06-1188 6094..6099 Hoosier Oncology Group Randomized Phase II Study of Docetaxel, Vinorelbine, and Estramustine in Combination in Hormone-Refractory Prostate Cancer with Pharmacogenetic Survival Analysis NoahM.Hahn,1SharonMarsh,3 William Fisher,4 RobertLangdon,5 Robin Zon,6 MarkBrowning,7 CynthiaS.Johnson,2 Tiffany J.Scott-Horton,3 Lang Li,2 Howard L.McLeod,3 andChristopher J.Sweeney1 Abstract Purpose:To determine the safety and efficacy of two docetaxel doublets inhormone-refractory prostate cancer (HRPC) patients and to examine the prognostic role of polymorphisms in host genes important to docetaxelmetabolismand transport. Experimental Design: Sixty-four chemotherapy-naive patients with HRPC were randomized to docetaxel and vinorelbine (D, 20 mg/m2 i.v. days1and 8; V, 25 mg/m2 i.v. days1and 8) or docetaxelandestramustinephosphate(D,60-70mg/m2i.v.day1;E,280mgoralthricedailydays 1-5) administered q21d. Primary end point was clinically significant toxicity. A pharmacogenetic analysis ofhostgenes was doneinpatients who receivedat leastone cycle ofdocetaxel therapy. Results: Grade 3/4 toxicity occurred in15.6% of DV patients andin 28.6% DE patients. Neither armexceededthethresholdofclinicallysignificant toxicity. IntheDVarm,objective responserate was 33%, prostate-specific antigen response rate was 20%, and median survival was 16.2 months. Inthe DE arm, objective response ratewas67%,prostate-specific antigenresponserate was43%,andmediansurvivalwas19.7months.Pharmacogeneticanalyses showedasignificant associationbetweensurvivalbeyond15months andtheABCG2421C>A(Q141K)polymorphism compared with the wild-type (C/C) genotype (66% versus 27%; P = 0.05). Conclusions: DV and DE doublets are active with a tolerable toxicity profile in patients with HRPC;however,efficacydoesnotseemsuperior tostandardsingle-agentdocetaxel.TheABCG2 421C>A(Q141K)polymorphismmaybeanimportantpredictorof responseandsurvival inHRPC patients treated with docetaxel-based chemotherapy. Prostate cancer afflicts 234,000 men yearly and results in >27,000 deaths (1). Recent trials have shown a survival benefit for men with hormone-refractory prostate cancer (HRPC) treated with a docetaxel-based regimen (2, 3). Vinorelbine is a Vinca alkaloid that inhibits the microtubular apparatus in malignant cells and has documented activity in HRPC(4–6).Vinorelbineanddocetaxelinhibitdifferentportions ofthemicrotubuleapparatusinasynergisticmannerin preclinical in vitro prostate cancer models (7–9). With this in mind, the Hoosier Oncology Group conducted a randomized phase II trial of weekly docetaxel and vinorelbine (DV) with docetaxel and estramustine phosphate (DE) as a parallel reference arm. This study also included a pharmacogenetic analysis of host genes with critical roles in docetaxel drug transport and metabolism. Clinical studies suggest that variant alleles in the thiopurine methyltransferase gene can identify patients at risk of severe hematologic toxicity after azathioprine, 6-mercaptopurine, or related agents (10). Similar toxicity asso- ciations are seen with dihydropyrimidine dehydrogenase gene mutations and 5-fluorouracil therapy (11) and with UGT1A1 mutations and irinotecan (12). Single-nucleotide polymor- phisms (and other genetic variants) have also been observed, which affect docetaxel metabolism and transport. These include alterations in the degradation pathway (cytochrome P450 3A4 and 3A5; refs. 13–15), microtubule associated pro- teins (MAP4 and MAPT; ref. 16), and drug transport proteins [multidrug resistance 1 (ABCB1) and breast cancer resistance protein (ABCG2); refs. 15, 17]. Polymorphisms in these genes Cancer Therapy: Clinical Authors’Affiliations: 1Department of Medicine and 2Division of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana; 3Department of Medicine,Washington University Schoolof Medicine, St. Louis, Missouri; 4Medical Consultants, P.C., Muncie, Indiana; 5Nebraska Methodist Hospital and Cancer Center, Omaha, Nebraska; 6Northern Indiana Cancer Research Consortium, South Bend, Indiana; and 7Oncology Hematology Associates of Southwest Indiana, Evansville, Indiana Received 5/16/06; accepted 8/3/06. Grant support: GlaxoSmithKline (C.J. Sweeney), Sanofi Aventis (C.J. Sweeney), Walther Cancer Institute (C.J. Sweeney), and NIH Pharmacogenetics Research Networkgrant GM63340 (H.L. McLeod). The costs ofpublicationof this articlewere defrayedinpartby thepaymentofpage charges.This article must therefore be hereby marked advertisement in accordance with18 U.S.C. Section1734 solely to indicate this fact. Note: Presented in abstract form at the Proceedings of theAmerican Society of ClinicalOncologyAnnualMeeting,May13-17,2005,Orlando,Florida,abstract4568. Requests for reprints: Christopher J. Sweeney, Division of Hematology and Oncology, Indiana University Cancer Center, 535 Barnhill Drive, Indiana University Cancer Pavilion, Room 473, Indianapolis, IN 46202. Phone: 317-274-3515; Fax: 317-274-3636; E-mail: chsweene@iupui.edu. F2006 American AssociationforCancerResearch. doi:10.1158/1078-0432.CCR-06-1188 www.aacrjournals.orgClin Cancer Res 2006;12(20) October15, 2006 6094 Research. on April 5, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from http://clincancerres.aacrjournals.org/ were assessed to identify genetic variants that predict for toxicity and efficacy of docetaxel-based chemotherapy. Materials and Methods Eligibility criteria. The protocol was approved by the institutional review boards of all participating centers. Key eligibility criteria are listed in Fig. 1. In addition, all patients had progressive disease after at least one hormonal therapy (orchiectomy, estrogens, luteinizing hormone–releasing hormone therapy, etc.) with castrate levels of testosterone (<30 ng/dL). Progressive disease was defined as one of the following: an increase in prostate-specific antigen (PSA) >50% over nadir on hormonal therapy measured on two successive occasions at least 2 weeks apart (18) and/or objective evidence of progressive disease on computed tomography scan and/or new symptomatic bone metastases. Patients treated with an antiandrogen must have also progressed after the antiandrogen had been discontinued for at least 4 weeks (or for 6 weeks for antiandrogens with a longer half-life, such as bicalutamide) and have continued evidence of disease progression (at least a 25% increase in PSA after discontinuing). There was no maximum number of hormonal therapies allowed. Patients were excluded if they had new or unstable central nervous system metastases, had received external beam radiation therapy to >25% of their bone marrow, or had a history of grade z2 peripheral neuropathy. Patients with active cardiac disease defined as active angina, symptomatic congestive heart failure, or myocardial infarction within the previous 6 months were excluded. Patients with a history of deep venous thrombosis, pulmonary embolism, or cerebrovascular attack within the last 6 months were also excluded. Patients who had received external beam radiation or samarium injection <4 weeks before enrollment and patients who had received a strontium injection <6 weeks before enrollment were ineligible. Patients with a history of prior malignancy <5 years disease-free with the exception of curatively treated basal and squamous skin cancers were excluded. Patients were excluded if they had received PC-SPES within 4 weeks before enrollment. In addition, the use of saw palmetto or lycopenes was not allowed. Treatment. Patients were randomized to one of two regimens. Patients on the DE arm received 280 mg of oral estramustine phosphate thrice daily 1 to 2 hours after a meal from days 1 to 5 with dairy products restricted on those days. Patients received i.v. infusion of docetaxel over 60 minutes at a dose of 60 mg/m2 on day 2. If no grade 3 or 4 toxicities occurred in the first cycle, docetaxel was increased to 70 mg/m2 for all subsequent cycles. Patients received 8 mg dexamethasone orally every 12 hours for five doses beginning the night before chemotherapy. From day 1 until 2 weeks after discontinuation of estramustine phosphate, patients received 325 mg of enteric-coated aspirin as prophylaxis for arterial thrombosis and 2 mg of oral warfarin. Patients randomized to the DV arm received i.v. vinorelbine over 6 to 10 minutes at a dose of 20 mg/m2 on days 1 and 8. Patients also received i.v. docetaxel over 30 minutes at a dose of 25 mg/m2 on days 1 and 8. All patients received 4 mg dexamethasone orally every 12 hours for 3 days beginning the day before treatment. In patients with a prior history of external beam radiation, vinorelbine and docetaxel were reduced to 15 and 20 mg/m2, respectively. In both DE and DV arms, treatments were repeated on a 21-day cycle up to a total of six cycles or until disease progression or dose-limiting toxicity. Patient evaluation. All patients had a history, physical exam, and PSA within 1 week before treatment. Other laboratories (complete blood count, aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, total bilirubin, testosterone, PSA, and coagula- tion studies) were done within 2 weeks before treatment. All radiologic studies (bone scan, abdomen and pelvis computed tomography scan, and chest X-ray) were done within 4 weeks before treatment. Before each cycle, a case report form, complete blood count, hepatic panel, PSA, and coagulations laboratories were recorded. Radiographic studies were repeated before each odd-numbered cycle and in follow-up for patients with documented objective responses to therapy. Disease assessment. Patients with measurable disease were assessed for response to therapy according to standard Response Evaluation Criteria in Solid Tumors criteria. Patients with an elevated PSA were assessed for a serologic PSA response. PSA complete response was defined as an undetectable PSA if prior prostatectomy and <4 ng/mL if prostate in place (after irradiation, no local therapy, etc.) on two consecutive measurements 3 weeks apart. PSA partial response was defined as a decline of PSA value by >50% for two consecutive determinations at least 3 weeks apart. PSA stabilization was defined as a <50% increase or decrease in PSA over a period of 3 months if baseline PSA >20 ng/mL. PSA progression was defined as an increase in PSA to >50% above baseline on two consecutive measures at least 3 weeks apart. PSA relapse was defined as a >50% increase in PSA over the minimum value recorded during a response as evidenced on three successive determinations. Pharmacogenetic evaluation. All patients who received at least 1 day of docetaxel therapy on either trial arm were eligible for inclusion in the companion pharmacogenetic study. Participation in the pharmacoge- netic study was not required for participation in the treatment portion of the trial. After a separate informed consent was obtained, a single 5-mL blood sample collected in an EDTA tube was drawn from each patient. Samples were stored at 2jC to 8jC for V5 days and shipped to Washington University (St. Louis, MO) for analysis. DNA was extracted from whole blood using the Gentra PureGene kit (Gentra Systems, Inc., Minneapolis, MN) following the manufacturer’s instruc- tions. Eight polymorphisms in six genes associated with taxane metabolism (CYP3A4*1B and CYP3A5*3C), transport (ABCB1 3435 C>T and ABCG2 421 C>A), and microtubule assembly (MAP4 68 G>A, MAP4 1280 C>A, MAPT-13 G>A, and MAPT 3674 A>G) were assessed using PCR and Pyrosequencing technology as described previously (19–22). Statistical analysis. The primary end point of the study was the incidence of clinically significant toxicity defined as any significant adverse event, which required reporting or need for RBC or platelet transfusion that was thought to be related to treatment. Events that required reporting were serious adverse events or reactions that were at least possibly related to therapy and resulted in death, or were life threatening, required inpatient hospitalization or prolongation of existing hospitalization, or resulted in persistent or significant disability.Fig.1. Keyeligibility criteria. Docetaxel,Vinorelbine, and Estramustine Phase II Study www.aacrjournals.org Clin Cancer Res 2006;12(20) October15, 20066095 Research. on April 5, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from http://clincancerres.aacrjournals.org/ Secondary end points included objective, PSA, and clinical response rates, time to progression, and pharmacogenetic analyses. Patients were stratified based on baseline performance status, pain requiring opiates, and presence of measurable disease. There was no intention to statistically compare the two arms. Eighteen patients were treated on each arm in the first stage. A study arm was to be terminated if z5 patients, among the first 18, experienced clinically significant toxicity. This did not occur in either arm; therefore, 14 additional patients were assigned to each arm in stage II. If z6 patients, among the total 32 of each arm, experienced clinically significant toxicity, the arm was to be considered not worthy of further testing. These numbers were determined by the Simon two- stage phase II design and provided the study with 90% power to detect a true toxicity rate of 30% with a 5% type I error rate. Confidence intervals for clinically significant toxicity rates of each arm were obtained using Jennison and Turnbull (1983) method. Confidence intervals for response rates were obtained using an exact method. Curves for time to progression and overall survival were estimated using Kaplan-Meier method. Toxicity grades were summarized using contin- gency tables. Pharmacogenetic polymorphism frequencies were ana- lyzed in relation to clinical benefit, PSA response, and overall survival using m2 proportional analyses. For the survival analysis, a cutoff of 15 months was chosen to approximate the expected median survival in study subjects. Progression-free survival was defined as the time from enrollment on study until either measurable progressive disease was noted, 50% increase in PSA from nadir was observed and confirmed at least 4 weeks later, or clinical deterioration (decline in Karnofsky performance score of z20 points from baseline, an increase in opiate requirements of 10 mg i.m. equivalents, or death due to any cause) was noted. Overall survival was defined as the time from enrollment on study until death due to any cause. Results Patients. From March 21, 2002 to April 21, 2004, 64 patients were randomized, 32 to each arm. The median age was 72 years. Median pretreatment PSA level was 136. The median Karnofsky performance score was 90 with a median pain score of 2 and median i.m. morphine equivalent opiate use of 0 mg/d. A total of 38 (59.4%) patients had measurable disease at baseline. Baseline characteristics are summarized in Table 1 for the entire group and each arm individually. There were no significant differences in any of the baseline stratification factors. Toxicity. In general, treatment was well tolerated. Nine of 32 (28.1%) patients [95% confidence interval (95% CI), 6.4-39.8] experienced grade 3 or 4 toxicity in the DE arm. Grade 3 toxicities included diarrhea, febrile neutropenia, hypoglycemia/ hypokalemia, increased lacrimation, hypersensitivity reactions, typhlitis, urinary retention, and elevated transaminases (one patient each). Grade 4 toxicities included hematuria, hypergly- cemia/fatigue, stroke, and elevated transaminases (one patient each). Five of 32 (15.6%) patients (95% CI, 5.0-32.0) expe- rienced grade 3 or 4 toxicity in the DV arm. Grade 3 toxicities included bone pain, edema, hyperglycemia, venous catheter- associated infection, and recurrent urinary tract infections (one patient each). Grade 4 toxicity was limited to a single episode of hypocalcemia. Neither arm exceeded the prespeci- fied limit of six clinically significant toxic events as previously defined. Chemotherapy administration. A total of 164 cycles of DE chemotherapy was given to patients. Of these, 87.2% of cycles were given without dose reduction or delay. The most common reasons for dose reduction or delay were infectious (2.4%; cellulitis, fever, neutropenic fever, and herpes zoster; all one patient each), hematologic (2.4%; four patients with neutropenia), unknown (1.8%; three patients), and gastroin- testinal toxicity (1.2%; hepatotoxicity and nausea/emesis; one patient each). In the DV arm, 150 cycles of chemotherapy were adminis- tered, 76.0% without dose reduction or delay. The most common causes for reduction or delay were prior radiotherapy (10.7%; 16 patients, mandated at baseline per treatment plan), hematologic (8.7%; 11 patients with neutropenia, 2 patients with thrombocytopenia), and infectious (2.7%; 4 patients with febrile neutropenia). Response and survival. Among patients with measurable disease in the DE arm, six of nine (66.7%) patients had a partial or complete response. A partial PSA response was observed in 42.9% of patients. Median progression-free survival was 5.7 months (95% CI, 5.1-7.1), with an overall survival of 19.7 months (95% CI, 12.0; not reached). In the DV arm, 4 of 12 (33.3%) patients with measurable disease had a partial or complete response. A partial PSA response was observed in 20.0% of patients. Median progres- sion-free survival was 6.2 months (95% CI, 5.0-8.0), with an overall survival of 16.2 months (95% CI, 13.1-25.2). Kaplan- Meier curves for progression-free survival and overall survival are summarized for both DE and DV arms in Figs. 2 to 3. Pharmacogenetic analysis. Samples for pharmacogenetic analyses were available for 51 patients. In this pilot analysis, it was noted that 4 of 6 (66%) patients with the ABCG2 421 C>A (Q141K) polymorphism were alive past 15 months compared with only 12 of 44 (27%) patients with wild-type (C/C; P = 0.05). Polymorphisms in other candidate genes (CYP3A4, CYP3A5, ABCB1, MAPT, and MAP4) were not Table 1. Baseline patient demographics All patients Arm DV Arm DE n Median (range) n Median (range) n Median (range) Age 67 72 (41-85) 35 70 (41-85) 32 72 (55-82) PSA 64 136.2 (7.3-1,881.0) 33 146.9 (7.3-1,881.0) 31 126.6 (10.1-1,333.9) KPS 64 90 (70-100) 33 90 (70-100) 31 90 (70-100) Mean pain score 61 2.0 (0.0-9.0) 32 2.2 (0.0-9.0) 29 2.0 (0.0-5.7) Median opiate use* 61 0 (0-50) 32 0 (0-50) 29 0 (0-24) Abbreviation: KPS, Karnofsky performance score. *I.m. morphine equivalents. Cancer Therapy: Clinical www.aacrjournals.orgClin Cancer Res 2006;12(20) October15, 2006 6096 Research. on April 5, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from http://clincancerres.aacrjournals.org/ associated with an increased chance of being alive beyond 15 months (Table 2). Pharmacogenomic analyses of toxicity or PSA response revealed no additional significant associations (all P > 0.05). Discussion Metastatic HRPC remains the third leading cause of cancer- related mortality among men in the United States. In 2004, Petrylak et al. (2) and Tannock et al. (3) reported results of two separate randomized controlled trials confirming a survival advantage for docetaxel-based chemotherapy. However, the potential toxic effects of chemotherapy remain a valid consideration as we evaluate new combinations. In previous studies combining estramustine phosphate and docetaxel in the treatment of HRPC patients, objective response rates of 6% to 50% were observed, with most responses characterized as partial responses. PSA responses range from 45% to 82%. Major toxicities include myelosuppression, fatigue, and nausea. Grade 3 or 4 neutropenia is seen in up to half of patients. With therapy, overall survival ranges between 12 and 20 months with variations depending on patient characteristics at baseline (23–26). In the present study, 9 of 32 (28.1%) patients (95% CI, 6.4-39.8) treated with DE experienced grade 3 or 4 toxicity. An objective response rate of 66.7%, a PSA response rate of 42.9%, and a median overall survival of 19.7 months were observed. These results compare favorably with the pivotal phase III randomized trials described above in terms of efficacy of every 3-week docetaxel and toxicity of the docetaxel and estramustine combination. Vinorelbine is a semisynthetic Vinca alkaloid that has shown activity against solid organ malignancies, including lung, breast, and germ cell tumors. Its preclinical synergism when combined with docetaxel in prostate cancer cell line experi- ments was a basis for its inclusion in this trial (27). Furthermore, as a single agent, it is tolerable in elderly patients with metastatic HRPC (28–31). In the present study, 5 of 32 (15.6%) patients (95% CI, 5.0-32.0) treated with DV experienced grade 3 or 4 toxicity and 1 had clinically significant toxicity per the protocol. An objective response rate of 33.3%, a PSA response rate of 20.0%, and a median overall survival of 16.2 months were observed. Koletsky et al. (32) have reported the only other phase II data combining vinorelbine and docetaxel to date. In the Koletsky study, 74% of patients experienced grade 3 or 4 neutropenia. One case of acute respiratory distress syndrome Fig. 3. A, overall survival for DE arm. B, overall survival for DVarm. Fig. 2. A, progression-free survival for DE arm. B, progression-free survival for DVarm. Docetaxel,Vinorelbine, and Estramustine Phase II Study www.aacrjournals.org Clin Cancer Res 2006;12(20) October15, 20066097 Research. on April 5, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from http://clincancerres.aacrjournals.org/ was also observed. An objective response was seen in 60% of patients with a PSA response in 60% of patients also. Patients in the Koletsky study had a significantly lower median PSA level at baseline (116 ng/mL) compared with our study population (242 ng/mL), which may account for the somewhat decreased response rates seen in our experience. Insufficient antitumor activity associated with weekly docetaxel dosing must also be considered. The weekly dosing of both docetaxel and vinor- elbine in our trial was based on prior results of a phase I dose escalation study in non–small cell lung cancer patients, which revealed a maximally tolerated dose of 20 mg/m2/wk of vinorelbine and 25 mg/m2/wk of docetaxel without scheduled treatment breaks (33). The total number of patients experiencing clinically signifi- cant toxicity with the combination of vinorelbine and docetaxel did not exceed our prestudy threshold. However, the efficacy results do not suggest an advantage compared with single-agent docetaxel. As such, we would not recommend further study of the combination regimen at this dosing schedule. However, clinicians should be reminded of the single-agent tolerability of vinorelbine, particularly in elderly patients often encountered with metastatic HRPC. The companion pharmacogenetic study assessed germ-line polymorphisms in genes known to play important roles in chemotherapy drug transport, metabolism, and mechanism of action. Analysis of candidate genes showed a significantly greater proportion of patients surviving beyond 15 months with docetaxel-based therapy in the presence of the ABCG2 421 C>A polymorphism. Previous studies of the ABCB1-associated P-glycoprotein-mediated drug efflux pump have shown mini- mal, if any, effect on docetaxel plasma concentrations when an inhibitor of this pump is administered (34). The effect of ABCG2 polymorphisms on docetaxel pharmacokinetics is unknown. The increased survival seen in individuals with an ABCG2 421 C>A polymorphism may suggest a less functional drug efflux pump, leading to increased intracellular (intra- tumoral) docetaxel concentrations and improved cytotoxic activity. This hypothesis should be interpreted cautiously due to the small patient sample size and potential confounding variables. In addition, this exploratory analysis was not subject to a multivariate analysis. Therefore, at most, these data are hypothesis generating and will be used as preliminary data for future large-scale studies. Nonetheless, the study shows the ability to successfully conduct translational pharmacogenetic studies in a community setting, such as the Hoosier Oncology Group. In summary, this randomized phase II study of two docetaxel-based regimens showed a tolerable toxicity profile with a weekly docetaxel and vinorelbine regimen. However, the efficacy data do not support evaluation in the phase III setting. A pharmacogenetic analysis of germ-line DNA showed that patients with an ABCG2 421 C>A genotype had an increased chance of being alive beyond 15 months if treated with docetaxel-based combination chemotherapy. This study shows that modern translational efforts can be accomplished beyond the confines of large tertiary academic medical centers and should serve as a model for future trial design. Table 2. Pharmacogenetic polymorphism analysis in patients surviving >15 months Gene Polymorphisms P ABCB1 3435 C/C, 4/10 (40%) C/T, 7/29 (24%) T/T, 6/12 (50%) 0.24 ABCG2 421 C/C, 12/44 (27%) C/A, 4/6 (67%) A/A, 0/0 (0%) 0.05 CYP3A4*1B A/A, 16/43 (37%) A/G, 0/5 (0%) G/G, 1/2 (50%) 0.10 CYP3A5*3C A/A, 1/2 (50%) A/G, 0/5 (0%) G/G, 16/44 0.10 MAP4 68 G/G, 14/46 (30%) G/A, 3/5 (60%) A/A, 0/0 (0%) 0.18 MAP4 1280 C/C, 7/25 (28%) C/A, 5/18 (28%) A/A, 3/5 (60%) 0.37 MAPT-13 G/G, 0/3 (0%) G/A, 7/18 (39%) A/A, 10/29 (34%) 0.26 MAPT 3674 A/A, 10/30 (33%) G/A, 6/17 (35%) G/G, 0/3 (0%) 0.29 Cancer Therapy: Clinical www.aacrjournals.orgClin Cancer Res 2006;12(20) October15, 2006 6098 References 1. Jemal A, Siegel R,Ward E, et al. Cancer Statistics, 2006. CA: A CancerJournal for Clinicians 2006;56: 106^30. 2. Petrylak DP,Tangen CM, Hussain MH, et al. Doce- taxel and Estramustine Compared with Mitoxantrone and Prednisone for Advanced Refractory Prostate Cancer. NEnglJMed 2004;351:1513^20. 3.Tannock IF, DeWit R, BerryWR, et al. Docetaxelplus Prednisone or Mitoxantrone plus Prednisone in Advanced Prostate Cancer. N Engl JMed 2004;351: 1502^12. 4.MatheG,ReizensteinP.PhaseIpharmacologicstudy of a newVinca alkaloid: navelbine. Cancer Lett1985; 27:285^93. 5. Fields-Jones S, KoletskyA,Wilding G, et al. Improve- ments in clinical benefit with vinorelbine in the treat- ment of hormone-refractory prostate cancer: a phase II trial. Ann Oncol1999;10:1307^10. 6. Sweeney CJ, Monaco FJ, Jung SH, et al. A phase II Hoosier Oncology Group study of vinorelbine and estramustine phosphate in hormone-refractory pros- tate cancer. Ann Oncol 2002;13:435^40. 7.BisseryMC,VrignaudP,LavelleF.Preclinicalprofileof docetaxel (taxotere): efficacy as a single agent and in combination. Semin Oncol1995;22 Suppl13:3^16. 8. Aoe K, Kiura K, Ueoka H, et al. Effect of docetaxel with cisplatin or vinorelbine on lung cancer cell lines. Anticancer Res1999;19:291^9. 9. KreisW, Budman DR, Calabro A. Unique synergism or antagonism of combinations of chemotherapeutic and hormonal agents in human prostate cancer cell lines. BrJUrol1997;79:196^202. 10. Black AJ, McLeod HL, Capell HA, et al.Thiopurine methyltransferase genotype predicts therapy-limiting severe toxicity from azathioprine. Ann Intern Med 1998;129:716^8. 11. MilanoG,McLeodHL.Candihydropyrimidinedehy- drogenase impact 5-fluorouracil-based treatment? EurJCancer 2000;36:37^42. 12. Innocenti F, Undevia SD, Iyer L, et al. Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irino- tecan. J Clin Oncol 2004;22:1382^8. 13. Tayeb MT, Clark C, Ameyaw MM, et al. CYP3A4 promoter variant in Saudi, Ghanaian, and Scottish Caucasian populations. Pharmacogenetics 2000;10: 753^6. 14. Kuehl P, ZhangJ, LinY, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nature Gene 2001;27:383^91. 15. AmeyawMM,RegateiroF,LiT,etal.MDR1pharma- cogenetics: frequency of the C3435T mutation in exon 26 is significantly influenced by ethnicity. Phar- macogenetics 2001;11:217^21. 16. Sangrajrang S, DenouletP, Millot G, et al.Estramus- tineresistancecorrelateswithH over-expressioninhu- man prostatic carcinoma cells. Int J Cancer 1998;77: 626^31. 17. Doyle LA, Ross DD. Multidrug resistance mediated Research. on April 5, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from http://clincancerres.aacrjournals.org/ Docetaxel,Vinorelbine, and Estramustine Phase II Study www.aacrjournals.org Clin Cancer Res 2006;12(20) October15, 20066099 by thebreastcancer resistanceproteinBCRP(ABCG2). Oncogene 2003;22:7340^58. 18. Bubley GJ, Carducci M, Dahut W, et al. Eligibility and response guidelines for phase II clinical trials in androgen-independent prostate cancer: recommen- dations from the Prostate-Specific AntigenWorking Group. JClin Oncol1999;17:3461^7. 19. Garsa AA, McLeod HL, Marsh S. CYP3A4 and CYP3A5 genotyping by Pyrosequencing. BMC Med Genet 2005;6:19. 20. MathijssenRH,MarshS,KarlssonMO,etal.Irinote- can pathway genotype analysis to predictpharmaco- kinetics. Clin Cancer Res 2003;9:3246^53. 21. Sparreboom A, Gelderblom H, Marsh S, et al. Diflomotecan pharmacokinetics in relation to ABCG2 421C>A genotype. Clin Pharmacol Ther 2004;76: 38^44. 22. Marsh S, King CR, Garsa AA, McLeod HL.Pyrose- quencing of clinically relevant polymorphisms. Meth- ods Mol Biol 2005;311:97^114. 23. PetrylakDP, Macarthur RB, O’ConnorJ, et al. Phase I trial of docetaxel with estramustine in androgen- independent prostate cancer. J Clin Oncol 1999;17: 958^67. 24. Kreis W, Budman DR, Fetten J, Gonzales AL, Barile B,Vinciguerra V. Phase I trial of the combina- tion of daily estramustine phosphate and intermit- tent docetaxel in patients with metastatic hormone refractory prostate carcinoma. Ann Oncol 1999;10: 33^8. 25. Savarese DM, Halabi S, HarsV, et al. Phase II study of docetaxel, estramustine, and low-dose hydrocorti- soneinmenwithhormone-refractoryprostatecancer: a final report of CALGB 9780. Cancer and Leukemia Group B. JClin Oncol 2001;19:2509^16. 26. Sinibaldi VJ, Carducci MA, Moore-Cooper S, Laufer M, Sahurak M, Eisenberger MA. Phase II eval- uation of docetaxel plus one-day oral estramustine phosphate in the treatment of patients with androgen independent prostate carcinoma. Cancer 2002;94: 1457^65. 27. Budman DR, Calabro A, KreissW. Synergistic and antagonistic combinations of drugs inhumanprostate cancer cell lines in vitro. AntiCancer Drugs 2002;13: 1011^6. 28. OudardS, CatyA, HumbletY,etal. Phase IIstudyof vinorelbine in patients with androgen-independent prostate cancer. Ann Oncol 2001;12:847^52. 29. Morant R, Hsu Schmitz SF, BernhardJ, et al.Vinor- elbine in androgen-independent metastatic prostatic carcinomaLa phase II study. EurJ Cancer 2002;38: 1626^32. 30.Tralongo P, Bollina R, Aiello R, et al.Vinorelbine and prednisone in older cancer patients with hormone- refractorymetastaticprostatecancer.AphaseIIstudy. Tumori 2003;89:26^30. 31. Robles C, Furst AJ, Sriratana P, et al. Phase II study of vinorelbine with low dose prednisone in the treat- ment of hormone-refractory metastatic prostate can- cer. Oncol Rep 2003;10:885^9. 32. KoletskyAJ,GuerraML,KronishL.PhaseIIstudyof vinorelbine andlow-dose docetaxel inchemotherapy- naI«ve patients with hormone-refractory prostate can- cer. CancerJ 2003;9:286^92. 33. JohnstonE,CrawfordJ,GarstJ,etal.PhaseItrialof weeklydocetaxel(D)andvinorelbine(V) inadvanced non-small cell lung cancer (NSCLC) [abstract 1838]. Proc Am Soc Clin Oncol1999;18:476. 34.VanZuylenL,VerweijJ,NooterK,BrouwerE,Stoter G, Sparreboom A. Role of intestinal P-glycoprotein in the plasma and fecal disposition of docetaxel in humans. Clin Cancer Res 2000;6:2598^603. Research. on April 5, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from http://clincancerres.aacrjournals.org/ 2006;12:6094-6099. Clin Cancer Res Noah M. Hahn, Sharon Marsh, William Fisher, et al. Survival Analysis Hormone-Refractory Prostate Cancer with Pharmacogenetic Docetaxel, Vinorelbine, and Estramustine in Combination in Hoosier Oncology Group Randomized Phase II Study of Updated version http://clincancerres.aacrjournals.org/content/12/20/6094 Access the most recent version of this article at: Cited articles http://clincancerres.aacrjournals.org/content/12/20/6094.full#ref-list-1 This article cites 33 articles, 6 of which you can access for free at: Citing articles http://clincancerres.aacrjournals.org/content/12/20/6094.full#related-urls This article has been cited by 2 HighWire-hosted articles. Access the articles at: E-mail alerts related to this article or journal.Sign up to receive free email-alerts Subscriptions Reprints and .pubs@aacr.orgDepartment at To order reprints of this article or to subscribe to the journal, contact the AACR Publications Permissions Rightslink site. (CCC) Click on "Request Permissions" which will take you to the Copyright Clearance Center's .http://clincancerres.aacrjournals.org/content/12/20/6094 To request permission to re-use all or part of this article, use this link Research. on April 5, 2021. © 2006 American Association for Cancerclincancerres.aacrjournals.org Downloaded from http://clincancerres.aacrjournals.org/content/12/20/6094 http://clincancerres.aacrjournals.org/content/12/20/6094.full#ref-list-1 http://clincancerres.aacrjournals.org/content/12/20/6094.full#related-urls http://clincancerres.aacrjournals.org/cgi/alerts mailto:pubs@aacr.org http://clincancerres.aacrjournals.org/content/12/20/6094 http://clincancerres.aacrjournals.org/