PATIENTS WITH RECURRENT malignant gliomas,

Similar documents
PROCARBAZINE, lomustine, and vincristine (PCV) is

Carmustine implants and Temozolomide for the treatment of newly diagnosed high grade glioma

Collection of Recorded Radiotherapy Seminars

Concomitant (without adjuvant) temozolomide and radiation to treat glioblastoma: A retrospective study

성균관대학교삼성창원병원신경외과학교실신경종양학 김영준. KNS-MT-03 (April 15, 2015)

Systemic Treatment. Third International Neuro-Oncology Course. 23 May 2014

Temozolomide in the treatment of recurrent malignant glioma in Chinese patients!"#$%&'()*+,-./0,1234

Contemporary Management of Glioblastoma

See the corresponding editorial in this issue, pp 1 2. J Neurosurg 115:3 8, An extent of resection threshold for newly diagnosed glioblastomas

Bevacizumab rescue therapy extends the survival in patients with recurrent malignant glioma

Incidence of Early Pseudo-progression in a Cohort of Malignant Glioma Patients Treated With Chemoirradiation With Temozolomide

UPDATES ON CHEMOTHERAPY FOR LOW GRADE GLIOMAS

High-grade glioma (World Health Organization Grades III and IV) Salvage Therapy in Patients with Glioblastoma. Is There any Benefit?

Introduction ORIGINAL RESEARCH

Prior to 1993, the only data available in the medical

Scottish Medicines Consortium

University of Zurich. Temozolomide and MGMT forever? Zurich Open Repository and Archive. Weller, M. Year: 2010

Advances in gastric cancer: How to approach localised disease?

Protocol Abstract and Schema

We have previously reported good clinical results

Locoregional treatment Session Oral Abstract Presentation Saulo Brito Silva

Research Article Prognostic Factors in Advanced Non-Small-Cell Lung Cancer Patients: Patient Characteristics and Type of Chemotherapy

A new score predicting the survival of patients with spinal cord compression from myeloma

Zurich Open Repository and Archive. Long-term survival of glioblastoma patients treated with radiotherapy and lomustine plus temozolomide

Practice of Interferon Therapy

Efficacy of neuroradiological imaging, neurological examination, and symptom status in follow-up assessment of patients with high-grade gliomas

Survival of Pediatric Patients After Relapsed Osteosarcoma: The St. Jude Children s Research Hospital Experience

NICE Single Technology Appraisal of cetuximab for the treatment of recurrent and /or metastatic squamous cell carcinoma of the head and neck

The role of cytoreductive. nephrectomy in elderly patients. with metastatic renal cell. carcinoma in an era of targeted. therapy

Zurich Open Repository and Archive. Long-term survival of glioblastoma patients treated with radiotherapy and lomustine plus temozolomide

Survival and Intracranial Control of Patients With 5 or More Brain Metastases Treated With Gamma Knife Stereotactic Radiosurgery

Technology appraisal guidance Published: 27 June 2007 nice.org.uk/guidance/ta121

Prognostic significance of surgery and radiation therapy in cases of anaplastic astrocytoma: retrospective analysis of 170 cases

SUPPLEMENTARY MATERIAL

Clinicopathological Factors Affecting Distant Metastasis Following Loco-Regional Recurrence of breast cancer. Cheol Min Kang 2018/04/05

3/8/2014. Case Presentation. Primary Treatment of Anal Cancer. Anatomy. Overview. March 6, 2014

GSK Medicine: Study Number: Title: Rationale: Study Period: Objectives: Indication: Study Investigators/Centers: Research Methods: Data Source

Hypofractionated radiation therapy for glioblastoma

Sponsor / Company: Sanofi Drug substance(s): Docetaxel (Taxotere )

CLINICAL INVESTIGATION of new agents and combination

Prognostic significance of K-Ras mutation rate in metastatic colorectal cancer patients. Bruno Vincenzi Università Campus Bio-Medico di Roma

Antiangiogenic drugs in unresectable glioblastoma. Dra. Carmen Balañá. /

trial update clinical

Treatment outcomes and prognostic factors of gallbladder cancer patients after postoperative radiation therapy

University of Alberta. Evaluation of Concomitant Temozolomide Treatment in Glioblastoma Multiforme Patients in Two Canadian Tertiary Care Centers

Interferon β and temozolomide combination therapy for temozolomide monotherapy refractory malignant gliomas

Precision Medicine Lessons from meta-analyses of 70,253 patients

A Randomized Phase III study: Comparison between Intravenous and Intraarterial ACNU Administration in Newly Diagnosed Primary Glioblastomas

Protocol Abstract and Schema

Arecent randomized controlled trial (RCT) established

INTRODUCTION TO SURVIVAL CURVES

J Clin Oncol 22: by American Society of Clinical Oncology INTRODUCTION

National Horizon Scanning Centre. Bevacizumab (Avastin) for glioblastoma multiforme - relapsed. August 2008

Best Papers. F. Fusco

Intraoperative Radiotherapy

Digital Washington University School of Medicine. Russell Schilder Fox Chase Comprehensive Cancer Center. Arturo Molina Biogen Idec

HSV1716 Dose levels and Cohort size Dose level No of Patients HSV1716 Dosage 1* 3 to 6 1 ml of 1 x 10 5 infectious units HSV1716 per ml 2

21/03/2017. Disclosure. Practice Changing Articles in Neuro Oncology for 2016/17. Gliomas. Objectives. Gliomas. No conflicts to declare

THE EFFECTIVE OF BRAIN CANCER AND XAY BETWEEN THEORY AND IMPLEMENTATION. Mustafa Rashid Issa

Supplementary Appendix to manuscript submitted by Trappe, R.U. et al:

Sergio Bracarda MD. Head, Medical Oncology Department of Oncology AUSL-8 Istituto Toscano Tumori (ITT) San Donato Hospital Arezzo, Italy

Enormous effort has been invested in clinical trials for malignant

The Impact of Adjuvant Chemotherapy in Pulmonary Large Cell Neuroendocrine Carcinoma (LCNC)

Factors associated with delayed time to adjuvant chemotherapy in stage iii colon cancer

Defining clinical and statistical improvement in consolidation or maintenance single-arm trials in Oncology

Viable Germ Cell Tumor at Postchemotherapy Retroperitoneal Lymph Node Dissection. Can We Predict Patients at Risk of Disease Progression?

CON: Removal of the Breast Primary in Patients with Metastatic Breast Cancer

The effect of delayed adjuvant chemotherapy on relapse of triplenegative

HIV/AIDS CLINICAL CARE QUALITY MANAGEMENT CHART REVIEW CHARACTERISTICS OF PATIENTS FACTORS ASSOCIATED WITH IMPROVED IMMUNOLOGIC STATUS

A Single Institution s Experience with Bevacizumab in Combination with Cytotoxic Chemotherapy in Progressive Malignant Glioma

Correlation of pretreatment surgical staging and PET SUV(max) with outcomes in NSCLC. Giancarlo Moscol, MD PGY-5 Hematology-Oncology UTSW

Prognostic Factors for Survival in Patients Treated With Stereotactic Radiosurgery for Recurrent Brain Metastases After Prior Whole Brain Radiotherapy

Efficacy of Treatment for Glioblastoma Multiforme in Elderly Patients (65+): A Retrospective Analysis

Temozolomide in combination with BCNU before and after radiotherapy in patients with inoperable newly diagnosed glioblastoma multiforme

Update: Chronic Lymphocytic Leukemia

Treatment With Bevacizumab and Irinotecan for Recurrent High-Grade Glial Tumors

Radioterapia no Tratamento dos Gliomas de Baixo Grau

Durable Responses Observed in Recurrent High Grade Glioma (rhgg) with Toca 511 & Toca FC Treatment

Accepted 28 April 2005 Published online 13 September 2005 in Wiley InterScience ( DOI: /hed.

Improved Survival after Gross Total Resection of Malignant Gliomas in Pediatric Patients from the HIT-GBM Studies

Criteria for evaluating patients undergoing chemotherapy for malignant brain tumors

Ependymoma Programme Synopsis

Liver Transplantation for Alcoholic Liver Disease in the United States: 1988 to 1995

Bevacizumab and dose-intense temozolomide in recurrent high-grade glioma

Principal Investigator: Robert J. Jones, MD, Beatson Cancer Center, 1053 Great Western Road, Glasgow; United Kingdom

Role of Primary Resection for Patients with Oligometastatic Disease

Citation Characteristics of Research Published in Emergency Medicine Versus Other Scientific Journals

To analyse whether ADC values have a correlation with survival or EGFR amplification status in glioblastoma

Corporate Medical Policy

A Population-Based Study on the Uptake and Utilization of Stereotactic Radiosurgery (SRS) for Brain Metastasis in Nova Scotia

Retroperitoneal Soft Tissue Sarcomas: Prognosis and Treatment of Primary and Recurrent Disease in 117 Patients

Selecting the Optimal Treatment for Brain Metastases

Supplementary Material

Synopsis (C1034T02) CNTO 95 Module 5.3 Clinical Study Report C1034T02

Durable Responses Observed in IDH1 Wildtype and Mutant Recurrent High Grade Glioma with Toca 511 & Toca FC Treatment

Timing of targeted therapy in patients with low volume mrcc. Eli Rosenbaum Davidoff Cancer Center Beilinson Hospital

Phase 2 study of BCNU and temozolomide for recurrent glioblastoma multiforme: North American Brain Tumor Consortium study 1

Corporate Medical Policy

ASSESSING LONG-TERM BENEFITS OF IMMUNOTHERAPY BASED ON EARLY TUMOR ASSESSMENT DATA

How to improve the reliability of Single Arm Trials

Transcription:

Outcomes and Prognostic Factors in Recurrent Glioma Patients Enrolled Onto Phase II Clinical Trials By Eric T. Wong, Kenneth R. Hess, Mary Jo Gleason, Kurt A. Jaeckle, Athanassios P. Kyritsis, Michael D. Prados, Victor A. Levin, and W.K. Alfred Yung PATIENTS WITH RECURRENT malignant gliomas, such as glioblastoma multiforme (GBM) and anaplastic astrocytoma (AA), have poor outcome. Repeat surgery may not be feasible because of tumor infiltration into eloquent areas of the brain, and additional irradiation has limited control on further tumor growth and would potentiate neurologic toxicity. Facing limited treatment options, patients are usually enrolled onto chemotherapy trials and receive investigational drugs. Published phase II trials for malignant gliomas often include a population with heterogeneous clinical characteristics. Although several prognostic factors are well established for patients newly diagnosed with malignant gliomas, those that are important at recurrence have not been extensively studied. Age, histology, extent of surgery, and performance status are important prognostic variables for malignant gliomas at From the Departments of Neuro-Oncology and Biomathematics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Neurological Surgery, University of California, San Francisco, CA. Submitted May 11, 1998; accepted March 19, 1999. Supported by National Cancer Institute grants no. CA55621, CA62412 and CA62431. Presented in part at the Thirty-Second Annual Meeting of the American Society of Clinical Oncology, Philadelphia, PA, May 18-21, 1996. Address reprint requests to W.K. Alfred Yung, MD, Department of Neuro-Oncology, Box 100, The University of Texas, M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030. 1999 by American Society of Clinical Oncology. 0732-183X/99/1708-2572 Purpose: To determine aggregate outcomes and prognostic covariates in patients with recurrent glioma enrolled onto phase II chemotherapy trials. Patients and Methods: Patients from eight consecutive phase II trials included 225 with recurrent glioblastoma multiforme (GBM) and 150 with recurrent anaplastic astrocytoma (AA). Their median age was 45 years (range, 15 to 82 years) and their median Karnofsky performance score was 80 (range, 60 to 100). Prognostic covariates were analyzed with respect to tumor response, progression-free survival (PFS), and overall survival (OS) by multivariate logistic and Cox proportional hazards regression analyses. Results: Overall, 34 (9%) had complete or partial response, whereas 80 (21%) were alive and progression-free at 6 months (APF6). The median PFS was 10 weeks and median OS was 30 weeks. Histology was a robust prognostic factor across all outcomes. GBM patients had significantly poorer outcomes than AA patients. The APF6 proportion was 15% for GBM and 31% for AA, whereas the median PFS was 9 weeks for GBM and 13 weeks for AA. Results were also significantly poorer for patients with more than two prior surgeries or chemotherapy regimens. Conclusion: Histology is a dominant factor in determining outcome in patients with recurrent glioma enrolled onto phase II trials. Future trials should be designed with separate histology strata. J Clin Oncol 17:2572-2578. 1999 by American Society of Clinical Oncology. initial diagnosis. First, age significantly affects survival 1 and strongly correlates with chemosensitivity of these tumors to nitrosourea in vitro 2 and in clinical series. 3 Second, histology, particularly the presence of necrosis as in GBM, is another important prognostic factor. 1,4 The median survival from initial diagnosis is 11 months for GBM as compared with 27 months for AA. 5 Third, although preoperative tumor size has no prognostic significance, a major reduction in postoperative size prolongs patient survival, particularly in those with minimal residual disease after at least two log units of tumor removal. 6 Lastly, multiple clinical trials have found performance status, in either Karnofsky performance score (KPS) or Eastern Cooperative Oncology Group scales, to be a significant prognostic variable. 5,7 However, the prognostic factors for patients with recurrent malignant gliomas remain unclear. Because phase II trials typically enroll patients with recurrent disease after they have failed established therapies, the small number of patients in each of these studies, typically 14 to 30 subjects, 8 precludes a robust analysis of prognostic variables because of insufficient statistical power. Eagan and Scott 9 combined 103 patients with recurrent malignant gliomas from five chemotherapy trials and found that none of the prognostic factors correlated with response or time to progression (TTP), except for performance score, which correlated with survival. However, their study was published more than a decade ago, and improvements in neuroimaging, surgical techniques, and chemotherapy may alter the prognosis of current patients. Moreover, a recently published phase II study on 51 adults with recurrent malignant gliomas sug- 2572 Journal of Clinical Oncology, Vol 17, No 8 (August), 1999: pp 2572-2578

OUTCOMES AND PREDICTORS FOR RECURRENT GLIOMA 2573 gested that histology, myelotoxicity, and prior chemotherapy had significant correlations with time to tumor progression and survival. 10 We combined 375 patients from eight phase II trials for analysis of outcomes and prognostic factors associated with recurrent GBM and recurrent AA. PATIENTS AND METHODS Patient Inclusion Patients were drawn from eight consecutive phase II chemotherapy trials conducted at The University of Texas M.D. Anderson Cancer Center. From 1986 to 1995, seven trials using interferon beta (IFN ), IFN combined with 13-cis-retinoic acid (CRA), menogaril, carboplatin, and a combination of carboplatin, fluorouracil (5-FU), and procarbazine were carried out at M.D. Anderson Cancer Center. One trial using difluoromethylornithine (DFMO) was carried out at M.D. Anderson Cancer Center and at the University of California, San Francisco. Menogaril, carboplatin, 5-FU, and procarbazine are known cytotoxic drugs, whereas IFN, CRA, and DFMO are thought to be cytostatic agents that mediate antitumor effect by blocking cell growth without effecting cell kill. All trials were approved by the respective institutional review boards, and informed consent was obtained from each patient or guardian before enrollment. Two trials administered intravenous IFN at a dose of 90 million IU once daily on Mondays, Wednesdays, and Fridays. Another trial used IFN at 6 or 18 million units daily on Mondays, Wednesdays, and Fridays, with or without CRA at 100 mg/m 2 /d. Menogaril was administered intravenously at a dose of 200 mg/m 2 initially and was increased to 225 mg/m 2 in subsequent cycles if tolerated; the drug was repeated every 4 weeks. DFMO was administered orally at a dose of 3.6 gm/m 2 on days 1 to 14, 22 to 35, and 43 to 56; the cycle was repeated on day 63. Carboplatin was given intravenously at a dose of 400 mg/m 2 initially and was increased to 475 and 525 mg/m 2 if tolerated; the drug was repeated every 4 weeks. Two trials used a combination of carboplatin, 5-FU, and procarbazine. Carboplatin and 5-FU were given by continuous intravenous infusion at a daily dose of 75 mg/m 2 /d and at 1,000 mg/m 2 /d on days 1 to 4, respectively. Oral procarbazine was administered at a dose of 75 mg/m 2 /d for 10 days on days 5 to 14. Patient eligibility and response criteria were similar among the trials. Briefly, patients with histologically identified malignant gliomas at recurrence were eligible for study. All eight studies required evidence of recurrence after prior radiotherapy and all had similar age criteria (eg, age 16 years) and performance criteria (eg, KPS 50). Each of the eight studies had a two-stage design (the first five were Gehan designs and the last three were Simon designs). 8,11 Three studies were stopped after the first stage and enrolled fewer than 15 patients. Two studies were two-arm randomized phase II trials and three studies used separate strata for patients with and without previous chemotherapy. Although five of the eight studies did not restrict entry on the basis of previous chemotherapy, one trial excluded patients with any previous chemotherapy and two trials excluded patients with more than one previous chemotherapy regimen. Patients enrolled onto the studies were evaluated by neurologic examinations and neuroimaging studies of the brain, contrast-enhanced computed tomography, gadolinium-enhanced magnetic resonance imaging, or both. 12 Evaluations were performed at 2-month intervals during the first year, at 3-month intervals during the second year, at 4-month intervals during the third year, or until disease progression. Response to chemotherapy was assessed by radiographic criteria and extent of corticosteroid use. Complete response (CR) was defined as complete resolution of tumor as determined by neuroimaging studies with the patient on a stable dose of corticosteroid. Partial response (PR) consisted of a decrease in tumor volume by 50% or greater in the product of two diameters with no increase in corticosteroid dosage. Minor response (MR) involved a decrease in tumor size by less than 50% with stable or decreasing corticosteroid dose. Stable disease (SD) referred to no change in tumor size with stable or decreasing corticosteroid dose. Progressive disease was defined as definite worsening or development of a new lesion on imaging study with stable or increasing corticosteroid dose. Patients who experienced medical complications, refused further participation in the study, developed toxicity, or died within 3 weeks of treatment were deemed nonassessable according to study protocols. To avoid postentry exclusion bias, these patients were included in our analysis. The response of those who suffered from an event at 3 weeks or later was based on the latest clinical and neuroimaging evaluations. Three binary outcomes were defined: (1) CR or PR; (2) CR, PR, MR, or SD; and (3) alive and progression-free at 6 months (APF6). We also defined two time-to-event end points: progression-free survival (PFS) and overall survival (OS). Both were measured from the date of enrollment. PFS measures TTP or death, whichever occurs first. PFS at 6 months is equivalent to APF6. Statistical Analysis To evaluate the effects of prognostic factors on outcomes, odds ratios for the three binary end points were estimated with logistic regression, 13 whereas hazard ratios for the two time-to-event end points were estimated using Cox regression. 14 Both univariate and multivariate analyses were performed for each end point. PFS and OS curves were constructed using the Kaplan-Meier method, 15 and estimates of medians and their respective 95% confidence intervals (CIs) were calculated from the Kaplan-Meier estimates. We also performed recursive partitioning in an attempt to identify prognostic subgroups. To perform the recursive partitioning analysis for censored data, we used the null martingale-based residual as input into the S-PLUS (MathSoft, Inc, Seattle, WA) implementation of tree-based methods. 16,17 The assumptions of the regression analyses were verified with residual analyses. Patient Characteristics The eight chemotherapy trials accounted for a total of 458 enrollments. Eighty-three patients were excluded from analysis: 60 did not have GBM or AA (histologies included 18 mixed gliomas, 14 astrocytomas, 10 anaplastic oligodendrogliomas, eight oligodendrogliomas, five unclassified gliomas, three gliosarcomas, and two ependymomas), 18 AA or GBM patients were enrolled in more than one of the eight trials and only the first enrollment was counted, four patients were retrospectively judged to be ineligible for enrollment, and one patient had a KPS of 50. The histology and KPS exclusions were performed to increase patient homogeneity. A total of 375 unique patients remain for analysis. The histologic diagnosis at the time of registration was recurrent GBM in 225 patients (60%) and recurrent AA in 150 patients (40%). The median age at enrollment was 45 years (range, 15 to 82 years). Six patients were younger than 20 years and five patients were older than 70. The median KPS at enrollment was 80 (range, 60 to 100). Twenty-two patients had KPS of 100 and 14 had KPS of 60. Two hundred eighty-five patients (77%) had received prior chemotherapy, whereas 92 patients (25%) had received at least two prior chemotherapy regimens. The maximum extent of previous surgery was biopsy in 46

2574 WONG ET AL patients (12%), partial resection in 194 patients (52%), and gross total resection in 135 patients (36%). One hundred twenty-eight patients (34%) had two prior surgeries, whereas 27 patients (7%) had more than two prior surgeries. We do not have data on the number of previous recurrences (ie, progressions), but this should be reflected in the number of previous chemotherapy regimens. Furthermore, although it is straight forward to determine whether a patient had prior chemotherapy, the lack of standardized diagnostic criteria make the determination of prior progression problematic. To separate out heavily pretreated (salvage) patients, we identified 41 patients with more than two prior chemotherapies and/or more than two prior surgeries. RESULTS Outcomes Among the 375 patients, 34 (9%) experienced CR (n 1) or PR (n 33), 18 (5%) experienced MR, and 94 (25%) experienced SD. Thus a total of 146 (39%) experienced CR, PR, MR, or SD. A total of 80 (21%) were alive and progression-free at 6 months (ie, the overall APF6 was 21%). The 95% CI for APF6 was 17% to 26%. A total of 343 patients experienced a progression of disease by the end of the study and a total of 298 patients had died by the end of the study. Fifty-nine patients were alive after progression and 14 patients died without radiographic evidence of progression (although most died from their brain cancer). These 14 patients constitute the difference between freedom from progression (in which case these patients would be censored and not count as events) and progression-free survival (in which case these patients are included as events). Thus our results for PFS would be similar to results using freedom from progression (357 events v 343 events). Eighteen patients were alive without progression with a median follow-up of 5.1 years (range, 35 weeks to 8.1 years). Nine patients with best protocol response of CR or PR had disease progression by 6 months after enrollment. The median OS time in the total patient group was 30 weeks, 6-month survival proportion was 55%, 1-year survival proportion was 32%, and 5-year survival proportion was 10%. The 95% CI for median OS time was 26 to 35 weeks. The median PFS time was 10 weeks, 6-month PFS proportion was 21%, 1-year PFS proportion was 12% and 5-year PFS proportion was 4%. The 95% CI for the median PFS time was 9 to 11 weeks. Figure 1 shows the OS and PFS curves for the 375 patients. Seventy-nine percent of patients had failed by 26 weeks, and 26 weeks marks the beginning of the long tail in the PFS curve shown in Fig 1. This suggests that 26 weeks would be a convenient time point to capture most of the failures (hence our use of AFP6). Prognostic Factors Table 1 shows the five outcomes according to the levels of the four covariates (histology, age 40 years, KPS 80, and salvage therapy). As listed in Tables 1 and 2, for all five outcomes, GBM patients had significantly poorer outcomes as compared with AA patients. APF6 was 15% for GBM Fig 1. OS and PFS in 375 patients. OS is 55%, 32%, and 10% at 26, 52, and 260 weeks, respectively; PFS is 21%, 12%, and 4% at 26, 52, and 260 weeks, respectively.

OUTCOMES AND PREDICTORS FOR RECURRENT GLIOMA 2575 Variable No. of Patients Table 1. Prognostic Factors CR/PR* C/P/M/S APF6 PFS at 1 year OS at 1 year Histology AA 150 14 48 31 20 47 GBM 225 6 33 15 8 21 Age 40 years 127 12 36 28 17 40 40 years 248 8 40 18 10 28 KPS 80 198 7 36 18 9 22 80 177 12 42 25 16 42 Salvage No 332 10 41 23 13 33 Yes 41 5 20 10 7 21 All patients 375 9 39 21 12 32 *CR/PR CR or PR. C/P/M/S CR or PR or MR or SD. APF6 PFS at 6 months (binary endpoint). Kaplan-Meier estimates. (95% CI, 10% to 19%) and 31% for AA (95% CI, 24% to 39%). Figure 2 shows PFS curves according to histology. The 5-year OS proportion was 2% for GBM and 21% for AA. The median PFS time was 9 weeks for GBM (95% CI, 8 to 10 weeks) and 13 weeks for AA (95% CI, 10 to 18 weeks). The median OS time was 25 weeks for GBM (95% CI, 21 to 28 weeks) and 47 weeks for AA (95% CI, 38 to 64 weeks). Table 3 shows the multivariate results for the five end points. Histology remains a significant predictor for all end points. Salvage treatment is significantly associated with poorer outcome for four of the five end points (the CR/PR analysis lacks adequate statistical power). KPS was significantly associated with survival only, whereas age was not related to outcome. The results for age and KPS were similar when these variables were analyzed as continuous covariates. We decided to use a salvage treatment dichotomy to summarize the previous treatment data because our data indicated that patients with fewer than three prior surgeries and fewer than three prior chemotherapies had similar responses regardless of the actual number of surgeries or Table 2. Outcomes: GBM v AA OR* HR 95% CI P CR/PR 2.7 1.3, 5.4.0081 CR/PR/MR/SD 1.9 1.2, 2.9.0036 APF6 2.7 1.6, 4.4.0002 PFS 1.6 1.3, 2.0.0001 OS 2.0 1.6, 2.6.0001 NOTE. Values greater than 1 indicate that GBM patients have higher risk of poor outcomes. Abbreviations: OR, odds ratio for nonresponse; HR, hazards ratio. chemotherapies. For example, among patients with only one prior surgery, the 48 patients with no prior chemotherapy had a 29% APF6, the 121 patients with one prior chemotherapy had a 20% APF6, and the 41 patients with two prior chemotherapies had a 24% APF6. Similarly, among patients with two prior surgeries, the 31 with no prior chemotherapy had a 23% APF6, the 63 with one prior chemotherapy had a 24% APF6, and the 28 with two prior chemotherapies had a 21% APF6. Thus, the 332 patients with no more than two prior chemotherapies and no more than two prior surgeries had an aggregate 23% APF6 proportion, compared with 10% for the 41 patients with more than two prior surgeries or more than two prior chemotherapies (ie, our salvage treatment patients). Additional analyses were performed to compare the effects of cytotoxic and cytostatic chemotherapies in our cohort. No statistical differences in outcomes were found between the patients treated with cytotoxic agents and those treated with cytostatic agents. For example, 21 (21%) of 102 patients who received cytotoxic agents were alive and progression-free at 6 months compared with 59 (22%) of the 273 patients who received cytostatic agents (odds ratio 0.9, 95% CI, 0.5 to 1.6, P.80). Recursive partitioning analysis confirmed the prognostic importance of histology. Histology was the first split for all five end points. Within the AA subgroup, recursive partitioning identified 119 patients with a KPS greater than 70 as having favorable long-term outcomes: median PFS time of 15 weeks compared with 9 weeks for 31 AA patients with a KPS of less than or equal to 70 (P.0090); median OS time of 59 weeks compared with 20 weeks (P.0002). No further meaningful prognostic subgroups were identified. We used a KPS of greater than 80 for our analyses because the results were similar to those obtained with using a KPS more than 70, because 85 was selected by recursive partitioning as the optimal cutoff for all five endpoints, and because a KPS greater than 80 yielded a more even split of the data, thus providing more precise effect estimates. We used an age cutoff of 40 years as a compromise between the optimal cutoffs for the five end points (which ranged from 28 to 52 years). We also chose 40 because it was close to the optimal cutoff for APF6 (41.5) and to the median age (45). DISCUSSION The entry criteria for these phase II studies are similar to those generally used for recurrent glioma. Thus, the results from our historical database should apply to patients eligible for such studies. The restrictions applied to histology, age, performance status, and (in some cases) prior chemotherapy should have some affect on the apparent relationships

2576 WONG ET AL Fig 2. PFS according to histology in 375 patients. PFS is 31%, 20%, and 10% at 26, 52, and 104 weeks, respectively, for AA patients, and 15%, 8%, and 4% at 26, 52, and 104 weeks, respectively, for GBM patients. between these factors and prognosis. Different results might be obtained if patients with additional histologies, younger ages, and lower performance status were included. In particular, the apparent effects of age and performance status would presumably be larger in a less restricted setting. Although the entry criteria for the studies included in our analysis were similar, they were not identical and this heterogeneity is a potential weakness of our study. Our results demonstrate the clear differences in outcomes between recurrent AA and recurrent GBM patients. Patients with GBM histology did consistently worse than patients with AA across all measures with respect to tumor response, progression, and survival. It seems clear that future trials should report results separately for AA and GBM. Ideally future trials would be designed with separate strata for AA and GBM. Furthermore, the relevance of the overall results of this historical database depends on future trials having roughly 60% GBM. Among prognostic factors examined in our study, histology was a highly significant prognostic variable for recurrent malignant gliomas. To a lesser extent, a KPS greater than 80 and salvage treatment were associated with poorer outcomes. A high KPS is expected to associate with a favorable outcome because it is probably a marker for overall tumor burden, cumulative treatment-related toxicity, or both. 18,19 A similar study reported more than a decade ago by Eagan and Scott 9 showed that none of their six factors examined, age, sex, tumor grade, performance score, TTP after irradiation, and prior chemotherapies, correlated with response or Table 3. Multivariate Results CR/PR CR/PR/MR/SD APF6 PFS OS Prognostic Factor OR 95% CI P OR 95% CI P OR 95% CI P HR 95% CI P HR 95% CI P Histology GBM v AA 2.4 1.1, 5.2.033 2.3 1.4, 3.6.0009 2.4 1.4, 4.1.0022 1.5 1.2, 2.0.0005 1.8 1.4, 2.4.0001 Age, 40 v 40 years 1.1 0.5, 2.5.73 0.6 0.4, 1.1.094 1.3 0.7, 2.3.37 1.0 0.8, 1.3.92 1.1 0.9, 1.5.37 KPS, 80 v 80 0.6 0.3, 1.3.24 0.9 0.6, 1.3.50 0.8 0.5, 1.3.36 0.8 0.7, 1.0.084 0.7 0.5, 0.8.0006 Salvage, yes v no 2.3 0.5, 10.28 2.9 1.3, 6.6.010 3.1 1.1, 9.3.038 1.5 1.1, 2.1.018 1.7 1.1, 2.4.0070 NOTE. Results are shown as effect measure (95% CI) P value. Effect measure is odds ratio for binary end points CR/PR, CR/PR/MR/SD, and APF6 and hazards ratio for time-to-event end points (PFS and OS). For binary end points, effects are shown for nonresponse so that the directions of the effects are consistent with those for PFS and OS (ie, progression and death).

OUTCOMES AND PREDICTORS FOR RECURRENT GLIOMA 2577 TTP. Performance score was the only variable correlated with survival. Our study had 375 patients combined from eight phase II trials, and this number would have enough statistical power to detect small differences in our prognostic variables (except perhaps for CR/PR where only 34 events were observed). This strategy of combining data from multiple small trials is similar to a meta-analysis, as in the one that established chemotherapy efficacy in newly diagnosed GBM and AA 20 even though multiple small trials had failed to do so previously. But unlike a typical metaanalysis, our trials were all performed at a single institution with similar entry criteria and methods of follow-up. This feature helps to minimize the extent of heterogeneity in our data. In addition, contrary to the study by Eagan and Scott, a large number of our patients had chemotherapies, and chemotherapy may accentuate small differences between GBM and AA. Nevertheless, the paucity of prognostic factors in recurrent malignant gliomas suggests that current treatments are still inadequate and much effort will be needed to develop better treatment strategies. Because our study included trials using cytotoxic and cytostatic agents, we performed an analysis to see whether patient outcome depends on the type of agents used. We found little difference in outcome. This result provides an important basis to combine the eight phase II trials in our study, and further suggests that recurrent malignant gliomas are relatively resistant to current second-line therapies, regardless of cytotoxic or cytostatic agents. In addition to prognostic factors, we established a set of reference outcome parameters, with respect to response, tumor progression, and survival, that can be used in future clinical trials for recurrent malignant gliomas. Although response to therapy remains a gold standard in current clinical trials for brain tumors, 21 PFS may play an increasingly important role in future studies. This is because emerging cytostatic drugs, such as biologic response modifiers 22 and antiangiogenic drugs, 23 are expected to stabilize tumor growth and would not effect tumor cell kill. As a result, a large number of patients would have SD, and cytostatic drugs would make an impact on patient outcome if their tumors remain stable for a significant period of time. Likewise, although survival is a frequently used end point in clinical trials, 24 PFS could be an alternative measure of treatment efficacy and would help validate response and survival data. APF6 is especially convenient because binary end points are easier to use for phase II trials. A major criticism for using survival as an end point is the multiplicity of treatments received by patients during the course of their illness. Separating out the effect on survival of a particular treatment in a study from other treatment effects would be difficult but not impossible. Furthermore, as evidenced by the steep slopes and long tails in the Kaplan-Meier PFS and survival curves (Fig 1), current treatments benefit only a small number of patients with recurrent malignant gliomas. ACKNOWLEDGMENT We thank Sandra Ictech, Brian Petter, and Mary Malec for help in data collection and Lissa Lawhorn for secretarial assistance. 1. Burger PC, Vogel FS, Green SB, et al: Glioblastoma multiforme and anaplastic astrocytoma, pathologic criteria and prognostic implications. Cancer 56:1106-1111, 1985 2. Rosenblum ML, Gerosa M, Dougherty DV, et al: Age-related chemosensitivity of stem cells from human malignant brain tumors. Lancet 1:885-887, 1982 3. Grant R, Liang BC, Page MA, et al: Age influences chemotherapy response in astrocytomas. Neurology 45:929-933, 1995 4. Nelson JS, Tsukada Y, Schoenfeld D, et al: Necrosis as a prognostic criterion in malignant supratentorial, astrocytic gliomas. Cancer 52:550-554, 1983 5. SC Schold, Jr, Burger PC, Mendelsohn DB, et al: Primary Tumors of the Brain and Spinal Cord. Boston, MA, Butterworth-Heinemann, 1997, pp 51-54 6. Wood JR, Green SB, Shapiro WR, et al: The prognostic importance of tumor size in malignant gliomas: A computed tomographic scan study by the Brain Tumor Cooperative Group. J Clin Oncol 6:338-343, 1988 7. Walker MD, Green SB, Byar DP, et al: Randomized comparisons of radiotherapy and nitrosoureas for the treatment of malignant glioma after surgery. N Engl J Med 303:1323-1329, 1989 REFERENCES 8. Gehan EA: The determination of the number of patients required in a preliminary and follow-up trial of a new chemotherapeutic agent. J Chronic Dis 13:346-353, 1961 9. Eagan RT, Scott M: Evaluation of prognostic factors in chemotherapy of recurrent brain tumors. J Clin Oncol 1:38-44, 1983 10. Rostomily RC, Spence AM, Duong D, et al: Multimodality management of recurrent adult malignant gliomas: Results of a phase II multiagent chemotherapy study and analysis of cytoreductive surgery. Neurosurgery 35:378-388, 1994 11. Simon R: Optimal two-stage designs for phase II clinical trials. Control Clin Trials 10:1-10, 1989 12. Levin VA, Wilson CB, Craft D, et al: Criteria for evaluating malignant brain tumor patients undergoing chemotherapy. J Neurosurg 47:329-335, 1977 13. Collett D: Modelling of Binary Data. New York, NY, Chapman and Hall, 1992 14. Cox DR, Oakes D: Analysis of Survival Data. New York, NY, Chapman and Hall, 1984 15. Kaplan EL, Meier P: Nonparametric estimation from incomplete observations. J Am Stat Assoc 53:475-481, 1958

2578 WONG ET AL 16. Clark LA, Pregibon D: Tree-based models, in Chambers JM, Hastie JJ (eds): Statistical Models in S. Belmont, CA, Wadsworth, 1992, pp 377-419 17. Therneau T, Grambsch P, Fleming T: Martingale based residuals for survival models. Biometrika 77:147-160, 1990 18. Salcman M: Resection and reoperation in neuro-oncology: Rationale and approach. Neurol Clin 3:831-842, 1985 19. Levin VA, Prados MD: Treatment of recurrent gliomas and metastatic brain tumors with a polydrug protocol designed to combat nitrosourea resistance. J Clin Oncol 10:766-771, 1992 20. Fine HA, Dear KBG, Loeffler JS, et al: Meta-analysis of radiation therapy with and without adjuvant chemotherapy for malignant gliomas in adults. Cancer 71:2585-2597, 1993 21. MacDonald DR, Cascino TL, Schold SC, et al: Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol 8:1277-1280, 1990 22. Yung WKA, Steck PA, Kelleher PJ, et al: Growth inhibitory effect of recombinant alpha and beta interferon on human glioma cells. J Neurooncol 5:323-330, 1987 23. Taki T, Ohnishi T, Arita N, et al: Anti-proliferative effects of TNP-470 on human malignant glioma in vivo: Potent inhibition of tumor angiogenesis. J Neurooncol 19:251-258, 1994 24. Bajan B, Ross G, Lim CC, et al: Survival in patients with recurrent glioma as a measure of treatment efficacy: Prognostic factors following nitrosourea chemotherapy. Eur J Cancer 12:1809-1815, 1994