1
SilenciX ® , novel stable knock-down cellular models to screen new molecular targets through the synthetic lethality approach PARP inhibitors are promising therapeutic drugs for cancers harboring BRCA1 and BRCA2 mutations. PARP is involved in the Base Excision Repair (BER) DNA repair pathway. Tumors with BRCA mutations are deficient in Homologous Recombination (HR), another DNA repair pathway. To maintain DNA integrity and prevent cell death, at least one of the BER or HE pathways must be functional, while the other may be inhibited or silenced (e.g. mutation, drug). In drug discovery, Synthetic Lethality (SL) used a cancer mutation and a drug in combination to cause the tumor cell death. SL is thus a promising approach to find new druggable targets by controlling the HR and BER DNA repair pathways in order to selectively kill mutated cancer cells. However, the main technical hurdle in SL approaches is to easily identify cancer-specific molecular interaction in various cancer types, while using robust and validated experi- mental cellular models. In this presentation we demonstrate how the SilenciX ® technology meets SL experimental requirements. BRCA1 and BRCA2 SilenciX ® cells were treated with PARP1 inhibitors (Olaparib, Veliparib, Rucaparib) and with Gemcitabine. All inhibitors except Gemcitabine were synthetic lethal in BRCA1 SilenciX ® cell lines. To mimic the cancer micro-environment, the model was tested under hypoxic conditions as the common denominator of growing solid tumors. Material & Method Proliferation assay on SilenciX ® HeLa cells were transfected using Lipofectamine 2000 with SilenciX ® vectors, containing siRNA for BRCA1, BRCA2, p53 and control sequence, as described by Biard (1) . We established BRCA1, BRCA2, p53 SilenciX ® cell lines and Control SilenciX ® cell lines. The cells were plated in 96-well plates at 3000 cells/cm 2 . After 24 h, cells were treated in triplicate with Olaparib, Veliparib, Rucaparib or Gemci- tabine at various concentrations from 100 µM to 0.1 nM. Cells were main- tained for 7 days at 37°C, 5% CO 2 . In hypoxia, cells were incubated at 1% O 2 in a C-chamber from Biospherix in which gas level was controlled by ProOx 110 oxygen controller. The cell proliferation assay was performed by utilizing the tetrazolium salt WST-8 according to the manufacturer’s instructions. From raw data, the Normalised Cell Survival was calculated as the percentage of living treated cells compared to untreated cells. To compare the level of synthetic lethality between control and knockdown cells, the results were expressed as IC50control / IC50KD ratio both in normoxia and in hypoxia. The experiment was performed three times with similar results. Figure 1a/1b. Immunocytochemical staining of knock-down BRCA1 and p53 HeLa SilenciX cells with primary antibody anti-BRCA1 and anti-p53. Control HeLa cells have been transformed with a control shRNA. BRCA1 DAPI Control HeLa SilenciX BRCA1 HeLa SilenciX p53 DAPI Control HeLa SilenciX p53 HeLa SilenciX Results I/ Validation of SilenciX ® cell lines Knock-down validation was performed by qRT-PCR for each silenced cell line compared to the control cell line. The silencing of p53, BRCA1 and BRCA2 was respectively 98%, 71% and 72%. Some data of Immunocytochemical staining of knock-down BRCA1 and p53 HeLa SilenciX ® cells were available and kindly provided by Dr D. Biard. Introduction II/ SilenciX as in vitro model for SL studies PARP inhibitors induce SL in cancer cells (2,3,4) . PARP activation is one of the cellular response to metabolic, chemical or radiation-induced single strand break DNA damage (2) . Our SilenciX ® cell lines, BRCA1KD, BRCA2KD and p53KD are used to mimic cancer cells with specific gene mutations in order to evaluate SL interactions between these mutations and PARP inhibitors. The three PARP inhibitors Olaparib, Veliparib and Rucaparib are in clinical development for cancer therapy (5, 6) . Moreover, hypoxia has important effects on chemosensitivity of cancer cells (7) but the consideration of its effect on the efficacy of synthetic lethal drugs is recent (8,9) . Therefore, the hypoxic conditions present in tumors need to be better considered to evaluate thera- peutic drugs. For this reason, cell survival assays were carried out to assess the sensitivity of BRCA1, BRCA2 and p53 SilenciX ® cells to PARP inhibitors (Olaparib, Veliparib and Rucaparib) and to a DNA synthesis inhibitor, Gemcitabine, in normoxic and hypoxic conditions. Figure 2a/2b. SilenciX ® Cell survival after 7 days drug treatment. SilenciX ® cells were incubated with Olaparib (A, B), Gemcitabine (C, D), Veliparib (E, F) and Rucaparib (G, H) and survival assays were carried out in normoxia (A, C, E, G) and hypoxia (B, D, F, H). Table 1. IC50 calculation from Cell survival assay. Cell survival assays showed that PARP inhibitors are less synthetic lethal in hypoxic conditions with increased IC50 and survival percentage at higher concentrations. These higher survival percentages were in part a result of a decrease of cell growth in hypoxic condition. However, the difference of IC50 probably involves the signaling pathway of PARP1. PARP1 is linked to the Hypoxia-inducible factor 1 (HIF-1) which is the key transcription factor regulating hypoxia-dependent gene expres- sion (10,11) . Indeed, PARP1 binds directly to HIF-1 and, thus, activates HIF1-dependent gene expression (10) . The low level of oxygen might disrupt the synthetic lethality relationship between PARP1 and BRCA1/2 through HIF-1 pathway. The IC50 ratios between Control SilenciX ® cells and knocked down cells revealed that olaparib is the most effective inhibitor in our in vitro model. In addition, olaparib, veliparib and rucaparib were more toxic to BRCA1-deficient cells. The DNA synthesis inhibitor, Gemcitabine, had no clear synthetic lethal relationship with BRCA1, BRCA2 and p53 genes. Cancer cells are characterized by their lack in oxygen delivery that leads to hypoxia. This has direct effects on the design of treatment of hypoxic tumors. Indeed tumor vessel normalization has recently been demonstrated with success in efficiently improving drug efficacy (12, 13) . The hypoxic microenvironment clearly alters DNA damage repair pathways (14) . The use of DNA repair inhibitors in combination with chemotherapy or radiotherapy can enhance tumor death but also increase toxicity towards normal cells. Targeting inter-gene relationship of synthetic lethality between DNA repair genes is an effective strategy to over- come this issue. Synthetic lethality is widely exploited to selectively kill to tumor cells and spare normal cells (15) . The best known SL relationship is between BRCA1/2 mutation and PARP1 inhibitors. Others have been reported as BRCA2 / ATM mutation and APE1 inhibitors (16) , PTEN mutation and PARP inhibitors (17) or XRCC1 mutation and ATM / DNA-PKcs inhibitors (18) . A simple, robust and stable in vitro model is a powerful tool for screening in this new SL therapeutic approach. Such a tool may be used to screen new drug libraries in order to find out new SL relationship. It can also help clarifying the underlying molecular mechanisms of a specific SL relationship. SilenciX ® by combining the RNAi technology with pEBV-derived vectors provides an exceptional opportunity to rapidly create a set of stable knock-down clones, able to be cultivated for long-term and with multiple freeze-thaw cycles. Such an in vitro model is still far from the tumor micro-environment. Indeed, the oxygen partial pressure varies between 11% and 1% O 2 in normal human tissues and represents the physioxic condition (19) . Working with cell cultures at an oxygen percentage of 20% could greatly affect the screening results. Moreover, within the tumor microenvironment, a gas gradient exists between the oxygenated exterior and anoxic core. More advanced physiologic in vitro models include multiple cell type 3D microspheroid models (20) which are not easily adaptable to High Throughput Screening. Our SilenciX ® model used in gas controlled cell culture environment represents a useful tool for screening in early drug discovery. Discussion References 1. Biard, Nucleic Acids Research, 2007. 2. Satoh, et al., Nature, 1992. 3. Farmer, et al., Nature, 2005. 4. Lord, et al., Curr. Opin. Pharmacol., 2008. 5. Reinbolt and Hays. Frontiers in oncology, 2013. 6. Rajan, et al., Clin Cancer Res, 2012. 7. Strese, et al., BMC Cancer, 2013. 8. Chan and Bristow, Clin Cancer Res, 2010. 9. Chan et al.,Cancer Res, 2010. 10. Elser, et al., Mol Cancer Res, 2008. 11. Martin-Oliva, et al., Cancer Res, 2006. 12. Kiéda, et al., J. Mel. Med., 2013. 13. Collet, et al., Mol Cancer Ther., 2013. 14. Bristow, et al., Nature Reviews Cancer, 2008. 15. Shaheen et al., Blood, 2011. 16. Sultana et al., Int J Cancer, 2012. 17. Mendes-Pereira et al., EMBO Mol Med., 2009. 18. Sultana et al., Cancer Res., 2013. 19. Carreau et al., J Cell Mol Med., 2011. 20. Thoma et al., Advanced Drug Delivery Reviews, 2014. We would like to address our thanks and acknowledgements to: - Denis Biard, inventor of the SilenciX ® technology, - Fabienne Fasani, for her contribution to the cell culture experiments in hypoxia conditions, - Stephen Durant, for his sponsoring to our participation at the 2014 AACR meeting. Eric Mennesson 1 *, Anne-Marie Renault 1 *, Isabelle Fixe 1 *, Catherine Grillon 2 , Claudine Kiéda 2 , Nadia Normand 1 * Authors contributed equally to this work - 1 tebu-bio, 39 rue de Houdan, 78612 Le Perray-en-Yvelines, France - 2 Centre de Biophysique Moléculaire, CNRS UPR 4301, Rue Charles Sadron, 45071 Orléans Cedex 2, France SilenciX ® is a registered trademark of tebu-bio, technology licensed from the CEA. Fig 1-a Fig 2-a Fig 1-b Fig 2-b Abstract #3733 Conclusion SilenciX ® models are ingenious gene-specific knock-down engineered cell lines without integration in the host genome and without off-target effects. They are suitable for a broad range of synthetic lethality studies. Cells other than HeLa cells may be used to build relevant cancer disease models. Such pathology models stably silenced for a specific gene can now be used to mimic tumor microenvironment. The access to SilenciX ® synthetic lethality approach and its use in oxygen controlled conditions similar to tumor physioxia open a new landscape for drug screening. Acknowledgements

SilenciX , novel stable knock-down cellular models to ... stable KD cellular... · * Authors contributed equally to this work - 1 tebu-bio, 39 rue de Houdan, 78612 Le Perray-en-Yvelines,

Embed Size (px)

Citation preview

SilenciX®, novel stable knock-down cellular models to screen new molecular targets through the synthetic lethality approach

PARP inhibitors are promising therapeutic drugs for cancers harboring BRCA1 and BRCA2 mutations. PARPis involved in the Base Excision Repair (BER) DNA repair pathway. Tumors with BRCA mutations are deficient in Homologous Recombination (HR), another DNA repair pathway. To maintain DNA integrityand prevent cell death, at least one of the BER or HE pathways must be functional, while the other maybe inhibited or silenced (e.g. mutation, drug). In drug discovery, Synthetic Lethality (SL) used a cancer

mutation and a drug in combination to cause the tumor cell death. SL is thus a promising approach tofind new druggable targets by controlling the HR and BER DNA repair pathways in order to selectivelykill mutated cancer cells. However, the main technical hurdle in SL approaches is to easily identifycancer-specific molecular interaction in various cancer types, while using robust and validated experi-mental cellular models. In this presentation we demonstrate how the SilenciX® technology meets SL

experimental requirements. BRCA1 and BRCA2 SilenciX® cells were treated with PARP1 inhibitors (Olaparib, Veliparib, Rucaparib) and with Gemcitabine. All inhibitors except Gemcitabine were synthetic lethal in BRCA1 SilenciX® cell lines. To mimic the cancer micro-environment, the modelwas tested under hypoxic conditions as the common denominator of growing solid tumors.

Material & Method

Proliferation assay on SilenciX®

HeLa cells were transfected using Lipofectamine 2000 with SilenciX®

vectors, containing siRNA for BRCA1, BRCA2, p53 and control sequence,as described by Biard(1). We established BRCA1, BRCA2, p53 SilenciX® celllines and Control SilenciX® cell lines. The cells were plated in 96-well plates at 3000 cells/cm2. After 24 h, cellswere treated in triplicate with Olaparib, Veliparib, Rucaparib or Gemci-tabine at various concentrations from 100 µM to 0.1 nM. Cells were main-tained for 7 days at 37°C, 5% CO2. In hypoxia, cells were incubated at1% O2 in a C-chamber from Biospherix in which gas level was controlledby ProOx 110 oxygen controller.The cell proliferation assay was performed by utilizing the tetrazoliumsalt WST-8 according to the manufacturer’s instructions. From raw data,the Normalised Cell Survival was calculated as the percentage of livingtreated cells compared to untreated cells. To compare the level of syntheticlethality between control and knockdown cells, the results were expressedas IC50control / IC50KD ratio both in normoxia and in hypoxia. The experiment was performed three times with similar results.

Figure 1a/1b. Immunocytochemical staining of knock-down BRCA1and p53 HeLa SilenciX cells with primary antibody anti-BRCA1 andanti-p53. Control HeLa cells have been transformed with a controlshRNA.

BRCA1 DAPI

Control HeLaSilenciX

BRCA1 HeLaSilenciX

p53 DAPI

Control HeLaSilenciX

p53 HeLa SilenciX Results

I/ Validation of SilenciX® cell lines Knock-down validation was performed by qRT-PCR for each silencedcell line compared to the control cell line. The silencing of p53, BRCA1and BRCA2 was respectively 98%, 71% and 72%. Some data of Immunocytochemical staining of knock-down BRCA1 and p53 HeLa SilenciX® cells were available and kindly provided by Dr D. Biard.

Introduction

II/ SilenciX as in vitro model for SLstudies PARP inhibitors induce SL in cancer cells (2,3,4). PARP activation is one ofthe cellular response to metabolic, chemical or radiation-induced singlestrand break DNA damage(2). Our SilenciX® cell lines, BRCA1KD, BRCA2KDand p53KD are used to mimic cancer cells with specific gene mutationsin order to evaluate SL interactions between these mutations and PARPinhibitors. The three PARP inhibitors Olaparib, Veliparib and Rucaparibare in clinical development for cancer therapy (5, 6).

Moreover, hypoxia has important effects on chemosensitivity of cancer cells (7) but the consideration of its effect on the efficacy ofsynthetic lethal drugs is recent (8,9). Therefore, the hypoxic conditionspresent in tumors need to be better considered to evaluate thera-peutic drugs. For this reason, cell survival assays were carried out toassess the sensitivity of BRCA1, BRCA2 and p53 SilenciX® cells to PARPinhibitors (Olaparib, Veliparib and Rucaparib) and to a DNA synthesisinhibitor, Gemcitabine, in normoxic and hypoxic conditions.

Figure 2a/2b. SilenciX® Cell survival after 7 days drug treatment. SilenciX®

cells were incubated with Olaparib (A, B), Gemcitabine (C, D), Veliparib(E, F) and Rucaparib (G, H) and survival assays were carried out innormoxia (A, C, E, G) and hypoxia (B, D, F, H).

Table 1. IC50 calculation from Cell survival assay.

Cell survival assays showed that PARP inhibitors are less syntheticlethal in hypoxic conditions with increased IC50 and survival percentageat higher concentrations. These higher survival percentages were inpart a result of a decrease of cell growth in hypoxic condition. However, the difference of IC50 probably involves the signaling pathway of PARP1. PARP1 is linked to the Hypoxia-inducible factor 1 (HIF-1) which is thekey transcription factor regulating hypoxia-dependent gene expres-sion (10,11). Indeed, PARP1 binds directly to HIF-1 and, thus, activatesHIF1-dependent gene expression (10). The low level of oxygen mightdisrupt the synthetic lethality relationship between PARP1 andBRCA1/2 through HIF-1 pathway. The IC50 ratios between Control SilenciX® cells and knocked down cells revealed that olaparib is themost effective inhibitor in our in vitro model. In addition, olaparib,veliparib and rucaparib were more toxic to BRCA1-deficient cells. The DNA synthesis inhibitor, Gemcitabine, had no clear synthetic lethal relationship with BRCA1, BRCA2 and p53 genes.

Cancer cells are characterized by their lack in oxygen delivery thatleads to hypoxia. This has direct effects on the design of treatmentof hypoxic tumors. Indeed tumor vessel normalization has recentlybeen demonstrated with success in efficiently improving drug efficacy (12, 13). The hypoxic microenvironment clearly alters DNA damage repair pathways (14).

The use of DNA repair inhibitors in combination with chemotherapyor radiotherapy can enhance tumor death but also increase toxicitytowards normal cells. Targeting inter-gene relationship of syntheticlethality between DNA repair genes is an effective strategy to over-come this issue. Synthetic lethality is widely exploited to selectivelykill to tumor cells and spare normal cells (15).The best known SL relationship is between BRCA1/2 mutation andPARP1 inhibitors. Others have been reported as BRCA2 / ATM mutationand APE1 inhibitors (16), PTEN mutation and PARP inhibitors (17) orXRCC1 mutation and ATM / DNA-PKcs inhibitors (18).

A simple, robust and stable in vitro model is a powerful tool forscreening in this new SL therapeutic approach. Such a tool may be usedto screen new drug libraries in order to find out new SL relationship. It can also help clarifying the underlying molecular mechanisms of a specific SL relationship. SilenciX® by combining the RNAi technologywith pEBV-derived vectors provides an exceptional opportunity to rapidlycreate a set of stable knock-down clones, able to be cultivated for long-term and with multiple freeze-thaw cycles.

Such an in vitro model is still far from the tumor micro-environment.Indeed, the oxygen partial pressure varies between 11% and 1% O2

in normal human tissues and represents the physioxic condition (19).Working with cell cultures at an oxygen percentage of 20% couldgreatly affect the screening results. Moreover, within the tumor microenvironment, a gas gradient exists between the oxygenatedexterior and anoxic core. More advanced physiologic in vitro modelsinclude multiple cell type 3D microspheroid models (20) which are noteasily adaptable to High Throughput Screening. Our SilenciX® modelused in gas controlled cell culture environment represents a usefultool for screening in early drug discovery.

Discussion References

1. Biard, Nucleic Acids Research, 2007.

2. Satoh, et al., Nature, 1992.

3. Farmer, et al., Nature, 2005.

4. Lord, et al., Curr. Opin. Pharmacol., 2008.

5. Reinbolt and Hays. Frontiers in oncology, 2013.

6. Rajan, et al., Clin Cancer Res, 2012.

7. Strese, et al., BMC Cancer, 2013.

8. Chan and Bristow, Clin Cancer Res, 2010.

9. Chan et al.,Cancer Res, 2010.

10. Elser, et al., Mol Cancer Res, 2008.

11. Martin-Oliva, et al., Cancer Res, 2006.

12. Kiéda, et al., J. Mel. Med., 2013.

13. Collet, et al., Mol Cancer Ther., 2013.

14. Bristow, et al., Nature Reviews Cancer, 2008.

15. Shaheen et al., Blood, 2011.

16. Sultana et al., Int J Cancer, 2012.

17. Mendes-Pereira et al., EMBO Mol Med., 2009.

18. Sultana et al., Cancer Res., 2013.

19. Carreau et al., J Cell Mol Med., 2011.

20. Thoma et al., Advanced Drug Delivery Reviews, 2014.

We would like to address our thanks and acknowledgements to:

- Denis Biard, inventor of the SilenciX® technology,

- Fabienne Fasani, for her contribution to the cell culture experimentsin hypoxia conditions,

- Stephen Durant, for his sponsoring to our participation at the 2014AACR meeting.

Eric Mennesson1*, Anne-Marie Renault1*, Isabelle Fixe1*, Catherine Grillon2, Claudine Kiéda2, Nadia Normand1

* Authors contributed equally to this work - 1 tebu-bio, 39 rue de Houdan, 78612 Le Perray-en-Yvelines, France - 2 Centre de Biophysique Moléculaire, CNRS UPR 4301, Rue Charles Sadron, 45071 Orléans Cedex 2, France

SilenciX® is a registered trademark of tebu-bio, technology licensed from the CEA.

Fig 1-a Fig 2-a

Fig 1-b Fig 2-b

Abstract # 3733

Conclusion

SilenciX® models are ingenious gene-specific knock-down engineered celllines without integration in the host genome and without off-target effects. They are suitable for a broad range of synthetic lethality studies.Cells other than HeLa cells may be used to build relevant cancer diseasemodels. Such pathology models stably silenced for a specific gene cannow be used to mimic tumor microenvironment. The access to SilenciX®

synthetic lethality approach and its use in oxygen controlled conditionssimilar to tumor physioxia open a new landscape for drug screening.

Acknowledgements