BRIEF COMMUNICATION

Role of Body Surface Area in Dosing of Investigational Anticancer Agents in Adults, 1991–2001

Sharyn D. Baker, Jaap Verweij, Eric K. Rowinsky, Ross C. Donehower, Jan H. M. Schellens, Louise B. Grochow, Alex Sparreboom

Affiliations of authors: S. D. Baker, R. C. Donehower, L. B. Grochow, Division of Experimental Therapeutics, The Sydney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; J. Verweij, J. H. M. Schellens, A. Sparreboom, Department of Medical Oncology, Erasmus MC-Daniel den Hoed Cancer Center, Rotterdam, The Netherlands; E. K. Rowinsky, The Institute for Drug Development, Cancer Therapy and Research Center, San Antonio, TX.

Correspondence to: Sharyn D. Baker, Pharm.D., Division of Experimental Therapeutics, The Sydney Kimmel Comprehensive Cancer Center at Johns Hopkins, 1650 Orleans St., Rm. 1M87, Baltimore, MD 21237 (e-mail: sdbaker{at}jhmi.edu).

ABSTRACT

The prescribed dose of anticancer agents is most commonly calculated using body surface area as the only independent variable, and it has been shown that this approach still results in large interpatient variability in drug exposure. Here, we retrospectively assessed the pharmacokinetics of 33 investigational agents tested in phase I trials from 1991 through 2001, as a function of body surface area in 1650 adult cancer patients. Twelve of the drugs were administered orally, 19 were administered intravenously, and two were administered by both routes. Body surface area-based dosing was statistically significantly associated with a reduction in interpatient variability in drug clearance for only five of the 33 agents: docosahexaenoic acid (DHA)–paclitaxel, 5-fluorouracil/eniluracil, paclitaxel, temozolomide, and troxacitabine. These results do not support the use of body surface area in dose calculations and suggest that alternate dosing strategies should be evaluated. We conclude that body surface area should not be used to determine starting doses of investigational agents in future phase I studies.


In clinical oncology, the traditional method by which individualized anticancer drug doses are determined uses body surface area, because use of this measurement is thought to reduce the interpatient variability of drug exposure and, hence, drug effects (1). The use of body surface area measurements arose from the extrapolation of drug doses used in experimental animals to those considered safe as starting doses for human cancer patients in phase I clinical trials (1). However, a rigorous scientific rationale for body surface area-based dosing of anticancer drugs in adults is lacking, especially when one considers that the difference in size between mice and humans is far greater than the difference in size between individual patients. Although the primary objective of phase I trials is to evaluate drug toxicity, antitumor activity is usually a secondary objective. Other measures, such as drug clearance, have also been used as surrogate markers of drug effects. However, it has been widely recognized that large interpatient variability in drug clearance exists despite the use of body surface area in drug-dose calculations (2). Indeed, for most drugs that are used in clinical practice today, clearance cannot be reliably predicted by body surface area, because other factors involved in drug disposition may be more important for clearance (1,35). For example, several recent studies (69) have highlighted the importance of genetic polymorphisms in drug-metabolizing enzymes and drug transporter proteins in explaining interindividual pharmacokinetic variability. As a follow-up to a preliminary report by Grochow et al. (10), we assessed the pharmacokinetics of 33 investigational agents in adult cancer patients as a function of body surface area to provide a pharmacokinetic rationale for selecting the appropriate starting doses for phase I evaluation.

Data were obtained from 1650 patients who were treated with 33 anticancer drugs (involving 21 classes of agents) that were developed in phase I trials over a 10-year period at three institutions. Twelve of the drugs were administered orally, 19 were administered intravenously, and two were administered by both routes. Detailed clinical and pharmacokinetic profiles for these agents have been described elsewhere (5,1168). All patients were at least 18 years old and had normal organ function, except for those enrolled in two studies that involved patients with varying degrees of renal and hepatic impairment (65,66). Drug clearance was calculated by using either noncompartmental or compartmental analysis (69) and was expressed either as liters per hour (L/h) or as L/h normalized to body surface area in meters squared (L/h/m2). Interpatient variation in drug clearance was calculated by dividing the standard deviation by the mean and was expressed as a percentage (i.e., the coefficient of variation [CV]). We used the following arbitrarily defined criteria to determine whether body surface area-based dosing was statistically significantly associated with a reduction in interpatient variation in clearance: 1) a linear regression coefficient (R) >= .50; 2) P<.01; and 3) a relative reduction in the variability of clearance >=15%, which was calculated by using the formula {[CV for clearance (L/h) – CV for clearance (L/h/m2)] / [CV for clearance (L/h)]} x 100. All three of these criteria had to be met for the reduction to be considered statistically significant.

The median body surface area for the entire patient population was 1.86 m2 (interquartile range = 1.68–2.00 m2) and the mean body surface area was 1.86 m2 (range = 1.25–3.06 m2). The CV for clearance, the correlation between body surface area and clearance, and the relative reduction in variability for clearance for each of the agents are listed in Tables 1 and 2GoGo. For all but five agents (i.e., docosahexaenoic acid [DHA]– paclitaxel, 5-fluorouracil/eniluracil, paclitaxel, temozolomide, and troxacitabine), body surface area-based dosing was not associated with a statistically significant reduction in the interpatient variability in drug clearance.


View this table:
[in this window]
[in a new window]
 
Table 1. Variability in drug clearance of orally administered agents*
 

View this table:
[in this window]
[in a new window]
 
Table 2. Variability in drug clearance of intravenously administered agents*
 
In the case of drugs for which renal function plays a principal role in drug elimination, BSA-based dosing may decrease variability in drug clearance among patients. For example, troxacitabine, an L-nucleoside analogue, and 5-fluorouracil coadministered orally with the dihydropyrimidine-dehydrogenase inactivator eniluracil, are primarily excreted in the urine as unchanged drugs ({approx}60% and 75% unchanged, respectively). Normalization of drug dose to body surface area was associated with a 22% reduction in the interpatient variation in troxacitabine clearance and a 15% reduction in the interpatient variation in 5-fluorouracil/eniluracil clearance (17,67). Body surface area-based dosing of pemetrexed, a novel multitargeted antifolate that is also mainly excreted in urine as unchanged drug (>70% unchanged) was also associated with a 16% reduction in interpatient variation in drug clearance (70). The known association between body surface area and glomerular filtration rate (71) may explain the observed relationship between body surface area and clearance of these renally excreted agents. However, the finding that differences in body surface area among patients account for only a small percentage (i.e., <=22%) of the total variability in drug clearance is consistent with results from a recent study (72) that reported that body surface area was poorly correlated with glomerular filtration rate (R<.22). Therefore, for agents that are excreted principally by the kidneys, dosing strategies that are based on the accurate assessment of glomerular filtration rate and not on body surface area should be associated with decreased interpatient variability in clearance (73).

Body surface area-based dosing may also be a preferred dosing strategy for drugs that are confined to blood volume because of the known relationship between body size and blood volume (74,75). For example, we found that body surface area was highly correlated with temozolomide clearance (R = .88, P<.001) and was associated with 35% of the variation in temozolomide clearance among patients. Temozolomide is an alkylating agent that undergoes pH-dependent breakdown to the active moiety 5-(3-methyltriazen-1-yl)imidazole-4-carboxamide immediately following drug administration and is virtually isolated to the central compartment. Interpatient variability in DHA–paclitaxel clearance was reduced by 26% when the dose of that agent was normalized to body surface area. DHA–paclitaxel has a low clearance (~0.11 L/h), has a small volume of distribution (~4 L), and is extensively (>99.6%) but nonspecifically bound to alpha1-acid glycoprotein and albumin (76). These characteristics indicate that DHA–paclitaxel is principally confined to blood volume, and that systemic exposure to total drug may be dictated by its capacity to bind plasma proteins. By contrast, drugs that bind a single protein with high affinity but low capacity [e.g., as 7-hydroxystaurosporine (UCN-01) binds alpha1-acid glycoprotein (77)] are more likely than DHA–paclitaxel to show wide variations in unbound (i.e., pharmacologically active) drug concentrations among patients. For agents with disposition characteristics like UCN-01, measurement of total drug concentrations in plasma is a poor surrogate for that of unbound drug (78), and accurate assessment of the relationship between body surface area and clearance requires measurement of unbound drug concentrations. Thus, the protein-binding characteristics of investigational agents across species should be characterized before those agents are evaluated in phase I trials.

Normalizing doses to body surface area may also provide an advantage for drugs, such as paclitaxel, that are formulated in vehicles that are known to affect drug disposition. For example, previous work (51,79) has shown that the distribution of paclitaxel in the blood depends on the duration of drug infusion and the dose of its formulation vehicle (Cremophor EL-dehydrated ethanol USP; Bristol-Myers Squibb, Wallingford, CT), which is likely due to the preferential affinity of paclitaxel for Cremophor EL in the circulation. It has been demonstrated that this vehicle has a distribution volume that approximates the blood volume and that body surface area is a statistically significant covariate for Cremophor EL clearance (80). Thus the impact of body surface area on the variability in paclitaxel pharmacokinetics is most likely associated with the affinity of paclitaxel for its vehicle in the circulation (81), the distribution of which is linked to total blood volume, and thus to body surface area (75).

For the majority of the anticancer agents we examined that underwent development in adult patients from 1991 through 2001, we found that body surface area-based dosing was not statistically significantly associated with a decrease in interpatient variability in clearance. For the few agents for which clearance was statistically significantly associated with body surface area, the relative reduction in variability in clearance was between 15% and 35%, which suggests that only up to one-third of the total variability can be explained by differences in body surface area. These results therefore do not support body surface area-based dosing for most anticancer agents but warrant the evaluation of alternate dosing strategies for phase I evaluation in adult humans. A non-body surface area-based dosing strategy (e.g., one based on a fixed dose) was successfully implemented in the development of five of the orally administered agents examined in our study [i.e., phenylbutyrate (20), PKI166 (21), R115777 (22), SCH66336 (23), and ZD9331 (33)], demonstrating that administration of a fixed total dose is feasible for the development of both cytotoxic and noncytotoxic targeted anticancer agents. We therefore recommend that the practice of calculating starting drug doses on the basis of body surface area in phase I trials should be abandoned and that future early clinical trials should instead evaluate the administration of fixed drug doses that are calculated on the basis of an average body surface area of 1.86 m2. For novel targeted agents, dose refinement should be based on finding an exposure that produces a biologic or molecular effect on a drug target that is associated with a desired therapeutic outcome or avoidance of a toxicologic outcome. For cytotoxic agents that have a narrow therapeutic window, efforts should continue to focus on defining individual doses that are based on patient characteristics that are known to affect drug clearance (e.g., age, sex, renal function, and use of concomitant medications). A combination of these strategies should yield more rational dosing schemes that can be implemented in oncology practice.

NOTES

Present address: J. H. M. Schellens, Netherlands Cancer Institute, Amsterdam, The Netherlands.

Present address: L. B. Grochow, National Cancer Institute, Bethesda, MD.

Present address: A. Sparreboom, National Cancer Institute, Bethesda, MD.

We thank Drs. Michael A. Carducci and Theresa Rogers (Baltimore, MD), Mats O. Karlsson (Uppsala, Sweden), Olaf van Tellingen (Amsterdam, The Netherlands), and Daniel D. Von Hoff (Tucson, AZ) for providing pharmacokinetic and patient demographic data.

REFERENCES

1 Gurney H. Dose calculation of anticancer drugs: a review of the current practice and introduction of an alternative. J Clin Oncol 1996;14:2590–611.[Abstract]

2 Sawyer M, Ratain MJ. Body surface area as a determinant of pharmacokinetics and drug dosing. Invest New Drugs 2001;19:171–7.[Medline]

3 Ratain MJ. Body-surface area as a basis for dosing of anticancer agents: science, myth, or habit? J Clin Oncol 1998;16:2297–8.[Medline]

4 de Jongh FE, Verweij J, Loos WJ, de Wit R, de Jonge MJ, Planting AS, et al. Body-surface area-based dosing does not increase accuracy of predicting cisplatin exposure. J Clin Oncol 2001;19:3733–9.[Abstract/Free Full Text]

5 Mathijssen RH, Verweij J, de Jonge MJ, Nooter K, Stoter G, Sparreboom A. Impact of body-size measures on irinotecan clearance: alternative dosing recommendations. J Clin Oncol 2002;20:81–7.[Abstract/Free Full Text]

6 Danesi R, De Braud F, Fogli S, Di Paolo A, Del Tacca M. Pharmacogenetic determinants of anti-cancer drug activity and toxicity. Trends Pharmacol Sci 2001;22:420–6.[Medline]

7 McLeod HL, Evans WE. Pharmacogenomics: unlocking the human genome for better drug therapy. Annu Rev Pharmacol Toxicol 2001;41:101–21.[Medline]

8 Innocenti F, Ratain MJ. Update on pharmacogenetics in cancer chemotherapy. Eur J Cancer 2002;38:639–44.[Medline]

9 Relling MV, Dervieux T. Pharmacogenetics and cancer therapy. Nat Rev Cancer 2001;1:99–108.[Medline]

10 Grochow LB, Baraldi C, Noe D. Is dose normalization to weight or body surface area useful in adults? J Natl Cancer Inst 1990;82:323–5.[Medline]

11 de Jonge MJ, Punt CJ, Gelderblom AH, Loos WJ, van Beurden V, Planting AS, et al. Phase I and pharmacologic study of oral (PEG-1000) 9-aminocamptothecin in adult patients with solid tumors. J Clin Oncol 1999;17:2219–26.[Abstract/Free Full Text]

12 de Jonge MJ, Verweij J, Loos WJ, Dallaire BK, Sparreboom A. Clinical pharmacokinetics of encapsulated oral 9-aminocamptothecin in plasma and saliva. Clin Pharmacol Ther 1999;65:491–9.[Medline]

13 Gelderblom H, Salazar R, Verweij J, O’Neill V, Sparreboom A, Principe P, et al. Oral bioavailability of BN80915 in adult patients with solid tumors. Proc Am Assoc Cancer Res 2001;42:2895.

14 Pronk LC, Vasey P, Sparreboom A, Reigner B, Planting AS, Gordon RJ, et al. A phase I and pharmacokinetic study of the combination of capecitabine and docetaxel in patients with advanced solid tumours. Br J Cancer 2000;83:22–9.[Medline]

15 Gilbert J, Dees EC, Baker SD, Summerson L, Carducci MA, Izumi T, et al. A phase I study of CS-682, an oral antimetabolite, in patients with refractory solid tumors. Clin Cancer Res 2000;6:4554s.

16 Brindley CJ, Morrison R, Gordon RJ, Devlin AJ, van der Gaast A, Verweij L, et al. Clinical pharmacokinetics of 2'-deoxy-2'-methylidenecytidine (DMDC), a deoxycytidine analogue antineoplastic agent. Clin Pharmacokinet 2000;38:475–91.[Medline]

17 Baker SD, Diasio RB, O’Reilly S, Lucas VS, Khor SP, Sartorius SE, et al. Phase I and pharmacologic study of oral fluorouracil on a chronic daily schedule in combination with the dihydropyrimidine dehydrogenase inactivator eniluracil. J Clin Oncol 2000;18:915–26.[Abstract/Free Full Text]

18 Eskens FA, Levitt NC, Sparreboom A, Choi L, Mather R, Verweij J, et al. Effect of food on the pharmacokinetics of oral MMI270B (CGS 27023A), a novel matrix metalloproteinase inhibitor. Clin Cancer Res 2000;6:431–3.[Abstract/Free Full Text]

19 Levitt NC, Eskens FA, O’Byrne KJ, Propper DJ, Denis LJ, Owen SJ, et al. Phase I and pharmacological study of the oral matrix metalloproteinase inhibitor, MMI270 (CGS27023A), in patients with advanced solid cancer. Clin Cancer Res 2001;7:1912–22.[Abstract/Free Full Text]

20 Gilbert J, Baker SD, Bowling MK, Grochow L, Figg WD, Zabelina Y, et al. A phase I dose escalation and bioavailability study of oral sodium phenylbutyrate in patients with refractory solid tumor malignancies. Clin Cancer Res 2001;7:2292–300.[Abstract/Free Full Text]

21 Hoekstra R, Dumez H, Eskens F, Sizer K, Cohen P, Ravera C, et al. A phase I and pharmacological study of intermittent dosing of PKI166, a novel epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, administered orally to patients with advanced cancer. Clin Cancer Res 2001;7:3771s.

22 Verweij J, Kehrer DF, Planting AS, de Jonge MJ, Klaren A, de Heus G, et al. Phase I trial of irinotecan in combination with the farnesyl transferase inhibitor (FTI) R115777. Proc ASCO 2001;20:81a.

23 Eskens FA, Awada A, Cutler DL, de Jonge MJ, Luyten GP, Faber MN, et al. Phase I and pharmacokinetic study of the oral farnesyl transferase inhibitor SCH 66336 given twice daily to patients with advanced solid tumors. J Clin Oncol 2001;19:1167–75.[Abstract/Free Full Text]

24 Hammond LA, Eckardt JR, Baker SD, Eckhardt SG, Dugan M, Forral K, et al. Phase I and pharmacokinetic study of temozolomide on a daily-for-5-days schedule in patients with advanced solid malignancies. J Clin Oncol 1999;17:2604–13.[Abstract/Free Full Text]

25 Schellens JH, Creemers GJ, Beijnen JH, Rosing H, de Boer-Dennert M, McDonald M, et al. Bioavailability and pharmacokinetics of oral topotecan: a new topoisomerase I inhibitor. Br J Cancer 1996;73:1268–71.[Medline]

26 Gerrits CJ, Burris H, Schellens JH, Planting AS, van den Burg ME, Rodriguez GI, et al. Five days of oral topotecan (Hycamtin), a phase I and pharmacological study in adult patients with solid tumours. Eur J Cancer 1998;34:1030–5.[Medline]

27 Gerrits CJ, Burris H, Schellens JH, Eckardt JR, Planting AS, van der Burg ME, et al. Oral topotecan given once or twice daily for ten days: a phase I pharmacology study in adult patients with solid tumors. Clin Cancer Res 1998;4:1153–8.[Abstract]

28 Creemers GJ, Gerrits CJ, Eckardt JR, Schellens JH, Burris HA, Planting AS, et al. Phase I and pharmacologic study of oral topotecan administered twice daily for 21 days to adult patients with solid tumors. J Clin Oncol 1997;15:1087–93.[Abstract]

29 de Jonge MJ, Loos WJ, Gelderblom H, Planting AS, van der Burg ME, Sparreboom A, et al. Phase I pharmacologic study of oral topotecan and intravenous cisplatin: sequence-dependent hematologic side effects. J Clin Oncol 2000;18:2104–15.[Abstract/Free Full Text]

30 Gelderblom AJ, Loos WJ, de Jonge MJ, Sparreboom A, Planting AS, van der Burg ME, et al. Phase I and pharmacological study of increased dose oral topotecan in combination with intravenous cisplatin. Ann Oncol 2000;11:1205–7.[Medline]

31 Gelderblom H, Sparreboom A, de Jonge MJ, Loos WJ, Wilms E, Mantel MA, et al. Dose and schedule-finding study of oral topotecan and weekly cisplatin in patients with recurrent ovarian cancer. Br J Cancer 2001;85:1124–9.[Medline]

32 Loos WJ, Gelderblom H, Sparreboom A, Verweij J, de Jonge MJ. Inter- and intrapatient variability in oral topotecan pharmacokinetics: implications for body-surface area dosage regimens. Clin Cancer Res 2000;6:2685–9.[Abstract/Free Full Text]

33 de Jonge MJ, Punt CJ, Sparreboom A, Planting AS, Peters ME, van de Schraaf J, et al. Phase I and pharmacologic study of ZD 9331, a novel non-polyglutamated thymidylate synthase inhibitor, in adult patients with solid tumors. J Clin Oncol 2002;20:1923–31.[Abstract/Free Full Text]

34 Sparreboom A, de Jonge MJ, Punt CJ, Nooter K, Loos WJ, Porro MG, et al. Pharmacokinetics and bioavailability of oral 9-aminocamptothecin capsules in adult patients with solid tumors. Clin Cancer Res 1998;4:1915–9.[Abstract]

35 Planting AS, Schellens JH, van der Burg ME, de Boer-Dennert M, Winograd B, Stoter G, et al. Phase I and pharmacologic study of BMS-181174 given as a 6 h infusion every 4 weeks to patients with advanced solid tumors. Anticancer Drugs 1999;10:821–7.[Medline]

36 van Tellingen O, Punt CJ, Awada A, Wagener DJ, Piccart MJ, Groot Y, et al. A clinical pharmacokinetics study of carzelesin given by short-term intravenous infusion in a phase I study. Cancer Chemother Pharmacol 1998;41:377–84.[Medline]

37 Dees EC, Whitfield LR, Grove WR, Rummel S, Grochow LB, Donehower RC. A phase I and pharmacologic evaluation of the DNA intercalator CI-958 in patients with advanced solid tumors. Clin Cancer Res 2000;6:3885–94.[Abstract/Free Full Text]

38 Louwerens M, Smorenburg CH, Sparreboom A, Loos WJ, Verweij J, de Wit R. Phase I and pharmacokinetic study of the combination of docetaxel and methotrexate in patients with solid tumors. Eur J Cancer 2002;38:497–504.[Medline]

39 van Zuylen L, Sparreboom A, van der Gaast A, van der Burg ME, van Beurden V, Bol CJ, et al. The orally administered P-glycoprotein inhibitor R101933 does not alter the plasma pharmacokinetics of docetaxel. Clin Cancer Res 2000;6:1365–71.[Abstract/Free Full Text]

40 van Zuylen L, Sparreboom A, van der Gaast A, Nooter K, Eskens FA, Brouwer E, et al. Disposition of docetaxel in the presence of P-glycoprotein inhibition by intravenous administration of R101933. Eur J Cancer 2002;38:1090–9.[Medline]

41 Kennedy MJ, Elza K, Donehower R, Hoker B, Chen TL, Rowinsky EK. Phase I and pharmacologic (PK) study of docetaxel and doxorubicin in patients (pts) with solid tumors: dose limiting toxicity at the first dose level. Proc ASCO 1997;16:240a.

42 Wolff AC, Krishnamurthi S, Sparano JA, Armstrong DK, Davidson NE, Fetting JH, et al. A pharmacokinetic (PK) and pharmacodynamic study of doxorubicin and docetaxel combinations plus marimastat (AT/M) in metastatic breast cancer (MBC). Proc ASCO 2001;20:98a.

43 Eskens FA, Dumez H, Verweij J, Brindley C, Perschi A, Kovar A, et al. Cilengitide (EMD 121974) inhibits angiogenesis by blocking {alpha}v{beta}3 and {alpha}v{beta}5 integrins: mature results of a phase I and pharmacological study. Clin Cancer Res 2000;6:4524s.

44 Schellens JH, Planting AS, van Acker BA, Loos WJ, de Boer-Dennert M, van der Burg ME, et al. Phase I and pharmacologic study of the novel indoloquinone bioreductive alkylating cytotoxic drug E09. J Natl Cancer Inst 1994;86:906–12.[Abstract]

45 Schellens JH, Dombernowsky P, Cassidy J, Epelbaum R, Dirix L, Cox EH, et al. Population pharmacokinetics and dynamics in phase II studies of the novel bioreductive alkylating cytotoxic indoloquinone EO9. Anticancer Drugs 2001;12:583–90.[Medline]

46 de Jonge MJ, Verweij J, de Bruijn P, Brouwer E, Mathijssen RH, van Alphen RJ, et al. Pharmacokinetic, metabolic, and pharmacodynamic profiles in a dose-escalating study of irinotecan and cisplatin. J Clin Oncol 2000;18:195–203.[Abstract/Free Full Text]

47 Kehrer DF, Sparreboom A, Verweij J, de Bruijn P, Nierop CA, van de Schraaf J, et al. Modulation of irinotecan-induced diarrhea by cotreatment with neomycin in cancer patients. Clin Cancer Res 2001;7:1136–41.[Abstract/Free Full Text]

48 Schellens JH, Heinrich B, Lehnert M, Gore ME, Kaye SB, Dombernowsky P, et al. Population pharmacokinetics and dynamic analysis of the topoisomerase I inhibitor lurtotecan in phase II studies. Invest New Drugs 2002;20:83–93.[Medline]

49 Rinaldi DA, Kuhn JG, Burris HA, Dorr FA, Rodriguez G, Eckhardt SG, et al. A phase I evaluation of multitargeted antifolate (MTA, LY231514), administered every 21 days, utilizing the modified continual reassessment method for dose escalation. Cancer Chemother Pharmacol 1999;44:372–80.[Medline]

50 Kehrer DF, Bos AM, Verweij J, Groen HJ, Loos WJ, Sparreboom A, et al. Phase I and pharmacologic study of liposomal lurtotecan (NX 211): urinary excretion predicts hematologic toxicity. J Clin Oncol 2002;20:1222–31.[Abstract/Free Full Text]

51 Gelderblom H, Mross K, ten Tije AJ, Behringer D, Mielke S, van Zomeren DM, et al. Comparative pharmacokinetics of unbound paclitaxel during 1- and 3-hour infusions. J Clin Oncol 2002;20:574–81.[Abstract/Free Full Text]

52 van der Gaast A, Polee M, Eskens F, Hoekstra R, van der Burg M, Sparreboom A. Phase I and pharmacokinetic study with weekly paclitaxel and increasing doses of weekly carboplatin in patients with esophageal or gastric junction cancer. Proc ASCO 2002;21:155a.

53 Henningsson A, Karlsson MO, Vigano L, Gianni L, Verweij J, Sparreboom A. Mechanism-based pharmacokinetic model for paclitaxel. J Clin Oncol 2001;19:4065–73.[Abstract/Free Full Text]

54 Fokkema E, Verweij J, van Oosterom AT, Uges DR, Spinelli R, Valota O, et al. A prolonged methoxymorpholino doxorubicin (PNU-152243 or MMRDX) infusion schedule in patients with solid tumours: a phase 1 and pharmacokinetic study. Br J Cancer 2000;82:767–71.[Medline]

55 de Jonge MJ, Wortelboer M, van der Gaast A, Morant R, Colajori E, Valota O, et al. PNU-159548, a novel alkycycline: phase I and pharmacokinetic studies in patients with advanced solid tumors. Proc ASCO 2000;19:201a.

56 de Jonge MJ, ten Tije AJ, van der Gaast A, Fiorentini F, Fowst C, Tursi J, et al. Phase I and pharmacokinetic study on PNU-166196, a novel minor groove binder potentiated by glutathione (GSH) and GSH-transferase, administered intravenously once every 3 weeks to patients with advanced cancer. Clin Cancer Res 2001;7:3797s.

57 Eskens FA, Greim GA, van Zuylen C, Wolff I, Denis LJ, Planting AS, et al. Phase I and pharmacological study of weekly administration of the polyamine synthesis inhibitor SAM 486A (CGP 48 664) in patients with solid tumors. European Organization for Research and Treatment of Cancer Early Clinical Studies Group. Clin Cancer Res 2000;6:1736–43.[Abstract/Free Full Text]

58 van Zuylen L, Eskens FA, Bridgewater J, Sparreboom A, Sklenar I, Planting AS, et al. The polyamine synthesis inhibitor SAM486A in combination with 5-FU/LV in metastatic colorectal cancer: results of a phase I and pharmacologic study. Proc ASCO 2000;19:193a.

59 Donehower R, Elze-Brown K, O’Reilly S, Isreal B, Grochow L. A phase I dose escalation, safety, tolerability, and pharmacokinetic study of TAS-103 in patients with refractory solid tumors. Proc ASCO 1998;17:209a.

60 Wolff AC, Baker SD, Bowling MK, Carducci MA, Bradley MO, Anthony FH, et al. Phase I study of taxoprexin DHA-paclitaxel (TXP), a novel taxane with unique preclinical activity, toxicity profile, and pharmacology. Clin Cancer Res 2000;6:4580s.

61 Rowinsky EK, Grochow LB, Hendricks CB, Ettinger DS, Forastiere AA, Hurowitz LA, et al. Phase I and pharmacologic study of topotecan: a novel topoisomerase I inhibitor. J Clin Oncol 1992;10:647–56.[Abstract]

62 Rowinsky EK, Kaufmann SH, Baker SD, Grochow LB, Chen TL, Peereboom D, et al. Sequences of topotecan and cisplatin: phase I, pharmacologic, and in vitro studies to examine sequence dependence. J Clin Oncol 1996;14:3074–84.[Abstract]

63 Rowinsky EK, Grochow LB, Sartorius SE, Bowling MK, Kaufmann SH, Peereboom D, et al. Phase I and pharmacologic study of high doses of the topoisomerase I inhibitor topotecan with granulocyte colony-stimulating factor in patients with solid tumors. J Clin Oncol 1996;14:1224–35.[Abstract]

64 Gelderblom H, Loos WJ, Sparreboom A, van Zomeren DM, de Jonge MJ, Verweij J. Influence of hydration schedule on cisplatin pharmacokinetics in the combination cisplatin followed by topotecan. Proc Am Assoc Cancer Res 2002;43:123.

65 O’Reilly S, Rowinsky EK, Slichenmyer W, Donehower RC, Forastiere AA, Ettinger DS, et al. Phase I and pharmacologic study of topotecan in patients with impaired renal function. J Clin Oncol 1996;14:3062–73.[Abstract]

66 O’Reilly S, Rowinsky E, Slichenmyer W, Donehower RC, Forastiere A, Ettinger D, et al. Phase I and pharmacologic studies of topotecan in patients with impaired hepatic function. J Natl Cancer Inst 1996;88:817–24.[Abstract/Free Full Text]

67 de Bono JS, Stephenson J Jr, Baker SD, Hidalgo M, Patnaik A, Hammond LA, et al. Troxacitabine, an L-stereoisomeric nucleoside analog, on a five-times-daily schedule: a phase I and pharmacokinetic study in patients with advanced solid malignancies. J Clin Oncol 2002;20:96–109.[Abstract/Free Full Text]

68 Dees EC, O’Reilly S, Figg WD, Elza-Brown K, Aylesworth C, Carducci M, et al. A phase I and pharmacologic study of UCN-01, a protein kinase C inhibitor. Proc ASCO 2000;19:205a.

69 Gibaldi M, Perrier D. Pharmacokinetics. New York (NY): Marcel Dekker, Inc.; 1982.

70 Takimoto CH, Baker SD, Sweeney CJ, Goetz A, Felton S, Hammond L, et al. Phase I and pharmacokinetic study of pemetrexed disodium (LY231514, MTA, Alimat) in patients (pts) with impaired renal function. Proc ASCO 2001;20:93a.

71 Smith HW. The kidney, structure and function in health and disease. New York (NY): Oxford University Press; 1951.

72 Dooley MJ, Poole SG. Poor correlation between body surface area and glomerular filtration rate. Cancer Chemother Pharmacol 2000;46:523–6.[Medline]

73 Calvert AH, Egorin MJ. Carboplatin dosing formulae: gender bias and the use of creatinine-based methodologies. Eur J Cancer 2002;38:11–6.[Medline]

74 Baker RJ, Kozoli DD, Meyer KA. The use of surface area for establishing normal blood volume. Surg Gynecol Obstet 1957;104:183–9.[Medline]

75 Wolner E, Domanig E Jr, Elkadi A, Helmer F, Romer P. On a simple relation between blood volume and body surface area. Z Kreislaufforsch 1968;57:79–84.[Medline]

76 Baker SD, Wolff AC, Zabelina Y, McIntyre G, Swindell CS, van Zomeren DM, et al. Disposition of Taxoprexin, DHA-paclitaxel (DHA-PAC), a novel taxane, in blood: in vitro and clinical pharmacokinetic (PK) studies. Clin Cancer Res 2001;7:3677s.

77 Fuse E, Tanii H, Kurata N, Kobayashi H, Shimada Y, Tamura T, et al. Unpredicted clinical pharmacology of UCN-01 caused by specific binding to human alpha1-acid glycoprotein. Cancer Res 1998;58:3248–53.[Abstract]

78 Sparreboom A, Nooter K, Loos WJ, Verweij J. The (ir)relevance of plasma protein binding of anticancer drugs. Neth J Med 2001;59:196–207.[Medline]

79 Sparreboom A, van Zuylen L, Brouwer E, Loos WJ, de Bruijn P, Gelderblom H, et al. Cremophor EL-mediated alteration of paclitaxel distribution in human blood: clinical pharmacokinetic implications. Cancer Res 1999;59:1454–7.[Abstract/Free Full Text]

80 Sparreboom A, Verweij J, van der Burg ME, Loos WJ, Brouwer E, Vigano L, et al. Disposition of Cremophor EL in humans limits the potential for modulation of the multidrug resistance phenotype in vivo. Clin Cancer Res 1998;4:1937–42.[Abstract]

81 van Zuylen L, Karlsson MO, Verweij J, Brouwer E, de Bruijn P, Nooter K, et al. Pharmacokinetic modeling of paclitaxel encapsulation in Cremophor EL micelles. Cancer Chemother Pharmacol 2001;47:309–18.[Medline]

Manuscript received April 8, 2002; revised October 7, 2002; accepted October 15, 2002.


This article has been cited by other articles in HighWire Press-hosted journals:


Correspondence about this Article

             
Copyright © 2002 Oxford University Press (unless otherwise stated)
Oxford University Press Privacy Policy and Legal Statement