9
Camptothecins in Cancer Therapy
Edited by: V. R. Adams and T. G. Burke © Humana Press Inc., Totowa, NJ 193
Clinical Experience
With 9-Aminocamptothecin
Lessons for New Drug Development
Chris H. Takimoto, MD , P h D
C
ONTENTSI
NTRODUCTIONH
ISTORICALB
ACKGROUNDC
LINICALD
EVELOPMENTL
ESSONS FORN
EWD
RUGD
EVELOPMENTC
ONCLUSIONSR
EFERENCES1. INTRODUCTION
In preclinical studies first published in 1989 by Giovanella and col- leagues, 9-aminocamptothecin (9-AC) demonstrated highly promising anti- tumor activity against human colorectal cancer xenografts (1). This led to an extensive drug development program for 9-AC initially under the guidance of the National Cancer Institute, and later in cooperation with pharmaceu- tical industry sponsors. However, despite this impressive preclinical activ- ity, 9-AC has not demonstrated clinically useful antitumor activity to date.
Thus, in contrast to its more successful brethren, irinotecan and topotecan,
9-AC is no longer under active clinical development. Nonetheless, testing
of other novel and new camptothecin derivatives remains an active area of
pharmacological research. Thus a careful review of the early clinical and
pharmacological trials of 9-AC may help us to understand why 9-AC has not
shown more meaningful antitumor activity and may also provide important
lessons for the future drug development of novel camptothecins and other
topoisomerase I (TOP-I) targeting agents.
2. HISTORICAL BACKGROUND
In 1986, Drs. Wani and Wall first synthesized the water-insoluble camptothecin derivative, 9-AC (2). Subsequent laboratory studies revealed that 9-AC was a potent inhibitor of TOP-I (3,4), and its ability to interfere with this enzyme correlated with its in vivo cytotoxic activity against murine L1210 leukemia cells (3,5). In 1989, Giovanella and colleagues published their impressive results in the journal Science documenting the activity of 9-AC in human colon cancer xenografts models in nude mice (1).
9-AC was highly active with minimal systemic toxicity and generated better antitumor responses than a number of different anticancer agents including 5-fluorouracil, doxorubicin, melphalan, methotrexate, vincristine, vinblas- tine, and others (1). Interest in clinically developing this compound was further heightened by the demonstration of a broad range of activity against a diverse group of human tumor xenografts including malignant melanoma
(6), acute leukemia (7), central nervous system tumors (8), and ovarian (9),prostate (10), breast (11), and bladder cancers (12).
In animal studies, the principal dose-limiting toxicities of 9-AC were neutropenia and gastrointestinal toxicity. Preclinical pharmacological stud- ies also revealed that brief intravenous infusions generating high peak con- centrations for short periods were toxic and relatively inactive against human tumor xenografts. Instead, subcutaneous administration of 9-AC as a sus- pension in Tween-80 resulted in sustained plasma concentrations of 9-AC lactone in the nanomolar range and generated antitumor responses (13).
3. CLINICAL DEVELOPMENT
In 1989, the US National Cancer Institute selected 9-AC as a high-prior- ity compound for further clinical development. However, clinical trials with this agent were delayed because of difficulties in formulating this com- pound resulting from poor aqueous solubility. In 1993, 9-AC was success- fully formulated in dimethylacetamide, polyethylene glycol, and phosphoric acid for use in phase I clinical trials. Later, in 1995, Pharmacia & Upjohn developed a newer colloidal dispersion formulation of 9-AC that was supe- rior in stability and in compatibility with standard intravenous infusions.
This newer colloidal dispersion formulation has been used in the more recent clinical studies of this agent.
3.1. Phase I Trials
Because preclinical studies highlighted the importance of prolonged
exposures to 9-AC lactone, the first phase I trials of 9-AC dimethylacetamide
used a continuous 72-hour intravenous infusion administered every 2 (14)
or every 3 (15,16) weeks (Table 1). Both schedules yielded similar toxicity
Table 1 9-Aminocamptothecin (9-AC) Phase I Clinical Trials 9-AC Recommended phase II regimenformulationPrincipal toxicitiesReference 0.84 mg/m2/day CIV × 3 days every 2 weeks, orDMANeutropenia14 1.13 mg/m2/day CIV × 3 days every 2 weeks with G-CSFNeutropenia and some thrombocytopenia 1.08 mg/m2/day CIV × 3 days every 3 weeksDMANeutropenia and some thrombocytopenia15 1.3 mg/m2/day CIV × 3 day every 3 weeksCDNeutropenia16 0.48 mg/m2/day CIV × 5 days weekly for 3 of 4 weeksDMANeutropenia17 Diarrhea 0.6 mg/m2/day CIV × 5 days weekly for 2 of 3 weeksCDNeutropenia 0.4 mg/m2/day CIV × 7 days every 4 weeksDMANeutropenia and some thrombocytopenia51 0.51-0.6 mg/m2/day CIV × 7 days every 4 weeksCD 1.65 mg/m2 CIV over 24 hours weekly for 4 of 5 weeksCDNeutropenia18 1.1 mg/m2/day IV over 30 minutes daily × 5 days every 3 weeksCDNeutropenia19 Oral Preparations No recommended oral dose daily × 5 every 2 weeksCD (oral)No dose-limiting toxicity determined20 0.84 mg/m2 orally daily × 14 days every 3 weeksPEG-1000Myelosuppression and diarrhea21 Specialized phase I studies Pediatric patients 1.25 mg/m2/day CIV × 3 days every 3 weeksDMANeutropenia Thrombocytopenia26 Acute leukemia1.4 mg/m2/day CIV over 7 daysCDMucositis27 IV, intravenous; CIV, continuous intravenous infusion; G-CSF, granulocyte-colony-stimulating factor; DMA, dimethylacetamide; CD, colloidal dispersion.
profiles with dose escalation limited by neutropenia and, less commonly, thrombocytopenia. Other toxicities included mild to moderate fatigue, ane- mia, nausea and vomiting, diarrhea, alopecia, and mucositis. In contrast to irinotecan, severe diarrhea was not observed on the 72-hour infusion sched- ules. Additional schedules of 9-AC administration tested in a phase I clini- cal trials include prolonged 120-hour infusions given weekly for 2 or 3 consecutive weeks followed by a week rest period (17) and a weekly 24- hour infusion given for 4 of every 5 weeks (18) (Table 1). For most of these regimens, the major dose-limiting toxicity was still myelosuppression;
however, diarrhea became more pronounced when prolonged administra- tion schedules were tested (17).
More recently, a short 15-minute infusion given daily for 5 days every 3 weeks was developed for clinical testing (19). On this schedule, neutropenia and thrombocytopenia were dose limiting and the recommended phase II dose was 1.1 mg/m
2/day infused over 15 minutes daily for 5 days every 3 weeks. The colloidal dispersion formulation of 9-AC has also been admin- istered to patients orally over a dose range of 0.2 to 0.68 mg/m
2/day daily
(20). However, in this study, no recommended phase II oral dose was deter-mined because of large interpatient variability in the area-under-the-con- centration versus time curve. Another oral study used 9-AC formulated as a polyethylene glycol 1000 capsule with much greater success (21). Admin- istration of 9-AC capsules over a dose range of 0.25 to 1.5 mg/m
2/day for 14 days every 3 weeks resulted in a recommended phase II dose of 1.1 mg/
m
2/day. Neutropenia and thrombocytopenia were dose limiting, and the oral bioavailability of 9-AC in capsular form was a relatively consistent 48
±17.8%.
3.2. Clinical Pharmacology
All camptothecin derivatives, including 9-AC, contain a terminal lactone
ring that rapidly hydrolyzes in aqueous solutions to form the more hydro-
philic open-ring carboxylic acid species. Under nonacidic conditions, the
equilibrium for this reaction very much favors a less active carboxylate
form. The pharmacokinetics of the lactone and carboxylate species of 9-AC
have been extensively examined (Table 2). Overall, the amount of active
lactone 9-AC circulating in plasma is quite low relative to the total (lactone
+ carboxylate) 9-AC, with most studies reporting a lactone-to-total 9-AC
plasma ratio of less than 0.16. This value is lower than the active lactone
forms of topotecan (0.16–0.2) (22) or irinotecan (0.51) (23,24) that circulate
in plasma. In addition, the plasma half-life for 9-AC lactone species is
relatively short, ranging from 4.5 to 10 hours (18,19,25–27). Urinary excre-
tion of unchanged drug ranges from 8.6 to 32.1% of the administered dose
(19,25).Table 2 9-Aminocamptothecin Clinical Pharmacology Volume ofPercent Terminaldistribution atUrinarylactone in half-lifesteady-stateCLexcretionplasma Dose and scheduleFormulationCompound(hours)(L/m2)(L/hour/m2)(%)(%)Reference 0.12–1.8 mg/m2/dayDMALactone4.47 ± 0.53195 ± 11424.5 ± 7.3NR8.7 ± 4.725 IV over 72 hoursTotal8.38 ± 2.123.6 ± 10.6NR32.1 ± 8.3 (over 24 hours) 1.08 mg/m2/day IVDMALactone42.2 ± 34.412.6 ± 3.655.2 ± 18.0NRNR15 over 72 hours 0.86–1.49 mg/m2/dayDMALactone7.1 ± 3.5135.3 ± 52.5NRNR10.8 ± 3.626 IV over 72 hours*Total8.1 ± 3.821.2 ± 13.3NRNR 0.7–1.9 mg/m2/dayCDLactone10.7 ± 6.7111.0 ± 72.318.0 ± 9.0NR7.9918 IV over 24 hours 0.9–1.55 mg/m2/dayCDLactone22.5 ± 8.5325 ± 14530.3 ± 4.5NRNR16 IV over 72 hours 0.45–1.4 mg/m2/dayCDLactone4.3 ± 2.758.4 ± 23.311.3 ± 4.9NRNR27 CIV over 7 days 0.48 mg/m2/day CIVDMALactone NRNR12.3 ± 7.1NR13.817 over 5 daysTotal NRNR1.65 ± 0.87NR 0.7–1.9 mg/m2/dayCDLactone10.7 ± 6.7111.0 ± 72.318.0 ± 9.0NR7.9918 IV over 24 hoursTotal7.7± 4.617.7 ± 6.02.36 ± 1.248.6 ± 4.0NR IV, intravenous,; CIV, continuous intravenous infusion; DMA, dimethylacetamide formulation; CD, colloidal dispersion formulation; NR, not reported. *Pediatric patients.
In a pharmacological study of 9-AC in brain cancer patients, subjects receiving anticonvulsant medications tolerated much higher doses of 9-AC consistent with an increased clearance of drug (28). One possible explana- tion is that 9-AC hepatic clearance may be increased by enzyme induction caused by antiepileptic drugs such as phenytoin, carbamazepine, phenobar- bital, primidone, felbamate, and valproic acid. However, hepatic metabo- lites of 9-AC have not been characterized (29). An alternative explanation is that these same enzyme inducing antiepileptic agents may also result in the increase expression of transport proteins leading to increased 9-AC clearance and biliary excretion (30).
Large interpatient variation in the plasma kinetics of 9-AC has been consistently observed in most studies, which is similar to clinical pharma- cological studies of other camptothecin derivatives such as topotecan (31) and irinotecan (23,24). In pharmacodynamic studies, moderate to strong correlations between dose-limiting myelosuppression and 9-AC steady state plasma concentrations (25,30) or overall area under the concentration-time curve (AUC) (18,19,32) have been observed.
3.3. Antitumor Activity
The antitumor activity of 9-AC administered as a 72-hour infusion has been examined in a variety of different tumors (Table 3). In phase II studies in advanced colorectal cancer, administration of 9-AC at 1.2 to 1.4 mg/m
2/ day over 72 hours every 2 weeks to 16 previously untreated patients resulted in no objective responses (33). However, the myelosuppressive toxicity of this regimen was substantial, with grade 4 neutropenia seen in 56% of patients and febrile neutropenia documented in 31% (33). No objective antitumor activity was also seen in another trial examining 9-AC administered at 0.84 mg/m
2/day over 72 hours every 2 weeks in 17 untreated colorectal cancer patients (34). In contrast, in advanced platinum refractory ovarian cancer patients, 72-hour infusions of 9-AC were active generating response rates of 21% (35) and 19% (36). Activity in relapsed or refractory lymphoma patients was also observed (37). Administration of 9-AC at 0.96 mg/m
2over 72 hours every 3 weeks with granulocyte colony-stimulating factor to 40 patients with advanced lymphoma resulted in a partial response rate of 25%
(95% confidence interval [CI] 13–41%) and a median survival of 12.5 months. In advanced cutaneous T-cell lymphoma, 9-AC at 1.1 mg/m
2/day every 2 weeks with granulocyte colony-stimulating factor resulted in partial responses in 2 of 12 patients for an overall rate of 17%, but myelosuppressive toxicity was high and 3 patients died from neutropenia and sepsis (38).
Minimal activity has also been reported in non-small-cell lung, central nervous system, and breast cancers (Table 3).
More prolonged infusion schedules administering 9-AC over 120 hours weekly for 3 of 4 weeks have also been tested in a limited number of studies.
In 17 previously untreated advanced colorectal cancer patients, administra-
tion of 0.48 mg/m
2/day for 120 hours weekly for 3 of 4 weeks again resulted in no objective responses (39). In lung cancer, administration of 120-hour infusions of 9-AC to stage IIIB and IV non-small-cell lung cancer patients generated only a single response in 12 previously untreated patients and produced a 1-year survival rate of 28% (95% CI = 5–58%) and a median survival of 10.2 months (40).
The rights to this agent were transferred in 1997 from the Pharmacia &
Upjohn Company to the IDEC Pharmaceutical Corporation. In this same year, phase II testing of 9-AC was initiated using a 1.1 mg/m
2intravenous infusion daily for a 5-day schedule (19). The results of phase II testing of 9- AC on this schedule have not been formally reported; however, in 1999 IDEC notified the US government that it was stopping the development of 9-AC based on preliminary phase II results (41). Thus, despite showing impressive activity in preclinical models of colorectal cancer, 9-AC did not produce clinically meaningful antitumor activity of 9-AC in patients with this disease. Modest activity has been observed in refractory lymphoma patients and in women with platinum-refractory ovarian cancer, but the overall phase II activity of 9-AC on any schedule has been largely disap- pointing.
4. LESSONS FOR NEW DRUG DEVELOPMENT This lack of clinically useful antitumor activity raises several key ques- tions. First, why were the preclinical animal xenograft studies of 9-AC so promising initially given the later clinical findings? Second, what pharma- cological characteristics make camptothecin derivatives such as topotecan and irinotecan so much more clinically successful than 9-AC?
The first question was explored in detail in an interesting study by Kirstein et al. (42). Careful analysis of the pharmacological characteristics of 9-AC in nude mouse experiments were compared with the clinical pharmacologi- cal findings seen in early human studies of 9-AC. They adopted a novel approach first suggested by Minderman and colleagues (43). This involved rigorously defining the minimally effective dose of 9-AC that caused objec- tive tumor regressions in animals (MEDOR). However, Kirstein and col- leagues expanded the MEDOR concept to include a minimally effective threshold exposure to 9-AC in plasma defined in terms of the AUC, thereby generating a new parameter that could more easily be applied across species to preclinical and clinical studies. The administration of 9-AC daily for 5 days for 2 consecutive weeks generated a MEDOR AUC of 690–1580 ng/
mL/hour in the nude mouse experiments (42). The mouse MEDOR AUC
was then compared with the AUC profiles generated in humans in Phase I
clinical trials. Unfortunately, dose-limiting myelosuppressive toxicity in
humans limited the 9-AC AUC to a substantially lower range of 126–493
ng/mL/hour. The greater sensitivity of human bone marrow to 9-AC limited
Table 3 9-Aminocamptothecin Phase II Clinical Trials PreviousMedianMedian therapy forduration ofduration of EvaluatedadvancedResponse rateResponseresponsesurvival Tumor type9-AC regimenpatientsdisease (%) type(months) (months)Reference Colorectal1.42 mg/m2/day IV for 72 hours14Yes0 (95% CI 0–20)0 PRNR9.533 every 2 weeks + GCSF(95% CI 7.3–12.9) Colorectal0.84 mg/m2/day IV for 72 hours17No00 PRNRNR34 every 2 weeks Colorectal0.48 mg/m2/day IV for 12017No00 PRNR8 (0.75–14.5)39 hours weekly for 3 of 4 weeks Nonsmall-cell lung1.42 mg/m2/day IV for 72 hours54No9 (95% CI 3–19)5 PR2.25–7.05.452 every 2 weeks + GCSF Nonsmall-cell lung0.6 mg/m2/day IV for 120 hours12No8 (95% CI 0–21)1 PRNR10.240 weekly for 2 of 3 weeks Refractory Hodgkin’s0.96 mg/m2/day IV for 72 hours40Yes25 (95% CI 13–41)10 PR5 (r 1–10)12.537 and non-Hodgkin’severy 3 weeks ± G-CSF lymphoma Central nervous0.85–1.78 mg/m2/day IV39NR31 PRNRNR53 systemfor 72 hours + 2 weeks Head and neck0.85–1.0 mg/m2/day IV14No00 PRNR6 (1–9+)54 for 72 hours + 2 weeks
Ovarian0.84–1.13 mg/m2/day IV28Yes216 PRNRNR35 for 72 hours + 2 weeks with GCSF Ovarian0.84 mg/m2/day IV for 72 hours27Yes19 (95% CI 6–38)5 PRNRNR36 + 2 weeks Breast1.1 mg/m2/day IV for 72 hours15Yes132 PR3.5-5NR55 every 2 weeks + GCSF Cutaneous T-cell1.1 mg/m2/day IV for 72 hours12Yes172 PRNR538 lymphomaevery 2 weeks +GCSF IV, intravenous; G-CSF, granulocyte-co;ony stimulating factor; PR, partial response; NR, not reported; CI, confidence interval.
the plasma AUC in humans to levels below the minimally effective expo- sures range required for optimal anticancer activity. Even modest decreases in achievable systemic exposure can mean the difference between success and failure for drugs, such as the camptothecins, that have steep dose response curves and narrow therapeutic indices. This analysis is also quite consistent with previous findings showing that mouse bone marrow pro- genitor cells are 6 to 11 times more resistant to 9-AC than their human counterparts (44). These studies predict, a priori, that maximal achievable concentrations of 9-AC in plasma would be lower in humans compared with mice because of marked differences in host tolerance to toxic drug effects.
Kirstein and colleagues recommended that careful pharmacokinetic and pharmacodynamic studies be performed both in preclinical experiments and in early clinical trials. Better communication between preclinical scien- tists and physicians designing and conducting phase I clinical trials may further help to streamline the drug development process.
The second question of which pharmacological characteristics of topotecan and irinotecan make them more clinically useful than 9-AC is difficult to answer with certainty. Differences in the clinical pharmacology of these camptothecin derivatives are quite prominent. For example, both irinotecan and topotecan are much more water-soluble than 9-AC and are easier to formulate in pharmaceutical preparations. Furthermore, the lac- tone forms of both of these camptothecins are more stable than 9-AC in plasma with higher percentage of the active lactone species circulating in plasma after intravenous administration (25). For topotecan it is 16–20%
(22), irinotecan (SN-38) 51–64% (23,24), and for 9-AC approximately 9%
(25). The tight protein binding of the open ring 9-AC carboxylate to plasma
albumin, which tends to destabilize the lactone species (45), may explain this difference. In contrast, the protein binding of topotecan is much less, and for SN-38, plasma protein binding interactions actually tend to favor the stability of the active lactone species (46). Another kinetic difference is found in the half-lives of these agents. The half-life of SN-38 is relatively long, 11 ± 3 hours, whereas for topotecan it is 3.01 ± 0.54 hours and for 9- AC it is 4.5 ± 0.5 hours. For this reason, irinotecan can be given as a rela- tively short infusion over 90 minutes every 3 weeks (23,24), whereas topotecan and 9-AC are administered on consecutive days or as prolonged infusions over many days.
Additional differences at the molecular level of action may also be impor-
tant in distinguishing between the different camptothecin analogues. For
example, SN-38 is highly potent as an inhibitor of TOP-I and generates
cleavable complexes in colon cancer cells that are more stabile than those
generated by 9-AC or topotecan (47). Molecular resistance mechanisms
may also differ. P-glycoprotein-associated multidrug resistance may have
some impact on the activity of some of these agents. Multidrug resistant
(MDR)-expressing cell lines are ninefold more resistant to topotecan and twofold more resistant to 9-AC (48) compared with parental wild-type cells.
In contrast, irinotecan and SN-38 are not effective substrates for the MDR drug efflux pump (49) and cross-resistance between irinotecan and vincris- tine or doxorubicin is not observed in P388 leukemia cells expressing the MDR phenotype (50).
In summary, 9-AC is more similar to topotecan than irinotecan. For example, both 9-AC and topotecan are not a prodrugs, they are a weak MDR substrates and both have relatively short half-lives in plasma after intrave- nous administration. Topotecan and 9-AC both exhibit dose-limiting toxici- ties consisting of myelosuppression rather than diarrhea and both have shown activity in ovarian cancer. Consequently, 9-AC was simply not unique enough to justify its further clinical development as a novel TOP-I- targeting agent.
5. CONCLUSIONS
The great promise of 9-AC seen in preclinical studies has not resulted in clinically useful antitumor activity. The reasons for this failure have been reviewed and provide some insights into further drug development in this area. However, there are no easy answers. Fundamentally, we still need to have a much better understanding of the pharmacology of these agents.
Important and as-yet poorly understood issues include how these different camptothecins function at the molecular level, what are their key determi- nants of antitumor response, and what are their clinically relevant mecha- nisms of drug resistance? Despite these challenges, there is no doubt that TOP-I is a validated molecular target for anticancer therapy and the future drug development efforts are sure to result in new generations of useful and better agents that target this enzyme.
REFERENCES
1. Giovanella BC, Stehlin JS, Wall ME, et al. 1989 DNA topoisomerase I—targeted chemotherapy of human colon cancer in xenografts. Science 246:1046–1048.
2. Wani MC, Nicholas AW, Wall ME. 1986 Plant antitumor agents. 23. Synthesis and antileukemic activity of camptothecin analogues. J Med Chem 29:2358–2363.
3. Jaxel C, Kohn KW, Wani MC, Wall ME, Pommier Y. 1989 Structure-activity study of the actions of camptothecin derivatives on mammalian topoisomerase I:
evidence for a specific receptor site and a relation to antitumor activity. Cancer Res 49:1465–1469.
4. Kingsbury WD, Boehm JC, Jakas DR, et al. 1991 Synthesis of water-soluble (aminoalkyl)camptothecin analogues: inhibition of topoisomerase I and antitumor activity. J Med Chem 34:98–107.
5. Hsiang YH, Liu LF, Wall ME, et al. 1989 DNA topoisomerase I-mediated DNA cleavage and cytotoxicity of camptothecin analogues. Cancer Res 49:4385–4389.
6. Pantazis P, Hinz HR, Mendoza JT, et al. 1992 Complete inhibition of growth followed by death of human malignant melanoma cells in vitro and regression of human melanoma xenografts in immunodeficient mice induced by camptothecins.
Cancer Res 52:3980–3987.
7. Jeha S, Kantarjian H, O’Brien S, Vitek L, Beran M. 1998 Activity of oral and intravenous 9-aminocamptothecin in SCID mice engrafted with human leukemia.
Leuk Lymph 32:159–164.
8. Potmesil M, Vardeman D, Kozielski AJ, Mendoza J, Stehlin JS, Giovanella BC.
1995 Growth inhibition of human cancer metastases by camptothecins in newly developed xenograft models. Cancer Res 55:5637–5641.
9. Pantazis P, Kozielski AJ, Mendoza JT, Early JA, Hinz HR, Giovanella BC. 1993 Camptothecin derivatives induce regression of human ovarian carcinomas grown in nude mice and distinguish between non-tumorigenic and tumorigenic cells in vitro. Int J Cancer 53:863–871.
10. de Souza PL, Cooper MR, Imondi AR, Myers CE. 19979-Aminocamptothecin: a topoisomerase I inhibitor with preclinical activity in prostate cancer. Clin Cancer Res 3:287–294.
11. Pantazis P, Kozielski AJ, Vardeman DM, Petry ER, Giovanella BC. 1993 Efficacy of camptothecin congeners in the treatment of human breast carcinoma xenografts.
Oncol Res 5:273–281.
12. Keane TE, El-Galley RE, Sun C, et al. 1998 Camptothecin analogues/cisplatin: an effective treatment of advanced bladder cancer in a preclinical in vivo model system. J Urol 160:252–256.
13. Potmesil M, Arbuck SG, Takimoto CH, Liebes L, Hochster H. 1996 9-Amino- camptothecin and beyond. Preclinical and clinical studies. Ann N Y Acad Sci 803:231–246.
14. Dahut W, Harold N, Takimoto C, et al. 1996 Phase I and pharmacologic study of 9-aminocamptothecin given by 72-hour infusion in adult cancer patients. J Clin Oncol 14:1236–1244.
15. Rubin E, Wood V, Bharti A, et al. 1995 A phase I and pharmacokinetic study of a new camptothecin derivative, 9- aminocamptothecin. Clin Cancer Res 1:269–276.
16. Eder JP, Jr., Supko JG, Lynch T, et al. 1998 Phase I trial of the colloidal dispersion formulation of 9-amino-20(S)- camptothecin administered as a 72-hour continu- ous intravenous infusion. Clin Cancer Res 4:317–324.
17. Thomas RR, Dahut W, Harold N, et al. 2001 A phase I and pharmacologic study of 9-aminocamptothecin administered as a 120-h infusion weekly to adult cancer patients. Cancer Chemother Pharmacol 48:215–222.
18. Siu LL, Oza AM, Eisenhauer EA, et al. 1998 Phase I and pharmacologic study of 9-aminocamptothecin colloidal dispersion formulation given as a 24-hour con- tinuous infusion weekly times four every 5 weeks. J Clin Oncol 16:1122–1130.
19. Herben VM, van Gijn R, Schellens JH, et al. 1999 Phase I and pharmacokinetic study of a daily times 5 short intravenous infusion schedule of 9-amino- camptothecin in a colloidal dispersion formulation in patients with advanced solid tumors. J Clin Oncol 17:1906–1914.
20. Mani S, Iyer L, Janisch L, et al. 1998 Phase I clinical and pharmacokinetic study of oral 9-aminocamptothecin (NSC-603071). Cancer Chemother Pharmacol 42:84–87.
21. de Jonge MJ, Punt CJ, Gelderblom AH, et al. 1999 Phase I and pharmacologic study of oral (PEG-1000) 9-aminocamptothecin in adult patients with solid tu- mors. J Clin Oncol 17:2219–2226.
22. Rowinsky EK, Grochow LB, Hendricks CB, et al. 1992 Phase I and pharmacologic study of topotecan: a novel topoisomerase I inhibitor. J Clin Oncol 10:647–656.
23. Rowinsky EK, Grochow LB, Ettinger DS, et al. 1994 Phase I and pharmacological study of the novel topoisomerase I inhibitor 7-ethyl-10-[4-(1-piperidino)-1- piperidino]carbonyloxycamptothecin (CPT-11) administered as a ninety- minute infusion every 3 weeks. Cancer Res 54:427–436.
24. Rothenberg ML, Kuhn JG, Burris HA, et al. 1993 Phase I and pharmacokinetic trial of weekly CPT-11. J Clin Oncol 11:2194–2204.
25. Takimoto CH, Dahut W, Marino MT, et al. 1997 Pharmacodynamics and pharma- cokinetics of a 72-hour infusion of 9- aminocamptothecin in adult cancer patients.
J Clin Oncol 15:1492–1501.
26. Langevin AM, Casto DT, Thomas PJ, et al. 1998 Phase I trial of 9-amino- camptothecin in children with refractory solid tumors: a pediatric oncology group study. J Clin Oncol 16:2494–2499.
27. Vey N, Kantarjian H, Tran H, et al. 1999 Phase I and pharmacologic study of 9- aminocamptothecin colloidal dispersion formulation in patients with refractory or relapsed acute leukemia. Ann Oncol 10:577–583.
28. Grossman SA, Hochberg F, Fisher J, et al. 1998 Increased 9-aminocamptothecin dose requirements in patients on anticonvulsants. NABTT CNS Consortium. The new approaches to brain tumor therapy. Cancer Chemother Pharmacol 42:118–126.
29. Takimoto CH, Dahut W, Harold N, et al. 1996 Clinical pharmacology of 9- aminocamptothecin. Ann N Y Acad Sci 803:324–326.
30. Minami H, Lad TE, Nicholas MK, Vokes EE, Ratain MJ. 1999 Pharmacokinetics and pharmacodynamics of 9-aminocamptothecin infused over 72 hours in phase II studies. Clin Cancer Res 5:1325–1330.
31. Grochow LB, Rowinsky EK, Johnson R, et al. 1992 Pharmacokinetics and phar- macodynamics of topotecan in patients with advanced cancer. Drug Metab Dispos 20:706–713.
32. de Jonge MJ, Verweij J, Loos WJ, Dallaire BK, Sparreboom A. 1999 Clinical pharmacokinetics of encapsulated oral 9-aminocamptothecin in plasma and saliva.
Clin Pharmacol Ther 65:491–499.
33. Saltz LB, Kemeny NE, Tong W, Harrison J, Berkery R, Kelsen DP. 1997 9- Aminocamptothecin by 72-hour continuous intravenous infusion is inactive in the treatment of patients with 5-fluorouracil-refractory colorectal carcinoma. Cancer 80:1727–1732.
34. Pazdur R, Diaz-Canton E, Ballard WP, et al. 1997 Phase II trial of 9-amino- camptothecin administered as a 72-hour continuous infusion in metastatic colorectal carcinoma. J Clin Oncol 15:2905–2909.
35. McCarthy N, Sarosy G, Minasian L, et al. 1999 Phase II and pharmacokinetic (PK) study of 9-aminocamptothecin (9AC) in recurrent epithelial ovarian cancer [abstract]. Proc Am Soc Clin Oncol 18:363a.
36. Speyer J, Hochster H, Mandeli J, Runowicz C, Krook J, Wadler S. 1999 Phase II study of 72 hr 9-amino-camptothecin (9AC) infusion in second-line therapy of ovarian cancer (a NYGOG and ECOG study) [abstract]. Proc Am Soc Clin Oncol 18:363a.
37. Wilson WH, Little R, Pearson D, et al. 1998 Phase II and dose-escalation with or without granulocyte colony- stimulating factor study of 9-aminocamptothecin in relapsed and refractory lymphomas [published erratum appears in J Clin Oncol 1998;16(8):2895]. J Clin Oncol 16:2345–2351.
38. Argiris A, Heald P, Kuzel T, et al. 2001 Phase II trial of 9-aminocamptothecin as a 72-h infusion in cutaneous T-cell lymphoma. Invest New Drugs 19:321–326.
39. Pazdur R, Medgyesy DC, Winn RJ, et al. 1998 Phase II trial of 9-amino- camptothecin (NSC 603071) administered as a 120-hr continuous infusion weekly for three weeks in metastatic colorectal carcinoma. Invest New Drugs 16:341–346.
40. Vokes EE, Gordon GS, Rudin CM, et al. 2001 A phase II trial of 9-amino- captothecin (9-AC) as a 120-h infusion in patients with non-small cell lung cancer.
Invest New Drugs 19:329–333.
41. IDEC Pharmaceuticals Corporation. Letter to Stockholders, July 22, 1999. San Diego, California.
42. Kirstein MN, Houghton PJ, Cheshire PJ, et al. 2001 Relation between 9- aminocamptothecin systemic exposure and tumor response in human solid tumor xenografts. Clin Cancer Res 7:358–366.
43. Minderman H, Cao S, Rustman YM. 1998 Rational design of irinotecan adminis- tration based on preclinical models. Oncology (Huntingt) 12:22–30.
44. Erickson-Miller CL, May RD, Tomaszewski J, et al. 1997 Differential toxicity of camptothecin, topotecan and 9- aminocamptothecin to human, canine, and murine myeloid progenitors (CFU- GM) in vitro. Cancer Chemother Pharmacol 39:467–472.
45. Mi Z, Malak H, Burke TG. 1995 Reduced albumin binding promotes the stability and activity of topotecan in human blood. Biochemistry 34:13722–13728.
46. Burke TG, Mi Z. 1994 The structural basis of camptothecin interactions with human serum albumin: impact on drug stability. J Med Chem 37:40–46.
47. Tanizawa A, Fujimori A, Fujimori Y, Pommier Y. 1994 Comparison of topoisomerase I inhibition, DNA damage, and cytotoxicity of camptothecin derivatives presently in clinical trials. J Natl Cancer Inst 86:836–842.
48. Chen AY, Yu C, Potmesil M, Wall ME, Wani MC, Liu LF. 1991 Camptothecin overcomes MDR1-mediated resistance in human KB carcinoma cells. Cancer Res 51:6039–6044.
49. Jansen WJ, Hulscher TM, van Ark-Otte J, Giaccone G, Pinedo HM, Boven E. 1998 CPT-11 sensitivity in relation to the expression of P170-glycoprotein and multidrug resistance-associated protein. Br J Cancer 77:359–365.
50. Tsuruo T, Matsuzaki T, Matsushita M, Saito H, Yokokura T. 1988 Antitumor effect of CPT-11, a new derivative of camptothecin, against pleiotropic drug- resistant tumors in vitro and in vivo. Cancer Chemother Pharmacol 21:71–74.
51. Hochster H, Liebes L, Speyer J, et al. 1997 Phase I and pharmacodynamic study of prolonged infusion 9-amino-camptothecin (9AC) in two formulations [abstract].
Proc Am Soc Clin Oncol 16:704.
52. Vokes EE, Ansari RH, Masters GA, et al. 1998 A phase II study of 9-amino- camptothecin in advanced non-small-cell lung cancer. Ann Oncol 9:1085–1090.
53. Hochberg F, Grossman SA, Mikkelsen T, Calabresi P, Fisher J, Piantadosi S. 1998 Efficacy of 9-aminocamptothecin (9-AC) in adults with newly diagnosed glioblas- toma multiforme (GBM) and recurrent high grade astrocytomas [abstract]. Proc Am Soc Clin Oncol 17:388a.
54. Lad T, Rosen F, Arietta R, Kuebler JP, Vokes E. 1998 Phase II trial of 9- aminocamptothecin (9AC/DMA) in patients with advanced squamous cell head and neck cancer [abstract]. Proc Am Soc Clin Oncol 17:392a.
55. Kraut EH, Balcerzak SP, Young D, et al. 2000 A phase II study of 9-amino- camptothecin in patients with refractory breast cancer. Cancer Invest 18:28–31.