Understanding Cisplatin Resistance Using Cellular Models. Britta Stordal 1 and Mary Davey 2

Similar documents
Cancer. Questions about cancer. What is cancer? What causes unregulated cell growth? What regulates cell growth? What causes DNA damage?

RESISTANCE RELATIONSHIPS BETWEEN PLATINUM AND PARP-INHIBITORS IN OVARIAN CANCER.

Overcoming the chemo-resistance in ovarian cancer

Cancer. October is National Breast Cancer Awareness Month

Brian T Burgess, DO, PhD, GYN Oncology Fellow Rachel W. Miller, MD, GYN Oncology

BCHM3972 Human Molecular Cell Biology (Advanced) 2013 Course University of Sydney

Biological activity of bis(carboxylato) cisplatin-based Pt(IV) prodrug candidates: how long the axial ligands should be?

Introduction to Genetics

Part-4. Cell cycle regulatory protein 5 (Cdk5) A novel target of ERK in Carb induced cell death

Targeting DNA base excision repair: a new strategy for personalised cancer therapy

Introduction to Cancer Biology

Multistep nature of cancer development. Cancer genes

Anti-cancer drugs. Introduction : Body : 1) Alkylating Agents

Principles of chemotherapy

Introduction. Cancer Biology. Tumor-suppressor genes. Proto-oncogenes. DNA stability genes. Mechanisms of carcinogenesis.

6.3 DNA Mutations. SBI4U Ms. Ho-Lau

ERCC-1 1 and response to chemotherapy. Jean-Charles SORIA, MD, PhD Institut Gustave Roussy

Chemoresistance and targeted therapies in ovarian and endometrial cancers

NFκB What is it and What s the deal with radicals?

Selenium / selenite in cancer prevention, therapy, and aftercare

Transformation of Normal HMECs (Human Mammary Epithelial Cells) into Metastatic Breast Cancer Cells: Introduction - The Broad Picture:

Radiation Oncology. Initial Certification Qualifying (Computer-based) Examination: Study Guide for Radiation and Cancer Biology

Radiobiology of fractionated treatments: the classical approach and the 4 Rs. Vischioni Barbara MD, PhD Centro Nazionale Adroterapia Oncologica

ATF3 as a Key Regulator of Cisplatin Cytotoxicity: Combining ATF3 Inducing Agents Enhances Cisplatin Activity in NSCLC

Anticancer Drugs. University of Sulaimani Faculty of Medical Sciences School of Pharmacy Pharmacology & Toxicology Dept.

2/21/2016. Cancer Precision Medicine: A Primer. Ovarian Cancer Statistics and Standard of Care in 2015 OUTLINE. Background

Development of Carcinoma Pathways

Hepatitis B Antiviral Drug Development Multi-Marker Screening Assay

Chapter 9. Cells Grow and Reproduce

through the cell cycle. However, how we administer drugs also depends on the combinations that we give and the doses that we give.

Biomarker for Response and Resistance in Ovarian Cancer

5/25/2015. Replication fork. Replication fork. Replication fork. Replication fork

Inhibidores de PARP Una realidad? dónde y cuando?

C-Phycocyanin (C-PC) is a n«sjfc&c- waefc-jduble phycobiliprotein. pigment isolated from Spirulina platensis. This water- soluble protein pigment is

Cellometer Image Cytometry for Cell Cycle Analysis

Early Embryonic Development

Molecular biology :- Cancer genetics lecture 11

Biochemistry 201: DNA repair January 24, 26, 2000 Gilbert Chu

CELL CYCLE MOLECULAR BASIS OF ONCOGENESIS

Investigation of ERCC1 and ERCC2 gene polymorphisms and response to chemotherapy and overall survival in osteosarcoma

Precision Therapeutics For Hard-To-Treat Cancers

IV Nutrient Therapy in Radiation Recovery

CELL CYCLE REGULATION AND CANCER. Cellular Reproduction II

Pt(IV) complexes in the treatment of Pt(II)-refractory tumors: an update. Mauro RAVERA

Targeted therapy & Tumor molecular profile. Anton Tikhonov V Bioinformatics Summer School, 2017

A Summary of the studies conducted with CV247

DNA/RNA: polynucleotide chains

IVC History, Cancer Research

Chapter 12. Regulation of Cell Division. AP Biology

Backgrounder. 1. What are targeted therapies? 2. How do targeted therapies work?

Smoke Like a Man, Survive Like a Woman

Regulation of cell cycle. Dr. SARRAY Sameh, Ph.D

Berberine Sensitizes Human Ovarian Cancer Cells to Cisplatin Through mir-93/ PTEN/Akt Signaling Pathway

Mechanistic Toxicology

Personalised Medicine in Practice. Dr Ingrid Slade MBChB, PhD, MRCPCH, MFPH Dr Chris Spencer DPhil Dr Gabriele De Luca MD, DPhil

Pre-clinical PK/PD modelling of combination therapies in oncology: abemaciclib and vemurafenib

TOUR REPORT. Report on participation of the ICMR International Fellow (ICMR IF) in Training / Research abroad

Inductively coupled plasma mass spectrometry

Fayth K. Yoshimura, Ph.D. September 7, of 7 HIV - BASIC PROPERTIES

Cell cycle and Apoptosis. Chalermchai Mitrpant

B. Incorrect! Compounds are made more polar, to increase their excretion.

Regulation of the Cell Cycle

Reviewing Immunotherapy for Bladder Carcinoma In Situ

Cancer Biology How a cell responds to DNA Damage

DNA codes for RNA, which guides protein synthesis.

The Presence and Persistence of Resistant and Stem Cell- Like Tumor Cells as a Major Problem in Ovarian Cancer

RESISTANT MECHANISMS OF CISPLATIN IN HUMAN LUNG ADENOCARCINOMA CELL LINE As49DDP

p53 and Apoptosis: Master Guardian and Executioner Part 2

Genomic instability. Amin Mahpour

The Need for a PARP in vivo Pharmacodynamic Assay

Regulation of Cell Division (Ch. 12)

Mitosis and the Cell Cycle

Section 9. Junaid Malek, M.D.

Oncolytic virus strategy

Why do patients take herbs and nutritional supplements?

Patrick: An Introduction to Medicinal Chemistry 5e Chapter 09

Necroptosis-Inducing Rhenium(V) Oxo Complexes

Tumour growth environment modulates Chk1 signalling pathways and sensitivity to Chk1 inhibition

Polyomaviridae. Spring

LESSON 3.2 WORKBOOK. How do normal cells become cancer cells? Workbook Lesson 3.2

CHK1 Inhibitor. Prexasertib, LY MsOH H 2 O. Drug Discovery Platform: Cancer Cell Signaling

Regulation of Cell Division. AP Biology

PARP inhibitors for breast cancer

number Done by Corrected by Doctor Maha Shomaf

Cancer Cells. It would take another 20 years and a revolution in the techniques of biological research to answer these questions.

Development of Drug Resistance and Strategies to Circumvent it: A Brief Look

Chemotherapy Treatment Algorithms for Urology Cancer

receive adjuvant chemotherapy

María José Mesa López

Tumor suppressor genes D R. S H O S S E I N I - A S L

2.1 The Importance of Cell Division

Co-exposure of Arsenite and Benzo(a)pyrene: : Effect of glutathione on DNA adduct levels

Section Chapter 14. Go to Section:

1. Basic principles 2. 6 hallmark features 3. Abnormal cell proliferation: mechanisms 4. Carcinogens: examples. Major Principles:

GPS Cancer. The Era of Complete Genomics and Proteomics is Here. Advanced molecular profiling to inform personalized treatment strategies

CONTENTS. Preface... xii

Division Ave. High School AP Biology

MOLECULAR BASIS OF ONCOGENESIS

Management of advanced non small cell lung cancer

Chemotherapy for Urological Cancers

Transcription:

Understanding Cisplatin Resistance Using Cellular Models Britta Stordal 1 and Mary Davey 2 1 Bill Walsh Cancer Research Laboratories, Royal North Shore Hospital and University of Sydney, St Leonards, NSW 2065, Australia. 2 Department of Medical and Molecular Biosciences, University of Technology Sydney, Ultimo, NSW 2007, Australia. Summary Many mechanisms of cisplatin resistance have been proposed from studies of cellular models of resistance including changes in cellular drug accumulation, detoxification of the drug, inhibition of apoptosis and repair of the DNA adducts. A series of resistant models were developed from CCRF-CEM leukaemia cells with increasing doses of cisplatin from 100 ng/ml. This produced increasing resistance up to 7-fold with a treatment dose of 1.6 µg/ml. Cisplatin resistance in these cells correlated with increases in the antioxidant glutathione, yet treatment with buthionine sulphoximine, an inhibitor of glutathione synthesis, had no effect on resistance, suggesting that the increase in glutathione was not directly involved in cisplatin resistance. Two models were developed from H69 SCLC cells, H69-CP and H69CIS200 using 100 ng/ml or 200 ng/ml cisplatin respectively. Both cell models were 2-4 fold resistant to cisplatin, and have decreased expression of p21 which may increase the cell s ability to progress through the cell cycle in the presence of DNA damage. Both the H69-CP and H69CIS200 cells showed no decrease in cellular cisplatin accumulation. However, the H69-CP cells have increased levels of cellular glutathione and are cross resistant to radiation whereas the H69CIS200 cells have neither of these changes. This suggests that increases in glutathione may contribute to cross-resistance to other drugs and radiation, but not directly to cisplatin resistance. There are multiple resistance mechanisms induced by cisplatin treatment, even in the same cell type. How then should cisplatin-resistant cancers be treated? Cisplatinresistant cell lines are often more sensitive to another chemotherapeutic drug paclitaxel (H69CIS200), or are able to be sensitised to cisplatin with paclitaxel pre-treatment (H69-

CP). The understanding of this sensitisation by paclitaxel using cell models of cisplatin resistance will lead to improvements in the clinical treatment of cisplatin resistant tumours. Keywords Cisplatin, Platinum, Resistance, Chemotherapy, Glutathione, p21, Paclitaxel, SCLC, Leukaemia, Cell Models Introduction Cisplatin has been used in the treatment of cancer for over 30 years, and is highly successful for many cancers, including testicular, ovarian and lung cancer. Upon entering the cell, cisplatin becomes positively charged, and so is able to interact with nucleophilic molecules including DNA, RNA and proteins. Cytotoxicity is believed mainly due to interaction with DNA, forming inter- and intra-strand adducts, hindering both RNA transcription and DNA replication, leading to cell cycle arrest and apoptosis. Inevitably, the use of cisplatin is limited by the development of drug resistance. Numerous cellular mechanisms potentially contributing to clinical cisplatin resistance have been proposed (1,2) including changes in cellular drug accumulation, detoxification of the drug, inhibition of apoptosis and repair of the DNA adducts, as summarised in Fig. 1. Understanding these mechanisms and their role in resistance is important for the continued success of cancer treatment. Cellular Models of Cisplatin Resistance We have developed several cellular models to attempt to understand the cellular adaptations underlying cisplatin resistance mechanisms, and potential strategies to reverse this resistance. Small cell lung cancer (SCLC) is an aggressive form of lung disease, with treatment involving combination chemotherapy including cisplatin. While this produces 90% response in patients, relapse is rapid with patients developing resistant disease. We have treated H69 SCLC cells with 100 ng/ml cisplatin, to produce the H69- CP (3) or 200 ng/ml cisplatin to obtain the H69CIS200 cells (4). These doses are below an IC 50 for cisplatin and are within the range achieved in the clinical use of cisplatin. The

cells were 2- to 4-fold resistant to cisplatin, but there was no decreased drug accumulation. To further identify molecular changes resulting from low, non-toxic doses of cisplatin, the model CCRF-CEM leukaemia cell was treated for 3-4 days with increasing doses of cisplatin from 100 ng/ml, a dose well below the IC 50 for cisplatin (540 ± 30 ng/ml) for these cells. This produced a series of cells with increasing cisplatin resistance up to 7-fold resistance with a treatment dose of 1.6 µg/ml, after which resistance, as determined in a 4-day cytotoxicity assay, decreased (Figure 2). Resistance was associated with decreased cisplatin accumulation, although, there were no changes in expression of the multidrug transport protein MRP2 which transports cisplatin conjugated to glutathione to explain the decreased intracellular drug as increased drug efflux (5). Detoxification mechanisms in cisplatin resistance Cisplatin is very reactive towards the cellular antioxidant glutathione, readily forming complexes. Resistance in the CEM cells reflected changes in glutathione (Figure 2). However, treatment of these cells with buthionine sulphoximine (BSO), an inhibitor of glutathione synthesis, had no effect on cellular resistance. This suggests that although a cellular response to cisplatin treatment was to increase their glutathione levels, this was not directly involved in cisplatin resistance. Glutathione changes have frequently been reported in cells treated with cisplatin, and may contribute to cross-resistance to other drugs and radiation, but not necessarily directly to cisplatin resistance. This proposal is supported by the SCLC cells which, although 2 to 4-fold resistant to cisplatin, the H69- CP cells had increased glutathione and cross-resistance to radiation while the H69CIS200 cells had no change in glutathione and were not radiation resistant. This is also supported by the fact that radiation resistant H69 cells with increased glutathione are highly resistant to cisplatin (6). However, glutathione is not the only thiol cellular redox system, and changes in the thioredoxin antioxidant system, thioredoxin reductase and thioredoxin, are also reported to confer cisplatin resistance (7). Increased thioredoxin reductase occurred in the cisplatin-resistant CEM cells, leading to cross- resistance to the thioredoxin reductase inhibitor auranofin, a gold compound clinically used as an antirheumatic drug. This

contrasts a recent report suggesting auranofin induces apoptosis in cisplatin resistant ovarian cancer cells, and so may be suitable to treat cisplatin resistant tumours (8). Again, the involvement of redox systems in cisplatin resistance is variable and may be dependent on cell type. Cisplatin resistance and the cell cycle In the CEM series of cisplatin-resistant cells, at higher levels of drug treatment the cells do not appear to be resistant as judged in a 4-day cytotoxicity assay. This is because cisplatin treatment causes the cells to stop growing. On removal of the drug, the cells then proliferate rapidly. While this resistance mechanism occurred at higher drug doses in the CEM cells, a similar response to cisplatin was evident after treatment with low levels of drug in the H69CIS200 cells (4), where cells rapidly grew on removal of drug. The contrast in resistant mechanisms developed in the H69CIS200 and H69-CP cells illustrates the diversity of mechanisms which may occur using similar treatment strategies even in the same cell line. As well as alterations in the cell cycle allowing rapid proliferation post drug treatment, the H69CIS200 cells also have several chromosomal rearrangements which are not associated with the resistant phenotype, suggesting an increase in genomic instability in the resistant cell lines (9). We hypothesise that there is a deregulation between the cell cycle and DNA repair in the H69CIS200 cells allowing proliferation in the presence of DNA damage which has created an increase in genomic instability. The cellular response to DNA damage as a result of cisplatin treatment would be induction of p53, causing cells to arrest, by regulating the expression of cyclins and cyclin-dependent kinases. Cisplatin however does not induce the cyclin-dependent kinase-inhibitor p21 in 2780CP cisplatin resistant cells, supporting the disruption of the normal response pathway in resistant cells (10). Both the H69CIS200 cells and the H69-CP cells have decreased p21 expression, which may increase the cell s ability to progress through the cell cycle despite the presence of DNA damage. This not only provides a resistance mechanism, but will contribute to the genomic instability of the cells which in turn will increase the mutagenic potential of the cells in response to further drug treatment.

DNA Repair Mechanisms Since the major effect of cisplatin is the formation of DNA adducts, increased DNA repair is a potential resistance mechanism. Nucleotide excision repair (NER) mainly repairs bulky DNA adducts such as those caused by interaction with cisplatin, and downregulation of ERCC1, a core protein required for NER, sensitised cells to cisplatin (11). We have found that the cisplatin-resistant H69CIS200 cells have no alteration in DNA repair capacity compared to the parental H69 cells. However, ERCC1 expression decreases in association with the cisplatin-induced cell cycle arrest in both sensitive and resistant cells rather than in association with any change in DNA repair. Both increased ERCC1 expression (12) and decreased ERCC1 expression (13,14) have been associated with sensitivity to cisplatin based combination chemotherapy. Cisplatin treatment may alter the expression of ERCC1 for reasons other than DNA repair. This may explain some of the contradictory results examining this gene as a marker for the clinical response to cisplatin therapy. The ability to differentiate between these different types of platinum resistance in the clinic will improve the choice of salvage chemotherapy in patients with cisplatin-resistant cancers. Conclusions It is apparent that there are multiple resistance mechanisms induced by cisplatin treatment, and as many of these are linked by the cellular stress response, it is difficult to determine which of these is more important in resistance. While many mechanisms have been identified, there is no consistent response, even in the same cell type to treatment with cisplatin. The question then is: how to treat cisplatin-resistant tumours. The cell models are useful not only for examining the potential of the new platinum drugs being developed, but also for looking for combinations of current drugs which may lead to improvements in response. A recent report demonstrated that combination of the cell cycle specific antagonist gemcitabine with cisplatin was more effective than either drug alone. This combination gave enhanced toxicity in cisplatin resistant cells, suggesting that gemcitabine reversed cisplatin resistance (15,16).

Of particular interest are the frequent reports of sensitivity to paclitaxel in cisplatin resistant cells. This was evident in the H69CIS200 cells which were 5-fold more sensitive to paclitaxel than the H69 cells. The other cisplatin resistant cells, although cross-resistant to many drugs, were not resistant to paclitaxel. However, treatment of these cisplatin resistant cells, but not the H69 cells, with non-cytotoxic doses of paclitaxel was able to sensitise the resistant cells not only to cisplatin, but to other drugs, and also to radiation (17,18,3) Paclitaxel sensitization occurred after at least a 12 hour pre-treatment of the cells, suggesting time is required for this response. Analysis of the protein profile of these cells showed that paclitaxel treatment reversed many of the cellular protein changes that accompanied the development of resistance (19). This activity of paclitaxel was independent of the cell cycle mediated effect of the drug, which suggests other signaling pathways are involved (18). Understanding this sensitization of cisplatin resistant cells would lead to improved treatment protocols for the treatment of all forms of cisplatin resistance, and suggests that while cisplatin resistance is multifactorial, the means to overcome resistance may lie in inhibition of one specific signaling pathway. Future studies using cell models of cisplatin resistance will lead to an understanding of ways to overcome cisplatin resistance and improve the treatment of cisplatin resistant tumours. References 1. Rabik,C.A. and Dolan,M.E. (2007) Molecular mechanisms of resistance and toxicity associated with platinating agents Cancer Treatment Reviews 33, 9-23. 2. Kartalou,M. and Essigmann,J.M. (2001) Mechanisms of resistance to cisplatin. Mutation Research 478, 23-43. 3. Locke,V.L., Davey,R.A., and Davey,M.W. (2003) Modulation of drug and radiation resistance in small cell lung cancer cells by paclitaxel Anti-Cancer Drugs 14, 523-531. 4. Stordal,B.K., Davey,M.W., and Davey,R.A. (2006) Oxaliplatin induces drug resistance more rapidly than cisplatin in H69 small cell lung cancer cells Cancer Chemotherapy & Pharmacology 58, 256-265. 5. Borst,P., Evers,R., Kool,M., and Wijnholds,J. (2000) A family of drug transporters: the multidrug resistance-associated proteins. Journal of the National Cancer Institute. 92, 1295-1302.

6. Henness,S., Davey,M.W., Harvie,R.M., and Davey,R.A. (2002) Fractionated irradiation of H69 small-cell lung cancer cells causes stable radiation and drug resistance with increased MRP1, MRP2, and topoisomerase IIalpha expression International Journal of Radiation Oncology, Biology, Physics. 54, 895-902. 7. Sasada,T., Nakamura,H., Ueda,S., Sato,N., Kitaoka,Y., Gon,Y., Takabayashi,A., Spyrou,G., Holmgren,A., and Yodoi,J. (1999) Possible involvement of thioredoxin reductase as well as thioredoxin in cellular sensitivity to cis-diamminedichloroplatinum (II) Free Radical Biology & Medicine 27, 504-514. 8. Marzano,C., Gandin,V., Folda,A., Scutari,G., Bindoli,A., and Rigobello,M.P. (2007) Inhibition of thioredoxin reductase by auranofin induces apoptosis in cisplatinresistant human ovarian cancer cells Free Radical Biology & Medicine 42, 872-881. 9. Stordal,B., Peters,G., and Davey,R. (2006) Similar chromosomal changes in cisplatin and oxaliplatin-resistant sublines of the H69 SCLC cell line are not associated with platinum resistance Genes, Chromosomes & Cancer 45, 1094-1105. 10. Siddik,Z.H., Mims,B., Lozano,G., and Thai,G. (1998) Independent pathways of p53 induction by cisplatin and X-rays in a cisplatin-resistant ovarian tumor cell line Cancer Research 58, 698-703. 11. Cummings,M., Higginbottom,K., McGurk,C.J., Wong,O.G., Koberle,B., Oliver,R.T., and Masters,J.R. (2006) XPA versus ERCC1 as chemosensitising agents to cisplatin and mitomycin C in prostate cancer cells: role of ERCC1 in homologous recombination repair Biochemical Pharmacology 72, 166-175. 12. Simon,G.R., Sharma,S., Cantor,A., Smith,P., and Bepler,G. (2005) ERCC1 expression is a predictor of survival in resected patients with non-small cell lung cancer Chest 127, 978-983. 13. Dabholkar,M., Vionnet,J., Bostick-Bruton,F., Yu,J.J., and Reed,E. (1994) Messenger RNA levels of XPAC and ERCC1 in ovarian cancer tissue correlate with response to platinum-based chemotherapy Journal of Clinical Investigation. 94, 703-708. 14. Lord,R.V., Brabender,J., Gandara,D., Alberola,V., Camps,C., Domine,M., Cardenal,F., Sanchez,J.M., Gumerlock,P.H., Taron,M., Sanchez,J.J., Danenberg,K.D., Danenberg,P.V., and Rosell,R. (2002) Low ERCC1 expression correlates with prolonged survival after cisplatin plus gemcitabine chemotherapy in non-small cell lung cancer Clinical Cancer Research 8, 2286-2291. 15. Smith,J.A., Gaikwad,A., Ramondetta,L.M., Wolf,J.K., and Brown,J. (2006) Determination of the mechanism of gemcitabine modulation of cisplatin drug resistance in panel of human endometrial cancer cell lines Gynecologic Oncology 103, 518-522. 16. Smith,J.A., Brown,J., Martin,M.C., Ramondetta,L.M., and Wolf,J.K. (2004) An in vitro study of the inhibitory activity of gemcitabine and platinum agents in human endometrial carcinoma cell lines Gynecologic Oncology 92, 314-319.

17. Su,G.M., Davey,M.W., and Davey,R.A. (1998) Induction of broad drug resistance in small cell lung cancer cells and its reversal by paclitaxel International Journal of Cancer 76, 702-708. 18. Locke,V., Davey,R., and Davey,M. (2001) Paclitaxel sensitization of multidrugresistant cells to chemotherapy is independent of the cell cycle Cytometry 43, 170-174. 19. Davey,R.A., Locke,V.L., Henness,S., Harvie,R.M., and Davey,M.W. (2004) Cellular models of drug- and radiation-resistant small cell lung cancer Anticancer Research 24, 465-471.

CYTOPLASM NUCLEUS Cl Cl Pt Cisplatin Decreased Uptake Increased Efflux + H 2 O Cl Detoxification by Glutathione GS Cl Pt Pt Nuclear Membrane DNA Pol. Platinum Adducts Causing Cytotoxicity Anti-Apoptotic Cell Membrane Pro-Apoptotic Inhibition of Apoptosis DNA Pol. Increased Bypass of Platinum Adducts Increased DNA Repair Removal of Platinum Adducts Figure 1. Cisplatin resistance mechanisms. Cisplatin is a neutral complex which on entering the cell becomes positively charged, and so able to interact with many molecules including DNA and proteins. Many mechanisms may contribute to cisplatin resistance including reduced uptake, increased efflux, increased detoxification, inhibition of apoptosis, increased ability to replicate DNA adducts and increased DNA repair. GS Glutathione, Pol Polymerase.

CEM 100 200 400 800 1600 3200 5000 ng/ml Cisplatin Fold Resistance 7.5 5.0 2.5 0 Resistance CP100 CP200 CP400 CP800 CP1600 CP3200 CP5000 Glutathione (% CEM) 200 100 Glutathione 0 CP100 CP200 CP400 CP800 CP1600 CP3200 CP5000 Figure 2. Cisplatin Resistance in CEM cells. CEM cells were treated for 3-4 days with cisplatin, commencing with 100 ng/ml. After 6 treatments, cells were stable to drug treatment and the doses increased. This developed a series of cisplatin-resistant cells. Cisplatin resistance (fold increase relative to the untreated CEM cells) is reflected in cellular glutathione levels.