ArticlePDF Available

Pennati M, Coltella G, Folini M, Citti L, Dandone MG, Zaffaroni MRibozyme-mediated attenuation of survivin expression sensitizes human melanoma cells to cisplatin-induced apoptosis. J Clin Invest 109: 285-286

Authors:
Survivin is a structurally unique mem-
ber of the inhibitor of apoptosis (IAP)
family of proteins that is potentially
involved in both control of cell divi-
sion and inhibition of apoptosis (1).
Specifically, its antiapoptotic function
is related to the ability to directly or
indirectly inhibit caspases (2). The
notion that survivin is overexpressed
in most of the common human
tumors (3, 4) but absent in normal
adult tissues with only a few excep-
tions (5, 6) has led to the proposal of
survivin as a promising therapeutic
target for novel anticancer therapies
(7). Indeed, in October 2001, Mesri et
al. (8) reported in the JCI that infec-
tion with a replication-deficient aden-
ovirus encoding a Thr34Ala mutant
of survivin caused apoptosis in
human tumor cell lines of different
histology and reduced tumor growth
in xenograft breast cancer models.
Moreover, inhibition of survivin
expression enhanced taxol-induced
cell death in tumor cells.
As an alternative strategy for survivin
inhibition we developed hammerhead
ribozymes targeting the 3end of the
CUA110 (RZ1) and the GUC294 (RZ7)
triplets in the survivin mRNA. In a cell-
free system, both ribozymes induced
cleavage of a synthetic RNA substrate
obtained by cloning a portion of sur-
vivin mRNA, with cleavage products
being detectable starting from a
ribozyme/substrate ratio of 1:0.5. Con-
versely, the catalytically inactive
mutRZ1 (which was produced by intro-
ducing a mutation in the catalytic core
of the active ribozyme RZ1 and was
used as control throughout the study)
did not show any cleavage activity. RZ1,
RZ7, and mutRZ1 sequences were
inserted into the pRC expression vector
under the control of the cytomega-
lovirus promoter and transfected into
the human metastatic melanoma cell
line JR8 overexpressing survivin. For the
present study we selected three stably
transfected clones proven to endoge-
nously express RZ1 (clone RZ1/C), RZ7
(clone RZ7/H), or mutRZ1 (clone
mutRZ1/B). RZ1/C and RZ7/H cells
were characterized by a markedly lower
survivin protein level (68% and 60%
lower, respectively) than JR8 parental
cells, whereas a negligible reduction
(13%) in survivin expression was
observed in mutRZ1/B cells (Figure 1a).
To evaluate the effect of survivin inhibi-
tion on the susceptibility of melanoma
cells to undergo cisplatin-induced apop-
tosis, we treated the different clones
with 10 µg/ml of the drug for 1 hour
and determined the presence of apop-
totic nuclei in cells stained with propid-
ium iodide under fluorescence micro-
scopy at 72 hours after treatment. A very
modest apoptotic response was ob-
served in the mutRZ1/B cells, whereas a
significant increase in the percentage of
apoptotic cells was observed in RZ1/C
(P = 0.01) and RZ7/H (P = 0.005) cells
(Figure 1b). Processing of caspase-3 to
its active subunits of approximately 17
and 19 kDa was observed in all three
drug-treated clones. However, the cas-
pase-3 catalytic activity as assessed by
hydrolysis of the fluorogenic substrate
N-Acetyl-Asp-Glu-Val-Asp-aldehyde
(Ac-DEVD-AMC) was about threefold
higher in RZ1/C and RZ7/H clones
than in the mutRZ1/B clone (Figure 1c).
These results are in agreement with
the previous finding of Grossman et
al. (9), who observed enhancement of
cisplatin-induced apoptosis by expres-
sion of the survivin Thr34Ala
mutant in YUSAC2 melanoma cells.
Unlike what was reported by these
authors, attenuation of survivin
expression in our melanoma cell sys-
tem was not sufficient to appreciably
trigger apoptosis in the absence of
other stimuli. Other antiapoptotic
factors besides survivin, such as bcl-2
and bcl-xL, are strongly expressed in
JR8 cells and may contribute to pre-
Ribozyme-mediated attenuation of survivin
expression sensitizes human melanoma cells
to cisplatin-induced apoptosis
The Journal of Clinical Investigation | January 2002 | Volume 109 | Number 2 285
Letters to the Editor
Figure 1
(a) Survivin protein expression in JR8 parental cells and melanoma cell clones transfected with the active ribozymes RZ1 (RZ1/C clone) and RZ7 (RZ7/H
clone) or with the mutant ribozyme mutRZ1 (mutRZ1/B clone). Western blots were probed with a polyclonal antibody for survivin. Proliferating cell
nuclear antigen (PCNA) was used as a control for loading. (band c) Induction of apoptosis in melanoma cell clones exposed to 10 µg/ml cisplatin for
1 hour. Seventy-two hours after treatment, cells were collected and the occurrence of apoptosis was determined. (b) The percentage of cells with an
apoptotic morphology with respect to the overall population was assessed by fluorescence microscopy after cell staining with propidium iodide. White
bars, control (no drug) cells; gray bars, cisplatin-treated cells. Data represent mean values ± SD of three experiments. (c) The caspase-3 catalytic activ-
ity was determined by hydrolysis of the fluorogenic substrate N-Acetyl-Asp-Glu-Val-Asp-aldehyde (Ac-DEVD-AMC) in the presence or absence of the
caspase-3 inhibitor Ac-DEVD-CHO. White bars, control (no drug) cells; gray bars, cisplatin-treated cells; black bars, cisplatin-treated cells + CHO.
Data represent mean values ± SD of three experiments.
In conclusion, the present results
obtained with the ribozyme-mediated
approach in melanoma cells extend
and corroborate earlier evidence indi-
cating that attenuating survivin expres-
sion renders these cells more suscepti-
ble to cisplatin-induced apoptosis.
These data also suggest a possible strat-
egy to enhance the chemosensitivity
profile of such a drug-refractory
human malignancy.
Marzia Pennati,1Gennaro Colella,1
Marco Folini,1Lorenzo Citti,2
Maria Grazia Daidone,1
and Nadia Zaffaroni1
1Department of Experimental Oncology,
Unit 10, Istituto Nazionale per lo Studio
e la Cura dei Tumori, Milan, Italy
2Istituto di Mutagenesi e Differenziamento,
Pisa, Italy
Address correspondence to: Nadia Zaffaroni,
Istituto Nazionale per lo Studio e la Cura dei
Tumori, Via Venezian 1, 20133 Milano, Italy.
Phone: 39-02-23903260; Fax: 39-0223903052;
E-mail: zaffaroni@istitutotumori.mi.it.
Marzia Pennati and Gennaro Colella
contributed equally to this work.
1. Altieri, D.C., and Marchisio, P.C. 1999. Survivin
apoptosis: an interloper between cell death and cell
proliferation in cancer. Lab. Invest. 79:1327–1333.
2. Reed, J.C. 2001. The survivin saga goes in vivo. J. Clin.
Invest. 108:965–969. DOI:10.1172/JCI200114123.
3. Ambrosini, G., Adida, C., and Altieri, D. 1997. A
novel anti-apoptosis gene, survivin, expressed in
cancer and lymphoma. Nat. Med. 3:917–921.
4. Velculescu, V.E., et al. 1999. Analysis of human
transcriptomes. Nat. Genet. 23:387–388.
5. Konno, R., et al. 2000. Expression of survivin and
Bcl-2 in the normal human endometrium. Mol.
Hum. Reprod. 6:529–534.
6. Chiodino, C., et al. 1999. Expression of the novel
inhibitor of apoptosis survivin in normal and
neoplastic skin. J. Invest. Dermatol. 113:415–418.
7.Grossman, D., McNiff, J.M., Li, F., and Altieri,
D.C. 1999. Expression and targeting of the apop-
tosis inhibitor, survivin, in human melanoma. J.
Invest. Dermatol. 113:1076–1081.
8. Mesri, M., Wall, N.R., Li, J., Kim, R.W., and Altieri,
D.C. 2001. Cancer gene therapy using a survivin
mutant adenovirus. J. Clin. Invest. 108:981–990.
DOI:10.1172/JCI200112983.
9. Grossman, D., Kim, P.J., Schechner, J.S., and
Altieri, D.C. 2001. Inhibition of melanoma tumor
growth in vivo by survivin targeting. Proc. Natl.
Acad. Sci. USA. 98:635–640.
venting programmed cell death in this
tumor model. However, it should be
stressed that in JR8 cells survivin
expression was attenuated but not
completely abrogated. It may be that
inhibition below a certain threshold is
insufficient to determine a proapop-
totic effect. Interestingly, and in accor-
dance with such a hypothesis, when
we transduced the human prostate
cancer cells DU145 with a Moloney-
based retroviral vector carrying the
catalytic sequence of the ribozyme
RZ7, we were able to select a
ribozyme-expressing clone character-
ized by an almost complete abroga-
tion of survivin expression (99.5%
lower compared with control cells, as
assessed by Western blotting, and lack
of detectable protein expression by
confocal microscopy). This ribozyme-
expressing clone also showed a
markedly higher percentage of apop-
totic cells than control culture (20%
and 3% of cells on the overall cell pop-
ulation, respectively).
286 The Journal of Clinical Investigation | January 2002 | Volume 109 | Number 2
... 9 Molecular probes such as antisense oligonucleotide, ribozyme, siRNA, and dominant negative mutant molecules all resulted in caspase-dependent cell death and increased drug-induced apoptosis. [10][11][12][13][14][15][16] Thus, survivin has become a significant and attractive drug target 17 due to not only its aberrant expression in cancers but also its dual roles in inhibiting apoptotic pathway and regulating cell cycle progression. 18 We previously demonstrated the ability to target the key residues in the hydrophobic dimerization domain of survivin with small-molecule inhibitors, leading to proteosomedependent destruction of survivin proteins. ...
... It is known that inhibiting survivin induces spontaneous apoptosis. [10][11][12][13][14][15][16] To determine if 7F1 induces apoptosis, annexin V staining of C4-2 and PC-3 cells following 7F1 treatments was performed. As shown in Fig. 4A, treatment with 200 nM 7F1 resulted in ~20% apoptosis in both PC-3 and C4-2 cells. ...
Article
Survivin, a member of the inhibitor of apoptosis protein family, exists as a homodimer and is aberrantly upregulated in a wide spectrum of cancers. It was thought to be an ideal target due to its lack of expression in most adult normal tissues and importance in cancer cell survival. However, it has been challenging to target survivin due to its “undruggable” nature. We previously attempted to target its dimerization domain with a hypothesis that inhibiting survivin dimerization would promote its degradation in proteasome, which led to identification of a lead small-molecule inhibitor, LQZ-7F. LQZ-7F consists of a flat tetracyclic aromatic core with labile hydrazone linking a 1,2,5-oxadiazole moiety. In this study, we tested the hypothesis that LQZ-7F could be developed as a prodrug because the labile hydrazone linker could be hydrolyzed, releasing the tetracyclic aromatic core. To this end, we synthesized the tetracyclic aromatic core (LQZ-7F1) using reported procedure and tested LQZ-7F1 for its biological activities. Here we show that LQZ-7F1 has a significantly improved potency with submicromolar IC50’s and induces spontaneous apoptosis in prostate cancer cells. It also more effectively inhibits survivin dimerization and induces survivin degradation in a proteasome-dependent manner than LQZ-7F. We also show that the combination of LQZ-7F1 and docetaxel have strong synergism in inhibiting prostate cancer cell survival. Together, we conclude that the hydrazone linker with the oxadiazole tail is dispensable for survivin inhibition and the survivin dimerization inhibitor, LQZ-7F, may be developed as a prodrug for prostate cancer treatment and to overcome docetaxel resistance.
... Studies on human melanoma cell lines transfected with hammerhead ribozymes like CUA110 (RZ7) and GUC294 (RZ1), result in reduction of survivin expression that contributes to cell death and sensitivity to drugs and radiation. Attenuation of survivin expression with ribozymes improves susceptibility of melanoma cells to anti-tumor activity of cisplatin and topotecan triggering apoptosis [201,202]. ...
Article
Full-text available
Survivin is one of the rare proteins that is differentially expressed in normal and cancer cells and is directly or indirectly involved in numerous pathways required for tumor maintenance. It is expressed in almost all cancers and its expression has been detected at early stages of cancer. These traits make survivin an exceptionally attractive target for cancer therapeutics. Even with these promising features to be an oncotherapeutic target, there has been limited success in the clinical trials targeting survivin. Only recently it has emerged that survivin was not being specifically targeted which could have resulted in the negative clinical outcome. Also, focus of research has now shifted from survivin expression in the overall heterogeneous tumor cell populations to survivin expression in cancer stem cells as these cells have proved to be the major drivers of tumors. Therefore, in this review we have analyzed the expression of survivin in normal and cancer cells with a particular focus on its expression in cancer stem cell compartment. We have discussed the major signaling pathways involved in regulation of survivin. We have explored the current development status of various types of interventions for inhibition of survivin. Furthermore, we have discussed the challenges involving the development of potent and specific survivin inhibitors for cancer therapeutics. Finally we have given insights for some of the promising future anticancer treatments.
... Surprisingly, the survivin level is elevated in different tumor types due to chemotherapeutic treatment with subsequent poor responses to chemo-/radio-therapy [92]. In addition, the low expression of survivin was found to enhance the anticancer effects of several conventional chemotherapeutic agents such as cisplatin [93]. Indirectly, survivin downregulation decreased the turnover rate of the P-glycoprotein efflux pump with consequent reversing of the multidrug resistance feature of CRC [94]. ...
Chapter
By 2030, the mortality rates of colon cancer are expected to increase by 60%. Therapy resistance (resistance to chemo- and targeted therapies) is the main reason for colorectal cancer-related mortality. Despite the development of novel and targeted therapies, chemo-resistance results in a high incidence of metastasis and recurrence. Chemo-sensitization using phytochemicals (natural products), with their multitarget potential and relatively low toxicity, is a recent and innovative strategy proposed to overcome chemo-resistance. They mainly aim to increase the cytotoxic potential of anticancer drugs, limiting their toxic side effects and delaying the appearance of acquired chemo-resistance. Yet, complex mechanisms of chemo-resistance exist that usually enable colorectal cancer cells to escape from the killing effects of the chemotherapeutic agent. Besides, chemo-resistance is a sum of complex phenomena involving many signaling cascades acting collectively for the sake of colorectal cancer cells’ survival. Herein, we summarized the details of several major resistant pathways utilized by CRC cells such as the autophagy, m-TOR pathway, tumor hypoxia, nuclear factor-κB (NF-κB), death receptors (TRAIL-receptors), survivin, and the ubiquitin-proteasome system. In addition, we shed the light on how some potentially promising phytochemicals (natural products) from plants and marine organisms can interfere with these CRC pro-survival resistant pathways.
Article
Full-text available
Isolated limb perfusion (ILP) involves the local administration of high doses of anticancer drugs into a limb affected by unresectable locally advanced tumors (with special regard to in-transit melanoma metastases), minimizing systemic side effects. Tumor response to anticancer drugs may depend on the expression of apoptosis-related genes, such as SURVIVIN and MDM2. This retrospective cohort study investigated the association between tumor SURVIVIN and MDM2 expression levels and treatment response or clinical outcomes in patients undergoing ILP for in-transit melanoma metastases. The study cohort consisted of 62 patients with in-transit metastases who underwent ILP with tumor necrosis factor (TNF) and melphalan. Tissue samples were taken from the in-transit metastases, and RNA was extracted for gene expression analysis. Patients’ response to treatment was assessed using clinical and radiological criteria two months after ILP, and disease response was classified as complete, partial, or stable/progressive disease. Disease-free survival (DFS) and overall survival (OS) were also analyzed. Expression of SURVIVIN and/or MDM2 was observed in 48% of patients; in these cases, complete response to ILP occurred in 40% of cases, with the overall response rate (complete + partial) being 85%. Patients with expression of MDM2 alone had a lower complete response rate (28%), while patients with expression of SURVIVIN alone had a higher complete response rate (50%). The combined expression of MDM2 and SURVIVIN resulted in a complete response rate of 30%. Patients without expression (of SURVIVIN or MDM2) had the highest complete response rate (58%). Survival analysis showed that high MDM2 expression was independently associated with a lower probability of a complete response to ILP. In addition, patients with MDM2 expression were three times more likely to have an incomplete response to ILP. This study highlights the importance of considering SURVIVIN and MDM2 expression in patients undergoing ILP for in-transit cutaneous melanoma metastases. High MDM2 expression was found to be an independent factor associated with a reduced likelihood of achieving a complete response to ILP, suggesting potential mechanisms of chemoresistance. These data support further research to explore the role of already available targeted therapies (i.e., MDM2 inhibitors) in improving tumor response to ILP in patients with in-transit melanoma metastases.
Article
Full-text available
The anticancer effects of taxanes are attributed to the induction of mitotic arrest through activation of the spindle assembly checkpoint. Cell death following extended mitotic arrest is mediated by the intrinsic apoptosis pathway. Accordingly, factors that influence the robustness of mitotic arrest or disrupt the apoptotic machinery confer drug resistance. Survivin is an inhibitor of apoptosis protein. Its overexpression is associated with chemoresistance, and its targeting leads to drug sensitization. However, Survivin also acts specifically in the spindle assembly checkpoint response to taxanes. Hence, the failure of Survivin-depleted cells to arrest in mitosis may lead to taxane resistance. Here we show that Survivin depletion protects HeLa cells against docetaxel-induced apoptosis by facilitating mitotic slippage. However, Survivin depletion does not promote clonogenic survival of tumor cells but increases the level of cellular senescence induced by docetaxel. Moreover, lentiviral overexpression of Survivin does not provide protection against docetaxel or cisplatin treatment, in contrast to the anti-apoptotic Bcl-xL or Bcl-2. Our findings suggest that targeting Survivin may influence the cell response to docetaxel by driving the cells through aberrant mitotic progression, rather than directly sensitizing cells to apoptosis.
Article
Survivin, a homodimeric member of the Inhibitor of Apoptosis Protein (IAP) family, is required for cancer cell survival and overexpressed in almost all solid tumors. However, targeting survivin has been challenging due to its “undruggable” nature. Recently, we used a novel approach to target the dimerization interface and identified inhibitors of two scaffolds that can directly bind to and inhibit survivin dimerization. One of the scaffolds, represented by the compound LQZ-7, con-tain an undesirable labile hydrazone linker and a potentially non-functional furazanopyrazine ring that we attempted to eliminate in this study. We found one compound, 7I, that is more active than the parent compound, LQZ-7, and when giv-en orally effectively inhibits xenograft tumor growth and induces survivin loss in tumors. These findings indicate that 7I with a stable linker and a quinoxaline ring can be used as a lead for further optimization of this novel class of survivin inhibitors.
Article
Full-text available
Abstract Survivin (also named BIRC5) is a well-known cancer therapeutic target. Since its discovery more than two decades ago, the use of survivin as a target for cancer therapeutics has remained a central goal of survivin studies in the cancer field. Many studies have provided intriguing insight into survivin’s functional role in cancers, thus providing promise for survivin as a cancer therapeutic target. Despite this, moving survivin-targeting agents into and through the clinic remains a challenge. In order to address this challenge, we may need to rethink current strategies in order to develop a new mindset for targeting survivin. In this Review, we will first summarize the current survivin mechanistic studies, and then review the status of survivin cancer therapeutics, which is classified into five categories: (i) survivin-partner protein interaction inhibitors, (ii) survivin homodimerization inhibitors, (iii) survivin gene transcription inhibitors, (iv) survivin mRNA inhibitors and (v) survivin immunotherapy. We will then provide our opinions on cancer therapeutics using survivin as a target, with the goal of stimulating discussion that might facilitate translational research for discovering improved strategies and/or more effective anticancer agents that target survivin for cancer therapy.
Article
Full-text available
Survivin is the smallest member of the inhibitor of apoptosis protein family, which has key roles in regulating cell division and inhibiting apoptosis by blocking caspase activation. Survivin is highly expressed in most human cancers, such as lung, pancreatic and breast cancers, relative to normal tissues. Aberrant survivin expression is associated with tumor cell proliferation, progression, angiogenesis, therapeutic resistance, and poor prognosis. Studies on the underlying molecular mechanisms indicate that survivin is involved in the regulation of cytokinesis and cell cycle progression, as well as participates in a variety of signaling pathways such as the p53, Wnt, hypoxia, transforming growth factor, and Notch signaling pathways. In this review, recent progress in understanding the molecular basis of survivin is discussed. Therapeutic strategies targeting survivin in preclinical studies are also briefly summarized.
Article
Hepatocellular carcinoma (HCC) is known to be resistant to chemotherapy. Survivin, a member of the inhibitor of apoptosis proteins, is overexpressed in most cancers but is absent in most normal adult tissue. The aim of this study was to investigate whether expression of survivin contributes to resistance to cisplatin-induced apoptosis. We confirmed induction of survivin expression in hepatoma in the N-diethylnitrosamine (DEN) induced rat and in the rat hepatoma cell line (K-251). We examined cell proliferation after treatment with cisplatin (CDDP) in the presence and absence of siRNA or the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 to suppress survivin or PI3K/ Akt, respectively. Survivin was expressed in DEN-induced rat HCC with RT-PCR and Western blotting. Expression of survivin was observed primary in the nuclei and in the cytoplasm with immunohistochemistry. However, survivin was not detected in non-tumor tissues. Expression of survivin was also observed primarily in the nuclei and in the cytoplasm of the K-251 rat hepatoma cell line. CDDP induced survivin expression, which was blocked by siRNA. LY294002 also attenuated survivin expression induced by CDDP. Our results indicate that survivin expression via PI3K contributes to resistance to CDDP-induced apoptosis in a rat hepatoma cell line.
Article
Full-text available
Survivin is a member of the inhibitors of apoptosis protein family. Here, we examined survivin expression and confirmed abundant survivin expression in bladder cancer cells. This expression pattern indicated that the transcriptional regulatory elements that control survivin expression could be utilized to discriminate cancer from normal cells. We therefore generated a novel adenovirus termed Ad5/35E1apsurvivinE4 with the following characteristics: 1) E1A and E4 protein expression was dependent on survivin promoter activity; 2) the green fluorescence protein gene was inserted into the genome under the control of the CMV promoter; 3) most of the E3 sequences were deleted, but the construct was still capable of expressing the adenovirus death protein with potent cytotoxic effects; and 4) the fiber knob was from serotype 35 adenovirus. As expected from the abundant survivin expression observed in bladder cancer cells, Ad5/35E1apsurvivinE4 replicated better in cancer cells than in normal cells by a factor of 106 to 102. Likewise, Ad5/35E1apsurvivinE4 exerted greater cytotoxic effects on all bladder cancer cell lines tested. Importantly, Ad5/35E1apsurvivinE4 inhibited the growth of Ku7-Luc orthotopic xenografts in nude mice. Taken together, Ad5/35E1apsurvivinE4 indicates that the survivin promoter may be utilized for the development of a replication-competent adenovirus to target bladder cancers.
Article
Full-text available
Survivin is a novel inhibitor of apoptosis. It has been reported that survivin is expressed during fetal development and in cancer tissues, but its expression has not been reported in adult tissues. We investigated the expression of survivin in the endometria of women with regular menstrual cycles using reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry, and compared these findings with Bcl-2, an apoptosis inhibitor. Survivin mRNA was detected by RT-PCR in all samples (nine of nine) of endometrium during the secretory phase, but in only four out of seven samples from endometrium during the proliferative phase, and in none of the atrophic endometrium. Immunohistochemistry demonstrated a survivin protein expression that was strongest in the nuclei of glandular epithelial cells during the late secretory phase. In the proliferative phase, glandular epithelial cells were not stained for survivin. The cyclic changes of survivin and Bcl-2 showed an inverse relationship, with Bcl-2 expression being strongest in the proliferative phase and survivin expression being strongest in the secretory phase. The up-regulation of survivin expression may be due to the concurrent rise in progesterone concentrations during the normal menstrual cycle. Moreover, survivin could play an important role independent of Bcl-2 in physiological homeostasis in the normal endometrium.
Article
Apoptosis plays a fundamental part in epidermal homeostasis, and apoptotic cells have been detected in normal and diseased skin. Little is known, however, on the inhibitory mechanisms of apoptosis at the skin level. In addition to bcl-2, a novel inhibitor of apoptosis designated survivin and structurally analogous to IAP apoptosis inhibitors has been recently identified. The expression of survivin in normal and pathologic skin was investigated. Immunohistochemical studies revealed that survivin is expressed in basal keratinocytes, but not in suprabasal epidermal layers, with a pattern similar to bcl-2. In western blots, the anti-survivin antibody recognized a single band of 16.5 kDa in protein extracts from normal human keratinocytes in culture, in agreement with the predicted size of survivin. In addition, survivin immunoreactivity was detected in benign and malignant melanocytic lesions, with strong expression in invasive lesions of melanomas. Whereas survivin staining was undetectable in benign epithelial tumors, such as seborrheic keratoses, it was observed in all epidermal layers in Bowen's disease. Interestingly, at variance with bcl-2, survivin was markedly expressed in squamous cell carcinoma, but virtually lacking in basal cell carcinoma, suggesting that these two apoptosis inhibitors may act through different anti-apoptotic pathways. Deregulation of survivin may influence both epidermal homeostasis and the development of melanoma and nonmelanoma skin cancer.
Article
A role of apoptosis (programmed cell death) in tumor formation and growth was investigated by targeting the apoptosis inhibitor survivin in vivo. Expression of a phosphorylation-defective survivin mutant (Thr(34)-->Ala) triggered apoptosis in several human melanoma cell lines and enhanced cell death induced by the chemotherapeutic drug cisplatin in vitro. Conditional expression of survivin Thr(34)-->Ala in YUSAC2 melanoma cells prevented tumor formation upon s.c. injection into CB.17 severe combined immunodeficient-beige mice. When induced in established melanoma tumors, survivin Thr(34)-->Ala inhibited tumor growth by 60-70% and caused increased apoptosis and reduced proliferation of melanoma cells in vivo. Manipulation of the antiapoptotic pathway maintained by survivin may be beneficial for cancer therapy.
A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma
  • G Ambrosini
  • C Adida
Ambrosini, G., Adida, C., and Altieri, D. 1997. A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat. Med. 3:917–921.
Inhibition of melanoma tumor growth in vivo by survivin targeting
  • D Grossman
  • P J Kim
  • J S Schechner
Grossman, D., Kim, P.J., Schechner, J.S., and Altieri, D.C. 2001. Inhibition of melanoma tumor growth in vivo by survivin targeting. Proc. Natl. Acad. Sci. USA. 98:635-640.
Cancer gene therapy using a survivin mutant adenovirus
  • M Mesri
  • N R Wall
  • J Li
  • R W Kim
Mesri, M., Wall, N.R., Li, J., Kim, R.W., and Altieri, D.C. 2001. Cancer gene therapy using a survivin mutant adenovirus. J. Clin. Invest. 108:981-990. DOI:10.1172/JCI200112983.