ArticlePDF Available

The Kaposi's Sarcoma-Associated Herpesvirus G Protein-Coupled Receptor as a Therapeutic Target for the Treatment of Kaposi's Sarcoma

Authors:

Abstract and Figures

The Kaposi's sarcoma-associated herpesvirus (KSHV) encodes a G protein-coupled receptor (vGPCR) that has been implicated in the initiation of Kaposi's sarcoma, identifying vGPCR as an attractive target for preventing Kaposi's sarcoma. However, as only a fraction of cells in advanced Kaposi's sarcoma lesions express vGPCR, it is unclear whether this unique viral oncogene contributes to Kaposi's sarcoma progression. We therefore set out to determine whether the few cells that express vGPCR in established tumors represent an appropriate therapeutic target for the treatment of patients with preexisting Kaposi's sarcoma. To this end, we generated endothelial cell lines stably expressing vGPCR or key KSHV latently expressed proteins (vCyclin, vFlip, and LANA1). The endothelial cell line expressing vGPCR was rendered sensitive to treatment with the nucleoside analogue ganciclovir by using a bicistronic construct coexpressing the herpes simplex virus 1 thymidine kinase. S.c. injection into nude mice with mixed-cell populations formed tumors that approximate the ratio of vGPCR-expressing and KSHV latent gene-expressing cells. These mice were then treated with ganciclovir to specifically target only the vGPCR-expressing cells. Surprisingly, despite the expression of KSHV latent genes in the vast majority of tumor cells, specifically targeting only the few vGPCR-expressing cells in established tumors resulted in tumor regression. Moreover, we observed an increase in apoptosis of latent gene-expressing cells after the pharmacologic deletion of the vGPCR-expressing cells. These findings indicate that vGPCR may play a key role in Kaposi's sarcoma progression and provide experimental justification for developing molecular-based therapies specifically targeting vGPCR and its effectors for the treatment of Kaposi's sarcoma patients.
Content may be subject to copyright.
2006;66:168-174. Cancer Res
Silvia Montaner, Akrit Sodhi, Amanda K. Ramsdell, et al.
of Kaposi's Sarcoma
Coupled Receptor as a Therapeutic Target for the Treatment
Associated Herpesvirus G ProteinThe Kaposi's Sarcoma
Updated version
http://cancerres.aacrjournals.org/content/66/1/168
Access the most recent version of this article at:
Cited Articles
http://cancerres.aacrjournals.org/content/66/1/168.full.html#ref-list-1
This article cites by 18 articles, 11 of which you can access for free at:
Citing articles
http://cancerres.aacrjournals.org/content/66/1/168.full.html#related-urls
This article has been cited by 9 HighWire-hosted articles. Access the articles at:
E-mail alerts
related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions
.permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
Research.
on June 9, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
The Kaposi’s Sarcoma–Associated Herpesvirus G Protein–Coupled
Receptor as a Therapeutic Target for the Treatment of
Kaposi’s Sarcoma
Silvia Montaner,
1,2
Akrit Sodhi,
3,4
Amanda K. Ramsdell,
3
Daniel Martin,
3
Jiadi Hu,
1
Earl T. Sawai,
4
and J. Silvio Gutkind
3
1
Department of Diagnostic Sciences and Pathology;
2
Greenebaum Cancer Center, University of Maryland, Baltimore, Maryland;
3
Oral and
Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland; and
4
Department of Comp arative Patholog y, University of California at Davis, Davis, California
Abstract
The Kaposi’s sarcoma–associated herpesvirus (KSHV) encodes
a G protein–coupled receptor (vGPCR) that has been implicat-
ed in the initiation of Kaposi’s sarcoma, identifying vGPCR as
an attractive target for preventing Kaposi’s sarcoma. However,
as only a fraction of cells in advanced Kaposi’s sarcoma lesions
express vGPCR, it is unclear whether this unique viral oncogene
contributes to Kaposi’s sarcoma progression. We therefore set
out to determine whether the few cells that express vGPCR in
established tumors represent an appropr iat e therapeutic
target for the treatment of patients with preexisting Kaposi’s
sarcoma. To this end, we generated endothelial cell lines stably
expressing vGPCR or key KSHV latently expressed proteins
(vCyclin, vFlip, and LANA1). The endothelial cell line expressing
vGPCR was rendered sensitive to treatment with the nucleoside
analogue ganciclovir by using a bicistronic construct coex-
pressing the herpes simplex virus 1 thymidine kinase. S.c.
injection into nude mice with mixed-cell populations formed
tumors that approximate the ratio of vGPCR-expressing and
KSHV lat ent gene-expressing cells. These mice were then
treated with ganciclovir to specifically target only the vGPCR-
expressing cells. Surprisingly, despite the expression of KSHV
latent genes in the vast majority of tumor cells, specifically
targeting only the few vGPCR-expressing cells in established
tumors resulted in tumor regression. Moreover, we observed an
increase in apoptosis of latent gene-expressing cells after the
pharmacologic deletion of the vGPCR-expressing cells. These
findings indicate that vGPCR may play a key role in Kaposi’s
sarcoma progression and provide experimental justification
for developing molecular-based therapies specifically targeting
vGPCR and its effectors for the treatment of Kaposi’s sarcoma
patients. (Cancer Res 2006; 66(1): 168-74)
Introduction
The Kaposi’s sarcoma–associated herpesvirus (KSHV or human
herpesvirus-8), the etiologic agent for Kaposi’s sarcoma (1), encodes
an arsenal of putative oncogenes that harbor transforming potential
in vitro (2). Several of these candidate oncogenes are latent genes,
expressed in the majority of Kaposi’s sarcoma tumor or spindle cells,
and are therefore thought to play an important role in Kaposi’s
sarcomagenesis. However, accumulating evidence suggests that the
expression of latent proteins may not be sufficient to initiate Kaposi’s
sarcoma. Conversely, expression of the KSHV-encoded viral G
protein–coupled receptor (vGPCR) as a transgene or by endothelial-
specific retroviral infection is sufficient to induce Kaposi’s sarcoma–
like tumors in mice (3–5), implicating this viral oncogene in the
initiation of Kaposi’s sarcoma (6, 7). This suggests that therapies
targeting this receptor or its downstream effectors (8) may be an
effective approach to prevent the formation of new Kaposi’s sarcoma
lesions in KSHV-infected patients. However, immunohistochemical
examination of biopsies from patients with established Kaposi’s
sarcoma lesions has revealed that expression of vGPCR is detected in
only a fraction of tumor cells (9), raising whether vGPCR would be an
appropriate therapeutic target in patients with preexisting Kaposi’s
sarcoma. Indeed, as the majority of Kaposi’s sarcoma tumor cells
primarily express latent genes, these viral gene products would be
expected to represent more suitable targets for the treatment of
Kaposi’s sarcoma (10). Unfortunately, due the lack of suitable animal
models to study Kaposi’s sarcoma promotion in vivo, it has been
difficult to assess the relative contribution of, and complex interplay
among, these genes to Kaposi’s sarcomagenesis.
We show here that coinjection of endothelial cells expressing
latent genes with a few endothelial cells stably expressing vGPCR—
at a ratio that approximates the proportion of vGPCR-expressing
and latent gene-expressing tumor cells found in human Kaposi’s
sarcoma—synergistically enhances latent gene-driven tumorigenic-
ity. Indeed, immunohistochemical analysis of tumors formed using
this system revealed that the majority of cells expressed the latent
genes, whereas vGPCR is expressed in only rare tumor cells,
analogous to human Kaposi’s sarcoma. Surpris ingly, however,
pharmacologic deletion of these rare vGPCR-expressing cells is
sufficient to cause tumor regression. We observed an increase in
apoptosis of tumor cells as a consequence of the pharmacologic
deletion of the vGPCR-expressing cells. Furthermore, although
expression of KSHV latent genes was still detected in few surviving
cells, these residual cells lost their tumorigenic potential in the
absence of the paracrine secretions from vGPCR -expressing cells.
Thus, using a Kaposi’s sarcoma model system that can recapitulate
the complex interplay between lytic and latent infected cells, we
provide evidence for the feasibility of specifically targeting vGPCR-
expressing cells as a therapeutic approach for the treatment of
patients with Kaposi’s sarcoma.
Materials and Methods
Expression plasmids and reagents. The expression plasmids for
vGPCR, vGPCR (R143A), vCyclin, vFlip, vCyclin/vFlip, Kaposin, vBcl2, vIRF1,
vIL6, and enhanced green flu orescent protein have been described
Requests for reprints: J. Silvio Gutkind, Oral and Pharyngeal Cancer Branch,
National Institute of Dental and Craniofacial Research, NIH, 30 Convent Drive,
Building 30, Room 211, Bethesda, MD 20892-4330. Phone: 301-496-6259; Fax: 301-402-
0823; E-mail: sg39v@nih.gov or Silvia Montaner; E-mail: SMontaner@umaryland.edu.
I2006 American Association for Cancer Research.
doi:10.1158/0008-5472.CAN-05-1026
Cancer Res 2006; 66: (1). January 1, 2006
168
www.aacrjournals.org
Research Article
Research.
on June 9, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
previously (3, 11). The expression plasmid for LANA1 was generously
provided by Dr. C. Boshoff (University College of London, London, United
Kingdom) and subsequently subcloned into the pCEFL euk ar yotic
expression vector. The expression plasmid for K1 was generously provided
by Dr. J. Jung (Harvard University, Cambridge, MA) and subsequently
subcloned into the pCEFL eukaryotic expression vector. The bicistronic
construct vGPCR-herpes simplex virus 1 thymidine kinase (HSV1-TK) was
obtained by first inserting vGPCR and then HSV1-TK into pCEFL internal
ribosome entry site (12). Ganciclovir was purchased from EMD Biosciences
(San Diego, CA). For in vitro studies, this compound was reconstituted in
PBS as a 50 mmol/L stock solution, which was further diluted to the
working concentration (0-1,000 nmol/L) in culture medium.
Cell lines and transfections. SV40 large T-antigen immortalized murine
endothelial cells (SVEC) were grown in DMEM supplemented with 10% fetal
bovine serum and 1% penicillin/streptomycin. Transfection was done using
Fugene reagent (Roche Applied Science, Indianapolis, IN) according to the
manufacturer’s protocol. Stable SVEC cell lines were obtained by stable
transfection of the corresponding pCEFL-derived plasmids as described
previously (3).
ELISA. Conditioned medium from EC-vGPCR, EC-R143A, or SVEC
parental cell lines was prepared as described previously (13). Assays for
cytokine secretion were done by Pierce Biotechnology (Rockford, IL).
Assessment of [
3
H]thymidine incorporation. Assessment of cell
proliferation by uptake of [
3
H]thymidine (ICN Pharmaceuticals, Inc., Costa
Mesa, CA) was essentially as described previously (13).
Establishment and treatment of tumor allografts in athymic nu/nu
mice. All animal studies were carried out using the appropriate NIH
animal care and user protocol. SVECs (10
6
cells) stably expressing vGPCR
were used to induce allografts in 6-week-old athymic (nu/nu) nude
females mice as described (3). For drug treatment, tumor-bearing
animals were randomly grouped (control, n = 10; test, n = 10) and
treated with ganciclovir (50 mg/kg/d) or an equal volume of diluent
(PBS). Treatment schedule was a single injection per animal given i.p.
for 4 consecutive days. For analysis, tumor weight was determined
as described previously (3), whereby tumor volume (LW
2
/ 2, where
L and W represent the longest length and shortest width of the tumor,
respectively) was converted to weight. Results of animal experiments
were expressed as mean F SE. At the end of the study period, animals
were euthanized for tissue retrieval, which was fixed (4% paraformalde-
hyde overnight before processing for paraffin embedding) for immuno-
histochemical analysis.
Immunohistochemistry. Tissues were fixed in 4% paraformaldehyde-1
PBS for 36 hours, transferred to 70% ethanol/PBS, and embedded in
paraffin. Immunohistochemical analysis of tissues has been described
previously (3).
Results
KSHV vGPCR potently renders expressing endothelial cells
tumorigenic. To establish a mouse model in which we could
study the contribution of candidate KSHV oncogenes to Kaposi’s
sarcomagenesis, we took advantage of the availability of non-
tumorigenic SVECs. When injected s.c. into nude mice, 10
6
SVECs are unable to form tumors up to 6 months after injection
(data not shown) but are rendered tumorigenic by stable
introduction of an oncogene, enabling rapid in vivo screening
for tumorigenic proteins. We generated SVEC lines stably
expressing key KSHV latent genes vCyclin (EC-vCyclin), vFlip
(EC-vFlip), LANA1 (EC-LANA1), and Kaposin (EC-Kaposin) or
lytic genes K1 (EC-K1), vBcl2 (EC-vBcl2), IRF1 (EC-IRF1), vIL6
(EC-vIL6), or vGPCR (EC-vGPCR). We first confirmed the
expression of these genes by Western blot analysis (data not
shown) before examining their tumorigenic potential in vivo.
Despite that many of these genes may harbor transforming
capability in vitro (2), only endothelial cells expressing the lytic
gene, vGPCR (EC-vGPCR), consistently formed tumors when
injected s.c. into nude mice (Fig. 1A). To assess whether latent
genes could cooperate in endothelial cell transformation, we also
prepared endothelial cell lines stably coexpressing vCyclin and
vFlip (EC-vCyclin/vFlip) or vCyclin, vFlip, and LANA1 (EC-
vCyclin/vFlip/LANA1). As shown in Fig. 1B, EC-vCyclin/vFlip
cells were only weakly tumorigenic. Surprisingly, addition of a
third KSHV latent gene, LANA1 (EC-vCyclin/vFlip/LANA1), failed
to further enhance the tumorigenic potential of expressing
endothelial cells (Fig. 1B), suggesting that cooperation among
latent genes may not be sufficient either to explain the
tumorigenic potential of KSHV infected cells. This is in striking
contrast to the potent tumorigenic potential of endothelial cells
expressing the KSH V lytic gene, vGPCR (Fig. 1A and B).
Collectively, prior studies (3–7) and these results suggest that
vGPCR may be one of the most potent oncogene encoded by
KSHV.
Figure 1. vGPCR is sufficient to render expressing endothelial cells oncogenic.
A, weight of tumors formed from s.c. injection of nude mice with 10
6
SVECs
stably expressing vCyclin (EC-vCyclin), vFlip (EC-vFlip), LANA1 (EC-LANA1),
Kaposin (EC-Kaposin), K1 (EC-K1), vBcl2 (EC-vBcl2), IRF1 (EC-IRF1), vIL6
(EC-vIL6), or vGPCR (EC-vGPCR). SVEC parental cells were used as a control.
B, weight of tumors formed from s.c. injection of nude mice with endothelial cell
lines stably expressing vCyclin and vFlip (EC-vCyclin/vFlip), vCyclin, vFlip, and
LANA1 (EC-vCyclin/vFlip/LANA1), vGPCR (EC-vGPCR), or the inactive vGPCR
R143A mutant (EC-R143A). SVEC parental cells were used as a control.
Tumor weight was estimated as described in Materials and Methods.
KSHV vGPCR as Therapeutic Target for Kaposi’s Sarcoma
www.aacrjournals.org
169
Cancer Res 2006; 66: (1). January 1, 2006
Research.
on June 9, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Figure 2. vGPCR promotes the tumorigenic potential of a combination of KSHV latent genes in a mouse allograft model for Kaposi’s sarcoma. A, secretion of
key Kaposi’s sarcoma cytokines, IL-6, murine IL-8/Gro-a homologue (KC), SDF-1, and IL-2 by endothelial cells expressing vGPCR (EC-vGPCR) or its inactive mutant,
R143A (EC-R143A). Columns, mean fold induction with respect to secretion by control (SVEC) cells; bars, SD. B, proliferation of EC-vCyclin/vFlip treated with
conditioned medium of cultured endothelial cells expressing vGPCR (EC-vGPCR) or its inactive mutant, R143A (EC-R143A), determined by the incorporation of
[
3
H]thymidine. Columns, mean fold induction with respect to results obtained using conditioned medium from control (SVEC) cells; bars, SD. C, schematic
representation of the Kaposi’s sarcoma allograft model in which mixed-cell populations of lytic (vGPCR) and latent gene-expressing endothelial are coinjected into nude
mice in a ratio that approximates their expression pattern in human Kaposi’s sarcoma. D, weight of tumors formed from coinjection with mixed-cell populations of these
cell lines. E, immunohistochemical analysis of tumors formed using this model system for Kaposi’s sarcoma showing rare tumor cells expressing vGPCR (left),
with the majority of cells expressing latent genes (right ).
Figure 3. vGPCR-expressing endothelial
cells are rendered sensitive to ganciclovir
treatment by coexpressing HSV1-TK.
A, schematic representation of an
endothelial cell line stably expressing a
bicistronic construct encoding both vGPCR
and HSV1-TK (EC-vGPCR/TK), rendering
this cell line sensitive to treatment with the
nucleoside analogue, ganciclovir (GAN ).
B, increasing doses (0.1-1,000 Amol/L)
of ganciclovir inhibits cell proliferation
of EC-vGPCR/TK but not EC-vGPCR
as determined by incorporation of
[
3
H]thymidine. C, extended treatment of
EC-vGPCR/TK with 10 Amol/L ganciclovir
induced cell death in f100% of treated
cells within 4 days. Cells treated with
vehicle control are indicated. EC-vGPCR
was completely insensitive to this dose of
ganciclovir, reaching confluence within
3 days.
Cancer Research
Cancer Res 2006; 66: (1). January 1, 2006
170
www.aacrjournals.org
Research.
on June 9, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
A mouse allograft model for Kaposi’s sarcoma. vGPCR is a
constitutively active GPCR closely related to the mammalian
cytokine receptor, CXCR2. Prior work suggests that the potent
oncogenic potential of vGPCR may, in part, be facilitated by the
paracrine secretions of vGPCR-expressing cells (7, 14–16). Indeed,
conditioned medium from EC-vGPCR cells showed elevated levels
of key Kaposi’s sarcoma cytokines, such as interleukin (IL)-6,
murine IL-8 (KC), and stromal cell–derived factor-1 (SDF-1;
Fig. 2A). To determine if these paracrine secretions could
contribute to the proliferation of latently infected endothelial cells,
we treated EC-vCyclin/vFlip cells with supernatants obtained from
cultured EC -vGPCR. Surprisingly, conditioned medium from
vGPCR-expressing cells promoted the proliferation of endothelial
cells expressing KSHV latent genes (Fig. 2B). These results
suggested that the paracrine secretions from vGPCR-expressing
cells may also promote the tumorigenic potential of latently
infected spindle cells.
To study the complex interplay among lytic and latent gene-
expressing endothelial cells in Kaposi’s sarcomagenesis in vivo,
we s.c. injected mixed-cell populations of EC-vGPCR along with
EC-vCyclin/vFlip into nude mice (Fig. 2C) in a ratio that
approximates the proportion of vGPCR-expressing and latent
gene-expressing tumor cells found in human Kaposi’s sarcoma
(3). Coinjection of EC-vCyclin/vFlip (10
6
cells) with a few
endothelial cells stably expressing vGPCR (EC-vGPCR; 10
5
cells)
synergistically enhanced EC-vCyclin/vFlip tumorigenicity (Fig. 2D).
Coexpression of a third latent gene, LANA1, failed to further
promote tumor growth (data not shown). Immunohistochemical
analysis of tumors formed using this allograft model for Kaposi’s
sarcoma revealed that vGPCR -expressing cells promote the
tumoral growth of EC-vCyclin/vFlip, as only few tumor cells
expressed vGPCR , whereas the majority of cells expressed
these latent genes (Fig. 2E), similar to human Kaposi’s sarcoma
lesions (9).
vGPCR-expressing endothelial cells can be rendered sensi-
tive to ganciclovir treatment by coexpressing HSV1-TK. These
results implicate vGPCR in Kaposi’s sarcoma tumor development
through the secretion of key Kaposi’s sarcoma cytokines and
growth factors, raising whether specifically targeting the vGPCR-
expressing cells, the reby quenchi ng the secretion of these
paracrine growth factors, could be an effective approach for
treating established Kaposi’s sarcoma lesions. To address this
question, we set out to establish a vGPCR-expressing cell line
that could be selectively and specifically targeted in vivo. To this
end, we took advantage of the fact that the HSV1-TK renders
expressing cells exquisitely sensitive to treatment with acyclic
guanidine analogues (e.g., ganciclovir; ref. 17). We employed
a bicistronic construct encoding both vGPCR and HSV1-TK
(Fig. 3A) to ensure that all cells expressing vGPCR coexpressed
HSV1-TK and were therefore rendered sensitive to ganciclovir
treatment. We first confirmed coexpression of both genes in
transiently transfected cells (data not shown) before generating
an endothelial cell line stably expressing this bicistronic
construct (EC-vGP CR/TK). We next tested the effects of
increasing doses of ganciclovir on the EC-vGPCR/TK cell line.
Proliferation of EC-vGPCR/TK, as determined by incorporation
of [
3
H]thymidine, was similar to that of EC-vGPCR during the
same time interval (data not shown), suggesting that coex-
pression of the HSV1-TK in the absence of ganciclovir did not
affect the proliferative potential of EC-vGPCR/TK. Conversely,
proliferation of EC-vGPCR/TK in the presence of ganciclovir was
dramatically reduced (Fig. 3B), with an IC
50
of f1 Amol/L
compared with f1,000 Amol/L for EC-vGPCR. Extended
treatment of EC-vGPCR/TK with 10 Amol/L ganciclovir induced
cell death in f100% of treated cells within 4 days (Fig. 3C);
terminal deoxynucleotidyl transferase–mediated dUTP nick end
labeling (TUNEL) assays revealed that cell death was not
through apoptosis (data not shown). Conversely, EC-vGPCR cells
Figure 4. vGPCR/TK-expressing endothelial cells are sensitive to ganciclovir
treatment in vivo . A, established tumors formed from s.c. injection of nude mice
with EC-vGPCR or EC-vGPCR/TK were treated with PBS (cont ) or a tolerable
dose of ganciclovir (50 mg/kg) for 4 consecutive days when they reached f50
mg. Tumor weight was calculated (LW
2
/ 2, where L and W represent the longest
length and shortest width of the tumor, respectively) and recorded biweekly.
B, immunohistochemical analysis of EC-vGPCR or EC-vGPCR/TK tumors
treated with PBS (control) or ganciclovir revealing a complete loss of all
vGPCR-expressing cells in EC-vGPCR/TK tumors 24 hours after completion
of the 4-day treatment cycle.
KSHV vGPCR as Therapeutic Target for Kaposi’s Sarcoma
www.aacrjournals.org
171
Cancer Res 2006; 66: (1). January 1, 2006
Research.
on June 9, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
were completely insensitive to this dose of ganciclovir and
continued to proliferate until reaching confluence (Fig. 3C).
Collectively, these results suggest that the EC-vGPCR/TK cell line
is specifically and exquisitely sensitive to cell death in the
presence of ganciclovir.
vGPCR/TK-expressing endothelial cells are sensitiv e to
ganciclovir treatment in vivo. We next set out to determine if
EC-vGPCR/TK was sensitive to ganciclovir treatment in vivo.To
this end, we established tumor allografts by injecting EC-vGPCR/
TK s.c. into nude mice. Tumors formed from EC-vGPCR/TK grew
rapidly and were similar in size to those formed from EC-vGP CR
during the same time interval (Fig. 4A), suggesting that coex-
pression of the HSV1-TK in the absence of ganciclovir did not
affect the ability of vGPCR-expressing cells to induce tumors.
However, treatment of animals with established EC-vGPCR/TK
tumors (100 mg) with a tolerable dose of ganciclovir (50 mg/kg/d;
ref. 18) for 4 consecutive days completely abolished tumor growth
and induced tumor regression (Fig. 4A), with only residual scar
tissue remaining up to 2 months after treatment (data not shown).
Conversely, EC-vGPCR formed tumors in the presence or absence
of ganciclovir, confirming the specificity of the targeted cells
in vivo. Immunohistochemical analysis of tumors formed from
EC-vGPCR/TK and treated for 4 consecutive days with ganciclovir
revealed a complete loss of all vGPCR-expressing cells within 1 day
of the completion of the treatment cycle (Fig. 4B). Conversely,
vGPCR-expressing cells were readily detected in tumors formed
from EC-vGPCR and treated with ganciclovir (Fig. 4B). Collectively,
these results show that coexpression of HSV1-TK in vGPCR/TK-
expressing cells renders these cells sensitive to treatment with
ganciclovir in vivo.
Selectively targeting only rare vGPCR-expressing tumor
cells in mixed-cell tumors composed primarily of cells
expressing latent KSHV genes induces tumor regression. We
next set out to determine if targeting the few vGPCR-expressing
cells in a tumor formed from mixed-cell populations of
EC-vGPCR/TK with EC-vCyclin/vFlip could affect the growth of
tumors formed primarily from cells expressing the KSHV latent
genes, vCyclin and vFlip. To this end , we coinjected 10
6
EC-vCyclin/vFlip cells with a smaller number (10
5
) of endothelial
cells stably expressing vGPCR and HSV1-TK (EC-vGPCR/TK;
Fig. 5A). Similar to the EC-vGPCR cell line, EC-vGPCR/TK
synergistically enhanced EC-vCyclin/vFlip tumorigenicity (Fig. 5B).
Figure 5. Selectively targeting only rare vGPCR-expressing tumor cells is sufficient to induce tumor regression. A, schematic representation of experiment in
which tumors formed from the s.c. injection of 10
5
EC-vGPCR/TK cells with 10
6
EC-vCyclin/vFlip cells are treated with ganciclovir to specifically target only the
vGPCR-expressing cells. B, established tumors arising from mixed-cell populations of 10
5
EC-vGPCR/TK cells with 10
6
EC-vCyclin/vFlip cells treated with PBS
(control) or ganciclovir (50 mg/kg) for 4 consecutive days when they reached f50 mg. Tumor weight was calculated (LW
2
/ 2, where L and W represent the longest
length and shortest width of the tumor, respectively) and recorded biweekly. C, size of tumors arising from mixed-cell populations of 10
5
EC-vGPCR or 10
5
EC-vGPCR/TK cells with 10
6
EC-vCyclin/vFlip 1 week after treatment with ganciclovir (1 wk Post-Tx ). Columns, mean fold induction with respect to tumor size before
treatment; bars, SD. D, representative TUNEL analysis showing an increase in apoptotic cells (*) within tumors generated by the indicated mixed-cell populations
1 week after treatment with ganciclovir. Original magnification,
20. E, immunohistochemical staining showing expression of KSHV latent genes within these tumors
1 week after treatment with ganciclovir. Original magnification,
40.
Cancer Research
Cancer Res 2006; 66: (1). January 1, 2006
172
www.aacrjournals.org
Research.
on June 9, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Surprisingly, although only few of the tumor cells expressed
vGPCR, treatment of animals with established tumors with
ganciclovir (50 mg/kg) for 4 consecutive days induced tumor
regression (Fig. 5B) and sustained inhibition of tumor growth for
up to 4 weeks following treatment (data not shown). In contrast,
the small tumors formed from EC-vCyclin/vFlip alone were
unaffected by treatment with ganciclovir (data not shown).
Furthermore, the administration of ganciclovir did not prevent
the rapid growth of tumors arising from EC-vCyclin/vFlip
cells mixed with EC-vGPCR, which served as a specificity control
(Fig. 5C). Immunohistochemical analysis of tumors 1 week after
treatment revealed an increase in apoptosis in the remaining
tumor cells in tumors that included EC-vGPCR/TK cells (Fig. 5D).
To further investigate the effect of specifically targeting only the
rare vGPCR-expressing cells on tumor cells expressing KSHV
latent genes, we examined tumors 1 week after treatment of
animals with 4 consecutive days of ganciclovir, after which
EC-vGPCR/TK cells could no longer be detected (Fig. 3 B; data not
shown). Surprisingly, immunohistochemical staining indicated
that although ganciclovir treatment halted tumor growth a
reduced number of cells expressing KSHV latent genes could be
detected (Fig. 5E), suggesting that the remaining cells expressing
KSHV latent genes are not sufficient for tumor growth in the
absence of the paracrine secretions from vGPCR -expressing cells.
These results suggest that established Kaposi’s sarcoma tumors
may be dependent on the presence of the rare vGPCR-expressing
cells for tumor growth.
Discussion
Despite over a century since its initial description, Kaposi’s
sarcoma remains a poorly understood disease. The recent discovery
of KSHV as the viral etiologic agent of Kaposi’s sarcoma has exposed
many potential therapeutic targets. Among all candidate oncogenes
encoded by KSHV, only vGPCR has been thus far shown to induce
Kaposi’s sarcoma–like lesions in several independent transgenic
animal models (3–5). Indeed, emerging evidence suggests that
dysregulated expression of this potent oncogene in nonlytic cells
may represent an early event initiating Kaposi’s sarcomagenesis (6).
Nonetheless, as expression of vGPCR is associated with only a
subpopulation of spindle cells in Kaposi’s sarcoma animal models
and in human Kaposi’s sarcoma, it is possible that the expression of
this receptor may create an environ ment permissive for the
subsequent tumor development driven by other KSHV survival
(latent) genes, after which receptor expression is no longer
necessary. This ‘‘hit-and-run’’ mechanism would imply that although
vGPCR could be an attractive therapeutic target in preventing the
initiation of Kaposi’s sarcoma it might not be an appropriate target
for the treatment of established Kaposi’s sarcoma lesions.
Surprisingly, however, using a Kaposi’s sarcoma allograft
mouse model and a novel approach to specifically eliminate
pharmacologically all vGPCR -expressing c ells, we obtained
evidence here that the paracrine secretions from the few
vGPCR-expressing cells in established Kaposi’s sarcoma lesions
may still be required to promote growth of established Kaposi’s
sarcoma lesions. Of note, the role of vGPCR-expressing cells in
paracrine-driven tumorigenesis is not without precedent. A
similar function has been attributed previously to Reed-Sternberg
cells in Hodgkin’s lymphoma. Thus, tumor cells expressing
vGPCR, although rare, might ser ve as vulnerable targets in
established Kaposi’s sarcoma lesions. Indeed, as cellular GPCRs
are the target of f60% of all current pharmaceutical drugs,
vGPCR and its downstream effectors (11–16, 19, 20) represent
attractive candidates for the development of novel therapies for
Kaposi’s sarcoma.
Of note, despite evidence of an increased rate of apoptosis in
mixed-cell tumors in which vGPCR-expressing cells have been
pharmacologically removed, few tumor cells still persisted after
ganciclovir treatment, which expressed vCyclin and vFlip. These
cells may have been protected from apoptosis by the prosurvival
effect of these KSHV latent genes. Nonetheless, these remaining
cells did not regrow tumors even after prolonged observation
(2 months). Collectively, these results, along with prior work,
suggest a model in which vGPCR plays a key role in the early
events of Kaposi’s sarcomagenesis by triggering endothelial cell
transformation and promoting the subsequent tumoral growth of
cells expressing KSHV latent genes. Ultimately, a collaborative
approach to Kaposi’s sarcoma treatment in which antiviral and
antiangiogenic therapies are combined with gene product-
targeted therapies directed against key KSHV latent and lytic
genes may ultimately prove to be the most effective therapeutic
strategy for the treatment of patients suffering from this still
enigmatic and disabling disease.
Acknowledgments
Received 3/25/2005; revised 9/5/2005; accepted 10/18/2005.
Grant support: In part by the Intramural Research Program of the National
Institutes of Health, NIDCR.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
KSHV vGPCR as Therapeutic Target for Kaposi’s Sarcoma
www.aacrjournals.org
173
Cancer Res 2006; 66: (1). January 1, 2006
References
1. Chang Y, C esarman E, Pessin MS, et al. Identificati on
of herpesvirus-like DNA sequences in AIDS-associated
Kaposi’s sarcoma. Science 1994;266:1865–9.
2. Boshoff C, Chang Y. Kaposi’s sarcoma-associated
herpesvirus: a new DNA tumor virus. Annu Rev Med
2001;52:453–70.
3. Montaner S, Sodhi A, Molinolo A, et al. Endothelial
infection with KSHV genes in vivo reveals that vGPCR
initiates Kaposi’s sarcomagenesis and can promote the
tumorigenic potential of viral latent genes. Cancer Cell
2003;3:23–36.
4. Yang TY, Chen SC, Leach MW, et al. Transgenic
expressi on of the chemokine receptor encoded by
human herpesvirus 8 induces an angioproliferative
disease resembling Kaposi’s sarcoma. J Exp Med 2000;
191:445–54.
5. Guo HG, Sadowska M, Reid W, Tschachler E, Hayward
G, Reitz M. Kaposi’s sarcoma-like tumors in a human
herpesvirus 8 ORF74 transgenic mouse. J Virol 2004;77:
2631–9.
6. Sodhi A, Montaner S, Gutkind JS. Does dysregulated
expression of a deregulated viral GPCR trigger Kaposi’s
sarcomagenesis? FASEB J 2004;18:422–7.
7. Cesarman E, Mesri EA, Gershengorn MC. Viral G
protein-coupled receptor and Kaposi’s sarcoma: a
model of paracrine neoplasia? J Exp Med 2000;191:
417–22.
8. Sodhi A, Montaner S, Gutkind JS. Viral hijacking of G-
protein-coupled-receptor signalling networks. Nat Rev
Mol Cell Biol 2004;5:998–1012.
9. Chiou CJ, Poole LJ, Kim PS, et al. Patterns of gene
expression and a transactivation function exhibited by
the vGCR (ORF74) ch emokine receptor protein of
Kaposi’s sarcoma-associated herpesvirus. J Virol 2002;
76:3421–39.
10. Staudt MR, Dittmer DP. Viral latent proteins as
targets for Kaposi’s sarcoma and Kaposi’s sarcoma-
associated herpesvirus (KSHV/HHV-8) induced lym-
phoma. Curr Drug Targets Infect Disord 2003;3:
129–35.
11. Sodhi A, Montaner S, Patel V, et al. Akt plays a central
role in sarcomagenesis induced by Kaposi’s sarcoma
herpesvirus-encoded G protein-coupled receptor. Proc
Natl Acad Sci U S A 1998;101:4821–6.
12. Montaner S, Sodhi A, Servitja JM, et al. The small
GTPase Rac1 links the Kaposi sarcoma -associated
Research.
on June 9, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
Cancer Research
Cancer Res 2006; 66: (1). January 1, 2006
174
www.aacrjournals.org
herpesvirus vGPCR to cytokine secretion and paracrine
neoplasia. Blood 2004;104:2903–11.
13. Montaner S, Sodhi A, Pece S, Mesri EA, Gutkind JS.
The Kaposi’s sarcoma-associated herpesvirus G protein-
coupled receptor promotes endothelial cell survival
through the activation of Akt/protein kinase B. Cancer
Res 2001;61:2641–8.
14. Pati S, Cavrois M, Guo HG, et al. Activation of NF-nB
by the human herpesvirus 8 chemokine receptor ORF74:
evidence for a paracrine model of Kaposi’s sarcoma
pathogenesis. J Virol 2001;75:8660–73.
15. Bais C, Santomasso B, Coso O, et al. G-protein-
coupled receptor of Kaposi’s sarcoma-associated her-
pesvirus is a viral oncogene and angiogenesis activator.
Nature 1998;391:86–9.
16. Sodhi A, Montaner S, Patel V, et al. The Kaposi’s
sarcoma-associated herpes virus G protein-coupled
receptor up-regulates vascular endothelial growth
factor expression and secretion through mitogen-
activated protein kinase and p38 pathways acting on
hypoxia-inducible factor 1a. Cancer Res 2000;60:
4873–80.
17. Fillat C, Carrio M, Cascante A, Sangro B. Suicide
gene therapy mediated by the herpes simplex vir us
thymidine kinase gene/ganciclovir system: fifteen years
of application. Curr Gene Ther 2003;3:13–26.
18. Hemminki A, Zinn KR, Liu B, et al. In vivo
molecular chemotherapy and noninvasive imaging with
an infectivity-enhanced adenovirus. J Natl Cancer Inst
2002;94:741–9.
19. Bais C, Van Geelen A, Eroles P, et al. Kaposi’s
sarcoma associated herpesvirus G protein-coupled
receptor immortalizes human endothelial cells by
activation of the VEGF receptor-2/KDR. Cancer Cell
2003;3:131–43.
20. Couty JP, Geras-Raaka E, Weksler BB, Gershengorn
MC. Kaposi’s sarcoma-associated herpesvirus G protein-
coupled receptor signals through multiple pathways in
endothelial cells. J Biol Chem 2001;276:33805–11.
Research.
on June 9, 2013. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from
... Neoplasms caused by the human herpesvirus-8 (HHV-8) or Kaposi's sarcoma-associated herpesvirus (KSHV) are characterized by angiogenesis and the proliferation of spindle cells, which have qualities of activated endothelial cells [5][6][7][8]. Within the viral genome, the viral G protein-coupled receptor (vGPCR) drives oncogenesis and angiogenesis martin [5,[8][9][10][11]; its persistent expression is necessary for tumor transformation and maintenance [9]. Although the incidence of AIDS-associated Kaposi's sarcoma has decreased markedly since the widespread implementation of highly active antiretroviral therapy (HAART), a significant percentage of patients with this condition never achieve full remission [6,12,13]. ...
... Neoplasms caused by the human herpesvirus-8 (HHV-8) or Kaposi's sarcoma-associated herpesvirus (KSHV) are characterized by angiogenesis and the proliferation of spindle cells, which have qualities of activated endothelial cells [5][6][7][8]. Within the viral genome, the viral G protein-coupled receptor (vGPCR) drives oncogenesis and angiogenesis martin [5,[8][9][10][11]; its persistent expression is necessary for tumor transformation and maintenance [9]. Although the incidence of AIDS-associated Kaposi's sarcoma has decreased markedly since the widespread implementation of highly active antiretroviral therapy (HAART), a significant percentage of patients with this condition never achieve full remission [6,12,13]. ...
... ROS levels have become relevant in the oncogenesis process. It has been reported that endothelial cells expressing vGPCR have high levels of ROS, which are important to stimulate angiogenesis [9]; however, a high concentration of ROS is cytotoxic in healthy cells. Since it has been shown that MNPs induce ROS production and consequently cytotoxicity [37], we investigated whether the range of MNPs concentration employed in this work increases the ROS levels in vGPCR cells. ...
Article
Full-text available
Magnetic nanosystems represent promising alternatives to the traditional diagnostic and treatment procedures available for different pathologies. In this work, a series of biological tests are proposed, aiming to validate a magnetic nanoplatform for Kaposi’s sarcoma treatment. The selected nanosystems were polyethylene glycol-coated iron oxide nanoparticles (MAG.PEG), which were prepared by the hydrothermal method. Physicochemical characterization was performed to verify their suitable physicochemical properties to be administered in vivo. Exhaustive biological assays were conducted, aiming to validate this platform in a specific biomedical field related to viral oncogenesis diseases. As a first step, the MAG.PEG cytotoxicity was evaluated in a cellular model of Kaposi’s sarcoma. By phase contrast microscopy, it was found that cell morphology remained unchanged regardless of the nanoparticles’ concentration (1–150 µg mL−1). The results, arising from the crystal violet technique, revealed that the proliferation was also unaffected. In addition, cell viability analysis by MTS and neutral red assays revealed a significant increase in metabolic and lysosomal activity at high concentrations of MAG.PEG (100–150 µg mL−1). Moreover, an increase in ROS levels was observed at the highest concentration of MAG.PEG. Second, the iron quantification assays performed by Prussian blue staining showed that MAG.PEG cellular accumulation is dose dependent. Furthermore, the presence of vesicles containing MAG.PEG inside the cells was confirmed by TEM. Finally, the MAG.PEG steering was achieved using a static magnetic field generated by a moderate power magnet. In conclusion, MAG.PEG at a moderate concentration would be a suitable drug carrier for Kaposi’s sarcoma treatment, avoiding adverse effects on normal tissues. The data included in this contribution appear as the first stage in proposing this platform as a suitable future theranostic to improve Kaposi’s sarcoma therapy.
... The most frequent type of tumor in AIDS patients is Kaposi sarcoma (KS), formed by spindle cells derived from endothelial cells transformed by KSHV (26,27). The product of orf 74 in the KSHV genome is a constitutively active G protein-coupled receptor (vGPCR) that plays an important role in the development of KSHV-induced oncogenesis (26,(28)(29)(30)(31). Only a few cells in KSlike lesions express vGPCR (28), however, down-regulation of the receptor in these cells results in a decreased expression of angiogenic factors and tumor regression (32,33). ...
... Inhibition of HO-1 expression or activity impairs the tumorigenesis induced by vGPCR in allograft tumor animal models (40). Several studies show that vGPCR contributes to KS development by switching on a complex network of signaling pathways (29,41). vGPCR activates downstream effectors by coupling to different subunits of heterotrimeric G proteins (42)(43)(44). ...
Article
Full-text available
Kaposi’s sarcoma (KS) is the most common tumor in AIDS patients. The highly vascularized patient’s skin lesions are composed of cells derived from the endothelial tissue transformed by the KSHV virus. Heme oxygenase-1 (HO-1) is an enzyme upregulated by the Kaposi´s sarcoma-associated herpesvirus (KSHV) and highly expressed in human Kaposi Sarcoma (KS) lesions. The oncogenic G protein-coupled receptor (KSHV-GPCR or vGPCR) is expressed by the viral genome in infected cells. It is involved in KS development, HO-1 expression, and vascular endothelial growth factor (VEGF) expression. vGPCR induces HO-1 expression and HO-1 dependent transformation through the Ga13 subunit of heterotrimeric G proteins and the small GTPase RhoA. We have found several lines of evidence supporting a role for Nrf2 transcription factors and family members in the vGPCR-Ga13-RhoA signaling pathway that converges on the HO-1 gene promoter. Our current information assigns a major role to ERK1/2MAPK pathways as intermediates in signaling from vGPCR to Nrf2, influencing Nrf2 translocation to the cell nucleus, Nrf2 transactivation activity, and consequently HO-1 expression. Experiments in nude mice show that the tumorigenic effect of vGPCR is dependent on Nrf2. In the context of a complete KSHV genome, we show that the lack of vGPCR increased cytoplasmic localization of Nrf2 correlated with a downregulation of HO-1 expression. Moreover, we also found an increase in phospho-Nrf2 nuclear localization in mouse KS-like KSHV (positive) tumors compared to KSHV (negative) mouse KS-like tumors. Our data highlights the fundamental role of Nrf2 linking vGPCR signaling to the HO-1 promoter, acting upon not only HO-1 gene expression regulation but also in the tumorigenesis induced by vGPCR. Overall, these data pinpoint this transcription factor or its associated proteins as putative pharmacological or therapeutic targets in KS.
... The use of KSHV oncoproteins as therapeutic targets has been gradually becoming more in focus as a novel strategy to treat and prevent the KS lesions growth. However, these studies are preliminary [75,[100][101][102][103][104][105][106]. ...
... This tumorigenesis is mediated through numerous pathways, the most important of which appears to be the phosphatidylinositol 3-kinase (PI3K) pathway ([100,120-124]) (Figure 1). PI3K is a lipid kinase that activates Akt, a serine-threonine kinase that has multiple targets, including the mammalian target of rapamycin (mTOR), a kinase that is associated with cell proliferation and survival in KS ( [100,124,125]). In vitro, cells expressing constitutively active vGPCR have high levels of activated Akt, inactivated TSC2 (a tumor suppressor which is inactivated by Akt), and activated mTOR ( [124,126,127]). This has been reversed with LY 294002, a PI3K inhibitor, or rapamycin, an mTOR inhibitor, in vitro and in vivo murine models; the latter was also associated with decreased tumor growth ( [124,127]) ( Figure 1). ...
Article
Full-text available
Kaposi’s sarcoma-associated herpesvirus (KSHV), also known as human gammaherpesvirus 8 (HHV-8), contains oncogenes and proteins that modulate various cellular functions, including proliferation, differentiation, survival, and apoptosis, and is integral to KSHV infection and oncogenicity. In this review, we describe the most important KSHV genes [ORF 73 (LANA), ORF 72 (vCyclin), ORF 71 or ORFK13 (vFLIP), ORF 74 (vGPCR), ORF 16 (vBcl-2), ORF K2 (vIL-6), ORF K9 (vIRF 1)/ORF K10.5, ORF K10.6 (vIRF 3), ORF K1 (K1), ORF K15 (K15), and ORF 36 (vPK)] that have the potential to induce malignant phenotypic characteristics of Kaposi’s sarcoma. These oncogenes can be explored in prospective studies as future therapeutic targets of Kaposi’s sarcoma.
... The previously mentioned lytic gene ORF74, encoding for the constitutively active vGPCR, activates MAP kinase and NFkB pathways [55], inducing the production of VEGF, IL-6, IL-8, and tumor necrosis factor alpha, and promoting tumorigenesis through autocrine and paracrine pathways [56][57][58][59][60]. Other lytic genes including vPK, viral IL-6 homolog (vIL-6/K2), viral interferon regulatory factor 1 (vIRF1/K9), and the membrane proteins K1 and K15 have oncogenic functions [56,59,60]. ...
... Other lytic genes including vPK, viral IL-6 homolog (vIL-6/K2), viral interferon regulatory factor 1 (vIRF1/K9), and the membrane proteins K1 and K15 have oncogenic functions [56,59,60]. KSHV vIL-6 can activate the JAK-STAT, PI3K-AKT, and MAPK-ERK pathways upon binding directly to gp130 dimers inducing the production of cellular IL-6, along with other viral proteins (Kaposins) [56][57][58]. It was shown more recently that vIL-6 decreases the expression of caveolin 1 and increases the expression of integrin β3 through its activation of STAT3, which contributes to its angiogenic-like behavior [59,60]. ...
Article
Full-text available
Simple Summary There are at least four forms of Kaposi’s sarcoma (KS) with the ‘HIV’-related form being the most aggressive and can involve mucosae or visceral organs. Kaposi’s sarcoma-associated herpes virus (KSHV) is the underlying cause of this disease. It can infect endothelial and/or mesenchymal cells and establish a latent phase in host cells in which latency proteins and various non-coding RNAs (ncRNAs) play a complex role in proliferation and angiogenesis. It also undergoes periods of sporadic lytic reactivation that are key for KS progression. Complex interactions with the microenvironment with production of inflammatory cytokines and paracrine signaling is a standout feature of KS development and maintenance. KSHV impairs the immune response by various mechanisms such as the degradation of a variety of proteins involved in immune response or binding to cellular chemokines. Treatment options include classical chemotherapy, but other novel therapies are being investigated. Abstract Kaposi’s sarcoma (KS) is a heterogeneous angioproliferative tumor that generally arises in the skin. At least four forms of this disease have been described, with the ‘HIV’-related form being the most aggressive and can involve mucosae or visceral organs. Three quarters of KS cases occur in sub-Saharan Africa (SSA) as geographic variation is explained by the disparate prevalence of KS-associated herpes virus (KSHV), which is the underlying cause of this disease. It can infect endothelial and/or mesenchymal cells that consequently transdifferentiate to an intermediate state. KSHV establishes a latent phase in host cells in which latency proteins and various non-coding RNAs (ncRNAs) play a complex role in proliferation and angiogenesis. It also undergoes periods of sporadic lytic reactivation triggered by various biological signals in which lytic stage proteins modulate host cell signaling pathways and are key in KS progression. Complex interactions with the microenvironment with production of inflammatory cytokines with paracrine signaling is a standout feature of KS development and maintenance. KSHV impairs the immune response by various mechanisms such as the degradation of a variety of proteins involved in immune response or binding to cellular chemokines. Treatment options include classical chemotherapy, but other novel therapies are being investigated.
... A vast amount of data on several viruses and their possible association with cancer development has been published [43][44][45][46][47][48][49][50][51][52]. While not the focus of this article, a brief review of the subject reveals the importance of the study of viral agents and their relation to occurrence of malignant disorders. ...
Chapter
Full-text available
Carcinogenic effects of Aspergillus spp. have been well established and generally attributed to a variety of mycotoxin productions, particularly aflatoxins. It is known that most carcinogenic mycotoxins, with the exception of fumonisins, are genotoxic and mutagenic, causing chromosomal aberrations, micronuclei, DNA single-strand breaks, sister chromatid exchange, unscheduled DNA synthesis etc. Some Aspergillus spp. are infected with mycoviruses which can result in loss of aflatoxin production. The effects of mycovirus containing Aspergillus on human health have not been fully evaluated. Recent studies in patients with acute lymphoblastic leukemia, in full remission, have revealed the existence of antibody to the products of a certain Aspergillus flavus isolate which harbored an unknown mycovirus. Exposure of blood mononuclear cells from these patients, but not controls, to the products of this organism had reproduced cell surface phenotypes and genetic markers, characteristic of acute lymphoblastic leukemia. Carcinogenic effects of Aspergillus spp. may not always be mycotoxin related and this requires further investigation.
Article
Within weeks of the first report of acquired immunodeficiency syndrome (AIDS) in 1981, it was observed that these patients often had Kaposi sarcoma (KS), a hitherto rarely seen skin tumor in the USA. It soon became apparent that AIDS was also associated with an increased incidence of high-grade lymphomas caused by Epstein-Barr virus (EBV). The association of AIDS with KS remained a mystery for more than a decade until Kaposi sarcoma-associated herpesvirus (KSHV) was discovered and found to be the cause of KS. KSHV was subsequently found to cause several other diseases associated with AIDS and human immunodeficiency virus (HIV) infection. People living with HIV/AIDS continue to have an increased incidence of certain cancers, and many of these cancers are caused by EBV and/or KSHV. In this review, we discuss the epidemiology, virology, pathogenesis, clinical manifestations, and treatment of cancers caused by EBV and KSHV in persons living with HIV.
Chapter
It was recently estimated that a virus infection is the central cause of more than 1,400,000 cancer cases annually, representing approximated 10% of the worldwide cancer burden. DNA viruses that have been confirmed to cause cancer are Epstein–Barr virus (EBV), hepatitis B virus (HBV), human papillomavirus (HPV), and human herpesvirus 8 (HSV-8). Some examples of cancer-causing RNA viruses are the human T lymphotropic virus type 1 (HTLV-1) and hepatitis C virus (HCV). The study of viruses and human cancer has sparked hope for developing novel ways to prevent cancer-causing viral infection. In this chapter, we decipher the general mechanism of cancer-causing virus and their general principles. We also summarize the common mechanism of direct and indirect carcinogenesis caused by virus infection.
Article
Endothelial-to-mesenchymal transition has been described in tumors as a source of mesenchymal stroma, while the reverse process has been proposed in tumor vasculogenesis and angiogenesis. A human oncogenic virus, Kaposi’s sarcoma herpes virus (KSHV), can regulate both processes in order to transit through this transition ‘boulevard’ when infecting KS oncogenic progenitor cells. Endothelial or mesenchymal circulating progenitor cells can serve as KS oncogenic progenitors recruited by inflammatory cytokines because KSHV can reprogram one into the other through endothelial-to-mesenchymal and mesenchymal-to-endothelial transitions. Through these novel insights, the identity of the potential oncogenic progenitor of KS is revealed while gaining knowledge of the biology of the mesenchymal-endothelial differentiation axis and pointing to this axis as a therapeutic target in KS.
Article
Introdução: A Síndrome da Imunodeficiência Adquirida (AIDS sigla em inglês para – Acquired Immunodeficiency Syndrome) é a manifestação clínica avançada decorrente de um quadro de imunodeficiência causado pelo vírus da Imunodeficiência Humana (HIV – Human Immunodeficiency Virus) identificado em 1981. O Sarcoma de Kaposi é um câncer que se desenvolve nos tecidos conectivos como cartilagem, osso, gordura, músculo, vasos sanguíneos e tecidos fibrosos. Este tipo de tumor recebe o tratamento especifico e diferencia para os indivíduos HIV + (soro positivo). Objetivo: Descrever os diferentes tipos de tratamento do Sarcoma de Kaposi para indivíduos HIV positivos. Método: Este estudo apresenta-se como uma revisão de literatura e utilizamos um método de analises de dados na qual buscamos manter o padrão de qualidade acima de tudo, filtramos assuntos relacionados ao tema de Sarcoma de Kaposi em HIV/AIDS positivo, utilizamos como descritores os seguintes: AIDS. HIV. KS. Sarcoma de Kaposi em HIV/AIDS. Todos os artigos utilizados nesta revisão foi retirado de bases eletrônicas, as quais foram: Literatura Latino Americana e do Caribe em Ciências da Saúde (LILLACS), Scientific Eletrônic Library Online (SCIELO) e National Library of Medicine (PUBMED). Foi definido como criterio de inclusão: artigos publicados entre os anos de 1990 e 2021 e se correlacionava ao tema principal deste trabalho. Resultados: A análise dos trabalhos sobre o Sarcoma de Kaposi em pacientes com HIV/AIDS contribuem para um melhor entendimento dos mecanismos que estão envolvidos o desenvolvimento destes tumores com a DST, e, com insto, conseguimos elucidar os mecanismos que interagem o Sarcoma com o HIV/AIDS. Conclusão: Com base nos artigos reunidos aqui, conseguimos notar que o primeiro tratamento para do Sarcoma de Kaposi utilizava-se quimioterapia e radioterapia, e, com o avanço da ciência conseguimos adaptar e criar novos medicamentos antirretrovirais que buscam conter o desenvolvimento do Sarcoma e até mesmo do próprio HIV/AIDS.
Article
The observation in 1981 of the emergence of Kaposi sarcoma (KS) among young men who had sex with men was one of the first harbingers of the HIV epidemic. With advances in HIV care, the incidence of HIV-associated KS (HIV ⁺ KS) has decreased over time in the United States. However, it remains a persistent malignancy among some HIV-infected populations and is one of the most common tumors in sub-Saharan Africa. Because of the relapsing and remitting nature of this cancer, patients with HIV ⁺ KS can experience significant, long-term, morbidity. Patients with severe HIV ⁺ KS may also have concurrent lymphoproliferative syndromes, malignancies, and/or infections that can contribute to mortality. Several chemotherapy agents were explored in clinical trials for HIV ⁺ KS during the early stage of the epidemic. As HIV ⁺ KS emerges with CD4 lymphopenia and immunodysregulation, T-cell–sparing options are important to consider. Here, we explore the pathogenesis of HIV ⁺ KS and the current evidence for immunotherapy and therapies that potentially target KS pathogenesis. This review provides the current landscape of therapies for HIV ⁺ KS and highlights management issues for patients with HIV and cancer.
Article
Full-text available
Human herpesvirus 8 (HHV8, also known as Kaposi's sarcoma [KS]-associated herpesvirus) has been implicated as an etiologic agent for KS, an angiogenic tumor composed of endothelial, inflammatory, and spindle cells. Here, we report that transgenic mice expressing the HHV8-encoded chemokine receptor (viral G protein–coupled receptor) within hematopoietic cells develop angioproliferative lesions in multiple organs that morphologically resemble KS lesions. These lesions are characterized by a spectrum of changes ranging from erythematous maculae to vascular tumors, by the presence of spindle and inflammatory cells, and by expression of vGPCR, CD34, and vascular endothelial growth factor. We conclude that vGPCR contributes to the development of the angioproliferative lesions observed in these mice and suggest that this chemokine receptor may play a role in the pathogenesis of KS in humans.
Article
Full-text available
The most recently identified human herpesvirus, Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) or human herpesvirus 8 (HHV-8), has been found to be a necessary, although perhaps not sufficient, etiologic agent for all forms of KS 12. This virus is also invariably present in a rare subset of malignant lymphomas, primary effusion lymphomas (PELs), and in a significant percentage of patients with multicentric Castleman's disease, an angiolymphoproliferative disorder 34. Both KS and PEL occur more frequently, but not exclusively, in HIV-infected individuals, and all cases of HIV-related multicentric Castleman's disease are infected with KSHV. These findings suggest an important role of immunosuppression and HIV infection in KSHV-mediated pathogenesis. Although it remains controversial whether KS is a malignant neoplasm, KS lesions probably evolve from a reactive, inflammatory/angioproliferative process into true clonal cancers. Thus, KS is generally considered a malignancy, especially because of its frequent multifocal and aggressive behavior. Given the definitive association of KSHV with two different human malignancies, it is not difficult to consider KSHV to be a human oncogenic virus.
Article
Full-text available
The elucidation of the molecular mechanisms governing the transition from a nonangiogenic to an angiogenic phenotype is central for understanding and controlling malignancies. Viral oncogenes represent powerful tools for disclosing transforming mechanisms, and they may also afford the possibility of investigating the relationship between transforming pathways and angiogenesis. In this regard, we have recently observed that a constitutively active G protein-coupled receptor (GPCR) encoded by the Kaposi's sarcoma-associated herpes virus (KSHV)/human herpes virus 8 is oncogenic and stimulates angiogenesis by increasing the secretion of vascular endothelial growth factor (VEGF), which is a key angiogenic stimulator and a critical mitogen for the development of Kaposi's sarcoma. Here we show that the KSHV GPCR enhances the expression of VEGF by stimulating the activity of the transcription factor hypoxia-inducible factor (HIF)-1alpha, which activates transcription from a hypoxia response element within the 5'-flanking region of the VEGF promoter. Stimulation of HIF-1alpha by the KSHV GPCR involves the phosphorylation of its regulatory/inhibitory domain by the p38 and mitogen-activated protein kinase (MAPK) signaling pathways, thereby enhancing its transcriptional activity. Moreover, specific inhibitors of the p38 (SKF86002) and MAPK (PD98059) pathways are able to inhibit the activation of the transactivating activity of HIF-1alpha induced by the KSHV GPCR, as well as the VEGF expression and secretion in cells overexpressing this receptor. These findings suggest that the KSHV GPCR oncogene subverts convergent physiological pathways leading to angiogenesis and provide the first insight into a mechanism whereby growth factors and oncogenes acting upstream from MAPK, as well as inflammatory cytokines and cellular stresses that activate p38, can interact with the hypoxia-dependent machinery of angiogenesis. These results may also help to identify novel targets for the development of antiangiogenic therapies aimed at the treatment of Kaposi's sarcoma and other neoplastic diseases.
Article
Full-text available
The Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor (KSHV-GPCR) is a key molecule in the pathogenesis of Kaposi's sarcoma, playing a central role in the promotion of vascular endothelial growth factor (VEGF)-driven angiogenesis and spindle cell proliferation. We previously have shown that KSHV-GPCR has oncogenic potential when overexpressed in fibroblasts and is responsible for the expression and secretion of VEGF through the regulation of different intracellular signaling pathways (A. Sodhi et al., Cancer Res., 60: 4873-4880, 2000; C. Bais et al., Nature, 391: 86-89, 1998). Here, we describe that this constitutively active G protein-coupled receptor is able to promote cell survival in primary human umbilical vein endothelial cells and that this effect is independent of its ability to secrete VEGF because it is not prevented by the expression of antisense constructs for VEGF or the addition of VEGF-blocking antibodies. Instead we found that ectopic expression of KSHV-GPCR potently induces the kinase activity of Akt/protein kinase B in a dose-dependent manner and triggers its translocation to the plasma membrane. This signaling pathway requires the function of phosphatidylinositol 3'-kinase and is dependent on betagamma subunits released from both pertussis toxin-sensitive and -insensitive G proteins. Furthermore, we found that KSHV-GPCR is able to protect human umbilical vein endothelial cells from the apoptosis induced by serum deprivation and that both wortmannin and the expression of a kinase-deficient Akt K179M mutant are able to block this effect. Finally, we observed that the Akt K179M protein also inhibits the activation of nuclear factor-KB induced by KSHV-GPCR, suggesting that this transcription factor may represent one of the putative downstream targets for Akt in the survival-signaling pathway. These results provide further knowledge in the elucidation of the signal transduction pathways activated by KSHV-GPCR and support its key role in promoting the survival of viral-infected cells. Moreover, the present findings also emphasize the importance of this G protein-coupled receptor in the development of KSHV-related neoplasias.
Article
Full-text available
Kaposi's sarcoma-associated herpesvirus (KSHV; human herpesvirus 8) encodes a chemokine-like G protein-coupled receptor (KSHV-GPCR) that is implicated in the pathogenesis of Kaposi's sarcoma (KS). Since endothelial cells appear to be targets for the virus, we developed an in vitro mouse lung endothelial cell model in which KSHV-GPCR is stably expressed and KSHV-GPCR signaling was studied. In mouse lung endothelial cells: 1) KSHV-GPCR does not exhibit basal signaling through the phosphoinositide-specific phospholipase C pathway but inositol phosphate production is stimulated by growth-related oncogene α (Gro-α) via a pertussis toxin (PTX)-insensitive pathway; 2) KSHV-GPCR signals basally through a PTX-sensitive pathway leading to a lowering of intracellular cAMP level that can be lowered further by Groα and increased by interferon γ-inducible protein 10; 3) KSHV-GPCR stimulates phosphatidylinositol 3-kinase via a PTX-insensitive mechanism; and 4) KSHV-GPCR activates nuclear factor-κB (NF-κB) by a PTX-sensitive Gβγ subunit-mediated pathway. These data show that KSHV-GPCR couples to at least two G proteins and initiates signaling via at least three cascades in endothelial cells thereby increasing the complexity of regulation of endothelial cell function by KSHV-GPCR that may occur during viral infection.
Article
Full-text available
Infection with human herpesvirus 8 (HHV-8), also known as Kaposi's sarcoma (KS)-associated herpesvirus, is necessary for the development of KS. The HHV-8 lytic-phase gene ORF74 is related to G protein-coupled receptors, particularly interleukin-8 (IL-8) receptors. ORF74 activates the inositol phosphate/phospholipase C pathway and the downstream mitogen-activated protein kinases, JNK/SAPK and p38. We show here that ORF74 also activates NF-kappaB independent of ligand when expressed in KS-derived HHV-8-negative endothelial cells or primary vascular endothelial cells. NF-kappaB activation was enhanced by the chemokine GROalpha, but not by IL-8. Mutation of Val to Asp in the ORF74 second cytoplasmic loop did not affect ligand-independent signaling activity, but it greatly increased the response to GROalpha. ORF74 upregulated the expression of NF-kappaB-dependent inflammatory cytokines (RANTES, IL-6, IL-8, and granulocyte-macrophage colony-stimulating factor) and adhesion molecules (VCAM-1, ICAM-1, and E-selectin). Supernatants from transfected KS cells activated NF-kappaB signaling in untransfected cells and elicited the chemotaxis of monocytoid and T-lymphoid cells. Expression of ORF74 conferred on primary endothelial cells a morphology that was strikingly similar to that of spindle cells present in KS lesions. Taken together, these data, demonstrating that ORF74 activates NF-kappaB and induces the expression of proangiogenic and proinflammatory factors, suggest that expression of ORF74 in a minority of cells in KS lesions could influence uninfected cells or latently infected cells via autocrine and paracrine mechanisms, thereby contributing to KS pathogenesis.
Article
The Kaposi's sarcoma herpesvirus (KSHV) has been identified as the etiologic agent of Kaposi's sarcoma (KS), but initial events leading to KS development remain unclear. Characterization of the KSHV genome reveals the presence of numerous potential oncogenes. To address their contribution to the initiation of the endothelial cell-derived KS tumor, we developed a novel transgenic mouse that enabled endothelial cell-specific infection in vivo using virus expressing candidate KSHV oncogenes. Here we show that transduction of one gene, vGPCR, was sufficient to induce angioproliferative tumors that strikingly resembled human KS. Endothelial cells expressing vGPCR were further able to promote tumor formation by cells expressing KSHV latent genes, suggestive of a cooperative role among viral genes in the promotion of Kaposi's sarcomagenesis.
Article
Representational difference analysis was used to isolate unique sequences present in more than 90 percent of Kaposi's sarcoma (KS) tissues obtained from patients with acquired immunodeficiency syndrome (AIDS). These sequences were not present in tissue DNA from non-AIDS patients, but were present in 15 percent of non-KS tissue DNA samples from AIDS patients. The sequences are homologous to, but distinct from, capsid and tegument protein genes of the Gammaherpesvirinae, herpesvirus saimiri and Epstein-Barr virus. These KS-associated herpesvirus-like (KSHV) sequences appear to define a new human herpesvirus.
Article
The Kaposi's sarcoma-associated herpesvirus (KSHV/HHV8) is a gamma-2 herpesvirus that is implicated in the pathogenesis of Kaposi's sarcoma and of primary effusion B-cell lymphomas (PELs). KSHV infects malignant and progenitor cells of Kaposi's sarcoma and PEL, it encodes putative oncogenes and genes that may cause Kaposi's sarcoma pathogenesis by stimulating angiogenesis. The G-protein-coupled receptor encoded by an open reading frame (ORF 74) of KSHV is expressed in Kaposi's sarcoma lesions and in PEL and stimulates signalling pathways linked to cell proliferation in a constitutive (agonist-independent) way. Here we show that signalling by this KSHV G-protein-coupled receptor leads to cell transformation and tumorigenicity, and induces a switch to an angiogenic phenotype mediated by vascular endothelial growth factor, an angiogenesis and Kaposi's-spindle-cell growth factor. We find that this receptor can activate two protein kinases, JNK/SAPK and p38MAPK, by triggering signalling cascades like those induced by inflammatory cytokines that are angiogenesis activators and mitogens for Kaposi's sarcoma cells and B cells. We conclude that the KSHV G-protein-coupled receptor is a viral oncogene that can exploit cell signalling pathways to induce transformation and angiogenesis in KSHV-mediated oncogenesis.
Article
Kaposi's sarcoma-associated herpesvirus (KSHV) is a newly identified gammaherpesvirus associated with all clinical forms of Kaposi's sarcoma (KS), body-cavity-based, primary effusion lymphomas (PELs), and a subset of Castleman's disease (CD). Sequence analysis of the KSHV genome demonstrates an extensive array of genes with homology to cellular genes involved in cell cycle regulation, cell proliferation, apoptosis, and immune modulation. Functional studies indicate that these genes may modify the host cell environment, contributing to the pathogenesis of KSHV-associated disorders. Several KSHV genes have been found to cause dysregulated cell proliferation or to interfere with established tumor suppressor pathways. The epidemiologic association of KSHV with malignancies and the coding features of its genome suggest that it is a new DNA tumor virus.