Veterinary Therapeutics • Vol. 4, No. 1, Spring 2003 Preclinical Tolerance and Pharmacokinetic Assessment of MU-Gold, a Novel Chemotherapeutic Agent, in Laboratory Dogs* Mary Lynn Higginbotham, DVMa Carolyn J. Henry, DVM, MSa Kattesh V. Katti, MS, PhD, FRSCb Stan W. Casteel, DVM, PhDc Patricia M. Dowling, DVM, MSd Nagavarakishore Pillarsetty, MSb aDepartment of Veterinary Medicine and Surgery for Radiological Research cVeterinary Medical Diagnostic Laboratory University of Missouri-Columbia Columbia, MO 65211 dVeterinary ■ ABSTRACT MU-Gold, tetrakis (trishydroxymethyl) phosphine gold(I) chloride, a novel gold compound, has cytotoxic effects against human androgen-dependent and -independent prostatic, gastric, and colonic carcinoma in cell culture and against malignant lymphoma in rodent models. A pilot study was conducted to evaluate the tolerance and pharmacokinetic properties of MU-Gold in normal dogs in anticipation of clinical trials in cancer-bearing dogs. MUGold (10 mg/kg) was administered by IV injection to three purpose-bred dogs. Serum was collected from all dogs for measurement of gold levels via atomic absorption spectrometry. In addition, complete blood counts and biochemical profiles were monitored for Dogs 2 and 3 every 7 days for 30 days. A twocompartment IV bolus model with first-order kinetics, mean elimination half-life of approximately 40 hours, and mean volume of distribution of 0.6 L/kg was established. Serum gold concentrations ranging from 10 to 50 µg/ml were sustained for 2 to 3 days with no clinically significant toxicities observed. Based on in vitro results in earlier studies and preliminary pharmacokinetic data collected in the present study, Phase I clinical trials should be conducted to define the optimal dosage, dose-limiting toxicities, and other characteristics of MUGold that will be used to design Phase II clinical trials. bCenter *Funding for this study was provided by the University of Missouri-Columbia, Department of Veterinary Medicine and Surgery Committee on Research, Columbia, MO. 76 Physiologic Sciences University of Saskatchewan Western College of Veterinary Medicine 52 Campus Drive Saskatchewan, Saskatoon, Canada S7N 5B4 ■ INTRODUCTION The role of chemotherapy has been defined as induction therapy, or therapy used as the primary treatment for disease in which no adjunctive therapies exist, adjuvant therapy for local disease, primary treatment for local disease, and site-specific therapy for sanctuary sites of particular cancers.1 Unfortunately, the ability to completely eradicate disease is limited both M. L. Higginbotham, C. J. Henry, K. V. Katti, S. W. Casteel, P. M. Dowling, and N. Pillarsetty P(CH2OH)3 Au + P(CH2OH)3 (HOH2C)3P P(CH2OH)3 Figure 1. MU-Gold chemical structure. by toxicity to normal tissues, such as bone marrow and the gastrointestinal system, and development of resistance of the neoplastic cells to chemotherapeutic agents.1 Although great strides have been made in the ability to treat cancer, there is still a need to develop new compounds with lower levels of toxicity and with known mechanisms of action that can be used in combination treatment protocols. In particular, the development of drugs that are not affected by multidrug resistance will improve the ability to treat neoplastic disease. Gold compounds are a class of drugs that are most commonly used in the treatment of rheumatoid arthritis. Although it is not precisely known how these gold agents work, they appear to reduce inflammation and occasionally may bring about remission of the disease. These agents have also been used for the treatment of other rheumatic diseases and various inflammatory skin disorders.2,3 In vitro and in vivo antitumor activity have also been described for gold compounds.3–5 Although the mechanism of action is not completely understood, one possibility is a preferential inhibition of DNA synthesis, similar to that exhibited by the platinum anticancer drug cisplatin.3 The oral gold compound auranofin showed in vivo activity against leukemia cells but was inactive against various solid tumor models.4,6 Various digold complexes, including [Au(dppe)2]Cl2 were later found to have antitumor activity but did not enter clinical trials due to cardiotoxicity.7 In fact, to the authors’ knowledge, no clinical trials CI – investigating gold compounds for use as anticancer agents have been completed to date.8,9 Gold agents that have been used clinically and experimentally for other applications have produced toxicities including cardiotoxicity,10 nephrotoxicity, thrombocytopenia,11 skin disorders, and possible hepatotoxicity.12 In an effort to develop new radionuclides for imaging and therapy, researchers at the University of Missouri-Columbia investigated a nonradioactive compound, MU-Gold (Figure 1), for comparison with radioactive gold compounds in tissue distribution studies.13 Using radiolabeling techniques, they found that this compound remained in the peripheral circulation too long to be of benefit for radiopharmaceutical applications. However, slow clearance may be beneficial for a compound intended for use as a chemotherapeutic agent in terms of achieving and maintaining adequate drug concentration. Further collaborative investigation with Dr. Hideo Kameia of Aichi-Gaikun University, Japan, demonstrated that MU-Gold has anticancer activity in vitro against several human tumor cell lines, including androgen-dependent and -independent prostatic carcinoma, gastric carcinoma, and colonic carcinoma. The compound slows cell growth in both tissue culture and rodent models of cancer, and has been shown to elongate the G1 phase (for gap 1) of the cell cycle.14 The G1 phase is considered the aDeceased. 77 Veterinary Therapeutics • Vol. 4, No. 1, Spring 2003 initial phase of the cell cycle in which cellular modifications occur in preparation for entry into the synthesis phase (S phase) of the cell cycle.15 In vitro and in vivo studies of MU-Gold demonstrated considerable tumor suppression in cell culture and remarkable increases in survival time of sarcoma-bearing mice. This agent was less toxic than cisplatin given at similar dosages in rodent models.14 It was determined, therefore, that MU-Gold may have potential as a new cytotoxic agent.14 Despite the absence of adverse side effects in rodent trials, specific toxicity testing of MUGold has not yet been performed for any species. Likewise, pharmacokinetic and pharmacodynamic studies of this agent are lacking. The currently marketed parenteral gold compounds, gold sodium thiomalate and aurothioglucose, are administered by IM injection due to poor water solubility. The drugs accumulate in the reticuloendothelial cells of the lymph nodes, kidneys, liver, spleen, and bone marrow.2 MUGold is unique among the gold compounds in that it is a water-soluble compound,13,16 thus permitting IV administration (Figure 1). The purpose of this preliminary pilot study was to evaluate the pharmacokinetics and tolerance of MU-Gold following administration of a single dose at 10 mg/kg IV to three healthy, purpose-bred dogs. The study presented here is considered the first step of preclinical development of this novel cytotoxic agent. Data collected in this preliminary investigation will be used to establish the model for pivotal toxicity and pharmacokinetic studies in dogs. ■ MATERIALS AND METHODS Three adult male, mixed-breed, purpose-bred dogs weighing 25.2 to 29.7 kg each were obtained for this study. This study was approved by and conformed to guidelines of the University of Missouri Animal Care and Use Committee. Baseline evaluations included a physical ex- 78 amination, complete blood count (CBC), serum biochemical profile, complete urinalysis, fecal flotation for parasite ova, heartworm status determination by Knott and ELISA tests, and Rickettsia rickettsia, Ehrlichia canis, and Ehrlichia platys titers by immunofluorescent assay. Components of the serum biochemical profile included glucose, urea nitrogen (BUN), creatinine, sodium, potassium, chloride, total carbon dioxide, anion gap, albumin, total protein, globulin, calcium, phosphorus, cholesterol, total bilirubin, alanine aminotransferase (ALT), alkaline phosphatase, and amylase. Dogs with azotemia (BUN greater than 28 mg/dl, creatinine greater than 1.8 g/dl), isosthenuria (urine specific gravity 1.008 to 1.012), increased ALT activity (greater than 122 U/L), neutropenia (absolute number of cells less than 3,000 per dl), thrombocytopenia (absolute number of cells less than 200,000 per dl), positive rickettsial or ehrlichial titers (greater than 1:80), or positive heartworm tests were excluded from the study. Seventy-two hours before drug administration, 6-Fr jugular catheters were placed for the administration of MU-Gold and for blood sampling. Sedation of the dogs for jugular catheter placement was achieved using morphine (0.4 mg/kg), xylazine (0.4 mg/kg), and atropine (0.04 mg/kg) administered by IM injection. After jugular catheter placement, yohimbine (one-quarter to one-half of the volume of xylazine) was administered IM to reverse the effects of xylazine. Heart rate, respiratory rate, and capillary refill time were monitored throughout the sedation period. Jugular catheters were kept patent by flushing with heparinized saline between placement and sampling times. Because MU-Gold had not previously been administered to dogs, this study was performed in two phases. Dog 1 received a single dose of MU-Gold (10 mg/ml concentration) by IV injection (Table 1). Subsequently, it was deter- M. L. Higginbotham, C. J. Henry, K. V. Katti, S. W. Casteel, P. M. Dowling, and N. Pillarsetty TABLE 1. Dosing Information for MU-Gold Administered by IV Injection to Healthy Dogs Dog Weight (kg) Dosage (mg/kg) MU-Gold Concentration (mg/ml) Dose (mg) Total Volume (ml) 1 2 3 29.7 26.5 25.2 10 10 10 10 100 100 297 265 252 29.7 2.65 2.52 mined that this concentration was unsuitable because of the length of time required to inject this volume and the inability to obtain Time 0 (designated to be the moment the injection of material was complete) samples. Therefore, MU-Gold was reformulated at a concentration of 100 mg/ml for Dogs 2 and 3. MU-Gold was given to these two dogs as a single dose at 10 mg/kg IV (Table 1). Serum was collected for gold measurement at 0, 3, 6, 9, 15, 25, 40, 60, 90 minutes and 2, 3, 4, 5, 6, 7, 8, 12, 18, 24, 48, and 72 hours after administration. Serum was separated within 30 minutes after collection and stored at –20˚C until analysis. Storage of serum has been shown to have no significant effects on the quantitative measurement of gold;17 therefore, storage of samples for batch analysis was considered appropriate. Elemental gold concentrations in serum were quantified using atomic absorption spectrometry, which has previously been established as an effective means for serum gold quantification.17–19 Gold concentrations were measured using a cathode adjusted to give maximum sensitivity at 242.8 nm, with the cathode lamp carrying a current of 14 to 15 mAmp. Gold concentrations were calculated from the height of the recorded peaks.17 Serum biochemical profiles and CBCs were evaluated every 7 days for 30 days after injection (Dogs 2 and 3) for a preliminary assessment of acute toxicity related to drug administration. The pharmacokinetic profile was established using computer software for compartmental modeling and kinetic analysis as well as for calculating noncompartmental analysis parameters (WinNonlin, Pharsight Corporation). Mean values for pharmacokinetic parameters were calculated. ■ RESULTS Physical examinations, serum biochemical profiles, and CBCs performed before jugular catheter placement and MU-Gold administration were considered within normal limits for all three dogs. Dog 2 exhibited a stress leukogram with mild elevations in both neutrophils and monocytes; however, these findings were considered inconsequential, and the dog was a suitable candidate for treatment and evaluation. Tests for heartworm and gastrointestinal parasites were all negative, and titers for R. rickettsia, E. canis, and E. platys were all less than 1:80. MU-Gold, as it was administered to Dog 1 was formulated at 10 mg/ml; however, the formulation was subsequently modified to 100 mg/ml for Dogs 2 and 3 because of difficulties with administering the large volume (approximately 30 ml) of solution in a given time frame to Dog 1 (Table 1). Clinical signs of toxicity were not seen immediately following administration of MUGold or within the 30 days the animals were evaluated. There were no apparent signs of toxicity in any serum biochemistry or CBC variables; however, slight increases in BUN values, not exceeding the normal range, were noted for Dogs 2 and 3 while the creatinine values remained relatively unchanged (Figure 2). 79 Veterinary Therapeutics • Vol. 4, No. 1, Spring 2003 MU-Gold at 10 mg/kg in preparation for the design of similar large30 scale studies. The data collected in the present study can be used to 25 design treatment protocols for fu20 ture in vitro and in vivo studies of 15 this compound. Novel chemotherapeutic agents for treatment of 10 Dog 2 cancers with decreased toxicity are 5 Dog 3 needed to improve our ability to 0 treat malignancies. Because the dog 0 7 14 21 28 offers a model of spontaneously ocTime after Treatment (days) A curring cancers that, in some cases, translate to human disease,20,21 evaluation of the tolerance and phar1 macokinetic properties was the first 0.9 0.8 step toward further evaluation of 0.7 MU-Gold for animal use. 0.6 Treatment of one of the three 0.5 dogs with MU-Gold formulated at 0.4 10 mg/ml demonstrated that the 0.3 time required for infusion of this 0.2 Dog 2 formulation was prolonged, and 0.1 Dog 3 the formulation was subsequently 0 0 7 14 21 28 modified to 100 mg/ml before Time after Treatment (days) B treating Dogs 2 and 3. This more concentrated formulation was satFigure 2. Serum (A) urea nitrogen and (B) creatinine levels measured at weekly intervals after IV administration of MU-Gold to healthy dogs. isfactory for administration of the gold compound providing for more accurate sampling for detection of gold levels in serum. Results indicated A two-compartment IV bolus model with first-order kinetics best described the eliminathat variability in the concentration of the gold tion properties of MU-Gold for the dogs in compound did not affect the serum gold conthis study. The mean elimination half-life (t1⁄2) centrations achieved. was 40 hours and the mean volume of distriThe relatively low volume of distribution and long plasma t1⁄2 for MU-Gold in these three bution was 0.6 L/kg. Serum gold concentranormal dogs may be beneficial in terms of its tions ranged from 10 to 50 µg/ml, and these ability to act as a cytotoxic agent for milevels were sustained for 2 or 3 days (Figure 3). crometastatic disease. Unfortunately, there is ■ DISCUSSION no significant evidence that chemotherapeutic The purpose of this study was to conduct a agents are effective at controlling macropreliminary evaluation of the tolerance and metastatic disease.22 Metastatic cells present in pharmacokinetics following a single IV dose of the vascular system may be exposed to the cySerum Creatinine (mg/dl) Serum Urea Nitrogen (mg/dl) 35 80 M. L. Higginbotham, C. J. Henry, K. V. Katti, S. W. Casteel, P. M. Dowling, and N. Pillarsetty in preparation for large-scale toxicity and organ distribution studies. Dog 1 In addition, this study was de50 Dog 2 signed to evaluate tolerance and Dog 3 40 not delayed toxicity. Delayed toxi30 cities are possible with this compound and further in vivo studies 20 are required to evaluate for poten10 tial long-term toxicity associated with MU-Gold. Data were insuffi0 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 54 57 60 63 66 69 72 75 78 81 84 87 90 cient for statistical analysis of pharmacokinetic properties or laboratoTime after Treatment (min) A ry tests. Future studies with larger numbers of dogs will be designed to allow for statistical analysis of the 20 Dog 1 data of interest. 18 Dog 2 16 Further pharmacokinetic evalua14 Dog 3 tion of MU-Gold elimination and 12 organ distribution following re10 peated dosing is warranted. Addi8 tional studies to determine dose6 limiting toxicities as well as studies 4 to determine the optimal dosage of 2 MU-Gold are planned. In addition, 0 0 4 8 12 16 20 24 28 32 36 40 44 48 52 56 60 64 68 72 in vitro studies to reveal the mechTime after Treatment (hr) B anism of action of MU-Gold are needed before this compound can Figure 3. Serum gold levels (A) 0 (immediately after material was injected) to 90 minutes and (B) 2 to 72 hours after IV administration of different be considered for practical use in concentrations of MU-Gold to healthy dogs. Dog 1 received 10 mg/ml con- the clinic setting. If the hypothesis of G1-phase arrest proves to be corcentration. Dogs 2 and 3 received 100 mg/ml concentration. rect, there is potential for synergism totoxic agent over a more prolonged period of between MU-Gold and either nonphase-speciftime as compared with drugs with a short t1⁄2. ic or G1-phase–specific cytotoxic drugs, which Gold levels that correspond to previously dewill cause the arrest of cycling cells in the G1 termined cytotoxic concentrations (in vitro)14 phase and expose them to a compound that is were sustained for 2 to 3 days in these dogs. cytotoxic within this phase. If such synergism is The authors acknowledge the small number found to exist, it would potentially increase the of dogs and brief posttreatment evaluation overall efficacy of cytotoxic therapy to cells that period are shortcomings to this study. However, are sensitive to MU-Gold. Further in vitro because MU-Gold had not yet been evaluated evaluation for cytotoxicity of MU-Gold against in this species, the minimum number of subhuman non-Hodgkin’s lymphoma is ongoing jects necessary was used to determine prelimias well as a Phase I clinical trial in dogs nary pharmacokinetic properties and tolerance with lymphoma. Gold Level (µ/ml) Gold Level (µ/ml) 60 81 Veterinary Therapeutics • Vol. 4, No. 1, Spring 2003 ■ CONCLUSION In this preliminary pilot study, MU-Gold administered at 10 mg/kg by IV injection to three normal laboratory dogs was well tolerated and did not produce signs of toxicity over a 30-day period. Administration of MU-Gold possesses pharmacokinetic properties consistent with a two-compartment IV bolus model with first-order kinetics. Based on in vitro results in earlier studies and preliminary pharmacokinetic data collected in the present study, Phase I clinical trials should be conducted to define the optimal dosage, dose-limiting toxicities, and other characteristics of MU-Gold that will be used to design Phase II clinical trials. ■ REFERENCES 1. Chu E, DeVita Jr VT: Principles of cancer management: Chemotherapy, in DeVita Jr VT, Hellman S, Rosenberg SA (eds): Cancer Principles and Practice of Oncology, ed 6. Philadelphia, Lippincott, Williams, & Wilkins, 2001, pp 289–291. 2. American Hospital Formulary Service: 1997 Drug Handbook. Bethesda, MD, American Society of Health-System Pharmacists, 1997, pp 2318–2327. 3. Fricker SP: Medicinal chemistry and pharmacology of gold compounds. Transition Met Chem 21: 377–383, 1996. 4. Berners-Price SJ, Mirabelli CK, Johnson RK, et al: In vivo antitumor activity and in vitro cytotoxic properties of bis[1,2-bis(diphenylphosphino)ethane]gold(I) chloride. Cancer Res 46:5486–5493, 1986. 5. Simon TM, Kunishima DH, Vibert GJ, Lorber A: Screening trial with the coordinated gold compound Auranofin using mouse lymphocytic leukemia P388. Cancer Res 41:94–97, 1981. 6. Mirabelli CK, Johnson RK, Sung CM, et al: Evaluation of the in vivo antitumor activity and in vitro cytotoxic properties of Auranofin, a coordinated gold compound, in murine tumor models. Cancer Res 45:32–39, 1985. 7. Hoke GD, Macia RA, Meunier PC, et al: In vivo and in vitro cardiotoxicity of a gold-containing antineoplastic drug candidate in the rabbit. Toxicol Appl Pharmacol 100:293–306, 1989. 8. Ni Dhubhghaill OM, Sadler PJ: Gold complexes in cancer chemotherapy: A review, in Keppler BK (ed): Metal Complexes in Cancer Chemotherapy. Weinheim, 82 VCH Publishers, 1993, pp 221–248. 9. Shaw III CF: Gold, in Fricker SP (ed): Metal Compounds in Cancer Therapy. London, Chapman and Hall, 1994, pp 46–64. 10. Fadok VA, Janney EH: Thrombocytopenia and hemorrhage associated with gold salt for bullous pemphigoid in a dog. JAVMA 181(3):261–262, 1982. 11. Guo Z, Sadler PJ: Metals in medicine. Angew Chem Int Ed 38:1512–1531, 1999. 12. Frazier DL, Hahn KA: Commonly used drugs, in Hahn KA, Richardson RC (eds): Cancer Chemotherapy: A Veterinary Handbook. Baltimore, Williams & Wilkins, 1995, pp 92–93. 13. Berning DE, Katti KV, Volkert WA, et al: 198-Au labeled hydroxymethyl phosphines as models for potential therapeutic pharmaceuticals. Nucl Med Biol 25:577–583, 1998. 14. Pillarsetty N, Katti KK, Hoffman TJ, et al: In vitro and in vivo antitumor properties of tetrakis ([trishydroxymethyl] phosphine) gold(I) chloride. J Med Chem, in press. 15. Kastan MB, Skapek SX: Molecular biology of cancer: The cell cycle, in DeVita VT, Hellman S, Rosenberg SA (eds): Cancer Principles and Practice of Oncology, ed 6. Philadelphia, Lippincott, Williams, & Wilkins, 2001, 91–109. 16. Katti KV, Gali H, Berning DE, et al: Design and development of functionalized water-soluble phosphines: Catalytic and biomedical implications. Acc Chem Res 32:9–17, 1999. 17. Dietz AA, Rubinstein HM: Serum gold. I. Estimation by atomic absorption spectroscopy. Ann Rheum Dis 32(2):124–127, 1973. 18. Lorber A, Cohen RL, Chancy CC, et al: Gold determinations in biological fluids by atomic absorption spectrophotometer: Application to chrysotherapy in rheumatoid arthritis patients. Arthritis Rheum 11:170–177, 1968. 19. Rooney TW, Lorber A, Veng-Pedersen P, et al: Gold pharmacokinetics in breast milk and serum of a lactating woman. J Rheumatol 14(6):1120–1122, 1987. 20. Vail DM, MacEwen GE: Spontaneously occurring tumors of companion animals as models for human cancer. Cancer Invest 18(8):781–792, 2000. 21. MacEwen GE: Spontaneous tumors in dogs and cats: Models for the study of cancer biology and treatment. Cancer Metastasis Rev 9:125–136, 1990. 22. Ogilvie GK, Straw RC, Jameson VJ, et al: Evaluation of single-agent chemotherapy for treatment of clinically evident osteosarcoma metastases in dogs: 45 cases (1987–1991). JAVMA 202(2):304–306, 1993.