperplasia. ...
Article
The insulin-like growth factors (IGFs) have mitogenic effects on normal and tumoral prostate epithelial cells and have been suggested to be involved in prostate cancer. Moreover, chronic GH and IGF-I excess causes prostate overgrowth in patients with acromegaly. This study was designed to investigate whether the suppression of GH and IGF-I levels by surgery or pharmacotherapy could induce the regression of prostatic hyperplasia in acromegalic patients. To this end, prostate volume (PV) as well as the occurrence of prostatic diseases were studied by transrectal ultrasonography in 23 untreated acromegalic patients (with elevated GH and IGF levels). None of the patients reported symptoms due to prostatic disorders or obstruction. At study entry, prostate hyperplasia was found in half patients. After 2 yr, GH, IGF-I, and IGFBP-3 levels were decreased, whereas prostate-specific antigen levels did not change. PV was decreased in the 16 patients who were well controlled. Among the 6 patients with prostate hyperplasia at study entry who achieved disease control, 4 regained a normal PV at the end of the 2 yr of treatment, whereas none of the 5 patients with prostate hyperplasia at study entry and not achieving disease control normalized their PV. When patients were divided according to age, prostate volume decreased after 2 yr only in the 8 controlled patients aged below 50 yr, but not in those controlled and with age above 50 yr despite similar decrease in GH, IGF-I, and IGFBP3 levels. No clinical, transrectal ultrasonography, or cytological evidence of prostate cancer was detected during the study period. These data suggest that hyperplasia, but not cancer, is frequent in acromegalic men, and that the GH-IGF axis and age are independently associated with the development of this process.
... ml versus 18.2 ml in healthy subjects of the same age), and 3 patients presented frank prostatic hypertrophy (volume > 30 ml). After treatment with octreotide in association with cabergoline in patients with coexisting hyperprolactinemia, the mean prostate volume showed a significant reduction (22. 1 ml), despite a simultaneous increase in testosterone levels to complete normalization [40]. The authors subsequently extended the evaluation to 46 acromegalic patients (26- 74 years), reporting not only a close correlation between the presence of prostatic hypertrophy and the degree of disease activity, but even that the prostate volume appears to be conditioned, even more than by age, by the duration of the disease. ...
... Therefore, studies capable of detecting its incidence necessarily require extremely long observation times and, moreover, no studies have systematically investigated this category of subjects with targeted invasive evaluations (e.g., prostate-specific antigen and prostate biopsy), so it can be expected that the actual incidence of prostate cancer has been underestimated. Therefore, although there has not been reported a marked increase in the incidence of prostate cancer in the work reported earlier in the text [38,[40][41][42][43], further investigation, with appropriate followup time and tools, is needed before definitive conclusions can be drawn on the risk of prostate cancer in subjects with acromegaly. ...
Article
Full-text available
Acromegaly is a rare pathology characterized by chronic hypersecretion of Growth Hormone (GH) and Insulin-like Growth Factor-1 (IGF-1) that causes somatic, metabolic, and systemic changes. The somatotropic axis acts physiologically favoring gonadal function, but when GH is produced in excess it has deleterious effects on many aspects of male sexuality. It is widely demonstrated, in fact, that acromegaly induces hypogonadism through different mechanisms, both through direct mass effect on gonadotropic cells and through increased plasma levels of prolactin. Moreover, hypogonadism is also one of the factors linking acromegaly to erectile dysfunction (ED), but also metabolic complications of acromegaly and, probably, GH itself contribute to the genesis of this disorder. There are few data in the literature on the impact of the disease on fertility and testicular volume. Finally, knowledge of the role of GH hypersecretion on the occurrence of prostatic diseases such as benign prostatic hypertrophy and prostatic cancer appears to be of fundamental clinical importance in the long-term management of these patients.
... 65 Indeed, BPH is reported in approximately half the acromegalic subjects 65,67 and the prevalence of parenchymal alterations (ie, cysts, calcifications and nodules) is also increased. 65,66,68 Slight increase in PSA levels and International Prostate Symptom Scores (IPSS) are also reported in these patients, although prostatic symptoms are often absent. 67,69 Moreover, as results from a large population study, prostatic cancer (PCa) incidence is increased both in healthy individuals with IGF-I levels in the upper reference range 70,71 and in acromegalic patients. ...
... 64 On the other side, effective pharmacological treatment of acromegaly for 1-2 years via somatostatin analogs results in a reduction of PV in young subjects (<50 years old), but not in older ones, despite a rise in testosterone levels. 66,74 ...
Article
Background: The hypothalamic-pituitary-gonadal (HPG) and hypothalamic-pituitary-somatotropic (HPS) axes are strongly interconnected. Interactions between these axes are complex and poorly understood. These interactions are characterized by redundancies in reciprocal influences at each level of regulation and the combination of endocrine and paracrine effects that change during development. Objectives: To comprehensively review the crosstalk between the HPG and HPS axes and related pathological and clinical aspects during various life stages of male subjects. Materials and methods: A thorough search of publications available in PubMed was performed using proper keywords. Results: Molecular studies confirmed the expressions of growth hormone (GH) and insulin-like growth factor-I (IGF-I) receptors on the HPG axis and reproductive organs, indicating a possible interaction between HPS and HPG axes at various levels. Insulin growth factors participate in sexual differentiation during fetal development, indicating that normal HPS axis activity is required for proper testicular development. IGF-I contributes to correct testicular position during minipuberty, determines linear growth during childhood, and promotes puberty onset and pace through gonadotropin-releasing hormone activation. IGF-I levels are high during transition age, even when linear growth is almost complete, suggesting its role in reproductive tract maturation. Patients with GH deficiency (GHD) and insensitivity (GHI) exhibit delayed puberty and impaired genital development; replacement therapy in such patients induces proper pubertal development. In adults, few studies have suggested that lower IGF-I levels are associated with impaired sperm parameters. Discussion and conclusion: The role of GH-IGF-I in testicular development remains largely unexplored. However, it is important to evaluate gonadic development in children with GHD. Additionally, HPS axis function should be evaluated in children with urogenital malformation or gonadal development alterations. Correct diagnosis and prompt therapeutic intervention are needed for healthy puberty, attainment of complete gonadal development during transition age, and fertility potential in adulthood.
... Chronic exposure to GH and IGF1 hypersecretions leads to soft tissue swelling of the tongue, heart, kidney, colon, and vocal cords and to periarticular and cartilaginous thickening [14]. Increased GH/IGF-1 levels were found to be associated with organ hyperplasias and tumourigenesis such as colon polyps [22], cardiomyopathy [23], goitre [24], thyroid nodules [25], prostate hyperplasia [26], submandibular gland volume increase [27], and myoma uteri [28]. The chronic overexpression of GH in transgenic mice results in systemically and locally increased IGF-1 levels and in adrenal enlargement [8]. ...
Article
Full-text available
Purpose: The aim of this study was to investigate the possible association of insulin-like growth factor-1 (IGF-1) with the pathogenesis of non-functioning adrenocortical adenomas (NFAs). Methods: This study included 50 female patients (mean age 54 years) with NFAs, 55 patients (mean age 48 years; 20 male, 35 female) with acromegaly and 38 female control subjects (mean age 58 years). Body mass index (BMI) and waist circumference (WC) of the subjects were recorded and blood samples for IGF-1 were taken. Insulin resistance was calculated using the homeostatic model assessment (HOMA) score. Since most of the acromegaly patients had been using medicine that could have effected insulin resistance, HOMA scores were calculated only in patients with NFAs and the controls. Computerized tomography or magnetic resonance imaging was taken of the acromegalics and controls to detect adrenal mass frequency. Results: The mean age was similar among the groups. As expected, the serum IGF-1 levels were significantly higher in patients with acromegaly than in patients with NFAs and the controls (p < 0.001). Although BMI, WC, and serum IGF-1 levels were significantly higher (p < 0.001) in patients with NFAs, the HOMA scores were similar between patients with NFAs and control groups. Although none of the control subjects had adrenal masses, NFAs were detected in 14 (25%) out of 55 acromegalic patients. Conclusions: Higher serum IGF-1 levels in patients with NFAs compared to the control group and an increased prevalence of NFAs in acromegaly patients compared to control subjects and the general population suggest an association of IGF-1 with the etiopathogenesis of NFA.
... These elevated levels are as a result of either pituitary tumours, or activating mutations in the GHRH signalling cascade, which result in hypersecretion of GH by pituitary somatotroph cells (Chhabra et al., 2011). The condition is associated with an increased risk of colon (Ezzat et al., 1991), breast (Cheung and Boyages, 1997), and prostate cancer (Colao et al., 1998;le Roux et al., 2000). Given that IGF1 has been shown to play a role in these cancers, it was therefore initially thought that the tumourigenic effects of GH were indirectly mediated by IGF1 rather than directly by GH. ...
Thesis
Full-text available
Squamous-cell carcinoma of the oesophagus is a formidable disease which poses a significant health risk in developing countries where incidence is high and survival is low. Investigating the poorly understood mechanisms involved in oesophageal tumourigenesis may provide a platform to develop improved diagnostic techniques and therapies. The growth hormone (GH) signalling axis is important for proper cellular and organ system function. The axis has been shown to play a role in a number of cancers. Additionally, GH has shown to induce neoplasms in vivo while forced autocrine signalling of GH was shown to promote carcinogenesis in several in vitro models, highlighting the role of GH in tumourigenesis. Several studies showed that the tumourigenic effects of GH are largely mediated by its receptor, growth hormone receptor (GHR). The aim of this project is to therefore identify the role GHR in oesophageal squamous-cell carcinoma (OSCC) using an in vitro model. mRNA analysis showed expression of endogenous GH mRNA in a majority of OSCC cell lines tested, while nearly all expressed some form of GHR. Few, however, expressed mRNA for the more-active isoform of GHR, d3GHR, indicating GH may play a role in OSCC, and may be mediated by full-length GHR (flGHR) in a majority of cell lines. Western blotting for GHR remained largely unsuccessful, indicative of the short half-life of mature GHR and the possible insensitivity of commercially-available GHR antibodies. However, unglycosylated, immature GHR is likely detected as a 95kDa band, while mature GHR may be detected as a 120kDa band. However, a non-specific 95kDa band was also detected, thereby interfering with detection of true levels of premature GHR in OSCC cell lines. GHR knockdown by small interfering RNA indicated that the GH axis may play a role in proliferation in a subset of patients that is mediated by GHR, but does not affect responses to the chemotherapeutic agents doxorubicin and cisplatin. Additionally, the mRNA expression of GHR did not correlate with the expression of functional GHR protein in a number of OSCC cell lines. These results together indicate while GHR may play a role in a subset of patients, the role is limited to proliferation when only investigating the effects of GH on proliferation and chemoresistance. However, investigating additional markers of tumourigenesis is needed to further elucidate the role of GH/GHR signalling in OSCC. Additionally, the potential tumourigenic effects of GH mediated by prolactin receptor (PRLR) may not be ignored.
... Furthermore, DISC1 mouse models display synaptic pathologies ( Lee et al. 2011) and show cognitive deficits reflecting those found in schizophrenia, such as impaired working memory ( Koike et al. 2006;Kvajo et al. 2008). Additionally, it has been shown that Cav-1 KO mice present with behavioral deficits similar to those seen in many schizo- phrenia-like symptoms such as altered motor function and altered emotion, as well as memory deficits ( Colao et al. 1998;Gioiosa et al. 2008;Head et al. 2008;Trushina et al. 2006). Because there has been much interest in understanding the neurobiology of schizophrenia over the past decade (Harrison and Weinberger 2005;Owen et al. 2005;Sawa and Snyder 2002), in the present study we investigated a potential interac- tion between DISC1 and Cav-1 on synapse biology. ...
Article
Background: Schizophrenia is a debilitating psychiatric disorder manifested in early adulthood. Disrupted-In-Schizophrenia-1 (DISC1) is a susceptible gene for schizophrenia (54, 76, 101) implicated in neuronal development, brain maturation and neuroplasticity (12, 20). Therefore, DISC1 is a promising candidate gene for schizophrenia, but the molecular mechanisms underlying its role in the pathogenesis of the disease are still poorly understood. Interestingly, Caveolin-1 (Cav-1), a cholesterol binding and scaffolding protein, regulates neuronal signal transduction and promotes neuroplasticity. Here we examined the role of Cav-1 in mediating DISC1 expression in neurons in vitro and the hippocampus in vivo. Methods and results: Overexpressing Cav-1 specifically in neurons using a neuron specific synapsin promoter (SynCav1) increased expression of DISC1 and proteins involved in synaptic plasticity (PSD95, synaptobrevin, synaptophysin, neurexin, and syntaxin-1). Similarly, SynCav1-transfected differentiated human neurons derived from induced pluripotent stem cells (hiPSCs) exhibited increased expression of DISC1 and markers of synaptic plasticity. Conversely, hippocampi from Cav-1 knockout (KO) exhibited decreased expression of DISC1 and proteins involved in synaptic plasticity. Finally, SynCav1 delivery to the hippocampus of Cav-1 KO mice and Cav-1 KO neurons in culture restored expression of DISC1 and markers of synaptic plasticity. Furthermore, we found that Cav-1 co-immunoprecipitated with DISC1 in brain tissues. Conclusion: These findings suggest an important role by which neuronal Cav-1 regulates DISC1 neurobiology with implications for synaptic plasticity. Therefore, SynCav1 might be a potential therapeutic target for restoring neuronal function in schizophrenia.
Article
Full-text available
Prostate cancer (PCa) is the most common malignancy in men worldwide, thus developing effective prevention strategies remain a critical challenge. Insulin-like growth factor 1 (IGF-1) is produced mainly in the liver by growth hormone signaling and is necessary for normal physical growth. However, several studies have shown an association between increased levels of circulating IGF-1 and the risk of developing solid malignancies, including PCa. Because the IGF-1 receptor is overexpressed in PCa, IGF-1 can accelerate PCa growth by activating phosphoinositide 3-kinase and mitogen-activated protein kinase, or increasing sex hormone sensitivity. Short-chain fatty acids (SCFAs) are beneficial gut microbial metabolites, mainly because of their anti-inflammatory effects. However, we have demonstrated that gut microbiota-derived SCFAs increase the production of IGF-1 in the liver and prostate. This promotes the progression of PCa by the activation of IGF-1 receptor downstream signaling. In addition, the relative abundance of SCFA-producing bacteria, such as Alistipes, are increased in gut microbiomes of patients with high-grade PCa. IGF-1 production is therefore affected by the gut microbiome, dietary habits, and genetic background, and may play a central role in prostate carcinogenesis. The pro-tumor effects of bacteria and diet-derived metabolites might be potentially countered through dietary regimens and supplements. The specific diets or supplements that are effective are unclear. Further research into the “Gut–IGF-1–Prostate Axis” may help discover optimal diets and nutritional supplements that could be implemented for prevention of PCa.
Article
Se l’acromegalia sia associata, o meno, ad un aumentato rischio di neoplasia rimane un problema assai dibattuto. La mancanza di univocità della letteratura è riconducibile a diversi motivi, tra i quali il più importante è verosimilmente l’effetto della mortalità competitiva per cause cardiovascolari che può aver portato ad una sottostima del rischio di neoplasia in molti studi. È verosimile che l’allungamento della vita media dei pazienti acromegalici possa produrre un netto aumento nell’incidenza di neoplasie così come accade nella popolazione generale. La neoplasia del colon è quella maggiormente studiata ed è la neoplasia per la quale esistono le più convincenti evidenze di un’aumentata frequenza nell’acromegalia. Gli studi che hanno impiegato come metodica di screening la colonscopia su casistiche numerose dimostrano che l’acromegalia si associa ad un moderato, ma significativo, aumento di rischio di neoplasia del colon. Pertanto, è indicato procedere a screening colonscopico in tutti i pazienti acromegalicici che non presentino controindicazioni alla procedura. È anche indicato procedere alla ripetizione della colonscopia nei pazienti con neoplasia benigna o maligna al primo esame, soprattutto se la malattia rimane attiva.
Article
Acromegaly is a rare disease associated with an increased risk of prostate enlargement. Severe prostate enlargement with severe lower urinary tract symptoms (LUTS) in an acromegalic patient is even more uncommon. Herein we report on a 55-year-old man who was diagnosed with acromegaly and prostate enlargement at 40 years of age. Transsphenoidal surgery, postoperative radiotherapy, and octreotide medical therapy failed to control the acromegaly, and growth hormone (GH) and insulin-like growth factor 1 (IGF-1) levels remained elevated. When the patient was 53 years of age, severe LUTS and prostate enlargement (prostate volume = 128 mL) were noted. However, LUTS improved and prostate volume decreased markedly after 5α-reductase inhibitors were used, despite the poorly controlled acromegaly (elevated GH and IGF-1 levels). This is the first long-term observation of LUTS and prostate enlargement in a poorly controlled acromegalic patient. Although the GH–IGF-1 axis was a factor contributing to prostate enlargement, the present case suggests that androgens may still play an essential role in prostate enlargement and symptoms in active acromegalic patients >50 years of age. Indeed, we should be aware that suppressing the GH–IGF-1 axis is not the only treatment choice for prostate enlargement in acromegalic patients, and even in poorly controlled acromegalic patients in whom suppression of the GH–IGF-1 axis is difficult. Symptomatic prostate enlargement in cases of active acromegaly can be treated with 5α-reductase inhibitors, as in general benign prostate hyperplasia populations.
Article
The objective of the present review was to consider the available data on the risk of development of prostatic hyperplasia and prostate cancer in patients presenting with acromegalia. It is shown that the incidence of prostatic diseases in acromegalic patients is significantly higher than in the general population. Prostatic hyperplasia occurs in patients of all age groups presenting with acromegalia while its treatment reduces the volume of the prostate gland. The reviewed publications do not report cases of prostate cancer.
Article
Full-text available
Regression of the rat ventral prostate occurs when the level of 5 alpha-dihydrotestosterone, the trophic hormone, drops below the threshold required to suppress apoptosis. The induction of apoptosis in the ventral prostate is accompanied by the increase in the steady-state level of a number of mRNAs coding for proteins that are involved in the latter stages of apoptosis and thus represent secondary thanatogens. These include proteases (cathepsins, plasminogen activators, and collagenase), clusterin, poly(ADP)ribose polymerase, tenascin, and several unidentified genes, as well as several RNases and the classical Ca2+,Mg(2+)-dependent endonuclease. In addition, insulin-like growth-factor-binding protein 5 (IGFBP-5) is induced de novo. We propose that IGFBP-5 may serve to trigger the apoptotic process through the attenuation of the insulin-like growth factor signalling system (which is necessary for cell survival), and as such, represents a primary thanatogen in the prostate.
Article
In this study, we investigated the involvement of GH in rat prostate function. First, we demonstrated that specific transcripts corresponding to the GH receptor (4.5 kilobases) and to the GH-binding protein (1.2 kilobases) were expressed in the normal rat prostate, but also in all prostatic carcinoma cell lines tested (LNCaP, PC-3, MAT-Lu, MAT-LyLu, and Pif-1). Moreover, these transcripts were much more abundant in the human and rat carcinoma cells than in the normal tissue. One-year-old dwarf rats were supplemented for 7 days with saline (group DR1) or highly purified rat GH (group DR2). Northern blotting and quantitation of prostatic messenger RNAs (mRNAs) revealed that GH increases the steady state levels of transcripts coding for androgen receptor (2.4-fold), type I and II 5 alpha-reductases (2.6- and 2.2-fold), and several androgen-dependent proteins [prostatein C3 subunit (3.6-fold), probasin (11.0-fold), and R. W. B. (Royal Winnipeg Ballet) (12.5-fold)]. This suggests that GH might either potentiat...
Article
Benign prostatic hyperplasia (BPH) is a local prostate disorder that generally develops in the peri-urethral and transitional zones, with nodular growth and a variable histological composition. The frequent association of these three elements in BPH in male patients suggests that there is a direct relationship between the normal physiological phenomena of prostate growth regulation and the development of BPH, and that there are intrinsic local differences in the development of parts of the prostate. BPH is thus above all linked to local abnormalities in the mechanisms by which tissue growth is regulated. Locally, prostate growth depends on non-specific control by growth factors. Several lines of evidence indicate that a number of families of growth factors are involved in BPH, such as the heparin binding growth factor (HBGF) family, which includes basic fibroblast growth factor (bFGF) and keratinocyte growth factor (KGF), the transforming growth factor (TGFß) family, the epidermal growth factor (EGF) family that includes EGF itself and transforming growth factor alpha (TGFα), platelet-derived growth factor (PDGF), nerve growth factor (NGF) and insulin-like growth factor type I (IGF-1). bFGF is linked to heparan sulfate in the extra-cellular matrix and stimulates growth of the stroma, while KGF and NGF, which are synthesized by the prostatic stroma, are mainly bound to epithelial receptors. TGFß is an inhibitor of both stromal and epithelial cells, and its production increases when the androgen supply drops. All these elements suggest that the growth factors, which are produced by prostatic tissue and which control the interactions between the stroma and the prostatic epithelium, play a part in the genesis of BPH. Nevertheless, the data presently available on this subject are insufficient to establish the mechanisms underlying the progression from a normal prostate to BPH.
Article
The somatostatin (SS) analog octreotide has been successfully used in the treatment of (neuro)endocrine tumors. The mechanism of action of the tumor (growth) inhibitory action by octreotide is not fully understood. We have investigated the effect of octreotide on 7315b rat pituitary tumor cell growth, PRL release, and intracellular PRL concentrations in vitro. When cultured in medium with 10% fetal calf serum, the number of high affinity SS receptors increased with increasing culture time. On days 7, 14, and 21 of culture, the number of SS receptors amounted to 978 +/- 217, 3588 +/- 705, and 5865 +/- 3332 fmol/mg protein, respectively, whereas they were not measurable on day 0. From days 0-7, 7-14, and 14-21 of culture, octreotide (1 pM to 1 microM) inhibited PRL release and the intracellular PRL concentration, with IC50 values in the nanomolar range. However, no inhibition of cell growth was observed by these octreotide concentrations from day 0-7 of culture, while octreotide inhibited cell growth in a dose-dependent fashion from days 7-14 and 14-21 of culture (maximal inhibition by 25% and 26%, respectively). In a series of nine consecutive experiments we found a significant positive correlation between the percent inhibition of cell growth induced by 1 microM octreotide and the number of SS receptors on 7315b cells (r = 0.7865; P = 0.012). Inhibition of PRL release did not correlate with SS receptor numbers. Octreotide (1 microM) inhibited forskolin (0.5 microM)-stimulated cell growth and intracellular PRL concentrations, while in the presence of a high concentration of forskolin (10 microM), octreotide had no effect on forskolin-stimulated cell growth and intracellular PRL concentrations. In addition, its PRL release inhibitory effect was significantly lower in forskolin-stimulated cultures. Pretreatment of the cells with pertussis toxin (10 micrograms/liter) completely prevented the inhibition of cell growth by octreotide and diminished the inhibitory effect of octreotide on PRL release. Finally, 1 microM octreotide significantly inhibited forskolin-stimulated cAMP production (by 29% and 53% on days 7 and 14 of culture, respectively). We conclude that 1) octreotide inhibits 7315b rat pituitary tumor cell proliferation via a pertussis toxin-sensitive GTP-binding protein- and adenylate cyclase-dependent mechanism; and 2) the number of SS receptors on 7315b pituitary tumor cells may determine whether octreotide exerts a direct antiproliferative effect, whereas its antihormonal effect occurs in the presence of relatively low numbers of SS receptors. This suggests a dissociation of the antiproliferative and antihormonal effects induced by octreotide.
Article
In this study we investigated the involvement of several different pituitary hormones on rat prostate development. 22-day-old Wistar rats, hypophysectomized (hypox) at 19 days of age were supplemented with highly purified human prolactin (hPRL), human luteinizing hormone (hLH), porcine follicle-stimulating hormone (pFSH), and bovine growth hormone (bGH) or with saline. Quantitative analysis of RNAs shows that treatment with either PRL or GH increases significantly steady-state mRNAs levels of the following genes in the prostate: androgen receptor (AR) (respectively 3.5- and 4.8-fold above hypox controls), IGF-I (5- and 2.7-fold), and IGF-I receptor (2.9- and 2.3-fold). LH and FSH, by contrast, have negative effects on these parameters. To test whether the enhancing effect of PRL and GH on AR-mRNA abundance was followed by increased content in the protein itself, binding assays were performed with the androgen agonist [3H]R1881 (131 and 153 fmol/mg protein while hypox controls contained 110 fmol/mg protein). In addition to the well-documented presence of prolactin receptors in prostatic tissues, we have further demonstrated, by means of nuclease S1 protection assays plus dot- and Northern-blot analyses, that a GH receptor mRNA is produced in the immature rat prostate. Moreover, we observed not only strong lactogenic but also purely somatogenic binding to be occurring in the immature prostates. Finally, we have studied IGF-I mRNA content in separated epithelial/stromal cell fractions and have concluded that IGF-I expression is principally located in the prostatic stroma. Taken together, these results suggest that PRL and GH are involved in regulating AR synthesis, at least partially by direct action on the organ. In this context IGF-I appears as a paracrine factor playing a role in epithelium/stroma interactions during prostatic development.
Article
The prostate gland contains three major glandular regions--the peripheral zone, the central zone, and the transition zone--which differ histologically and biologically. The central zone is relatively resistant to carcinoma and other disease; the transition zone is the main site of origin of prostate hyperplasia. There are also several important nonglandular regions concentrated in the anteromedial portion of the gland. Each glandular zone has specific architectural and stromal features. In all zones, both ducts and acini are lined by secretory epithelium. In each zone, there is a layer of basal cells beneath the secretory lining, as well as interspersed endocrine-paracrine cells. Frequent deviations from normal histology include post-inflammatory atrophy, basal cell hyperplasia, benign nodular hyperplasia, atypical adenomatous hyperplasia, and duct-acinar dysplasia. These lesions may at times be confused with carcinoma, especially in biopsy material.
Article
I. Introduction MALE accessory sex glands such as the prostate, seminal vesicle, and bulbourethral gland have served in various capacities as models for investigating the action of androgens in regulating epithelial growth, RNA and protein synthesis, and secretory activity. The most extensively studied gland of this group is the prostate. This gland is found exclusively in mammals and produces many components of semen such as fructose, zinc ions, and various proteins important for the formation of the copulatory plug in rodents. The impetus for investigating regulation of prostatic growth and function stems in part from the many pathological complications which affect this gland. The prostate is the site of various types of inflammatory and infectious conditions (1) as well as benign and malignant proliferative changes in aging males. Despite extensive research, little of the pathogenesis or natural history of these diseases has been elucidated (2). Benign prostatic hypertrophy (BPH), a disease in which t...
Article
After lung cancer, prostate cancer is the leading cause of deaths from cancer among men in the United States. It will claim 40,000 lives in 1995.1 Studies in the early 1990s demonstrated that levels of prostate-specific antigen (PSA), a serine protease, are elevated in most men with clinically important prostate cancer and that measuring them is the best means for early detection of the disease.2–5 In 1993, the American Cancer Society recommended that clinicians measure PSA in all men 50 years of age and older as part of an annual prostate examination and that PSA screening should begin at . . .
Article
In this study, we investigated the involvement of GH in rat prostate function. First, we demonstrated that specific transcripts corresponding to the GH receptor (4.5 kilobases) and to the GH-binding protein (1.2 kilobases) were expressed in the normal rat prostate, but also in all prostatic carcinoma cell lines tested (LNCaP, PC-3, MAT-Lu, MAT-LyLu, and Pif-1). Moreover, these transcripts were much more abundant in the human and rat carcinoma cells than in the normal tissue. One-year-old dwarf rats were supplemented for 7 days with saline (group DR1) or highly purified rat GH (group DR2). Northern blotting and quantitation of prostatic messenger RNAs (mRNAs) revealed that GH increases the steady state levels of transcripts coding for androgen receptor (2.4-fold), type I and II 5 alpha-reductases (2.6- and 2.2-fold), and several androgen-dependent proteins [prostatein C3 subunit (3.6-fold), probasin (11.0-fold), and R. W. B. (Royal Winnipeg Ballet) (12.5-fold)]. This suggests that GH might either potentiate the action of androgens on the prostate or act directly on this gland by a mechanism that does not depend on testicular androgens. To address this question, we supplemented hypophysectomized and castrated adult rats for 7 days with saline (group HC1), rat GH (group HC2), testosterone propionate (group HC3), or GH plus testosterone (group HC4), starting 3 days after castration. In this animal model, the abundance of C3 mRNA increased in all hormone-treated rats; the stimulation factors were 3.5 (group HC2), 25.5 (group HC3), and 9.5 (group HC4) compared to group HC1. Analysis of prostatein synthesis by Western blotting confirmed these results at the protein level. The same trend was observed for probasin and RWB mRNA levels. Probasin mRNA increased 4.5-fold in group HC2 and 12-fold in group HC3, but did not increase in group HC4 (both hormones combined); enhancement of RWB mRNA was, respectively, 5.0-, 28.0-, and 15.0-fold in groups HC2, HC3, and HC4. GH did not affect the abundance of androgen receptor mRNA. As described previously, the level of this mRNA dropped significantly in group HC3. GH alone did not significantly alter the level of either 5 alpha-reductase mRNA, whereas testosterone, alone or with GH, produced a 2-fold increase in type II 5 alpha-reductase mRNA (groups HC3 and HC4). Type I isoenzyme mRNA reached 1.6 times the control level (group HC1) in groups HC3 and HC4.(ABSTRACT TRUNCATED AT 400 WORDS)