throbber
ICANCER RESEARCH 50, 1220-1225, February 15. 1990]
`
`Phase I and Pharmacokinetic
`NSC 281272)
`
`Study of Arabinofuranosyl-5-azacytosine
`
`(Fazarabine,
`
`Antonella Surbone, Harry Ford, Jr., James A. Kelley, Noa Ben-Baruch, Rose V. Thomas, Robert Fine, and
`Kenneth H. Cowan1
`
`Medicine Branch ¡A.S., N. B-B., R. V. T., R. F., K. H. C.J and Laboratory of Medicinal Chemistry [H. F., J. A. K.J, National Cancer Institute, Bethesda, Maryland 20892
`
`ABSTRACT
`
`(ara-
`A Phase I clinical trial of l-i8-D-arabinofuranosyl-5-azacytosine
`AC or fazarabine) given as a 72-h continuous infusion on a 21-day cycle
`was conducted in 27 adult patients with refractory cancer. The major
`toxicity was reversible granulocytopenia and thrombocytopenia. Dose-
`limiting toxicity was observed at a dose rate of 1.96 mg/mz/h in which
`Grade IV leukopenia (WBC < 1.0(1(1,mm')occurred in 4 of 11 patients
`and Grade IV thrombocytopenia (platelets <25,000/mm') occurred in 3
`of 11 patients. Plasma steady-state levels ranged from 0.13 to 0.6 /JMfor
`doses of 1.25 to 5.94 mg/m2/h. Mean total body clearance was 647 ml/
`min nr. Minor clinical responses were seen in one patient with testicular
`cancer, one patient with colon cancer, one patient with breast cancer, and
`one patient with acute nonlymphocytic leukemia. Another patient with
`adenocarcinoma of unknown primary had stable disease during 13 cycles
`of therapy. Based on the results of this study, the recommended dose for
`Phase II studies of l-/9-r>-arabinofuranosyl-5-azacytosine administered
`as a 72-h continuous infusion is 2.0 mg/m2/h (48 mg/m2/day).
`
`to 5-AC (which undergoes intracellular phosphoryla
`resistant
`tion by this enzyme) but sensitive to both ara-C and ara-AC
`(10).
`Although the mechanism of ara-AC cytotoxicity is not well
`understood,
`this analogue is incorporated into cellular DNA in
`a dose- and time-dependent manner
`(5, 6, 11). Furthermore,
`ara-AC inhibition of DNA synthesis
`in LI210 cells occurs at
`drug doses that do not affect RNA or protein synthesis. Thus,
`the mechanism of ara-AC cytotoxicity,
`like that of ara-C,
`is
`apparently related to its ability to inhibit DNA synthesis.
`One of the most interesting features of ara-AC is its relatively
`broad spectrum of activity in both in vivo and in vitro preclinical
`screens (8, 12-15).
`In contrast
`to both parent compounds,
`ara-
`C and 5-AC, which show little if any activity against
`solid
`tumors, ara-AC demonstrated
`significant activity in a National
`Cancer Institute panel of human tumor xenografts.
`In addition
`to the above-mentioned
`activity against human and mouse
`leukemia cell
`lines, ara-AC displayed activity against human
`colon,
`lung, breast, and ovarian cancer xenografts.
`studies have
`Preclinical pharmacokinetic
`and toxicological
`been performed on both mice and dogs (12, 15) and limiting
`toxicities appeared to be myelosuppression
`and gastrointestinal
`distress, with the occurrence of bloody diarrhea
`and emesis
`(15). Other
`toxicities
`that were not dose limiting included
`lethargy,
`tremors,
`and ataxia. Histological
`evidence of drug-
`related toxicity included bone marrow depletion,
`lymphoid
`organ depletion, necrosis of gastrointestinal
`epithelium,
`and
`maturation arrest with necrosis of the seminiferous
`tubules of
`the testis (15).
`activity, we
`its broad spectrum of preclinical
`Because of
`initiated a Phase I clinical study of this agent. Preclinical studies
`indicated that
`the activity of this agent
`is schedule dependent
`and that continuous
`infusion and frequent
`intermittent
`admin
`istration are apparently more effective than single-dose sched
`ules (12-15). Thus,
`in this Phase I trial ara-AC was adminis
`tered as a 72-h infusion to patients with clinically refractory
`cancer.
`
`MATERIALS AND METHODS
`
`INTRODUCTION
`ara-AC2 (fazarabine; NSC 281272)
`is a synthetic pyrimidine
`features of two effective
`nucleoside containing
`the structural
`antineoplastic
`agents, ara-C and 5-AC (Fig. 1). ara-AC com
`bines the arabinose sugar of ara-C (with its inverted hydroxyl
`group at
`the 2'-position) with the triazine base of 5-AC,
`in
`which nitrogen
`is substituted
`for
`the C-5 of cytidine. This
`triazine ring is highly susceptible to reduction and nucleophilic
`reactions. Thus, ara-AC,
`like 5-AC, undergoes decomposition
`in aqueous solution to inactive products
`through the addition
`of water to its 5-6 double bond with concomitant
`ring opening.
`In contrast, when ara-AC is dissolved in organic solvents such
`as DMSO,
`its stability is markedly enhanced and there is little
`hydrolysis of the triazine ring (3, 4).
`ara-AC,
`like ara-C, apparently enters cells via a nucleoside
`transport mechanism and is subsequently
`activated through
`phosphorylation
`by deoxycytidine
`kinase (5, 6). Degradative
`pathways
`for both ara-AC and ara-C involve phosphatases
`which can cleave phosphate groups,
`from the arabinofuranosyl
`moiety. While ara-C can be converted to an inactive metabolite,
`l-/3-D-arabinofuranosyluridine,
`by cytidine deaminase (7), ara-
`AC is a poor substitute for this enzyme and is relatively refrac
`tory to inactivation by deamination (5).
`The similarity in activation via deoxycytidine kinase suggests
`that cross-resistance between the two nucleoside analogues, ara-
`C and ara-AC, can occur (8, 9). Indeed, P388 murine leukemia
`cells that are resistant
`to ara-C because of a decrease in deoxy
`cytidine kinase activity are also cross-resistant
`to ara-AC (9).
`In contrast, HL-60 cells deficient
`in uridine/cytidine
`kinase are
`
`revised 10/20/89; accepted 11/9/89.
`Received 4/17/89;
`The costs of publication of this article were defrayed in pan by the payment
`of page charges. This article must
`therefore be hereby marked advertisement
`in
`accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
`' To whom requests for reprints
`should be addressed, at Medicine Branch.
`National Cancer Institute. NIH, Bldg. 10, Rm. 12N226. Bethesda. MD 20892.
`2The abbreviations used are: ara-AC,
`l-#-D-arabinofuranosyl-5-azacytosine;
`ara-C,
`I-0-D-arabinofuranosylcytosine;
`5-AC, 5-azacytidine; HPLC, high-per
`formance liquid chromatography; C„plasma steady-state concentration; DMSO,
`dimethyl sulfoxide.
`
`I.
`are shown in Table
`characteristics
`Patient Selection. Patient
`ranging in age from
`Twenty-seven (26 évaluablefor toxicity) patients,
`25 to 72 years were entered into the study. All patients had a histolog-
`ically proved diagnosis of malignant disease with definitive evidence of
`metastatic spread and/or of inoperable local recurrence. One patient
`with treatment-induced
`acute nonlymphoblastic
`leukemia was given
`ara-AC under a special exemption;
`this patient was included in the
`pharmacokinetic
`evaluation but not in the toxicity evaluation since he
`was neutropenic and thrombocytopenic before entering this study. The
`patients in this study were, in general, heavily pretreated. All but four
`patients had previously received chemotherapy.
`In addition to chemo
`therapy,
`13 patients
`(48%) were treated previously with radiation
`therapy.
`Prior to beginning treatment with ara-AC, each patient underwent a
`comprehensive evaluation including complete history, physical exami
`nation, and evaluation of measurable disease by appropriate
`radio
`CELGENE 2121
`1220
`APOTEX v. CELGENE
`IPR2023-00512
`
`© 1990 American Association for Cancer Research.
`
`

`

`PHASE I ARA-AC (FAZARABINE)
`
`resulting in a final ara-AC concentra
`in water (Tera Pharmaceuticals)
`tion of 25 mg/ml. A 24-h supply of drug was diluted to a final volume
`of 12 ml with 70% DMSO and placed in a 12-ml Luer-lock syringe
`(Monoject, Model 512936). The syringe was replaced every 24 h with
`one containing freshly reconstituted drug solution. The drug was deliv
`ered by an Autosyringe infusion pump (Travenol Laboratories; Model
`AS2F) as a "piggyback" into an i.v. solution of 5% dextrose in water
`and infused at 20 ml/h. A polyolefin-lined extension set (Pacesetter
`Infusion, Ltd.; Model 126) was used to connect
`the syringe to the i.v.
`set. This diluted solution of ara-AC was administered through a pe
`ripheral vein or a central
`line.
`Treatment Plan. The starting dose was 0.2 mg/m2/h administered as
`a continuous infusion over 72 h. This starting dose was determined on
`the basis of the tolerated dose in mice during 72-h continuous infusion
`(10 mg/m2/h) with an additional consideration given to the potential
`increased metabolic activation of this drug in humans. Some studies
`suggest
`that human cells may contain higher
`levels of activity of
`deoxycytidine kinase,
`the enzyme that converts ara-AC to its toxic
`metabolites (15). Indeed, previous clinical experience had shown that
`fludarabine, an agent
`that
`is also converted to toxic species by deoxy
`cytidine kinase, was considerably more toxic in humans that
`it was in
`mice and other animals (15). Based on this,
`the starting dose of ara-
`AC in this Phase I clinical study was chosen to be 0.2 mg/m2/h for 72
`h. Dose escalation was allowed in individual patients. A minimum of
`three patients were treated at each dose level before escalation to the
`subsequent dose level was permitted in new patients. The ara-AC dose
`was initially escalated to 0.4 mg/m2/h and then to 0.8 mg/m2/h for 72
`h with subsequent doses being escalated by 25% of the previous dose
`level (as indicated in Table 3). ara-AC was administered every 21 days
`if the patients had recovered from toxicity of the previous cycle;
`otherwise, administration of the next dose was delayed for 1 week. The
`objective of the study was to determine the maximally tolerated dose
`of ara-AC when given as a 72-h continuous
`infusion and to determine
`the pharmacokinetics of this agent in humans. The maximally tolerated
`dose was defined as the dose at which greater than 50% of the patients
`treated at a given dose experienced Grade III or greater
`toxicity.
`Toxicity was graded according to the guidelines recommended by the
`Cancer Therapy Evaluation Program, National Cancer Institute.
`In Vitro Stability. An appropriate volume of 1.0 x 10~3M ara-AC in
`
`DMSO was added to fresh heparinized human plasma (pH 7.6) from a
`normal volunteer and to 0.1 M phosphate-buffered saline solution (pH
`7.2) at 37°Cto give a final ara-AC concentration of 1 Mg/m' (4.1 JUM).
`A 0.5-ml aliquot was taken from each sample before incubation (t = 0),
`and the remainder of the sample was then incubated at 37 ±0.2°C.At
`predetermined intervals 0.5-ml aliquots were removed from each sam
`ple, and ara-AC concentrations were measured by the method described
`below. Sampling was continued for a minimum of three half-lives.
`Pharmacokinetic Studies. Blood samples were obtained in 10 ml
`heparinized tubes both prior to administration
`of ara-AC and at 24-h
`intervals after the initiation of therapy. Each sample was maintained
`on ice to retard hydrolysis of the parent drug, and plasma was separated
`from cells as soon as possible by centrifugation at 1000 x g for 10 min.
`Because of limited sample stability, an aliquot of plasma was processed
`and analyzed within 4 h using a modification of a reverse-phase high-
`performance liquid chromatography assay (16).
`Briefly, 2 ¿ilof 2.25 x IO"3 M (1.0 ¿ig)2'-deoxy-5-azacytidine were
`added to a 0.5-ml aliquot of plasma as an internal standard. The sample
`was vortexed for 10s to mix the internal standard. A 1.0-ml phenyl-
`boronic acid solid-phase extraction cartridge (Bond Elut PBA; Analy-
`tichem International, Harbor City, CA) was activated by washing with
`1 ml of methanol
`followed by 1 ml of 0.01 M phosphate buffer, pH 8.O.
`Attached to this by an adaptor was a 3.0 ml disposable cartridge to
`which the plasma sample was transferred. The sample was then slowly
`pushed through the cartridge and the eluant was collected. The cartridge
`was rinsed with 0.5 ml of 0.01 M phosphate buffer, pH 8.0, and this
`was then ultrafiltered by centrifugation at 1000 x g for 30 min at 4°C
`in a Centrifree partition system (Amicon Corp., Danvers, MA).
`A 100-fil aliquot was analyzed using a Model 204W liquid chroma
`tography system (Walters Associate, Milford, MA) consisting of a U6K
`injector, a Model 6000 A solvent delivery system, and a Model 116
`1221
`
`Ara-C
`5-AC
`Fig. l. Chemical structures of S-AC, Ara-C. Ara-AC. D, changes in structure
`compared to cytidine.
`
`Ara-AC
`
`27
`26
`82(79)
`
`52
`25-74
`
`17/10
`82
`
`24
`10
`1
`12
`
`Table 1 Patient characteristics
`No. of patients entered
`No. évaluable
`Total no. of cycles (évaluable)
`
`Age. yr
`Mean
`Range
`
`Male/female
`Mean performance status (Karnofsky)
`
`No. of patients with prior therapy
`Chemotherapy only
`Radiation therapy only
`Chemotherapy and radiation therapy
`
`Diagnosis
`Acute nonlymphocytic leukemia
`Adenocarcinoma
`liver, unknown primary
`Adrenal cancer
`Breast cancer
`Colon cancer
`Diffuse lymphoma
`Embryonal cell carcinoma
`Fibrohistiocytoma
`Glioma
`Nodular lymphoma
`Non-small cell lung cancer
`Osteogenic cancer
`Ovarian cancer
`Pancreatic carcinoma
`Renal cell carcinoma
`Small cell lung cancer
`Squamous cell cancer, anus
`Uterine sarcoma
`
`graphic studies. Patients in this study had to have a performance status
`of greater than 50% on a Karnofsky scale. Pretreatment evaluation also
`included complete blood cell count, with WBC differential,
`serum
`chemistries, coagulation studies, serum creatinine, and creatinine clear
`ance. All patients had adequate liver (bilirubin <1.5 mg/100 ml and
`serum transaminases
`less than twice the normal
`range) and renal
`(creatinine clearance of 45 mi/min or a serum creatinine <1.5 mg/100
`ml) function, as well as normal serum electrolytes and normal coagu
`lation studies. All patients evaluated for toxicity had adequate periph
`eral blood cell counts (WBC > 3,000/mm3, platelets > 100,000/mm')
`prior
`to initiation of study. Complete blood cell counts and liver
`function tests were done weekly while the patients received therapy and
`they were seen at least once every 3 weeks prior to a new cycle at which
`time all laboratory tests were repeated. At each visit, patients were
`asked to report and subjectively rate every possible drug-related side
`effect. All patients gave written informed consent prior to therapy in a
`manner consistent with institutional guidelines. Patients who developed
`progressive disease after two cycles were removed from study.
`Drug Formulation and Dosage. ara-AC was supplied by the Division
`of Cancer Treatment, National Cancer Institute (Bethesda, MD), as a
`sterile lyophilized powder in 250-mg vials. Because of limited aqueous
`stability,
`the vials were reconstituted with 9.9 ml of 70% (v/v) DMSO
`
`© 1990 American Association for Cancer Research.
`
`

`

`PHASE l ARA-AC (FAZARABINE)
`
`Table 2 Pharmacokinetic parameters of ara-AC
`Clearance*
`(ml/min/m2)
`
`Dose
`(mg/mVh)
`1.25
`1.75
`2.44
`3.83
`4.75
`5.94
`
`(ng/ml)
`32
`50
`57
`71
`108
`137
`
`651
`583
`713
`899
`733
`729
`
`Cycle
`
`578
`
`10
`II
`13
`
`Patient
`W. P.
`
`33127
`
`1.962.441.96
`
`724761627l
`
`1.962.96
`
`527695
`
`
`
`1143122111.961.562.441.562.441.961.962.442.442.447437
`6270455654685862441703454865536
`65637190458360S598701656
`
`Q.W.J.
`
`F.R.
`
`D.M.
`
`T.K.
`
`S.E.W.V.
`
`N.P.S.D.
`
`W.H.W.T.
`
`S.R.
`
`J.32
`°Measured values of 30-69 ng/ml are in the range of less-certain quanlitation.
`" Mean ±SD. 647 ±141 (N = 21).
`
`V = 3 146 + 21.7X
`r = 0.9660
`
`150
`
`100
`
`1
`
`2345
`
`6
`
`-
`E
`
`UV detector (Gilson Medical Electronics. Middleton, WI). The sample
`was separated on a 4.6- x 250-mm, 5-^m Ultrasphere octadecylsilane
`analytical column (Altex/Beckman. Berkeley. CA) with a mobile phase
`of 0.5% CHjCN in 0.01 M phosphate buffer. pH 6.8. with a flow rate
`of l ml/min. The detector
`signal was integrated by a dual channel
`Spectra-Physics 4200 computing and recording integrator, which was
`interfaced with a ChromStation-AT data system (Compaq 386 version;
`Spectra-Physics, Santa Clara, CA) for further data reduction and stor
`age.
`Standard curves of ara-AC in plasma were prepared for each patient's
`samples by addition of know n amounts of ara-AC to the corresponding
`pretreatment
`sample. These spiked standards were processed in the
`same manner as above. A standard curve was prepared for analysis each
`day for the range 0 to 400 ng/ml. This cune was the best straight
`line
`defined by least squares regression analysis and possessed a correlation
`coefficient >0.997. Ratios of peak height of ara-AC standard to peak
`height of internal
`standard were obtained for a minimum of four
`concentrations each day. Samples were run concurrently, and concen
`trations were calculated from the appropriate
`standard curve. ara-AC
`peaks below the automatic detection threshold of the integrator were
`measured using the data system software. UV detection at 240 nm
`allowed a limit of quantitation (S/N >5) of 70 ng/ml
`(0.3 MM)and a
`limit of detection of 30 ng/ml
`(0.12 //M). ara-AC concentrations
`be
`tween these two limits could also be estimated; but, since these values
`have a greater error,
`they are indicated as falling in the range of less-
`certain quantitation (17) in Table 2.
`Kinetic Calculations. For determination of in vitro stability, half-lives
`were calculated from the best straight
`line obtained by linear regression
`analysis of the logarithm of ara-AC concentration versus time. C«was
`the average of measured 24-h ara-AC plasma concentrations,
`since ara-
`AC levels were observed to reach a plateau by 8 h in patients given
`higher doses on a 24-h infusion schedule (16). The apparent
`total body
`clearance was then defined as the rate of infusion divided by Ca.
`
`RESULTS
`
`In Vitro Stability. ara-AC was unstable in both phosphate-
`buffered saline and human plasma at the clinically achievable
`concentration
`of 1 pg/m\
`(4.1 ^M) (18). The disappearance
`of
`ara-AC in these in vitro experiments was apparent
`first order,
`with the half-life of 2.5 ±0.2 h (n = 2) in fresh human plasma
`incubated at 37°Cbeing much more rapid than the 9.8 ±0.3 h
`(n = 3) observed during incubation in phosphate-buffered
`saline
`under the same conditions.
`Pharmacokinetics.
`ara-AC was measured in the plasma of 13
`patients receiving 21 cycles of therapy at doses above 1.25 mg/
`nr/h. Css ranged from 32 ng/ml
`(0.13 //M) to 137 ng (0.6 JUM)
`(Table 2). For Patient W. P., who received 13 cycles of therapy
`and had the most extensive blood level study, a good linear
`relationship of measured Css with dose was calculated (Fig. 2).
`A relatively rapid mean clearance of 647 ml/min/m2 was ob
`served for all patients (Table 2).
`Toxicity. Twenty-six évaluablepatients were treated with a
`total of 79 cycles of ara-AC. The dose-limiting
`toxicity was
`reversible leukopenia
`and thrombocytopenia
`(Table 3). Grade
`IV leukopenia was observed in 1 of 7 patients
`(1 of 7 cycles)
`given ara-AC at 1.25 mg/nr/h,
`in 1 of 8 patients
`(2 of 11
`cycles) at 1.56 mg/nr/h,
`and in 4 of 11 patients (5 of 14 cycles)
`at 1.96 mg/nr/h. Overall, Grade III or Grade IV leukopenia
`occurred in 6 of 11 patients treated at 1.96 mg/nr/h
`and in 4
`of 7 treated at 2.44 mg/nr/h.
`This was not a reflection of
`cumulative drug toxicity,
`inasmuch as 2 of 4 new patients who
`entered treatment with ara-AC at 1.96 mg/nr/h
`and 4 of 4 new
`patients who entered at 2.44 mg/nr/h
`experienced Grade III
`or IV neutropenia. Thrombocytopenia
`also occurred but overall
`was less prominent
`than leukopenia. Grade IV thrombocyto
`penia was observed in 1 of 7 patients (1 of 7 cycles) treated at
`1222
`
`Dose (mg/m?/hl
`(ng/ml or UM)at
`Fig. 2. Correlation between ara-AC plasma concentrations
`steady state and dose (mg/m2/h)
`in a single patient (W. P.) given multiple cycles
`of therapy.
`
`1.25 mg/m2/h, 1 of 8 patients (2 of 11 cycles) treated with 1.56
`mg/nr/h,
`and 3 of 11 patients (3 of 14 cycles) treated with 1.96
`mg/nr/h
`for 72 h. The median time to WBC nadir was 18 days
`(range, 7 to 31), while the median time to the corresponding
`platelet nadir was 13 days (range, 7 to 22).
`There were five episodes of fever and neutropenia
`
`requiring
`
`© 1990 American Association for Cancer Research.
`
`

`

`PHASE [ ARA-AC (FAZARAB1NE)
`
`Table 3 Hemalological
`
`loxicily of ara-AC
`nadir*IV111411I33113II
`
`of
`patients
`of
`Dose
`III12ii31IV113
`
`(new/total)3/33/53/61/64/74/84/114/70/30/10/10/1No.II111
`cycles357771114174112WBCI
`(mg/inVh)0.20.40.81.01.251.561.962.443.053.834.755.94No.
`
`21
`22
`211nadir"III222323Platelet
`
`leukocytes/mm3: Grade I 3.000-3.999; Grade II. 2.000-2.999; Grade
`"Total
`III, 1,000-1,999; Grade IV, <1,000.
`* Platelets/mm3: Grade 1, 75,000-90,000: Grade II. 50.000-74.000; Grade III,
`25,000-49,000; Grade IV, <25,000.
`
`during this study.
`treatment
`for i.v. antibiotic
`hospitalization
`In three cases no source of infection was detected. One patient
`developed Candida and herpes esophagitis during the period of
`fever and neutropenia. One patient with adrenocortical
`cancer
`developed Listeria meningitis
`at which time the WBC was
`1400/mm3. She was treated successfully and recovered without
`sequelae. Another patient became neutropenic during his 12th
`cycle of ara-AC therapy. He subsequently developed staphylo-
`coccal septicemia
`and Candida
`endocarditis
`on a prosthetic
`mitral valve (he had a previous history of Candida endocarditis).
`Although his WBC returned to normal, he subsequently died
`of cardiac complications. This was the only treatment-related
`death on the study.
`Increases in liver function tests were observed in 14 patients
`treated with ara-AC. This was predominantly manifested by
`increases in alkaline phosphatase although there were also some
`minor increases in transaminases. Overall,
`there was no appar
`ent relationship between ara-AC dose and liver toxicity. Grade
`I liver toxicity (increased alkaline phosphatase
`or
`transami
`nases) was seen in 19 cycles (occurring in doses ranging from
`0.4 to 2.44 mg/m2). Grade II in 13 cycles (in doses ranging
`from 0.8 to 5.94 mg/m2), Grade III in one cycle (at 1.56 mg/
`m2), and Grade IV toxicity in 2 cycles (at 1.96 mg/m2). In each
`case,
`the hepatic abnormality was transient
`and returned to
`baseline prior to the next cycle. One-half (7 of 14) of the patients
`who developed elevated liver function tests in this study had
`evidence of liver involvement with tumor,
`thus complicating
`the evaluation for hepatotoxicity.
`There were few other side effects reported in this trial. Since
`ara-AC was reconstituted
`in DMSO in order
`to enhance
`its
`stability during prolonged infusions,
`the odor of DMSO was
`noticeable to everyone who entered the patients'
`rooms. How
`ever, the patients themselves did not complain of the odor, nor
`did they complain of any garlic-like taste. Nausea and vomiting
`were uncommonly
`(7 of 27 patients)
`reported
`in this trial.
`Overall,
`there were only 5 episodes of Grade I and 2 Grade II
`nausea and vomiting. One patient
`received a total of 13 cycles
`with an escalation from 0.2 mg/m2/h to 5.94 mg/m2/h for 72
`h. This 44-year-old patient complained
`of sleep disturbances
`(nightmares
`and excessive tiredness) beginning after Cycle 4
`and of
`impotence
`after Cycle 6. These toxicities were not
`observed in any other patient.
`Clinical Activity. In this Phase I study, both prolonged disease
`stabilization in one patient and minor clinical responses in four
`other patients were observed. One patient
`(W. P.) with an
`adenocarcinoma
`of the liver of unknown primary had stable
`disease without any evidence of progression by abdominal com
`
`scan during 13 cycles of therapy
`tomographic
`puterized axial
`with ara-AC. Of the four patients who were noted to have minor
`clinical
`responses, one (E. W.) entered the study with rapidly
`progressing liver métastasesfrom colon carcinoma. Following
`two cycles of ara-AC the size of the liver as noted by physical
`examination
`diminished
`from 28 cm to 19 cm. This patient
`developed disease progression in the retroperitoneum after four
`cycles of therapy. One patient
`(V. L.) with metastatic breast
`cancer had some shrinkage in the size and character of extensive
`chest wall
`lesions after
`three cycles of ara-AC but developed
`progressive disease after the fourth cycle. One patient
`(T. R.)
`previously treated with chemotherapy and radiation therapy for
`Hodgkin's disease had developed acute nonlymphocytic
`leuke
`mia. Following treatment with ara-AC,
`there was a diminution
`in the number of circulating blasts, a decrease in láclatedehy-
`drogenase,
`a decrease
`in fever, and a feeling of subjective
`improvement. After two cycles of therapy,
`there was a complete
`disappearance
`of biopsy-proved
`leukemic skin lesions. How
`ever, bone marrow biopsies failed to show a decrease in the
`proportion of blasts, and the patient's
`therapy was stopped after
`three cycles.
`cell
`(K. S.) in this study had embryonal
`Another patient
`carcinoma
`and had been previously treated with two different
`salvage combination
`chemotherapy
`regimens. At
`the time of
`entering this trial,
`this patient had bilateral adrenal masses, a
`testicular mass, pleural effusions, and ascites which required
`twice-weekly paracentesis. After
`the first cycle of ara-AC, he
`no longer required paracentesis, and after the fourth cycle, there
`was no evidence of ascites by abdominal
`computerized
`axial
`tomographic
`scan. He was subsequently treated with an addi
`tional 9 cycles of ara-AC. Despite clinical
`improvement
`there
`was no apparent change in the size of his adrenal or testicular
`masses and his «-fetoprotein increased during this time from
`600 to >11,000. An orchiectomy was performed which showed
`an embryonal cell carcinoma with yolk sac elements, which had
`not been a prominent
`feature of his previous biopsies.
`It
`is
`possible that ara-AC promoted the differentiation of the tumor
`toward a yolk sac tumor
`resulting in enhanced «-fetoprotein
`production.
`Indeed, ara-AC does induce differentiation of HL-
`60 cells (8), a property presumed to be related to its ability to
`inhibit DNA methylation (11). It is also possible that the testes
`represented a sanctuary site. Following orchiectomy,
`this pa
`tient's a-fetoprotein
`returned to normal. The patient developed
`neutropenia
`during a subsequent
`cycle of ara-AC which was
`complicated by staphylococcal
`sepsis and Candida endocarditis
`on a prosthetic mitral valve (the patient had developed Candida
`endocarditis
`during earlier
`treatments
`for testicular
`cancer).
`The patient
`subsequently
`died of cardiac complications. At
`autopsy,
`there were only necrotic masses in the abdomen and
`adrenals without evidence of tumor. He did, however, have two
`small sites of residual embryonal cancer in his lung.
`
`DISCUSSION
`
`In this Phase I trial, ara-AC was given as a 72-h infusion to
`adult patients with refractory cancer. The dose-limiting toxicity,
`which was reversible leukopenia, was reached at 1.96 mg/nr/h
`for 72 h in this heavily pretreated patient population. Neutro
`penia was more prominent
`than thrombocytopenia. Other
`tox
`icities included mild elevations in liver function abnormalities.
`Hepatotoxicity was difficult
`to evaluate in this patient popula
`tion since many of the patients had metastatic disease in the
`liver. Nausea and vomiting were not prominent
`side effects of
`this drug, despite the obvious odor produced by the DMSO
`1223
`
`© 1990 American Association for Cancer Research.
`
`

`

`PHASE l ARA-AC (FAZARABINE)
`
`REFERENCES
`
`of
`10 months) was seen in one patient with adenocarcinoma
`unknown origin involving the liver. Minor clinical
`responses
`were observed in one patient with acute nonlymphocytic
`leu
`kemia,
`in one patient with colon cancer metastatic to liver, in
`one patient with breast cancer, and in one patient with em
`bryonal
`testicular cancer. This wide range of possible clinical
`activity mirrors the breadth of observed preclinical activity.
`ara-AC is thus a new antineoplastic
`agent which appears to
`combine the chemical and clinical features of ara-C and 5-AC.
`The 70% DMSO formulation
`of ara-AC is well tolerated by
`patients when it
`is piggy-backed into a rapidly dripping i.v.
`solution and given continuously for 72 h. Although all of our
`patients have been treated in the hospital,
`it should be possible
`to deliver ara-AC as an outpatient
`regimen. The only limiting
`feature to outpatient
`therapy may be the noxious odor produced
`by the DMSO. The recommended dose for Phase II studies in
`heavily pretreated
`patients
`is 2.0 mg/nr/h
`for 72 h. Higher
`dose levels should be tolerated by individuals who have received
`little or no prior chemotherapy.
`Preclinical
`studies and this
`Phase I study suggest
`that ara-AC may have a broad spectrum
`of activity against human solid tumors.
`
`which was required for the effective formulation of the clinical
`dose.
`The mean clearance for the 21 cycles of therapy for which
`ara-AC could be measured was 647 ±141 ml/min/nr,
`which
`is not statistically different
`from the mean clearance (571 ml/
`min/nr) observed in the Phase I pediatrie trial of this agent for
`a dose of 15 mg/m2/h over 24 h (18). These clearances also fell
`into the 480-
`to 1157-ml/min/m2
`range observed in non human
`primates
`for ara-AC doses of 200 mg/kg given over either 15
`min or l h (19). This rapid clearance of ara-AC is about one-
`half that of ara-C (20), the nucleoside that ara-AC most closely
`resembles biochemically. While ara-C is rapidly metabolized by
`cytidine deaminase, araAC is refractory to catabolism by this
`enzyme (5). Thus, any clearance of ara-AC through deamina-
`tion, even considering the high levels of this enzyme in human
`liver and kidney (21), is expected to be minimal as is the case
`for 2',3'-dideoxycytidine,
`another cytosine nucleoside which is
`a poor substrate for mammalian cytidine deaminase (22). Based
`on in vitro stability, however, ara-AC probably undergoes sub
`stantial hydrolytic degradation in vivo. If one assumes a steady-
`state volume of distribution equivalent
`to total body water (42
`liters for a 70-kg person), which is what has been observed for
`ara-AC in monkeys (19), a maximum hydrolytic contribution
`to plasma clearance of 110 ml/min/m2 can be calculated (23).
`This indicates that other as yet to be defined processes such as
`1. Beisler. J. A., Abbasi, M. M.. and Driscoll, J. S. The synthesis and antitumor
`activity of arabinosyl-5-azacytosine. Biochem. Pharmacol.. 26: 2469-2472,
`distribution into other compartments
`(e.g., intracellular
`trans
`1977.
`port),
`renal excretion,
`and anabolic metabolism may account
`2. Beisler, J. A., Abbasi, M. M., and Driscoll, J. S. Synthesis and antitumor
`activity of 5-azacytosine arabinoside. J. Med. Chem., 22: 1230-1234, 1979.
`for the majority of plasma clearance.
`3. Mojaverian, P., and Repta, A. J. Development of an intravenous formulation
`Steady plasma levels of ara-AC ranged from 32 to 137 ng/
`for the unstable investigational cytoloxic nucleosides S-azacytosine arabino
`ml (0.13 to 0.6 MM)for patients receiving drug at rates between
`side (NSC 281272) and 5-azacytidine (NSC 102816). J. Pharm. Pharmacol.,
`Jo: 728-733, 1984.
`1.25 and 5.94 mg/nr/h
`(Table 2; Fig. 2). Studies with ara-AC
`4. Notar!, R. E., and DeYoung, J. L. Kinetics and mechanisms of degradation
`in several
`in vitro systems suggest
`that
`these observed concen
`of the antileukemic agent S-azacytidine in aqueous solutions. J. Pharm. Sci.,
`64: 1148-1156, 1975.
`trations may be sufficient
`for biological activity, especially if
`5. Townsend, A., Ledere, J. M., Dutschman, G., Cooney, D. A., and Cheng,
`they are maintained for the better part of 72 h. Incubation of
`Y. C. Metabolism of l-$-r>arabinosyI-5-azacytosine
`and incorporation into
`Molt-4 human T-lymphoblastic
`leukemia cells with 1.0 ^M ara-
`DNA of human T-lymphoblastic cells (Molt-4). Cancer Res., 45:3522-3528,
`1985.
`AC for 24 h resulted in 98% inhibition in clonogenic assays
`6. Vesely, J., and Piskala, A. Mechanism of action of 1-0-D-arabinofuranosyl-
`(5). Although a direct comparison is not possible because drug
`5-azacytosine and its effects in LI 210 leukemia cells. Neoplasma, 33: 3-10,
`concentrations with respect
`to time are not known for the in
`1986.
`In: B. A. Chabner (éd.).Pharmacologie
`7. Chabner, B. A. Cytosine arabinoside.
`vitro study, drug exposure was probably similar in both cases.
`Principles of Cancer Treatment, pp. 387-401. Philadelphia: W. B. Saunders
`If one takes into account
`the half-life of ara-AC in RPMI 1640
`Co., 1982.
`(19),
`then the average drug concentration
`for
`the 24 h of
`8. Dalai, M., Plowman, J., Breitman, T. R., Schuller, H. M., Del Campo, A.
`A., Vistica, D., Cooney, D. A., and Johns, D. G. Arabinofuranosyl-5-
`incubation is 0.2 ¿ÕM,a level that should be achievable at
`the
`azacytosine: antitumor and cytotoxic properties. Cancer Res., 46: 831-838,
`recommended
`dose of 2.0 mg/m2/h for Phase II studies.
`In
`1986.
`J.
`9. Ahluwalia, G. S., Cohen, M. B., Kang, G.-J., Arnold, S. T., McMahon,
`addition,
`the inhibitory concentration
`for 50% control growth
`B., Dala, M., Wilson, Y. A., Cooney, D. A., Balzarini, J., and Johns, D. G.
`for a 24-h exposure of ara-AC to P388 murine leukemia in vitro
`Arabinosyl-5-azacytosine: mechanisms of native and acquired resistance.
`was found to be 1.9 ^M, and exposure of these same cells to 2
`Cancer Res., 46:4479-4485,
`1986.
`10. Grant, S., Bhalla, K., and Gleyzer, M. Effect of uridine on response of 5-
`fiM ara-AC for 3 day

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