<<

METABOLISM OF IN DOGS AND COMPARATIVE CYTOTOXICITY OF LOMUSTINE AND ITS MAJOR METABOLITES AND O6-METHYLGUANINE-DNA METHYLTRANSFERASE EXPRESSION IN CANINE CELLS

BY

THUSHARA CHAKKATH

DISSERTATION

Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in VMS - Comparative Biosciences in the Graduate College of the University of Illinois at Urbana-Champaign, 2014

Urbana, Illinois

Doctoral Committee:

Associate Professor Levent Dirikolu, Chair Professor Duncan C. Ferguson Professor Margarethe Hoenig Associate Professor David Bunick Associate Professor Timothy M. Fan

ABSTRACT

The drug lomustine is used clinically for treating a wide variety of malignancies, most commonly brain tumors and lymphoma. Lomustine undergoes hydrolysis in vivo to form isomeric metabolites, primarily trans-4-hydroxylomustine (trans-4) and cis-4- hydroxylomustine (cis-4) in various animal species including humans. Despite its widespread usage to treat canine lymphoma, the metabolism of lomustine has not been studied in dogs. It is reported that 4’-hydroxylation products of lomustine (trans-4 and cis-4) have enhanced alkylating activity and reduced toxic effects relative to lomustine, resulting in a better therapeutic index of each of the metabolites relative to the parent compound. Our results show that the metabolic profile of lomustine in dogs is similar to that in humans with trans-4 being the major metabolite and cis-

4 as the minor metabolite. Comparative cytotoxicity studies of lomustine and its trans-4 and cis-4 metabolites in canine lymphoma cell lines 17-71 and GL-1 show that there is no difference in the cytotoxicity of the three compounds. In addition, a concentration and time-dependent cell killing was seen in both of these cell lines. Also primary canine cells like peripheral blood mononuclear cells (PBMC) from lymphoma dogs and canine hepatocytes for normal dogs did not show any sensitivity towards lomustine and its metabolites.

Alkylation is thought to be the reason for the efficacy of lomustine while carbamylation has been implicated as the cause for the side effects seen with lomustine treatment such as hepatotoxicity. In the alkylation study we show that lomustine and its metabolites form similar levels of the DNA adducts N7 hydroxyethylguanine and O6 hydroxyethyldeoxyguanosine. These adducts may be used as molecular dosimeters of therapeutic response thus correlating to the similar cytotoxicity of these compounds. In terms of carbamylation, lomustine showed greater extent of

ii carbamylation in the canine hepatocytes and lymphoma cell lines. We hypothesize that if carbamylation of proteins is indeed one of the reasons for the side effects of lomustine, then using the metabolites instead may be a better option in terms of side effects.

The DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT) causes resistance of tumor cells to bifunctional , like lomustine. There is no data available regarding MGMT expression/activity in canine cells or tissues. Our study shows that there is low

MGMT activity in the canine lymphoma cell line 17-71 while the GL-1 cells did not show any detectable enzyme activity or mRNA expression. The MGMT enzyme activity measured in canine hepatocytes is about 250-350 fmol/mg protein as compared to about 90 fmol/mg protein in 17-71 cells. We also show that MGMT mRNA expression in 17-71 cells and canine hepatocytes positively correlates with its enzyme activity in these cells.

iii

ACKNOWLEDGEMENTS

I would like to express my gratitude to my adviser, Dr. Levent Dirikolu, for his continual guidance, support and encouragement throughout the course of my doctoral research. I would also like to thank my advisory committee members, Dr. Duncan Ferguson, Dr. Margarethe Hoenig, Dr.

Timothy Fan and Dr. David Bunick for their guidance and valuable comments.

I am grateful to the following people for helping me with my research work; Dr. Sidonie

Lavergne for helping design the cytotoxicity studies, Dr. Lucas Li for LC/MS/MS analysis of DNA samples and Dr. Matthew Allender for help with statistical analysis. I would like to acknowledge the students and staff of Bagchi, Das and Flaws labs in Comparative biosciences and Fan lab in the Small Animal Clinic for their courtesy and cooperation. I am extremely grateful to the staff and fellow students in Comparative Biosciences for their assistance and their friendship.

I would also like to thank my family in India and all my friends for their support and encouragement. Finally a big thank you to my husband, Dr. Manas Gartia, for all the love and support and for making life more enjoyable.

iv

TABLE OF CONTENTS

List of abbreviations and symbols ...... vii

Chapter I Introduction ...... 1 1.1 Canine Lymphoma ...... 1 1.2 Side effects of lomustine ...... 5 1.3 Mechanism of action of lomustine and role of O6-methylguanine-DNA methyltransferase .....6 1.4 Figures and Legends ...... 12 1.5 References ...... 14

Chapter II Development of validated analytical method for detection of lomustine and its metabolites and metabolism characteristics of lomustine in dogs…………………………...19 2.1 Introduction ...... 19 2.2 Materials and Methods ...... 21 2.3 Results ...... 26 2.4 Discussion ...... 30 2.5 Figures and Legends ...... 34 2.6 References ...... 44

Chapter III Cytotoxicity of lomustine and its trans-4-hydroxylomustine and cis-4- hydroxylomustine metabolites in canine lymphoma cell lines and primary cells…………..46 3.1 Introduction ...... 46 3.2 Materials and Methods ...... 47 3.3 Results ...... 53 3.4 Discussion ...... 55 3.5 Figures and Legends ...... 58 3.6 References ...... 69

Chapter IV Alkylating and carbamylating effects of lomustine, trans-4-hydroxylomustine and cis-4-hydroxylomustine in canine lymphoma cell lines and hepatocytes………...…….71 4.1 Introduction ...... 71 4.2 Materials and Methods ...... 73 4.3 Results ...... 79 4.4 Discussion ...... 79 4.5 Figures and Legends ...... 83 4.6 References ...... 87

Chapter V MGMT mRNA expression and enzyme activity in canine lymphoma cell lines and hepatocytes...... 89 5.1 Introduction ...... 89 5.2 Materials and Methods ...... 91 5.3 Results ...... 96 5.4 Discussion ...... 98 5.5 Figures and Legends ...... 102 5.6 References ...... 112

v

Chapter VI Conclusion ...... 115 6.1 Conclusion ...... 115 6.2 References ...... 121

vi

LIST OF ABBREVIATIONS AND SYMBOLS

ALT Alanine transaminase ANOVA Analysis of variance BCA Bicinchoninic acid bp Base pair CBL Carbamylated lysine CBL-BSA Carbamylated lysine-bovine serum albumin Cblood Concentration of drug in blood cDNA Complementary DNA CHOP , Hydroxydaunorubicin, Oncovin, Prednisone Cplasma Concentration of drug in plasma CRBC Concentration of drug in RBC CCNU 1-(2-Chloroethyl)-3-cyclohexyl-1-nitrosourea Cis-4 Cis-4-hydroxylomustine CV Coefficient of variation DMEM Dulbecco's modified Eagle medium DNA Deoxyribonucleic acid DTT Dithiothreitol D3-O6 methylguanine Deuterated O6 methylguanine ED50 Effective dose for 50 percent of the group EDTA Ethylenediaminetetraacetic acid ELISA Enzyme-linked immunosorbent assay ESI Electrospray ionization FITC Fluorescein isothiocyanate FBS Fetal bovine serum g Centrifugal acceleration force GI tract Gastrointestinal tract HEPES 4-(2-Hydroxyethyl)piperazine-1-ethanesulfonic acid HBSS Hank's Balanced Salt Solution HEX Hexachloro-fluorescein HPRT-1 Hypoxanthine phosphoribosyltransferase 1 IC50 Half maximal inhibitory concentration Ke/p Erythrocyte plasma partitioning coefficient L15 medium Liebovitz L 15 medium LC Liquid chromatography LC/MS/MS Liquid chromatography-tandem mass spectrometry LD10 Lethal dose for 10 percent of the group LOD Limit of detection LOQ Limit of quantitation mAU Arbitrary unit for intensity MGMT O6-methylguanine-DNA methyltransferase min Minutes MNNG Methylnitronitrosoguanidine MNU N-methyl-N-nitrosourea

vii mRNA messenger RNA MRM Multiple reaction monitoring m/z Mass-to-charge ratio Oligo Oligonucleaotide PAC-1 First procaspase-3 activating compound PBMC Peripheral blood mononuclear cells PBS Phosphate buffered saline PI Propidium iodide PTFE Polytetrafluoroethylene QPCR Quantitative polymerase chain reaction r2 Coefficient of determination RBC Red blood cells RNA Ribonucleic acid RPMI-1640 (Roswell Park Memorial Institute)1640 medium RT-PCR Real time polymerase chain reaction SD Standard deviation TBE Tris/Borate/EDTA TMCS Chlorotrimethylsilane Trans-4 Trans-4-hydroxylomustine tRNA transfer RNA V Volts VC Vehicle control WHO World Health Organization XTT 2,3-Bis-(2-methoxy-4-5-sulfophenyl)-2H-terazolium-5- carboxanilide

viii

CHAPTER I

Introduction

1.1 Canine lymphoma

Lymphoma is one of the most common hematopoietic neoplasias seen in dogs, accounting for up to 20% of all cancers diagnosed in canines (Vail et al., 2001). The disease more commonly occurs in middle age dogs but younger dogs are also known to be affected by it. Four anatomic forms of canine lymphoma have been described: multicentric, alimentary, mediastinal and extranodal (renal, central nervous system, cutaneous, ocular, bone, etc.) with multicentric being the most common anatomic form accounting for over 80% of all diagnosed cases (Madewell &

Theilen, 1987). A common manifestation of multicentric lymphoma is the development of nonpainful generalized lymphadenopathy with peripheral lymph nodes often bilaterally and symmetrically enlarged (Meuten et al., 2008). Lymphoma is also classified based on the clonality of the lymphocytes involved, i.e. T-cell or B-cell. Diffuse large B-cell lymphoma accounts for majority of the lymphoma cases (Richards et al., 2013).

Diagnosis of multicentric lymphoma involves physical examination for palpation of lymph nodes and obtaining tissue or cytological specimens for definitive diagnosis. For deep seated cancers, X-ray, ultrasound, bone marrow biopsy may be needed. The WHO (Swerdlow et al.,

2008) has classified different stages of lymphoma based on the degree of metastasis and invasiveness as:

1

Stage I: Ailment restricted to single lymph node

Stage II: Regional lymphadenopathy

Stage III: Generalized lymphadenopathy

Stage IV: Enlargement of the liver and spleen (with or without lymphadenopathy)

Stage V: Bone marrow, central nervous system or involvement of other extranodal sites.

The treatment approach for canine lymphoma is determined by the stage of the disease.

The current standard of care for dogs with lymphoma involves the use of multiagent , which achieves response rates of 65% to 96% and first-remission durations of 6 to 9 months

(Morrison, 2004). There are various multiagent chemotherapy protocols such as different variations of ‘CHOP’ (cyclophosphamide, , , prednisolone). Despite favorable treatment outcomes initially, the majority of dogs will experience relapse, necessitating the reinstitution of additional for the management of refractory disease. Second remission is harder to achieve and may be of shorter duration (Moore et al., 1999). Some relapsed dogs become refractory to the initial treatment and hence may require a different chemotherapeutic agent/rescue therapy to achieve second remission. For rescue therapy, again a multiagent protocol is followed; however, a single agent therapy may be preferred for clients with financial or logistical restrictions. Also cross-resistance between chemotherapeutic agents can decrease the efficacy of rescue protocols. Alkylating agents seldom show cross resistance and are quite effective in achieving clinical remission (Tew et al., 2006). Alkylating agents include chemotherapeutic drug classes such as nitrosoureas, nitrogen mustards and alkyl sulfonates and function by attaching an alkyl group to the DNA.

2

The nitrosourea drug, lomustine (CCNU, CeeNU, C9H16ClN303, 1-(2-chloroethyl)-3- cyclohexyl-1-nitrosourea), is used clinically for treating a wide variety of human malignancies, most commonly brain tumors and lymphoma (Mitchell & Schein, 1986; Tew et al., 1996). In veterinary medicine, lomustine has been successfully used primarily for the treatment of resistant lymphoma (Moore et al., 1999; Saba et al., 2007; Sauerbrey et al., 2007), but also for the treatment of mast cell tumors (Rassnick et al., 1999), intracranial meningioma (Jung et al., 2006), epitheliotropic lymphoma (Williams et al., 2006) and histiocytic sarcoma (Skorupski et al., 2007) in dogs either alone or in combination with other chemotherapeutic agents. There are several studies in the literature indicating good efficacy of lomustine in combination with other chemotherapeutic agents such as L-, doxorubicin, temozolamide, etc. for the treatment of resistant canine lymphoma (Moore et al., 1999; Saba et al., 2007; Sauerbrey et al.,

2007), however, its potential as a single agent for first line therapy or to treat relapsed canine lymphoma is less promising. In a study that evaluated the efficacy of lomustine and prednisolone as first line treatment of muticentric lymphoma, 9 out of 17 dogs showed complete or partial response with a median duration of remission of 39.5 days (range 20-130 days) (Sauerbrey et al.,

2007). In another study of lomustine as single agent chemotherapy for relapsed canine lymphoma in 11 dogs, 28% showed complete or partial remission with median response duration of 86 days

(range 36-212 days) (Moore et al., 1999). However, lomustine as a single agent may be preferred when treatment costs prohibit exploring other options or when a multidrug treatment is not tolerated by the patient due to extreme side effects.

In a clinical setting, lomustine is usually administered to dogs orally at 50-110 mg/m2 every three weeks (Kristal et al., 2004; Moore et al., 1999; Sauerbrey et al., 2007). Lomustine is a highly

3 lipophilic nitrosourea compound, which undergoes hydrolysis in vivo to form isomeric metabolites, primarily trans-4-hydroxylomustine (trans-4) and cis-4-hydroxylomustine (cis-4)

(Figure 1.1) in various animal species including humans (Hilton & Walker, 1975; Kastrissios et al., 1996; Lee et al., 1985). The comparative biological effects of lomustine and its metabolites were studied in mice. The single dose LD10 (dose causing death of 10% of the mice when administered intraperitonially) and single dose ED50 (dose producing 45 day survival in 50% of the mice after intraperitoneal implantation of 105 L1210 cells) were compared to determine the therapeutic index for each compound (Wheeler et al., 1977). The metabolites showed an approximately two-fold greater therapeutic index compared to the parent drug. It was therefore suggested that hydroxylation at the 4’-position of the cyclohexyl ring of lomustine, confers both isomers (cis-4 and trans-4) of the metabolite with enhanced alkylating activity and reduced toxic effects relative to lomustine, resulting in better therapeutic index of each of the metabolites relative to the parent compound (Wheeler et al., 1977).

Hence it is plausible that the metabolites of lomustine could be better options for chemotherapy, with greater efficacy and favorable side effect profile than lomustine itself. In addition to the above metabolites, minor metabolites are also formed due to hydroxylation at the second and third position of the cyclohexyl ring (Hilton & Walker, 1975; Reed, 1984; Wheeler et al., 1977). However, there is no data available regarding the metabolism pattern of lomustine in dogs.

4

1.2 Side effects of lomustine

One of the major toxic effects of lomustine in humans is delayed myelosuppression

(Hoogstraten et al., 1973; Moertel et al., 1972). The hematological toxicity associated with lomustine is known to be dose-dependent with greater toxicity seen at higher doses. Another toxicity associated with lomustine treatment is hepatotoxicity, but unlike myelosuppression, these reactions are believed to be not dose-dependent or predictable. Toxicity trials in healthy beagle dogs and clinical trials in tumor bearing dogs have shown lomustine to induce hepatotoxicity and myelotoxicity at doses that are clinically used (50-110 mg/m2 every three weeks) (Fulton &

Steinberg, 1990; Kristal et al., 2004; Moore et al., 1999; Rassnick et al., 1999).

The reported incidence of hepatotoxicity in dogs due to lomustine treatment is variable.

Retrospective studies have reported elevation in liver enzymes in up to 86% of dogs receiving lomustine for cancer treatment (Kristal et al., 2004; Risbon et al., 2006; Williams et al., 2006) but only 3-6% of them showed clinical signs of serious liver injury. Another retrospective study concluded that elevation of alanine aminotransferase (ALT) is common during lomustine chemotherapy in dogs and that severe elevation can develop on a sudden onset not preceded by a mild ALT elevation (Hosoya et al., 2009). A prospective study of lomustine induced hepatotoxicity in tumor bearing dogs reported that out of 28 dogs that received lomustine as single agent chemotherapy, 7 dogs (25%) required discontinuation of lomustine due to severe elevation of ALT

(Skorupski et al., 2007). In some rare cases fatalities due to terminal liver damage after lomustine treatment have also been reported (Williams et al., 2006). Based on the available data on this topic, it may be concluded that elevated ALT is a common occurrence following lomustine treatment,

5 however some dogs may show severe liver enzyme elevation and also clinical signs of liver injury necessitating discontinuation of lomustine. The underlying mechanism for hepatic injury due to lomustine is unclear.

It is believed that chemical breakdown of lomustine, in addition to the alkylating intermediates, also produces isocyanates that carbamylate proteins and this may contribute to the side effects of lomustine (Babson & Reed, 1978; Lemoine et al., 1991; Reed, 1981; Sariban et al.,

1984). Carbamylation is a non-enzymatic post-translational modification of proteins that occurs due to binding of isocyanic acid to proteins and is usually associated with partial or complete loss of protein function (Lemoine et al., 1991). Although the highly reactive nature of the isocyanate species precludes their identification, lomustine derived isocyanates could participate in the chemotherapeutic and cytotoxic responses by inactivating enzymes some of which could be involved in vital functions of the cell such as replication and DNA repair. (Ducastelle et al., 1988;

Fornace et al., 1978; Kann et al., 1974). It is thought that the metabolites of lomustine may be safer in terms of their side effects as compared to the parent drug, lomustine (Wheeler et al., 1977; Heal et al., 1978). The comparative carbamylating effects of lomustine and its major metabolites, have not been investigated in canine cells.

1.3 Mechanism of action of lomustine and role of O6-methylguanine-DNA methyltransferase

Like other nitrosoureas, lomustine and its metabolites lead to the covalent attachment of alkyl groups on the purine and pyrimidine bases of DNA. Although multiple adducts may be formed due to exposure to alkylating agents, not all adducts have the potential to cause permanent

6

DNA alteration. Of particular interest is the methyl adduct at the O6 position of guanine (Figure

1.2) which causes mispairing and is associated with mutagenicity (Allay et al., 1999; Kadlubar et al., 1984; Swenberg et al., 1985). Adducts at the O6 position formed by chloroethylating compounds like lomustine cause inter-strand crosslinking in the DNA (Kohn, 1977). During DNA replication this crosslinking can ultimately lead to double strand breaks which is a potent inducer of apoptosis leading to cell death (Kaina et al., 2007; Ochs & Kaina, 2000). In addition to O6 guanine, adducts at the N7 position of guanine (Figure 1.2) are formed in high concentration after nitrosourea exposure. In fact, N7 guanine adducts makes up 70-90% of the total adducts formed by alkylating agents (Rinne et al., 2005). However, they have not been implicated as a causal factor for mutagenesis like the O6 alkylguanine (Lawley, 1976; Pegg, 1977; Singer, 1975; Tong et al.,

1982). But due to its abundant formation following nitrosourea treatment, measuring the

N7alkylguanine especially after exposure to low concentrations of lomustine and its metabolites

(when other adducts may be below detection limits) can be used as a molecular dosimeter for therapeutic response or genotoxic drug exposure to cells.

The direct repair of adducts at O6 guanine position is via the enzyme O6-methylguanine-

DNA methyltransferase (MGMT). MGMT is a DNA protective enzyme that is known to be present in the cytoplasm and nucleus of mammalian cells. The cytoplasmic MGMT is thought to be translocated to the nucleus rapidly in the event of DNA alkylation (Kaina et al., 2007). MGMT constitutes an evolutionary highly conserved DNA repair mechanism for the correction of O6- guanine alkyl adducts. While most other DNA repair pathways involve the combined action of multiple proteins, repair by MGMT is a direct reversal of damage, in which the offending alkyl group is transferred to a cysteine residue within the acceptor pocket of the MGMT protein

7

(Margison et al., 2003). This reaction is stoichiometric in nature i.e. one MGMT molecule will bind to a single adduct and will be directed to ubiquitination and cannot be reused, thus appropriately being called a ‘suicide enzyme’ (Kaina et al., 2007). Hence the cell’s capacity to use

MGMT for repair of alkylation depends on the number of MGMT molecules already present and its ability for de novo synthesis. Due to the stoichiometry and autoinactivation mechanism, cells overwhelmed with alkylation can be depleted of MGMT and can be more susceptible to O6 alkylating agents (Kaina et al., 2007) such as lomustine.

This DNA repair enzyme (MGMT) plays a role in maintaining the integrity of the DNA in normal cells but also protects tumor cells against alkylating chemotherapeutic agents, resulting in drug resistance (Christmann et al., 2011). MGMT is the major mechanism of resistance of tumor cells to bifunctional nitrosoureas, and its depletion with suicide inhibitors, such as O6 benzylguanine, has been shown to repeatedly sensitize tumor cells to alkylating agents (Gerson,

2002). Previous studies have shown that mice deficient in MGMT gene are more susceptible to the toxic effects of alkylating anti-cancer agents (Sakumi et al., 1997), while in transgenic models, mice expressing increased levels of MGMT are more resistant to alkylating carcinogens (Dumenco et al., 1993; Nakatsuru et al., 1993). Although MGMT is ubiquitously expressed in all cells of the body, there is significant variation in its level in different tissues, the same tissues between individuals and also between neoplastic and non-neoplastic tissues (Christmann et al., 2011). Data from humans show that the liver cells have high MGMT activity while brain and bone marrow cells are known to have low MGMT expression (Christmann et al., 2011; Margison et al., 2003).

This low MGMT level may be the reason for the dose dependent myelotoxicity seen with nitrosourea compounds (Maze et al., 1996). The MGMT level in different tumor cells is also

8

variable with certain tumors showing low level of MGMT like gliomas which explains the relative

sensitivity of gliomas to alkylating anticancer agents (Christmann et al., 2011). However, the low

MGMT in glioma cells is an average value with some gliomas completely lacking MGMT (Kaina

et al. 2007), while in others the variability was over 10 fold (Bodell et al 2001).

Thus it is believed that MGMT may be playing a significant role in the efficacy of

alkylating chemotherapeutic compounds in tumor tissue and also in the toxicity of these

compounds in normal tissues. There is no data available regarding the MGMT levels in canine

cells and tissues.

The primary goal of this project was to study the metabolism of lomustine in dogs and to

compare the cytotoxicity of lomustine and its major metabolites. Another goal was to measure the

MGMT level in canine cells and to correlate it with the cytotoxicity of lomustine and its

metabolites. The hypotheses were:

1. Lomustine and its two major monohydroxylated metabolites (trans-4 and cis-4) have different and

concentration dependent in vitro cytotoxic capabilities that are related to their ability to cause DNA

alkylation and they also have different carbamylating activity on proteins.

2. MGMT, a DNA repair enzyme, is negatively correlated with the cytotoxicity caused by lomustine

and its metabolites in normal canine cells and also in canine lymphoma cell lines.

Specific Aim # 1: To study the metabolism of lomustine in dogs and to compare the concentration

dependent cytotoxicity of lomustine and its major metabolites in malignantly transformed canine

lymphoma cell lines, peripheral blood mononuclear cells (PBMC) from lymphoma dogs and

9 normal canine hepatocytes. The comparative cytotoxic effects of lomustine and its metabolites in canine cells have not been investigated previously. We propose that the anticancer activities of lomustine are primarily exerted by the monohydroxylated metabolites. In addition, lomustine and its two major monohydroxylated metabolites may have different cytotoxic activities in canine primary cells and canine lymphoma cell lines.

Specific Aim # 2: To compare the carbamylation caused by lomustine and its major metabolites in canine lymphoma cell lines and canine primary hepatocytes. This will test the hypothesis that the breakdown of lomustine and its metabolites produces reactive intermediates capable of carbamylating proteins. We will investigate the ability of these compounds to induce carbamylation in canine lymphoma cell lines and hepatocytes. It is believed that carbamylation of proteins could contribute to the off-target toxicity of lomustine like hepatotoxicity. Also the metabolites of lomustine are thought to cause carbamylation to a lesser extent than parent lomustine.

Specific Aim # 3: To detect and quantify O6 hydroxyethyldeoxyguanosine and N7 hydroxyethylguanine adducts in canine lymphoma cell lines and canine primary hepatocytes exposed to lomustine, trans-4 and cis-4. The extent of DNA alkylation may act as a molecular dosimeter of therapeutic response and correlate with the cytotoxicity of lomustine and its metabolites.

Specific Aim # 4: To measure the MGMT enzyme activity and mRNA expression in canine lymphoma cell lines and canine primary hepatocytes and to correlate it with the cytotoxicity of

10 lomustine and its metabolites. This will help to determine whether or not the gene expression and enzyme activity of MGMT, a DNA repair enzyme, is negatively correlated with the cytotoxicity caused by lomustine and its metabolites in canine cells.

In this dissertation, chapters 2-5 describe in detail the aforementioned aims of the study.

Chapter 6 elaborates the overall conclusions of the study and also includes suggestions for future work based on the results obtained.

11

1.4 Figures and Legends

Figure 1.1 Chemical structure of lomustine (A), trans-4-hydroxylomustine (B) and cis-4- hydroxylomustine (C).

(B) (A)

1 4

NO

(C) H N N Cl 1 4 O HO

12

Figure 1.2 Schematic showing the N7 (A) and O6 (B) position of guanine

(B) (A)

7 6

13

1.5 References

Allay, E., Veigl, M., & Gerson, S. L. (1999). Mice over-expressing human O (6)alkylguanine- DNA alkyltransferase selectively reduce O(6)methylguanine mediated carcinogenic mutations to threshold levels after N-methyl-N-nitrosourea. Oncogene, 18(25), 3783-3787.

Babson, J. R., & Reed, D. J. (1978). Inactivation of glutathione reductase by 2-chloroethyl nitrosourea-derived isocyanates. Biochemical and Biophysical Research Communications, 83(2), 754-762.

Bodell, W.J., Giannini, D.D., Hassenbusch, S., & Levin V.A. (2007). Levels of N7-(2- hydroxyethyl) guanine as a molecular dosimeter of drug delivery to human brain tumors. Neuro Oncology 3(4), 241–245.

Christmann, M., Verbeek, B., Roos, W. P., & Kaina, B. (2011). O-6-methylguanine-DNA methyltransferase (MGMT) in normal tissues and tumors: Enzyme activity, promoter methylation and immunohistochemistry. Biochimica Et Biophysica Acta-Reviews on Cancer, 1816(2), 179- 190.

Ducastelle, T., Raguenezviotte, G., Fouinfortunet, H., Matysiak, M., Hemet, J., & Fillastre, J. (1988). The hepatotoxicity of 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (ccnu) in rats - ultrastructural evidence of a delayed microtubular toxicity. Cancer Chemotherapy and Pharmacology, 22(2), 153-162.

Dumenco, L. L., Allay, E., Norton, K., & Gerson, S. L. (1993). The prevention of thymic lymphomas in transgenic mice by human O6-alkylguanine- alkyltransferase. Science, 259(5092), 219-222.

Fornace, A., Kohn, K., & and Kann, H. J. (1978). Inhibition of the ligase step of excision repair by 2-chloroethylisocyanate, a decomposition product of 1,3-bis(2-chloroethyl)-1-nitrosourea. . Cancer Res, 38, 1064.

Fulton, L., & Steinberg, H. (1990). Preliminary-study of lomustine in the treatment of intracranial masses in dogs following localization by imaging techniques. Seminars in Veterinary Medicine and Surgery-Small Animal, 5(4), 241-245.

Gerson, S. (2002). Clinical relevance of MGMT in the treatment of cancer. Journal of Clinical Oncology, 20(9), 2388-2399.

Heading, K. L., Brockley, L. K., & Bennett, P. F. (2011). CCNU (lomustine) toxicity in dogs: A retrospective study (2002-07). Australian Veterinary Journal, 89(4), 109-116.

Heal, J. M., Fox, P. A., Doukas, D., & Schein, P. S. (1978). Biological and biochemical properties of 2-hydroxyl metabolites of 1-2-chloroethyl)-3-cyclohexyl-1-nitrosourea. Cancer Research, 38(4), 1070-1074.

14

Hilton, J., & Walker, M. (1975). Hydroxylation of 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea. Biochemical Pharmacology, 24(23), 2153-2158.

Hoogstraten, B., Gottlieb, J., & Caoili, E. (1973). CCNU (1-[2-chloroethyl]-3-cyclohexyl-1- nitrosourea, NSC-79037) in the treatment of cancer. phase II study. Cancer, 32, 38.

Hosoya, K., Lord, L. K., Lara-Garcia, A., Kisseberth, W. C., London, C. A., & Couto, C. G. (2009). Prevalence of elevated alanine transaminase activity in dogs treated with CCNU (lomustine)*. Veterinary and Comparative Oncology, 7(4), 244-255.

Jung, D., Kim, H., Park, C., Kim, J., Kang, B., Lim, C., & Park, H. (2006). Long-term chemotherapy with lomustine of intracranial meningioma occurring in a miniature schnauzer. Journal of Veterinary Medical Science, 68(4), 383-386.

Kadlubar, F., Beranek, D., Weis, C., Evans, F., Cox, r., & Irving, C. (1984). Charecterization of the purine ring-opened 7-methylguanine and its persistence in rat bladder epithelial DNA after treatment with carcinogen N-methylnitrosourea. Carcinogenesis, 4, 58.

Kaina, B., Christmann, M., Naumann, S., & Roos, W. P. (2007). MGMT: Key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents. DNA Repair, 6(8), 1079-1099.

Kann, H. J., Kohn, K., & Lyles, J. (1974). Inhibition of DNA repair by the 1,3-bis(2-chloroethyl)- 1-nifrosourea breakdown product, 2-chloroethylisocyanate. Cancer Res, 34, 398.

Kastrissios, H., Chao, N., & Blaschke, T. (1996). of high dose oral CCNU in bone marrow transplant patients. Cancer Chemotherapy and Pharmacology, 38(5), 425-430.

Kohn, K. (1977). Interstrand cross-linking of DNA by 1,3-bis(2-chloroethyl)-1-nitrosourea and other 1-(2-haloethyl)-1-nitrosoureas. Cancer Res., 37(5), 1450.

Kristal, O., Rassnick, K., Gliatto, J., Northrup, N., Chretin, J., Morrison-Collister, K., & Moore, A. (2004). Hepatotoxicity associated with CCNU (lomustine) chemotherapy in dogs. Journal of Veterinary Internal Medicine, 18(1), 75-80.

Lawley, P. (1976). Methylation of DNA by carcinogens: Some applications of chemical analytical methods. In M. Montesano, H. Bartsch & L. and Tomatis (Eds.), Screening tests in chemical carcinogenesis (pp. 181). Lyon: IARC Scientific Publications.

Lee, F., Workman, P., Roberts, J., & Bleehen, N. (1985). Clinical pharmacokinetics of oral ccnu (lomustine). Cancer Chemotherapy and Pharmacology, 14(2), 125-131.

Lemoine, A., Lucas, C., & Ings, R. M. J. (1991). Metabolism of the chloroethylnitrosoureas. Xenobiotica, 21(6), 775-791.

15

Madewell, B. R., & Theilen, G. H., (1987). Haematopoietic neoplasms, sarcomas and related conditions (IV). In G. H. Theilen, & B. R. Madewell (Eds.) Veterinary Cancer Medicine, 2nd edn. (pp. 392-407) Lea & Febiger, Philadelphia.

Margison, G. P., Povey, A. C., Kaina, B., & Santibanez Koref, M. F. (2003). Variability and regulation of O6-alkylguanine-DNA alkyltransferase. Carcinogenesis, 24(4), 625-635.

Maze, R., Carney, J. P., Kelley, M. R., Glassner, B. J., Williams, D. A., & Samson, L. (1996). Increasing DNA repair methyltransferase levels via bone marrow stem cell transduction rescues mice from the toxic effects of 1,3-bis(2-chloroethyl)-1-nitrosourea, a chemotherapeutic alkylating agent. Proceedings of the National Academy of Sciences of the United States of America, 93(1), 206-210.

Meuten, D.J, Jacobs, R.M., Messick, J.B., & Valli V.E. (2008). Tumors of the Hemolymphatic System. In D.J. Meuten (Eds.) Tumors in Domestic Animals, 4th edn (pp.138)

Mitchell, E., & Schein, P. (1986). Contributions of nitrosoureas to cancer-treatment. Cancer Treatment Reports, 70(1), 31-41.

Moertel, C., Schutt, A., Reitemeier, R., & Hahn, R. (1972). Cancer Res, 32, 1278.

Moore, A., London, C., Wood, C., Williams, L., Cotter, S., L'Heureux, D., & Frimberger, A. (1999). Lomustine (CCNU) for the treatment of resistant lymphoma in dogs. Journal of Veterinary Internal Medicine, 13(5), 395-398.

Morrison, W. (2004). Lymphoma in dogs and cats. Jackson, WY: Teton NewMedia.

Nakatsuru, Y., Matsukuma, S., Nemoto, N., Sugano, H., Sekiguchi, M., & Ishikawa, T. (1993). O6-methylguanine-dna methyltransferase protects against nitrosamine-induced hepatocarcinogenesis. Proceedings of the National Academy of Sciences of the United States of America, 90(14), 6468-6472.

Ochs, K., & Kaina, B. (2000). Apoptosis induced by DNA damage O-6-methylguanine is bcl-2 and caspase-9/3 regulated and fas/caspase-8 independent. Cancer Research, 60(20), 5815-5824.

Pegg, A. (1977). Formation and metabolism of alkylated nucleosides: Possible role in carcinogenesis by nitroso compoundsand alkylating agents. In G. Klein, & S. and Weinhouse (Eds.), Adv.cancer res. (pp. 195). New York: Academic Press.

Rassnick, K., Moore, A., Williams, L., London, C., Kintzer, P., Engler, S., & Cotter, S. (1999). Treatment of canine mast cell tumors with CCNU (lomustine). Journal of Veterinary Internal Medicine, 13(6), 601-605.

16

Reed, D. (1981). Metabolism of nitrosoureas . In A. Prestayko, S. Crooke, L. Baker, S. Caster & P. Sehein (Eds.), Nitrosoureas: Current status and new developments (pp. 51). London: Academic.

Reed, D. J. (1984). Synthesis and characterization of nitrosoureas. Ottenbrite, R.M.and G.B.Butler (Ed.).Drugs and the Pharmaceutical Sciences, Vol.24.Anticancer and Interferon Agents: Synthesis and Properties.Xi+325p.Marcel Dekker, Inc.: New York, N.Y., Usa: Basel, Switzerland.Illus, , 177-202.

Richards, K.L., Motsinger-Reif, A.A., Chen, H., Fedoriw, Y., Fan,C., Nielsen, D.M., Small, G.W., Thomas, R., Smith, C., Dave, S.S., Perou, C.M., Breen, M., Borst, L.B., & Suter, S.E. (2013). Gene profiling of canine B-cell lymphoma reveals germinal center and postgerminal center subtypes with different survival times, modeling human DLBCL. Cancer Research. 73; 5029

Rinne, M. L., He, Y., Pachkowski, B. F., Nakamura, J., & Kelley, M. R. (2005). N-methylpurine DNA glycosylase overexpression increases alkylation sensitivity by rapidly removing non-toxic 7-methylguanine adducts. Nucleic Acids Research, 33(9), 2859-2867.

Risbon, R. E., de Lorimier, L. P., Skorupski, K., Burgess, K. E., Bergman, P. J., Carreras, J., Clifford, C. A. (2006). Response of canine cutaneous epitheliotropic lymphoma to lomustine (CCNU): A retrospective study of 46 cases (1999-2004). Journal of Veterinary Internal Medicine, 20(6), 1389-1397.

Saba, C. F., Thamm, D. H., & Vail, D. M. (2007). Combination chemotherapy with L- asparaginase, lomustine, and prednisone for relapsed or refractory canine lymphoma. Journal of Veterinary Internal Medicine, 21(1), 127-132.

Sakumi, K., Shiraishi, A., Shimizu, S., Tsuzuki, T., Ishikawa, T., & Sekiguchi, M. (1997). Methylnitrosourea-induced tumourigenesis in MGMT gene knockout mice. . Cancer Res., 57, 2415.

Sariban, E., Erickson, L., & Kohn, K. (1984). Effects of carbamoylation on cell survival and DNA repair in normal human embryo cells (IMR-90) treated with various 1-(2-chloroethyl)-1- nitrosoureas. Cancer Res., 44(4), 1352.

Sauerbrey, M. L., Mullins, M. N., Bannink, E. O., Van Dorp, T. E. Renate, Kaneene, J. B., & Obradovich, J. E. (2007). Lomustine and prednisone as a first-line treatment for dogs with multicentric lymphoma: 17 cases (2004–2005). Journal of the American Veterinary Medical Association, 230(12), 1866-1869.

Sauerbrey, M. L., Mullins, M. N., Bannink, E. O., Van Dorp, T. E. R., Kaneene, J. B., & Obradovich, J. E. (2007). Lomustine and prednisone as a first-line treatment for dogs with multicentric lymphoma: 17 cases (2004-2005). Javma-Journal of the American Veterinary Medical Association, 230(12), 1866-1869.

17

Singer, B. (1975). The chemical effect of nucleic acid alkylation and their relation to mutagenesis and carcinogenesis. In W. Cohen (Ed.), Progr. nucleic acid res. and mot. biol (pp. 219). New York: Academic Press.

Skorupski, K. A., Clifford, C. A., Paoloni, M. C., Lara-Garcia, A., Barber, L., Kent, M. S., Sorenmo, K. U. (2007). CCNU for the treatment of dogs with histiocytic sarcoma. Journal of Veterinary Internal Medicine, 21(1), 121-126.

Swenberg, J. A., Richardson, F. C., Boucheron, J. A., & Dyroff, M. C. (1985). Relationships between dna adduct formation and carcinogenesis. Environmental Health Perspectives, 62(OCT), 177-183.

Swerdlow, S., Campo, E., Harris, N., Jaffe, E., Pileri, S., Stein, H., . . . Vardiman, J. W. (2008). WHO classification of tumours of haematopoietic and lymphoid tissues. (4th ed.). Lyon, France.: IARC Press.

Tew, K., Coluin, M., & Chabner, B. (1996). Alkylating agents. In B. Chabner, & D. Longo (Eds.), Cancer chemotherapy and biotherapy (2nd ed., pp. 297). Philadelphia PA: Lippincott-Raven.

Tew, K., Coluin, O., & Jones, R. (2006). Clinical and high-dose akylating agents
. In D. L. BA Chabner (Ed.), Cancer chemotherapy and biotherapy (4th ed., pp. 283). Philadelphia, PA: Lippincott Willimas & Wilkins.

Tong, W. P., Kohn, K. W., & Ludlum, D. B. (1982). Modifications of dna by different haloethylnitrosoureas. Cancer Research, 42(11), 4460-4464.

Vail, D., MacEwen, E., & Young, K. (2001). Canine lymphoma and lymphoid leukemia. In M. E. Withrow SJ (Ed.), Small animal clinical oncology (3rd ed. ed., pp. 558) WB Saunders Co.

Wheeler, G., Johnston, T., Bowdon, B., McCaleb, G., Hill, D., & Montgomery, J. (1977). Comparison of the properties of metabolites of CCNU. Biochem Pharmacol, 26, 2331.

Williams, L., Rassnick, K., Power, H., Lana, S., Morrison-Collister, K., Hansen, K., & Johnson, J. (2006). CCNU in the treatment of canine epitheliotropic lymphoma. Journal of Veterinary Internal Medicine, 20(1), 136-143.

18

CHAPTER II

Development of validated analytical method for detection of lomustine and its metabolites

and metabolism characteristics of lomustine in dogs

2.1 Introduction

There are several studies in the literature indicating good efficacy of lomustine in combination with other chemotherapeutic agents for the treatment of resistant canine lymphoma

(Moore et al., 1999; Saba et al., 2007; Sauerbrey et al., 2007). Lomustine is usually administered to dogs orally at 50-110 mg/m2 every three weeks (Kristal et al., 2004; Moore et al., 1999;

Sauerbrey et al., 2007). One of the unique characteristics of lomustine is that it undergoes extensive first-pass hepatic metabolism to the geometric isomeric forms of a monohydroxylated metabolite in different animal species and humans (Kastrissios et al., 1996; Weinkam & Lin,

1982). Wheeler et al. have concluded from their toxicity study in mice that the hydroxylation at the 4-position of the cyclohexyl ring confers both isomers (cis-4 and trans-4) of the metabolite with enhanced alkylating activity and reduced toxic effects relative to the lomustine, resulting in an approximately two-fold increase in the therapeutic index of each of the metabolite relative to the parent compound (Wheeler et al., 1977). Unfortunately, metabolism of lomustine is associated with large interindividual variability. A reduction in the metabolism of lomustine to its hydroxylated isomeric forms is associated with a narrower therapeutic margin, and may manifest clinically as an increased risk of toxicity (Chang et al., 1993; Kastrissios et al., 1996). Although numerous studies have reported efficacy of lomustine for canine lymphoma, the metabolism of

19 lomustine has not been studied in dogs previously. In order to do that, we first developed an analytical method for the detection of lomustine and its monohydroxylated metabolites in canine plasma.

Stability of the drugs is a major issue in such studies as nitrosoureas are known to be a highly labile class of drugs. For many of the drugs in this class, there is about 5% degradation when a solution is kept at room temperature in phosphate buffer saline or normal saline for 1 hour

(Bosanquet, 1985). Also serum proteins are known to affect the stability of lomustine (Chatterji et al., 1978; Montgomery et al., 1967; Weinkam et al., 1980). It has been shown that human serum albumin has catalytic effect on lomustine thought to be due to the increased stress from distortion of bond angles of an already inherently unstable molecule (Weinkam et al., 1980). In aqueous media, lomustine gets rapidly ionized and undergoes uncatalyzed decomposition (Chatterji et al.,

1978). In our project, the stability study in methanol and plasma was very important as the stock solutions of lomustine and its metabolites and the blood/plasma samples from dogs that received lomustine may have to be stored for a few hours before analysis. In addition, to assess the in vitro cytotoxicity of lomustine and its metabolites in canine cells (elaborated in chapter 3), their stability in cell culture conditions had to be evaluated.

In this chapter we first describe the development of an HPLC method for the quantitation of lomustine and its metabolites. We also describe studies to evaluate the stability of the compounds. In addition, the metabolism pattern of lomustine in dogs is evaluated.

20

2.2 Materials and Methods

Analytical detection of lomustine and its monohydroxylated metabolites

Sample preparation

The monohydroxylated metabolites of lomustine, trans-4 (trans-(1-(2-Chloroethyl)-3-

(trans-4-hydroxycyclohexyl)-1-nitrosourea) and cis-4 (1-(2-Chloroethyl)-3-(cis-4- hydroxycyclohexyl)-1-nitrosourea) were synthesized and purified by Obiter Research, LLC

(Champaign, IL). A standard solution of 1 mg lomustine (Sigma-Aldrich), trans-4 and cis-4 was prepared in 1 ml HPLC grade ice-cold methanol. Standards were prepared by the addition of a specified amount of lomustine, trans-4 and cis-4 in ice-cold methanol to blank canine plasma samples, 0.5 ml each, over a range from 25 to 5,000 ng/ml. Phenytoin (Sigma-Aldrich) was used as an internal standard. The internal standard was prepared in 1 ml ice-cold methanol (1 mg/ml) and diluted 1 to 10 in methanol to yield 100 ng/l standard solution. To each sample, 20 l of 100 ng/l internal standard was added.

Extraction method

Clean 16 × 150 mm Pyrex screw-cap culture tubes with polytetrafluoroethylene (PTFE) cap liners were silanized with trimethylchlorosilane (TMCS; Pierce Chemical) before use by rinsing them first with methanol, followed by a 5% solution of TMCS in hexane (Fisher Scientific).

This step was followed by rinsing with methanol (Fisher Scientific) twice. The silanized tubes

21 were allowed to air-dry at room temperature. The analytical method used was as follows: Plasma samples (0.5 ml) were pipetted into 15-ml screw-cap tubes. Twenty µl of 100 ng/l internal standard was added to each tube, followed by 2 ml of dichloromethane. The tubes were then vortex mixed for a few seconds, then centrifuged at 2000 rpm for 15 minutes. The top aqueous layer was discarded off and the organic layer was transferred into clean tubes and dried under a stream of nitrogen at 20ºC in a water bath. The residue was reconstituted with 120 l of ice-cold methanol, vortex mixed briefly, and then transferred to an autosampler vial equipped with a 300 l spring- loaded insert. All samples were analyzed within 24 hours post-extraction.

Instrumentation

The HPLC procedure was adapted from that described by Lee et al. (Lee et al., 1985). The instrument employed was Agilent HP 1100 HPLC system with quaternary solvent delivery pumps, vacuum degasser, thermostatted column compartment, 1100 series photodiode array detector and

1100 series autosampler. The column was an Inertsil ODS, 5 m particle size, 4.6 mm x 250 mm column size. The mobile phase consisted of 34% acetonitrile in water (Solvent A), 44% acetonitrile in water (Solvent B), and 64% acetonitrile in water (Solvent C) run with a flow rate of

0.8 ml/minute. Samples were eluted by running two-step linear gradient commencing at the time of injection, with an initial condition of solvent A and proceeding to solvent B within 6 minutes, and then to solvent C within 8 minutes with a total running time of 30 minutes. Acetonitrile (A998-

4, Fisher Scientific) and methanol (MX0488-1, EM Science) were HPLC grade. The diode array detector was set up for single wavelength acquisition at 254 nm with a 12 nm span. Injections were made with a 20 l sample loop.

22

Standard Curve Generation and Method Validation

Standard curves were constructed by plotting standard lomustine, trans-4 and cis-4 concentrations versus the ratio of lomustine, trans-4 and cis-4: internal standard peak areas.

Standard curves were generated with Sigma Plot for Windows (Aspire Software International).

The areas of peaks corresponding to lomustine, trans-4, cis-4 and internal standard were recorded, and the internal standard values thereby used to normalize the lomustine, trans-4 and cis-4 areas.

Integrated peak values were entered into QuattroPro for Windows (Borland Software

Coorporation, Scotss valley) for statistical analysis of standards and for calculation of amounts of lomustine, trans-4 and cis-4. Each standard sample was run in duplicate.

The quantitative HPLC method of lomustine, trans-4 and cis-4 was validated by examining the measurement of consistency of results (within-run and between-run), correlation (coefficient of determination of the standard curve), and extraction efficiency of the assay. The within-run precision was calculated from similar responses from six repeats of three control samples (50, 500,

2500 ng/ml) in one run. The between-run precision was determined by comparing the calculated response (in ng/ml backfit of the standard curve) of the low (50 ng/ml), middle (500 ng/ml), and high (2500 ng/ml) control samples over three consecutive daily runs (total of six runs). The assay accuracy for within-run and between-run was established by determining the ratio of calculated response to expected response for low (50 ng/ml), middle (500 ng/ml), and high (2500 ng/ml) control samples over six runs.

23

Standard curve correlation was measured by the mean coefficient of determination (r2) for six consecutive daily runs. The extraction efficiency was determined by comparing the response

(in area) of low (50 ng/ml), middle (500 ng/ml), and high (2500 ng/ml) standards, and the internal standard (2000 ng/ml) spiked into blank plasma eluent before evaporation to the equivalent extracted standards. The lower limit of detection (LOD) was calculated utilizing the analyte’s peak height compared to the baseline noise. By this method, the LOD was defined as the lowest concentration of analyte producing a peak greater than or equal to three times the baseline noise of the chromatogram.

Stability studies

Stability in methanol and canine plasma

The stability of lomustine, cis-4 and trans-4 (1000 ng/ml, n=3) in methanol stored at room temperature, 4oC (refrigerated) and –20oC for 1, 3 and 7 days were investigated. In this experiment, instrument responses from incubated samples were compared with that of freshly prepared samples using freshly prepared standard samples spiked with freshly prepared internal standard right before extraction. The stability of lomustine, cis-4 and trans-4 in canine plasma samples were also investigated in refrigerated (4oC) and frozen samples (-20oC and -80oC) stored for 1, 3, 7 and 14 days (1000 ng/ml, n=3 for each experiment)

24

Stability in blood, plasma and culture media at 37ºC

The drugs were added to culture media DMEM and L-15 canine whole blood and plasma at the concentration of 500 ng/ml and incubated at 37ºC. The drug concentration was measured at

30 minutes, 1, 2, 4, 6 and 8 hours by HPLC. L-15 is a bicarbonate free media that is buffered by salts, amino acids and galactose. The culture media were supplemented with 10% FBS and 1% penicillin/streptomycin. Four replicates were used for each experiment.

Metabolism characteristics of lomustine in dogs

The in vivo metabolism characteristics of lomustine was studied in two research hounds.

The dogs received 1.7 mg/kg (53.6 mg/m2) oral administration of commercially available lomustine capsules and blood samples were collected at 15, 30, 45 minutes, 1, 2, 4, 6, 8 and 12 hours after drug administration. Lomustine was also administered orally to a lymphoma dog at the same dose and the concentration of lomustine and its metabolites were analyzed in the blood samples using the HPLC method described above.

Red blood cell partitioning of lomustine and its metabolites

Certain drugs that have greater lipophilicity may penetrate RBCs where it can be stored temporarily and this may bring down the plasma concentration of the drugs. Hence studying the

RBC partitioning of drugs can help to select the appropriate matrix among whole blood, plasma or serum for assaying pharmacokinetic behavior of the drug (Hinderling, 1997). For this study,

25 commercially purchased canine whole blood in EDTA (The Veterinarians’ Blood Bank) was used.

Two sets of blood samples (4 samples in each set, 1 ml/sample) were spiked with lomustine, trans-

4 and cis-4 at the concentration of 500 ng/ml. After incubating the samples at 37ºC for different time intervals, one set of blood samples was directly analyzed for drug concentration and in the other set, the plasma was separated and the drug concentration was measured in plasma.

The concentration in RBCs was calculated as:

CRBC= [(Cblood - Cplasma) x (1 - Hematocrit)] Hematocrit

where Cblood and Cplasma are the concentrations of the drug in blood and plasma, respectively.

Further the extent of partitioning of the drugs was measured by the following formula:

Ke/p= Concentration of drug in RBC Concentration of drug in plasma

2.3 Results

Analytical detection of lomustine and its monohydroxylated metabolites

The HPLC diode array detection method reported here readily detects lomustine, cis-4 and trans-4 in canine plasma, with LOD of lomustine, cis-4 and trans-4 in plasma of about 10 ng/120

µl, 5 ng/120 µl, and 5 ng/120 µl, respectively based on signal to noise ratio (data not shown). The mean extraction efficiency values for lomustine, cis-4 and trans-4 were 73%, 89% and 90%, respectively from canine plasma samples on HPLC (Table 2.1). The lomustine, cis-4 and trans-4

26 peak eluted at around 25 minutes (min) (0.3 min), 10.9 min (0.3 min), 10 min (0.3 min), respectively and the internal standard peak eluted at 13.4 min (0.3 min) (Figure 2.1).

The resultant standard curve was curvilinear from 25 ng/ml to 5000 ng/ml for lomustine, cis-4 and trans-4 (Figure 2.2). Both linear and curvilinear regression analysis were evaluated for generation of standard curves for lomustine. Interestingly, back-calculated concentrations for each standard sample were within 20% of known amount sometimes when linear and sometimes when curvilinear regression analyses were used. Therefore, based on back-calculated concentrations for each standard sample either curvilinear or linear regression analyses were used for the data evaluation.

For within-run, the mean coefficient of variations (CV) were 5.1%, 1.9%, and 2.0% for lomustine, cis-4 and trans-4, respectively (Table 2.2). In addition, the mean within-run accuracies for this method (n = 6 for each value) were 94.5%, 96.7% and 100.4% for lomustine, cis-4 and trans-4, respectively (Table 2.2). For between-run, the mean CV were 10.9%, 8.5%, and 5.7% for lomustine, cis-4 and trans-4, respectively (Table 2.3). The mean between-run accuracies for this method (n = 6 for each value) were 97.3%, 99.2%, and 101.7% for lomustine, cis-4 and trans-4, respectively (Table 2.3).

Stability studies

Results of HPLC analysis demonstrated 105 ± 6.0%, 98 ± 1.5% and 92 ± 3.7% recovery of lomustine, cis-4 and trans-4, respectively stored in methanol at room temperature for 24 hours

27

(n=3 for each compound) (Table 2.4). Here, 6.0%, 1.5% and 3.7% denotes the respective percent standard deviations (SD). The stability studies in methanol demonstrated 111 ± 0.3% (SD), 103 ±

0.6% (SD) and 100 ± 1.1% (SD) recovery and 110 ± 0.1% (SD), 102 ± 0.6% (SD) and 99.7 ± 0.8% recovery of lomustine, cis-4 and trans-4 in refrigerated and frozen (-20oC) samples, respectively stored for 24 hr (Table 2.4). The stability of lomustine, cis-4 and trans-4 in refrigerated and frozen

(-20oC) samples were 110 ± 3.9% (SD), 101 ± 4.7% (SD) and 100 ± 0.8% (SD), and 107 ± 0.1%

(SD), 111 ± 0.5% (SD) and 102 ± 0.8%, respectively stored in methanol solution for 3 days (Table

4). The stability of lomustine, cis-4 and trans-4 in refrigerated and frozen (-20oC) samples were

108 ± 0.6% (SD), 80.9 ± 0.4% (SD) and 94 ± 0.1% (SD), and 108 ± 2.2% (SD), 81.5 ± 0.3% (SD) and 96 ± 0.4%, respectively stored in methanol solution for a week (Table 2.4).

Results of HPLC analysis demonstrated 71 ± 0.8% (SD), 95 ± 2.3% (SD) and 76 ± 0.6% recovery in refrigerated, 81 ± 1% (SD), 109 ± 2.3% (SD) and 89 ± 4.2% recovery in –20oC, and

81 ± 1.0% (SD), 103 ± 8.3% (SD) and 88 ± 3.3% recovery in –80oC samples of lomustine, cis-4 and trans-4, respectively stored in plasma for 24 hours (n=3 for each compound) (Table 2.5).

The stability studies in canine plasma demonstrated 37 ± 0.8% (SD), 80 ± 1.8% (SD) and

70 ± 1.3% (SD) recovery in refrigerated, 72 ± 3.2% (SD), 97 ± 2.7% (SD) and 119 ± 4.5% recovery in –20oC, and 81 ± 3.5% (SD), 93 ± 1.5% (SD) and 111 ± 5.9% recovery in –80oC samples of lomustine, cis-4 and trans-4, respectively, stored for 3 days (Table 2.5). The stability of lomustine, cis-4 and trans-4 in canine plasma stored for a week in refrigerator were 20 ± 1.1% (SD), 26 ±

1.5% (SD) and 16 ± 1.1% (SD), respectively, in –20oC samples were 73 ± 3.8% (SD), 79 ± 1.9%

(SD) and 68 ± 0.9%(SD), respectively, in –80oC samples were 83 ± 9.3% (SD), 72 ± 2.4% (SD)

28 and 79 ± 1.5%, respectively. The stability of lomustine, cis-4 and trans-4 in canine plasma stored for two-week in refrigerator were 7 ± 0.1% (SD), 7 ± 1.9% (SD) and 14 ± 1.7% (SD), respectively, in –20oC samples were 62 ± 0.9% (SD), 77 ± 2.2% (SD) and 83 ± 0.2, respectively, in –80oC samples were 69 ± 0.8% (SD), 88 ± 4.0% (SD) and 89 ± 2.1%, respectively.

The stability studies indicated that lomustine and its two monohydroxylated metabolites are relatively stable in methanol solution at room temperature for 24 hours. Lomustine and its metabolites are also relatively stable in methanol solution both in refrigerated and frozen (-20oC) samples for up to a week. Based on our data, cis-4 seems to be less stable than lomustine and trans-4 in methanol solution when stored for a week in refrigerated and frozen (-20oC) samples.

In the present study, the stability data indicated less stability of lomustine and its metabolites in canine plasma samples than in methanol solution especially in refrigerated samples.

For example, we only had 37 ± 0.8% (SD), 80 ± 1.8% (SD) and 70 ± 1.3% (SD) recovery in refrigerated canine samples of lomustine, cis-4 and trans-4, respectively stored only for 3 days.

Based on the data, the best recovery was obtained when canine plasma samples were stored at -

80oC.

The results of the stability study at 37ºC show that lomustine and its metabolites degrade in the culture media, blood and plasma at 37ºC, the degradation process being quickest in DMEM

(Figure 2.3). Lomustine, trans-4 and cis-4 seem to be more stable in the bicarbonate free media L-

15 as compared to DMEM. Lomustine and its metabolites were only detected up to 2 hours in the culture media and 4 hours in whole blood and plasma. The results indicate that in cell culture

29 conditions, there is rapid breakdown to lomustine and its metabolites and that the drugs may not be detected beyond 3-4 hours.

Metabolism characteristics of lomustine in dogs

Lomustine was not detected in plasma samples of the three dogs following oral administration of the drug. Figure 2.4A shows that trans-4 is the major metabolite of lomustine and the maximum concentration was about 325 ng/ml detected 1 hour after drug administration for both the research hounds. The maximum concentration of cis-4 in the three dogs was around

40-65 ng/ml (Figure 2.4B).

Red blood cell partitioning of lomustine and its metabolites

The results show that there is a minor partitioning of these drugs into/onto RBC (Table

2.6). About 9-15% of the drug was found to be in the RBC compartment. For our metabolism studies, drug concentrations were measured in plasma and not whole blood. This small difference in the drug levels in plasma and whole blood could have led to a slight underestimation of the actual drug concentrations in blood.

2.4 Discussion

Lomustine has been identified as a potentially important therapeutic agent for use in the treatment of various cancers in dogs; as such we needed a sensitive and reliable analytical method

30 to perform pharmacokinetic experiments with this agent in dogs. In particular, the method selected needed to be sufficiently sensitive to detect low concentrations of lomustine and its possible monohydroxylated metabolites in canine plasma from test animals.

The resultant HPLC method is a sensitive analytical method with the LOD of lomustine, cis-4 and trans-4 in plasma of about 10 ng/120 µl, 5 ng/120 µl, and 5 ng/120 µl, respectively based on signal to noise ratio. Additionally, satisfactory recovery was obtained for lomustine and its monohydroxylated metabolites extracted from plasma samples. The developed extraction procedure is very simple (one step liquid/liquid extraction procedure) and suitable to analyze multiple samples in a short-time period. Additionally, with the mentioned HPLC method, we were able to separate chromatographic peaks of cis-4 and trans-4 isomers by almost 1 minute. The CV for within-run and between-run samples were less than 15% for lomustine, trans-4 and cis-4 indicating good method precision.

The stability studies indicated that lomustine and its two monohydroxylated metabolites are relatively stable in methanol solution at room temperature for 24 hours and also when refrigerated and frozen (-20oC) for up to a week. In the present study, the stability data indicated less stability of lomustine and its metabolites in canine plasma samples than in methanol solution especially in refrigerated samples. Based on the data, the best recovery was obtained when canine plasma samples were stored at -80oC. Due to stability issues, unknown canine samples following administration of lomustine should be stored at -80oC and should be analyzed within a week for better accuracy when determining the pharmacokinetic parameters of lomustine in dogs.

31

Lomustine is known to be an unstable compound and various factors accelerate the process of degradation. At around pH 6 and above, there is a drastic increase in degradation of lomustine in aqueous buffer (Loftsson & Fridriksdottir, 1990). Another study looked at the stability of lomustine in different culture media at 37ºC and found that in media that contain bicarbonate, the half-life of lomustine is lower than that of in bicarbonate free media (Lemoine et al., 1991).

Presence of serum in the media also has a lowering effect on the half-life of lomustine (Weinkam et al., 1980). The above data suggest that physiological body conditions are not conducive for the stability of lomustine and its metabolites. After first pass metabolism, lomustine forms the hydroxylated metabolites (trans-4 and cis-4) that are more water soluble but these metabolites are also very unstable (Lemoine et al., 1991). It is believed that lomustine and its metabolites form extremely reactive alkylating intermediates, like carbonium ions, which are responsible for the cytotoxic effects of the drug (Heal et al., 1978; Lemoine et al., 1991).

The metabolic profile of oral lomustine in dogs in our study is similar to that reported in humans and rats with trans-4 being the major metabolite followed by cis-4 and the complete absence of parent lomustine. However, since the study was only done in three dogs, these results have to be considered as preliminary. In our study, the dogs received lomustine at the dose of 1.7 mg/kg and the two research hounds showed similar peak levels of trans-4 (about 325 ng/ml), but the levels for the same in the lymphoma dog was lower. The peak cis-4 lomustine level was between 40-65 ng/ml. In a published study of four human patients that received lomustine at the dose of 4.1 mg/kg, parent lomustine was not detected in blood. The average peak concentration of trans-4 was 530 ng/ml and that for cis-4 was 320 ng/ml (Lee et al., 1985). In another study in human patients that used high dose lomustine (15 mg/kg), the peak plasma concentration detected

32 ranged between 410-2310 ng/ml for trans-4 and 180-1820 ng/ml for cis-4 (Kastrissios et al., 1996), a 5 and 10 fold difference between the maximum concentrations observed among patients, respectively. This supports the premise that there is a large interindividual difference in the metabolic profile of lomustine.

As parent lomustine was not detected in the blood at any time after administration of the drug, the RBC partitioning behavior of lomustine and its metabolites was studied. The results showed that there is a minor partitioning of these drugs into RBC. But the RBC partitioning values are not high enough to explain why lomustine was not detected in plasma samples obtained from three dogs after oral lomustine dosing. This study suggests that like in other animal species, the absence of lomustine in dogs’ plasma is most likely due to rapid “first pass” metabolism and not due to significant RBC partitioning.

In conclusion, we have developed and validated relatively sensitive, simple and reliable

HPLC method to detect and quantify lomustine and its two monohydroxylated metabolites for future pharmacokinetic/toxicokinetic studies of lomustine in dogs. This method was utilized to study the stability of compounds and also to measure the drug concentration in the dog blood samples. Lomustine and its metabolites are highly unstable and therefore consideration must be given to the stability of these compounds while storing and using them for experimental purposes.

Finally, the metabolic profile of lomustine in dogs was studied and was found to be similar to that in humans.

33

2.5 Figures and Legends

Figure 2.1 A typical HPLC chromatogram of trans-4-hydroxylomustine, cis-4-hydroxylomustine and lomustine with internal standard (phenytoin) in methanol as extracted from spiked sample (1000 ng/0.5 ml plasma). Absorbance at 254 nm is plotted vs. retention time.

10.01 10.88 Cis-4-hydroxylomustine

16 Trans-4-hydroxylomustine 14 12 13.42 25.08 mAU 10 Lomustine

8 Internal standard 6 4 2 0 7.5 10 12.5 15 17.5 20 22.5 25 27.5 min u

34

Figure 2.2 Typical calibration curves for cis-4-hydroxylomustine (A), trans-4-hydroxylomustine (B), and lomustine (C) extracted from canine plasma samples, indicating coefficient of determination (R2) of 0.998, 0.997 and 0.991, respectively. "Instrument Response" on the x-axis refers to Area/internal Standard Area; "Concentration" on the y-axis is in ng/ml.

(A) (B)

6000 6000

5000 5000

)

l 4000

)

l m 4000

/

m

g

/

n

g

(

n

(

n

o

n i 3000

t

o

i

a 3000

t

r

a

t

r

t

n

n

e

e

c

c

n

n o 2000 o 2000

C

C

1000 1000

0 0 0 2 4 6 8 10 12 0 2 4 6 8 10 12 Instrument Response Instrument Response

(C)

6000

5000

)

l 4000

m

/

g

n

(

n

o i 3000

t

a

r

t

n

e

c

n

o 2000

C

1000

0 0 2 4 6 8 10 12 Instrument Response

35

Figure 2.3 Stability of lomustine (A), trans-4-hydroxylomustine (B) and cis-4-hydroxylomustine (C) in canine whole blood, canine plasma and culture media DMEM and L15. The ‘X-axis’ represents time in minutes and the ‘Y-axis’ represents concentration in ng/ml.

(A) (B)

(C)

36

Figure 2.4 Plasma concentration of trans-4-hydroxylomustine (A) and cis-4-hydroxylomustine (B) following single 1.7 mg/kg oral administration of lomustine in three dogs.

(A) (B)

37

Table 2.1 Extraction efficiencies of HPLC assay used to quantify cis-4-hydroxylomustine, trans- 4-hydroxylomustine and lomustine in canine plasma samples.

Theoretic Extraction Efficiency Extraction Efficiency Extraction Efficiency Concentration (Cis-4-hydroxylomustine (Trans-4-hydroxylomustine (Lomustine (%) (ng/ml) (%) (mean ± SD) (n = 5) (%) (mean ± SD) (n = 5) (mean ± SD) (n = 5) 50 90 ± 3.5 87 ± 2.5 72 ± 8.1 500 90 ± 3.4 90 ± 2.9 74 ± 3.5 2500 91 ± 5.9 89 ± 6.1 73 ± 12.7 Mean 90 89 73

38

Table 2.2 Within-run precisions of HPLC assay used to quantify lomustine and its metabolites in canine plasma samples.

Compound Theoretic Measured concentration Accuracy (%) Coefficient of Concentration (ng/ml; mean ± SD) (mean ± SD) Variation (%) (ng/ml) (n = 6) Trans-4- 53 ± 1.6 106.2 ± 2.3 2.2 hydroxylomustine 50 500 496 ± 6.0 99.2 ± 1.2 1.2 2500 2395 ± 66.3 95.8 ± 2.4 2.5 Mean 100.4 2.0 Cis-4- 50 54 ± 1.4 107.1 ± 2.8 2.6 hydroxylomustine 455 ± 5.9 90.8 ± 1.3 1.4 500 2301 ± 42.3 92.3 ± 1.7 1.8 2500 Mean 96.7 1.9 Lomustine 50 54 ± 3.1 107.1 ± 6.3 5.9 500 450 ± 15.3 90.1 ± 3.1 3.4 2500 2158 ± 131.0 86.3 ± 5.2 6.1 Mean 94.5 5.1

39

Table 2.3 Between-run precisions of HPLC assay used to quantify lomustine and its metabolites in canine plasma samples.

Compound Theoretic Measured concentration Accuracy (%) Coefficient of Concentration (ng/ml; mean ± SD) (mean ± SD) Variation (%) (ng/ml) (n = 6) Trans-4- 51 ± 3.0 101.5 ± 6.0 5.9 hydroxylomustine 50 500 519 ± 27.5 103.8 ± 5.5 5.3 2500 2495 ± 144.0 99.8 ± 5.8 5.8

Mean 101.7 5.7 Cis-4- 50 49 ± 4.0 97.7 ± 8.0 8.1 hydroxylomustine 507 ± 50.0 101.3 ± 10.0 9.9 500 2468 ± 186.4 98.7 ± 7.5 7.6 2500

Mean 99.2 8.5 Lomustine 50 51 ± 5.4 102.3 ± 10.8 10.5 500 482 ± 58.5 96.5 ± 11.7 12.1 2500 2326 ± 233.9 93 ± 9.4 10.1

Mean 97.3 10.9

40

Table 2.4 Stability of cis-4-hydroxylomustine, trans-4-hydroxylomustine and lomustine in methanol.

Duration of Temperature % Recovery of Cis-4- % Recovery of Trans- % Recovery of Incubation hydroxylomustine 4- hydroxylomustine Lomustine (Mean ± SD, n=3) (Mean ± SD, n=3) (Mean ± SD, n=3)

24 hrs Room 98 ± 1.5 92 ± 3.7 105 ± 6 temperature Refrigerated 103 ± 0.6 100 ± 1.1 111 ± 0.3

-20oC 102 ± 0.6 99.7 ± 0.8 110 ± 0.1

3 days Refrigerated 101 ± 4.7 100 ± 0.8 110 ± 3.9

-20oC 111 ± 0.5 102 ± 0.8 107 ± 0.1

1 week Refrigerated 80.9 ± 0.4 94 ± 0.1 108 ± 0.6

-20oC 81.5 ± 0.3 96 ± 0.4 108 ± 2.2

41

Table 2.5 Stability of cis-4-hydroxylomustine, trans-4-hydroxylomustine and lomustine in canine plasma.

Duration of Temperature % Recovery of Cis- % Recovery of % Recovery of Incubation 4- Trans-4- Lomustine hydroxylomustine hydroxylomustine (Mean ± SD, (Mean ± SD, n=3) (Mean ± SD, n=3) n=3) 24 hours Refrigerated 95 ± 2.3 76 ± 0.6 71 ± 0.8

-20oC 109 ± 2.3 89 ± 4.2 81 ± 1

-80oC 103 ± 8.3 88 ± 3.3 81 ± 1

3 days Refrigerated 80 ± 1.8 70 ± 1.3 37 ± 0.8

-20oC 119 ± 4.5 97 ± 2.7 72 ± 3.2

-80oC 111 ± 5.9 93 ± 1.5 81 ± 3.5

1 week Refrigerated 16 ± 1.1 26 ± 1.5 20 ± 1.1

-20oC 68 ± 0.9 79 ± 1.9 73 ± 3.8

-80oC 72 ± 2.4 79 ± 1.5 83 ± 9.3

2 week Refrigerated 7 ± 1.9 14 ± 1.7 7 ± 0.1

-20oC 77 ± 2.2 83 ± 0.2 62 ± 0.9

-80oC 88 ± 4.0 89 ± 2.1 69 ± 0.8

42

Table 2.6 In vitro concentrations of lomustine, trans-4-hydroxylomustine and cis-4- hydroxylomustine (ng/ml) in whole blood, plasma and red blood cells.

Drug Plasma mean ± SD Whole blood mean ± SD RBC Ke/p

Lomustine 287 ± 27 310 ± 14 37.49 0.12

Trans-4- 256 ± 41 273 ± 16 27.71 0.10 hydroxylomustine

Cis-4-hydroxylomustine 229 ± 12 254 ± 16 40.75 0.17 *All samples spiked at 500 ng/ml. Hematocrit for the blood sample was 0.38 (38%).

43

2.6 References

Bosanquet, A. (1985). Stability of solutions of antineoplastic agents during preparation and storage for invitro assays - general-considerations, the nitrosoureas and alkylating-agents. Cancer Chemotherapy and Pharmacology, 14(2), 83-95.

Chang, T., Weber, G., Crespi, C., & Waxman, D. (1993). Differential activation of cyclophosphamide and ifosphamide by cytochrome-P-450-2b and cytochrome-P450-3a in human liver-microsomes. Cancer Research, 53(23), 5629-5637.

Chatterji, D., Greene, R., & Gallelli, J. (1978). Mechanism of hydrolysis of halogenated nitrosoureas. Journal of Pharmaceutical Sciences, 67(11), 1527-1532.

Heal, J. M., Fox, P. A., Doukas, D., & Schein, P. S. (1978). Biological and biochemical properties of 2-hydroxyl metabolites of 1-2-chloroethyl)-3-cyclohexyl-1-nitrosourea. Cancer Research, 38(4), 1070-1074.

Hinderling, P. H. (1997). Red blood cells: A neglected compartment in pharmacokinetics and pharmacodynamics. Pharmacological Reviews, 49(3), 279-295.

Kastrissios, H., Chao, N., & Blaschke, T. (1996). Pharmacokinetics of high dose oral CCNU in bone marrow transplant patients. Cancer Chemotherapy and Pharmacology, 38(5), 425-430.

Kristal, O., Rassnick, K., Gliatto, J., Northrup, N., Chretin, J., Morrison-Collister, K., Moore, A. (2004). Hepatotoxicity associated with CCNU (lomustine) chemotherapy in dogs. Journal of Veterinary Internal Medicine, 18(1), 75-80.

Lee, F., Workman, P., Roberts, J., & Bleehen, N. (1985). Clinical pharmacokinetics of oral ccnu (lomustine). Cancer Chemotherapy and Pharmacology, 14(2), 125-131.

Lemoine, A., Lucas, C., & Ings, R. M. J. (1991). Metabolism of the chloroethylnitrosoureas. Xenobiotica, 21(6), 775-791.

Loftsson, T., & Fridriksdottir, H. (1990). Degradation of lomustine (ccnu) in aqueous-solutions. International Journal of Pharmaceutics, 62(2-3), 243-247.

Montgomery, J., James, R., McCaleb, G., & Johnston, T. (1967). The modes of decomposition of 1,3-bis(2-chloroethyl)-1-nitrosourea and related compounds. J Med Chem., 10(4), 668.

Moore, A. S., London, C. A., Wood, C. A., Williams, L. E., Cotter, S. M., L'Heureux, D. A., & Frimberger, A. E. (1999). Lomustine (CCNU) for the treatment of resistant lymphoma in dogs. Journal of Veterinary Internal Medicine / American College of Veterinary Internal Medicine, 13(5), 395-398.

44

Saba, C. F., Thamm, D. H., & Vail, D. M. (2007). Combination chemotherapy with L- asparaginase, lomustine, and prednisone for relapsed or refractory canine lymphoma. Journal of Veterinary Internal Medicine, 21(1), 127-132.

Sauerbrey, M. L., Mullins, M. N., Bannink, E. O., Van Dorp, T. E. Renate, Kaneene, J. B., & Obradovich, J. E. (2007). Lomustine and prednisone as a first-line treatment for dogs with multicentric lymphoma: 17 cases (2004–2005). Journal of the American Veterinary Medical Association, 230(12), 1866-1869.

Weinkam, R., & Lin, H. (1982). Chloroethylnitrosourea cancer chemotherapeutic-agents. Advances in Pharmacology and Chemotherapy, 19, 1-33.

Weinkam, R., Liu, T., & Lin, H. (1980). Protein mediated chemical-reactions of chloroethylnitrosoureas. Chemico-Biological Interactions, 31(2), 167-177.

Wheeler, G., Bowdon, B., & Struck, R. (1975). Carbamoylation of amino acid, peptides, and proteins by nitrosoureas. Cancer Res., 35(11), 2974.

Wheeler, G., Bowdon, B., Grimsley, J., & Lloyd, H. (1974). Interrelationships of some chemical, physicochemical, and biological activities of several 1-(2-haloethyl)-1-nitrosoureas. Cancer Res., 34(1), 194.

Wheeler, G., Johnston, T., Bowdon, B., McCaleb, G., Hill, D., & Montgomery, J. (1977). Comparison of the properties of metabolites of CCNU. Biochem Pharmacol, 26, 2331.

45

CHAPTER III

Cytotoxicity of lomustine and its trans-4-hydroxylomustine and cis-4-hydroxylomustine

metabolites in canine lymphoma cell lines and primary cells

3.1 Introduction

Lomustine is a highly lipophilic nitrosourea compound, which undergoes hydrolysis in vivo to form isomeric metabolites, primarily trans-4 and cis-4 in various animal species including humans (Hilton & Walker, 1975; Kastrissios et al., 1996; Lee et al., 1985). From our metabolism studies described in chapter 2, we conclude that this is true in dogs as well. It is reported that hydroxylation at the 4’-position of the cyclohexyl ring of lomustine, confers both isomers (cis-4 and trans-4) of the metabolite with enhanced alkylating activity and reduced toxic effects relative to lomustine, resulting in an approximately two-fold increase in the therapeutic index of each of the metabolites relative to the parent compound (Wheeler et al., 1977). This may suggest that the metabolites might have advantage over lomustine for clinical use.

Hepatotoxicity is known to be a side effect of lomustine treatment. Based on the published studies on this topic, it can be concluded that elevated ALT is a common occurrence following lomustine treatment (Hosoya et al., 2009; Kristal et al., 2004), however some dogs may show severe liver enzyme elevation and also clinical signs of liver injury necessitating discontinuation of lomustine (Hosoya et al., 2009). The mechanism by which lomustine causes hepatotoxicity is unclear.

46

The goal of this study was to compare the concentration and time-dependent cytotoxicity of lomustine and its trans-4 and cis-4 metabolites in canine cells. The study was done in canine lymphoma cell lines, PBMC from lymphoma dogs and canine primary hepatocytes. Hepatotoxicity is one of the side effects seen with lomustine and it is believed that the metabolites may have a better side effect profile as compared to parent lomustine (Wheeler at al., 1977). The primary aim of using PBMC from lymphoma dogs for cytotoxicity study is to determine if PBMC, which are primary cells, are more sensitive to lomustine and its metabolites as compared to the immortalized cell lines. We hypothesize that the PBMC may show greater cell killing with lomustine and its metabolites as compared to the canine lymphoma cell lines.

3.2 Materials and Methods

Cytotoxicity of lomustine and its metabolites in canine lymphoma cell lines and PBMC

The canine lymphoma cell lines 17-71 (Steplewski et al., 1987; Suter et al., 2005) and GL-

1 (Nakaichi et al., 1996) and PBMC obtained from lymphoma dogs were used for this assay.

Peripheral blood mononuclear cells (PBMC) isolation

Blood samples were collected from 3 lymphoma dogs that were brought to the veterinary teaching hospital. The dogs did not have any prior history of being treated for lymphoma. Four milliliters of whole blood were collected into one 5 ml heparinized tubes. Procedure for isolation of PBMC from lymphoma dogs was as described by Lavergne et al. (Lavergne et al., 2006). Tubes

47 were placed on ice immediately and PBMC were isolated using density centrifugation. Blood was kept on ice and cell isolation was begun within 30 minutes of venipuncture. Whole blood was diluted with two volumes of prewarmed (37ºC) HBSS with 5 mM EDTA. This was carefully overlaid on 5 ml of prewarmed (37ºC) Lymphocyte Separation Media (Cellgro). After centrifugation, the buffy coat was harvested and washed with 3–4 volumes of HBSS with 5 mM

EDTA. Lysing buffer (2 ml; 15.5 mM NH4Cl, 10 mM KHCO3, 1 mM EDTA, pH 7.4) was added to pelleted cells to lyse the residual erythrocytes. After washing with HBSS with 5 mM EDTA, cells were re-suspended in PBS and an aliquot was used to quantitate viable cells using Trypan blue dye exclusion.

Cytotoxicity assays

Cytotoxicity was evaluated after 48 hours by XTT cell proliferation assay and Annexin-PI assay (flow cytometry). The cell lines 17-71 and GL-1 were grown and maintained at 37°C in 5%

CO2, and cultures were passaged as necessary to maintain high cell viability (above 90%) in culture media DMEM supplemented with 10% FBS, 1% penicillin/streptomycin. The PBMC were maintained in culture media RPMI-1640 supplemented with 10% FBS, 1% penicillin/streptomycin. The cell viability was assessed by Trypan blue dye exclusion test. For the

XTT assay, the cells were plated in 96 well plates at a density of 20,000 cells/well. The cells were exposed to fresh media (DMEM supplemented with 10% FBS, 1% penicillin/streptomycin for the cell lines and RPMI-1640 supplemented with 10% FBS, 1% penicillin/streptomycin for PBMC) containing varying concentrations of lomustine, trans-4, or cis-4 metabolites (300, 1000, 3000 and

10,000 ng/ml). Staurosporine (1 µM for 24 hours) was used as positive control for apoptosis in

48

GL-1 cells and PBMC while PAC-1 (first procaspase-3 activating compound, 10 µM for 48 hours) was used as positive controls for apoptosis in 17-71 cells. The vehicle control was 0.1% methanol corresponding to the concentration of methanol present in each of the drug treated samples. Forty eight hours after drug exposure, the cytotoxicity was measured by using XTT Cell Proliferation

Assay Kit (Promega). For the Annexin/PI assay, 100,000 cells were plated/well in a 12 well plate.

The drug treatment protocol was similar to that in the XTT assay. In addition to the apoptosis controls, a necrosis control was also used which was Triton-X (0.03% for 10 minutes). At the end of 48 hours of incubation, the cells were stained using the Annexin V-FITC Apoptosis detection

Kit (BD biosciences) and analyzed by flow cytometry (BD Accuri C6). The experiments were done 3 separate times in the lymphoma cell lines with two technical replicates used for each assay.

The experiment was done on PBMC obtained from 3 lymphoma dogs (n=3) with two technical replicates for each assay.

Time dependent cytotoxicity of lomustine and its metabolites

In this experiment, the cytotoxicity of lomustine, trans-4 and cis-4 was measured in canine cell lines 17-71 and GL-1 and primary canine hepatocytes at 48, 96 and 144 hours after drug exposure in order to see if there was a time dependent cytotoxic effect of these drugs.

Isolation of canine hepatocytes

Hepatocytes were isolated from six dogs that were euthanized at the Champaign county animal control facility. Isolation of canine hepatocytes was done by the by the method of Spotorno

49 et al. (Spotorno et al., 2006) with few modifications. Liver samples (about 25 grams) from freshly euthanized dogs were obtained aseptically and immediately transported in PBS to the lab. About

10 grams of the sample were cut out from the center of the collected liver tissue and transferred to a petri dish containing PBS with 1% penicillin/streptomycin. The tissue was finely minced with scissors and washed a couple of times with PBS. The minced sample was transferred to a flask containing 100 ml of cold collagenase solution (Collagenase II, 300 units/mg, 500 ng/ml in HBSS-

HEPES buffer) and softly stirred for about 10 minutes. At the end of incubation, 10 ml of FBS and

50 ml of ice cold HBSS were added. The suspension was filtered through a nylon cell strainer (100 micron pore size) and the filtrate was centrifuged at 50 g for 5 minutes at 4ºC. The supernatant was discarded and the pellet washed twice with PBS with 1% penicillin/streptomycin and centrifuged twice at 150 g for 2 minutes each and the pellet suspended in 5 ml PBS. The cell suspension was layered over 15 ml cushion of 60% Percoll in PBS in a 50 ml polycarbonate tube and centrifuged at 70 g at 4ºC for 5 minutes. The supernatant was discarded and the pellet washed with PBS. Cell viability was determined by Trypan blue dye exclusion test. The cells were suspended in DMEM (supplemented with 10% FBS and 1% penicillin/streptomycin) at a density of 0.6 million/ml and were incubated at 37ºC for 48 hours to acclimatize and form a monolayer.

After 48 hours the media was replaced by hepatocyte maintenance media (Triangle research Labs,

NC) and were ready for cytotoxicity assays. No additional FBS or penicillin/streptomycin was added to the commercially purchased hepatocyte maintenance media. The maintenance media was changed every day.

50

Commercially purchased canine hepatocytes

In addition, fresh plated canine hepatocytes (96-well plates) were purchased from Triangle research labs (Research Park, NC). The canine hepatocytes were purchased 2 separate times and were harvested from 2 separate dogs (n=2). Similar to the manually isolated hepatocytes, hepatocyte maintenance media (Triangle research Labs, NC) was used in these cells (without additional FBS or penicillin/streptomycin) and was replaced every 48 hours.

Cytotoxicity assays

The cytotoxicity of lomustine, trans-4 and cis-4 in canine cell lines 17-71 and GL-1 and primary canine hepatocytes was measured at 48, 96 and 144 after exposure to the drugs. The experimental set up was similar to that mentioned above for the 48 hour cytotoxicity in the canine cells. Since the cytotoxicity had to be measured for longer time, the media was partially changed every 48 hours in order to avoid acid build-up. Half of the maintenance media was replaced every

48 hours by slowly aspirating the media from the top and replacing with the same volume of new media. At the end of 48, 96 and 144 hours, cytotoxicity was measured by XTT and Annexin/PI assay. For the Annexin/PI assay the hepatocytes were trypsinized as follows. After aspirating out all the media from the wells, about 20µl trypsin (0.25%) was added to each well and the plates were incubated at 37ºC for 1-2 minutes. After that time, DMEM containing 10% FBS was added to each well to inactivate the trypsin. The plates were returned to the incubator for 5 minutes. The cells were then washed with PBS and stained with Annexin V and PI (propidium iodide) for flow cytometry. Staurosporine (1 µM for 24 hours) was used as positive control for apoptosis. Two

51 technical replicates were used for each assay. For the lymphoma cell lines the experiments were done 3 separate times with two technical replicates used for each assay. The experiment was done in hepatocytes isolated from six dogs (n=6) and commercially purchased hepatocytes from two dogs (n=2). Two technical replicates were used for each assay.

Statistical analysis

Statistical analysis was done using SPSS software (IBM, NY). Descriptive statistics were produced for all continuous variables. Mean and standard deviation were calculated. Normality of data was assessed using the Shapiro-Wilks test. Paired t-tests were performed to evaluate differences between technical repeats of each drug and dose combination. Multiple one way

ANOVAs and Tukey’s post hoc test were performed to evaluate the following within and between group differences: Between drugs within each dose in each cell type at each time point, between doses (concentrations) within each drug in each cell type at each time, between time points within dose in cell types for the each drug. The p value was set at 0.05. For the canine lymphoma cell lines, two out of seventy two groups did not show normal distribution using Shapiro-Wilks test.

However when these groups were further evaluated for normality using kurtosis, skewness and Q-

Q plots, the data showed normal distribution. Hence a parametric test was used to reduce the probability of type II error.

Power analysis: Cytotoxicity data from two canine lymphoma cell lines were used to estimate sample size requirements for PBMC. Using the data from the 17-71 and GL-1 cell lines, based on a priori information of alpha=0.05, power of 0.8 and expected difference in mean of 3.77

52

(corresponding to the maximum standard deviation seen at any concentration), we would require

3 samples. The lowest concentration of 300 ng/ml was excluded from the power analysis as no statistically significant cell killing was seen in either of the cell lines with any of the drugs at that concentration.

3.3 Results

Cytotoxicity of lomustine and its metabolites in canine lymphoma cell lines and PBMC

The results obtained from XTT assay show that there was significant cell killing at 3000 and 10,000 ng/ml concentrations, as compared to control for lomustine, trans-4 and cis-4 metabolites in canine lymphoma cell line 17-71(Figure 3.1). In the canine lymphoma cell line GL-

1, in addition to the above concentrations, 1000 ng/ml also showed significant cytotoxicity compared to the vehicle control. The Annexin/PI assay also showed similar pattern of reduction in cell viability. After treating the cells with the highest drug concentration, 10,000 ng/ml, about 50% and 40% of the cells were still viable at 48 hours in 17-71 and GL-1 cells respectively (Figure 3.2).

The results obtained from both XTT and Annexin/PI assay do not show any significant difference in the cytotoxicity caused by lomustine, trans-4, and cis-4 in 17-71 and GL-1 cells at 48 hours.

Lomustine and its metabolites did not show any cell killing in the PBMC obtained from three lymphoma dogs at 48 hours after exposure to lomustine, trans-4 and cis-4 (Figure 3.3). Our results show that terminally differentiated lymphocytes in circulation may not be as sensitive to alkylating agents like lomustine. It is also possible that PBMC may require longer incubation time for the cytotoxicity of lomustine to become apparent.

53

Time dependent cytotoxicity of lomustine and its metabolites

All the three compounds showed a greater degree of cell killing at 96 and 144 hours as compared to 48 hours thus confirming the premise that cytotoxicity with these drugs is delayed in nature (Figures 3.4-3.7). However, no statistically significant difference was seen in the cytotoxicity among the three compounds. In the 17-71 cell, the 300 ng/ml samples did not show any cell killing or reduction in viability even at 144 hours after drug treatment. As evident from the results, the 3000 and 10,000 ng/ml concentration resulted in a comparable level of cell killing at 144 hours. Also, at 10,000 ng/ml at 96 hours, most of the cells were non-viable. Due to this

‘saturation’ in cell killing very little difference was seen in cytotoxicity at 144 hours as compared to 96 hours at 10,000 ng/ml. These results show that there is a delay in cell killing brought about by lomustine and its metabolites and hence measuring cell viability/apoptosis after short incubation time may not be the best indicator of the cytotoxic potential of the compounds.

The commercially purchased hepatocytes did not show any time or concentration dependent sensitivity to lomustine or its metabolites both by XTT and Annexin/PI assay (Figures

3.8 and 3.9). The same was true for the hepatocytes that were isolated from the six dogs euthanized at the animal control facility (Figures 3.10 and 3.11). This may be suggestive that the primary cells are not as responsive to lomustine or its metabolites as compared to the cell lines.

54

3.4 Discussion

As mentioned previously it has been reported that the major hydroxylated metabolites of lomustine may have enhanced alkylating activity and reduced toxic effects relative to parent lomustine (Wheeler et al., 1977). In this project we wanted to investigate, through in-vitro studies, if the major hydroxylated metabolites show potential as a more effective treatment option compared to lomustine. The metabolism studies discussed in chapter 2 show that trans-4 and cis-

4 are the major metabolites in dogs after oral treatment with lomustine, trans-4 being in greater proportion.

The drug concentrations for the cytotoxicity study were derived from the metabolism studies in dogs where we found that the maximum concentration of trans-4 was about 300 ng/ml and that for cis-4 was 50 ng/ml. Hence we used log incremental concentrations starting at 300 ng/ml up till 10,000 ng/ml as we did not know at what concentration the in vitro biological effects of the drugs would be seen. In our cytotoxicity study to evaluate the time dependent effect of lomustine and its metabolites, the culture media was partially changes every 48 hours. Stability studies in culture media (elaborated in chapter 2) showed that lomustine and its metabolites do not persist in the culture media and blood beyond 4 hours at 37ºC. Due to the short half-life of the drugs, changing the culture media every 48 hours would not be a confounding factor for comparing cytotoxicity at different time points.

We found no significant difference in the cytotoxicity caused by lomustine and its trans-4 and cis-4 metabolites in both the canine lymphoma cell lines 17-71 and GL-1. At 48 hours after

55 treatment with 10,000 ng/ml concentration of lomustine or its metabolites, 40-50 % of the cells were still viable. However, after 96 hours of incubation this number had dropped to about 20% indicating a time dependent effect of lomustine cytotoxicity. In the PBMCs from lymphoma dogs, none of the drugs showed any significant cell killing compared to the control at 48 hours after lomustine treatment. Interestingly, the in-vitro IC50 concentration for lomustine mentioned in literature varies from 50-250 μM (11,650-58,400 ng/ml) (Astier et al., 2005; Shinwari et al., 2008;

Wilkoff & Dulmadge, 1986). These studies have utilized different cell lines and also used different techniques of detecting the cell killing. However a systematic study of the time dependent nature of cell killing by lomustine has not been done before. Our finding may be suggestive that apoptosis of cells after lomustine administration is not an immediate event but rather a delayed one. This is also supported by the fact that peak neutropenia, which is a dose limiting side effect of lomustine treatment usually occurs 7-10 days after drug administration (Kristal et al., 2004; Moore et al.,

1999; Rassnick et al., 1999).

Another aim of this study was to compare the cytotoxicity of the lomustine and its metabolites in primary hepatocytes from normal dogs. Published studies on lomustine inducted hepatotoxicity show that hepatotoxicity after lomustine treatment is very common and may also result in serious irreversible hepatopathy. A recent study published in 2009 that looked at ALT elevation after lomustine administration as single agent chemotherapy (Hosoya et al., 2009) puts the overall prevalence of major ALT elevation (>5-fold upper reference limit) at 29% (32 of 109).

This developed most commonly after one to three doses of lomustine out of which 53% (17/32) of the cases occurred without preceding mild ALT elevation. Hence, to compare the cytotoxic potential of lomustine and its trans-4 and cis-4 metabolites in hepatocytes seemed promising as it

56 is thought that the metabolites may have lower side effects as compared to lomustine. In our study however, we were not able to verify this as none of the three compounds showed any apparent cell killing in the canine hepatocytes, the ones that were commercially purchased or isolated from euthanized dogs.

One reason for the non-significant cytotoxic response in hepatocytes could be that primary cells that are non-replicating like hepatocytes in vitro, may require much longer time to show apparent cell killing with alkylating compounds. Nitrosoureas are known to form adducts on the

DNA and primarily the adducts formed at the O6 position of guanine are thought to be toxic to the cells (Kaina, 2003; Ludlum, 1990; Sedgwick, 2004). During cell replication, this epigenetic modification may lead to double strand breaks which is known to be a potent inducer of apoptosis

(Kaina, 2003; Kaina et al., 2007). This may explain why we did not see any cell killing in the hepatocytes even six days after lomustine treatment and also in the PBMCs which are terminally differentiated.

From this study we conclude that there is no difference in the cytotoxicity of lomustine, trans-4 and cis-4 in two canine lymphoma cell lines 17-71 and GL-1. Longer incubation after drug exposure showed greater reduction in cell viability indicating a time dependent cell killing by all the three compounds. Also primary cells like PBMC and hepatocytes did not show any cell killing with lomustine or its metabolites and may require much longer time after drug exposure to show apparent cell killing.

57

3.5 Figures and Legends

Figure 3.1 Graphs showing the results of XTT cell proliferation assay in 17-71 (A) and GL-1 (B) cells.VC refers to vehicle control which is 0.1% methanol. PAC-1 (10 µM) and staurosporine (1 µM) were used as positive control for apoptosis for 17-71 and GL-1 cells, respectively. The results show mean and standard deviation of three independent experiments. * indicates statistical significant difference compared to vehicle control. (p=0.05)

(A)

* * * * * * *

(B)

* * * * * * * * *

58

Figure 3.2 Graphs showing the percent viable cells obtained from Annexin/PI assay in 17-71 (A) and GL-1 (B) cells. VC refers to vehicle control which is 0.1% methanol. PAC-1 (10 µM) and staurosporine (1 µM) were used as positive control for apoptosis for 17-71 and GL-1 cells, respectively. The results show mean and standard deviation of three independent experiments. * indicates statistical significant difference compared to vehicle control. (p=0.05)

(A)

* * * * * *

(B)

* * * * * * * * *

59

Figure 3.3 Graphs showing the results of XTT (A) and Annexin/PI (B) assays in PBMC isolated from three lymphoma dogs .VC refers to vehicle control which is 0.1% methanol. Staurosporine (1 µM) was used as positive control for apoptosis. The graphs show mean and standard deviation of results obtained from three dogs. No statistically significant difference was seen in any of the drug treated samples compared to vehicle control.

(A)

(B)

60

Figure 3.4 Graphs showing the result of XTT cell proliferation assay in 17-71 cells at 48 (A), 96 (B) and 144 (C) hours after treatment with lomustine, trans-4-hydroxylomustine and cis-4- hydroxylomustine. VC refers to vehicle control which is 0.1% methanol. PAC-1 (10 µM) was used as positive control for apoptosis. The results show mean and standard deviation of three independent experiments. * indicates significant difference compared to vehicle control. (p=0.05). Δ indicates significant difference compared to the same drug concentration at the immediately earlier time point.

(A)

* * * * * *

(B)

* * * Δ Δ Δ * * * Δ Δ Δ * * *

(C)

Δ * Δ Δ * * Δ Δ Δ * * * Δ Δ Δ * * *

61

Figure 3.5 Graphs showing the results of XTT cell proliferation assay in GL-1 cells at 48 (A), 96 (B) and 144 (C) hours after treatment with lomustine, trans-4-hydroxylomustine and cis-4- hydroxylomustine. VC refers to vehicle control which is 0.1% methanol. Staurosporine (1 µM) was used as positive control for apoptosis. The results show mean and standard deviation of three independent experiments. * indicates significant difference compared to vehicle control. (p=0.05). Δ indicates significant difference compared to the same drug concentration at the immediately earlier time point.

(A)

* * * * * * * * *

(B)

* * * Δ Δ Δ * * * Δ Δ Δ * * *

(C)

* * * Δ Δ Δ * Δ Δ Δ * * * * *

62

Figure 3.6 Graphs showing the results of Annexin/PI assay assay in 17-71 cells at 48 (A), 96 (B) and 144 (C) hours after treatment with lomustine, trans-4-hydroxylomustine and cis-4- hydroxylomustine. VC refers to vehicle control which is 0.1% methanol. PAC-1 (10 µM) was used as positive control for apoptosis. The results show mean and standard deviation of three independent experiments. * indicates significant difference compared to vehicle control. (p=0.05). Δ indicates significant difference compared to the same drug concentration at the immediately earlier time point.

(A)

* * * * * *

(B)

Δ Δ Δ * * * Δ Δ Δ * * * Δ Δ Δ * * *

(C)

Δ Δ Δ * * * Δ Δ Δ * * * * * *

63

Figure 3.7 Graphs showing the percent viable cells obtained from Annexin/PI assay in GL-1 cells at 48 (A), 96 (B) and 144 (C) hours after treatment with lomustine, trans-4-hydroxylomustine and cis-4-hydroxylomustine. VC refers to vehicle control which is 0.1% methanol. Staurosporine (1 µM) was used as positive control for apoptosis. The results show mean and standard deviation of three independent experiments. * indicates significant difference compared to vehicle control. (p=0.05). Δ indicates significant difference compared to the same drug concentration at the immediately earlier time point.

(A)

* * * * * * * * *

(B)

Δ Δ Δ * * * Δ Δ Δ * * Δ Δ Δ * * * *

(C)

* * * * * * Δ Δ Δ * * * * * *

64

Figure 3.8 Graphs showing the results of XTT assay in commercially purchased canine hepatocytes at 48 (A), 96 (B) and 144 (C) hours after treatment with lomustine, trans-4- hydroxylomustine and cis-4-hydroxylomustine. VC refers to vehicle control which is 0.1% methanol. Staurosporine (1 µM) was used as positive control for apoptosis. The graphs show mean and standard deviation of results obtained from two dogs. No statistically significant difference was seen in any of the drug treated samples compared to vehicle control.

(A)

(B)

(C)

65

Figure 3.9 Graphs showing the percent viable cells obtained from the Annexin/PI assay in commercially purchased canine hepatocytes at 48 (A), 96 (B) and 144 (C) hours after treatment with lomustine, trans-4-hydroxylomustine and cis-4-hydroxylomustine. VC refers to vehicle control which is 0.1% methanol. Staurosporine (1 µM) was used as positive control for apoptosis. The graphs show mean and standard deviation of results obtained from two dogs. No statistically significant difference was seen in any of the drug treated samples compared to vehicle control.

(A)

(B)

(C)

66

Figure 3.10 Graphs showing the results of XTT assay in hepatocytes from six dogs at 48 (A), 96 (B) and 144 (C) hours after treatment with lomustine, trans-4-hydroxylomustine and cis-4- hydroxylomustine. VC refers to vehicle control which is 0.1% methanol. Staurosporine (1 µM) was used as positive control for apoptosis. The graphs show mean and standard deviation of results obtained from six dogs. No statistically significant difference was seen in any of the drug treated samples compared to vehicle control.

(A)

(B)

(C)

67

Figure 3.11 Graphs showing the percent viable cells obtained from the Annexin/PI assay in hepatocytes from six dogs at 48 (A), 96 (B) and 144 (C) hours after treatment with lomustine, trans-4-hydroxylomustine and cis-4-hydroxylomustine. VC refers to vehicle control which is 0.1% methanol. Staurosporine (1 µM) was used as positive control for apoptosis. The graphs show mean and standard deviation of results obtained from six dogs. No statistically significant difference was seen in any of the drug treated samples compared to vehicle control.

(A)

(B)

(C)

68

3.6 References

Astier, A., Ferrer, M., & Gibaud, S. (2005). Enhancement by 1-(2-chloroethyl)-3-cyclohexyl-1- nitrosourea (CCNU) of the cytotoxic properties of 1-β-d-arabinofuranosylcytosine (ara-C) on human promyelocytic leukemia cell line HL-60 . EJHP Science, 11(3), 62.

Hilton, J., & Walker, M. (1975). Hydroxylation of 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea. Biochemical Pharmacology, 24(23), 2153-2158.

Hosoya, K., Lord, L. K., Lara-Garcia, A., Kisseberth, W. C., London, C. A., & Couto, C. G. (2009). Prevalence of elevated alanine transaminase activity in dogs treated with CCNU (lomustine)*. Veterinary and Comparative Oncology, 7(4), 244-255.

Kaina, B. (2003). DNA damage-triggered apoptosis: Critical role of DNA repair, double-strand breaks, cell proliferation and signaling. Biochemical Pharmacology, 66(8), 1547-1554.

Kaina, B., Christmann, M., Naumann, S., & Roos, W. P. (2007). MGMT: Key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents. DNA Repair, 6(8), 1079-1099.

Kastrissios, H., Chao, N., & Blaschke, T. (1996). Pharmacokinetics of high dose oral CCNU in bone marrow transplant patients. Cancer Chemotherapy and Pharmacology, 38(5), 425-430.

Kristal, O., Rassnick, K., Gliatto, J., Northrup, N., Chretin, J., Morrison-Collister, K., & Moore, A. (2004). Hepatotoxicity associated with CCNU (lomustine) chemotherapy in dogs. Journal of Veterinary Internal Medicine, 18(1), 75-80.

Lavergne, S., Kurian, J., Bajad, S., Maki, J., Yoder, A., Guzinski, M., & Trepanier, L. (2006). Roles of endogenous ascorbate and glutathione in the cellular reduction and cytotoxicity of sulfamethoxazole-nitroso. Toxicology, 222(1-2), 25-36.

Lee, F., Workman, P., Roberts, J., & Bleehen, N. (1985). Clinical pharmacokinetics of oral ccnu (lomustine). Cancer Chemotherapy and Pharmacology, 14(2), 125-131.

Ludlum, D. (1990). Dna alkylation by the haloethylnitrosoureas - nature of modifications produced and their enzymatic repair or removal. Mutation Research, 233(1-2), 117-126.

Moore, A. S., London, C. A., Wood, C. A., Williams, L. E., Cotter, S. M., L'Heureux, D. A., & Frimberger, A. E. (1999). Lomustine (CCNU) for the treatment of resistant lymphoma in dogs. Journal of Veterinary Internal Medicine / American College of Veterinary Internal Medicine, 13(5), 395-398.

Nakaichi, M., Taura, Y., Kanki, M., Mamba, K., Momoi, Y., Tsujimoto, H., & Nakama, S., (1996). Establishment and characterization of a new canine B-cell leukemia cell line. The Journal of veterinary medical science 58: 469–71.

69

Rassnick, K., Moore, A., Williams, L., London, C., Kintzer, P., Engler, S., & Cotter, S. (1999). Treatment of canine mast cell tumors with CCNU (lomustine). Journal of Veterinary Internal Medicine, 13(6), 601-605.

Sedgwick, B. (2004). Repairing DNA-methylation damage. Nature Reviews Molecular Cell Biology, 5(2), 148-157.

Shinwari, Z., Manogaran, P. S., Alrokayan, S. A., Al-Hussein, K. A., & Aboussekhra, A. (2008). Vincristine and lomustine induce apoptosis and p21(WAF1) up-regulation in and normal human epithelial and fibroblast cells. Journal of Neuro-Oncology, 87(2), 123-132.

Spotorno, V. G., Hidalgo, A., Barbich, M., Lorenti, A., & Zabal, O. (2006). Culture of bovine hepatocytes: A non-perfusion technique for cell isolation. Cytotechnology, 51(2), 51-56.

Steplewski, Z., Jeglum, K.A., Rosales, C., & Weintraub, N., (1987). Canine lymphoma-associated antigens defined by murine monoclonal antibodies. Cancer Immunology, Immunotherapy, 24(3):197-201.

Suter, S.E., Chein, M.B., Messling, V., Yip, B., Cattaneo, R., Vernau, W., Bruce R. Madewell, B.R., & London, C.A., (2005). In vitro canine distemper virus infection of canine lymphoid cells: A prelude to oncolytic therapy for lymphoma. Clinical Cancer Research. 11(4):1579-87.

Wheeler, G., Johnston, T., Bowdon, B., McCaleb, G., Hill, D., & Montgomery, J. (1977). Comparison of the properties of metabolites of CCNU. Biochemical Pharmacology, 26, 2331.

Wilkoff, L. J., & Dulmadge, E. A. (1986). Sensitivity of proliferating cultured murine pancreatic tumor cells to selected antitumor agents. Journal of the National Cancer Institute, 77(5), 1163- 1169.

70

CHAPTER IV

Alkylating and carbamylating effects of lomustine, trans-4-hydroxylomustine and cis-4-

hydroxylomustine in canine lymphoma cell lines and hepatocytes

4.1 Introduction

It is known that nitrosoureas decompose at physiological conditions to yield alkylating and carbamylating intermediates (Montgomery et al., 1967), however the relative contribution of these events to the toxicity and therapeutic effects are unclear (Wheeler et al., 1974). Decomposition of lomustine in aqueous produces chloroethyl carbonium ion which is an alkylating intermediate and also cyclohexyl isocyanate which is a carbamylating agent (Colvin et al., 1976; Weinkam et al,

1980; Wheeler et al., 1975).

Alkylating agents bind to the DNA at various sites based on the relative electrophilicity and nucleophilicity. These compounds are electrophilic in nature and attack the nucleophilic sites on the DNA to form an alkylated product (Ludlum, 1990). The N7 position of guanine is the most reactive site (Boysen et al., 2009; Ludlum, 1990) with all alkylating agents and hence about 70-

90% of the adducts are formed at this position (Sedgwick, 2004; Rinne et al., 2005). However the relative mutagenicity and carcinogenicity of alkylating agents correlate with the reagents ability to bind to oxygen sites (Beranek, 1990; Singer, 1985). Chloroethylating compounds like lomustine form adducts at the O6 position of guanine (Sedgwick, 2004). If not repaired, these adducts form

71

DNA interstrand cross links (Ludlum, 1990) which may lead to double strand breaks and ultimately, apoptosis of the cell (Kaina, 2003).

In our cytotoxicity study (elaborated in chapter 3), lomustine and its metabolites showed similar degree of cell killing in the canine cell lines 17-71 and GL-1 and hepatocytes. As alkylation is thought to be the main reason for the cytotoxic effects of these drugs (Ludlum, 1990), we wanted to investigate if the compounds have similar pattern of alkylation. It is believed that the concentration of an internal dosimeter such as DNA adducts is a more accurate measure of the amount of mutagen reaching the target site than the applied dose (Beranek, 1990). In a recent study published, it was shown that N7 hydroxyethylguanine and O6 hydroxyethyldeoxyguanosine are some of the main DNA alkylation products formed after exposure to chloroethylating agents

(Bodell, 2009).

It is believed that the dominant role of carbamylating activity is in determining the toxicity of compounds which may be manifested as side effects (Wheeler et al., 1974). The isocyanate intermediates formed by the breakdown of lomustine react with the lysine residues in protein leading to post translational modification of proteins (Lemoine et al., 1991). This could result in partial or complete loss of protein function (Jaisson et al., 2011). The amount of isocyanates produced can vary depending on the chemical structure of the compound which may result in variable degree of carbamylation of intracellular proteins (Schmall et al., 1973; Wheeler et al.,

1974). Carbamylation of proteins is believed to contribute to the side effects seen with lomustine

(Wheeler et al., 1974).

72

It is reported that the cis-4 and trans-4 metabolites of lomustine may have reduced toxic effects relative to lomustine (Wheeler et al., 1977) and therefore the metabolites might have advantage over lomustine for clinical use (Heal et al., 1978). The aim of this study was to compare the alkylation and carbamylation potential of lomustine and its trans-4 and cis-4 metabolites in canine lymphoma cell lines and hepatocytes. The O6 hydroxyethyldeoxyguanosine and N7 hydroxyethylguanine adducts was measured by LC/MS/MS and the carbamylation was assessed by ELISA.

4.2 Materials and Methods

DNA adduct detection assay

The O6 hydroxyethyldeoxyguanosine and N7 hydroxyethylguanine adducts were measured in canine lymphoma cell lines 17-71 and GL-1 and primary canine hepatocytes (isolated from six dogs that were euthanized at the Champaign county animal control facility) after exposure to lomustine, trans-4 and cis-4 by the LC/MS/MS method as mentioned above. The lymphoma cell lines were grown and maintained at 37°C in 5% CO2, and cultures were passaged as necessary to maintain high cell viability (above 90%) in culture media DMEM supplemented with 10% FBS,

1% penicillin/streptomycin.

73

Isolation of canine hepatocytes

Hepatocytes were isolated from six dogs that were euthanized at the Champaign county animal control facility. Isolation of canine hepatocytes was done by the by the method of Spotorno et al. (Spotorno et al., 2006) with few modifications. Liver samples (about 25 grams) from freshly euthanized dogs were obtained aseptically and immediately transported in PBS to the lab. About

10 grams of the sample were cut out from the center of the collected liver tissue and transferred to a petri dish containing PBS with 1% penicillin/streptomycin. The tissue was finely minced with scissors and washed a couple of times with PBS. The minced sample was transferred to a flask containing 100 ml of cold collagenase solution (Collagenase II, 300 units/mg, 500 ng/ml in HBSS-

HEPES buffer) and softly stirred for about 10 minutes. At the end of incubation, 10 ml of FBS and

50 ml of ice cold HBSS were added. The suspension was filtered through a nylon cell strainer (100 micron pore size) and the filtrate was centrifuged at 50 g for 5 minutes at 4ºC. The supernatant was discarded and the pellet washed twice with PBS with 1% penicillin/streptomycin and centrifuged twice at 150 g for 2 minutes each and the pellet suspended in 5 ml PBS. The cell suspension was layered over 15 ml cushion of 60% Percoll in PBS in a 50 ml polycarbonate tube and centrifuged at 70 g at 4ºC for 5 minutes. The supernatant was discarded and the pellet washed with PBS. Cell viability was determined by Trypan blue dye exclusion test. The cells were suspended in DMEM (supplemented with 10% FBS and 1% penicillin/streptomycin) at a density of 0.6 million/ml and were incubated at 37ºC for 48 hours to acclimatize and form a monolayer.

After 48 hours the media was replaced by hepatocyte maintenance media (Triangle research Labs,

NC). No additional FBS or penicillin/streptomycin was added to the commercially purchased hepatocyte maintenance media. The maintenance media was changed every day.

74

Sample preparation

About 106 cells were used per sample. The cells were exposed to fresh media (hepatocyte maintenance media for the canine hepatocytes and DMEM supplemented with 10% FBS and 1% penicillin/streptomycin for the canine cell lines 17-71 and GL-1) containing varying concentrations of lomustine, trans-4 or cis-4 metabolites (300, 1000, 3000, 10,000 and 50,000 ng/ml) for 4 hours. This time point was based on a published study that looked at DNA alkylation products after exposure to lomustine and adducts were measured between 1 and 6 hours. The results showed that the adducts were formed rapidly and were detected at 1 hour after drug exposure (Bodell, 2009). In our initial experimentation, the 1 hour samples showed very low adduct level and hence we chose to detect the adducts at 4 hours. At the end of 4 hours, the DNA was isolated using Qiagen blood mini kit (Qiagen). The extracted DNA was lyophilized and concentrated to 100 µg. The DNA was enzymatically digested by a method described by Singer et al (Singer et al., 1978). The DNA sample digestion was carried out as follows: per 50 μl DNA, 10

μl of 0.5 M Tris (pH 7.3), 2 μl of 1 M MgCl2, 10 μl DNAse I (5 mg/ml, 2303 units/mg,

Worthington), 10 μl of snake venom phosphatase (5 mg/ml, 36 units/mg, Worthington) and 4 μl of bacterial alkaline phosphatase (5 mg/ml, 35 units/mg, Worthington) was added and final volume adjusted to 150 μl with deionized water. The mixture was incubated overnight at 37ºC and then heated to 100ºC for 90 seconds to inactivate the enzymes. The samples were briefly centrifuged to sediment the proteins. The supernatant was collected and analyzed by LC/MS/MS.

Preliminary experiments showed that N7 adduct was detected at the concentration of 10,000 ng/ml and above while the mutagenic adduct O6 hydroxyethyldeoxyguanosine was only detected

75 at 50,000 ng/ml drug concentration. Hence 10,000 ng/ml and 50,000 ng/ml concentrations were used to compare the adducts formed by the three drugs. For the cell lines, the experiment was done two separate times, with two technical replicates each. For the canine hepatocytes, the experiment was done once with duplicate samples.

LC/MS/MS method for detection of DNA adducts

An LC/MS/MS method was developed to quantify the O6 hydroxyethyldeoxyguanosine and N7 hydroxyethylguanine adducts in the DNA samples.

Standards preparation

Pure standards of O6 hydroxyethyldeoxyguanosine and N7 hydroxyethylguanine were purchased from Toronto research company (Toronto, Canada). Deuterated O6 methylguanine (D3-

O6 methylguanine) was used as internal standard (Sigma Aldrich). Standards were dissolved in methanol to prepare stock solutions. Standard curve range was 0.1-10 ng for each compound and

50 ng of internal standard was added to each sample. The final volume was adjusted to 150 µl with water.

LC/MS/MS conditions

Samples were analyzed with the QTRAP 5500 LC/MS/MS system (AB Sciex, Foster City,

CA) with the 1200series HPLC system (Agilent Technologies, Santa Clara, CA) including a

76 degasser, an autosampler, and a binary pump. The LC separation was performed on an Agilent

SB-Aq column (4.6 mm × 50 mm, 5µ) (Santa Clara, CA) with mobile phase A (0.1% formic acid in water) and mobile phase B (0.1% formic acid in acetonitrile). The flow-rate was 0.35 ml/min. The linear gradient was as follows: 0-1 min, 100% A; 8-11 min, 25% A; 12-18 min, 100%

A.

The autosampler was set at 5ºC and the injection volume is 5 µL. Positive mass spectrometry was achieved with electrospray ionization (ESI). The ion spray voltage was 5500

V. The source temperature was 400 ºC. The curtain gas, ion source gas 1, and ion source gas 2 were 32, 65, and 50, respectively. Multiple reaction monitoring (MRM) was used to quantify O6- hydroxyethyldeoxyguanosine (m/z 312.2 à m/z 110.1), N7-hydroxyethylguanine (m/z 196.1 à m/z

110.1), and the internal standard deuterated O6-methylguanine (m/z 169.1 à m/z 70.1) (Figure 4.1).

Carbamylation assay

Carbamylation was measured in primary hepatocytes harvested from six dogs euthanized at the Champaign county animal control facility (hepatocyte isolation procedure as mentioned above) and canine cell lines 17-71 and GL-1 after treatment with lomustine and its metabolites using Oxyselect™ carbamylation ELISA kit. The quantity of carbamylated lysine (CBL) in protein samples is determined by comparing its absorbance with that of a known CBL-BSA standard curve.

77

About 106 cells were used per sample. The cells were exposed to fresh media (hepatocyte maintenance media for the canine hepatocytes and DMEM supplemented with 10% FBS and 1% penicillin/spreptomycin for the canine cell lines 17-71 and GL-1) containing varying concentrations of lomustine, trans-4 or cis-4 metabolites (300, 1000, 3000 and 10,000 ng/ml).

Carbamylation was measured at 4 hours after exposure to drugs as per the ELISA kit instructions.

Carbamylation is a rapid process occurring within a few minutes and previously published studies have measured carbamylation at similar time points (Heal et al., 1978; Wheeler et al., 1974). Initial experimentation did not show any carbamylation at 300, 1000 and 3000 ng/ml drug concentration.

Thus 10,000 ng/ml concentration was used to compare the carbamylation effects of the three drugs.

For the cell lines, the experiment was done two separate times, with two technical replicates each.

For the canine hepatocytes, the experiment was done once with duplicate samples.

Statistical analysis

Statistical analysis was done using SPSS (IBM, NY). Descriptive statistics were produced for all continuous variables. Mean and standard deviation were calculated. Normality of data was assessed using the Shapiro-Wilks test. Paired t-tests were performed to evaluate differences between technical repeats of each drug. One way ANOVA and Tukey’s post hoc test were performed to evaluate the between drug differences. The p value was set at 0.05.

78

4.3 Results

There is no significant difference in the levels of adducts formed by lomustine, trans-4 or cis-4. This can be correlated with the similar cytotoxicity observed in the cells after treatment with the three compounds. The N7 hydroxyethylguanine adduct was detected at similar level in 17-71,

GL-1 and the hepatocytes from 6 dogs after exposure to lomustine, trans-4 and cis-4 at 10,000 ng/ml and 50,000 ng/ml concentration (Figure 4.2). However the mutagenic adduct O6 hydroxyethyldeoxyguanosine was detected at a significantly lower level (2-3 fold) in the hepatocytes as compared to the cell lines when treated with the three drugs at 50,000 ng/ml concentration (Figure 4.3).

In the canine cell lines 17-71 and GL-1 and in the canine hepatocyte samples, lomustine caused significantly greater carbamylation (about 1.5 fold) as compared to its metabolites (Figure

4.4). Both the metabolites showed similar carbamylation levels. In all the six canine hepatocyte samples tested, the carbamylation caused by all the three drugs was 1.5-2 fold greater than that seen in 17-71 and GL-1 cells. Among the six dog hepatocyte samples, two dogs showed about 2 fold higher level of carbamylation compared to the others with lomustine, trans-4 and cis-4).

4.4 Discussion

In our study we found that there is no significant difference in the N7 hydroxyethylguanine and O6 hydroxyethyldeoxyguanosine adduct level formed by lomustine, trans-4 and cis-4 which correlates with the similar cytotoxicity of these compounds. The mutagenic O6 adduct was higher

79 in the canine cell lines 17-71 and GL-1 as compared to the hepatocytes which may indicate either lower adduct formation or rapid repair. Rapid repair may be a possibility since all the canine hepatocyte samples showed greater MGMT level (elaborated in chapter 5). It can be speculated that as MGMT is a DNA repair enzyme, an enhanced repair of O6 adducts could contribute to the reduced sensitivity of hepatocytes to the alkylating action of lomustine and its metabolites.

It should however be noted that we have only detected the levels of two individual adducts after drug exposure. The cell killing action of nitrosoureas may be orchestrated by the formation and cumulative action of multiple adducts that these drugs are capable of forming. Also, in case of chloroethylating agents like lomustine, cross linking of DNA is known to be harmful lesion that can lead to double stand breaks and ultimately apoptosis (Lemoine et al., 1991). This cross linking of DNA is initiated by the formation of O6 chloroethyldeoxyguanosine adduct. This adduct is highly unstable and cross links with the cytosine on the opposite stand forming the cytotoxic cross links (Lemoine et al., 1991; Ludlum, 1990). Thus in our alkylation study, measuring the two adducts can act only as a molecular dosimeter of therapeutic response and not a measure of the full extent of alkylation caused by the drugs.

Carbamylation was measured in the canine lymphoma cell lines and hepatocytes by an

ELISA kit. The assay measures the amount of carbamylated lysine (CBL) in the samples. Our results show that lomustine causes greater carbamylation as compared to its metabolites in canine cell lines and hepatocytes. All the three compounds caused greater carbamylation in hepatocytes as compared to the canine cell lines. Also hepatocyte samples from two dogs showed greater carbamylation following treatment with lomustine and its metabolites compared to the others. But

80 it should be noted that all of the dog hepatocyte samples showed no cytotoxicity to lomustine or its metabolites up to six days of incubation (elaborated in chapter 3). Hence it is unlikely that greater carbamylation in the hepatocytes in the two dogs would lead to cell death in our experiments, although it may be inhibiting various proteins in these cells. The same would also hold true for the cell lines 17-71 and GL-1 where we see lower carbamylation but greater cytotoxicity after treatment with the drugs.

Even though the hepatocyte samples showed greater carbamylation than the cell lines it is not known if this is of clinical relevance with regards to hepatotoxicity caused by lomustine since carbamylation levels have not been measured in dogs that show hepatotoxicity after lomustine treatment. There is no clear correlation between the carbamylating potential of the isocyanates formed from nitrosourea and the cytotoxicity as nitrosoureas that have low carbamylating activity retain their antitumor activity (Lemoine et al., 1991). However carbamylation of proteins may interfere with the proper functioning or replication of the cells which may not necessarily result in cell death. It has also been shown that nitrosoureas which form isocyanates are particularly able to inhibit the polymerization of purified brain tubulin in a dose dependent manner (Brodie et al.,

1980). In a study where rats were given lomustine by oral gavage at 50 mg/kg dose, and sacrificed at different time points, the hepatocytes remained normal at early time points although the bile canaliculi became dilated and contained bile thrombi (Kretschmer et al, 1987). In another study in rats that were administered lomustine at 20 mg/kg, hepatocytes were not markedly altered with normal nuclei and cell organelles. The main alteration of the parenchymal cells occurred after the

6th day with modification of the cytoskeletal components and reduction in number of

(Ducastelle et al., 1988). It is suggested that the isocyanates carbamylate tubulin in hepatocytes

81 leading to mitotic spindle inhibition and G2 arrest or may cause disruption of secretory functions such as bile secretion which in turn results in cholestasis and secondary hepatic injury

(Ducastelle et al., 1988; Laquerriere et al., 1991). Thus carbamylation of proteins may not necessarily manifest as immediate cell killing.

In this study we have shown that lomusine and its metabolites form similar levels of the

DNA adduct N7 hydroxyethylguanine and O6 hydroxyethyldeoxyguanosine which corresponds to the similar cytotoxicity of these compounds. In terms of carbamylation, lomustine showed greater carbamylation in the canine hepatocytes and lymphoma cell lines. Thus if carbamylation of proteins is indeed one of the reasons for the side effects of lomustine, then using the metabolites instead may be a better option in terms of side effects.

82

4.5 Figures and Legends

Figure 4.1 LC/MS/MS Multiple Reaction Monitoring (MRM) analysis for O6 hydroxyethyldeoxyguanosine, N7 hydroxyethylguanine and D3-O6 methylguanine

D3- O6 methylguanine – internal standard

O6 hydroxyethyldeoxyguanosine

N7 hydroxyethylguanine

83

Figure 4.2 Graphs showing the N7 hydroxyethylguanine adduct formed after treatment with lomustine, trans-4-hydroxylomustine and cis-4-hydroxylomustine metabolites at 10,000 ng/ml (A) and 50,000 ng/ml (B) concentration in 17-71 and GL-1 cells and hepatocytes from 6 dogs (n=6). No statistically significant difference was seen in the N7 hydroxyethylguanine levels in 17-71 and GL-1 cells and canine hepatocytes with lomustine, trans-4-hydroxylomustine and cis-4- hydroxylomustine.

(A)

(B)

84

Figure 4.3 Graph showing the O6 hydroxyethyldeoxyguanosine adduct formed after treatment with lomustine, trans-4-hydroxylomustine and cis-4-hydroxylomustine metabolites at 50,000 ng/ml concentration in 17-71 and GL-1 cells and hepatocytes from 6 dogs (n=6). No statistically significant difference was seen in the O6 hydroxyethyldeoxyguanosine levels formed by lomustine, trans-4-hydroxylomustine and cis-4-hydroxylomustine in the same cell type.* denotes statistical significance as compared to the 17-71 and GL-1 cells. p=0.05

* * *

85

Figure 4.4 Graphs showing the carbamylation induced by lomustine, trans-4-hydroxylomustine and cis-4-hydroxylomustine at 10,000 ng/ml concentration in 17-71 and GL-1 cells (A) and hepatocytes from 6 dogs (B). The data represents the mean and standard deviation. * indicates statistical significance compared to trans-4-hydroxylomustine and cis-4-hydroxylomustine. p=0.05

(A)

* *

(B)

* * * * * *

86

4.6 References

Beranek, D. (1990). Distribution of methyl and ethyl adducts following alkylation with monofunctional alkylating-agents. Mutation Research, 231(1), 11-30.

Bodell, W. J. (2009). DNA alkylation products formed by 1-(2-chloroethyl)-1-nitrosourea as molecular dosimeters of therapeutic response. Journal of Neuro-Oncology, 91(3), 257-264.

Boysen, G., Pachkowski, B. F., Nakamura, J., & Swenberg, J. A. (2009). The formation and biological significance of N7-guanine adducts. Mutation Research-Genetic Toxicology and Environmental Mutagenesis, 678(2), 76-94.

Brodie, A., Babson, J., & Reed, D. (1980). Inhibition of tubulin polymerization by nitrosourea- derived isocyanates. Biochemical Pharmacology, 29(4), 652-654.

Colvin, M., Brundrett, R., Cowens, W., Jardine, I., & Ludlum, D. (1976). A chemical basis for the antitumor activity of chloroethylnitrosoureas. Biochem Pharmacol, 25(6), 695.

Ducastelle, T., Raguenezviotte, G., Fouinfortunet, H., Matysiak, M., Hemet, J., & Fillastre, J. (1988). The hepatotoxicity of 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (ccnu) in rats - ultrastructural evidence of a delayed microtubular toxicity. Cancer Chemotherapy and Pharmacology, 22(2), 153-162.

Heal, J. M., Fox, P. A., Doukas, D., & Schein, P. S. (1978). Biological and biochemical properties of 2-hydroxyl metabolites of 1-2-chloroethyl)-3-cyclohexyl-1-nitrosourea. Cancer Research, 38(4), 1070-1074.

Jaisson, S., Pietrement, C., & Gillery, P. (2011). Carbamylation-derived products: Bioactive compounds and potential biomarkers in chronic renal failure and atherosclerosis. Clinical Chemistry, 57(11), 1499-1505.

Kaina, B. (2003). DNA damage-triggered apoptosis: Critical role of DNA repair, double-strand breaks, cell proliferation and signaling. Biochemical Pharmacology, 66(8), 1547-1554.

Kretschmer, N., Boor, P., Elazhary, R., Ahmed, A., & Reynolds, E. (1987). Studies on the mechanism of 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU)-induced hepatotoxicity .3. ultrastructural characterization of bile-duct injury. Cancer Chemotherapy and Pharmacology, 19(2), 109-117.

Laquerriere, A., Raguenez-Viotte, G., Paraire, M., Bizzari, J.P., Paresy, M., Fillastre, J.P., Hemet, J. (1991). Nitrosoureas lomustine, and induced hepatotoxic perturbations in rats: biochemical, morphological and flow cytometry studies. European Journal of Cancer, 27(5):630-8.

87

Lemoine, A., Lucas, C., & Ings, R. M. J. (1991). Metabolism of the chloroethylnitrosoureas. Xenobiotica, 21(6), 775-791.

Ludlum, D. (1990). Dna alkylation by the haloethylnitrosoureas - nature of modifications produced and their enzymatic repair or removal. Mutation Research, 233(1-2), 117-126.

Montgomery, J., James, R., McCaleb, G., & Johnston, T. (1967). The modes of decomposition of 1,3-bis(2-chloroethyl)-1-nitrosourea and related compounds. J Med Chem., 10(4), 668.

Rinne, M. L., He, Y., Pachkowski, B. F., Nakamura, J., & Kelley, M. R. (2005). N-methylpurine DNA glycosylase overexpression increases alkylation sensitivity by rapidly removing non-toxic 7-methylguanine adducts. Nucleic Acids Research, 33(9), 2859-2867.

Schmall, B., Cheng, C., Fujimura, S., Gersten, N., Grunberger, D., & Weinstein, I. (1973). Modification of proteins by 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (NSC 79037) in vitro. Cancer Res, 33, 1921.

Sedgwick, B. (2004). Repairing DNA-methylation damage. Nature Reviews Molecular Cell Biology, 5(2), 148-157.

Singer, B. (1985). In vivo formation and persistence of modified nucleosides resulting from alkylating agents. Environ Health Perspect., 62, 41.

Singer, B., Bodell, W., Cleaver, J., Thomas, G., Rajewsky, M., & Thon, W. (1978). Oxygens in DNA are main targets for ethylnitrosourea in normal and xeroderma pigmentosum fibroblasts and fetal rat-brain cells. Nature, 276(5683), 85-88.

Weinkam, R., Liu, T., & Lin, H. (1980). Protein mediated chemical-reactions of chloroethylnitrosoureas. Chemico-Biological Interactions, 31(2), 167-177.

Wheeler, G., Bowdon, B., & Struck, R. (1975). Carbamoylation of amino acid, peptides, and proteins by nitrosoureas. Cancer Res., 35(11), 2974.

Wheeler, G., Bowdon, B., Grimsley, J., & Lloyd, H. (1974). Interrelationships of some chemical, physicochemical, and biological activities of several 1-(2-haloethyl)-1-nitrosoureas. Cancer Res., 34(1), 194.

Wheeler, G., Johnston, T., Bowdon, B., McCaleb, G., Hill, D., & Montgomery, J. (1977). Comparison of the properties of metabolites of CCNU. Biochem Pharmacol, 26, 2331.

88

CHAPTER V

MGMT mRNA expression and enzyme activity in canine lymphoma cell lines and

hepatocytes

5.1 Introduction

The DNA protective enzyme O6-methylguanine-DNA methyltransferase (MGMT) is known to play a critical role in the defense against alkylating compounds which produce the O6 alkylguanine lesion. This defense is thought to be crucial at the cellular level as there are many environmental carcinogens that also act as alkylating agents (Kaina et al., 2007). MGMT is not essential for life as MGMT knockout mice are viable and phenotypically normal, however they are hypersensitive to cell killing by alkylating agents (Kaina et al., 2007). Mice that were engineered to be deficient in MGMT, when administered methylnitrosourea (MNU) died early as compared to the wild type and also showed greater frequency of tumor formation (Chaney &

Sancar, 1996; Sakumi et al., 1997).

Unlike other DNA repair mechanisms such as, base excision repair or nucleotide excision repair, MGMT is unique in that it can repair the DNA without the help of other enzymes or cofactors. Repair of the O6 alkylguanine adduct via MGMT is a rapid single step process in which the offending alkyl group is transferred to the cysteine residue of MGMT thereby restoring the affected DNA (Gerson, 2002; Kaina et al., 2007; Sedgwick, 2004). The reaction is stoichiometric in nature where one MGMT molecule removes a single DNA adduct. Once used, the MGMT

89 molecule cannot be recycled and is directed towards ubiquitination (Gerson, 2002; Srivenugopal et al., 1996) and degraded by the proteasome (Xu-Welliver & Pegg, 2002). Therefore a cell’s capacity to repair the O6 adducts on the DNA depends on the number of MGMT molecules already present and also the rate of de novo synthesis (Kaina et al., 2007).

The MGMT level is highly variable among individuals and also among the various tissues in the same individual (Christmann et al., 2011). A long term study of MGMT in PBMC from healthy human subjects showed that there is a 7 fold variation in MGMT activity with very little change over time (Janssen et al., 2001). Data from human studies have shown that liver has the highest MGMT activity, while the brain has the lowest (Christmann et al., 2011). Also the MGMT activity varies between normal and malignant tissues. However it is unclear if MGMT activity changes during carcinogenesis (Christmann et al., 2011).

Due to its functional role in DNA repair, MGMT may have a significant impact on the outcome of cancer therapy with alkylating agents. Certain tumors that are high expressers of

MGMT were found to be less sensitive to treatment with nitrosoureas while tumors that were deficient in MGMT or showed relatively low expression responded better to nitrosourea therapy

(Christmann et al., 2011). MGMT levels in normal tissues may also have significant impact on the choice of therapy to be used. A dose limiting side effect of most nitrosourea compound is myelotoxicity, and it is thought that this could be due to the low MGMT expression in bone marrow cells (Gerson, 2002; Maze et al., 1996). There is data indicating that MGMT expression is inducible (via transcriptional activation) by alkylating agents, X-ray or other DNA damaging agents (Grombacher & Kaina, 1995). Studies in rat liver cells have shown a 2-5 fold MGMT gene

90 induction with maximum induction happening around 24 to 48 hours after exposure to X-ray or alkylating agent, methylnitronitrosoguanidine (MNNG) (Grombacher et al., 1996; Grombacher &

Kaina, 1995).

In this study, we evaluated the MGMT expression and enzyme activity in canine lymphoma cell lines and primary canine hepatocytes. There is no published data available regarding the

MGMT level in canine cells and tissues. Also, we tried to correlate the MGMT levels with the cytotoxicity of lomustine and its metabolites in these cells and also the level of DNA adducts formed by these compounds.

5.2 Materials and Methods

MGMT mRNA expression in 17-71 and GL-1 cells and canine primary hepatocytes

The lymphoma cell lines 17-71 and GL-1 were grown and maintained at 37°C in 5% CO2, and cultures were passaged as necessary to maintain high cell viability (above 90%) in culture media DMEM supplemented with 10% FBS, 1% penicillin/streptomycin. For the MGMT gene induction study, MGMT gene expression was measured in canine lymphoma cell lines 17-71 and

GL-1 at 6, 12, 24 and 48 hours after exposure to varying concentrations of lomustine (1000, 3000 and 12,000 ng/ml) to study MGMT induction. The cells were only treated with 12,000 ng/ml concentration of trans-4 and cis-4, based on the initial results obtained after lomustine treatment study. Untreated samples were used to measure basal MGMT mRNA expression in these cells.

91

Isolation of canine hepatocytes

Hepatocytes were isolated from six dogs that were euthanized at the Champaign county animal control facility. Isolation of canine hepatocytes was done by the by the method of Spotorno et al. (Spotorno et al., 2006) with few modifications. Liver samples (about 25 grams) from freshly euthanized dogs were obtained aseptically and immediately transported in PBS to the lab. About

10 grams of the sample were cut out from the center of the collected liver tissue and transferred to a petri dish containing PBS with 1% penicillin/streptomycin. The tissue was finely minced with scissors and washed a couple of times with PBS. The minced sample was transferred to a flask containing 100 ml of cold collagenase solution (Collagenase II, 300 units/mg, 500 ng/ml in HBSS-

HEPES buffer) and softly stirred for about 10 minutes. At the end of incubation, 10 ml of FBS and

50 ml of ice cold HBSS were added. The suspension was filtered through a nylon cell strainer (100 micron pore size) and the filtrate was centrifuged at 50 g for 5 minutes at 4ºC. The supernatant was discarded and the pellet washed twice with PBS with 1% penicillin/streptomycin and centrifuged twice at 150 g for 2 minutes each and the pellet suspended in 5 ml PBS. The cell suspension was layered over 15 ml cushion of 60% Percoll in PBS in a 50 ml polycarbonate tube and centrifuged at 70 g at 4ºC for 5 minutes. The supernatant was discarded and the pellet washed with PBS. Cell viability was determined by Trypan blue dye exclusion test. The cells were suspended in DMEM (supplemented with 10% FBS and 1% penicillin/streptomycin) at a density of 0.6 million/ml and were incubated at 37ºC for 48 hours to acclimatize and form a monolayer.

After 48 hours the media was replaced by hepatocyte maintenance media (Triangle research Labs,

NC) without FBS or penicillin/streptomycin.

92

MGMT mRNA expression assay

The total RNA from samples was extracted using RNeasy mini kit (Qiagen). Relative expression levels of the MGMT gene was carried out using Taqman Real-Time quantitative RT-

PCR (QPCR) analysis on an ABI Prism 7500 thermal cycler. An ABI canine MGMT specific

Taqman probe (cf02626419_m1) spanning the exon 1/2 junction was used for all analyses. HPRT-

1 was used as internal reference for normalization (ABI Taqman probe cf02626258_m1). For this analysis, 1 µg aliquot of total RNA from a given sample was converted to cDNA using Superscript

III first strand synthesis system (Life technologies). The resulting cDNA was used in separate

Taqman QPCR analyses, each including triplicate technical repeats. Expression comparisons was made between treated and untreated (control) samples and also between different cell types. The mRNA expression was measured two separate times in all samples (canine cell lines and hepatocytes). In order to ascertain that the Taqman assay is able to detect the gene expression correctly, MGMT gene expression was measured in three canine glioma cell lines J3T-Bg, SDT-

3 and G06A (York et al., 2011).

MGMT enzyme activity in 17-71 and GL-1 cells and canine hepatocytes

A fluorometric oligonucleotide assay was used for measuring MGMT enzyme activity

(Kreklau et al., 2001). The following 18 base pair (bp) 5’hexachlorofluoroscein phosphoramitide

(HEX) labelled oligonucleaotide (oligo) containing O6-methylguanine lesion within a PvuII restriction site and its complementary oligo were custom made (Sigma Aldrich).

93

Hex labeled oligo: 5’HEX-GCCCGGCCAG CTGCAGTT

Complementary oligo: 3’-CGGGCCGGTCGACGTCAA

PvuII restriction site

G = O6 methylguanine

Oligonucleotide strands were annealed to complementary strands in 10 mM Tris–HCl, 1 mM

EDTA, 33 mM NaCl, pH 8.0, by heating to 90°C for 2 minutes to denature and cooling to room temperature overnight. The 17-71 and GL-1 cells and hepatocytes were maintained in culture media as mentioned above. Cells (106) were harvested and resuspended in serum-containing culture medium (DMEM), then centrifuged at 1500 g for 5 minutes.

The cells were washed with 5–10 ml of PBS and recentrifuged, then resuspended in 0.5 ml assay buffer (50 mM Tris–HCl, 1 mM DTT, 0.5 mM EDTA, 5% glycerol, pH 8.0) and kept on ice.

Protein from cells (about 106 cells/sample) was extracted by pulse-sonication on ice five times for

5 seconds each, then centrifuged at 14,000 g at 4°C for 30 minutes. The total protein was quantitated by measuring absorbance at 595 nm using the Pierce BCA kit reagent. To measure the

MGMT activity the 200 fmols of HEX labeled oligo was incubated with cell protein (500 µg) at

37°C for 2 hours. The oligo was extracted by phenol:chloroform:isoamyl alcohol (25:24:1) extraction and using sodium acetate and ethanol with tRNA (10 µg) as a carrier. Equal volume of phenol:chloroform:isoamyl alcohol solution was mixed with the oligo cell protein mixture along with 50 µl sodium acetate and 10µg tRNA. This was vortex mixed briefly for a few seconds until an emulsion was formed. This mixture was centrifuged at 12,000 g for 3-5 minutes at room

94 temperature. The upper aqueous phase was transferred to a new tube and the interface and bottom organic phases were discarded. The purified oligo was digested with a restriction enzyme PvuII

(Promega) for 2 hours in a total reaction mixture of 20 µl following manufacturer’s instructions.

PvuII cleaves the oligo at the O6-methylguanine lesion producing two short fragments (10 bp and

8 bp). Then formamide loading buffer (10 µl), without dyes was added to each sample and the samples were denatured by heating at 95°C for 5 minutes. Sample solutions (15 µl) were applied to a pre-warmed 20% polyacrylamide gel containing 8 M in 1× Tris–borate EDTA (TBE) buffer and run at 50 mA for 15 minutes. Detection and quantitation of the fluorescent HEX labeled oligonucleotides was done by using a fluorescent imaging system (Typhoon 9410 Variable Mode

Imager).

The MGMT enzyme activity was quantified by comparing the intensity of the bands (18 bp) between the PvuII treated and PvuII nontreated (control) samples. The MGMT activity was calculated by comparing the intensity (density volume obtained from the fluorescent reader) of the top band (18 bp) in the samples with that in the controls. The deficit in intensity in the sample bands compared to the control bands represents the amount of cleavage which corresponds to the

MGMT enzyme activity. For the cell lines and hepatocytes, the experiment was done with duplicate samples.

For measuring the lower limit of detection (LOD) and limit of quantitation (LOQ) of the method, serial dilution of the HEX labelled oligo was made from 1-200 fmols in deionized water and the fluorescent intensity was measured and this was plotted against the oligo concentration.

The lowest concentration showing 3 times the fluorescent intensity compared to the blank sample

95 was chosen as the LOD and the lowest concentration with CV less than or equal to 15% was chosen as the LOQ of the method.

Statistical analysis

Statistical analysis was done using SPSS (IBM, NY). Descriptive statistics were produced for all continuous variables. Mean and standard deviation were calculated. Kruskal-Wallis one way ANOVA was used to compare the means of the drug treated and non-treated groups at different time points in the MGMT gene induction study. Correlation between MGMT mRNA expression and enzyme activity was evaluated by Pearson’s correlation. The p value was set at

0.05.

5.3 Results

MGMT gene expression

GL-1 cells did not show any MGMT mRNA expression in untreated samples. When GL-1 samples were treated with lomustine, trans-4 and cis-4 at 1000, 3000 and 12,000 ng/ml and incubated for 12, 24 and 48 hours, still no MGMT mRNA expression was detected. In the 17-71 cells, a low basal level of MGMT expression was seen in the untreated samples (Figure 5.1). Upon treatment with various concentrations of lomustine, trans-4 and cis-4 and incubated for different time points, no statistically significant difference was seen in any of the drug treated samples at any time point as compared to the vehicle control (Figures 5.1 and 5.2). Two of the glioma cell

96 lines SDT-3 and J3G-Bg showed high MGMT mRNA expression (Figure 5.3). The results show that the Taqman qPCR assay is able to detect higher transcript levels of MGMT gene and that the previously measured low MGMT gene expressions in 17-71 cells are indeed true values.

The MGMT gene expression measured in the hepatocytes of the 6 dogs was 2-3 fold greater than that seen in the 17-71 cells (Figure 5.4). The variation in MGMT mRNA expression among the 6 dogs was low except for one dog which showed a lower MGMT level.

MGMT enzyme activity

The LOD of the fluorometric MGMT enzyme activity assay was 5 fmols while the LOQ was 10 fmols (Figure 5.5). For GL-1 and 17-71 cell line, using 100 µg and 200 µg of cell protein did not show any MGMT activity. In order to ascertain the functionality of the assay, human recombinant MGMT (Cayman chemicals) was used as a positive control. The results showed that with increasing concentration of MGMT, greater cleavage of the oligo was seen (Figure 5.6). Thus the protein amount in the assay was increased to 500 µg and in the 17-71 samples showed visible cleavage of the oligo (Figure 5.7). The GL-1 cell lines did not show any MGMT enzyme activity even when 500 µg cell protein was used for the assay.

The MGMT activity in the 17-71 cells was about 90 fmols/mg protein (range 83-96 fmols/mg protein, SD 10.11). In the GL-1 cell line, as no MGMT gene expression was detected, consequently, MGMT enzyme activity was also absent. The enzyme activity measured in the hepatocytes was greater than in 17-71 cells, proportional to the mRNA levels. The MGMT enzyme

97 activity measured in canine hepatocytes is about 250-350 fmols/mg protein (Figures 5.8 and 5.9).

There was good correlation between MGMT gene expression and enzyme activity in the 17-71 and the canine hepatocytes (Figure 5.10).

5.4 Discussion

This data is of significance as there is no prior published report of the MGMT gene expression or enzyme levels in canine cells or tissues. The canine lymphoma cell line 17-71 showed low MGMT mRNA expression and enzyme activity (about 90 fmols/mg protein). MGMT activity data from humans show that similar level of activity is seen in brain tissue (about 100 fmols/mg protein) which is one of the organs expressing the lowest level of MGMT activity in the body (Christmann et al., 2011). This may be one of the reasons for the higher success rate of nitrosourea therapy on brain tumors (Christmann et al., 2011). The GL-1 cells did not show any detectable MGMT mRNA expression or enzyme activity. It has been reported that while MGMT is expressed in all normal human cell types and tissues, 20- 30% of human tumor or more than

50% of virally transformed cell lines are completely deficient in MGMT expression (Day et al.,

1980; Tsujimura et al., 1987). It is not clear yet if the process of immortalization may be leading to loss of MGMT expression (Harris et al., 1996). The mechanism of suppression of the MGMT gene in MGMT negative cells is not yet understood. However, it is known that lack of expression is not due to deletion of the gene (Tano et al., 1990) and may possibly be an epigenetic event

(Cairnssmith & Karran, 1992; Vonwronski & Brent, 1994).

98

MGMT induction is known to result from transcriptional activation of the MGMT gene in response to DNA damage (Grombacher et al., 1998). In our gene induction study, the 17-71 cells did not show any significant increase in gene expression after 48 hours of exposure to lomustine and its metabolites. The experiment was only done twice and the percent SD between samples from the two experiment was 7- 35% which probably led to the non-significant values. Time dependent expression of MGMT gene has been measured after exposure to different types of DNA damages including X-ray treatment and methylating agents, and the maximum induction (2-5 fold) was seen at 24 to 72 hours (Grombacher et al., 1996). Also the half-life of MGMT mRNA is known to be over 10-12 hours and that for the protein is over 15-20 hours (Kroes & Erickson, 1995; Pegg,

1977).

The hepatocytes obtained from the six dogs showed a 2-3 fold greater MGMT mRNA expression as compared to the canine cell line 17-71. The MGMT enzyme activity measured in the hepatocytes is about 250-350 fmol/mg protein as compared to 90 fmol/mg protein in 17-71 cells. This indicates a positive correlation between the MGMT mRNA expression and the enzyme activity in the canine cell lines and the hepatocytes. Data from humans also suggest that MGMT activity corresponds with the MGMT protein and RNA level (Christmann et al., 2011). The average MGMT activity in human liver is reported to be around 600 fmol/mg protein, however there is wide inter-individual variation (Christmann et al., 2011; Margison et al., 2003). In our

MGMT activity assay in hepatocytes, there was over 70% enzymatic cleavage of the substrate oligo (Figure 5.7). This may indicate a sub-optimal enzyme-substrate ratio which could lead to a lower rate of reaction. It is suggested that enzyme reactions be conducted under initial rate conditions with total consumption of substrate less than 20% (Wenham et al. 2006). Hence it is

99 possible that the MGMT activity measured in the canine hepatocytes in our study is lower than the actual values.

In chapter 3 we have seen that lomustine and its metabolites showed a time and concentration dependent toxicity in canine lymphoma cell lines 17-71 and GL-1. However the canine hepatocytes did not show any detectable cell killing even 6 days after exposure to the drugs.

When the toxic O6 guanine adduct was measured in the cells after exposure to lomustine and the metabolites, the canine cell lines showed higher levels of the adduct as compared to the hepatocytes. It is known that MGMT directly repairs the O6 adducts in a rapid stoichiometric reaction. In our MGMT study we saw that the hepatocytes had 2-3 fold greater enzyme activity compared to the 17-71 cell line. Correlating all this data together it is plausible that higher MGMT enzyme activity may be a reason for the lower levels of O6 adducts detected in the hepatocyte samples. The higher level of MGMT may cause repair of adducts, and hence contributing to the lower cytotoxicity of lomustine and its metabolites in hepatocytes. However this interpretation must be viewed with caution, as no significant difference in the O6 adduct levels between 17-71 cells and GL-1 cells was noted even though we were unable to detect any MGMT activity in the

GL-1 cells. Also lomustine and its metabolites form multiple adducts at the O6 guanine position out of which we chose the O6 hydroxyethyldeoxyguanosine as a marker or indicator of the genotoxic potential of the compounds. It should be noted that MGMT is only one of the several ways in which alkylation damage in DNA is repaired by the cell. Other pathways such as base excision repair and mismatch repair are also known to repair damaged DNA depending on the stage and type of the damage. Thus it may also be possible that the lower O6

100 hydroxyethyldeoxyguanosine measured in the hepatocytes may be due to lower formation or a

DNA repair mechanism other than MGMT.

In summary, we showed that MGMT mRNA expression in 17-71 cells and canine hepatocytes correlates with the MGMT enzyme activity in these cells. There was low MGMT activity in the canine lymphoma cell line 17-71 while the GL-1 cells did not show any detectable enzyme activity or mRNA expression. The canine hepatocytes have greater MGMT activity as compared to the 17-71 cell line.

101

5.5 Figures and Legends

Figure 5.1 Graph showing MGMT mRNA expression in 17-71 cells before (untreated) and after treatment with lomustine at 1000, 3000 and 10,000 ng/ml and incubated for 6, 12, 24 and 48 hours. No statistical significance was seen in MGMT mRNA expression in any of the drug treated samples at any time point as compared to the untreated samples.

MGMT mRNA expression in 17-71 cells

3 1 - 2.5 2 1.5 1 0.5 0

(fold difference) (fold Untreated 6 12 24 48 normalized to HPRT to normalized

MGMT mRNA expression mRNA MGMT Time (hours)

1000 3000 12000

102

Figure 5.2 Graph showing MGMT mRNA expression in 17-71 cells before (untreated) and after treatment with trans-4-hydroxylomustine and cis-4 hydroxylomustine at 12,000 ng/ml concentration and incubated for 6, 12, 24 and 48 hours. No statistical significance was seen in MGMT mRNA expression in any of the drug treated samples at any time point as compared to the untreated samples.

MGMT mRNA expression in 17-71 cells 3

1 1 2.5 - 2 1.5 1 0.5 0

(fold difference) (fold Untreated 6 12 24 48

normalized to HPRT to normalized Time (hours) MGMT mRNA expression mRNA expression MGMT

Lomustine Trans-4 Cis-4

103

Figure 5.3 Graph showing MGMT mRNA expression in canine glioma cell lines J3T-Bg, SDT-3 and G06A and canine lymphoma cell line 17-71.

MGMT mRNA expression in canine glioma cell lines

16 14 12 10 8 6 4 (fold difference) (fold 2 0

MGMT gene expression/HPRT 1 expression/HPRT gene MGMT J3T-Bg SDT-3 G06A 17-71 Canine cell lines

104

Figure 5.4 Graphs showing the MGMT mRNA expression in canine lymphoma cell line 17-71 cells and primary hepatocytes from 6 dogs.

MGMT mRNA expression

4

1 3.5

- 3 2.5 2 1.5 1

normalized to HPRT to normalized 0.5 MGMT mRNA expression mRNA MGMT 0 Dog 1 Dog 2 Dog 3 Dog 4 Dog 5 Dog 6 17-71

105

Figure 5.5 Serially diluted oligo from 1-200 fmols to detect LOD and LOQ of the method. Amount of oligo in each lane is as follows: Lane 1=1 fmol, lane 2=5 fmols, lane 3=10 fmols, lane 4=20 fmols, lane 5=50 fmols, lane 6=100 fmols, lane 7=150 fmols and lane 8=200 fmols.

1 2 3 4 5 6 7 8

106

Figure 5.6 Oligo treated with increasing concentration of human recombinant MGMT. Lane 1 represents untreated oligo. Oligos in lane 2-7 were treated with MGMT at 5, 10, 20, 50, 100 and 200 ng/ml concentration. Upper and lower bands represent 18 bp substrate and 10 bp cleavage product, respectively.

1 2 3 4 5 6 7

107

Figure 5.7 MGMT activity in canine lymphoma cell line 17-71: lanes 1-3 represent replicate oligo samples treated with 500 μg cell protein from 17-71 cells. Lanes labeled ‘N’ are untreated oligo samples (control). Upper and lower bands represent 18 bp substrate and 10 bp cleavage product, respectively.

1 2 3 N N

108

Figure 5.8 MGMT activity in primary hepatocytes obtained from 6 dogs: lanes 1-6 represent oligo samples treated with 500 μg cell protein from hepatocytes obtained from 6 dogs. Lanes labeled ‘N’ are untreated oligo samples (control). Upper and lower bands represent 18 bp substrate and 10 bp cleavage product, respectively.

1 2 3 4 5 6 N N

109

Figure 5.9 Graph showing MGMT enzyme activity in the primary hepatocytes obtained from 6 dogs and in the canine lymphoma cell line 17-71.

MGMT Enzyme activity 400 350 300 250 200 150 100

50 fmol MGMT/mg fmol MGMT/mg protein 0 Dog 1 Dog 2 Dog 3 Dog 4 Dog 5 Dog 6 17-71

110

Figure 5.10 Graph showing correlation between MGMT gene expression and enzyme activity in canine lymphoma cell line 17-71 and the hepatocytes obtained from 6 dogs. Pearson’s correlation was 0.806 which is highly significant with p value of 0.028. (p value set at 0.05).

111

5.6 References

Cairnssmith, S., & Karran, P. (1992). Epigenetic silencing of the dna-repair enzyme O(6)- methylguanine-dna methyltransferase in mex- human-cells. Cancer Research, 52(19), 5257-5263.

Chaney, S., & Sancar, A. (1996). DNA repair: Enzymatic mechanisms and relevance to drug response. Journal of the National Cancer Institute, 88(19), 1346-1360.

Christmann, M., Verbeek, B., Roos, W. P., & Kaina, B. (2011). O-6-methylguanine-DNA methyltransferase (MGMT) in normal tissues and tumors: Enzyme activity, promoter methylation and immunohistochemistry. Biochimica Et Biophysica Acta-Reviews on Cancer, 1816(2), 179- 190.

Day, R., Ziolkowski, C., Scudiero, D., Meyer, S., Lubiniecki, A., Girardi, A., & Bynum, G. (1980). Defective repair of alkylated dna by human-tumor and Sv40-transformed human cell strains. Nature, 288(5792), 724-727.

Gerson, S. (2002). Clinical relevance of MGMT in the treatment of cancer. Journal of Clinical Oncology, 20(9), 2388-2399.

Grombacher, T., Mitra, S., & Kaina, B. (1996). Induction of the alkyltransferase (MGMT) gene by DNA damaging agents and the glucocorticoid dexamethasone and comparison with the response of base excision repair genes. Carcinogenesis, 17(11), 2329-2336.

Grombacher, T., & Kaina, B. (1995). Constitutive expression and inducibility of O6- methylguanine-DNA methyltransferase and N-methylpurine-DNA glycosylase in rat liver cells exhibiting different status of differentiation. Biochimica Et Biophysica Acta (BBA) - Molecular Basis of Disease, 1270(1), 63-72.

Harris, L., vonWronski, M., Venable, C., Remack, J., Howell, S., & Brent, T. (1996). Changes in O-6-methylguanine-DNA methyltransferase expression during immortalization of cloned human fibroblasts. Carcinogenesis, 17(2), 219-224.

Janssen, K., Eichhorn-Grombacher, U., Schlink, K., Nitzsche, S., Oesch, F., & Kaina, B. (2001). Long-time expression of DNA repair enzymes MGMT and APE in human peripheral blood mononuclear cells. Archives of Toxicology, 75(5), 306-312.

Kaina, B., Christmann, M., Naumann, S., & Roos, W. P. (2007). MGMT: Key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents. DNA Repair, 6(8), 1079-1099.

Kreklau, E., Limp-Foster, M., Liu, N., Xu, Y., Kelley, M., & Erickson, L. (2001). A novel fluorometric oligonucleotide assay to measure O-6-methylguanine DNA methyltransferase, methylpurine DNA glycosylase, 8-oxoguanine DNA glycosylase and abasic endonuclease activities: DNA repair status in human breast carcinoma cells overexpressing methylpurine DNA glycosylase. Nucleic Acids Research, 29(12), 2558-2566.

112

Kroes, R., & Erickson, L. (1995). The role of messenger-rna stability and transcription in O-6- methylguanine dna methyltransferase (mgmt) expression in mer(+) human tumor-cells. Carcinogenesis, 16(9), 2255-2257.

Margison, G. P., Povey, A. C., Kaina, B., & Santibanez Koref, M. F. (2003). Variability and regulation of O6-alkylguanine-DNA alkyltransferase. Carcinogenesis, 24(4), 625-635.

Maze, R., Carney, J. P., Kelley, M. R., Glassner, B. J., Williams, D. A., & Samson, L. (1996). Increasing DNA repair methyltransferase levels via bone marrow stem cell transduction rescues mice from the toxic effects of 1,3-bis(2-chloroethyl)-1-nitrosourea, a chemotherapeutic alkylating agent. Proceedings of the National Academy of Sciences of the United States of America, 93(1), 206-210.

Pegg, A. (1977). Formation and metabolism of alkylated nucleosides: Possible role in carcinogenesis by nitroso compoundsand alkylating agents. In G. Klein, & S. and Weinhouse (Eds.), Adv.cancer res. (pp. 195). New York: Academic Press.

Sakumi, K., Shiraishi, A., Shimizu, S., Tsuzuki, T., Ishikawa, T., & Sekiguchi, M. (1997). Methylnitrosourea-induced tumourigenesis in MGMT gene knockout mice. . Cancer Res., 57, 2415.

Sedgwick, B. (2004). Repairing DNA-methylation damage. Nature Reviews Molecular Cell Biology, 5(2), 148-157.

Srivenugopal, K., Yuan, X., Friedman, H., & AliOsman, F. (1996). Ubiquitination-dependent proteolysis of O-6-methylguanine-DNA methyltransferase in human and murine tumor cells following inactivation with O-6-benzylguanine or 1,3-bis(2-chloroethyl)-1-nitrosourea. Biochemistry, 35(4), 1328-1334.

Tano, K., Shiota, S., Collier, J., Foote, R., & Mitra, S. (1990). Isolation and structural characterization of a cdna clone encoding the human dna-repair protein for O-6-alkylguanine. Proceedings of the National Academy of Sciences of the United States of America, 87(2), 686-690.

Grombacher, T., Eichhorn, U., & Kaina, B (1998). p53 is involved in regulation of the DNA repair gene O6-methylguanine-DNA methyltransferase (MGMT) by DNA damaging agents. Oncogene, 17(7), 845-851.

Tsujimura, T., Zhang, Y., Fujio, C., Chang, H., Watatani, M., Ishizaki, K., & Ikenaga, M. (1987). O-6-methylguanine methyltransferase activity and sensitivity of japanese tumor-cell strains to 1- (4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosourea hydrochloride. Japanese Journal of Cancer Research, 78(11), 1207-1215.

Vonwronski, M., & Brent, T. (1994). Effect of 5-azacytidine on expression of the human dna- repair enzyme O-6-methylguanine-dna methyltransferase. Carcinogenesis, 15(4), 577-582.

113

Wenham, D., Illy, C., St.Pierre, J.A., and Bouchard, N. (2006). Development of high throughput screening assays for kinase drug targets using Alphascreen technology. In L.K. Manor (Eds.) Handbook of assay development in drug discovery. (pp 56). CRC, London.

Xu-Welliver, M., & Pegg, A. (2002). Degradation of the alkylated form of the DNA repair protein, O-6-alkylguanine-DNA alkyltransferase. Carcinogenesis, 23(5), 823-830.

York, D., Higgins, R.J., LeCouteur, R.A., Wolfe, A.N., Grahn, R., Olby, N., Campbell, M., & Dickinson, P.J. (2012). TP53 Mutations in Canine Brain Tumors. Veteterinary Pathology 49: 796.

114

CHAPTER VI

Conclusion

6.1 Conclusion

The primary goal of this project was to study the metabolism of lomustine in dogs and to compare the cytotoxicity of lomustine and its trans-4 and cis-4 metabolites, as these metabolites have shown better therapeutic index compared to the parent drug in human patients (Wheeler et al., 1977). Another goal was to measure the level of the DNA protective enzyme MGMT in canine cells, which is thought to play a role in the cytotoxicity of lomustine.

Chapter 2 discussed the stability and metabolism study of lomustine in dogs. From the stability study we can conclude that lomustine, trans-4 and cis-4 are relatively stable in methanol but labile in canine plasma. Thus, unknown canine samples following administration of lomustine should be stored at -80oC and analyzed within a week for better accuracy when determining the pharmacokinetic parameters of lomustine in dogs. The results of the metabolism study show that the metabolic profile of lomustine in dogs was similar to that in humans with trans-4 being the major metabolite and cis-4 as the minor metabolite. Also parent lomustine was not detected in the blood samples from the dogs at any time after oral administration of the drug. The data obtained from this metabolism study may be considered as preliminary as it was carried out only in three dogs. Data from humans show about 5-10 fold difference between the maximum concentrations of trans-4 observed among patients (Kastrissios et al., 1996). This supports the premise that there is a large inter-individual difference in the metabolic profile of lomustine. However, based on our

115 results, we may conclude that studying the pharmacokinetics of lomustine in dogs may not yield substantial information that could alter therapy. Our preliminary metabolism study results indicate that the metabolites may be carrying out a major part of the chemotherapeutic effects of lomustine as only the metabolites were detected in blood after oral administration of lomustine. But due to their unstable nature that causes them to spontaneously breakdown at physiological body conditions, studying pharmacokinetics of lomustine or its metabolites may not yield any useful data which could be used to modify treatment protocol of lomustine.

Chapter 3 elaborates the comparative cytotoxicity of lomustine and its major metabolites in canine lymphoma cell lines 17-71 and GL-1, PBMC from lymphoma dogs and canine primary hepatocytes. From this study we conclude that there is no difference in the cytotoxicity of lomustine, trans-4 and cis-4 in the canine lymphoma cell lines, 17-71 and GL-1. In addition to the concentration-dependent cell killing, all the three compounds also showed a time dependent-cell killing in the cell lines. Thus, exposure to higher concentration and longer incubation afterwards, showed greater reduction in cell viability. Interestingly, the primary cells like PBMC and hepatocytes did not show any sensitivity towards lomustine and its metabolites. One reason for the non-significant cytotoxic response in hepatocytes could be that primary cells that are non- replicating, may require much longer time to show apparent cell killing with alkylating compounds. Nitrosoureas like lomustine are known to form toxic adducts on the DNA and during cell replication, this epigenetic modification may lead to double strand breaks which is known to be a potent inducer of apoptosis (Kaina, 2003; Kaina et al., 2007). This may explain why we did not see any cell killing in the hepatocytes even six days after lomustine treatment and also in the

PBMC which are terminally differentiated. Evaluating the long term cytotoxic effects may show

116 that the lomustine and its metabolites cause cell killing in hepatocytes, however the time duration till the cytotoxicity becomes apparent, is unpredictable. In some clinical studies, it is reported that hepatoxicity after lomustine treatment may be delayed or cumulative in nature occurring after a few doses (Kristal et al., 2004).

In chapter 4, we compared the alkylating and carbamylating potential of lomustine and its metabolites in canine lymphoma cell lines and hepatocytes. Alkylation is thought to be the reason for the efficacy of the drugs while carbamylation has been implicated as the cause for the side effects seen with lomustine treatment (Lemoine et al., 1991). In the alkylation study we have shown that lomustine and its metabolites form similar levels of the DNA adducts N7 hydroxyethylguanine and O6 hydroxyethyldeoxyguanosine. These adducts may be used as molecular dosimeter of therapeutic response which corresponds to the similar cytotoxicity of these compounds. However the mutagenic adduct O6 hydroxyethyldeoxyguanosine was detected at a significantly lower level (2-3 fold) in the hepatocytes as compared to the cell lines which may indicate either lower adduct formation or rapid repair. Rapid repair of the O6 adduct may be a possibility since all the canine hepatocyte samples showed greater MGMT level.

In terms of carbamylation, lomustine caused greater carbamylation in the canine hepatocytes and lymphoma cell lines. All the three compounds caused greater carbamylation in hepatocytes as compared to the canine cell lines. Also hepatocyte samples from two dogs showed greater carbamylation following treatment with lomustine and its metabolites compared to the others. But it should be noted that all of the dog hepatocyte samples showed no cytotoxicity to lomustine or its metabolites up to six days of incubation. Hence it is unlikely that greater

117 carbamylation in the hepatocytes in the two dogs would lead to cell death in our experiments, although it may be inhibiting various proteins in these cells. The same would also hold true for the cell lines 17-71 and GL-1 where we see lower carbamylation but greater cytotoxicity after treatment with the drugs. Thus, carbamylation may not necessarily lead to cell killing.

Even though the hepatocyte samples showed greater carbamylation than the lymphoma cell lines after exposure to lomustine and its metabolites, the clinical relevance of this with regards to hepatotoxicity is not evident. There is no clear correlation between the carbamylating potential of the isocyanates formed from nitrosourea and the cytotoxicity as nitrosoureas that have low carbamylating activity retain their antitumor activity (Lemoine et al., 1991). The role of carbamylation in the hepatotoxicity caused by lomustine needs to be studied further. Clinical studies need to be done in order to compare the carbamylation level in hepatocytes of dogs that develop hepatotoxicity after lomustine administration versus those that do not. This can be done by collecting needle biopsy samples from the liver of dogs receiving lomustine treatment and evaluating the extent of carbamylation.

Finally, in chapter 5 we studied the MGMT enzyme activity and mRNA expression in canine lymphoma cell lines and hepatocytes. In our gene induction study, the 17-71 cells did not show any significant increase in gene expression after 48 hours of exposure to lomustine and its metabolites. There was low MGMT activity in the canine lymphoma cell line 17-71 while the GL-

1 cells did not show any detectable enzyme activity or mRNA expression. The canine hepatocytes showed 2-3 fold greater gene expression as compared to 17-71 cells. We showed that MGMT mRNA expression in 17-71 cells and canine hepatocytes correlates with the MGMT enzyme

118 activity in these cells. The MGMT enzyme activity measured in the hepatocytes is conservatively greater than 250-350 fmol/mg protein as compared to about 90 fmol/mg protein in 17-71 cells. In our MGMT activity assay in hepatocytes, there was over 70% enzymatic cleavage of the substrate oligo. This may indicate a sub-optimal enzyme-substrate ratio which could lead to a lower rate of reaction thus resulting in underestimation of MGMT activity in hepatocytes. The MGMT activity in hepatocytes needs to be verified by redoing the fluorometric assay and using either lower amount of cell protein or higher amount of oligo substrate such that the reaction does not consume more than 20% of the substrate.

Previously, we have seen that lomustine and its metabolites showed a time and concentration dependent toxicity in canine lymphoma cell lines 17-71 and GL-1. However the canine hepatocytes did not show any detectable cell killing even 6 days after exposure to the drugs.

Also, the canine hepatocytes showed lower levels of the cytotoxic O6 guanine adduct as compared to the lymphoma cell lines. It is plausible that higher MGMT enzyme activity detected in the hepatocytes may be a reason for the lower levels of O6 adducts detected in these cells. The higher level of MGMT may cause repair of adducts, and hence contributing to the lower cytotoxicity of lomustine and its metabolites in hepatocytes. However this interpretation must be viewed with caution, as no significant difference in the O6 adduct levels between 17-71 cells and GL-1 cells was noted even though we were unable to detect any MGMT activity in the GL-1 cells.

In our study, the role of MGMT in the cytotoxicity of lomustine and its metabolites is inconclusive. One way to study the impact of MGMT on the cytotoxic effects of lomustine and its metabolites would be to compare the cytotoxicity of the drugs in cells showing high MGMT level

119

(in our case, canine glioma cell lines SDT-3 and J3T-Bg or canine hepatcocytes) before and after treatment of the cells with an MGMT inhibitor like O6 benzylguanine. O6 benzylguanine acts as a pseudosubstrate for MGMT and causes its depletion (Koch et al., 2007). This will help determine if the level of MGMT impacts the cytotoxicity of lomustine and its metabolites.

120

6.2 References

Kastrissios, H., Chao, N., & Blaschke, T. (1996). Pharmacokinetics of high dose oral CCNU in bone marrow transplant patients. Cancer Chemotherapy and Pharmacology, 38(5), 425-430.

Kristal, O., Rassnick, K., Gliatto, J., Northrup, N., Chretin, J., Morrison-Collister, K., & Moore, A. (2004). Hepatotoxicity associated with CCNU (lomustine) chemotherapy in dogs. Journal of Veterinary Internal Medicine, 18(1), 75-80.

Koch, D., Hundsberger, T., Boor, S., & Kaina, B. (2007). Local intracerebral administration of O6-benzylguanine combined with systemic chemotherapy with of a patient suffering from a recurrent . Journal of Neuro-Oncology, 82(1), 85-89.

Lemoine, A., Lucas, C., & Ings, R. M. J. (1991). Metabolism of the chloroethylnitrosoureas. Xenobiotica, 21(6), 775-791.

Wheeler, G., Johnston, T., Bowdon, B., McCaleb, G., Hill, D., & Montgomery, J. (1977). Comparison of the properties of metabolites of CCNU. Biochemical Pharmacology, 26, 2331.

121