Categories
Articles

ABBS 2008,40(06): Experimental cancer gene therapy by multiple anti-survivin hammerhead ribozymes

Original Paper

Pdf

file on Synergy OPEN

omments

Acta Biochim Biophys

Sin 2008, 40: 466-477

doi:10.1111/j.1745-7270.2008.00430.x

Experimental cancer gene therapy by multiple anti-survivin

hammerhead ribozymes

Qi Fei1,2, Hongyu Zhang2, Lili Fu2, Xinlan Dai2, Baomei Gao2, Min Ni2, Chao Ge2, Jinjun Li2, Xia Ding3, Yuwen Ke3, Xuebiao Yao3*, and Jingde Zhu2*

1 Fudan University School of Medicine,

Shanghai 200032, China

2 Cancer Epigenetics and Gene Therapy Program,

The State Key Laboratory for Oncogenes and Related Genes, Shanghai Cancer

Institute, Shanghai Jiaotong University, Shanghai 200032, China

3 Division of Cellular Dynamics, Hefei

National Laboratory, University of Science and Technology of China, Hefei

230027, China

Received: April 6,

2008       

Accepted: May 5,

2008

This work was

supported by the grants from the Shanghai Science Foundation (No. 07DJ14074 to

JZ), the National Science Foundation (Nos. 30570850 and 10574134 to JZ), the National

Natural Science Foundation of China (No. 2004CB518804 to JZ and No.

2002CB713700 to XY), the National Basic Research Program of China (Nos.

2006AA02Z320 and 2006AA02Z197 to JZ), and the European 6th Program (No. LSHB-CT-2005-019067

to JZ). XY is a GCC Eminent Scholar

*Corresponding

authors:

Jingde Zhu:

Tel/Fax, 86-21-64224285; E-mail, [email protected]

Xuebiao Yao:

Tel/Fax, 86-551-3607141; E-mail, [email protected]

To improve the efficacy of gene therapy for

cancer, we designed four hammerhead ribozyme adenoviruses (R1 to R4) targeting

the exposed regions of survivin mRNA. In addition to the in vitro

characterization, which included a determination of the sequence specificity of

cleavage by primer extension, assays for cell proliferation and for in vivo

tumor growth were used to score for ribozyme efficiency. The resulting

suppression of survivin expression induced mitotic catastrophe and cell death

via the caspase-3-dependent pathway. Importantly, administration of the

ribozyme adenoviruses inhibited tumor growth in a hepatocellular carcinoma

xenograft mouse model. Co-expression of R1, R3 and R4 ribozymes synergistically

suppressed survivin and, as this combination targets all major forms of the survivin

transcripts, produced the most potent anti-cancer effects. The adenoviruses

carrying the multiple hammerhead ribozymes described in this report offered a

robust gene therapy strategy against cancer.

Keywords        hammerhead ribozyme; survivin;

hepatocellular carcinoma; gene therapy

Defects in apoptosis are a key hallmark of the malignant cells

resulting from such factors as the activation of anti-apoptotic genes and the

repression of pro-apoptotic genes [1]. A well-known anti-apoptotic factor is

survivin [2], which is overexpressed in all types of cancers but is

undetectable in normal adult cells. Survivin participates in a variety of

cellular processes such as cell division, angiogenesis, apoptosis etc [3].

Survivin negatively regulates apoptosis by interfering with

caspase-9-processing [4]. Despite our early findings that it binds to

Smac/DIABLO released from mitochondria [5], how survivin directs crossroad

traffic between cell division and apoptosis remains elusive.Besides serving as a biomarker for cancer diagnosis and prognosis

[6,7], survivin has been selected as a target for cancer intervention [3].

Forced expression of CDK1-nonphosphorylatable survivinT34A

mutant has displayed dominant-negative effects and induces dramatic cell death

[8,9]. Down-regulation of survivin expression by antisense oligonucleotide has

exhibited a promising outcome in xenograft models and phase I clinical trials

[10,11]. Other down-regulation approaches involving RNA interference (RNAi) also

have produced therapeutic effects in xenograft models [1214]. However,

there has been a long debate as to the specificity and durability of antisense

oligonucleotide and RNAi approaches in clinical therapy.Ribozyme is a powerful new therapeutic tool used to diminish RNA

molecules in cells [15]. The hammerhead ribozyme is the smallest and best

characterized. It consists of three parts: an invariant catalytic domain with

13 conserved nucleotides in a stem-loop structure and two arms located on each

side of the catalytic domain stretched complementary to the target RNA.

Hammerhead ribozymes cleave after NUH (where N can be any nucleotide, and H can

be any nucleotide except G) sequences of target mRNA to suppress the target

protein expression [16]. The substrate specificity of the

ribozyme is determined by the paired regions flanking the cleavage site.

Binding of ribozymes to its target mRNA is significantly more stringent than

short interfering RNA approaches, which often produce off-target effects [17,18].

We have used the expression cassette in pGVaL to ensure a high level

of expression of the stable ribozyme RNAs for better physical contact with

substrate RNA [19]. The substrate RNA has been systematically assessed in

vitro, in cell cultures and in mouse tumor xenograft model systems with the

human hepatocellular carcinoma as the targeted tumor type.

Materials and Methods

Anti-survivin ribozyme expression plasmid vectorsThe coding region (the A of ATG: 1 to A of TGA: 426) of the human survivin mRNA sequence (GenBank accession No.

BC008718) was analyzed for exposed regions (loops) using the MFOLD program (http://mfold.bioinfo.rpi.edu/cgi-bin/rna-form1.cgi).

The ribozyme sequences targeting H of the NUH triplet in each of four loops

were designated R1 to R4 and +61, +83, +232 and +358, respectively (Fig. 1).

Each pair of the oligonucleotides (Table 1) were phosphorylated,

annealed and cloned at SalI and PstI sites of pGVaL plasmid

vector [19] [Fig. 2(A)]. The constructs were designated pGVaL-R1 to R4.

Survivin expression plasmid vectorThe survivin coding region (the A of ATG: 1 to A of TGA: 426)

was amplified by plaque-forming unit-polymerase chain reaction from the cDNA

from SMMC-7721 liver cancer cells with the primers SurL 5?-CCGctcgag­atg­g­gtgccccgacg-3? and SurR 5?-GAagatctatcca­t­ggcagccagct-3? (BglII and XhoI recognition sequences are underlined,

respectively). This was followed by cloning in the downstream phase of the

three hemagglutinin (HA) tag sequences of pcDNA-HA vector to form HA-survivin

(HAS). The HindIII (blunt) and XbaI luciferase fragment of

pGL-3-basic (Promega, Madison, USA) was cloned at the BglII (blunt) and XbaI

sites of HAS to generate the plasmid HA-survivin-luciferase (HASL) [Fig.

2(C)].

Non-replicative adenoviral-ribozyme vectors The BamHI/NheI (blunt) fragment of pGVaL and pGVaL-R1

to R4, was placed at BglII/HindIII (blunt) sites of pDC vector

using the AdMax™ system (Microbix, Calgary, Canada) to create pDC-GVaL and

pDC-R1 to R4, respectively [Fig. 2(B)]. The EcoRI/SacI

(blunt) fragment of pDC-R1 was placed at EcoRI/SmaI sites of

pDC-R3 to create pDC-R13. The EcoRI/SacI (blunt) fragment of

pDC-R13 was placed at EcoRI/SmaI sites of pDC-R4 to create

pDC-R134. The adenoviral particles were made by the CreLoxP (3CreLoxP) mediated

recombination between each pDC vector and adenoviral vector (pBHGE3?E1) in

HEK 293 cells. The sequenced adenoviro/ribozyme particles were amplified,

titered and kept at 20 ?C until used. The adenoviruses were batch-produced and purified

by Shanghai Sino-Gene Company (Shanghai, China).The BamHI/NheI (blunt) fragment of pGVaL and pGVaL-R1

to R4, was placed at BglII/HindIII (blunt) sites of pDC vector

using the AdMax™ system (Microbix, Calgary, Canada) to create pDC-GVaL and

pDC-R1 to R4, respectively [Fig. 2(B)]. The EcoRI/SacI

(blunt) fragment of pDC-R1 was placed at EcoRI/SmaI sites of

pDC-R3 to create pDC-R13. The EcoRI/SacI (blunt) fragment of

pDC-R13 was placed at EcoRI/SmaI sites of pDC-R4 to create

pDC-R134. The adenoviral particles were made by the CreLoxP (3CreLoxP) mediated

recombination between each pDC vector and adenoviral vector (pBHGE3?E1) in

HEK 293 cells. The sequenced adenoviro/ribozyme particles were amplified,

titered and kept at 20 ?C until used. The adenoviruses were batch-produced and purified

by Shanghai Sino-Gene Company (Shanghai, China).

In vitro analysis of the anti-survivin

ribozymesBoth ribozymes and HASL RNAs were made by the in vitro

transcription of 2 mg linearized pGVaL/pGVaL-R4 (by XbaI) and 2 mg HASL (by HindIII),

respectively, in vitro transcribed with T7 RNA polymerase (TaKaRa,

Dalian, China). This was followed by the removal of the DNA template by DNase

I. The transcripts were then purified and quantified by semi-quantitative

reverse transcription-polymerase chain reaction (RT-PCR) using a known amount

of plasmid DNA as a reference.For the in vitro ribozyme cleavage reaction of HASL RNAs,

equal molar amounts (1 mmol) of ribozyme and the HASL RNAs were heat denatured

in 10 ml of 50 mM Tris (pH 7.5)/1 mM EDTA, quickly cooled and followed by

the addition of MgCl2 for a final concentration of 10 mM to

initiate the cleavage reaction at 37 ?C for 60 min. The cleavage products were

NH4Ac/ethanol precipitated. They were analyzed for cleavage specificity

by primer analysis and for cleavage efficacy by determining the relative

luciferase activity and protein amount (Western blot analysis with the HA

antibody) of the in vitro translated products. The non-virus control

(mock) was made from the cleavage reaction with the in vitro transcribed

RNAs from pGVaL.Two-thirds of both mock and cleaved transcripts were subjected to

primer extension analysis with the 5?-end [32P]-labeled

DNA primer by T4 polynucleotide kinase [SPE1 for R1 and R2, SPE2 for R3 and

SPE3 for R4, Fig. 1(C)]. The sequence ladders were made with the same

primer, and the HASL template was created by T7 DNA polymerase-based

manual-sequencing (Amersham, Buckinghamshire, UK). The remaining one-third of the in

vitro cleavage products were in vitro translated (Rabbit

Reticulocyte Lysate System; Promega), one-fifth of which were measured for

luciferase activity (Promega) in a LB 9506 Lumat luminometer (EG&G,

Gaithersburg, USA) and plotted. The cleavage reaction by all four ribozymes was

also included in this analysis. The remaining four-fifths of the products were

analyzed with anti-HA antibody (Santa Cruz Biotechnology, Santa Cruz, USA) and

visualized by SuperSignal West Pico Chemiluminescent Substrate (Pierce,

Rockford, USA). The relative densitometric reading of the cleaved over that of

the mock (as 100%) was calculated.

Evaluation of the anti-survivin adeno/ribozyme effect in

hepatocellular cellsSMMC-7721 liver cancer cell line (Cell Bank No. TCHu68; China), and PLC

hepatocarcinoma cells (ATCC No. CRL-8024) were cultured at 37 ?C with 5% CO2 in Dulbecco’s modified eagle’s medium (DMEM; Invitrogen, Carlsbad,

USA) supplemented with 10% fetal bovine serum (PAA, Pasching, Austria) or 10%

newborn bovine serum (Sijiqing, Hangzhou, China). The log-phase cells were

continuously infected for 72 h by each virus followed by a

3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) reading for

cell viability. The relative MTT reading was calculated by the following formula.

The cells were collected at 48 h for RT-PCR and Western blot analysis.

Eq.

The semi-quantitative RT-PCR analyses were carried out for survivin,

b-actin and ribozymes, respectively, with the primer pairs (Table

2). The expression level of the survivin mRNA was presented as a

ratio (the survivin over b-actin densitometric quantification) of the infected cells over the

mock control. Western blot analyses of survivin (anti-survivin antibody; Santa

Cruz Biotechnology) and proliferating cell nuclear antigen (PCNA) (anti-PCNA

antibody; Santa Cruz Biotechnology) were performed on a 15% SDS-polyacrylamide

gel. Western blot analysis of poly(ADP-ribose) polymerase (PARP) (85 kDa;

Promega)/PCNA was carried out on a 7.5% SDS-polyacrylamide gel. The relative

abundance of the survivin or PARP protein was presented in the density ratio of

that over PCNA of the viral-infected cells over that (arbitrarily as 100%) of

the non-infected cells.

Immunofluorescence microscopySMMC-7721 cells were infected with the control (GVaL) and survivin

ribozymes (R3 or R134), respectively, followed by fixation in 4% formaldehyde

24 h after infection. Coverslips were then permeabilized with 0.2% Triton X-100

and blocked with PBS containing 0.05% Tween-20 with 1% bovine serum albumin

(Sigma, St. Louis, USA). Cells were incubated with primary antibodies (tubulin

mouse antibody DM1A, 1:5000 dilution; human anti-centromere antibody ACA,

1:2000 dilution) in a humidified chamber for 1 h and then washed three times in

PBS containing 0.05% Tween-20. Texas red-conjugated goat antihuman IgG+IgM and

fluorescein-isothiocyanate-conjugated rabbit antimouse IgG (Jackson Immuno­Research,

West Grove, USA) were used as the secondary antibodies for visualization of

appropriate antigens. DNA was stained with TOTO-3 dye (Invitrogen). Slides were

examined with a Leica SP5 confocal microscope (Leica, Heerbrugg, Switzerland)

and images were presented with Photoshop.

Inhibitory effect of the anti-survivin adeno/ribozyme on tumor

xenograft modelThe log-phase SMMC-7721 cells were infected with the virus at

multiplicity of infection (MOI) of 10 or 25 for 24 h at 37 ?C before being

injected subcutaneously into both flanks (3?106 cells/flank) of each mouse (BALB/c, nu/nu).

Each group consists of three mice; the left flank of each mouse was injected

with SMMC-7721 cells or adeno/GVaL (non-ribozyme viral control) while the right

flank was injected with the cells infected with each adeno/ribozyme virus under

study. Tumor growth was monitored and measured until the experiment terminated

at week 4, when the tumor mass was measured. The weight ratio of the tumor over

the mock was determined for each mouse. All animal experiments were approved by

the Institutional Review Board and the Animal Research Committee.The tumor was fixed, sectioned, hematoxylin and eosin (H&E)

stained or immunostained for the expression of survivin (1:10 dilution of

anti-survivin antibody; FL-142, Santa Cruz Biotechnology) and ki67 protein

(1:10 dilution of anti-Ki67 antibody; Novocastra, Newcastle, UK), and

visualized by the EnVision System (Dako, Carpinteria, USA).

Results

Design of the anti-survivin hammerhead ribozymes Four hammerhead ribozymes were designed to cleave respectively at

nucleotide +61 (R1), +83 (R2), +232 (R3), and +358 (R4) of the predicted open

frames of survivin mRNA (Fig. 1). The genes were then embedded in

the adenoviral Va I RNA of the pGVaL vector [19] [Fig. 2(A)]. By placing

the ribozyme genes at the center of a loop formed from an inverted repeat

sequence that was artificially introduced into the Val I gene [19]

[pGVaL-Rx in Fig. 2(A)], the ribozyme sequences were expected to

protrude from the Va I RNA region. The BamHI and NheI fragment of

each pGVaL-R was cloned into the adenoviral vector (Microbix pDC-316 based) to

facilitate analysis of both the cellular level and the entire animal and that

will ultimately be used to treat cancer in clinical trials [Fig. 2(B)].

The HA-tagged survivin-luciferase fusion gene was put into a pcDNA 3.1-based

vector to create HASL [Fig. 2(C)], so that evaluation of the ribozyme

mediated cleavage in vitro of survivin RNA could be readily

carried out by primer extension (for both cleaving specificity and efficacy)

and luciferase activity (for cleaving efficacy).

Evaluation of survivin mRNA’s ribozyme-cleavage specificity

and efficacy Except for R3, which failed to target DEx3, all the remaining

ribozymes were expected to cleave all the three major forms of survivin

mRNAs [20,21] [Fig. 3(A)]. Equal amounts of both ribozymes and the survivin

RNAs made by in vitro transcription were incubated in the presence of Mg2+ for the cleavage reaction. Primer extension analysis of the

resulted survivin RNAs showed that each ribozyme cleaved the survivin

RNA at the predicted sites [arrowed in Fig. 3(B)]. Judged from its band

density relative to its expected size, R2 appeared to be least efficient

ribozyme. The in vitro translated products of the RNA remaining from the

cleavage reaction were, respectively, assayed for luciferase activity [Fig.

3(C)] and for the size of the HA-associated protein band in Western blot

analysis [Fig. 3(D)] to semi-quantitatively determine the efficacy of

the ribozyme cleavage. The luciferase activity from the mock (100%) was reduced

to 62.7% in R1, 70.0% in R2, 40.4% in R3, 57.0% in R4, and 28.7% in a

combination of the four ribozyme experiments [Fig. 3(C)] and the

HA-associated band density to 48.8% in R1, 124.4% in R2, 28.7% in R3, 61.0% in

R4 and 16.5% in a combination of the four ribozyme experiments [Fig. 3(D)].

As the in vitro analyses indicated, R3 was the most effective, followed

by R1, R4 and R2. There was an additive effect in cleavage by all the four

ribozymes in combination [Fig. 3(C,D)]. In view of the protein-free RNA

in vitro analyses [Fig. 3], the exposed region of the substrate

RNA by the MFOLD program was indeed informative for the rational design of the

hammerhead ribozyme.

Ribozyme-mediated cleavage of survivin mRNA-induced apoptosis

and mitotic catastrophe in cells SMMC-7721 hepatocellular carcinoma cells were challenged with the

non-replicative adenoviral particles carrying a single ribozyme gene (R1 to

R4), three ribozymes (R1, R3 and R4) in tandem (R134) or no ribozyme control

(GVaL) for 72 h. The loss of cell viability then was assayed by MTT assay [Fig.

4(A)], by a semi-quantitative PCR analysis for repression of survivin

expression [Fig. 4(B)] and by Western blot analysis [Fig. 4(C)].

Western blot analysis was also used to assess the apoptotic index for the level

of PARP protein [Fig. 4(D)]. Consistent with the in vitro observations (Fig. 3), R2

was the least effective ribozyme; infection at MOI 50 reduced the MMT reading

by no more than 20%. R134 was more effective than the most potent single

ribozyme, R3; infection at MOI 12.5 reduced the MTT reading by 23.2% versus

38.6% and at MOI 25 by 15.2% versus 27.8% [Fig. 4(A)]. The levels of

ribozyme and b-actin RNAs were comparable, but the survivin mRNA level

varied with the ribozyme viruses: GVaL, 91.4% of that of the SMMC-7721 cells;

R1, 36.1%; R2, 108.3%; R3, 33.8%; R4, 60.5%; and R134, 22.0% [Fig. 4(B)].

These results reflected the relative potency of the ribozyme against survivin

mRNA, which was also confirmed by Western blot analysis of survivin protein [Fig.

4(C)]. PARP protein (85 kDa) was a specific cleaved product of PARP-1 by

caspase-3 in the cell undergoing apoptosis [22]; the protein’s level increased

from 100% in mock infected cells to 164.0% by R2, 479.9% by R1, 768.8% by R3,

460.1% by R4 and 1339.3% by R134 in viral-infected cells [Fig. 4(D)]. We

also performed the same set of analyses on another hepatocellular cell line PLC

(ATCC No. CRL-8024) and yielded comparable results (Fig. 5).An immunofluorescence study showed that when survivin was depleted

of R3 and R134, respectively, SMMC-7721 cells exhibited aberrant chromosome

segregation [Fig. 4(E), d’, d”], a phenotype associated with

mitotic catastrophe. Multipolar spindles were typical phenotypes seen in 83%±7%

of the infected cells where mitotic chromosomes failed to align at the equator

and enter into anaphase [Fig. 4(E), c’, c”], compared to 5%±3% in

GVaL-infected control (n=4). The microtubule integrity and centromere

labeling by anti-centromere antibody ACA remained unaltered, confirming the

specificity of the ribozyme treatment [Fig. 4(E), a’, a”].

Adenoviral-ribozymes prevented and suppressed growth of the

hepatocellular tumor xenografts in mice SMMC-7721 cells were infected with viruses at MOI 25 for 24 h prior

to injection into mice. The right flanks of nude mice were injected with

SMMC-7721 cells that had been pre-infected with the ribozyme viruses, while the

left flank received cells with either no virus (mock) or the empty vector-virus

(GVaL) pre-infection [Fig. 6(A)]. While no difference in growth profile

and tumor mass was observed in the mock-infected SMMC-7721 cells [Fig. 6(B,D)],

all the ribozyme viruses, except for R2, prevented tumor growth. The tumors

from the cells pretreated with R2, the least effective ribozyme, grew

significantly more slowly than those pretreated with the mock, and at 35 d when

the experiment was ended, had a smaller mass by a ratio of 0.64: 1 [Fig.

6(C,D)]. Again neither R3 nor R134 at MOI 10 led to tumor growth; the cells

that received the R4 virus grew more slowly than the GVaL-infected cells [Fig.

6(EG)]. Consistent with the reduced tumor

growth, expressions of both survivin [Fig. 6(I)] and ki67 proteins (a

marker for cell proliferation) [Fig. 6(J)] were significantly repressed

in tumors derived from the cells pre-infected with either R2 at MOI 25 or R4 at

MOI 10. The growth profile of the GVaL-infected SMMC-7721 cells might have

reflected the potential to support tumor expansion that varied with individual

animals. The observation that tumor mass in R134 (100%) by the GVaL infection

was 2-fold bigger than that in R3 (31%) experiments [Fig. 6(K,L)]

favored the conclusion from the previous analyses that three ribozymes acting

together in R134 would have a greater anti-cancer effect than any single

ribozyme (Figs. 3 and 4).To mimic the clinical practice of cancer gene therapy, R3 or R134

viruses were injected in a regime of 3?106 three times every other day into the SMMC-7721

cell-derived tumor (at a volume of approximately 150 mm3) at the

right flank. This resulted in a reduction of tumor growth by 69.5% (R3) and

55.5% (R134) as well as a reduction in tumor volume by 64.8% (R3) and 57.3%

(R134) [Fig. 7(AC)]. Both survivin and ki67 expression

was significantly reduced in the tumors injected R3 or R134 viruses than the

control viruses [Fig. 7(E,F)].

Discussion

The survivin gene has been regarded as a rational target for

cancer treatment using new molecular antagonists, cancer vaccines and gene

therapeutic agents [3]. Small-molecule antagonists against survivin, including

tetra-O-methyl nordihydroguaiaretic acid to repress Sp1 dependent survivin gene

expression and 17-allyl-amino-geldanamycin or shepherdin to disrupt

survivin/Hsp90 interaction [23–26], have entered phase I and II clinical

trials. The use of hammerhead ribozymes seems more advantageous than both RNAi,

because of its target specificity, and antisense, because of its deliverability

by conventional gene therapeutic vectors, including the adenoviral vector used

in this report. The sequence targeted by the R1 in this report is identical to

CUA110, the ribozyme described by Pennati et al. that was also placed

directly within an adenoviral Va I RNA gene in a retroviral vector [25].

However, all characterizations, including cell viability, apoptosis and in

vivo tumor growth, have been performed in stable transformed tumor cell

lines. Choi et al. constructed a hammerhead ribozyme against the same

sequence motif as R3 in this work, where the ribozyme RNA was expressed on its

own in an adenoviral system [14]. Both in vitro and in cell results

demonstrate its ability to cleave survivin RNA as expected and to induce

apoptosis; however, the ability of in vivo to inhibit tumor growth has

not been demonstrated.Here, we show both the specificity and efficacy of each of the four

hammerhead ribozymes to cleave the survivin RNA in vitro (Fig. 3),

in cell (Figs. 4,5), and in vivo (Figs. 6,7). A

combination of three ribozymes (R1, R3 and R4) synergistically suppress

survivin expression [19], which is likely attributable to the fact that there

are three alternately processed survivin transcripts, designated as survivin

(full length), survivin-DEx3 (lacking exon 3) and survivin-2B (retaining part of intron 2 as

a cryptic exon). All forms of survivin RNAs are required for full-blown

apoptosis and aberrant mitosis [20]. A high level of survivin-DEx3 reportedly

correlates with a poorer prognosis in acute myeloid leukemia and cannot be

cleaved by R3 [21], the most potent single ribozyme. Therefore, using R134

virus, which combines three effective ribozymes, should be advantageous, as it

has a higher efficacy at cleaving all the spliced forms of survivin RNA.In addition to the demonstration of the robust therapeutic potential

of hammerhead ribozyme-mediated depletion of survivin expression, this report

has detailed a strategy to develop an effective hammerhead ribozyme that aims

to repress the expression of a given gene, has a rational design that provides

both specificity and efficacy in vitro, in cell culture and in animal,

and includes vectors that will address most, if not all, major technical

concerns.

References

 1   Hanahan D, Weinberg RA. The hallmarks of

cancer. Cell 2000, 100: 5770

 2   Salvesen GS, Duckett, CS. IAP proteins:

blocking the road to death’s door. Nat Rev Mol Cell Biol 2002, 3: 401410

 3   Altieri DC. Survivin, cancer networks and

pathway-directed drug discovery. Nat Rev Cancer 2008, 8: 6170

 4   Dohi T, Beltrami E, Wall NR, Plescia J,

Altieri DC. Mitochondrial survivin inhibits apoptosis and promotes

tumorigenesis. J Clin Invest 2004, 114: 11171127

 5   Song Z, Yao X, Wu M. Direct interaction

between survivin and Smac/DIABLO is essential for the anti-apoptotic activity

of survivin during taxol-induced apoptosis. J Biol Chem 2003, 278: 2313023140

 6   Schultz IJ, Witjes JA, Swinkels DW, de Kok JB.

Bladder cancer diagnosis and recurrence prognosis: comparison of markers with

emphasis on survivin. Clin Chim Acta 2006, 368: 2032

 7   Parker AS, Kosari F, Lohse CM, Thompson RH,

Kwon ED, Murphy L, Riehle DL et al. High expression levels of survivin

protein independently predict a poor outcome for patients who undergo surgery

for clear cell renal cell carcinoma. Cancer 2006, 107: 3745

 8   Mesri M, Wall NR, Li J, Kim RW, Altieri DC.

Cancer gene therapy using a survivin mutant adenovirus. J Clin Invest 2001,

108: 981990

 9   Tu SP, Cui JT, Liston P, Huajiang X, Xu R,

Lin MC, Zhu YB et al. Gene therapy for colon cancer by adeno-associated

viral vector-mediated transfer of survivin Cys84Ala mutant. Gastroenterology

2005, 128: 361375

10  Li F, Ackermann EJ, Bennett CF, Rothermel AL,

Plescia J, Tognin S, Villa A et al. Pleiotropic cell-division defects

and apoptosis induced by interference with survivin function. Nat Cell Biol

1999, 1: 461466

11  Zangemeister-Wittke U. Antisense to apoptosis

inhibitors facilitates chemotherapy and TRAIL-induced death signaling. Ann N Y

Acad Sci 2003, 1002: 9094

12  Caldas H, Holloway MP, Hall BM, Qualman SJ,

Altura RA. Survivin-directed RNA interference cocktail is a potent suppressor

of tumour growth in vivo. J Med Genet 2006, 43: 119128

13  Uchida H, Tanaka T, Sasaki K, Kato K, Dehari

H, Ito Y, Kobune M et al. Adenovirus-mediated transfer of siRNA against

survivin induced apoptosis and attenuated tumor cell growth in vitro and

in vivo. Mol Ther 2004, 10: 162171

14  Choi KS, Lee TH, Jung MH. Ribozyme-mediated

cleavage of the human survivin mRNA and inhibition of antiapoptotic function of

survivin in MCF-7 cells. Cancer Gene Ther 2003, 10: 8795

15  Sullivan SM. Development of ribozymes for gene

therapy. J Invest Dermatol 1994, 103: 85S89S

16  Haseloff J, Gerlach WL. Simple RNA enzymes

with new and highly specific endoribonuclease activities. Nature 1988, 334: 585591

17  Furue H. Topoisomerase inhibitors developing

in Japan. Gan To Kagaku Ryoho 1993, 20: 4249

18  Tanner NK. Ribozymes: the characteristics and

properties of catalytic RNAs. FEMS Microbiol Rev 1999, 23: 257275

19  Lieber A, Strauss M. Selection of efficient

cleavage sites in target RNAs by using a ribozyme expression library. Mol Cell

Biol 1995, 15: 540551

20  Caldas H, Jiang Y, Holloway MP, Fangusaro J,

Mahotka C, Conway EM, Altura RA. Survivin splice variants regulate the balance

between proliferation and cell death. Oncogene 2005, 24: 19942007

21  Wagner M, Schmelz K, Wuchter C, Ludwig WD,

Dorken B, Tamm I. In vivo expression of survivin and its splice variant

survivin-2B: impact on clinical outcome in acute myeloid leukemia. Int J Cancer

2006, 119: 12911297

22  Ame JC, Spenlehauer C, de Murcia G. The PARP

superfamily. Bioessays 2004, 26: 882893

23  Chang CC, Heller JD, Kuo J, Huang RC.

Tetra-O-methyl nordihydroguaiaretic acid induces growth arrest and cellular

apoptosis by inhibiting Cdc2 and survivin expression. Proc Natl Acad Sci U S A

2004, 101: 1323913244

24  Sausville EA, Tomaszewski JE, Ivy P. Clinical

development of 17-allylamino, 17-demethoxygeldanamycin. Curr Cancer Drug

Targets 2003, 3: 377383

25  Plescia J, Salz W, Xia F, Pennati M, Zaffaroni

N, Daidone MG, Meli M et al. Rational design of shepherdin, a novel

anticancer agent. Cancer Cell 2005, 7: 457468

26  Gyurkocza B, Plescia J, Raskett CM, Garlick

DS, Lowry PA, Carter BZ, Andreeff M et al. Antileukemic activity of

shepherdin and molecular diversity of hsp90 inhibitors. J Natl Cancer Inst

2006, 98: 10681077