Affiliation of authors: National Cancer Institute of Canada Clinical Trials Group, Queens University, Kingston, Ontario, Canada.
Correspondence to: Wendy Parulekar, MD, FRCPC, National Cancer Institute of Canada Clinical Trials Group, 10 Stuart St., Queens University, Kingston, ON, Canada K7L 3N6 (e-mail: wparulekar{at}ctg.queensu.ca)
![]() |
ABSTRACT |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
![]() |
INTRODUCTION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
The emergence of targeted, so-called "non-cytotoxic" therapies as anticancer agents may challenge the traditional phase I study paradigm in a variety of ways (26). Unlike cytotoxic agents, most of which act on DNA or tubulin, these new therapies have myriad targets, including membrane receptors, components of cytoplasmic signaling pathways, cell cycle regulator proteins, and proteins or factors important in angiogenesis. Because the resulting antineoplastic effects may be cytostatic (i.e., inhibit tumor growth or prevent metastases) rather than cytotoxic, early efficacy trials may need to incorporate measures of antitumor behavior other than changes in tumor size. In addition to different mechanisms of action and potential antitumor effects, these novel compounds may also be characterized by a lack of clinically significant organ toxicity compared with conventional chemotherapy. Thus, although determination of the recommended phase II dose using toxicity as a surrogate endpoint for activity may be unnecessary or unachievable in the phase I setting for these agents and therapies, demonstration that the agents have the desired target effect is an important aspect of their early clinical development.
Alternatives to toxicity as a surrogate endpoint for phase I dose escalation trials evaluating non-cytotoxic therapies can include measurement of target inhibition and/or pharmaco- kinetic analysis. Although measurement of a molecular target effect seems logical, it is associated with several challenges. First, given the complexity of cellular pathways and signaling processes, it may be difficult to define the appropriate measure of achieved target effects for a specific drug. Second, restricting patient enrollment to those with accessible disease for assessment of the drug effect on the tumor decreases the eligible population and puts an additional level of ethical and administrative burden on the conduct of the trial. Even if patients consent to tumor biopsy, serial tumor sampling is invasive and associated with sampling errors resulting from the hetero- geneous tissue composition of cancers. The use of surrogate tissues such as skin, mucosa, or peripheral blood may be an appropriate solution to these problems, provided that changes in the surrogate tissue parallel those in the tumor in preclinical studies. Third, the optimal level of "target inhibition" must be defined. Finally, a reliable assay for measurement of the drug effect must be available. Pharmacokinetic endpoints, such as achieving target plasma levels of the drug, may help with phase I study dose selection of the non-cytotoxic drug. However, pharmacokinetic endpoints are appropriate only if sufficient preclinical data exist demonstrating a convincing pharmacokineticpharmaco- dynamic relationship.
Drug toxicity, however, remains an important part of phase I drug evaluation for all drugs. Drug toxicities can be determined and reported relatively easily because of the existence of standardized criteria. Furthermore, even if toxicity is not the primary endpoint of the dose escalation study, its description remains a necessary part of early testing of new agents. Although the use of toxicity for dose selection may not be appropriate for agents that have maximal target inhibition at nontoxic doses, this method of dose selection minimizes the possibility that a subtherapeutic dose will be chosen.
To understand how investigators carrying out phase I trials have responded to these challenges and what adaptations, if any, have been made to the traditional phase I trial design when evaluating novel targeted agents, we reviewed recently reported completed phase I studies of targeted non-cytotoxic therapies. Our review had three objectives: first, to describe study conduct, including choice of patient population, starting dose, and dose escalation schemes; second, to describe methods of dose selection, including which measures limited dose escalation and led to the selection of a recommended phase II dose; and third, to characterize the contribution of correlative studies to dose selection.
![]() |
MATERIALS AND METHODS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
|
|
![]() |
RESULTS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
We retrieved 60 completed single-agent phase I reports involving 31 agents representative of the most common targets of interest in the oncology literature (Tables 1 and 2) (770). Of the 31 agents, 20 were classified as small molecules (e.g., BAY 43-9006, ZD1839, and OSI-774), six as antibodies (e.g., trastuzumab), four as antisense oligodeoxynucleotides (e.g., ISIS 5132), and one as other (endostatin) (Table 1).
In general, the patient populations for the phase I studies were not selected on the basis of target expression. Only one trial (56) restricted patient entry to those expressing the target of interest (patients with breast cancer expressing HER2) and another five trials (11,20,46,61,65) enrolled patients likely to express the target on the basis of preclinical and clinical data (e.g., patients with nonsmall-cell lung cancer or head and neck cancer for treatment with EGFR inhibitors).
Starting Dose and Dose Escalation Method
Most studies did not state an explicit rationale for the choice of starting dose or a description of the dose escalation scheme (data not shown). However, most studies did state clearly the actual doses and routes of the administered drug and the number of patients in each cohort.
Determination of the Maximum Administered Dose
In 36 of 60 trials, toxicity was the primary basis for halting dose escalation (Table 3). Eight trials reported halting dose escalation on the basis of pharmacokinetic measurements (e.g., steady state plasma levels were achieved or the area under the curve [AUC] was greater than those determined to be active in preclinical studies). Less frequent reasons reported for determining the maximum administered dose included achievement of maximum planned dose level (five trials), limited drug supply (four trials), information obtained from other phase I studies that would preclude further escalation (two trials), and demonstration of drug activity (one trial). The reason for halting dose escalation was not stated in four trials.
|
A recommended phase II dose was reported in 52 trials involving 27 different agents (Table 4). The primary basis for the dose recommendation was toxicity in 35 of the 52 trials, consistent with toxicity being the most common reason stated for halting dose escalation. Pharmacokinetic data were the primary basis for the final dose recommendation in 11 of the 52 studies. Other less commonly cited primary reasons for the recommended phase II dose selection included toxicity information from another trial, demonstration of clinical activity, surrogate tissue findings (i.e., alterations in peripheral blood mononuclear cells), changes in measures of target effect in tumors, and convenience of dosing. Six studies did not provide a recommended phase II dose, and two studies did not recommend the drug or drug dosing schedule for further study.
|
|
|
|
Use of correlative laboratory studies to measure drug effects in tumor or surrogate tissue is appealing, especially when evaluating drugs with a specific target and relatively little toxicity. However, we found that only five trials incorporated correlative studies in tumor tissue (e.g., measures of EGFR downstream signaling) into phase I studies (Table 8). Furthermore, tumor correlative studies were the primary basis for dose selection in only one trial that evaluated an EGFR antibody given to patients with nonsmall-cell lung cancer or head and neck cancer (46).
|
Functional imaging studies were included in six trials (Table 8). Information derived from these studies was used to recommend a phase II dose in only one trial (49).
![]() |
DISCUSSION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Our most noteworthy finding was that toxicity was the primary basis for halting dose escalation and selecting the recommended phase II dose for the majority of targeted agents. In the context of our review, this finding suggests that, despite the promotion of alternative endpoints for phase I trials of these agents, toxicity remained the familiar gauge for determining the recommended dose. In a broader context, this finding underscores the importance of detailed characterization of side effects of targeted agents and highlights the concept that "non-cytotoxic" does not mean "free from toxicities." In addition, the hypersensitivity pneumonitis and cardiac toxicity associated with gefitinib (71) and trastuzumab (72), respectively, are useful reminders that vigilance is needed beyond the phase I setting because rare but potentially serious or cumulative toxicities may become apparent only with treatment of greater numbers of patients for longer periods than is usually the case in phase I trials.
After toxicity, pharmacokinetic measures were the next most common primary determinant for dose selection. Plasma drug levels also provided supportive evidence for the dose selected primarily by toxicity in several instances. The relatively frequent reliance on plasma levels as a guide for selecting a phase II dose presupposes that there is strong preclinical evidence demonstrating an association between drug levels and target inhibition or antitumor activity. We also found that measurements of target inhibition in tumor or surrogate tissue were infrequently incorporated into study design and, when undertaken, usually provided supporting rather than primary data for selecting a phase II dose. This finding was somewhat surprising, given the level of importance often assigned to proof-of-principle studies for targeted agents. Functional imaging studies were rarely used in the phase I trials we reviewed, perhaps because of the nature of the agents selected, and had limited usefulness for determining the recommended phase II dose.
The purpose of our study was to obtain a general sense of the current methods of dose selection for targeted agents, and our findings should be interpreted with an understanding of the potential weaknesses of our study. First, although we attempted to include a large number of published reports, this cannot be considered an exhaustive review of all phase I studies of targeted anticancer drugs. In addition, in those studies published in abstract form only, the reported data were limited in detail and may not have included all elements of interest. To determine whether the type of publication was a source of reporting bias, we examined all studies to see if there was a difference between published manuscripts and abstracts regarding inclusion and use of correlative studies as an endpoint. Twenty-four of the retrieved reports were in abstract form; 36 were full manuscripts. The inclusion of correlative studies (such as changes in tumor or surrogate tissue markers, functional imaging, or other measures such as blood VEGF levels) was similar in both types of publications (54% of the abstracts and 44% of the full papers). The use of correlative studies for selecting a recommended phase II dose was also similar. Thus, irrespective of the type of publication, the majority of correlative studies provided data that were supportive rather than primarily responsible for dose selection.
Second, we may have reviewed trials that had secondary publications of correlative studies that were not included in the primary report because multiple publications on the same trial sometimes occur.
Third, although the main reason for halting dose escalation was generally apparent from the study reports, the determinants of the recommend phase II dose required interpretation on our part. In general, we identified the primary determinant of dose selection as that which halted dose escalation if the next lower dose was chosen as the recommend phase II dose. In other situations, we relied on the presentation of other data if the values fell within desired ranges (on the basis of preclinical or clinical data) at the recommend phase II dose. Similarly we classified data, such as plasma pharmacokinetic levels, as "supplementary" if they were not presented as the primary determinant for selecting the recommended dose but if the authors cited them as providing additional evidence that the selected dose was likely to be in an active dose range.
In conclusion, to date, phase I studies of targeted, non-cytotoxic anticancer drugs generally use the traditional endpoints of toxicity and, to a lesser degree, pharmacokinetic information for selecting a recommended phase II dose. Measures of molecular drug effects in tumor or surrogate tissue were not routinely incorporated into study design and rarely formed the primary basis for dose selection in the studies we reviewed. Whether this approach has been wise or necessary cannot be concluded from our analysis. However, we note that several of the drugs we reviewed have produced objective responses in phase II trials at the doses recommended in phase I trials (7376), and others, such as bevacizumab and trastuzumab, have already been shown to improve survival or time to progression in randomized studies (7779). Yet it is clear that further work is needed to optimize dose selection and drug development strategies for targeted therapies in light of recent failures of high-profile agents, such as negative results from trials involving R115777 (a fanesyl transferase inhibitor), ISIS 3521 (an antisense agent to protein kinase C alpha), and ZD1839 ("Iressa," an EGFR kinase inhibitor) (8084).
Understanding the impact of the agent on the tumor target and identification of the patient population most likely to benefit is an important step in the drug development process if future failures are to be minimized. Because our review demonstrates that these questions remain largely unanswered in the phase I evaluation of these agents, the question thus becomes, What are the minimum criteria that must be satisfied in the phase I setting to continue drug development? Current practice indicates that these criteria include compelling preclinical evidence of proposed target inhibition, antitumor effect in animal models, and phase I evidence of drug tolerability in a dose (range) supported by pharmacokinetic data. This practice seems reasonable because questions regarding drug activity (on both the target and the tumor) and selection of optimal patient and/or disease subgroups for further testing may be more appropriately addressed in phase II studies of novel design using the dose (or dose range) established by toxicity and pharmacokinetic measures in phase I studies. As with conventional chemotherapy, phase III studies will ultimately define clinical utility because, regardless of the type of anticancer agent, the ultimate goal remains the sameprolongation of life and/or better relief of symptoms.
![]() |
REFERENCES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
1 Eisenhauer EA, ODwyer PJ, Christian M, Humphrey JS. Phase I clinical trial design in cancer drug development. J Clin Oncol 2000;18:68492.
2 Gelmon KA, Eisenhauer EA, Harris AL, Ratain MF, Workman P. Anticancer agents targeting signaling molecules and cancer cell environment: challenges for drug development? J Natl Cancer Inst 1999;91:12817.
3 Stadler WM, Ratain MJ. Development of target-based antineoplastic agents. Invest New Drugs 2000;18:716.[CrossRef][ISI][Medline]
4 Ratain MJ, Stadler WM. Clinical trial designs for cytostatic agents. J Clin Oncol 2001;19:31545.
5 Korn EL, Arbuck SG, Pluda JM, Simon R, Kaplan RS, Christian MC. Clinical trial designs for cytostatic agents: are new approaches needed? J Clin Oncol 2001;19:26572.
6 Parulekar WR, Eisenhauer EA. Novel endpoints and design of early clinical trials. Ann Oncol 2002;13 Suppl 4:13943.
7 Adjei AA, Erlichman C, Davis JN, Cutler DL, Sloan JA, Marks RS, et al. A phase I trial of the farnesyl transferase inhibitor SCH66336: evidence for biological and clinical activity. Cancer Res 2000;60:18717.
8 Awada A, Eskens FA, Piccart M, Cutler DL, van der Gaast A, Bleiberg H, et al.; EORTC Early Clinical Studies Group. Phase I and pharmacological study of the oral farnesyltransferase inhibitor SCH 66336 given once daily to patients with advanced solid tumours. Eur J Cancer 2002;38:22728.[CrossRef][ISI][Medline]
9 Awada A, Hendlisz A, Gil T, Munoz R, Bartholomeus S, de Valeriola D, et al. Final results of a clinical and pharmacokinetic (PK) phase I study of the Raf kinase inhibitor BAY 43-9006 in refractory solid cancers: a promising anti-tumor agent. Eur J Cancer 2002;38:S52.
10 Baselga J, Pfister D, Cooper MR, Cohen R, Burtness B, Bos M, et al. Phase I studies of anti-epidermal growth factor receptor chimeric antibody C225 alone and in combination with cisplatin. J Clin Oncol 2000;18:90414.
11 Baselga J, Rischin D, Ranson M, Calvert H, Raymond E, Kieback DG, et al. Phase I safety, pharmacokinetic, and pharmacodynamic trial of ZD1839, a selective oral epidermal growth factor receptor tyrosine kinase inhibitor, in patients with five selected solid tumor types. J Clin Oncol 2002;20:4292302.
12 Brahmer JR, Kelsey S, Scigalla P, Hill G, Bello C, Elza-Brown K, et al. A phase I study of SU6668 in patients with refractory solid tumors [abstract]. Proc ASCO 2002;21:84a.
13 Britten CD, Rosen LS, Kabbinavar F, Rosen P, Mulay M, Hernandez, et al. Phase I trial of SU6668, a small molecule receptor tyrosine kinase inhibitor, given twice daily in patients with advanced cancers [abstract]. Proc ASCO 2002;21:28b.
14 Britten CD, Rowinsky EK, Soignet S, Patnaik A, Yao SL, Deutsch P, et al. A phase I and pharmacological study of the farnesyl protein transferase inhibitor L-778,123 in patients with solid malignancies. Clin Cancer Res 2001;7:3894903.
15 Camacho LH, Soignet SL, Pezzulli S, Canales C, Aghajanian C, Spriggs DS, et al. Dose escalation study of oral farnesyl transferase inhibitor (FTI) BMS-214662 in patients with solid tumors [abstract]. Proc ASCO 2001;20:79a.
16 Cropp G, Rosen Le, Mulay M, Langecker P, Hannah A. Pharmacokinetics and pharmacodynamics of SU5416 in a phase I, dose escalating trial in patients with advanced malignancies [abstract]. Proc ASCO 1999;18:161a.
17 Crul M, de Klerk GJ, Swart M, vant Veer LJ, de Jong D, Boerrigter L, et al. Phase I clinical and pharmacologic study of chronic oral administration of the farnesyl protein transferase inhibitor R115777 in advanced cancer. J Clin Oncol 2002;20:272635.
18 Cunningham CC, Holmlund JT, Schiller JH, Geary RS, Kwoh TJ, Dorr A, et al. A phase I trial of c-raf kinase antisense oligonucleotide ISIS 5132 administered as a continuous intravenous infusion in patients with advanced cancer. Clin Cancer Res 2000;6:162631.
19 Davis AJ, Gelmon KA, Siu LL, Moore MF, Britten CD, Mistry N, et al. Phase I and pharmacologic study of the human DNA methyltransferase antisense oligodeoxynucleotide MG98 given as a 21-day continuous infusion every 4 weeks. Invest New Drugs 2003;21:8597.[CrossRef][ISI][Medline]
20 Divgi CR, Welt S, Kris M, Real FX, Yeh SD, Gralla R, et al. Phase I and imaging trial of indium 111-labeled anti-epidermal growth factor receptor monoclonal antibody 225 in patients with squamous cell lung carcinoma. J Natl Cancer Inst 1991;83:97104.[Abstract]
21 Eder JP Jr, Supko JG, Clark JW, Puchalski TA, Garcia-Carbonero R, Ryan DP, et al. Phase I clinical trial of recombinant human endostatin administered as a short intravenous infusion repeated daily. J Clin Oncol 2002;20:377284.
22 Erlichman C, Hidalgo M, Forero L, Boni J, Quinn S, Zacharchuk C, et al. EKB-569, an irreversible inhibitor of the epidermal growth factor receptor: phase I trial results in patients with advanced solid tumors. Eur J Cancer 2002;38:64.[ISI][Medline]
23 Eskens FA, Awada A, Cutler DL, de Jonge MJ, Luyten GP, Faber MN, et al.; European Organization for Research and Treatment of Cancer Early Clinical Studies Group. 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;19:116775.
24 Forouzesh B, Buckner J, Adjei A, Marks R, Hammond L, Molpus K, et al. Phase I, bioavailability and pharmacokinetic study of oral dosage of CCI-779 administered to patients with advanced solid malignancies. Eur J Cancer 2002;38:S54.[Medline]
25 Gordon MS, Margolin K, Talpaz M, Sledge GW, Holmgren E Jr, Benjamin R, et al. Phase I safety and pharmacokinetic study of recombinant human anti-vascular endothelial growth factor in patients with advanced cancer. J Clin Oncol 2001;19:84350.
26 Grochow L, OReilly S, Humphrey R, Sundaresan P, Donehower R, Sartorius S, et al. Phase I and pharmacokinetic study of the matrix metallo-proteinase inhibitor (MMPI), BAY12-9566 [abstract]. Proc ASCO 1998;17:213a.
27 Gupta E, Huang M, Mao Y, Patel R, Tarby C, Daniels R, et al. Pharmacokinetic evaluation of BMS-275291, a matrix metalloproteinase inhibitor in cancer patients [abstract]. Proc ASCO 2001;20:76a.
28 Heath EI, OReilly S, Humphrey R, Sundaresan P, Donehower RC, Sartorius S, et al. Phase I trial of the matrix metalloproteinase inhibitor BAY 12-9566 in patients with advanced solid tumours. Cancer Chemother Pharmacol 2001;48:26974.[CrossRef][ISI][Medline]
29 Herbst RS, Hess KR, Tran HT, Tseng JE, Mullani NA, Charnsangavej C, et al. Phase I study of recombinant human endostatin in patients with advanced solid tumors. J Clin Oncol 2002;20:3792803.
30 Herbst RS, Maddox AM, Rothenberg ML, Small EJ, Rubin EH, Baselga J, et al. Selective oral epidermal growth factor receptor tyrosine kinase inhibitor ZD1839 is generally well-tolerated and has activity in non-small-cell lung cancer and other solid tumors: results of a phase I trial. J Clin Oncol 2002;20:381525.
31 Hidalgo M, Siu LL, Nemunaitis J, Rizzo J, Hammond LA, Takimoto C, et al. Phase I and pharmacologic study of OSI-774, an epidermal growth factor receptor tyrosine kinase inhibitor, in patients with advanced solid malignancies. J Clin Oncol 2001;19:326779.
32 Hilger RA, Kredtke S, Hedley D, Moeller JG, Bauer RJ, Stellberg W, et al. Inhibition of ERK phosphorylation and clinical outcome in patients treated with the raf kinase inhibitor BAY 43-9006 [abstract]. Proc ASCO 2002;21:27b.
33 Hirte H, Goel R, Major P, Seymour L, Huan S, Stewart D, et al. A phase I dose escalation study of the matrix metalloproteinase inhibitor BAY 12-9566 administered orally in patients with advanced solid tumours. Ann Oncol 2000;11:157984.[Abstract]
34 Hirte H, Miller W, Major P, Goss G, Stewart D, Batist G, et al. Initial results of part 2 of a phase I/II pharmacokinetics, pharmacodynamic and biological activity study of ZD1839: NCIC CTG IND 122. Clin Cancer Res 2001;7:3761s.
35 Hirte H, Moore M, Hotte S, Oza A, Siu L, Harris H, et al. Final results of a phase I study of the raf-1 kinase inhibitor BAY 43-9006 in patients with advanced refractory solid tumours. Eur J Cancer 2002;38:55.
36 Hurwitz H, Humphrey J, Williams K, Ness E, Conway A, Lockhart AC, et al. A phase I trial of BMS-275291: a novel, non-hydroxamate, Sheddase-Sparing matrix metalloproteinase inhibitor (MMPI) with no dose limiting arthritis [abstract]. Proc ASCO 2001;20:98a.
37 Hurwitz H, Holden SN, Eckhardt SG, Rosenthal M, de Boer R, Rischin D, et al. Clinical evaluation of ZD6474, an orally active inhibitor of VEGF signalling, in patients with solid tumors [abstract]. Proc ASCO 2002;21:82a.
38 Kuenen B, Ruijter R, Hoekman K, Scigalla P, Giaccone G, Pinedo H. Dose finding study of SU6668 given thrice daily by oral route under fed conditions in patients with advanced malignancies [abstract]. Proc ASCO 2002;21:110a.
39 LoRusso PM, Adjei AA, Meyer MB, Wozniak A, Gadgeel SM, Hanson LJ, et al. A phase I clinical and pharmacokinetic evaluation of the oral MEK inhibitor, CI-1040, administered for 21 consecutive days, repeated every 4 weeks in patients with advanced cancer [abstract]. Proc ASCO 2002;21:81a.
40 Mitchell DY, Reid JM, Parchment RE, Meyer MB, Leopold JS, Herrera R, et al. Pharmacokinetics and pharmacodynamics of the oral MEK inhibitor, CI-1040, following multiple dose administration to patients with advanced cancer [abstract]. Proc ASCO 2002;21:81a.
41 Morimoto A, West KA, Yuen HA, Mathews WR, Bello C, Cropp GF, et al. Biomarker analysis of patient samples from a phase I clinical trial using the angiogenesis inhibitor, SU6668, for treatment of advanced solid tumors [abstract]. Proc ASCO 2002;21:85a.
42 Morris MJ, Tong WP, Cordon-Cardo C, Drobnjak M, Kelly WK, Slovin SF, et al. Phase I trial of BCL-2 antisense oligonucleotide (G3139) administered by continuous intravenous infusion in patients with advanced cancer. Clin Cancer Res 2002;8:67983.
43 Negoro S, Nakagawa K, Fukuoka M, Kudoh S, Tamura T, Yoshimura N, et al. Final results of a phase I intermittent dose-escalation trial of ZD1839 (Iressa) in Japanese patients with various solid tumours [abstract]. Proc ASCO 2001;20:324a.
44 Nemunaitis J, Holmlund JT, Kraynak M, Richards D, Bruce J, Ognoskie N, et al. Phase I evaluation of ISIS 3521, an antisense oligodeoxynucleotide to protein kinase c-alpha, in patients with advanced cancer. J Clin Oncol 1999;17:358695.
45 ODonnell A, Trigo J, Banerji U, Raynaud F, Padhani A, Hannah A, et al. A phase I trial of the VEGF inhibitor SU5416, incorporating dynamic contrast MRI assessment of vascular permeability [abstract]. Proc ASCO 2000;19:177a.
46 Perez-Soler R, Donato NJ, Shin DM, Rosenblum MG, Zhang HZ, Tornos C, et al. Tumor epidermal growth factor receptor studies in patients with non-small-cell lung cancer or head and neck cancer treated with monoclonal antibody RG 83852 [Erratum in: J Clin Oncol 1994;12:1526.]. J Clin Oncol 1994;12:7309.[Abstract]
47 Punt CJ, van Maanen L, Bol CJ, Seifert WF, Wagener DJ. Phase I and pharmacokinetic study of the orally administered farnesyl transferase inhibitor R115777 in patients with advanced solid tumors. Anti-Cancer Drugs 2001;12:1937.[CrossRef][ISI][Medline]
48 Ranson M, Hammond LA, Ferry D, Kris M, Tullo A, Murray PI, et al. ZD1839, a selective oral epidermal growth factor receptor-tyrosine kinase inhibitor, is well tolerated and active in patients with solid, malignant tumors: results of a phase I trial. J Clin Oncol 2002;20:224050.
49 Raymond E, Faivre S, Vera K, Delbaldo C, Robert C, Spatz A, et al. for the SU11248 Working Group; Institute Gustave Roussy, Ville-juif, France; Pharmacia Co. and SUGEN Inc., Ville-juif, France. Final results of a phase I and pharmacokinetic study of SU11248, a novel multi-target tyrosine kinase inhibitor, in patients with advanced cancers [abstract]. Proc ASCO 2003;22:192.
50 Rosemurgy A, Harris J, Langleben A, Casper E, Goode S, Rasmussen H. Marimastat in patients with advanced pancreatic cancer: a dose finding study. Am J Clin Oncol 1999;22:24752.[CrossRef][ISI][Medline]
51 Rosen L, Mulay M, Mayers A, Kabbinavar F, Rosen P, Cropp G, et al. Phase I dose-escalating trial of SU5416, a novel angiogenesis inhibitor in patients with advanced malignancies [abstract]. Proc ASCO 1999;18:161a.
52 Rowinsky E, Humphrey R, Hammond LA, Aylesworth C, Smetzer L, Hidalgo M, et al. Phase I and pharmacologic study of the specific matrix metalloproteinase inhibitor BAY 12-9566 on a protracted oral daily dosing schedule in patients with solid malignancies. J Clin Oncol 2000;18:17886.
53 Rubin R, Abbruzzese J, Morrison B, Mazina K, Lee Y, Zamek R, et al. Phase I trial of the farnesyl protein transferase (FPTase) inhibitor L-778123 on a 14- or 28-day dosing schedule [abstract]. Proc ASCO 2000;19:178a.
54 Rudek MA, Figg WD, Dyer V, Dahut W, Turner ML, Steinberg SM, et al. Phase I clinical trial of oral COL-3, a matrix metalloproteinase inhibitor, in patients with refractory metastatic cancer. J Clin Oncol 2001;19:58492.
55 Rudin CM, Holmlund J, Fleming GF, Mani S, Stadler WM, Schumm P, et al. Phase I trial of ISIS 5132, an antisense oligonucleotide inhibitor of c-raf-1, administered by 24-hour weekly infusion to patients with advanced cancer. Clin Cancer Res 2001;7:121420.
56 Shak S. Overview of the trastuzumab (Herceptin) anti-HER2 monoclonal antibody clinical program in HER2-overexpressing metastatic breast cancer. Semin Oncol 1999;26:717.[ISI][Medline]
57 Stevenson JP, Yao KS, Gallagher M, Friedland D, Mitchell EP, Cassella A, et al. Phase I clinical/pharmacokinetic and pharmacodynamic trial of the c-raf-1 antisense oligonucleotide ISIS 5132 (CGP 69846A). J Clin Oncol 1999;17:222736.
58 Stewart DJ, Donehower RC, Eisenhauer EA, Wainman N, Shah AK, Bonfils C, et al. A phase I pharmacokinetic and pharmacodynamic study of the DNA methyltransferase 1 inhibitor MG98 administered twice weekly. Ann Oncol 2003;14:76674.
59 Stopeck A, Sheldon M, Vahedian M, Cropp G, Gosalia R, Hannah A. Results of a phase I dose-escalating study of the antiangiogenic agent, SU5416, in patients with advanced malignancies. Clin Cancer Res 2002;8:2798805.
60 Strumberg D, Voliotis D, Moeller JG, Hilger RA, Richly H, Beling C, et al. A clinical phase I and biomarker study of the raf kinase inhibitor BAY 43-9006: preliminary evidence of activity in patients with advanced solid tumors. Eur J Cancer 2002;38:S53.
61 Tabernero J, Rojo F, Jimenez E, Montaner I, Snatome L, Guix M, et al. A phase I pharmacokinetic and serial tumor and skin pharmacodynamic study of weekly, every 2 weeks or every 3 weeks 1-hour infusion EMD72000, a humanized monoclonal anti-epidermal growth factor receptor antibody, in patients with advanced tumours known to overexpress the EGFR. Eur J Cancer 2002;38:S69.[Medline]
62 Tabernero J, Sonnichsen D, Albanell J, Damle B, Rojo F, Christopher L, et al. A phase I pharmacokinetic (PK) and serial tumor and PBMC pharmacodynamic (PD) study of weekly BMS-214662, a farnesyltransferase (FT) inhibitor, in patients with advanced solid tumors [abstract]. Proc ASCO 2001;20:77a.
63 Thomas A, Morgan B, Decatris A, Henry A, Pfister C, Gano C, et al. A phase I study of the oral vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitor PTK787/ZK 222584 on a twice daily schedule in patients with advanced cancer. Eur J Cancer 2002;38:77.
64 Thomas JP, Arzoomanian RZ, Alberti D, Marnocha R, Lee F, Friedl A, et al. Phase I pharmacokinetic and pharmacodynamic study of recombinant human endostatin in patients with advanced solid tumors. J Clin Oncol 2003;21:22331.
65 van Oosterom AT, Judson IR, Verweij J, Stroobants S, Dumez H, Donato di Paola E, et al. Update of phase I study of imatinib (STI571) in advanced soft tissue sarcomas and gastrointestinal stromal tumors: a report of the EORTC Soft Tissue and Bone Sarcoma Group. Eur J Cancer 2002;38 Suppl 5:S837.[Medline]
66 Voi M, Tabernero T, Cooper MR, Marimon I, Van Vreckem A, Albanell J, et al. A phase I study of the farnesyltransferase (FT) inhibitor BMS-214662 administered as a weekly 1-hour infusion in patients (pts) with advanced solid tumors: clinical findings [abstract]. Proc ASCO 2001;20:79a.
67 Waters JS, Webb A, Cunningham D, Clarke PA, Raynaud F, di Stefano F, et al. Phase I clinical and pharmacokinetic study of bcl-2 antisense oligonucleotide therapy in patients with non-Hodgkins lymphoma. J Clin Oncol 2000;18:181223.
68 Wojtowicz-Praga S, Torri J, Johnson M, Steen V, Marshall J, Ness E, et al. Phase I trial of Marimastat, a novel matrix metalloproteinase inhibitor, administered orally to patients with advanced lung cancer. J Clin Oncol 1998;16:21506.[Abstract]
69 Yuen AR, Halsey J, Fisher GA, Holmlund JT, Geary RS, Kwoh TJ, et al. Phase I study of an antisense oligonucleotide to protein kinase C-alpha (ISIS 3521/CGP 64128A) in patients with cancer. Clin Cancer Res 1999;5:335763.
70 Zujewski J, Horak ID, Bol CJ, Woestenborghs R, Bowden C, End DW, et al. Phase I and pharmacokinetic study of farnesyl protein transferase inhibitor R115777 in advanced cancer. J Clin Oncol 2000;18:92741.
71 Cohen MH, Williams GA, Sridhara R, Chen G, Pazdur R. FDA drug approval summary: gefitinib (ZD1839) (Iressa) tablets. Oncologist 2003;8:3036.
72 Perez EA, Rodeheffer R. Clinical cardiac tolerability of trastuzumab. J Clin Oncol 2004;22:3229.
73 Kris MG, Natale RB, Herbst RS, Lynch TJ, Prager D, Belani CP, et al. A phase II trial of ZD 1839 (Iressa) in advanced non-small cell lung cancer patients who had failed platinum- and docetaxel-based regimens (IDEAL2) [abstract]. Proc ASCO 2002;21:292a.
74 Fukuoka M, Yano S, Giaccone G, Tamura T, Nakagawa K, Douillard JY, et al. Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer. J Clin Oncol 2003;21:223746.
75 Saltz L, Meropol NJ, Loehrer PJ, Waksal H, Needle MN, Mayer RJ. Single agent IMC-225 (Erbitux) has activity in CPT-11-refractory colorectal cancer that expresses the epidermal growth factor receptor [abstract]. Proc ASCO 2002;21:127a.
76 Demetri GD, von Mehren M, Blanke CD, Van den Abbeele AD, Eisenberg B, Roberts PJ, et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med 2002;347:47280.
77 Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ, Topalian SL, et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med 2003;349:42734.
78 Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001;344:78392.
79 Hurwitz H, Fehrenbacher L, Cartwright T, Hainsworth J, Heim W, Berlin J, et al. Bevacizumab (a monoclonal antibody to vascular endothelial growth factor) prolongs survival in first-line colorectal cancer (CRC): results of a phase III trial of bevacizumab in combination with bolus IFL (irinotecan, 5-fluorouracil, leucovorin) as first-line therapy in subjects with metastatic CRC. 2003. Available at: http://www.asco.org/hurwitz_no3646.
80 Miller KD, Rugo HS, Cobleigh MA, Marcom PK, Chap LI, Holmes FA, et al. Phase III trial of capecitabine (Xeloda) plus bevacizumab (Avastin) verus capecitabine alone in women with metastatic breast cancer previously treated with an anthracycline and a taxane. Breast Cancer Res Treat 2002;76:36.
81 Van Cutsem E, Karasek P, Oettle H, Vervenne A, Szawlowski P, Schoffski P, et al. Phase III trial comparing gemcitabine plus R115777 (Zamestra versus gemcitabine plus placebo in advanced pancreatic cancer [abstract]. Proc ASCO 2002;21:130a.
82 Johnson DH, Herbst R, Giaccone G, Schiller J, Natale RB, Miller V, et al. ZD1839 (Iressa) in combination with paclitaxel and carboplatin in chemotherapy-naive patients with advanced non-small cell lung cancer (NSCLC): results from a phase III clinical trials (INTACT 2). Ann Oncol 2002;13(Suppl 5):127.
83 Giaconne G, Johnson DH, Manegold C, Scagliotti GV, Rosell R, Wolf M, et al. A phase III clinical trial of ZD1839 (Iressa) in combination with gemcitabine and cisplatin in chemotherapy-naive patients with advanced non-small cell lung cancer (INTACT 1). Ann Oncol 2002;13(Suppl 5):2.
84 Lynch TJ, Raju R, Lind M, Riviere A, Gatzemeier U, Drorr A, et al. Randomized phase III trial of chemotherapy and antisense oligonucleotide LY900003 (ISIS 3521) in patients with advanced NSCLC: initial report [abstract]. Proc ASCO 2003;22:623.
Manuscript received November 3, 2003; revised April 19, 2004; accepted May 12, 2004.
This article has been cited by other articles in HighWire Press-hosted journals:
Editorial about this Article
![]() |
||||
|
Oxford University Press Privacy Policy and Legal Statement |