throbber
P h a s e I S t u d y o f a n O r a l F o r m u l a t i o n o f Z D 9 3 3 1 A d m i n i s t e r e d
`D a i l y f o r 2 8 D a y s
`
`By Michael B. Sawyer, Mark J. Ratain, Donna Bertucci, Robert P. Smith, Richard L. Schilsky, Nicholas J. Vogelzang, Keith Shulman,
`Edwin C. Douglass, and Gini F. Fleming
`
`Purpose: To define the maximum-tolerated dose and
`dose-limiting toxicities (DLTs) of an oral formulation of
`ZD9331, a novel thymidylate synthase inhibitor that is not a
`substrate for folylpolyglutamate synthase.
`Patients and Methods: Patients had Cancer and Leukemia
`Group B performance status < 2 and refractory solid tumors.
`Initially, patients received ZD9331 daily for 2 weeks, with the
`duration of treatment escalated to a maximum of 4 weeks,
`followed by a 2-week rest period. Once the maximum-toler-
`ated duration of treatment was determined, the dose of
`ZD9331 was increased until DLT occurred.
`Results: Fifty-five patients were enrolled at eight dose
`levels. The DLTs were thrombocytopenia and neutropenia. At
`3 mg/d, two of 19 patients developed DLT; one patient had
`
`grade 3 thrombocytopenia and grade 4 neutropenia, and the
`other patient had grade 3 thrombocytopenia only. Anemia
`was common, with a median hemoglobin nadir of 75% of
`baseline, before recovery or transfusion. The apparent oral
`clearance of ZD9931 was 11.6 ⴞ 6.3 mL/min. Dose-limiting
`myelosuppression was associated with both an increased
`24-hour ZD9931 concentration and blood urea nitrogen.
`Conclusion: The recommended phase II dose on this
`schedule is 3 mg/d for 4 weeks, followed by a 2-week rest
`period. ZD9331 seems to have a manageable toxicity pro-
`file, although it should be used with caution in patients with
`renal impairment.
`J Clin Oncol 21:1859-1865. © 2003 by American
`Society of Clinical Oncology.
`
`T HYMIDYLATE SYNTHASE catalyzes conversion of de-
`
`oxyuridine monophosphate to thymidylate and is the only
`de novo source of thymidylate for DNA synthesis.1 The central
`role of thymidylate synthase in nucleotide synthesis has led to
`the development of several folate analogs that inhibit thymidy-
`late synthase. CB3717, the first antifolate to specifically target
`thymidylate synthase, was abandoned because of its unpredictable
`nephrotoxicity,2 which was thought to be due to its low water
`solubility.3 Because CB3717 had significant antitumor activity, the
`Cancer Research Campaign Center for Cancer Therapeutics devel-
`oped antifolates with improved water solubility.
`This research led to the development of raltitrexed (Tomudex;
`ZD1694; AstraZeneca, Macclesfield, UK), which had improved
`water solubility and decreased nephrotoxicity in animal models.3
`Subsequent clinical studies demonstrated activity of ZD1694 in
`colorectal cancer and led to its approval in Australia, Canada,
`and Europe for treatment of metastatic colorectal cancer.4 In a
`later study in the United States, patients with metastatic colo-
`rectal cancer were randomly assigned to either fluorouracil and
`leucovorin or ZD1694; a slight survival advantage was found for
`fluorouracil and leucovorin.5 On the basis of this study, ZD1694
`was not developed further in the United States. ZD1694 required
`polyglutamation by folylpolyglutamate synthase to effectively
`inhibit thymidylate synthase.6 In vitro studies have shown that
`resistance to antifolates can occur by cancer cells decreasing
`accumulation of polyglutamated antifolates.7,8 In addition, vari-
`ability in polyglutamation may be a factor in the interpatient
`variability of antifolate pharmacokinetics.
`ZD9331 was developed to overcome decreased polyglutama-
`tion as a mechanism of resistance to antifolates and have more
`predictable pharmacokinetics and less interpatient variability
`than ZD1694. Preclinical studies demonstrated that ZD9331 was
`
`active against lymphoid and leukemia cell lines and small-cell
`lung, gastric, and colorectal cancer xenografts.9 ZD9331 was
`more active when given by a protracted schedule than an
`intermittent schedule. In vitro studies using L5178Y TK⫺/⫺
`mouse lymphoma cell lines demonstrated that exposure to 10
`␮mol/L of ZD9331 for 4 hours inhibited colony formation by
`80%, whereas exposure to 0.1 ␮mol/L of ZD9331 for 24 hours
`inhibited colony formation by 99.96%. In vivo studies also
`showed that ZD9331 had superior activity administered on a
`protracted schedule. Mice were implanted with L5178Y TK
`⫺/⫺ cells and treated with ZD9331; nine of 16 mice treated with
`100 mg/kg of ZD9331 given by continuous infusion for 7 days
`were cured, whereas none of the mice treated with 100 mg/kg of
`ZD9331 given by intraperitoneal injection were cured.10 Preclin-
`ical pharmacokinetic studies showed good oral bioavailability:
`30% to 60% in rats at 6 mg/m2 and 80% in dogs at 2 mg/m2.
`These preclinical studies demonstrating increased activity
`with prolonged exposure to ZD9331 and good oral bioavailabil-
`
`From the Committee on Clinical Pharmacology, Department of Medicine,
`Cancer Research Center, and Section of Hematology/Oncology, University
`of Chicago; Committee on Clinical Pharmacology and Cancer Research
`Center, Chicago, IL; AstraZeneca Pharmaceuticals, Macclesfield, UK, and
`Wilmington, DE.
`Submitted January 30, 2002; accepted February 13, 2003.
`Supported in part by AstraZeneca and grant no. CA 14599 from the
`National Cancer Institute, National Institutes of Health.
`Michael B. Sawyer was the Gordon E. Richards Fellow of the Canadian
`Cancer Society.
`Address reprint requests to Mark J Ratain, MD, 5841 S Maryland Ave,
`MC2115, Chicago, IL 60637; email: mratain@medicine.bsd.uchicago.edu.
`© 2003 by American Society of Clinical Oncology.
`0732-183X/03/2109-1859/$20.00.
`
`Journal of Clinical Oncology, Vol 21, No 9 (May 1), 2003: pp 1859-1865
`DOI: 10.1200/JCO.2003.01.148
`
`Amerigen Exhibit 1103
`Amerigen v. Janssen IPR2016-00286
`Downloaded from ascopubs.org by Univ of Chicago Library on January 3, 2017 from 205.208.061.064
`Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
`
`1859
`
`

`
`1860
`
`ity prompted this phase I study of oral ZD9331 with prolonged
`administration. Because the intravenous formulation of ZD9331
`had demonstrated that clearance was independent of body-
`surface area,11 this oral formulation study used fixed dosing
`rather than dosing that was based on body-surface area.
`
`PATIENTS AND METHODS
`
`Patient enrollment began in April 1998, and the study was closed in
`February 2000. Eligible patients had a solid tumor that was refractory to
`standard therapies or for which no standard therapy existed; Cancer and
`Leukemia Group B performance status ⱕ 2; age ⱖ 18 years; a life
`expectancy ⱖ 12 weeks; a baseline platelet count ⱖ 100 ⫻ 109/L; a total
`WBC count ⱖ 3.5 ⫻ 109/L or an absolute neutrophil count ⱖ 2 ⫻ 109/L;
`serum bilirubin concentration ⱕ 1.25 times the upper limit of normal; ALT
`and AST ⱕ 2.5 times the upper limit of normal in the absence of liver
`metastases and ⱕ 5 times the upper limit of normal if liver metastases
`were present; serum creatinine ⱕ 1.25 times the upper limit of normal; no
`severe or uncontrolled systemic disease, such as uncompensated respira-
`tory or cardiac conditions; no use of folate-containing vitamin supple-
`ments; and no chlorambucil, mitomycin, or nitrosoureas for more than 6
`months total duration.
`
`Treatment Plan
`
`Patients took ZD9331 as a single daily dose, between 8 and 10 AM (either
`30 minutes before food or 2 hours after). The initial patient was treated with
`ZD9331 0.5 mg/d given for 14 days, followed by a 14-day rest period.
`Because no toxicity was observed, the next patient was treated for 21 days,
`followed by a 2-week rest period. The treatment period was increased to 28
`days followed by a 2-week rest period for the next patient. The treatment
`period then remained at 28 days for the duration of the study. The dose of
`ZD9331 was doubled until the development of any grade of drug-related
`toxicity, with a minimum of one patient per dose level. Subsequent dose
`escalations followed a modified Fibonacci scheme starting at the 67%
`dose-escalation level, with a minimum of three patients enrolled per dose
`level.
`If any patient developed dose-limiting toxicity (DLT),
`three
`additional patients were enrolled at that dose level. At least 12 patients
`were to be enrolled at
`the recommended phase II dose, which was
`anticipated to be the dose at which less than one third of patients
`experienced DLT in the first cycle.
`DLT was defined as grade 4 neutropenia of any duration with fever, grade
`4 neutropenia without fever for at least 7 days, or grade 4 thrombocytopenia.
`Patients stopped treatment if they developed grade 2 neutropenia or throm-
`bocytopenia while still taking ZD9331, and this was considered a DLT.
`Grade 3 or 4 nonhematologic toxicity that was not ameliorated by symp-
`tomatic directed therapy (with the exception of reversible elevations of AST
`or ALT) was also considered a DLT.
`Chemistry and hematologic measurements were repeated weekly during
`the first two cycles and within 3 days of starting the next cycle. Tumor
`measurements were repeated every two cycles.
`Patients remained on study until there was evidence of tumor progression,
`unacceptable toxicity, or until the patient or investigator felt continuing
`treatment with ZD9331 was not in the patient’s interest. Patients who
`experienced DLT but who were clinically benefiting from ZD9331 could
`continue to receive treatment at the investigator’s discretion at a reduced
`dose. The University of Chicago institutional review board approved the
`protocol and the consent form. Written informed consent was obtained from
`all patients.
`
`Pharmacokinetics
`
`Samples for pharmacokinetic analysis were collected on day 1 pretreat-
`ment and at 1, 2, 4, 6, 8, and 24 hours after starting treatment. Samples were
`also taken at predose and 1, 2, 4, and 6 hours postdose 1 week before the end
`of dosing, although these results are not presented here. The AstraZeneca
`Safety of Medicines Laboratory determined the plasma levels of ZD9331
`
`Table 1. Patient Characteristics
`
`SAWYER ET AL
`
`No. of Patients
`(n ⫽ 55)
`
`36:19
`
`Female/male
`Age, years
`Median
`Range
`CALGB performance status
`0
`1
`2
`Prior treatment
`Chemotherapy only
`Chemotherapy and radiation
`Chemotherapy regimens
`Median
`Range
`Cancer diagnosis
`Colorectal
`Ovarian
`Endometrial
`Esophagus
`Hepatocellular
`Others
`
`58
`33-79
`
`2
`1-8
`
`23
`29
`3
`
`36
`19
`
`31
`6
`2
`2
`2
`12
`
`Abbreviation: CALGB, Cancer and Leukemia Group B.
`
`(AstraZeneca) using a high-performance liquid chromatography–mass spec-
`trometry–mass spectrometry assay as previously described.8
`A noncompartmental approach was used to characterize the pharmacoki-
`netic parameters of ZD9331. The pharmacokinetic software used to analyze
`the plasma concentrations was WinNonlin (version 2.1; Pharsight Corp,
`Cary, NC). The area under the concentration-time curve (AUC) was
`calculated to the 24-hour sampling point (after the first dose) using the linear
`trapezoidal rule. The terminal half-life was estimated from the terminal part
`of the log concentration-time curve. The apparent oral clearance (Cl/F) was
`calculated as dose divided by AUC. Creatinine clearance was estimated
`using the formula of Cockcroft and Gault.12
`
`Statistics
`
`Stata 6 (version 6.0; Stata Corp, College Station, TX) was used to perform
`the statistical analysis. Pharmacokinetic parameters were examined for
`possible relationships with the development of myelosuppression using the
`Student’s t test. Exploratory analyses (using univariate and multivariate
`regression) were conducted to assess the possible relationship of various
`patient characteristics (eg, age, renal function) to ZD9331 clearance. In
`addition, an exploratory pharmacodynamic analysis was conducted after log
`transformation of the hematologic parameters. This analysis included an
`assessment of the relative contributions of variability in peak (maximum
`concentration; Cmax) and trough concentrations (24-hour minimum concen-
`tration; Cmin) and the AUC.
`
`RESULTS
`
`Demographics
`
`A total of 55 patients were enrolled, from April 1998 to
`February 2000. Patient characteristics are outlined in Table 1.
`Patients were assessable for toxicity if they were able to
`complete cycle 1 of treatment. Forty-five patients were assess-
`able for toxicity. One patient who developed suspected cellulitis
`during cycle 1 was withdrawn and therefore was not assessable
`for toxicity. Two patients died while on study, but the deaths
`were not considered to be related to ZD9331. One patient
`
`Downloaded from ascopubs.org by Univ of Chicago Library on January 3, 2017 from 205.208.061.064
`Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
`
`

`
`PHASE I STUDY OF AN ORAL ZD9331 FORMULATION
`
`1861
`
`presented to her local hospital in the third week of treatment with
`acute shortness of breath; at presentation her WBC count was
`9.6 ⫻ 109/L, creatinine was 0.5 times the upper limit of normal,
`and blood urea nitrogen (BUN) was 9 mg/dL. Her respiratory
`status deteriorated, and she died from a presumed pulmonary
`embolus. At enrollment, she had a creatinine clearance of 81
`mL/min and BUN of 13.5 mg/dL. The other patient had a history
`of recurrent urinary tract infections and was taking nitrofurantoin
`to prevent recurrent infections. At enrollment, she had a calcu-
`lated creatinine clearance of 151 mL/min and a BUN of 9 mg/dL.
`She developed urosepsis in the fifth week of treatment and
`presented to her local hospital. She progressed to septic shock
`and her advance directive precluded inotropic support. She died
`despite maximal support outside of the intensive care unit. The
`remaining patients not assessable for toxicity either withdrew
`consent or developed progressive disease, preventing completion
`of one cycle.
`
`Dose Escalation
`
`A total of 128 cycles were administered, and the median
`number of cycles completed by each patient was two (range, one
`to seven cycles). The treatment duration was escalated to 28 days
`(at a dose of 0.5 mg/d) without the occurrence of DLT. The dose
`of ZD9331 was then escalated from 0.5 to 5 mg/d. No DLTs
`occurred until the 5 mg/d dose level, at which two patients
`developed DLTs. At the prior dose level of 3 mg/d, no significant
`drug-related toxicity was observed, which led us to examine 4
`mg/d. The three initial patients evaluated at 4 mg/d experienced
`more severe toxicity than patients treated at 5 mg/d. A prelimi-
`nary analysis suggested that patients who experienced DLT at
`the 4 and 5 mg/d dose levels had decreased drug clearance and
`that this was correlated with an elevated BUN. Seven additional
`patients were enrolled at the 4-mg level to test this hypothesis;
`three patients had a high BUN, ⱖ 20 mg/dL (4.0H), and four
`patients had a low BUN,
`less than 20 mg/dL (4.0L). An
`additional 16 patients were subsequently enrolled at the recom-
`mended phase II dose of 3 mg/d to better define the toxicity and
`pharmacodynamics at this dosage level.
`
`Hematologic Toxicity
`
`The DLT of ZD9331 was myelosuppression, both thrombo-
`cytopenia and neutropenia (Table 2). Patients developed throm-
`bocytopenia at a median of 14 days after starting therapy (range,
`11 to 14 days). Platelet counts returned to baseline at a median
`of 10 days (range, 7 to 14 days). Neutropenia occurred at a
`median of 21 days (range, 11 to 21 days), with a median duration
`of 7 days before recovery (range, 7 to 11 days). At 3 mg/d, the
`recommended phase II dose, two of 19 patients developed DLTs;
`one patient had grade 3 thrombocytopenia and grade 4 neutro-
`penia, and the other patient had grade 3 thrombocytopenia only.
`Anemia was a common toxicity at doses ⱖ 3 mg/d (Fig 1), but
`was not dose-limiting. This was managed with erythropoietin
`and RBC transfusions. Midway through the study, one patient
`developed anemia associated with a low haptoglobin, elevated
`bilirubin and lactate dehydrogenase (LDH), and a negative
`Coombs test. After this index case, all subsequent patients had
`
`Table 2. Hematologic Toxicity by Dose Level (fully assessable patients only)
`
`Dose of
`ZD9331
`(mg/d)
`
`0.5
`0.5
`0.5
`1.0
`1.5
`3.0
`5.0
`
`4.0
`
`3.0
`
`4.0H*
`
`4.0L*
`
`Cycle Length
`(days)
`
`No. of
`Patients
`
`Grade 3 and 4 Adverse Events
`(no. of patients)
`
`Dose-Limiting
`Toxicity
`(no. of patients)
`
`14
`21
`28
`28
`28
`28
`28
`
`28
`
`28
`
`28
`
`28
`
`2
`2
`2
`3
`2
`3
`5
`
`3
`
`16
`
`3
`
`4
`
`—
`—
`—
`—
`—
`—
`Grade 3 leukopenia (1), grade
`3 thrombocytopenia (1),
`grade 3 neutropenia (1)
`Grade 3 thrombocytopenia
`(1), grade 3 neutropenia
`(1), grade 4 neutropenia (1)
`Grade 3 thrombocytopenia
`(2), grade 3 anemia (1),
`grade 4 neutropenia (1)
`Grade 3 thrombocytopenia
`(1), grade 2
`thrombocytopenia (1)
`Grade 4 thrombocytopenia
`(1), grade 2 nausea and
`vomiting (1)
`
`—
`—
`—
`—
`—
`—
`2
`
`2
`
`2
`
`2
`
`2
`
`*4.0H (high) blood urea nitrogen ⱖ 20 mg/dL, 4.0L (low) blood urea nitrogen ⬍
`20 mg/dL.
`
`haptoglobin levels measured along with routine biochemistry. At
`the 3-mg dose level, five of 16 patients had anemia with
`undetectable haptoglobin levels. As Fig 2 demonstrates, the
`decrease in hemoglobin was gradual over several weeks. There
`was no significant difference in nadir hemoglobin, bilirubin, or
`LDH between patients with or without low haptoglobin levels.
`
`Nonhematologic Toxicity
`
`The most frequent toxicities encountered at the 3-mg dose
`level are listed in Table 3. The most common toxicity was
`fatigue, which occurred in almost all patients. Nausea was a
`frequent side effect but was not dose-limiting. Four patients
`developed an erythematous maculopapular rash, which preceded
`the development of myelosuppression. Six patients developed
`elevated AST and ALT while receiving therapy, but the ALT and
`AST levels returned to baseline during the rest period and did not
`exceed grade 2 toxicity.
`
`Tumor Response
`
`Patients were evaluated after the first two cycles and every
`two cycles thereafter. There were no objective minor or major
`responses observed, but 14 patients had prolonged stabilization
`of disease. The median duration of stable disease was 12 weeks
`(range, 12 to 48 weeks). At the 3-mg dose level, five of 19
`patients achieved stable disease, including two patients who had
`prolonged stable disease (36 and 48 weeks). The majority of
`patients with stable disease had colorectal cancer (n ⫽ 10), but
`two patients with ovarian cancer also had disease stabilization.
`
`Pharmacokinetics
`
`None of the patients with DLT reached the day 21 sampling
`time. Therefore, to avoid biasing the pharmacokinetic analysis,
`
`Downloaded from ascopubs.org by Univ of Chicago Library on January 3, 2017 from 205.208.061.064
`Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
`
`

`
`1862
`
`SAWYER ET AL
`
`Fig 1. Relationship between dose
`and development of anemia. Symbols
`differentiate dose levels.
`
`the pharmacokinetic analysis is limited to the day 1 pharmaco-
`kinetic samples that are available for all patients. ZD9331
`concentrations were measured in 51 patients on day 1 (Table 4).
`A sample concentration-time profile of a patient at the 3-mg dose
`level is shown in Fig 3. Peak concentrations occurred within the
`first 2 hours, followed by a rapid decline. Some patients who had
`decreased Cl/F reached a plateau in the terminal part of the
`concentration-time curve. There was a rebound in concentra-
`tions seen at 6 to 8 hours, which may represent enterohepatic
`circulation. The mean terminal half-life at 3 mg/d was 25 ⫾
`
`22 hours (range, 9 to 109 hours). The mean half-life is likely
`an underestimate of the true half-life; only 24-hour data were
`available on all patients because patients with an estimated
`long half-life came off study early, before the late pharma-
`cokinetic sample times.
`As the dose of ZD9331 increased from 3 to 5 mg (a 67%
`increase in dose), the mean AUC increased from 2,839 to
`3,431 ng/mL 䡠 h (a 21% increase), and the mean peak
`concentrations were 206 ng/mL and 298 ng/mL, respectively
`(a 45% increase).
`
`Time course of the develop-
`Fig 2.
`ment of anemia. Symbols differentiate
`time points.
`
`Downloaded from ascopubs.org by Univ of Chicago Library on January 3, 2017 from 205.208.061.064
`Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
`
`

`
`PHASE I STUDY OF AN ORAL ZD9331 FORMULATION
`
`Table 3. Common Toxicities at the 3 mg/d Dose Level
`(n ⴝ 19)
`
`Fatigue
`Nausea
`Anorexia
`Constipation
`Alopecia
`Diarrhea
`Vomiting
`Increased AST
`Dyspnea
`Increased ALT
`Ankle edema
`Rash
`Mucositis
`Skin hyperpigmentation
`Fever
`Increased bilirubin
`
`1
`
`8
`14
`10
`10
`6
`6
`5
`5
`5
`3
`4
`4
`3
`2
`1
`
`2
`
`9
`1
`5
`
`2
`1
`1
`1
`
`2
`
`1
`
`Grade
`
`3
`
`1
`1
`1
`
`4
`
`1
`
`1
`
`1
`
`The Cl/F, AUC, Cmax, and Cmin for each dose level are listed
`in Table 4. At doses of ⱖ 3 mg/d, there was no relationship
`between dose and the pharmacokinetic parameter AUC or Cmin.
`Thus data for the 3-, 4-, and 5-mg dose levels were combined for
`the purpose of analysis of the determinants of pharmacokinetic
`variability. At doses ⱖ 3 mg/d, there was no significant corre-
`lation between AUC and creatinine clearance (r ⫽ 0.14) but
`BUN was correlated with AUC (r ⫽ 0.42; P ⫽ .007). At doses ⱖ
`3 mg/d, Cmin was significantly related to BUN (r ⫽ 0.49; P ⬍
`.001) but not to creatinine clearance (r ⫽ 0.20).
`
`Pharmacodynamics
`
`In phase I studies, the large number of dose levels studied can
`confound analysis of determinants of pharmacodynamic vari-
`ability.13 Toxicity is usually correlated with dose administered
`and therefore dose level. The relationship between dose and
`AUC will lead to a false-positive association between AUC and
`toxicity unless the confounding issue of dose is removed. At the
`3-, 4-, and 5-mg dose levels, there was no correlation between
`dose and AUC. The pharmacodynamic analysis was therefore
`confined to the 3-, 4-, and 5-mg dose levels to eliminate potential
`confounding by dose level.
`Demographic characteristics, pharmacokinetic parameters,
`and organ function were examined for possible relationships
`
`1863
`
`Fig 3.
`
`Typical concentration profile of ZD9331.
`
`with thrombocytopenia and anemia. In the univariate analysis,
`significant predictors of log platelet nadir were RBC folate, dose,
`Cmin, half-life, AUC, clearance, bilirubin, BUN, and log Cmin
`(Table 5). Serum folate, Cmax, performance status, albumin
`creatinine clearance, BUN/creatinine ratio, age, and log baseline
`platelet count were not associated with log platelet nadir. In the
`multivariate analysis, only log Cmin and BUN were correlated
`with log platelet nadir (r2 ⫽ 0.44; P ⬍ .001).
`In univariate analysis, significant predictors of log hemoglobin
`nadir were baseline hemoglobin, dose, Cmin, bilirubin, albumin,
`creatinine clearance, and log Cmin. Serum folate, Cmax clearance,
`AUC, performance status, BUN, and BUN/creatinine ratio did
`not reach statistical significance. In multivariate analysis, signif-
`icant predictors of log nadir hemoglobin were log baseline
`hemoglobin, albumin, and log Cmin (r2 ⫽ 0.61; P ⬍ .001).
`AUC (during the first 24 hours), Cmin, creatinine clearance,
`and BUN were examined as predictors for dose-limiting myelo-
`suppression (Table 6). BUN levels were significantly higher in
`patients who developed dose-limiting myelosuppression com-
`pared with patients who did not (Student’s t test, P ⫽ .04). In
`addition, Cmin was higher in patients who developed dose-
`limiting myelosuppression than in patients who did not (Stu-
`dent’s t test, P ⫽ .03).
`
`DISCUSSION
`
`The recommended phase II dose for ZD9331 is 3 mg/d for 4
`weeks followed by a 2-week rest period. Some patients were able
`to tolerate ZD9331 doses of 4 and 5 mg/d without toxicity.
`Because preliminary pharmacokinetic analysis suggested that
`
`Table 4. Pharmacokinetic Parameters
`
`Dose Level
`(mg/d)
`
`0.5
`1.0
`1.5
`3.0
`4.0
`5.0
`
`No. of Patients
`
`Cl/F (mL/min)
`
`AUC (ng/mL 䡠 h)
`
`Cmin (ng/mL)
`
`Cmax (ng/mL)
`
`6
`3
`1
`23
`12
`6
`
`9.9 ⫾ 4.8
`22.1 ⫾ 9.5
`13.8
`12.1 ⫾ 8.8
`10.0 ⫾ 4.5
`17.9 ⫾ 8.2
`
`423 ⫾ 125
`414 ⫾ 138
`1203
`2,839 ⫾ 857
`3,441 ⫾ 1241
`3,431 ⫾ 1045
`
`13 ⫾ 6
`12 ⫾ 4
`27
`79 ⫾ 35
`110 ⫾ 50
`80 ⫾ 50
`
`32 ⫾ 13
`34 ⫾ 14
`96
`206 ⫾ 66
`215 ⫾ 58
`298 ⫾ 70
`
`Abbreviations: Cl/F, apparent oral clearance; AUC, area under the curve; Cmin, minimum concentration; Cmax, maximum
`concentration.
`
`Downloaded from ascopubs.org by Univ of Chicago Library on January 3, 2017 from 205.208.061.064
`Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
`
`

`
`1864
`
`SAWYER ET AL
`
`Table 5. Univariate Analysis
`
`Correlation
`
`P
`
`Log platelet nadir variable
`RBC folate
`Dose
`Cmin
`Half-life
`AUC
`Clearance
`Bilirubin
`BUN
`Log Cmin
`Log hemoglobin nadir variable
`RBC folate
`Log baseline hemoglobin
`Dose
`Cmin
`Bilirubin
`Albumin
`Creatinine clearance
`Log Cmin
`
`⫺0.30
`⫺0.43
`⫺0.60
`⫺0.36
`⫺0.48
`0.38
`⫺0.30
`⫺0.48
`⫺0.50
`
`⫺0.32
`0.59
`⫺0.32
`⫺0.38
`⫺0.31
`0.59
`0.36
`⫺0.44
`
`.04
`.004
`.0001
`.02
`.001
`.01
`.02
`.001
`.0004
`
`.04
`.0001
`.03
`.01
`.04
`.0001
`.02
`.003
`
`Abbreviations: Cmin, minimum concentration; AUC,
`area under the curve; BUN, blood urea nitrogen.
`
`elevated BUN predisposed patients to develop ZD9331 toxicity,
`we therefore re-examined the 4 mg/d dose level, enrolling
`patients on the basis of BUN levels. However, even in the group
`with low BUN, we could not safely administer ZD9331 at 4
`mg/d and could not recommend 4 mg/d as the initial phase II
`dose. Given that only two of 19 patients at 3 mg/d had DLT,
`patients who tolerate ZD9331 3 mg/d might be considered for
`escalation to a dose of 4 mg/d.
`The DLT of ZD9331 on this oral schedule was neutropenia
`and thrombocytopenia; this contrasts with other studies in which
`cutaneous toxicity and myelosuppression were found to be the
`DLTs.11,14 In the study by de Jonge et al,14 dose-limiting
`cutaneous toxicity occurred at 10 mg twice a day given for 5
`days. In the study by Goh et al,11 dose-limiting skin toxicity
`occurred at 12 mg/m2 and 25 mg/d given daily for 5 days. In
`this study,
`the daily administration of ZD9331 was more
`prolonged at 4 weeks, and the highest daily dose administered
`was 5 mg/d. The total dose of ZD9331 administered per cycle
`in the three studies was not substantially different (100 v 125
`v 84 mg/cycle). ZD9331 was administered four times more
`rapidly in the two schedules associated with skin toxicity than
`in this study. This indicates that skin toxicities may be related
`
`Table 6. Parameters Examined for Relationship to Dose-Limiting
`Myelosuppression (fully assessable patients only)
`
`Parameter
`
`AUC
`Cmin
`Creatinine clearance
`BUN
`Half-life
`
`Patients Without
`Myelosuppression
`
`2,975 ⫾ 794
`77.6 ⫾ 32.6
`94 ⫾ 34
`14 ⫾ 4
`20.8 ⫾ 10.9
`
`Patients With
`Myelosuppression
`
`3,877 ⫾ 1,249
`128 ⫾ 57.1
`86 ⫾ 30
`22 ⫾ 9
`39.2 ⫾ 10.2
`
`P
`
`.07
`.03
`NS
`.04
`.12
`
`Abbreviations: AUC, area under the curve; Cmin, minimum concentration; BUN,
`blood urea nitrogen; NS, not significant.
`
`to peak concentrations and the rate at which thymidylate
`synthase was inhibited.
`Anemia was a common toxicity at or above the 3-mg dose
`level, but was not dose-limiting and was easily managed with
`erythropoietin and RBC transfusions. Midway through the study,
`one patient developed anemia associated with a low haptoglobin
`level. The decrease in hemoglobin was gradual over several
`weeks, which is more likely secondary to decreased production
`than hemolysis.
`Three studies have implicated methotrexate (in combina-
`tion with other agents) with elevations of LDH or anemia
`associated with a low haptoglobin level. Klimo and Connors15
`first reported elevation of LDH levels in the majority of
`patients treated with a regimen consisting of methotrexate,
`doxorubicin, cyclophosphamide, vincristine, prednisone, and
`bleomycin. The patients with elevated LDH were in complete
`or partial remission and had no elevations in other hepatic
`enzymes. In another study of methotrexate, doxorubicin,
`cyclophosphamide, vincristine, prednisone, and bleomycin,
`McAdam et al16 noted an increase in LDH during treatment
`that returned to baseline before the next cycle. In a study of
`cyclophosphamide, doxorubicin, methotrexate, bleomycin,
`vincristine, etoposide,
`ifosfamide, and prednisolone, Ma-
`ruyama et al17 observed an elevation of LDH isoenzymes 1
`and 2. In addition, six patients had undetectable haptoglobin
`levels. We speculate that anemia with elevation of LDH and
`suppressed haptoglobin may be an unappreciated side effect
`of the antifolate class of chemotherapeutic agents as a result
`of ineffective erythropoiesis.
`Given orally, ZD9331 showed a less than dose-proportional
`increase of AUC at doses ⱖ 3 mg/d. Potential explanations
`are solubility-limited absorption (discounted because of the
`known physiochemical properties of the compound), satura-
`ble gastrointestinal absorption, nonlinear protein-binding, or
`saturable renal tubular reabsorption. Although interpretation
`is difficult because of the variability of the data and low
`numbers of subjects at lower doses, the increase of Cmax with
`dose could suggest that saturable gastrointestinal absorption
`is not the entire explanation. Because preclinical studies did
`not demonstrate saturable protein-binding, it is possible that
`the nonlinearity may be secondary to saturation of
`the
`extensive renal reabsorption of this folate analog.
`The dominant role of reabsorption in the renal handling of
`ZD9331 may explain the relationship between Cmin and BUN of
`ZD9331 and the poor correlation between Cmin and calculated
`creatinine clearance; the latter is an estimation of the amount of
`blood that is filtered at the glomerulus and enters the proximal
`tubule. Creatinine clearance is a useful surrogate for predicting
`the clearance of drugs that are predominantly filtered, such as
`carboplatin, but may not be well correlated with the ability of
`the proximal tubule to reabsorb solutes and electrolytes. BUN
`is a better surrogate for tubular function because urea is
`reabsorbed. The parallels between urea reabsorption and
`ZD9331 reabsorption may be the basis for the apparent
`relationship between Cmin and BUN. Thus ZD9331, like other
`
`Downloaded from ascopubs.org by Univ of Chicago Library on January 3, 2017 from 205.208.061.064
`Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
`
`

`
`PHASE I STUDY OF AN ORAL ZD9331 FORMULATION
`
`1865
`
`renally excreted drugs, should be used with caution in patients
`with renal insufficiency or in combination with drugs that
`affect proximal tubular reabsorption.
`Plasma 2⬘-deoxyuridine (dUrd) is the substrate of thymidylate
`synthase that is elevated when thymidylate synthase is inhibited.
`Jackman et al18 and Mitchell et al19 have shown that plasma
`levels of dUrd are correlated with the degree of thymidylate
`synthase inhibition. We had planned to measure dUrd levels in
`
`this study as a measure of thymidylate synthase inhibition;
`unfortunately, plasma interference prevented analysis of dUrd in
`samples obtained.
`In this study, we observed disease stabilization of colorectal
`and ovarian cancers at a dose of 3 mg/d given for 28 days
`followed by a 2-week rest period. The results from this study
`indicate that phase II studies of ZD9331 are warranted in
`metastatic colorectal and ovarian cancer.
`
`REFERENCES
`
`1. Hillman RS: Hematopoietic agents: Growth factors, minerals and
`vitamins, in Hardman JG, Limbird LE, Molinoff PB, et al (eds): Goodman
`and Gilman’s Pharmacological Basis of Therapeutics (ed 9). New York, NY,
`McGraw-Hill, 1996, pp 1311-1340
`2. Vest S, Bork E, Hansen HH: A phase I evaluation of N10-propargyl-
`5,8-dideazafolic acid. Eur J Cancer Clin Oncol 24:201-204, 1988
`3. Jodrell DI, Newell DR, Morgan SE, et al: The renal effects of
`N10-propargyl-5,8-dideazafolic acid (CB3717) and a non-nephrotoxic ana-
`logue ICI D1694, in mice. Br J Cancer 64:833-838, 1991
`4. Clarke SJ, Hanwell J, de Boer M, et al: Phase I trial of ZD1694, a new
`folate-based thymidylate synthase inhibitor, in patients with solid tumors.
`J Clin Oncol 14:1495-1503, 1996
`5. Blackledge G: New developments in cancer treatment with the novel
`thymidylate synthase inhibitor raltitrexed (Tomudex). Br J Cancer 77:29-37,
`1998 (suppl)
`6. Jackman AL, Boyle FT, Harrap KR: Tomudex (ZD1694): From
`concept to care, a programme in rational drug discovery. Invest New Drugs
`14:305-316, 1996
`7. Pizzorno G, Mini E, Coronnello M, et al: Impaired polyglutamylation
`of methotrexate as a cause of resistance in CCRF-CEM cells after short-term,
`high-dose treatment with this drug. Cancer Res 48:2149-2155, 1988
`8. Rhee MS, Wang Y, Nair MG, et al: Acquisition of resistance to
`antifolates caused by enhanced gamma-glutamyl hydrolase activity. Cancer
`Res 53:2227-2230, 1993 (suppl)
`9. Jackman AL, Kimbell R, Brown M, et al: The antitumour activity of
`ZD9331, a non-polyglutamatable quinazoline thymidylate synthase inhibitor.
`Adv Exp Med Biol 370:185-188, 1994
`10. Jackman AL, Kimbell R, Aherne GW, et al: Cellular pharmacology
`and in vivo activity of a new anticancer agent, ZD9331: A water-soluble,
`
`nonpolyglutamatable, quinazoline-based inhibitor of thymidylate synthase.
`Clin Cancer Res 3:911-921, 1997
`11. Goh BC, Ratain MJ, Bertucci D, et al: Phase I study of ZD9331 on
`short daily intravenous bolus infusion for 5 days every 3 weeks with fixed

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket