throbber
REVIEW ARTICLE
`Oral Chemotherapy: Rationale and Future Directions
`
`By Mark D. DeMario and Mark J. Ratain
`
`Purpose and Methods: The expanding role of oral
`chemotherapy in oncology is suggested by the abun-
`dance of orally formulated agents currently in develop-
`ment. The pharmacoeconomic principles that drive oral
`drug formulation are discussed. Patient preference for
`oral therapy is identified as a second major impetus for
`the design of oral cytotoxics. While the rationale for oral
`formulations is apparent, substantial patient compli-
`ance and pharmacokinetic limitations have been identi-
`fied for this route of administration. Specific aspects of
`bioavailability limitations and patient compliance are
`discussed. Relevant pharmacokinetic data for each
`orally formulated chemotherapy agent are compared
`and selected novel oral cytotoxics and cytotoxic modu-
`lators are discussed.
`Results: A review of pharmacokinetic literature sug-
`gests substantial variability in bioavailability for many
`orally formulated cancer cytotoxics. While these find-
`ings are observed for all classes of oral drugs, the issue
`is especially critical for cancer chemotherapy, in which a
`narrow therapeutic index is frequently observed. Im-
`proved bioavailability and reduced interpatient biovari-
`
`AS WITH VIRTUALLY ALL office-based and academic
`
`medicine, the issue of cost-effectiveness must be
`addressed by the medical oncologist. Cost may no longer be
`considered as an aside, but will likely become a central issue
`in therapeutic decision-making, particularly in the limita-
`tions of the palliative setting. At the beginning of the next
`century, it is estimated that health care expenses will amount
`to $1.5
`trillion dollars or 15% of the projected Gross
`National Product.' In 1990, an estimated $35 billion was
`spent on direct costs for cancer care. 2 The direct cost of
`chemotherapy agents represented only a small fraction of
`this sum. A cost survey of one outpatient cancer center noted
`direct chemotherapy charges accounted for only 5.4% of
`total Medicare cost claims.2 In an analysis of adjuvant and
`metastatic breast cancer settings, direct drug charges ac-
`counted for only 19% to 36% of total care costs.3 These
`examples of the relatively small contribution of chemother-
`apy agents to the overall cost of oncology care are represen-
`tative of national health care expenditures as a whole. In
`1991, prescription drugs represented only 6.4% of total
`health care expenditures.4
`While cost-effectiveness has been substantiated for both
`adjuvant and palliative chemotherapy, 5-8 it is likely these
`therapies will continue to be scrutinized in terms of their
`component charges, primarily direct drug costs and adminis-
`
`ability are therefore desirable for new cytotoxic formu-
`lations. Pharmacologic manipulations to improve
`bioavailability and reduce costs are examined.
`Conclusion: Oral chemotherapy represents a funda-
`mental change in contemporary oncology practice,
`driven by pharmacoeconomic
`issues, patient conve-
`nience, and the potential for improved patient quality of
`life. Novel cytostatic therapies that require protracted
`drug administration periods will also favor an oral
`formulation. While the use of oral chemotherapy may
`initially be limited to metastatic disease palliation, dem-
`onstration of equivalent efficacy would allow for its
`subsequent use in adjuvant settings. This efficacy is
`contingent on circumventing bioavailability limitations
`and patient noncompliance. The development of spe-
`cific, low-toxicity inhibitors of CYP3A4, P-glycoprotein
`(P-gp), and other drug metabolizing enzymes such as
`dihydropyrimidine dehydrogenase represents a major
`innovative step in the successful formulation of oral
`chemotherapy.
`J Clin Oncol 16:2557-2567. © 1998 by American
`Society of Clinical Oncology.
`
`tration costs. Administration costs traditionally have incorpo-
`rated some or all of the following charges: hospitalization,
`physician fees, salaries of nursing and technical support
`personnel, infusion equipment supply costs, administration
`supply product fees, and overhead charges (capital equip-
`ment, regulatory compliance costs, and liability insurance
`costs). In the evolving era of managed care, the impact of
`cost-effective strategies on these charges is evident to many
`in practice-based oncology. From 1991 to 1996, the average
`Medicare reimbursement for a chemotherapy administration
`declined 57% from $155.51 to $67.01.9 Additionally, the
`Health Care Finance Administration (HCFA) formally elimi-
`nated payment for all supplies used to administer chemother-
`apy in 1992. In a recent appraisal of cost and reimbursement
`patterns for commonly used infusional and bolus lymphoma,
`
`From the Section ofHematology/Oncology, Department of Medicine,
`Committee on Clinical Pharmacology, and Cancer Research Center
`University of Chicago, Chicago, IL.
`Submitted August 12, 1997; accepted March 24, 1998.
`Address reprint requests to Mark J. Ratain, MD, University of
`Chicago Medical Center, 5841 S Maryland Ave, MC 2115, Chicago, IL
`60637-1470; Email mjratain@mcis.bsd.uchicago.edu.
`© 1998 by American Society of Clinical Oncology.
`0732-183X/98/1607-0002$3.00/0
`
`Journal of Clinical Oncology, Vol 16, No 7 (July), 1998: pp 2557-2567
`
`2557
`
`Downloaded from ascopubs.org by 63.247.71.66 on January 9, 2017 from 063.247.071.066
`
`Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
`
`Breckenridge Exhibit 1084
`DeMario
`Page 001
`
`

`

`2558
`
`breast, and colon regimens, Lokich et all' noted Medicare
`endorsement averaged just 21%
`to 36 % of total charge
`claims. The larger implications for reimbursement denials of
`novel, off-label, or investigational infusional regimens are
`obvious. Pharmacoeconomics, heralded by Medicare, and,
`increasingly, private-payer capitation constraints will pro-
`mote increasingly cost-effective treatment regimens. From
`the pharmacoeconomic viewpoint, the rapid emergence of
`orally available oncologic agents might be as readily antici-
`pated as the transition from hospital- to outpatient-based
`chemotherapy administration.
`An administrative cost analysis of ganciclovir therapy
`provides an illustration of the economic issues for orally
`formulated agents.,n1 2 A late complication of AIDS, cyto-
`megalovirus retinitis requires a protracted ganciclovir treat-
`ment course. Sullivan et all 2 have examined cost issues of
`ganciclovir administered over an 80-day schedule. While the
`direct drug costs of oral ganciclovir were nearly 170% those
`of the intravenous formulation, mean oral total therapy costs
`averaged just 58% those of intravenous therapy ($4,938 v
`$8,583). Cost variables examined include administration
`and nursing, central intravenous catheter, and infusion pump
`costs, and costs related to adverse effects of intravenous
`ganciclovir administration, such as line sepsis. A significant
`cost differential was noted for induction, adverse-event
`treatment, and combined home care, nursing administration,
`and monitoring charges, for which oral therapy costs repre-
`sented only 54%, 36%, and 14% of intravenous therapy
`costs, respectively. 12 Thus, prescription of oral ganciclovir
`results in increased revenues for its manufacturer and
`decreased revenues to health care providers and hospitals.
`Aside from economic considerations, it seems intuitive
`that patient preferences and quality-of-life issues will be
`another major impetus for the development of orally formu-
`lated chemotherapy. Recognizing the limitations of cyto-
`toxic therapy in many metastatic cancers, patient quality of
`life is increasingly becoming a central consideration in
`palliative treatment regimens. The issue of patients' prefer-
`ences regarding chemotherapy administration has recently
`been examined by Liu et al.' 3 Patients with advanced
`malignancies were asked about their preferred method of
`treatment before initiation of chemotherapy. Of 103 patients,
`more than 90% indicated a preference for orally adminis-
`tered agents, provided significant reductions in efficacy or
`duration of response would not result from this mode of
`treatment. The reasons for patients' preferences included
`convenience (57%), current concerns or previous difficulties
`with intravenous access lines (55%), or preference to control
`the chemotherapy administration environment (33%). If
`equivalent safety and efficacy are demonstrated, an all oral
`regimen would likely first be realized in the palliative
`setting, for example, an oral fluoropyrimidine replacing
`
`DeMARIO AND RATAIN
`
`intravenous fluorouracil (5-FU) in the treatment of meta-
`static colorectal carcinoma.
`
`DRUG ADMINISTRATION CONSIDERATIONS
`To have efficacy in an oral formulation, a chemotherapeu-
`tic drug must be sufficiently bioavailable. Bioavailability
`concerns the rate and extent to which a drug is absorbed into
`the systemic circulation. The bioavailability of oral agents is
`contingent on adequate intestinal absorption and the circum-
`vention of intestinal and, subsequently, hepatic metabolic
`systems. In considering absorption, the limitations of satura-
`bility and structural stability in gastric and intestinal pH
`must be addressed. The importance of saturable absorption
`is exemplified by oral etoposide and oral leucovorin. The
`large bioavailability variation between low- and high-dose
`oral etoposide suggests a saturable absorption system.
`Hande et a114 noted a 76% ± 22% bioavailability of 100 mg
`etoposide, while a 400-mg dose was only 48% ± 18%
`bioavailable. A linear increase of etoposide bioavailability at
`doses up to 200 mg has been demonstrated by other
`investigators, which confirms the saturable absorptive kinet-
`ics. 15 Leucovorin represents a second agent for which
`limited oral bioavailability is clinically relevant. While
`leucovorin's relative bioavailability is 78% in a 40-mg oral
`dose, bioavailability is reduced to only 31% in a 200-mg
`dose.' 6 These constraints would limit the use of oral
`leucovorin in some commonly used adjuvant regimens for
`colon carcinoma.
`The importance of structural stability in gastric and
`intestinal pH is also demonstrated by the etoposide model.
`Etoposide stability is optimal at pH 5. Joel et al' 7 attempted
`to improve oral etoposide bioavailability through coadminis-
`tration of agents that improve stability of etoposide in
`intestinal fluid (ethanol or bile salts) or increase pH (cimeti-
`dine). 17 Although the bioavailability and area under the
`concentration-time curve (AUC) of etoposide were not
`significantly enhanced with the modifications imposed in
`this study, it is nevertheless valuable to consider pH
`manipulations for structural stability in etoposide and,
`potentially, other oral chemotherapeutic drugs.
`An ideal chemotherapeutic drug would have little interpa-
`tient variability (in absorption and AUC) and, more impor-
`tantly, little intrapatient variability with successive doses.
`Comparing bioavailability from 143 studies in several drug
`classes (including antineoplastics), Hellriegel et al' 8 noted a
`significant inverse correlation between decreasing absolute
`bioavailability and intersubject variability in absolute bio-
`availability. It follows that caution must be taken in prescrib-
`ing an oral chemotherapy drug with low bioavailability. The
`generally narrow therapeutic index of these agents means
`significant interpatient variability would predispose some
`
`Downloaded from ascopubs.org by 63.247.71.66 on January 9, 2017 from 063.247.071.066
`
`Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
`
`Breckenridge Exhibit 1084
`DeMario
`Page 002
`
`

`

`ORAL CHEMOTHERAPY: A REVIEW
`
`individuals to excessive toxicity, or, conversely, inadequate
`efficacy.
`There are numerous examples of clinically significant
`variability in bioavailability for orally formulated chemo-
`therapeutic agents. While etoposide bioavailability averages
`50%, significant interpatient and intrapatient biovariability
`produces a bioavailability range of 25% to 75%.19 For
`busulfan, Hassan et al20 have noted up to a twofold and
`sixfold variability in bioavailability in adult and young
`pediatric populations, respectively. Similarly, Lebbe et al21
`observed a greater than fivefold range of AUC in patients
`who received low-dose oral methotrexate. Noting a fivefold
`interpatient variability in mercaptopurine AUC following
`oral administration, Zimm et al22 questioned the efficacy of
`the standardized oral dose used as maintenance therapy for
`acute lymphoblastic leukemia.
`
`Other Bioavailability Limitations Specific for Oral Drugs
`Intestinal CYP3A4. As mentioned previously, intestinal
`metabolic systems represent a "first-pass" bioavailability
`limitation unique to oral drugs. Principal among these is
`enterocyte CYP3A4. CYP3A4 is an enzyme subtype of
`CYP3, itself one of three major subclasses of cytochrome
`P-450 enzymes (CYPI, CYP2, and CYP3). The P-450
`enzymes are primarily responsible for phase I metabolism,
`facilitating drug excretion through nonconjugation reac-
`tions. CYP3A represents the major P-450 subclass, which
`accounts for as much as 25% of phase I drug metabolism in
`humans. 23 While hepatocytes contain the highest concentra-
`tions of P-450, these enzymes have been isolated from the
`endoplasmic reticulum of several cell types. Four distinct
`intestinal CYP3A P-450s have been characterized in hu-
`mans, CYP3A3, CYP3A4, CYP3A5, and CYP3A7. These
`enterocyte P-450s lie below the surface of the jejunal
`microvillus. In humans, CYP3A4 is the major intestinal
`P-450.24
`While it is difficult to distinguish between the activity of
`enteric and hepatic CYP3A4, the importance of this entero-
`cyte enzyme in oral bioavailability limitations is suggested
`by clinical studies of cyclosporine metabolism. The brief
`anhepatic phase of orthotopic liver transplantation provides
`a system to analyze isolated metabolic activity of enterocyte
`CYP3A4. Measuring cyclosporine Ml and M2 metabolites
`following jejunal drug delivery, Kolars et al25 concluded that
`as much as 50% of cyclosporine is metabolized by entero-
`cyte P-450 3A4.
`Table 1 lists representative chemotherapeutic substrates
`of CYP3A4. Orally formulated etoposide and cyclophospha-
`mide are subject to substantial first-pass metabolism by
`enterocyte CYP3A4. The exact contribution of enterocyte
`CYP3A4 in the metabolism of oral etoposide is not known;
`however, inhibition studies discussed below suggest the
`
`2559
`
`Table 1. Investigational and Available Oral Chemotherapeutic Agents
`That Are Substrates of CYP3A4 and P-gp
`
`Agent
`
`CYP3A4
`
`P-gp
`
`References
`
`Cyclophosphamide
`Etoposide
`Idarubicin
`Paclitaxel*
`Topotecan*
`Vinorelbine
`
`*Investigational agents.
`
`x
`x
`
`x
`
`x
`
`x
`x
`x
`x
`x
`
`26
`27, 28
`29
`28, 30
`31
`32, 33
`
`possibility for a substantial role.34 Further investigation will
`be required to elucidate the role of enterocyte CYP3A4 in
`the first-pass metabolism of other orally available cytotoxic
`agents.
`In determining the potential pharmacokinetic significance
`of enterocyte CYP3A4 for a given oral agent, it is necessary
`to consider the significant interpatient variability in CYP3A4
`expression. It is known that hepatic CYP3A4 concentrations
`and catalytic activity vary at least 10-fold among individu-
`als. 35 This has practical implications in the metabolism of
`many drugs, including cyclosporine. Likewise, Lown et al 36
`have reported a greater than sixfold interpatient variability in
`enterocyte CYP3A4 metabolic activity. This substantial
`heterogeneity may also have important clinical implications
`in oral drug metabolism. While hepatic CYP3A4 catalytic
`activity may be quantified by the erythromycin breath test
`(ERMBT), this test is not useful for enterocyte CYP3A4.3 6
`Developing a noninvasive, quantitative probe for enterocyte
`CYP3A4 will be necessary to further our understanding of
`this enzyme's contribution in oral drug metabolism.
`Perhaps the most important reason for fully elucidating
`CYP3A4 metabolism is that bioavailability may be substan-
`tially enhanced through pharmacologic manipulations of
`this system. CYP3A4 has numerous inducers, principally
`rifampin, phenobarbital, and dexamethasone. 23,37 Con-
`versely, erythromycin, quinidine, ketoconazole, and cyclos-
`porine serve as inhibitors of CYP3A4.38-40 An appreciation
`of the utility of these agents may be gained by examining the
`cyclosporine model. Herbert et a141 demonstrated a signifi-
`cant reduction in cyclosporine oral bioavailability with the
`concomitant administration of rifampin, whereas Gupta et
`al 42 markedly increased oral cyclosporine bioavailability
`with the coadministration of erythromycin. Similarly, Gomez
`et al43 noted coadministration of ketoconazole increased oral
`cyclosporine bioavailability from 22% to 56%. The cost
`implications of increasing cyclosporine bioavailability
`through CYP3A4 inhibition have been examined. Keogh et
`al44 noted an annual savings of $5,200 per patient for cardiac
`transplant patients who received cyclosporine coadminis-
`tered with ketoconazole.
`Kobayashi et a134 have recently examined the effect of
`ketoconazole modulation on the metabolism of etoposide.
`
`Downloaded from ascopubs.org by 63.247.71.66 on January 9, 2017 from 063.247.071.066
`
`Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
`
`Breckenridge Exhibit 1084
`DeMario
`Page 003
`
`

`

`2560
`
`Etoposide
`is known
`to be metabolized by CYP3A4-
`mediated O-demethylation.27 With coadministration of the
`potent CYP3A4 inhibitor ketoconazole, a 44% increase in
`plasma etoposide AUC was noted.34 This model suggests
`CYP3A4 modulation may be of great value in improving the
`oral bioavailability of chemotherapeutic agents that are its
`substrates.
`Intestinal P-glycoprotein. P-glycoprotein (P-gp) is a
`drug efflux pump well known to confer chemotherapy
`resistance. Orally formulated cytotoxic drugs known to be
`P-gp substrates include etoposide, idarubicin, and topotecan
`(investigational formulation).29,31 Encoded by MDR1, P-gp
`has been isolated in several human tissues, including the
`intestinal mucosa.45 There are preclinical data that suggest
`P-gp limits the intestinal absorption of paclitaxel, docetaxel,
`and vinblastine.46,47 Moreover, Leu et al48 reported a signifi-
`cant increase in etoposide bioavailability in rat jejunal and
`ileal loops first exposed to intravenous quinidine, a known
`P-gp inhibitor. The effect was realized at quinidine concen-
`trations at or below the therapeutic range. A monoclonal
`antibody against P-gp similarly produced a marked decrease
`in etoposide efflux from this rat jejunal system. Two
`preclinical studies of oral paclitaxel have demonstrated the
`significant bioavailability limitations
`imposed by P-gp.
`Sparreboom et al47 noted a sixfold increase in paclitaxel
`AUC in mdr la (-/-) mice, which lack intestinal P-gp. Using
`a murine model, Van Asperen et al49 have recently examined
`the effects of the P-gp blocker SDZ PSC 833 on the oral
`bioavailability of paclitaxel. The effect of this potent P-gp
`blocker was substantial; SDZ PSC 833 pretreatment resulted
`in a 10-fold increase in paclitaxel AUC. The bioavailability
`limitations imposed by P-gp must therefore be considered in
`the development of orally formulated chemotherapy. Trials
`that use P-gp inhibitors such as quinidine, verapamil, or
`SDZ PSC 833 may be anticipated in the successful formula-
`tion of oral paclitaxel or other agents.
`Table 1 lists representative chemotherapeutic substrates
`of P-gp.28
`
`Patient Compliance Issues
`
`Aside from bioavailability limitations, patient noncompli-
`ance represents a second potential major obstacle for orally
`formulated chemotherapy. Bonadonna and Valagussa 5o un-
`derscored
`the implications of an incomplete treatment
`course in the adjuvant breast setting; markedly inferior
`disease-free survival was experienced in patients who re-
`ceived less than 65% of planned therapy. Various studies
`have examined noncompliance in patient self-administered
`chemotherapy regimens. Lebovits et a151 noted a noncompli-
`ance rate of 43% in 51 breast cancer patients treated over 26
`weeks with an outpatient, oral cyclophosphamide regimen.
`Factors associated with higher rates of noncompliance
`
`DeMARIO AND RATAIN
`
`included lower socioeconomic status or treatment in a
`community-based setting. Levine et al 52 examined outpa-
`tient allopurinol and prednisone compliance in a cohort of
`patients who received concomitant chemotherapy for hema-
`tologic malignancies. With no interventions, a full allopuri-
`nol compliance rate of only 17% was noted in patients
`responsible for one component of a potentially curative
`treatment regimen.52 Moreover, pharmacokinetic analysis
`showed actual compliance was less than half that suggested
`by patient self-report. Importantly, measures designed to
`increase compliance, including patient education, home
`psychologic support, and exercises in pill taking, were able
`to increase compliance nearly threefold. The impact of side
`effects, complexity of treatment regimen, and age on compli-
`ance rates has subsequently been investigated.53 The occur-
`rence, frequency of occurrence, or severity of physical side
`effects correlated with clinic appointment noncompliance,
`but not with compliance of self-administered chemotherapy
`medications.
`For self-administered oral regimens, quantifying compli-
`ance rates will be essential for the accurate determination of
`regimen efficacy. In this regard, a novel electronic model has
`been suggested by Lee et al.54 An electronically activated
`tablet bottle scored opening to indicate daily outpatient
`compliance with an oral chemotherapy agent. A compliance
`rate of 110.6% ± 20.6% was demonstrated in a cohort of 21
`patients responsible for a self-administered component of an
`outpatient lymphoma chemotherapy regimen. It is also
`encouraging that, using the identical electronic model, these
`investigators were able to demonstrate a 93.2% ± 12%
`compliance rate in a cohort of small-cell lung carcinoma
`patients receiving low-dose oral etoposide. 55 The high
`compliance rate was maintained despite the cohort's gener-
`ally poor overall prognosis.
`
`ORALLY AVAILABLE AGENTS AND OVERVIEW OF
`SELECTED NOVEL CYTOTOXICS
`
`Table 2 lists relevant pharmacokinetic and cost data for
`the orally formulated chemotherapeutic agents. For several
`of these agents, bioavailability varies substantially between
`patients, with increasing drug dose, or with food. Given the
`narrow therapeutic index for many of these agents, it is
`desirable to improve bioavailability and reduce intrapatient
`biovariability through mechanisms discussed earlier. Where
`applicable, an oral/intravenous cost ratio is given for equiva-
`lent milligram doses of drug at average wholesale prices.
`These ratios do not consider the extensive costs of intrave-
`nous drug administration. While price data for the investiga-
`tional oral agents are not currently available, it is likely their
`price will be considerably greater than that of the intrave-
`nous preparation, as in the previously discussed case of
`
`Downloaded from ascopubs.org by 63.247.71.66 on January 9, 2017 from 063.247.071.066
`
`Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
`
`Breckenridge Exhibit 1084
`DeMario
`Page 004
`
`

`

`Table 2. Comparison of Oral Chemotherapeutic Agents
`
`Oral
`Bioavailability
`(%)
`
`0-80
`122
`100
`61
`
`16-50
`14-46
`20-90
`100
`
`48-76
`85
`100
`100
`
`58-95
`100
`70-80
`
`Terminal
`Half-life
`(hrs)
`
`0.22
`4.5
`6-16
`0.80
`
`1.5
`11
`3-15
`2.0-3.0
`
`6.6
`2.8
`0.16
`1.3-2.9
`1.3-2.5
`0.95-1.8
`2.3-3.4
`2.0
`
`6.0-7.7
`
`Cost Ratio
`(oral/
`intravenous)
`
`Comments
`
`122% mean bioavailability and 4.5-hour half-life
`reflect 5-FU modulation with 5-ethynyluracil
`
`Extensive first-pass gut metabolism to thiouric acid
`
`4.56
`
`Bioavailability substantially limited over 30 mg/m2
`
`0.46
`1.53
`
`0.15
`
`Bioavailability reduced at doses >100 mg
`
`Half-lifes are of cis- and trans-4-OH CCNU
`metabolites, respectively
`
`Half-life of platinum in ultra-filtrate with 120
`mg/m2/d x 5 JM216 schedule
`
`Undergoes reversible pH-dependent conversion to
`hydroxy acid
`A colloidal dispersion is poorly bioavailable
`
`2561
`
`References
`
`56-61
`
`62-65
`66
`
`22, 67, 68
`69-71
`72, 73
`70
`
`14
`74
`70,75
`76
`
`77, 78
`71,79, 80
`78
`
`81, 82, 83
`
`84, 85
`
`86-88
`
`89, 90
`
`ORAL CHEMOTHERAPY: A REVIEW
`
`Class
`
`Agent
`
`Pyrimidine antimetabolites
`
`5-FU
`
`Other antimetabolites
`
`Epipodophyllotoxins
`Alkylating agents
`
`UFT
`Capecitabine
`
`Mercaptopurine
`Thioguanine
`Methotrexate
`Hydroxyurea
`
`Etoposide
`Cyclophosphamide
`Procarbazine
`Lomustine
`
`Melphalan
`Busulfan
`Chlorambucil
`
`Platinum derivatives
`
`JM-216
`
`Vinca alkaloids
`
`Vinorelbine
`
`26-45
`
`24-56
`
`Camptothecins
`
`Topotecan
`
`30
`
`2.82
`
`9-aminocamptothecin
`
`27-49
`
`7.5-24.3
`
`ganciclovir. Selected novel oral cytotoxic agents are now
`discussed.
`
`Novel Oral Fluoropyrimidines and Modulators
`
`UFT UFT is an oral preparation of 1-(2-tetrahydrofuryl)-
`5-FU (tegafur) and uracil admixed in a 1:4 molar ratio. UFT
`is a unique oral prodrug that undergoes both hepatic P-450
`and target tissue metabolism to 5-FU.91 The 1:4 tegafur-to-
`uracil ratio was noted to provide the highest intratumoral
`levels of 5-FU.92 The oral bioavailability of UFT is excel-
`lent; Antilla et a162 noted a comparative oral/intravenous
`AUC of 115% ± 8%. Phase I investigations of 28-
`consecutive-day administration with concomitant high- or
`low-dose leucovorin have suggested feasibility and accept-
`able toxicity at doses less than 350 mg/m 2/d.93,94 UFT has
`been used extensively in Japan. Examining pooled phase II
`of more than 400 patients with cholangiocarcinoma, colorec-
`tal, gastric, and breast cancers, Ota et al95 noted single-agent
`UFT response rates that ranged from 25% to 32%.95 More
`recently, Pazdur et a196 noted a 42% response rate in
`therapy-naive colon carcinoma patients. Comparing oral
`ftorafur and intravenous 5-FU in a small cohort of patients
`with advanced colorectal cancer, Andersen et al97 noted
`equivalent gastrointestinal toxicity with significantly lower
`hematologic toxicity in ftorafur-treated patients. There was
`
`no significant difference in median or overall survival in
`UFT- or 5-FU-treated patients.97 Confirmatory phase III
`trials will be necessary to justify UFT in a metastatic or,
`ultimately, adjuvant colorectal setting. The ability to admin-
`ister UFT over protracted schedules may allow for the
`simulation of continuous infusion 5-FU kinetics, which
`suggests a role as a radiosensitizing agent.
`Capecitabine. Capecitabine represents an orally bioavail-
`able fluoropyrimidine designed with unique tumor selectiv-
`ity. An oral 5'-deoxy-5-fluorocytidine derivative, it is ab-
`sorbed unchanged from intestinal mucosa and converted
`to 5' deoxy-5-fluorocytidine ribonucleotide (5'-DFCR) via
`hepatic acylamidases. The drug is subsequently converted to
`5'-DFUR by cytidine deaminase, an enzyme preferentially
`located in hepatic and tumor tissues. Finally, the conversion
`of 5' deoxy-5-fluorouridine ribonucleotide. (5'-DFUR) to
`5-FU occurs intratumorally via pyrimidine nucleoside phos-
`phorylases (PyNPases), uridine phosphorylase, and thymi-
`dine phosphorylase.98,99 Specificity is achieved with PyN-
`Pase, which is constitutively expressed at high levels within
`tumors.
`Capecitabine phase I data have been determined for
`6-week continuous and intermittent administration sched-
`ules.100 A maximum-tolerated dose (MTD) of 1,657 mg/
`m 2/d was determined for capecitabine on a 6-week daily
`
`Downloaded from ascopubs.org by 63.247.71.66 on January 9, 2017 from 063.247.071.066
`
`Copyright © 2017 American Society of Clinical Oncology. All rights reserved.
`
`Breckenridge Exhibit 1084
`DeMario
`Page 005
`
`

`

`2562
`
`administration schedule. At this dose level, plasma 5-FU
`concentrations were similar to those obtained for continuous
`intravenous infusion 5-FU. Direct examination of resected
`colorectal tumor and surrounding tissue showed a tumor-to-
`normal tissue capecitabine ratio of 2.9 in pretreated pa-
`tients. 101 Phase II studies that examined capecitabine on
`12-week continuous and 12-week intermittent schedules
`with and without leucovorin showed response rates of 19%,
`28%, and 24%, respectively. 102 Based on these results, phase
`III trials of capecitabine versus single-agent 5-FU might be
`anticipated in the setting of metastatic colon carcinoma.
`Capecitabine recently has been approved by the Food and
`Drug Administration for use in refractory breast carcinoma.
`5-Ethynyluracil (eniluracil). 5-FU has long been recog-
`nized as an active agent in the treatment of a variety of
`human malignancies.
`Improved efficacy for 5-FU has
`previously been achieved with modulatory agents such as
`leucovorin and N-(phosphonacetyl)-L-asparatate
`(PALA).
`5-Ethynyluracil (eniluracil) is a novel modulator shown to
`improve the efficacy of 5-FU in preclinical models through
`the selective, irreversible inhibition of dihydropyrimidine
`dehydrogenase (DPD) metabolism. In individuals with nor-
`mal levels of DPD, 5-FU is quickly metabolized in both its
`intravenous and oral formulations, which results in a parent
`drug half-life of only 5 to 20 minutes.103 Moreover, the
`extremely variable bioavailability (0% to 80%) of oral 5-FU,
`which limits its clinical use, is felt to be a reflection of
`It is
`intrapatient variability in intestinal DPD activity.'"
`therefore anticipated that a DPD inhibitor such as 5-ethynylu-
`racil would increase both 5-FU half-life and oral bioavailabil-
`ity. A recent phase I investigation showed complete absorp-
`tion (average bioavailability, 122%) and a substantially
`prolonged half-life (4.5 hours) for oral 5-FU when it was
`concomitantly administered with 5-ethynyluracil.61 The in-
`terpatient variability of 5-FU bioavailability was also re-
`duced (coefficient of variation [CV], 33%), while systemic
`5-FU clearance was reduced more than 20-fold to a rate
`comparable to that of renal blood flow. This study noted
`neutropenia as the dose-limiting toxicity at a dose of 25
`mg/m 2/d on a daily-times-five every-28-day schedule. The
`impact of improved 5-FU oral bioavailability and prolonged
`half-life has been demonstrated in preclinical models, which
`have shown up to a sixfold increase in the 5-FU therapeutic
`index with coadministration of 5-ethynyluracil.105,106 A
`recent phase II investigation examined the 5-day administra-
`tion of oral 5-FU with 5-ethynyluracil with and without
`leucovorin
`to patients with metastatic colorectal carci-
`noma.107 A 33% response rate was noted in previously
`untreated patients treated with 20 mg/m 2 x 5 oral 5-FU, 50
`mg/d oral leucovorin, and 5-ethynyluracil (eniluracil).10 7
`Additional studies using a 28-day protracted 5-FU schedule
`with 776C85 DPD inhibition are currently underway to
`
`DeMARIO AND RATAIN
`
`clarify the optimal administration schedule for this promis-
`ing new modulatory agent.
`
`Other Oral Cytotoxics in Development
`
`JM-216. Bis-acetato-amine-dichloro-cyclohexyl-amine-
`platinum (IV) or JM216 represents a unique platinum-based
`agent with oral bioavailability. Phase I single-dose data
`suggest linear pharmacokinetics at doses less than 120
`mg/m 2; however, saturability was suggested at doses greater
`than 200 mg/m2.81 Significant interpatient variability (CV)
`to 63%) and AUC
`of the maximum concentration (21%
`(26% to 63%) was demonstrated in this single-dose phase I
`study. JM216 displays a notable biotransformation follow-
`ing oral administration, with six metabolite species. JM-118
`has been identified as the principal cytotoxic species. 83
`JM216 is unique among platinum derivatives for its schedule-
`dependency; in vivo models suggest a maximal therapeutic
`8 An MTD of
`index with 5-consecutive-day administration.
`120 to 140 mg/m 2 daily on a 5-day administration schedule
`has been determined from phase I testing. 82,109 Linear AUC
`pharmacokinetics were observed up to 140 mg/m2/d x 5.82
`Significant toxicities observed for JM216 to date include
`nausea/emesis, diarrhea, neutropenia, and thrombocytope-
`nia. 82,109 A significant correlation between the thrombocyto-
`penia and platinum ultrafiltrate AUC has been observed. 82
`Myelosuppression may be delayed relative to that observed
`for cisplatin or carboplatin.' 10 Importantly, JM216 has not
`demonstrated significant renal or neurotoxicity in preclinical
`or phase I/II trials to date, a significant toxicity advantage
`over cisplatin." 0-112 A recent phase II trial of single-agent
`JM216 for hormone-refractory prostate cancer noted a
`prostate-specific antigen (PSA) reduction in 42% of treated
`patients.110 Phase II 5-day dosing studies have also demon-
`strated responses in small-cell lung cancer.113 A phase II trial
`in previously untreated non-small-cell lung
`of JM-216
`cancer found no objective responses among 13 assessable
`patients treated on a 120 mg/m 2/d x 5 schedule.114
`Vinorelbine. Vinorelbine is unique among

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