GBM Fellow Talk 2009

Embed Size (px)

Citation preview

  • 7/30/2019 GBM Fellow Talk 2009

    1/69

    Glioblastoma Multiforme:The Multidisciplinary Approach

    to Treatment (2009)

    H. Ian Robins MD, PhDUniversity of Wisconsin

    School of medicine and Public HealthProfessor, Departments of Medicine,

    Human Oncology, and Neurology

  • 7/30/2019 GBM Fellow Talk 2009

    2/69

    Background

    Primary Brain Neoplasia: Incidence

    16,500 new cases predicted1

    13,000 deaths/year1

    1.4% of all cancers and 2.4% of all cancer deaths2

    21% of childhood cancers2,3

    60% are malignant glioma3

    1Greenlee RT, et al. CA Cancer J Clin. 2000;50:733.2http://www.cancer.org. Accessed May 2005.3Chamberlain MC, et al. West J Med. 1998;168:114120.

  • 7/30/2019 GBM Fellow Talk 2009

    3/69

    Background

    Primary Brain Neoplasia: Frequency*

    *Total is >100 due to rounding.

    GBM = glioblastoma multiforme.

    Levin VA, et al. In: Cancer Principles and Practice of Oncology. 1997;2022-2082.

  • 7/30/2019 GBM Fellow Talk 2009

    4/69

    Background

    Glioma: Grading

    Grade Tumor Type Glioma %

    I/II Well-differentiated(low-grade) astrocytoma

    15 to 20

    III Anaplastic astrocytoma 30 to 35

    IV Glioblastoma multiforme 40 to 50

    Chamberlain MC, et al. West J Med. 1998;168:114-120.

  • 7/30/2019 GBM Fellow Talk 2009

    5/69

    Background

    Genetic Risk Factors

    A small percentage of CNS tumors attributable to inherited cancersyndromes or neurologic disorders1,2

    von Hippel-Lindau

    Li-Fraumeni

    tuberous sclerosis neurofibromatosis

    Heritable factors may account for 5% to 12% of all CNS cancers and 2%of CNS tumors in children1,2

    Risk of brain cancer reported to be elevated in people with first-degreerelatives with Hodgkins disease or stomach, colon, or prostate cancer1

    Familial clusters may be due to genetic and/or environmental factors3,4

    1Hill DA, et al. CancerEpidemiol Biomarkers Prev. 2003;12:14431448.2Hemminki K, Li X. Cancer Epidemiol Biomarkers Prev. 2003;12:11371142.3Malmer B, et al. Int J Cancer. 2003;106:260263.4Grossman SA, et al. CancerInvest. 1999;17:299308.

  • 7/30/2019 GBM Fellow Talk 2009

    6/69

    Background

    Median Survival:

    Importance of Histologic Grading Pathologic diagnosis is crucial in determining treatment

    and prognosis1

    Tumor Type

    Median

    Survival,years

    Low-grade oligodendroglioma 4-102

    Low-grade astrocytoma 53

    Anaplastic oligodendroglioma 3-42

    Anaplastic astrocytoma 33

    Glioblastoma multiforme

  • 7/30/2019 GBM Fellow Talk 2009

    7/69

    Background

    Glioblastoma Multiforme (GBM):

    Overall Characteristics

    Grade IV malignant glioma

    Most malignant, invasive, difficult-to-treat primary brain tumor

    Frequency: most common in older adults (peak age, 55 to 65 years)

    Recurrence: rapid growth; size may double every 10 days

    Median survival: ~1 year

  • 7/30/2019 GBM Fellow Talk 2009

    8/69

    Background

    Features of Glioblastoma Multiforme

    Rapid progression

    Enhancing tumor

    Surrounding edema

    Contains tumor

  • 7/30/2019 GBM Fellow Talk 2009

    9/69

    Background

    Glioblastoma Multiforme

    Classic Pseudopallisading

  • 7/30/2019 GBM Fellow Talk 2009

    10/69

    Background

    Anaplastic Astrocytoma:Overall Characteristics

    Grade III malignant glioma

    Less aggressive than GBM; malignant but somewhat better prognosis

    Frequency: highest in young adults (30 to 40 years)

    Recurrence: often as a higher grade glioma

    Challenge: difficult to remove completely with surgery

    Median survival: approximately 3 to 4 years

    GBM = glioblastoma multiforme.

  • 7/30/2019 GBM Fellow Talk 2009

    11/69

    Background

    Anaplastic Astrocytoma

    Ill-defined Borders High Cellularity

    Nuclear Atypia

    High Proliferation Index

    Ki 67

    Diffusely Infiltrating

  • 7/30/2019 GBM Fellow Talk 2009

    12/69

    Tumor Type WHO Grade Chromosomal Abnormality

    Astrocytoma Grade II p53 gene; chromosome 22q

    Anaplastic astrocytoma Grade III Chromosome 9p, 13q, 19q

    Glioblastoma multiforme Grade IV Chromosome 10

    Louis DN, et al. Available at: http://brain.mgh.harvard.edu/MolecularGenetics.htm.

    Background

    Molecular Biology of Grade II-IVAstrocytic Tumors

  • 7/30/2019 GBM Fellow Talk 2009

    13/69

    Gliomagenesis: Mol Targets

    Glial Cell

    LGG

    AA

    AO

    GBM

    1p/19q loss

    p53 mut

    22q loss

    Rb mut

    19q loss

    p15/p16 loss

    MDM2 amp

    CDK4 amp

    Chr 10 loss

    Chr 10 loss

    EGFR amp

    PTEN mut: PI3kChr 10 loss

    Angiogenesis: VEGF, VEGFR, PDGF, FGF

  • 7/30/2019 GBM Fellow Talk 2009

    14/69

    Surgical Management

    Surgery: Objectives in Malignant Glioma

    Bulk reduction

    Resection and potential for cure

    Palliation

    Biopsy for tissue diagnosis

    Overall, the objectives of surgery in the treatment of CNSmalignancies include:

  • 7/30/2019 GBM Fellow Talk 2009

    15/69

    Surgical Management

    Therapeutic Impact of Radical Surgery inGlioblastoma Multiforme

    0

    2

    4

    6

    8

    10

    12

    14

    98%

    All patients

    (n=416)

    No prior treatment

    (n=233)

    P

  • 7/30/2019 GBM Fellow Talk 2009

    16/69

    Surgical Management

    Surgical Implantation of ChemotherapyWafers: Gliadel

    Gliadelis a trademark of Guilford Pharmaceuticals.

  • 7/30/2019 GBM Fellow Talk 2009

    17/69

    Surgical Management

    GliadelTrial Protocol

    Randomized, double-blind, placebo-controlled trial1

    240 patients with newly diagnosed malignant glioma1

    Glioblastoma multiforme: 207

    Anaplastic oligoastrocytoma: 10

    Anaplastic oligodendroglioma: 9

    Anaplastic astrocytoma: 2

    Wafers implanted at the time of surgery 120 active/120 placebo1

    Most patients received 6-8 wafers2

    Mean tumor resection was 89.9% in the Gliadel group and88.3% in the placebo group

    77.5% of Gliadel patients and 81.7% of controls received 55-60 Gy

    of irradiation beginning 3 weeks after surgery2

    14% of Gliadel patients and 10% of controls also received systemicchemotherapy during the study2

    Participants followed for up to 30 months1

    1Westphal M, et al. Neuro-oncol. 2003;5:79-88.2Gliadel product information. Baltimore, MD: Guilford Pharmaceuticals, Inc.

  • 7/30/2019 GBM Fellow Talk 2009

    18/69

    Surgical Management

    Overall Survival Intent-to-Treat Group

    Note:GBM patients (n=207) did not achieve survival

    benefit; survival at 22months ~10%; no improvement in PFS

    Westphal M, etal , Neuro-Oncol, 20035:79-88

  • 7/30/2019 GBM Fellow Talk 2009

    19/69

    Radiation Therapy

  • 7/30/2019 GBM Fellow Talk 2009

    20/69

    Radiation Therapy

    Objectives of Radiationin Malignant Glioma

    Potential for cure Prolong survival times

    Control local infiltration of cancer cells

    Palliation

    Overall, the objectives of radiation in the treatment of CNSmalignancies include:

  • 7/30/2019 GBM Fellow Talk 2009

    21/69

    Radiation Therapy

    Adverse Effects of Radiation Therapyon Brain Tissue

    Acute reaction due to radiation-induced edema; symptoms include:

    Headache

    Nausea/vomiting

    Somnolence

    Late effect: fatigue

    Levin VA, et al. In: Cancer Principles and Practice of Oncology. 1997;2022-2082.

  • 7/30/2019 GBM Fellow Talk 2009

    22/69

    Radiation Therapy

    Radiation Treatment for AnaplasticAstrocytoma and Glioblastoma Multiforme

    Efficacy confirmed in early randomized trials1-3

    Significantly improved survival over supportive care alone orwith chemotherapy

    Addition of chemotherapy did not improve survival compared withradiotherapy alone

    External beam radiotherapy

    Cornerstone of therapy4

    Fractionation improves therapeutic ratios5

    Anaplastic astrocytoma and glioblastoma treated with larger fields4

    Radiosensitizers6

    1Walker MD, et al. J Neurosurg. 1978;49:333-343.2Walker MD, et al. N Engl J Med. 1980;303:1323-1329.3Kristiansen K, et al. Cancer. 1981;47:649-652.4Soo EW, et al. Available at: http://www.cancernetwork.com. Accessed March 31, 2005.5Levin VA, Leibel SA, Gutin PH. Neoplasms of the central nervous system. In: DeVita VT, et al, eds. Cancer Principles and Practice of

    Oncology. 6th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2001:21002160.

    6Knisely JP, et al. Neuroimaging Clin N Am. 2002;12:525-536.

  • 7/30/2019 GBM Fellow Talk 2009

    23/69

    Radiation Therapy

    Stereotactic Radiosurgery

    A focal, single-fraction radiation delivery method1

    Linear accelerator

    Gamma knife

    Delivers a high dose of radiation to a small, discrete,

    well-defined target1

    Rapid dose fall-off

    External beam method to deliver brachytherapy boost

    Focal therapy for infiltrative disease

    Functional neuroimaging2

    No selection bias in clinical trials3

    1Levin VA, Leibel SA, Gutin PH. Neoplasms of the central nervous system. In: DeVita VT, et al, eds.

    Cancer Principles and Practice of Oncology. 6th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2001:21002160.2McDermott MW, et al. J Neuro-oncol. 2004;69:83-100.

    3Lustig RA, et al.Am J Clin Oncol. 2004;27:516-521.

  • 7/30/2019 GBM Fellow Talk 2009

    24/69

    Radiation Therapy

    Stereotactic Radiosurgery

  • 7/30/2019 GBM Fellow Talk 2009

    25/69

    Radiation Therapy

    RTOG 9305: Newly Diagnosed GBMStereotactic Radiosurgery Phase III Trial

    Souhami L, et al. Int J Radiat Oncol Biol Phys. 2004;60:853-860.

  • 7/30/2019 GBM Fellow Talk 2009

    26/69

    Chemotherapy

  • 7/30/2019 GBM Fellow Talk 2009

    27/69

    Conventional Chemotherapy for Malignant Gliomas

    Other alkylating agents (e.g., procarbazine, temozolomide)

    Response rate in GBM 20% - 35% in newly diagnosed 5% - 15% in recurrent cases

    Nitrosoureas (e.g., BCNU, CCNU)

    Effect on survival modest

    Salvage agents used include platinoids, CPT-11, and others

    1Rieger J, et al. Neurol Psychiatr Brain Res. 1999;7:37-46.

    2Levin VA, Leibel SA, Gutin PH. Neoplasms of the central nervous system.

    In: DeVita VT, et al, eds. Cancer Principles and Practice of Oncology. 6th ed.

    Philadelphia, PA: Lippincott Williams and Wilkins

  • 7/30/2019 GBM Fellow Talk 2009

    28/69

    Limitations of Chemotherapy:Perfusion

    Rieger J, et al. Neurol Psychiatr Brain Res. 1999;7:37-46.

  • 7/30/2019 GBM Fellow Talk 2009

    29/69

    Chemotherapy

    Limitations of Chemotherapy in TreatingBrain Tumors: Drug Interactions

    Enzyme inducers1

    Anticonvulsants (paclitaxel and CPT-11 clearance)1

    Carbamazepine

    Oxycarbazepine

    Phenobarbital

    Phenytoin

    Enzyme inhibitors1

    Valproic acid

    Cytoprotective effect Corticosteroids2

    1Vecht CJ, et al. Semin Oncol. 2003;30:49-52.

    2Weller M, et al. Neurology. 1997;48:1704-1709.

  • 7/30/2019 GBM Fellow Talk 2009

    30/69

    Chemotherapy

    Meta-analysis of Chemotherapy inHigh-Grade Glioma

    Stewart LA. Lancet. 2002;359:10111018.

  • 7/30/2019 GBM Fellow Talk 2009

    31/69

    0

    5

    10

    15

    20

    25

    30

    35

    40

    45

    50

    1-year survival 2-year survival

    %o

    fpatients

    No chemotherapy

    Chemotherapy

    Chemotherapy

    Meta-analysis of Chemotherapy inHigh-Grade Glioma

    Stewart LA. Lancet. 2002;359:10111018.

  • 7/30/2019 GBM Fellow Talk 2009

    32/69

    Chemotherapy

    Limited Role for Chemotherapyin the Treatment of GBM

    Randomized trials

    with adjuvant PCV

    chemotherapyhave failed to

    demonstrate any

    survival benefit

    Medical Research Council Brain Tumor Working Party. J Clin Oncol. 2001;19:509-518.

  • 7/30/2019 GBM Fellow Talk 2009

    33/69

    Temozolomide:

    Mechanism of Action

    and Pharmacokinetics

  • 7/30/2019 GBM Fellow Talk 2009

    34/69

    Temozolomide

    Temozolomide: Second-GenerationAlkylating Agent

    TMZ spontaneously converts to MTIC at physiologic pH: no hepaticor renal metabolism required: therefore, drug levels not altered byanticonvulsant use

    NN

    O

    NN

    CNH2

    NO CH3

    pH > 7.0

    Spontaneous

    hydrolysis NN

    O

    NN

    CNH2

    N

    HCH3

    O

    NN

    CNH2

    NH2

    Temozolomide MTIC AIC Methyldiazoniumion

    + N NCH3

    MTIC, 5-(3-methyltriazen-1-yl) imidazole-4-carboxamide.

    Denny BJ, et al. Biochemistry. 1994;33:90459051.

  • 7/30/2019 GBM Fellow Talk 2009

    35/69

    *High cytotoxicity.

    Denny BJ, et al. Biochemistry. 1994;33:9045-9051.

    N3 adenine

    9%

    Other sites

    16%

    O6

    guanine5%*

    N7 guanine

    70%

    Position/base total adducts (%)

    Temozolomide

    Temozolomide Exerts Cytotoxic Effectsvia Methylation of DNA

  • 7/30/2019 GBM Fellow Talk 2009

    36/69

    Temozolomide

    Temozolomide PharmacokineticCharacteristics

    Key characteristics of temozolomide include1,2: Excellent bioavailability

    Rapid, complete absorption Peak plasma concentrations in 1 h

    Rapid elimination: mean t1/2 = 1.8 h

    Newlands ES, et al. Br J Cancer. 1992;65:287-291.

    TEMODAR Prescribing Information.

  • 7/30/2019 GBM Fellow Talk 2009

    37/69

    Temozolomide

    Temozolomide Drug Interactions

    In a multiple-dose study, administration of temozolomide withranitidine did not change the Cmax or AUC values for temozolomideor MTIC

    Coadministration of valproic acid decreases the clearance oftemozolomide by 5%

    Coadministration of the following drugs had no influence ontemozolomide clearance: Dexamethasone

    Carbamazepine

    Prochlorperazine

    Ondansetron

    Phenytoin H2-receptor antagonists

    Phenobarbital

  • 7/30/2019 GBM Fellow Talk 2009

    38/69

    Temozolomide

    Primary Mechanisms of Resistance

    High levels of O6-alkylguanine DNA alkyltransferase (AGT)

    Applies to methylating and chloroethylating agents

    A Principal cytotoxic mechanism is DNA damage caused by failureof DNA mismatch repair (MMR): cells with deficiency in MMR do not

    recognize O6

    -MG adducts allowing replication without apoptosis.Applies to methylating agents only

    Poly(ADP- ribose) polymerase (PARP)

    Friedman HS, et al. Clin Cancer Res. 2000;6:2585-2597.

  • 7/30/2019 GBM Fellow Talk 2009

    39/69

    Poly(ADP-ribose) polymerase

    Can we exploit the other DNA alkylation events associated withtemozolomide?

    Only about 7% of the alkylation caused by temozolomide involves the O-6position of Guanine

    Far more common is alkylation of the N-7 position of Guanine and the N-

    3 position of Adenine

    These are repaired by base exicision pathway enzymes,which can beinhibited by PARP inhibitors

  • 7/30/2019 GBM Fellow Talk 2009

    40/69

    Temozolomide in Newly DiagnosedGlioblastoma Multiforme

  • 7/30/2019 GBM Fellow Talk 2009

    41/69

    Temo zolom ide in GBM

    Phase II Study in Newly DiagnosedGlioblastoma Multiforme

    Study design and objectives

    Two center, open-label pilot study

    Evaluate safety, tolerability, and overall survival

    Inclusion criteria

    Adults with newly diagnosed GBM ECOG performance status 2

    Adequate hematologic, renal, and hepatic function

    Stupp R, et al. J Clin Oncol. 2002;20:1375-1382.

  • 7/30/2019 GBM Fellow Talk 2009

    42/69

    Patients (n=64)

    Median age = 52

    Performance status 0 or 1 in 86%

    Complete resection in 42%

    Treatment Temozolomide discontinued in 4 patients in concomitant phase due to infection

    or thrombocytopenia

    Maintenance therapy administered in 49 patients for median 5.5 cycles

    Thirty-nine percent received all planned temozolomide

    Temo zolom ide in GBM

    Phase II Study: Patients andTreatment Delivery

  • 7/30/2019 GBM Fellow Talk 2009

    43/69

    Hematologic toxicity

    Concomitant phase: Grade 3/4 neutropenia and thrombocytopenia in 6% ofpatients each

    Maintenance phase: Grade 3/4 neutropenia or thrombocytopenia in 2% and 6%of cycles, respectively

    Pneumocystis cariniipneumonia in 2 patients

    Non-hematologic toxicity

    Most mild-to-moderate severity

    Nausea, vomiting, and fatigue most common

    Temo zolom ide in GBM

    Phase II Study: Adverse Events

    Stupp R, et al. J Clin Oncol. 2002;20:1375-1382.

  • 7/30/2019 GBM Fellow Talk 2009

    44/69

    Temo zolom ide in GBM

    Phase II Study: Adverse Events

    Hematologic toxicity

    Concomitant phase: Grade 3/4 neutropenia and thrombocytopenia in 6% ofpatients each

    Maintenance phase: Grade 3/4 neutropenia or thrombocytopenia in 2% and 6%of cycles, respectively

    Pneumocystis cariniipneumonia in 2 patients*

    Non-hematologic toxicity

    Most mild-to-moderate severity

    Nausea, vomiting, and fatigue most common

    Stupp R, et al. J Clin Oncol. 2002;20:1375-1382.

  • 7/30/2019 GBM Fellow Talk 2009

    45/69

    Temo zolom ide in GBM

    Phase II Study: Survival

    Stupp R, et al. J Clin Oncol. 2002;20:1375-1382.

    Median survival = 16 months

    n=64

  • 7/30/2019 GBM Fellow Talk 2009

    46/69

    Temo zolom ide in GBM

    Radiotherapy plus Concomitant andAdjuvant Temozolomide for Glioblastoma

    R Stupp, WP Mason, MJ van den Bent, et al.

    N Engl J Med. 2005;352:987-996.

  • 7/30/2019 GBM Fellow Talk 2009

    47/69

    Temo zolom ide in GBM

    Phase III EORTC/NCIC Trial:Study Design

    Phase III, randomized, open-label, multicenter study

    Patients randomly assigned to receive either radiotherapy (RT) 5days/week for 6 weeks or radiotherapy plus concomitant temozolomide(TMZ) followed by maintenance TMZ for 6 cycles

    StuppR, et al. N Engl J Med. 2005;352:987996.

  • 7/30/2019 GBM Fellow Talk 2009

    48/69

    Temo zolom ide in GBM

    Inclusion Criteria

    Newly diagnosed, histologically proven GBM

    Age 18 70 years

    WHO performance status 2

    6 weeks since biopsy or resection

    Adequate bone marrow, hepatic, and renal function

    No other severe underlying disease

    Stratification factors:

    Age

    Resection vs. biopsy Performance status 0 to 1 vs. 2

    Institution

  • 7/30/2019 GBM Fellow Talk 2009

    49/69

    Temozolomide 75 mg/m2 po qd for 6 weeks,

    then 150200 mg/m2 po qd d15 every 28 days for 6 cycles

    Focal RT daily 30 x 200 cGy

    Total dose 60 Gy

    Concomitant

    TMZ/RT*Adjuvant TMZ

    Weeks6 10 14 18 22 26 30

    RT Alone

    R

    EORTC 26981-22981 and NCIC CE.3: Schema

    0

    *PCP prophylaxis was required for patients receiving TMZ during the concomitant phase.

    Stupp, NEJM,2005

  • 7/30/2019 GBM Fellow Talk 2009

    50/69

    Temo zolom ide in GBM

    Patient Characteristics

    Characteristic Radiotherapy

    n=286

    TEMODAR plus Radiotherapyn=287

    Age, yrsMedianRange

    5723-71

    5619-70

    WHO performance status, n (%)012

    110 (38)141 (49)35 (12)

    113 (39)136 (47)38 (13)

    Sex, n (%)MaleFemale

    175 (61)11 (39)

    185 (64)102 (36)

    Extent of surgery, n (%)

    BiopsyDebulking 45 (16)241 (84) 48 (17)239 (83)

    Corticosteroid therapy, n (%)YesNoMissing data

    215 (75)70 (24)12 (4)

    193 (67)94 (33)10 (3)

  • 7/30/2019 GBM Fellow Talk 2009

    51/69

    Treatment delivered, n (%) Radiotherapyn=286

    Radiotherapy plustemozolomide

    n=287

    Radiotherapy

    Received >90% of doseEarly discontinuation

    264 (92%)19 (7%)

    273 (95%)14 (5%)

    Completed both radiotherapy andtemozolomide

    N/A 243 (85%)

    Discontinued temozolomide due totoxicity

    N/A 14 (5%)

    Temo zolom ide in GBM

    Treatment Delivery: Concomitant Phase

  • 7/30/2019 GBM Fellow Talk 2009

    52/69

    Temo zolom ide in GBM

    Treatment Delivery: Maintenance Phase

    Treatment delivered Patients

    n=287

    Started maintenance therapy, n (%) 223 (78%)

    Cycles completed, median (range) 3 (0-7)

    Completed 6 cycles, n (%) 105 (47%)

    Dose escalated at cycle 2, n (%) 149 (67%)

    Reasons for discontinuation, n (%)Disease progressionToxicityOther

    86 (39%)17 (8%)15 (7%)

  • 7/30/2019 GBM Fellow Talk 2009

    53/69

    Temo zolom ide in GBM

    Treatment Delivery: Salvage Therapy

    Radiotherapy

    n=286

    Radiotherapy plusTemozolomide

    n=287

    Progressive disease, n (%) 268 (94) 244 (85)

    Second surgery, % 23 23

    Chemotherapy, % 72 58

    Temozolomide, % 60 25

    Stupp R, et al. N Engl J Med. 2005;352:987-996.

  • 7/30/2019 GBM Fellow Talk 2009

    54/69

    EORTC Phase III Trial Overall Survival

    months

    0 6 12 18 24 30 36 420

    1020

    30

    40

    50

    6070

    80

    90

    100RT TMZ/RT

    Median OS, mo: 12.1 14.6 p

  • 7/30/2019 GBM Fellow Talk 2009

    55/69

    Temozolom ide Safety

    Adverse Events in Newly DiagnosedGlioblastoma Multiforme

    ConcomitantPhase

    Radiotherapy +Temozolomide

    (n=288)

    MaintenancePhase

    Temozolomide(n=224)

    Adverse Event Number (%) of patients

    All Grade >3 All Grade >3

    Alopecia 199 (69) 0 124 (55) 0

    Fatigue 156 (54) 19 (7) 137 (61) 20 (9)

    Nausea 105 (36) 2 (1) 110 (49) 3 (1)

    Vomiting 57 (20) 1 (

  • 7/30/2019 GBM Fellow Talk 2009

    56/69

    Conclusions

    Temozolomide given concomitantly with radiotherapy and asmaintenance therapy significantly improves survival in patients withnewly diagnosed glioblastoma multiforme compared with radiotherapyalone.

    Temozolomide provides a measurable response in patients withtreatment-tolerability profile.

    Non-cumulative myelosuppression is the dose-limiting adverse effect.

  • 7/30/2019 GBM Fellow Talk 2009

    57/69

    Phase II German Trial

    Used 50 mg/m2 TMZ during XRT Used no post-XRT TMZ

    Results:

    53 pts with GBM; 60 Gy/6 wks

    Resection: complete 14, subtotal 22, Bx 17

    Median PFS: 8 mos; Median survival: 19 mos

    1-yr survival: 72%; 2-yr survival: 29%

    No adjuvant TMZ; used lower daily dose

    Combs, ASCO 2004.

  • 7/30/2019 GBM Fellow Talk 2009

    58/69

    Wedge SR, et al. Ant icancer Drugs1997;8:92-97.

    TMZ & XRT: Increased Inhibition of Cell Growth*

    * In-vitro studies of U373MG

    glioblastoma cell line

    Radiation (Gray)

    35

    30

    25

    20

    15

    10

    5

    00 1 2

    InhibitionofCellG

    rowth(%) RT only

    RT + 5 M TMZ

    RT + 10 M TMZ

  • 7/30/2019 GBM Fellow Talk 2009

    59/69

    Overall & Progression-Free Survival by

    Treatment

    Variable RT (n=286) RT+TMZ (n=287)

    Value (95% Confidence Interval)

    Median Overall Survival (mos) 12.1 (11.2-13.0) 14.6 (13.2-16.8)

    Overall Survival (%)

    at 12 months 50.6 (44.7-56.4) 61.1 (55.4-66.7)

    at 24 months 10.4 (6.8-14.1) 26.5 (21.2-31.7)

    Median PFS (mos) 5.0 (4.2-5.5) 6.9 (5.8-8.2)

    Progression-Free Survival (%)

    at 12 months 9.1 (5.8-12.4) 26.9 (21.8-32.1)

    at 24 months 1.5 (0.1-3.0) 10.7 (7.0-14.3)

    Adapted from Stupp R, et al. N Engl J Med. 352:10:987-996.

    Overall Survival by MGMTPromoter

  • 7/30/2019 GBM Fellow Talk 2009

    60/69

    yMethylation Status

    Hegi ME, et al. N Engl J Med. 352:10:997-1003.

  • 7/30/2019 GBM Fellow Talk 2009

    61/69

    Survival with Methylated MGMTPromoter(44.7% had methylated MGMT)

    Promoter Status & Outcome RT (n=100) RT+TMZ (n=106)

    Number of Patients 46 46

    Progression-Free Survival

    Median (months) 5.9 (5.3-7.7) 10.3 (6.5-14.0)

    at 6 months 47.8 (33.4-62.3) 68.9 (55.4-82.4)

    Hazard Ratio for Death 1.00 0.48 (0.31-0.75)

    Overall Survival

    Median (months) 15.3 (13.0-20.9) 21.7 (17.4-30.4)

    at 2 years (%) 22.7 (10.3-35.1) 46.0 (31.2-60.8)

    Hazard Ratio for Death 1.00 0.51 (0.31-0.84)

    Adapted from Hegi ME, et al. N Engl J Med. 352:10:997-1003.

    Note: Numbers in parentheses are 95% con f idence intervals.

  • 7/30/2019 GBM Fellow Talk 2009

    62/69

    Survival with Unmethylated MGMTPromoter

    Promoter Status & Outcome RT (n=100) RT+TMZ (n=106)

    Number of Patients 54 60

    Progression-Free Survival

    Median (months) 4.4 (3.1-6.0) 5.3 (5.0-7.6)

    at 6 months 35.2 (22.5-47.9) 40.0 (27.6-52.4)

    Hazard Ratio for Death 1.00 0.62 (0.42-0.92)

    Overall Survival

    Median (months) 11.8 (9.7-14.1) 12.7 (11.6-14.4)

    at 2 years (%)

  • 7/30/2019 GBM Fellow Talk 2009

    63/69

    Months0 4 8 12 16 20 24 28 32 36 40

    0

    10

    20

    30

    40

    50

    60

    70

    80

    90

    100

    O N Patients at risk, n

    54 54 28 9 0 0 0 0 0 0 0

    53 60 44 18 8 8 8 7 5 3 1

    45 46 33 15 7 3 2 1 0 0 0

    40 46 35 28 18 14 10 6 3 1 0

    Unmeth, RTaloneUnmeth, TMZ/ RTMeth, RT aloneMeth, TMZ/ RT

    Overall Wald test: P< .0001 (df = 3)

    Progression-free

    survival

    Meth, TMZ/RT

    Meth, RT

    Unmeth,TMZ/RTUnmethRT

    Progression-Free Survival by MGMT and +/- TMZ

    Survival by EGFR and MGMT Status

  • 7/30/2019 GBM Fellow Talk 2009

    64/69

    Survival by EGFRand MGMTStatus

    Months

    Survi

    val

    Low EGFR,Meth

    Low EGFR, Unmeth

    High EGFR,

    Unmeth

    High EGFR,Meth

    EGFRexpression data based on GeneChip U133 Plus 2.0

    Wald test, p= 0.0031

    Hegi, SNO 2004

    MGMT I hibiti /D L l

  • 7/30/2019 GBM Fellow Talk 2009

    65/69

    -100

    -80

    -60

    -40

    -20

    0

    0 7 15 22

    Days

    75mg/m2 100 125 150 175

    %C

    hange

    50-150 mg/m2/d 21 d with 1 wk rest

    Tolcher AW, et al. Br J Cancer, 2003;88:10041011.

    MGMT Inhibition/Dose Level

  • 7/30/2019 GBM Fellow Talk 2009

    66/69

    TMZ Inactivates DNA Repair

    Schedule A

    50 - 175 mg/m2/day x 7 days repeated q 2weeks

    Schedule B

    50 - 150 mg/m2/day x 21 days repeated q 4weeks

    Serial serum samples collected for 52 of the 72 patients

    Inactivation of AGAT noted after 7 days of TMZ treatment

    (P

  • 7/30/2019 GBM Fellow Talk 2009

    67/69

    R

    E

    G

    I

    ST

    E

    R

    RT ( 60 Gy) plus

    TMZ 75 mg/m2

    /d

    R

    A

    N

    D

    O

    M

    I

    Z

    E*

    Arm 1: Standard

    TMZ days 1 - 5 q 28days

    Maximum 12 cycles

    Arm 2: Experimental

    TMZ days 1 - 21 q 28days

    Maximum 12 cycles

    RTOG/EORTC 0525 Phase III GBM Trial

    *Strat ify b y RTOG RPA class and AGAT statu s

    N = 834 to detect 25% relative improvement in median survival

    corresponding to hazard ratio = 0.80.

    PIs: Gilbert and Mehta

    BackgroundMellinghoff et al 11/05

  • 7/30/2019 GBM Fellow Talk 2009

    68/69

    Grade IV astrocytomas express EGFRvIII

    which is constituatively active, strongly

    activating PI3K

    PTEN inhibits this pathway and is commonly

    lost in these tumors

    TK inhibitors target the cytoplasmic domain

    for blockage-depriving cell from proliferation

    (ras) and apoptosis (Akt)

    Mellinghoff et al 11/05

    KrishnanetalFrontiersinBioscience1/03

  • 7/30/2019 GBM Fellow Talk 2009

    69/69

    Before I came here I was confused

    about this subject.

    Now having listened to your lecture, Iam still confused, but at a higher

    level.

    Enrico Fermi, 1938 Nobel Laureate in

    Physics