ArticlePDF Available

Khan, AS, Smith, LC, Anscombe, IW, Cummings, KK, Pope, MA and Draghia-Akli, R. Growth hormone releasing hormone plasmid supplementation, a potential treatment for cancer cachexia, does not increase tumor growth in nude mice. Cancer Gene Ther 12:54-60

Authors:

Abstract and Figures

Growth hormone releasing hormone (GHRH) is known to have multiple anabolic effects and immune-stimulatory effects. Previous studies suggest that treatment with anabolic hormones also has the potential to mitigate the deleterious effects of cancer cachexia in animals. We studied the effects of plasmid-mediated GHRH supplementation on tumor growth and the role of antitumor immune cells with two different human tumor cell lines, NCI-H358 human bronchioalveolar carcinoma and MDA-MB-468 human breast adenocarcinoma, subcutaneously implanted in nude mice. GHRH supplementation by delivery of human GHRH from a muscle-specific GHRH expression plasmid did not increase tumor progression in tumor-bearing nude mice. Male animals implanted with the NCI-H358 tumor cell line and treated with the GHRH-expressing plasmid exhibited a 40% decrease in the size of the tumors (P<.02), a 48% increase in white blood cells (P<.025) and a 300% increase in monocyte count (P<.0001), as well as an increase in the frequency of activated CD3+ and CD4+ cells in the tumors, compared to tumors of control animals. No adverse effects were observed in animals that received the GHRH-plasmid treatment. The present study shows that physiological stimulation of the GHRH-GH-IGF-I axis in mice with cancer does not promote tumor growth and may provide a viable treatment for cancer cachexia in humans.
Content may be subject to copyright.
Growth hormone releasing hormone plasmid supplementation, a
potential treatment for cancer cachexia, does not increase tumor
growth in nude mice
Amir S Khan,
1
Louis C Smith,
1
Ingrid W Anscombe,
1
Kathleen K Cummings,
1
Melissa A Pope,
1
and Ruxandra Draghia-Akli
1,2
1
ADViSYS, Inc., The Woodlands, Texas 77381, USA; and
2
Department of Molecular and Cellular Biology,
Baylor College of Medicine, Houston, Texas 77030, USA.
Growth hormone releasing hormone (GHRH) is known to have multiple anabolic effects and immune-stimulatory effects. Previous
studies suggest that treatment with anabolic hormones also has the potential to mitigate the deleterious effects of cancer cachexia in
animals. We studied the effects of plasmid-mediated GHRH supplementation on tumor growth and the role of antitumor immune
cells with two different human tumor cell lines, NCI-H358 human bronchioalveolar carcinoma and MDA-MB-468 human breast
adenocarcinoma, subcutaneously implanted in nude mice. GHRH supplementation by delivery of human GHRH from a muscle-
specific GHRH expression plasmid did not increase tumor progression in tumor-bearing nude mice. Male animals implanted with
the NCI-H358 tumor cell line and treated with the GHRH-expressing plasmid exhibited a 40% decrease in the size of the tumors
(Po.02), a 48% increase in white blood cells (Po.025) and a 300% increase in monocyte count (Po.0001), as well as an increase
in the frequency of activated CD3
þ
and CD4
þ
cells in the tumors, compared to tumors of control animals. No adverse effects were
observed in animals that received the GHRH-plasmid treatment. The present study shows that physiological stimulation of the
GHRH–GH–IGF-I axis in mice with cancer does not promote tumor growth and may provide a viable treatment for cancer cachexia
in humans.
Cancer Gene Therapy (2005) 12, 54–60. doi:10.1038/sj.cgt.7700767
Published online 17 September 2004
Keywords: plasmid; GHRH; cachexia; electroporation; muscle
T
hree of the hormones largely responsible for postnatal
growth in animals and humans include growth
hormone releasing hormone (GHRH), which stimulates
growth hormone (GH) production and secretion from the
anterior pituitary,
1
and insulin-like growth factor-I (IGF-
I) that is responsible for many of the indirect effects of
GH.
2
The effects of these hormones on development,
growth, metabolism and regeneration have been widely
documented.
3,4
Recent studies in different animal models
and humans have also shown that GHRH has immune-
stimulatory effects, both through stimulation of the GH
axis and direct actions as an immune-modulator.
5
Conflicting data exist regarding the role of the GHRH–
GH–IGF-I axis in tumorigenesis and cancer-associated
pathology. Some studies have suggested that carcinogen-
esis is dependent upon critical plasma levels of GH and
IGF-I.
6
GH- or IGF-I-deficient animals are resistant to
chemically induced carcinogenesis.
7
Circulating IGF-I
levels play an important role in tumor development and
metastasis
8
and IGF-II mRNA levels are increased in
some tumor lines.
9
By contrast, other studies have failed
to demonstrate an effect of GH on cancer development
10
or have concluded that GH may actually improve the
efficacy of cancer chemotherapy.
11
Preclinical studies in rodents have suggested that
anabolic hormones, such as GH and IGF-I, may reverse
the catabolic state associated with cachexia, one of the
major complications of cancer and cancer therapies,
12
as
well as inhibit metastases in tumor-bearing animals.
13,14
In this study, the use of species-specific GHRH was not
necessary. Numerous studies have shown that GHRH of
different mammalian origin or GHRH analogs exert
similar effects in species such as dogs, pigs, cattle and
rodents.
15,16
In a previous study in immunocompetent
mice with implanted LL-2 adenocarcinoma cell line, we
showed that stimulation of the GH axis by intramuscular
delivery of a GHRH plasmid increased serum IGF-I
concentrations by 13% (an indicator of GHRH activity),
decreased growth of the tumor by 20% in males and 11%
in females, attenuated tumor metastases by up to 57%,
and prevented muscle atrophy. These results suggest a
role for plasmid-mediated GHRH therapy in reversing
the catabolic processes associated with cancer cachexia.
17
Received April 9, 2004.
Address correspondence and reprint requests to: Dr Ruxandra
Draghia-Akli, MD, PhD, Vice President of Research, ADViSYS,
Inc., 2700 Research Forest Drive, Suite 180, The Woodlands,
TX 77381, USA. E-mail: ruxandradraghia@advisys.net or
ada@bcm.tmc.edu
Cancer Gene Therapy (2005) 12, 5460
r
2005 Nature Publishing Group All rights reserved 0929-1903/05 $30.00
www.nature.com
/
cgt
Similar results were obtained in dogs with spontaneous
malignancies.
18
Long-term evaluation of the cancer-
afflicted dogs that received plasmid-mediated GHRH
supplementation showed a significant improvement in
hematological parameters and quality of life.
19
Clinical
use of GHRH-expressing plasmids for cancer cachexia,
however, requires that ectopic GHRH expression does
not upregulate tumor growth or increase tumor pro-
gression.
In lieu of a constitutively active system of GHRH
delivery, regulated expression of GHRH may be required
under some circumstances. A mifepristone (MFP)-indu-
cible plasmid vector system was used in this case.
20
An
earlier version of this MFP-inducible GHRH system has
been shown to increase serum IGF-I, lean body mass,
body weight, and bone mineral density in SCID mice.
21
In the present study on nude mice with implanted NCI-
H358 human bronchioalveolar carcinoma
22
or MDA-
MB-468 breast adenocarcinoma cells,
23
we tested the
hypothesis that physiologic stimulation of the GHRH-
GH-IGF-I axis through a plasmid-based GHRH expres-
sion system does not stimulate tumor growth in immune-
deficient animals, and thus may prove beneficial as a
treatment for cancer cachexia.
Materials and methods
Cell culture
NCI-H358 cells (ATCC CRL-5807) and MDA-MB-468
cells (ATCC HTB-132) were obtained from ATCC
(Manassas, VA) and stored in liquid nitrogen. Cells were
rapidly thawed and plated in DMEM media (GIBCO,
Grand Island, NY) with 10% fetal bovine serum
(GIBCO, Grand Island, NY) and 1% penicillin/strepto-
mycin. Cells were grown to 80% confluence, removed by
adding 0.1% trypsin-EDTA (Gibco), centrifuged and
resuspended in 1 PBS. Cells were counted before
implantation.
DNA constructs
The plasmid pSPc5-12 contained a 360 bp SacI/BamHI
fragment of the SPc5-12 synthetic promoter.
24
To
generate pSP-hGHRH(1-40), the human GHRH cDNA
was modified by site-directed mutagenesis of human (1-
44)OH GHRH cDNA, and cloned into the BamHI/Hind
III sites of pSP-GHRH, followed by the 3
0
untranslated
region and poly(A) signal of hGH gene.
25
The GHRH-
inducible (IS) is a two-plasmid system. Plasmid pGS1633
codes for GeneSwitch regulatory protein, version 4.0 and
is controlled by a muscle-specific promoter (Valentis,
Burlingame, CA). Plasmid pGHRH(1-40) is an inducible
human GHRH plasmid. The GeneSwitch and inducible
GHRH plasmids were mixed to generate a 1:10 mol/mol
solution.
21
Control plasmid, pSP-b-gal, contained the
Escherichia. coli b-galactosidase gene under control of the
same muscle-specific promoter. Plasmids were grown in
E. coli DH5a, (GIBCO, Grand Island, NY). Endotoxin-
free plasmid (Qiagen Inc., Chatsworth, CA) preparations
were diluted to 0.8 mg/mL in sterile water and stored at
801C prior to use. Mifepristone (Sigma, St. Louis, MO)
was diluted in sesame oil and 360 mg/kg was administered
by gavage three times a week starting at Day 7.
Animals
Hsd:Athymic Nude-nu mice were obtained from Harlan
(Indianapolis, IN) or Charles River (Raleigh, NC) and
allowed to acclimate for 2 weeks. Rodents were housed
five per cage on Tek-Fresh Autoclavable Bedding
(Harlan) and given ad libitum access to food (Autocla-
vable Rodent Lab Diet 5010, Harlan) and water (reverse-
osmosis, UV-treated, sterile-filtered from municipal water
supply). No known contaminants that would interfere
with the outcome of the study were present in feed or
water. Animals were housed at 22731C, with a relative
humidity of range between 30 and 80% on a 12- hour
light/dark cycle.
Injection, electroporation, and experimental procedure
Animal groups were: Group 1 (constitutive pSP-GHRH),
Group 2 (control pSP-bgal), Group 3 (GHRH-inducible
system (GHRH-IS), no MFP), Group 4 (GHRH-IS,
MFP), Group 5 (control pSP-bgal, MFP), and Group 6
(no plasmid). All animals received a pre-experiment
physical examination by a registered veterinary technician
prior to selection for testing. At Day 7, all animals were
weighed, bled, and randomly assigned to one of six
groups (n ¼ 20/group, 10 of each sex). On Day 1,
animals were weighed, bled, and injected subcutaneously
in the flank with tumor cells in 30 mL PBS. Nude mice
received either 2 10
7
NCI-H358 cells or 1 10
7
MDA-
MB-468 cells. Mice were anesthetized on Day 0 with 0.5–
0.7mL/kg of a combination anesthetic: ketamine
(42.8 mg/mL), xylazine (8.2 mg/mL) and acepromazine
(0.7 mg/mL). A measure of 25 mL of the thawed plasmid
stock was injected into the lateral gastrocnemius muscle
using 3/10 cm
3
syringe with 26-gauge needle. At 2 minutes
after injection, the injected muscle was electroporated (3
pulses, 150 V/cm, 50 milliseconds) with a BTX ECM 830
electroporator and two-needle electrodes (BTX, San
Diego, CA), as described.
26
Animals were weighed and
bled once a week, while tumor volume was evaluated
twice a week using Promax NSK Electronic Digital
Calipers (Fred Fowler Co., Newton, MA) and Gage
Wedge for Sylvac Measuring Tools software (TAL
Technologies, Philadelphia, PA). Length, width, and
depth of the tumors were separately measured and then
used to calculate tumor volume.
Necropsy and histopathology
Animals were weighed and then euthanized by CO
2
inhalation on Days 42–43 (NCI-H358 mice) and Days 33–
34 (MDA-MB-468 mice). Organs (lungs, heart, liver,
spleen, kidneys, and the injected gastrocnemius) were
excised, weighed and checked for gross pathologies.
Gastrocnemius and any of the excised organs with
macroscopic abnormalities were fixed in 10% buffered
GHRH does not increase tumor growth in nude mice
AS Khan et al
55
Cancer Gene Therapy
formalin overnight, washed in PBS, and transferred to
70% ethanol for storage. Tumor was also excised,
weighed, fixed in 10% buffered formalin overnight, and
stored in 70% ethanol. For MDA-MB-468 males, a
complete histopathological examination was performed
on internal organs (brain, heart, lung, liver, spleen, and
kidneys), injected muscle, and tumor (IDEXX Labora-
tories, Inc., West Sacramento, CA). The tissues were
paraffin-embedded, sectioned at 4–5 mm, stained with
hematoxylin/eosin and examined microscopically by
IDEXX Laboratories. An independent licensed veterinary
pathologist read slides of each organ, including tumors of
each animal and data were recorded. Tissue from NCI-
H358 mice was paraffin embedded, sectioned at 4–5 mm,
stained with hematoxylin/eosin and examined microsco-
pically for micrometastases.
CD3 and CD4 immunohistochemistry
Tumor sections were selected from NCI-H358 male
animals from Groups 1 and 2. Sections were deparaffi-
nized and subsequently washed in PBS. Slides were
stained using goat ABC staining system (Santa Cruz
Biotechnology, Santa Cruz, CA) following the manufac-
turer’s instructions with slight modifications. Briefly, the
sections were first incubated in 0.03% hydrogen peroxide
in methanol solution to block endogenous peroxidases,
then incubated in the blocking solution (1.5% donkey
serum (Santa Cruz Biotechnology, Santa Cruz, CA) in
PBS), and finally, incubated overnight at 41C in the
primary antibody, CD3-e
0
(M-20) or CD-4 (C-18) (Santa
Cruz Biotechnology, Santa Cruz, CA), diluted 1:2000
(CD3-e
0
) and 1:1500 (CD-4) in blocking solution. After
PBS washes, the secondary antibody was applied for 30
minutes at room temperature and slides were incubated in
ABC solution for 30 minutes, as per kit instructions.
Slides were washed in PBS between each step of the
procedure. Peroxidase activity was revealed using diami-
nobenzidine (DAB) as substrate (Vector Laboratories,
Burlingame, CA) for 4 minutes. The stained sections were
visualized on an Olympus
s
BX51 microscope (Leeds
Instruments, Irving, TX) with a 20 objective, and
digital images of the sections were captured using an
Optronics MagnaFire digital color camera with the
MagnaFire 2.0 software (Optronics, Goleta, CA). The
observer was blinded to the treatment groups and counted
a random set of 9–18 fields per group. The within-animal
average was corrected for tumor volume, and then used to
calculate an average of the CD3
þ
and CD4
þ
cell counts/
tumor volume per group.
CBC and biochemistries
At necropsy, whole blood was collected in Microtainer
Brand tubes with EDTA (Becton Dickinson, Franklin
Lakes, NJ) for CBC analysis and in Microtainer Serum
Separator tubes (Becton Dickinson, Franklin Lakes, NJ)
for serum biochemistries. All tests were performed by
IDEXX Contract Research Services (West Sacramento,
CA). Parameters tested in the biochemical analysis were:
ALT (alanine aminotransferase), AST (aspartate amino-
transferase), creatinine kinase, albumin, total protein,
bilirubin, cholesterol, glucose, calcium, phosphorous,
bicarbonate, chloride, potassium, and sodium.
IGF-I radioimmunoassay
Serum was aliquoted for serum IGF-I measurement using
a mouse-specific IGF-I kit (Diagnostic Systems Labora-
tories, Inc., Webster, TX). The intra-assay variability was
6.6% for NCI-H358 males and 5.0% for NCI-H358
females.
Statistical analysis
A Microsoft Excel statistics analysis package was used.
The mean values were compared with paired t-test or
ANOVA with subsequent Student’s t-test as post hoc test.
Po.05 was taken as the level of statistical significance.
Results
Body weight
No significant differences in body weight were observed
among groups during the treatment period.
Tumor volume
As an initial observation, tumor growth rate was higher
for the NCI-H358 males (Fig 1a) than for the females
(Fig 1b). In NCI-H358 males, animals treated with the
constitutive GHRH plasmid exhibited a 40% decline in
tumor volume by Day 40 (Po.02), compared to b-
galactosidase (pSP-b-gal) plasmid controls. In NCI-H358
animals, the females treated with the constitutive GHRH
plasmid exhibited a 33% decrease in tumor volume by
Day 40 (P ¼ .14), compared to pSP-b-gal plasmid controls
and 37% decline (P ¼ .15) in final tumor weight at
necropsy. Male and female mice that received the GHRH-
IS activated at 7 days after tumor implantation, had a
23% reduction in tumor growth (Po.05 for males, and
P ¼ .15 for females, due to high variance within the
groups). Tumor volumes in MDA-MB-468 males were
not statistically significantly different (data not shown)
among groups. Female animals implanted with MDA-
MB-468 were removed from analysis due to abnormalities
in growth rate of MDA-MB-468 cells in culture and,
unlike other experiments, had inconsistent tumor devel-
opment.
CBC and serum biochemistry
In the NCI-H358 male mice treated with the constitutively
active GHRH plasmid, white blood count (WBC) was
increased 48.8% (Po.025) (Fig 2a) and percent mono-
cytes were increased 300% (Po.00005) (Fig 2b) com-
pared to pSP-b-gal plasmid controls. In MDA-MB-468
males treated with the constitutively active GHRH
plasmid, lymphocyte count was increased by 20%
(Po.045) relative to pSP-b-gal plasmid controls (Fig
2c). In the NCI-H358 female mice, with overall smaller
tumors and tumor growth rates, no biologically or
GHRH does not increase tumor growth in nude mice
AS Khan et al
56
Cancer Gene Therapy
statistically significant changes were found in the CBC or
biochemistry values.
Necropsy
Tumor weights at necropsy were 45% smaller (P ¼ .017)
in Group 1 relative to Group 2 in NCI-H358 male
animals (Fig 3a), and were 37% smaller in the NCI-H358
females, although this did not attain statistical signifi-
cance due to individual variation of tumor size (P ¼ .15)
(Fig 3b). Tumor weights were not significantly different
between Groups 1 and 2 in the MDA-MBA-468 animals.
No biologically significant changes were found in the
mean necropsy organ weights (mean organ weight/mean
body weight) of male or female mice implanted with either
cell line.
Histopathology
A complete histopathological examination was performed
on internal organs (brain, heart, lung, liver, spleen, and
kidneys), injected muscle and tumor of MDA-MBA-468
animals killed at Days 41–42. Analysis showed similar
tumors in all examined animals, with expansile and locally
invasive nodular tumors composed of cords of neoplastic
epithelial cells. There was no difference in tumor
vascularization between groups. Only one micrometasta-
sis was found, in the liver of a control animal.
Extramedullary hematopoiesis was noted in the spleen
of all animals. Two animals in the pSP-bgal plasmid
group also exhibited lymphoid hyperplasia in the spleen.
Focal areas of generally mild muscle atrophy were present
Figure 1 (a) Tumor growth as measured by tumor volume in MALE
mice implanted with NCI-H358 human bronchioalveolar carcinoma
cells. Tumors were measurable from Day 5 of the experiments, and
tumor volume measurements were taken until Day 40. (b) Tumor
growth as measured by tumor volume in Groups 1 and 2 of the
FEMALE mice implanted with NCI-H358 human bronchioalveolar
carcinoma cells. Tumors were measurable from Day 4 of the
experiments, and tumor volume measurements were taken until Day
40. *Po.02.
Figure 2 CBC values in tumor-bearing mice either treated with
constitutively active GHRH plasmid (pSP-GHRH) or nontreated
controls: (a) White blood cell counts (WBC) in NCI-H358 males,
*Po.025 (b) monocyte percentage in NCI-H358 males, *Po.00005
and (c) lymphocyte percentage in MDA-MB-468 male mice,
*Po.045.
GHRH does not increase tumor growth in nude mice
AS Khan et al
57
Cancer Gene Therapy
in many animals across all groups. Macrophage infiltra-
tion was also found in most animals and did not appear to
be treatment related. A survey of liver and lung sections
from NCI-H358 mice did not reveal any apparent
differences in the numbers of micrometastases.
Immunohistochemistry
Tumors collected at necropsy (Days 41–42) were exam-
ined for tumor-infiltrating T cells. CD3
þ
cell counts
corrected for tumor volume were increased 66.7%
(P ¼ .18) and CD4
þ
cell counts corrected for tumor
volume were increased 87.2% (P ¼ .15) in Group 1
animals versus Group 2 animals (Fig 4).
Serum IGF-I values
Serum IGF-I levels, while increased in the GHRH-treated
groups, were not significantly different among treatment
groups for the duration of the experiments.
Discussion
NCI-H358 human bronchioalveolar carcinoma and
MDA-MB-468 human breast adenocarcinoma cell lines
have previously been used to investigate tumor progres-
sion in mice.
27,28
This report demonstrates that plasmid-
mediated, physiologic GHRH supplementation did not
increase tumor progression in tumor-bearing nude mice.
Furthermore, male animals implanted with the NCI-H358
tumor cell line and treated with a GHRH-expressing
plasmid showed a decrease in tumor volume, increased
white blood cells and monocyte count, as well as increased
intratumoral activated CD3
þ
and CD4
þ
cells, as
compared to control animals. Finally, this work confirms
earlier work in immunocompetent animals
17
and suggests
that the mechanism responsible for tumor growth
reduction involves immune stimulation. These results
provide support for the use of GHRH-expressing
plasmids in the treatment of cancer cachexia.
The present data confirm the results of a previous study
that used LL-2 adenocarcinoma cell line in immunocom-
petent mice.
17
In both studies, pSP-GHRH-treated male
mice exhibited the highest decrease in tumor volume at
the end of the study compared to controls. In the present
study, tumor weights at necropsy were smaller in pSP-
GHRH-treated animals as compared to animals that
received GHRH-IS inducible system that was activated at
7 days after tumor implantation. Thus, during tumor-
igenesis, it is preferable to increase circulating GHRH
levels early rather than late for therapeutic success. Body
weight was not changed in any of the experiments,
confirming that at lower doses the direct effects of GHRH
on tissues may be more important than the effects
mediated through the GH axis. It is known that IGF-I
is highly dependent on metabolic state, and substantially
decreased in subjects with cancer cachexia.
29
Importantly,
the nude mice in the present experiment maintained
circulating IGF-I concentrations within physiologic lim-
its. As seen in GH-deficient patients administered
physiologic doses of GH,
30
the maintenance of normal
IGF-I levels results in improved clinical outcome, while
the adverse effects of IGF-I overstimulation are avoided.
Sexual dimorphism in the neuroendocrine regulation of
the GH axis in nude mice is largely unknown. In humans,
a gender dissociation within the GH/IGF-I axis is evident
in protracted critical illness, with men showing greater
Figure 3 Tumor weights at necropsy (Day 42 in males, Day 43 in
females) in (a) NCI-H358 male, *P ¼ .017 and in (b) NCI-H358
female mice.
Figure 4 Intratumoral CD3
þ
and CD4
þ
cell counts corrected for
tumor volume in NCI-H358 male mice treated with constitutive
GHRH plasmid, as compared to nontreated controls.
GHRH does not increase tumor growth in nude mice
AS Khan et al
58
Cancer Gene Therapy
loss of pulsatility and regularity within the GH secretory
pattern than women (despite indistinguishable total GH
output) and concomitantly lower IGF-I levels;
31
as a
clinical consequence, females appear to be protected
against, at least in part, adverse outcome from prolonged
critical illness. In the present study, we found that certain
tumors develop at a slower rate in female compared to
male mice, remarkably consistent with our previously
published study.
17
Additional studies are required to
elucidate the mechanism of sexual dimorphism of the
GHRH axis on tumor progression.
Enhancement of immune function is also one of the
possible mechanisms of the decline in tumor growth
observed in the present study. A substantial body of
research exists to support the production of GHRH, GH
and IGF-I by cells of the immune system.
32
In the present
study, tumor-bearing male mice treated with GHRH
plasmid demonstrated significant increases in WBC,
monocytes, and lymphocytes, confirming a similar
observation in dogs with late-stage malignancy.
18
Furthermore, the numbers of intratumoral CD3
þ
and
CD4
þ
cells were higher in male NCI-H358 tumor-bearing
animals treated with GHRH plasmid compared to
controls, which could reflect an immune response against
the tumor itself.
33
The data support the presence of an antitumor immune
function in athymic nude mice. Since nude mice lack a
normal thymus, they are characterized by low numbers of
mature T cells.
34
These animals have measurable numbers
of Thy-1
þ
and CD8
þ
and small numbers of CD4
þ
T
cells can be found. In addition, a second T-cell receptor
(TCR), gamma delta-TCR, is expressed in the spleens of
nude mice. Lake et al
35
demonstrate the quantitative
measurement of TCR expression by T cells that mature in
athymic nude mice, and they suggest that the extrathymic
environment, although inefficient, is nevertheless permis-
sive for the maturation of alpha/beta and gamma/delta
TCR-expressing T cells. Kennedy et al
36
provide further
evidence of extrathymic T-cell maturation and that nude
mice accrue increasing numbers of lymphocytes bearing
Thy-1, CD3, CD4, and CD8 with age. Radzikowski et al
37
have reported that splenocytes from young immunodefi-
cient mice retain their cytotoxic immune response to a
challenge. This suggests a possible mechanism for the
antitumor response demonstrated by the mice injected
with GHRH-expressing plasmid in this study.
Schally and colleagues have introduced evidence that
GHRH antagonists may inhibit tumor growth,
38
through
specific splice-variant GHRH receptors on tumor cells.
39
Synthetic GHRH antagonists do not inhibit the bioactiv-
ity of endogenous GHRH, but act primarily through the
inhibition of autocrine production of growth factors in
tumor cells.
40,41
Thus, an endocrine stimulation of the
GHRH axis that results in augmentation of immune
function will likely benefit affected patients, while not
stimulating malignant cell growth.
GHRH expression does not increase tumor growth in
immunodeficient, tumor-bearing mice. The benefits of
plasmid-mediated GHRH treatment may be related to
increased immune activity against the tumor in addition
to anabolic effects on the tumor-bearing animal. An
improved metabolic status of a patient in advanced stages
of cancer may increase the likelihood of the patient
surviving the radiotherapy and/or chemotherapy cancer
treatment. The findings in the present study suggest that
physiological stimulation of the GHRH-GH-IGF-I axis
in patients with cancer will likely enhance the quality of
life and may increase survival time.
Acknowledgments
We particularly thank Dr Malcolm Brenner and the
Center for Cell and Gene Therapy for continuous support
and useful discussions. We also thank Dr Jeff Nordstrom
and Ms Catherine Tone for the editorial correction of this
manuscript. We acknowledge support for this study from
ADViSYS, Inc. (The Woodlands, TX).
References
1. Muller EE, Locatelli V, Cocchi D. Neuroendocrine control
of growth hormone secretion. Physiol Rev. 1999;79:511–607.
2. Laron Z. Insulin-like growth factor 1 (IGF-1): a growth
hormone. Mol Pathol. 2001;54:311–316.
3. Thorner MO, Chapman IM, Gaylinn BD, Pezzoli SS,
Hartman ML. Growth hormone-releasing hormone and
growth hormone-releasing peptide as therapeutic agents to
enhance growth hormone secretion in disease and aging.
Recent Prog Horm Res. 1997;52:215–244 discussion 244-246.
4. Piwien-Pilipuk G, Huo JS, Schwartz J. Growth hormone
signal transduction. J Pediatr Endocrinol Metab. 2002;
15:771–786.
5. Khorram O, Garthwaite M, Golos T. The influence of aging
and sex hormones on expression of growth hormone-
releasing hormone in the human immune system. J Clin
Endocrinol Metab. 2001;86:3157–3161.
6. Shim M, Cohen P. IGFs and human cancer: implications
regarding the risk of growth hormone therapy. Horm Res.
1999;51(Suppl 3):42–51.
7. Ramsey MM, Ingram RL, Cashion AB, et al. Growth
hormone-deficient dwarf animals are resistant to dimethyl-
benzanthracine (DMBA)-induced mammary carcinogenesis.
Endocrinology. 2002;143:4139–4142.
8. Wu Y, Yakar S, Zhao L, Hennighausen L, LeRoith D.
Circulating insulin-like growth factor-I levels regulate colon
cancer growth and metastasis. Cancer Res. 2002;62:1030–
1035.
9. Guo N, Ye JJ, Liang SJ, et al. The role of insulin-like
growth factor-II in cancer growth and progression evi-
denced by the use of ribozymes and prostate cancer
progression models. Growth Horm IGF Res. 2003;13:44–53.
10. Beentjes JA, van Gorkom BA, Sluiter WJ, de Vries EG,
Kleibeuker JH, Dullaart RP. One year growth hormone
replacement therapy does not alter colonic epithelial cell
proliferation in growth hormone deficient adults. Clin
Endocrinol (Oxf). 2000;52:457–462.
11. Cherbonnier C, Deas O, Vassal G, et al. Human growth
hormone gene transfer into tumor cells may improve cancer
chemotherapy. Cancer Gene Ther. 2002;9:497–504.
12. Kotler DP. Cachexia. Ann Intern Med. 2000;133:622–634.
GHRH does not increase tumor growth in nude mice
AS Khan et al
59
Cancer Gene Therapy
13. Torosian MH. Growth hormone and prostate cancer
growth and metastasis in tumor-bearing animals. J Pediatr
Endocrinol. 1993;6:93–97.
14. Wang W, Iresjo BM, Karlsson L, Svanberg E. Provision of
rhIGF-I/IGFBP-3 complex attenuated development of
cancer cachexia in an experimental tumor model. Clin Nutr.
2000;19:127–132.
15. Hashizume T, Ohtsuki K, Sasaki K, et al. Effects of growth
hormone-releasing hormone (GRF) analogs, bovine and rat
GRF on growth hormone secretion in cattle in vivo. Endocr
J. 1997;44:811–817.
16. Campbell RM, Stricker P, Miller R, et al. Enhanced stability
and potency of novel growth hormone-releasing factor
(GRF) analogues derived from rodent and human GRF
sequences. Peptides. 1994;15:489–495.
17. Khan AS, Anscombe IW, Cummings KK, Pope MA, Smith
LC, Draghia-Akli R. Effects of plasmid-mediated growth
hormone releasing hormone supplementation on LL-2
adenocarcinoma in mice. Mol Ther. 2003;8:459–466.
18. Draghia-Akli R, Hahn KA, King GK, Cummings K,
Carpenter RH. Effects of plasmid mediated growth hor-
mone releasing hormone in severely debilitated dogs with
cancer. Mol Ther. 2002;6:830–836.
19. Tone CM, Cardoza DM, Carpenter RH, Draghia-Akli R.
Long-term effects of plasmid-mediated growth hormone
releasing hormone in dogs. Cancer Gene Ther. 2004;11:389–
396.
20. Abruzzese RV, Godin D, Mehta V, et al. Ligand-dependent
regulation of vascular endothelial growth factor and
erythropoietin expression by a plasmid-based autoinducible
GeneSwitch system. Mol Ther. 2000;2:276–287.
21. Draghia-Akli R, Malone PB, Hill LA, Ellis KM, Schwartz
RJ, Nordstrom JL. Enhanced animal growth via ligand-
regulated GHRH myogenic-injectable vectors. FASEB J.
2002;16:426–428.
22. McLemore TL, Eggleston JC, Shoemaker RH, et al.
Comparison of intrapulmonary, percutaneous intrathoracic,
and subcutaneous models for the propagation of human
pulmonary and nonpulmonary cancer cell lines in athymic
nude mice. Cancer Res. 1988;48:2880–2886.
23. Cailleau R, Olive M, Cruciger QV. Long-term human breast
carcinoma cell lines of metastatic origin: preliminary
characterization. In vitro. 1978;14:911–915.
24. Li X, Eastman EM, Schwartz RJ, Draghia-Akli R.
Synthetic muscle promoters: activities exceeding naturally
occurring regulatory sequences. Nat Biotechnol. 1999;17:
241–245.
25. Draghia-Akli R, Fiorotto ML, Hill LA, Malone PB, Deaver
DR, Schwartz RJ. Myogenic expression of an injectable
protease-resistant growth hormone-releasing hormone aug-
ments long-term growth in pigs. Nat Biotechnol. 1999;17:
1179–1183.
26. Khan AS, Fiorotto ML, Hill LA, et al. Nonhereditary
enhancement of progeny growth. Endocrinology. 2002;143:
3561–3567.
27. Zhang L, Akbulut H, Tang Y, et al. Adenoviral vectors with
E1A regulated by tumor-specific promoters are selectively
cytolytic for breast cancer and melanoma. Mol Ther. 2002;
6:386–393.
28. Fernandez Y, Gu B, Martinez A, Torregrosa A, Sierra A.
Inhibition of apoptosis in human breast cancer cells: role in
tumor progression to the metastatic state. Int J Cancer.
2002;101:317–326.
29. Simons JP, Schols AM, Buurman WA, Wouters EF. Weight
loss and low body cell mass in males with lung cancer:
relationship with systemic inflammation, acute-phase re-
sponse, resting energy expenditure, and catabolic and
anabolic hormones. Clin Sci (Lond). 1999;97:215–223.
30. Pincelli AI, Bragato R, Scacchi M, et al. Three weekly
injections (TWI) of low-dose growth hormone (GH) restore
low normal circulating IGF-I concentrations and reverse
cardiac abnormalities associated with adult onset GH
deficiency (GHD). J Endocrinol Invest. 2003;26:420–428.
31. Van den Berghe G, Baxter RC, Weekers F, Wouters P,
Bowers CY, Veldhuis JD. A paradoxical gender dissociation
within the growth hormone/insulin-like growth factor I axis
during protracted critical illness. J Clin Endocrinol Metab.
2000;85:183–192.
32. Burgess W, Liu Q, Zhou J, et al. The immune-endocrine
loop during aging: role of growth hormone and insulin-like
growth factor-I. Neuroimmunomodulation. 1999;6:56–68.
33. Wakabayashi O, Yamazaki K, Oizumi S, et al. CD4(+) T
cells in cancer stroma, not CD8(+) T cells in cancer cell
nests, are associated with favorable prognosis in human
non-small cell lung cancers. Cancer Sci. 2003;94:1003–1009.
34. Ferrick DA, Sambhara SR, Chadwick BS, Miller RG, Mak
TW. The T-cell receptor repertoire is strikingly similar in
older nude mice compared to normal adult mice. Thymus.
1989;13:103–111.
35. Lake JP, Pierce CW, Kennedy JD. CD8
+
alpha/beta or
gamma/delta T cell receptor-bearing T cells from athymic
nude mice are cytolytically active in vivo. J Immunol.
1991;147:1121–1126.
36. Kennedy JD, Pierce CW, Lake JP. Extrathymic T cell
maturation. Phenotypic analysis of T cell subsets in
nude mice as a function of age. J Immunol. 1992;148:
1620–1629.
37. Radzikowski C, Rygaard J, Budzynski W, et al. Strain- and
age-dependent natural and activated in vitro cytotoxicity in
athymic nude mice. APMIS. 1994;102:481–488.
38. Kiaris H, Koutsilieris M, Kalofoutis A, Schally AV. Growth
hormone-releasing hormone and extra-pituitary tumorigen-
esis: therapeutic and diagnostic applications of growth
hormone-releasing hormone antagonists. Expert Opin In-
vestig Drugs. 2003;12:1385–1394.
39. Kineman RD. Antitumorigenic actions of growth hormone-
releasing hormone antagonists. Proc Natl Acad Sci USA.
2000;97:532–534.
40. Chatzistamou I, Schally AV, Varga JL, et al. Inhibition of
growth and reduction in tumorigenicity of UCI-107 ovarian
cancer by antagonists of growth hormone-releasing hor-
mone and vasoactive intestinal peptide. J Cancer Res Clin
Oncol. 2001;127:645–652.
41. Chatzistamou I, Schally AV, Varga JL, et al. Inhibition of
growth and metastases of MDA-MB-435 human estrogen-
independent breast cancers by an antagonist of growth
hormone-releasing hormone. Anticancer Drugs. 2001;12:
761–768.
GHRH does not increase tumor growth in nude mice
AS Khan et al
60
Cancer Gene Therapy
... Ghrelin is a 28-amino acid peptide which acts as the endogenous ligand for the ghrelin receptor (GRLN receptor, formally known as GHS-R1a) [7]. Administration of ghrelin to animals and humans has been shown to stimulate gastric acid secretion and motility, increase food intake and appetite leading to weight gain, promote anabolic activity, and inhibit production of pro-inflammatory cytokines, and thus may provide a viable target for cancer-related cachexia [8][9][10][11]. Ghrelin activity is thought to be mediated by both growth hormone (GH)-dependent and GH-independent mechanisms [8]. However, the short half-life (∼30 min), and parenteral administration requirement of ghrelin has limited its clinical usefulness, and interest has switched to the development of orally available ghrelin mimetics [9][10][11]. ...
... Ghrelin activity is thought to be mediated by both growth hormone (GH)-dependent and GH-independent mechanisms [8]. However, the short half-life (∼30 min), and parenteral administration requirement of ghrelin has limited its clinical usefulness, and interest has switched to the development of orally available ghrelin mimetics [9][10][11]. One of these, anamorelin (ONO-7643, formally known as RC-1291), is a GRLN receptor agonist currently in development for the treatment of non-small-cell lung cancer (NSCLC)-related anorexia and cachexia. ...
... The finding in our study that ghrelin and anamorelin do not promote tumor growth, even in the presence of elevated GH and IGF-1, is consistent with findings from other studies evaluating GH-based therapies in tumor-bearing animals [9,[23][24][25][26], as well as in formal carcinogenicity studies in tumor-free animals [27], as summarized below. ...
Article
Full-text available
Purpose Anamorelin (ONO-7643) is an orally active ghrelin receptor agonist in development for non-small cell lung cancer (NSCLC)-related anorexia/cachexia. It displays both orexigenic and anabolic properties via ghrelin mimetic activity and transient increases in growth hormone (GH). However, increasing GH and insulin-like growth factor-1 in cancer patients raises concerns of potentially stimulating tumor growth. Therefore, we investigated the effect of ghrelin and anamorelin on tumor growth in a murine NSCLC xenograft model. Methods Female nude mice (15–21/group) with established A549 tumors were administered ghrelin (2 mg/kg i.p.), anamorelin (3, 10, or 30 mg/kg p.o.), or vehicle controls daily for 28 days. Tumor growth, food consumption, and body weight were monitored. Murine growth hormone (mGH) and murine insulin-like growth factor-1 (mIGF-1) were measured in plasma. Results Tumor growth progressed throughout the study, with no significant differences between treatment groups. Daily food consumption was also relatively unchanged, while the percentage of mean body weight gain at the end of treatment was significantly increased in animals administered 10 and 30 mg/kg compared with controls (p < 0.01). Peak mGH levels were significantly higher in ghrelin- and anamorelin-treated animals than in controls, while peak mIGF-1 levels were slightly elevated but not statistically significant. All regimens were well tolerated. Conclusions These findings demonstrate that neither anamorelin nor ghrelin promoted tumor growth in this model, despite increased levels of mGH and a trend of increased mIGF-1. Together with anamorelin’s ability to increase body weight, these results support the clinical development of ghrelin receptor agonist treatments for managing NSCLC-related anorexia/cachexia.
... It was found that these GHRH agonists do not stimulate tumor growth or neoplastic transformation. Similarly, in a work conducted by Khan et al., a vector-based expression of GHRH did not stimulate and actually inhibited tumor growth in nude mice xenografted with human bronchioloalveolar and breast carcinoma [49]. ...
Article
Full-text available
Decreased or impaired proliferation capability of dermal fibroblasts interferes with successful wound healing. Several growth factors tested failed to fully restore the growth of fibroblasts, possibly due to their rapid degradation by proteases. It is therefore critical to find new agents which have stimulatory effects on fibroblasts while being highly resistant to degradation. In such a scenario, the activities of two agonistic analogs of growth hormone releasing hormone (GHRH), MR-409 and MR-502, were evaluated for their impact on proliferation and survival of primary human dermal fibroblasts. In vitro, both analogs significantly stimulated cell growth by more than 50%. Under serum-depletion induced stress, fibroblasts treated with MR-409 or MR-502 demonstrated better survival rates than control. These effects can be inhibited by either PD98059 or wortmannin. Signaling through MEK/ERK1/2 and PI3K/AKT in an IGF-1 receptor-independent manner is required. In vivo, MR-409 promoted wound closure. Animals treated topically with MR-409 healed earlier than controls in a dose-dependent manner. Histologic examination revealed better wound contraction and less fibrosis in treated groups. In conclusion, MR-409 is a potent mitogenic and anti-apoptotic factor for primary human dermal fibroblasts. Its beneficial effects on wound healing make it a promising agent for future development.
... The results of this study are consistent with findings from other studies evaluating GH-based therapies in tumor-bearing animals as well as in formal carcinogenicity studies in tumorfree animals. Khan et al. [33] demonstrated that treatment with a GH releasing hormone (GHRH)-expressing plasmid in nude mice implanted with a human bronchoalveolar carcinoma cell line did not increase the growth of the tumor, but rather it reduced tumor volume by 40%, suggesting that the therapeutic role of GH-based therapies may not be limited to cachexia. In a study by Perboni et al., was highlighted that the addition of the ghrelin agonist growth hormone releasing peptide-2 (GHRP-2) to cytotoxic therapy with 5-fluorouracil (5-FU) avoid the anorexia associated with chemotherapy in tumor-bearing cachectic BALB/c mice, and there was also a trend of improved survival in the 5-FU + GHRP-2 treated mice compared with those with 5-FU alone [34]. ...
Article
Cancer cachexia is a multi-organ, multifactorial and often irreversible syndrome affecting many patients with cancer. Cancer cachexia is invariably associated with weight loss, mainly from loss of skeletal muscle and body fat, conditioning a reduced quality of life due to asthenia, anorexia, anaemia and fatigue. Treatment options for treating cancer cachexia are limited. The approach is multimodal and may include: treatment of secondary gastrointestinal symptoms, nutritional treatments, drug, and non-drug treatments. Nutritional counselling and physical training may be beneficial in delaying or preventing the development of anorexia-cachexia. However, these interventions are limited in their effect, and no definitive pharmacological treatment is available to address the relevant components of the syndrome. Anamorelin is a first-in-class, orally active ghrelin receptor agonist that binds and stimulates the growth hormone secretagogue receptor centrally, thereby mimicking the appetite-enhancing and anabolic effects of ghrelin. It represents a new class of drug and an additional treatment option for this patient group, whose therapeutic options are currently limited. In this review we examine the mechanisms of anamorelin by which it contrasts catabolic states, its role in regulation of metabolism and energy homeostasis, the data of recent trials in the setting of cancer cachexia and its safety profile.
... Taken together, data suggest that ghrelin may play an important role in stimulating appetite and food intake in CACS; however, the short half-life (<30 min), and the parenteral administration requirement of ghrelin has limited its clinical usefulness. Therefore, interest has switched to the development of orally-available ghrelin mimetics [55][56][57] . One of these mimetics, anamorelin (ONO-7643, formerly known as RC-1291), is a ghrelin receptor agonist currently in development for the treatment of non-small-cell lung cancer (NSCLC)related cachexia. ...
Article
Introduction: Cancer anorexia-cachexia syndrome (CACS) is associated with increased morbidity and mortality. Anamorelin is a novel, orally active ghrelin receptor agonist in clinical development for the treatment of CACS in NSCLC. The aim of this review is to summarize preclinical and clinical studies evaluating anamorelin as a potential promising treatment for CACS in NSCLC. Areas covered: Pharmacodynamics, pharmacokinetics and metabolism, clinical efficacy, safety and tolerability of anamorelin for the treatment of CACS in NSCLC were reviewed. Anamorelin administration may lead to increases in food intake, body weight and lean body mass, and a stimulatory effect on growth hormone secretion in NSCLC patients. Anamorelin is well tolerated with no dose-limiting toxicities identified to date. Expert opinion: Targeting ghrelin receptors presents the advantage of potentially addressing multiple mechanisms of CACS simultaneously including appetite, muscle protein balance, adipose tissue metabolism, energy expenditure and inflammation. Clinical data suggest that anamorelin is well tolerated and it effectively increases appetite, body weight and lean mass in patients with advanced NSCLC. Long-term safety remains unknown at this time. The potential synergistic effects of anamorelin with nutritional support or exercise as well as its efficacy/safety in other tumor types are also unknown.
... Notably, the A549 NSCLC adenocarcinoma tumor cell line used in that study has been shown to possess a high degree of expression of the IGF-1 receptor [35]. Other studies evaluating GH-based therapies in tumor-bearing animal models [14,[36][37][38][39] and in formal carcinogenicity studies in tumor-free rats and mice [40] support the findings that ghrelin and ANAM do not stimulate tumor growth. Furthermore, in a Phase II clinical study in NSCLC patients, no statistically significant effect on long-term overall survival was noted after 12-weeks of treatment with 50 mg or 100 mg anamorelin compared with placebo [26]. ...
Article
Full-text available
Background Anamorelin HCl (ANAM) is a novel, orally active, ghrelin receptor agonist in clinical development for the treatment of cancer cachexia. We report in vitro and in vivo studies evaluating the preclinical pharmacologic profile of ANAM. Methods Fluorescent imaging plate reader and binding assays in HEK293 and baby hamster kidney cells determined the agonist and antagonist activity of ANAM, and its affinity for the ghrelin receptor. Rat pituitary cells were incubated with ANAM to evaluate its effect on growth hormone (GH) release. In vivo, rats were treated with ANAM 3, 10, or 30 mg/kg, or control orally, once daily for 6 days to evaluate the effect on food intake (FI) and body weight (BW), and once to assess GH response. In pigs, single (3.5 mg/kg) or continuous (1 mg/kg/day) ANAM doses were administered to assess GH and insulin-like growth factor (IGF-1) response. Results ANAM showed significant agonist and binding activity on the ghrelin receptor, and stimulated GH release in vitro. In rats, ANAM significantly and dose-dependently increased FI and BW at all dose levels compared with control, and significantly increased GH levels at 10 or 30 mg/kg doses. Increases in GH and IGF-1 levels were observed following ANAM administration in pigs. Conclusion ANAM is a potent and highly specific ghrelin receptor agonist with significant appetite-enhancing activity, leading to increases in FI and BW, and a stimulatory effect on GH secretion. These results support the continued investigation of ANAM as a potential treatment of cancer anorexia-cachexia syndrome.
Article
Full-text available
Cancer mediated activation of the ActRIIB-ALK4/5 heterodimer by myostatin is strongly associated with muscle wasting. We investigated in vitro and in vivo the efficacy of ALK4/5 receptor blockers SB431542 and GW788388 in preventing muscle wasting, and explored synergy with IGF-I analogue LONG R3 (LR3) IGF-I. In vitro, C2C12 skeletal muscle cells were treated with vehicle, SB431542, GW788388 and LR3 IGF-I. A C26-CD2F1 cachexia model was used to induce cachexia in vivo. Mice were allocated as non-tumour bearing (NTB) or C26 tumour-bearing (C26 TB) vehicle control, treated with SB431542, LR3 IGF-I, SB431542 and LR3 IGF-I, or GW788388 (intraperitoneally or orally). In vitro, differentiation index and mean nuclei count increased using SB431542, GW788388, LR3 IGF-I. In vivo, GW788388 was superior to SB431542 in limiting loss of bodyweight, grip-strength and gastrocnemius weight. and downregulated Atrogin-1 expression comparable to NTB mice. LR3 IGF-I treatment limited loss of muscle mass, but at the expense of accelerated tumour growth. In conclusion, treatment with GW788388 prevented cancer cachexia, and downregulated associated ubiquitin ligase Atrogin-1.
Article
Full-text available
Background: Cancer cachexia is a catabolic syndrome associated with uncontrolled muscle breakdown. There may be associated fat loss. Occurring in high frequency in advanced cancer, it is an indicator of poor prognosis. Besides weight loss, patients experience a cluster of symptoms including anorexia, early satiety, and weakness. The 3 stages of cachexia include stages of precachexia, cachexia, and refractory cachexia. Refractory cachexia is associated with active catabolism or the presence of factors that make active management of weight loss no longer possible. Patients with refractory cachexia often receive glucocorticoids or megasterol acetate. Glucocorticoid effect is short and responses to megasterol are variable. Anamorelin is a new agent for cancer anorexia-cachexia, with trials completed in advanced lung cancer. Acting as an oral mimetic of ghrelin, it improves appetite and muscle mass. This article reviews the pharmacology, pharmacodynamics, and effect on cancer cachexia. Methods: A PubMed search was done using the Medical Subject Headings term anamorelin. Articles were selected to provide a pharmacologic characterization of anamorelin. Results: Anamorelin increases muscle mass in patients with advanced cancer in 2-phase 3 trials. Conclusions: Anamorelin improves anorexia-cachexia symptoms in patients with advanced non-small-cell lung cancer.
Article
AIM: To investigate the effects of recombinant human growth hormone (rhGH) on tumor growth and VEGF expression in subcutaneous xenografts derived from human gastric carcinoma SGC-7901 cells in nude mice. METHODS: The expression of growth hormone receptor (GHR) in human gastric carcinoma cell line SGC-7901 was detected by immunocytochemistry. Thirty nude mice bearing subcutaneous xenografts derived from carcinoma SGC-7901 cells were randomly divided into three groups: control group, low-dose rhGH group and high-dose rhGH group. The low- and high-dose rhGH groups were injected with rhGH at doses of 0.5 and 2.5 U/(kg·d) once a day for two weeks, respectively, while the control group was injected with equal volumes of normal saline for the same duration. The changes in body weight and tumor volume were recorded. The content of serum VEGF in peripheral blood was analyzed by enzyme-linked immunosorbent assay (ELISA). The expression of VEGF mRNA and protein in tumor tissue was detected by reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry, respectively. RESULTS: GHR is highly expressed in SGC-7901 cells. After treatment with rhGH for three days, the tumor volume was significantly larger in the two rhGH groups than in the control group (both P < 0.05). High-dose rhGH revealed stronger tumor growth-promoting effect than low-dose one (P < 0.05). No significant difference was found in the body weight of nude mice among the three groups (all P > 0.05). The content of serum VEGF was elevated more obviously in the high-dose rhGH group than in the low-dose rhGH group and the control group. (252.94 ng/L ± 15.32 ng/L vs 167.60 ng/L ± 9.54 ng/L and 49.94 ng/L ± 5.73 ng/L, respectively; both P < 0.05). The expression level of VEGF protein in tumor tissue was significantly higher in the two rhGH groups than in the control group. The relative expression level of VEGF mRNA was much higher in the high-dose rhGH group than in the low-dose rhGH group and the control group (0.647 ± 0.0447 vs 0.412 ± 0.0351 and 0.323 ± 0.0258, respectively; both P < 0.05). CONCLUSION: RhGH can promote tumor growth and VEGF expression in subcutaneous xenografts derived from human gastric carcinoma SGC-7901 cells in nude mice.
Article
Objective: To investigate the effects of recombinant human growth hormone (rhGH) on tumor growth and tumor angiogenesis factor relevant to Janus kinase 2-signal transducer and activator of transcription 3 of human gastric carcinoma xenografts in nude mice with different expressions of growth hormone receptor (GHR). Methods: Immunocytochemical method was used to pick out one GHR-positive and one GHR-negative cell line. The cells were subcutaneously injected into 26 nude mice separately, then the patterns of xenografts in nude mice with different expressions of GHR were established. The nude mice bearing two different kinds of human gastric caicinoma were equally randomized into control group, low-dose rhGH group, and high-dose rhGH group, and were treated with drugs for 14 days. Changes of tumor volumes and body weight of nude mice were record. The protein and mRNA expressions of tumor angiogenesis factor in tumor tissue were detected by RT-PCR and Western blot, respectively. Results: GHR was highly expressed in SGC-7901 cells but negative in MKN-45 cells. For nude mice bearing GHR + SGC-7901 xenografts, the tumor volumes were significantly larger in low-dose rhGH group [(1.141 ± 0.234) cm 3] and high-dose rhGH group [(2.106 ± 0.260) cm 3] than in control group [(0.612 ± 0.156) cm 3] (P = 0.034, P = 0.001), and the high-dose rhGH group revealed greater effect (P = 0.043). Body weight was not significantly different among three groups. Compared with the control group, the mRNA expressions of tumor angiogenesis factor were significantly increased in low-does rhGH group, and the P values of GHR, Janus kinase 2, signal transducer and activator of transcription 3, vascular endothelial growth factor (VEGF), hypoxia-inducible factor-1α (HIF-1α), fibroblast growth factor, and matrix metalloproteinases-2 (MMP-2) was 0.001, 0.011, 0.042, 0.045, 0.040, 0.002, and 0.003, respectively; however, the high-does rhGH group did not show the greater effects. The protein expressions were significantly increased in low-does rhGH group, and the P value of phosphorylation-signal transducer and activator of transcription 3, signal transducer and activator of transcription 3, VEGF, HIF-1α, and MMP-2 was 0.015, 0.003, 0.010, 0.008, and 0.005, respectively; furthermore, the high-does group revealed the further greater effects, and the P value of VEGF, HIF-1α, and MMP-2 was 0.012, 0.025, and 0.046, respectively. On the contrary, for nude mice bearing GHR - MKN-45 xenografts, the body weights of low-dose rhGH group [(24.94 ± 0.517) g] and high-dose rhGH group [(26.97 ± 0.686) g] were significantly higher than that of control group [(22.78 ± 0.418) g] (P = 0.040, P = 0.012), while tumor growth as well as the expressions of mRNA and protein of tumor angiogenesis factor in tumor tissue were not significantly different. Conclusions: rhGH can promote tumor growth and up-regulate the expression of tumor angiogenesis factor in the GHR-highly-expressed SGC-7901 xenograft tumor model; However, such effects do not exist in GHR-negatively-expressed MKN-45 xenograft tumor model. The existence of GHR may be a key target by which rhGH influences the tumor growth and the expressions of tumor angiogenesis factor, which is probably achieved through Janus kinase 2-signal transducer and activator of transcription 3 activation.
Article
Growth hormone-releasing hormone (GHRH) is mostly thought to act by stimulating the production and release of growth hormone from the pituitary. However, this neuropeptide emerges as a rather pleiotropic hormone in view of the identification of various extrapituitary sources for GHRH production, as well as the demonstration of a direct action of GHRH on several tissues other than the pituitary. Non-pituitary GHRH has a wide spectrum of activity, exemplified by its ability to modulate cell proliferation, especially in malignant tissues, to regulate differentiation of some cell types, and to promote healing of skin wounds. These findings extend the role of GHRH and its analogs beyond its accepted regulation of somatotropic activity and indicate new possibilities for therapeutic intervention.
Article
Full-text available
Cachexia is a common manifestation of late stage malignancy and is characterized by anemia, anorexia, muscle wasting, loss of adipose tissue, and fatigue. Although cachexia is disabling and can diminish the life expectancy of cancer patients, there are still no effective therapies for this condition. We have examined the feasibility of using a myogenic plasmid to express growth hormone-releasing hormone (GHRH) in severely debilitated companion dogs with naturally occurring tumors. At a median of 16 days after intramuscular delivery of the plasmid, serum concentrations of insulin-like growth factor I (IGF-I), a measure of GHRH activity, were increased in 12 of 16 dogs (P < 0.01). These increases ranged from 21 to 120% (median, 49%) of the pretreatment values and were generally sustained or higher on the final evaluation. Anemia resolved posttreatment, as indicated by significant increases in mean red blood cell count, hematocrit, and hemoglobin concentrations, and there was also a significant rise in the percentage of circulating lymphocytes. Treated dogs maintained their weights over the 56-day study and did not show any adverse effects from the GHRH gene transfer. We conclude that intramuscular injection of a GHRH-expressing plasmid is both safe and capable of stimulating the release of growth hormone and IGF-I in large animals. The observed anabolic responses to a single dose of this therapy might be beneficial in patients with cancer-associated anemia and cachexia.
Article
The secretion of growth hormone (GH) is regulated through a complex neuroendocrine control system, especially by the functional interplay of two hypothalamic hypophysiotropic hormones, GH-releasing hormone (GHRH) and somatostatin (SS), exerting stimulatory and inhibitory influences, respectively, on the somatotrope. The two hypothalamic neurohormones are subject to modulation by a host of neurotransmitters, especially the noradrenergic and cholinergic ones and other hypothalamic neuropeptides, and are the final mediators of metabolic, endocrine, neural, and immune influences for the secretion of GH. Since the identification of the GHRH peptide, recombinant DNA procedures have been used to characterize the corresponding cDNA and to clone GHRH receptor isoforms in rodent and human pituitaries. Parallel to research into the effects of SS and its analogs on endocrine and exocrine secretions, investigations into their mechanism of action have led to the discovery of five separate SS receptor genes encoding a family of G protein-coupled SS receptors, which are widely expressed in the pituitary, brain, and the periphery, and to the synthesis of analogs with subtype specificity. Better understanding of the function of GHRH, SS, and their receptors and, hence, of neural regulation of GH secretion in health and disease has been achieved with the discovery of a new class of fairly specific, orally active, small peptides and their congeners, the GH-releasing peptides, acting on specific, ubiquitous seven-transmembrane domain receptors, whose natural ligands are not yet known.
Article
Nineteen human breast carcinoma cell lines have been established as continous cultures during the past 6 years in our laboratory. This preliminary report is designed to list the lines by their designated code numbers (MDA-MB) and present a brief summary of their morphological, cytogenetic and biochemical characteristics. Sixteen of our lines were obtained from pleural effusions, two from brain metastases, and one from pericardial fluid. All lines have been shown to be distinct entities and are uncontaminated by HeLa cells or each other. A lq marker chromosome is present in all but one of the lines examined.
Article
Purpose: To evaluate the tumor inhibitory activities of antagonists of growth hormone-releasing hormone (GH-RH) and vasoactive intestinal peptide (VIP) in UCI-107 human ovarian cancer model, and to investigate the role of the insulin-like growth factor (IGF) system in the response. Methods: In the present study we investigated the effects of GH-RH antagonist JV-1-36 and VIP antagonist JV-1-52, on the growth and tumorigenicity of UCI-107 ovarian cell carcinoma xenografted into nude mice. Studies on the effects of hGH-RH(1-29)NH2, IGF-I, IGF-II, JV-1-36, and JV-1-52 on the proliferation of UCI-107 cells cultured in vitro were also performed. Results: After 22 days of therapy with JV-1-36 or JV-1-52 at the dose of 20 microg/day, the final volume of UCI-107 tumors was significantly (P<0.05) decreased by 50.5% and 56%, respectively, compared to controls. The concentration of IGF-II in tumors was reduced by 66% in the JV-1-36-treated group and by 62% in the group given JV-1-52 (both P < 0.05). Exposure in vitro to 1 microM concentrations of JV-1-36 or JV-1-52 for 24 h decreased the tumorigenicity of UCI-107 cells in nude mice. All ten mice injected with cells treated with medium alone developed tumors within 23 days after cell inoculation, while only eight of ten and four of ten mice injected with cells exposed to JV-1-36 or JV-1-52, respectively, had tumors. In vitro exposure of UCI-107 cells to 5-35 ng/ml IGF-II produced a significant suppression in the rate of cell proliferation (P < 0.01). Conclusion: Our results suggest that GH-RH and VIP antagonists inhibit the growth of UCI-107 ovarian cell carcinoma by mechanisms that appear to involve direct effects on the cancer cells.
Article
Nineteen human breast carcinoma cell lines have been established as continuous cultures during the past 6 years in our laboratory. This preliminary report is designed to list the lines by their designated code numbers (MDA-MB) and present a brief summary of their morphological, cytogenetic and biochemical characteristics. Sixteen of our lines were obtained from pleural effusions, two from brain metastases, and one from pericardial fluid. All lines have been shown to be distinct entities and are uncontaminated by HeLa cells or each other. A lq marker chromosome is present in all but one of the lines examined.
Article
T cell maturation in an extrathymic environment has been studied using as a model the congenitally athymic nude mouse. Phenotypic analyses as a function of age were conducted on lymphocytes obtained from the spleens and lymph nodes of nude mice through use of mAb recognizing T cell surface markers and multiparameter flow cytometry. The data show that nude mice accumulate increasing numbers of lymphocytes bearing Thy-1, CD3, CD4, and CD8 with age characterized by a progression from heterogeneous dim to more homogeneous bright expression. In contrast, the expression of heat-stable Ag (HSA), a marker of immature thymocytes, decreases with age. By analogy to intrathymic maturation, spleens and lymph nodes in nude mice contain T cells defined as immature, transitional, and mature based on the expression of these markers. Although the proportion of CD4+ and CD8+ T cells associated with bright CD3 expression increases with age, at no age are significant numbers of CD4+8+ cells observed, in contrast to intrathymic T cell maturation. In addition to the frequently observed inversion in the ratio of CD4 to CD8, the CD8 T cell subpopulation in older nude mice contains mainly mature cells (CD8+, CD3+, HSA-) whereas only 50% of CD4+ T cells express the mature (CD4+, CD3+, HSA-) phenotype. At any age, the spectrum of phenotypes observed indicates that lymph nodes contain more mature T cells than spleen, suggesting a role for environmental Ag in driving extrathymic maturation, a process occurring most efficiently among CD8+ T cells. Because extrathymic maturation mirrors some but not all aspects of the intrathymic pathway, we propose that the nude mouse may be a useful model for further dissecting those interactions crucial to establishing the T cell repertoire in euthymic individuals as well as elucidating the contribution of extrathymically derived T cells to the peripheral immune system.
Article
Phenotypic analysis of lymphocytes that mature extrathymically in congenitally athymic nude mice has revealed a large population of CD3+ CD8+ T cells that express gamma/delta-TCR. In euthymic mice, significant numbers of cells with this phenotype are found only in the intestinal epithelium. Intestinal intraepithelial lymphocytes have been shown to be cytolytically active in vivo, as measured by the redirected lysis assay. In this communication, freshly harvested T cell subsets obtained from pooled nude mouse spleen and lymph nodes and separated by flow cytometric cell sorting were assayed for their ability to lyse FcR+ P815 targets in the presence of mAb to the epsilon-chain of the CD3 complex. CD8+, but not CD4+ or CD4- CD8-, T cells in nude mice were cytolytically active. CD8+ alpha/beta- and gamma/delta-TCR-bearing T cells from the spleen and lymph nodes of nude mice demonstrated similar cytolytic activity. No cytolytic activity of purified cell subsets was apparent in the absence of anti-CD3 mAb, even when NK-susceptible target cells were used. These data indicate that, in contrast to euthymic mice, a large proportion of CD8+ cells from the spleen and lymph nodes of nude mice are cytolytically active in vivo. In addition, these results suggest that the intestinal epithelium is not the only anatomical location where constitutively cytolytic CD8+ alpha/beta- or gamma/delta TCR-bearing T cells may be found.
The secretion of growth hormone (GH) is regulated through a complex neuroendocrine control system, especially by the functional interplay of two hypothalamic hypophysiotropic hormones, GH-releasing hormone (GHRH) and somatostatin (SS), exerting stimulatory and inhibitory influences, respectively, on the somatotrope. The two hypothalamic neurohormones are subject to modulation by a host of neurotransmitters, especially the noradrenergic and cholinergic ones and other hypothalamic neuropeptides, and are the final mediators of metabolic, endocrine, neural, and immune influences for the secretion of GH. Since the identification of the GHRH peptide, recombinant DNA procedures have been used to characterize the corresponding cDNA and to clone GHRH receptor isoforms in rodent and human pituitaries. Parallel to research into the effects of SS and its analogs on endocrine and exocrine secretions, investigations into their mechanism of action have led to the discovery of five separate SS receptor genes encoding a family of G protein-coupled SS receptors, which are widely expressed in the pituitary, brain, and the periphery, and to the synthesis of analogs with subtype specificity. Better understanding of the function of GHRH, SS, and their receptors and, hence, of neural regulation of GH secretion in health and disease has been achieved with the discovery of a new class of fairly specific, orally active, small peptides and their congeners, the GH-releasing peptides, acting on specific, ubiquitous seven-transmembrane domain receptors, whose natural ligands are not yet known.