58
Glioblastoma Newer Advances Dr. Vibhay Pareek

Glioblastoma vibhay

Embed Size (px)

Citation preview

Page 1: Glioblastoma vibhay

GlioblastomaNewer Advances

Dr. Vibhay Pareek

Page 2: Glioblastoma vibhay

Introduction to Glioblastoma

Page 3: Glioblastoma vibhay

3

CNS and the Blood-Brain Barrier• CNS includes the brain and spinal cord[1]

– Brain is major control network for every physiological, motor, and thought process[4]

• CNS cell types include[2] – Neurons: Structural and

functional cells of the CNS– Glial cells: Perform neuronal

support and immune functions

Blood-brain barrier

Blood-brain barrier (BBB) protects the brain and maintains

a stable brain environment[3]

Astrocyte Oligodendrocytes

Microglial cell

Capillary

Neurons

CNS, central nervous system.

1. SEER Training Modules- Anatomy and Function Areas of the Brain and CNS. Available at: http://training.seer.cancer.gov/brain/tumors/anatomy/. Accessed December 17, 2015.

2. SEER Training Modules- Neurons and Glial Cells. Available at: http://training.seer.cancer.gov/brain/tumors/anatomy/neurons.html. Accessed December 17, 2015.

3. Yilmaz A et al. J Antimicrob Chemother. 2011. doi:10.1093/jac/dkr492.4. Krucik G. Healthline: Brain. Available at

http://www.healthline.com/human-body-maps/brain#seoBlock. Accessed December 17, 2015.

5. Allen NJ. Nature. 2009;457(7230):675-677.

Adapted from Allen NJ 2009.[5]

Page 4: Glioblastoma vibhay

4

Primary CNS Tumors• CNS tumors arise from CNS cells and are categorized according to the cell

type/tissue from which they originate[1]

• Gliomas arise from glial cells and neuronal precursors, and constitute 80% of all malignant primary brain and CNS tumors[2]

CNS, central nervous system; NOS, Not Otherwise Specificed1. DeAngelis LM. N Engl J Med. 2001;344(2):114-123.2. Ostrom QT et al. Neuro Oncol. 2013;15(Suppl 2):ii1-ii56.

3. Brain Tumor Information. Available at http://www.braintumor.org/brain-tumor-information/. Accessed December 17, 2015.

Brain metastases are more common than primary brain tumors[3]

Benign (63%)[2,3] Malignant (37%)[3]

Primary CNS Tumors (28%–40%)[3]

Other (20%)[2]

Glioblastoma (54%)

Meningioma

Tumors of the Pituitary

Vestibular Schwannoma

Hernangioma

Craniopharyngioma

Ependymal Tumors

Lymphoma

Embryonal Tumors

Meningioma

Germ Cell Tumors

Diffuse Astrocytoma

NOS Malignant Glioma

Ependymal Tumors

Oligodendroglioma

Anaplastic Astrocytoma

Pilocytic Astrocytoma

Oligoastrocytic Tumors

Gliomas (80%)[2]

Brain Mets (60%–72%)[3]

Page 5: Glioblastoma vibhay

5

2. DeAngelis LM. N Engl J Med. 2001;344(2):114-123.3. Louis DN et al. Acta Neuropathol. 2007;114(2):97-109.4. Burkhard C et al. J Neurosurg. 2003;98(6):1170-1174.5. NCCN Guidelines®. Central Nervous System Cancers. V1.2015. 6. Kleihues P, Ohgaki H. Neuro Oncol. 1999;1(1):44-51.

• Brain tumors are typically graded according to cellular origin and aggressiveness [1]

• WHO classification combines tumor type with degree of malignancy[1-3]

World Health Organization (WHO) Grades of CNS Tumors

• Low proliferative potential• Potentially curable with surgical resection aloneGrade I[3]

• Infiltrative properties• Tendency to recur and progress to malignancy

despite low-level proliferationGrade II[3]

Includes malignant astrocytomas• Histological evidence of malignancy• Often recur as higher grade tumors

Grade III[3,5,6]

Includes glioblastoma and variants*• Cytologically malignant• Rapid pre- and postoperative disease evolution

Grade IV[3]Hig

h-gr

ade

L

ow-g

rade >10[4]

>5[3]

3[3]

1[1]

mOS (yrs)

* Gliosarcoma, giant cell glioblastoma, and small cell glioblastoma.[1]

CNS, central nervous system; mOS, median Overall Survival1. Wen PY, Kesari S. N Engl J Med. 2008;359(5):492-507.

Page 6: Glioblastoma vibhay

6

Incidence and Mortality of Brain and Other Nervous System Cancer

2012 Worldwide Brain and CNS Cancer Statistics*Global[1] EU[1] Asia[1] US FR DE JP

New cases 256,213 43,136 131,452 22,850†[2] 4,770[4] 7,120[4] 5,700†[5]

Glioblastoma NA NA NA 11,140[3] 2,200[3] 3,740[3] 2,700[3]

Incidence rate‡ 3.4 6.9 3.0 5.3[1] 5.1[1] 5.3[1] 2.8[1]

Deaths 189,382 32,960 95,732 15,320†[2] 3,290[4] 5,660[4] 2,100†[5]

Mortality rate‡ 2.5 4.9 2.2 3.3[1] 3.2[1] 3.5[1] 1.0[1]

• CNS cancers are the 13th most commonly diagnosed cancer worldwide[1]

– Glioblastoma accounts for 54% of new glioma and 45% of primary malignant tumors[6]

* Estimated 2012 unless otherwise noted.† Estimated 2015.‡ Estimated ASRs (W) per 100,000. Both sexes, all ages.

3. Decision Resources: Glioblastoma Multiforme. September 2013.4. Ferlay J et al. Eur J Cancer. 2013;49(6):1374-1403. 5. National Cancer Center. Available at:

http://ganjoho.jp/en/public/statistics/short_pred.html. Accessed December 21, 2015. 6. Ostrom QT et al. Neuro Oncol. 2013;15:ii1-ii56.

ASR, age standardized rate; CNS, central nervous system; W, world.

1. GLOBOCAN 2012: Population Fact Sheets. Available at: http://globocan.iarc.fr/Pages/fact_sheets_population.aspx. Accessed December 21, 2015.

2. American Cancer Society. Cancer Facts and Figures 2015. Available at: http://www.cancer.org/acs/groups/content/ @research/documents/webcontent/acspc-044552.pdf. Accessed December 21, 2015.

Page 7: Glioblastoma vibhay

7

2012

2013

2014

2015

2016

2017

2018

2019

2020

2021

2022

02,0004,0006,0008,000

10,00012,00014,00016,000

USFranceGermanyJapan

Regional Incidence Trends for Glioblastoma• Number of newly diagnosed cases of glioblastoma is expected to

increase in the US, France, Germany, and Japan[1]

Projections for 2012–2022

2.01.31.01.0

Growth (%/yr)

1. Decision Resources: Glioblastoma Multiforme. September 2013.

Page 8: Glioblastoma vibhay

8

Glioblastoma 5-Year Survival Rates by Region

4.7%

US[1]

2.7%

Europe[2]

6.3%

Japan[3]

• Grade IV glioblastoma has the poorest prognosis of all primary brain tumors[4]

– Overall 5-year survival worldwide <3%[4]

1. Ostrom QT et al. Neuro Oncol. 2013;15:ii1-ii56.2. Sant M et al. Int J Cancer. 2012;131(1):173-185. 3. Nomura K et al. Int J Clin Oncol. 2000;5(6):355-360.4. Roche Glioblastoma Backgrounder. Available at:

http://www.roche.com/backgrounder_glioblastoma__concise_guide.pdf. Accessed December 17, 2015.

Page 9: Glioblastoma vibhay

9

• Etiology of brain tumors is not well understood[1]

– Ionizing radiation is the only established environmental risk factor[1,2]

Risk Factors for Glioblastoma

Non-ionizing radiation

Ionizing radiation

Genetically inherited

syndromes

Family history

Male gender

Urban residence

Age

Caucasian origin

Glioblastoma Risk

Factors[1-3]

1. Grossman SA et al. Cancer Invest. 1999;17(5):299-308. 2. Neglia JP et al. J Natl Cancer Inst. 2006;98(21):1528-1537.3. Deorah S et al. Neurosurg Focus 2006;20(4):E1.

Page 10: Glioblastoma vibhay

10

Prognostic Factors for Glioblastoma

Younger ageSingle most

powerful predictor of outcome[1]

Methylated MGMT

status[1,2]

Higher KPS score[1]

Tumor resectability (size, location, and number)[3,4]

Factors associated with better prognosis

1. Hegi ME et al. N Engl J Med. 2005;352:997-1003.2. Arvold ND et al. Clin Interv Aging. 2014;9:357-367.3. Kawano H et al. Br J Neurosurg. 2014;14:1-7. 4. NCCN Guidelines®. Central Nervous System Cancers. V1.2015.

KPS, Karnosfsky performance status ; MGMT, O6-methylguanine DNA methyltransferase.

Page 11: Glioblastoma vibhay

11

Prognostic Factors for Glioblastoma: Age

Survival Rates by Age Group[2]

Age Group (yrs)

1-Year Survival, %

5-Year Survival, %

0-19 57.2 19.220-44 66.5 16.945-54 52.7 5.955-64 40.7 3.865-74 23.7 1.775+ 9.2 0.8

Glioblastoma incidence

0

20–44

10

20

30

40

45–54 55–64 65–74 75+Age Groups

Age

-Adj

uste

d In

cide

nce

Rat

e pe

r 100

,000

Incidence Rate by Age Group[2]

• Elderly* patients represent ~50% of newly diagnosed glioblastoma[1]

– Virtually all elderly glioblastoma tumors are primary and characterized by genetic differences[1]

• Glioblastoma incidence: increases with age[1,2]

• Glioblastoma survival rates: decrease with age[1,2]

1. Arvold ND et al. Clin Interv Aging. 2014;9:357-367.2. Ostrom QT. Neuro Oncology. 2013;15(Suppl 2):ii1-ii56.* Definition of “elderly” varies, with most randomized trials including

patients aged 60, 65, or 70 years and older.[1]

Page 12: Glioblastoma vibhay

12

• Potential of prognostic biomarkers in identifying specific patient populations has not yet been fully realized[6]

• MGMT methylation status is the only biomarker with predictive implications on treatment outcomes identified to date[6]

Select Biomarkers in Glioblastoma

Biomarker Prognostic Indication Favorable Poor

MGMT methylation[1]

• Methylated in 30%–60% of cases• Methylated MGMT increases response to chemotherapy• Unmethylated MGMT decreases response to

chemotherapy

IDH1/2 mutations[2,3]• More common in lower grade glial tumors• IDH1/2 mutation occurs in approximately 3.7% of primary

GBMs versus 73.3% in secondary GBM

EGFR amplification[4,5] Observed in ~50% of primary glioblastomas

EGFRvIII mutation[4]• EGFR-amplified cells often contain EGFRvIII mutation,

which confers constitutive activity• 30% glioblastoma tumors express EGFRvIII

Prognostic Association

1. Preusser M et al. Ann Neurol. 2011;70(1):9-21. 2. Nobusawa S et al. Clin Cancer Res. 2009;15(19):6002-6007.3. Yan H et al. N Engl J Med. 2009;360(8):765-773.4. Johnson H et al. Mol Cell Proteomics. 2012;11(12):1724-1740.5. Stupp R et al. Ann Oncol. 2014;25(Suppl 3):iii93-iii101.6. McNamara MG et al. Cancers. 2013;5(3):1103-1119.

EGFR, epidermal growth factor receptor; IDH1/2, isocitrate dehydrogenase 1/2; MGMT, O6-methylguanine DNA methyltransferase.

Page 13: Glioblastoma vibhay

13

• Glioblastomas are highly heterogeneous at the molecular level[1]

• Four transcriptional subclasses have recently been defined based on tumor expression profiles[2]

Toward a Molecular Signature: The Cancer Genome Atlas Histological Subtypes of Glioblastoma

Classical

EGFR amplification/mutations

Significantly decreased mortality

Mesenchymal

High expression of tumor necrosis factor pathway

genes

Significantly decreased mortality

Neural

Neuronal marker expression

Efficacy suggested

Proneural

IDH1 mutations

No difference in survival

Transcriptional profile includes:[2]

Response to intensive treatment*:[2]

• Classifying glioblastoma tumors according to expression profiles may help predict response to certain treatments and assess patient prognosis[2]

1. Olar A, Aldape KD. J Pathol. 2014;232(2):165-177.2. Verhaak RG et al. Cancer Cell. 2010;17(1):98-110.

* Concurrent chemotherapy and radiation or more than four cycles of chemotherapy.EGFR, epidermal growth factor receptor; IDH1, isocitrate dehydrogenase.

Page 14: Glioblastoma vibhay

14

Summary of Treatment Approaches and Unmet Needs

NATURE AND PREVALENCE OF

TUMORS

• Glioblastomas are the most common and most aggressive primary brain cancer[1]

• Lower-grade gliomas (WHO grade II and III gliomas) eventually progress into grade IV within 5–10 years[2]

BLOOD-BRAIN BARRIER

• Physical, metabolic, and immunological privilege status of the brain limits therapeutic potential[3]

BIOMARKERS• Potential of prognostic/predictive biomarkers in

identifying specific patient populations has not been fully realized[4]

1. Olar A, Aldape KD. J Pathol. 2014;232(2):165-177.2. Zong H et al. Expert Rev Mol Diagn. 2012;12(4): 383-394.3. Wei X et al. Acta Pharmaceutica Sinica B. 2014;4(3):193-201.4. McNamara MG et al. Cancers. 2013;5(3):1103-1119. WHO, World Health Organization

Page 15: Glioblastoma vibhay

Treatment Guidelinesin Glioblastoma

Page 16: Glioblastoma vibhay

16

• Time between onset of symptoms and diagnosis of glioblastoma is often short[1]

– Headaches, memory loss, and motor weakness are among the most common presenting symptoms[1]

• Early diagnosis and treatment do not improve disease outcomes[2]

Glioblastoma Presentation

Glioblastoma presenting symptoms

General[1]

(Related to increased ICP)• Headaches• Slowed cognitive function• Seizures• Nausea/vomiting• Personality changes

Focal[2]

(Related to tumor location)• Aphasia• Hemiparesis• Sensory/visual loss

1. Chang SM. JAMA. 2005;293(5):557-564.2. Omuro A, DeAngelis LM. JAMA. 2013;310(17):1842-1850. ICP, intracranial pressure.

Page 17: Glioblastoma vibhay

17

• MRI: Preferred imaging modality for high-grade glioma diagnosis and treatment-planning[3]

– BBB disruption results in enhancement on contrast MRI[4]

• Challenging to distinguish between grade III and IV glioma by MRI

• No lab studies can currently suggest or confirm diagnosis of glioblastoma[2]

– Tissue diagnosis is mandatory[6]

Glioblastoma Workup and Diagnosis

1. Uddin ABMS. Medscape, Neurologic manifestations of glioblastoma multiforme workup. Available at: http://emedicine.medscape.com/article/1156220-workup#showall. Accessed December 17, 2015.

2. Bruce JN. Medscape. Glioblastoma multiforme workup. Available at: http://emedicine.medscape.com/article/283252-workup#showall. Accessed December 17, 2015.

3. Omuro A, DeAngelis LM. JAMA. 2013;310(17):1842-1850. 4. DiStefano AL et al. Biomed Res Int. 2014;2014:154350.5. Pope WB, Hessel C. AJNR Am J Neuroradiol. 2011;32(5)794-797.6. Stupp R et al. Ann Oncol. 2014;25(Suppl 3):iii93-iii101.

T1-weighted MRI*Contrast-enhanced[1]

Irregular margins may make defining

exact tumor size challenging[2]

T2-weighted/ FLAIR MRI*†

Not contrast-enhanced[1,2]

May result in improved definition of tumor volume[5]

* MRI images of same glioblastoma tumor.[1]

† Image shows T2 MRI.BBB, blood-brain barrier; FLAIR, fluid-attenuated inversion recovery; MRI, magnetic resonance imaging.

Page 18: Glioblastoma vibhay

18

• There are no curative therapies for glioblastoma[1]

– Glioblastoma recurrence rate is nearly 100%• Treatment goals are focused on preserving PS/QOL, neurological function, and

extending survival[2]

– NCCN recommends glioblastoma patients receive palliative care from diagnosis[3]

Goals of Therapy for Glioblastoma

Surgery[4]

• Pathological diagnosis• Alleviate mass effect• Increase survival

(mainly GTR)• Facilitate adjuvant

therapy• Decrease

corticosteroid need

Radiotherapy[5]

• Increase survival after surgery in newly diagnosed patients

Chemotherapy[6]

• Extend survival• Potentially increase

therapeutic effect of RT[1]

1. Rulseh AM et al. World J Surg Oncol. 2012;10:220.1-6.2. Henriksson R et al. J Neurooncol. 2011;104(3):639-646.3. NCCN Guidelines®. Central Nervous System Cancers. V1.2015. 4. Sanai N, Berger MS. Neurosurgery. 2008;62(4):753-766.5. Valduvieco I et al. Clin Transl Oncol. 2013;15(4):278-282.6. Stupp R et al. J Clin Oncol. 2007;25(26):4127-4136.

GTR, gross total resection; NCCN, National Comprehensive Cancer Network; PS, performance status; QOL, quality of life; RT, radiotherapy.

Page 19: Glioblastoma vibhay

Guidelines for Standard of Care

Diagnosis GBMCraniotomyMaximal Surgical resection or Biopsy

FitKPS>60

UnfitKPS<60

Age <70

Age >70

Radiotherapy short course or Temozolamide or BSC: steroids, anticonvulsants etc.

Radiotherapy 60Gy/30#/6weeks+ concurrent Temozolamide 75mg/m2

Followed by 6 cycles adjuvant Temozolamide 150 mg/m /day for 5d every 28d

Methylated

Unmethylated

1. NCCN Guidelines®. Central Nervous System Cancers. V2.2014. 2. Stupp R et al. Ann Oncol. 2014;25(Suppl 3):iii93-iii101.3. Weller M et al. Lancet Oncol. 2014;15(9):e395-e403

KPS 60 - Ambulatory and capable of most self-care but requires occasional assistance. Unable to carry out any work activities. KPS 50 - Capable of only limited self-care, confined to bed or chair more than 50% of waking hours. Requires considerable assistance and frequent medical care

Temozolamide

Radiotherapy 60Gy/30#/6weeks

Page 20: Glioblastoma vibhay

20

• Virtually all patients relapse and there is no defined SOC[1,2]

• Clinical trial enrollment is suggested by ESMO and always recommended by NCCN[1]

Treatment of Recurrent Glioblastoma

Good PS [1,3]

Poor PS

Re-resection† ± / + BCNU†

Surgery to alleviate mass

effect‡

BSCChemotherapy

TMZNU

PCVBEV ± IRI

± NU ± TMZ ± Carb

CP Carb Erlotinib Imatinib

Re-irradiation TTFBevacizumab use in glioblastoma is increasingly widespread[1]*

2A 4C

2A 2B1A

2B

2A 3C

2B

2C

2B 4C 3

BSC

NCCN

ESMO

Both

1. NCCN Guidelines®. Central Nervous System Cancers. V1.2015. 2. Decision resources. Glioblastoma Multiforme. 2013.3. Stupp R et al. Ann Oncol. 2014;25(Suppl 3):iii93-iii101.4. Wick W et al. J Clin Oncol. 2010;28(12):e188-e189.

IFNβ-based regimens are also used in Japan[4]

BCNU, carmustine; BEV, bevacizumab; BSC, best supportive care; Carb, carboplatin; CP, cisplatin; EMA, European Medicines Agency; ESMO, European Society of Medical Oncology; IFNβ, interferon beta; IRI, irinotecan; NCCN, National Comprehensive Cancer Network; NU, nitrosourea; PCV, vincristine; PS, performance status; SOC, standard of care; TMZ, temozolomide; TTF, tumor treating fields.

* BEV is used in parts of EU despite not having received EMA approval[4]; † For local recurrent disease, the NCCN Guidelines recommend re-resection, when feasible[1]; ‡ Recommended by NCCN for recurrent disease with diffuse/multiple tumors.[1]

Page 21: Glioblastoma vibhay

21

• All patients should receive BSC[1]

• Neurological function is closely monitored and lab tests are performed according to treatment regimen[2]

• MRI scans are routinely performed– Early detection of recurrence is desirable[1]

– Management of recurrent tumors is guided by patient condition and extent of disease[1]

Follow-Up/Monitoring

NCCN[1]

• Serial scans starting at 2–6 weeks after RT to titrate corticosteroid dose

• Then every 2–4 months for 2–3 years to monitor for recurrence

ESMO[2]

• Scans every 3–4 months unless otherwise clinically indicated

Monitoring MRI scans

1. NCCN Guidelines®. Central Nervous System Cancers. V1.2015. 2. Stupp R et al. Ann Oncol. 2014;25(Suppl 3):iii93-iii101.

BSC, best supportive care; ESMO, European Society for Medical Oncology; MRI, magnetic resonance imaging; NCCN, National Comprehensive Cancer Network; RT, radiotherapy.

Page 22: Glioblastoma vibhay

22

• Treatment-induced changes in brain permeability introduce challenges in detecting response to therapy and disease recurrence by MRI[1]

Challenges in Response Assessment: Pseudoprogression and Pseudoresponse

PseudoprogressionApparent increase of tumor lesion

on imaging that is not due to actual tumor growth[1-3]

PseudoresponseApparent decrease of tumor lesion

on imaging that does not reflect true tumor reduction[1]

Often occurs with antiangiogenic agents such as bevacizumab[1]

Observed in 5%–31% patients* after RT/chemotherapy[2]

• “Pseudoprogression” has also been observed after treatment with immunotherapies, but is often followed by tumor regression[4,5]

– Apparent increases in tumor lesions in these cases may be a result of immune-cell infiltration of the tumor[4]

* More common in MGMT-methylated patients.[2]

MGMT, O6-methylguanine DNA methyltransferase; MRI, magnetic resonance imaging; RT, radiotherapy.

1. Hygino da Cruz LC et al. AJNR Am J Neuroradiol. 2011;32(11):1978-1985.

2. Stuplich M et al. J Clin Oncol. 2012;30(31):e180-183.3. Brandsma D et al. Lancet Oncol. 2008;9(5):453-461.4. Wolchok JD et al. Clin Cancer Res. 2009;15(23)7412-7420.5. Lipson EJ. Oncoimmunology. 2013;2(4):e23661.

Page 23: Glioblastoma vibhay

23

iRANO Criteria for Glioblastoma Assessment

RANO Criteria[1,2] CR PR SD PD*

T1 enhancing disease None ≥ 50% < 50% if ↓ but

< 25% if ↑ ≥ 25% ↑

T2/FLAIR Stable/improved Stable/improved Stable/improved Worsened

New lesion None None None Present

Corticosteroid use None Stable or ↓ Stable or ↓ NA†

Clinical status Stable/improved

Stable/improved

Stable/improved Declined

iRANO[2]

• Further guidance on confirmation scans

• New lesions are incorporated into total lesion area

• Further guidance on considerations for corticosteroid use

* Progression occurs when any one of these criteria are present[1]; † Increase in corticosteroids alone will not determine PD without persistent clinical deterioration.[1]

1. Wen PY et al. J Clin Oncol. 2010;28(11):1963-1972. 2. Okada H et al. Lancet Oncol. 2015;16(15):e534-e542.

CR, complete response; FLAIR, fluid-attenuated inversion recovery; iRANO, Immunotherapy RANO; NA, not available; PD, progressive disease; PR, partial response; RANO, Response Assessment in Neuro-Oncology; SD, stable disease.

Page 24: Glioblastoma vibhay

24

Summary of and Challenges in Glioblastoma Treatment

Diagnosis • Early diagnosis and treatment do not improve outcomes for glioblastoma, precluding the utility of screening efforts[1]

Biomarkers• Although molecular characterization is specified in guidelines

for glioblastoma (eg, MGMT status), biomarkers have not yet translated into making clinical decisions[2]

Treatment• SOC for glioblastoma is MSR/Biopsy→RT/TMZ→TMZ[3,4]

• PS is the main criterion in deciding to administer SOC[3,4]

Recurrence • Virtually all glioblastomas recur, and there is nodefined SOC for this setting[3,5]

MGMT, O6-methylguanine DNA methyltransferase; MSR, maximal surgical resection.; PS, performance status; RT, radiotherapy; SOC, standard of care; TMZ, temozolomide.

1. Omuro A. JAMA. 2013;310(17):1842-1850. 2. Jackson CM et al. Clin Cancer Res. 2014;20(14):3651-3659.3. NCCN Guidelines®. Central Nervous System Cancers. V1.2015. 4. Stupp R et al. Ann Oncol. 2014;25(Suppl 3):iii93-iii101.5. Decision resources. Glioblastoma multifigure. 2013.

Page 25: Glioblastoma vibhay

Treatment Options for Glioblastoma

Module 3

Page 26: Glioblastoma vibhay

26

• RT with concurrent and adjuvant temozolomide is SOC for newly diagnosed patients[1-3]

Current Treatment Options for Newly Diagnosed Glioblastoma

Newly Diagnosed• Surgery (MSR or biopsy) ± carmustine wafer

− 5-ALA dye*• Radiation• Systemic chemotherapy

− Temozolomide• Clinical trials (NCCN)• Bevacizumab (Japan only†)

* Approved in Europe, Japan, and Canada.[4]

† As part of SOC.[3]

5-ALA, aminolevulinic acid; MSR, maximal safe resection; NCCN, National Comprehensive Cancer Network; RT, radiotherapy; SOC, standard of care.

1. NCCN Guidelines. Central Nervous System Cancers. V1. 20152. Stupp R et al. Ann Oncol. 2014;25(Suppl3):iii93-iii101.3. Takano S et al. Onco Targets Ther. 2014;7:1551-1562.4. Roberts DW et al. Neurosurg Clin N Am. 2012;23(3):271-377.

Page 27: Glioblastoma vibhay

27

• Adjuvant radiotherapy has long been a mainstay of glioblastoma therapy[1]

– RT is administered to T1-enhanced tumor region + 2–3-cm margin on T2 or FLAIR abnormality[2]

• Key randomized clinical trials demonstrated mOS benefit of 5 mo for RT in newly diagnosed patients:

Radiotherapy

Trial Setting N Experimental Arms Control Arms mOS

NCI (1978)[3] 1L, Anaplastic gliomas 303

RT

BSC

36 wks

14 wksRT + IV BCNU 34.5 wksIV BCNU 18.5 wks

Scandinavian Glioblastoma Study Group[4]

1L 118RT+ bleomycin

BSC10.8 mo

5.2 moRT 10.8 mo

1L, first line; BCNU, 3-bis (2-chloroethyl 1)-1-nitrosourea (carmustine); BSC, best supportive care; EBRT, external beam radiation therapy; IFRT, involved-field radiation therapy; FLAIR, fluid-attenuated inversion recovery; IV, intravenous; MGMT, O6-methylguanine DNA methyltransferase; mOS, median overall survival; NCI, National Cancer Institute; PS, performance score; RT radiotherapy.

1. Brandes AA et al. J Clin Oncol. 2009;(8):1275-12792. Dhermain F. Chin J Cancer. 2014;33(1):16-24.3. Walker MD et al. J Neurosurg. 1978;49(3):333-343.4. Kristiansen K et al. Cancer. 1981;47(4):649-652.5. NCCN Guidelines. Central Nervous System Cancers. V1. 2015.6. Malmstrom A et al. Lancet Oncol. 2012;13(9):916-926.

• Fractionated EBRT and IFRT: Most commonly used RT modalities[5]

• Hypofractionated radiotherapy: Often preferred in elderly/poor PS patients, especially when lacking MGMT promoter methylation[6]

Page 28: Glioblastoma vibhay

28

Chemotherapy: Carmustine Wafer (BCNU) • Biodegradable polymer wafers loaded with nitrosourea carmustine (BCNU) are

implanted into the surgical cavity at the time of initial resection[1]

• A key phase III trial demonstrated survival benefit* for BCNU wafers in newly diagnosed malignant glioma patients (N=240)[2,3]

* P=0.03.[2]

BCNU, 3-bis (2-chloroethyl 1)-1-nitrosourea (carmustine); CI, confidence interval; Gliadel, poly [carboxyphenoxy-propane/sebacic acid] anhydride wafers containing 3.85% carmustine; HR, hazard ratio; NS, not significant; OS, overall survival; PBO, placebo.

100

90

80

70

60

50

40

30

20

10

00 6 12 14 16 18 22 24

Months From Implant Surgery

Sur

viva

l Rat

e (%

)

42 20108

BCNU

Placebo

• At 3-year follow-up, glioblastoma patients who received BCNU had 22% reduced risk of death (NS)[3]

• Survival benefit of BCNU wafer for glioblastoma remains unclear[2,3]

• Implanting wafers may impact clinical trial enrollment[4]

1. Valtonen S et al. Neurosurgery. 1997;41(1):44-49.2. Westphal M et al. Neuro Oncol. 2003;5(2):79-88.3. Westphal M et al. Acta Neurochir (Wein). 2006;148(3):269-275.4. NCCN Guidelines. Central Nervous System Cancers. V1. 2015.

Median OS (mo)BCNU

(n=101) 13.5 HR (95% CI):0.76 (0.55–1.05)

P=0.10PBO (n=106) 11.4

Glioblastoma Subgroup (n=207)[2]

Minimum follow-up:

12 mo

Page 29: Glioblastoma vibhay

29

Chemotherapy: Temozolomide (TMZ)

TMZ is an alkylating agent prodrug• Alkylating agents bind to DNA, impairing

replication/transcription and ultimately leading to cell death[1]

• TMZ is able to cross the BBB and is spontaneously converted into the active metabolite in the CNS[2]

• TMZ is a standard treatment for newly diagnosed glioblastoma[3]

− Administered daily concomitantly with RT and as maintenance thereafter*[3]

− Available in oral and IV formulation[4]

* Alternative TMZ dosing regimens have not demonstrated improved efficacy, but continue to be investigated (see Module 4 for further details).

Tumorcell

TMZ

ReplicationTranscription

1. Fu D et al. Nat Rev Cancer. 2012;12(2):104-120.2. Agarwala SS, Kirkwood JM. Oncologist. 2000;5(2):144-151.3. NCCN Guidelines. Central Nervous System Cancers. V1. 2015.4. TEMODAR [package insert]. Whitehouse Station, NJ:

Merck & Co, Inc; 2014. 5. Wesolowski JR et al. Am J Neuroradiol. 2010;31(8):1383-1384.

BBB, blood-brain barrier; CNS, central nervous system; DNA, deoxynucleic acid; IV, intravenous; RT, radiotherapy; TMZ, temozolomide.

Adapted from Wesolowski et al 2010.[5]

Page 30: Glioblastoma vibhay

30

TMZ in Glioblastoma: Phase III EORTC/NCIC Trial

Daily TMZ + RT→ 6 cycles TMZ

Key Inclusion Criteria[1]

• 18–70 years old• Newly diagnosed

glioblastoma• WHO PS<2

R

RT alone

N=573

• ~2 month increase in mOS[1]

• 2-yr survival: 26.5% vs 10.4%[1]

• 5-yr survival: 10% vs 2%[2]

00 6 12 18 24 30 36 42

102030405060708090

100

Months

Prob

abili

ty o

f Ove

rall

Surv

ival

(%)

RT + TMZRT

The widespread use of TMZ in glioblastoma is based on the EORTC/NCIC trial

1. Stupp R et al. N Engl Med. 2005;352(10):987-996.2. Stupp R et al. Lancet Oncol. 2009;10(5):459-466.

CI, confidence interval; EORTC, European Organization for Research and Treatment of Cancer; HR, hazard ratio; mOS, median OS; NCIC, National Cancer Institute of Canada; OS, overall survival; PS, performance status; R, randomization; RT, radiotherapy; TMZ, temozolomide; WHO, World Health Organization.

Median OS (mo)[1]

RT+TMZ(n=287) 14.6 HR (95% CI):

0.63 (0.52–0.75)P<0.001RT

(n=286) 12.1

Page 31: Glioblastoma vibhay

31

• Alkylating agents, such as TMZ, cause cell death by binding to DNA[1,2]

• The MGMT protein can reverse alkylation*[1]

• MGMT methylation leads to loss of MGMT protein and thus, ineffective DNA repair[1]

‒ TMZ remains effective• Unmethylated MGMT retains

MGMT protein synthesis and therefore, DNA repair[1]

‒ TMZ ineffective

MGMT Methylation Status in Glioblastoma: Predictive Biomarker for TMZ

100 EORTC/NCIC Trial[2]

80

60

40

20

00 6 12 18 24 30 36 42

Months

Prob

abili

ty o

f OS

(%)

MethylatedMGMT

promoter

UnmethylatedMGMT

promoter P<0.001

1. Esteller M et al. N Engl J Med. 2000;343(19):1350-1354.2. Hegi ME et al. N Engl J Med. 2005;352(10):997-1003.

* If alkylation is at the O6 position of guanine.[1]

EORTC, European Organisation for Research and Treatment of Cancer; MGMT, O-methylguanine DNA methyltransferase; OS, overall survival; NCIC, National Cancer Institute of Canada; TMZ, temozolomide.

Page 32: Glioblastoma vibhay

32

• Non-invasive medical device that applies tumor-treating fields (TTF) via electrodes placed on the scalp, shown to have antimitotic activity[1,2]

• Phase III trial in newly diagnosed glioblastoma was terminated at interim analysis due to early success[3]

– Control arm pts are now crossing over to receive SOC + TTF[3]

TTF for Newly Diagnosed Glioblastoma

1. Vymazal J, Wong ET. Semin Oncol. 2014;41(Suppl6):S14-24.2. Stupp R et al. Eur J Cancer. 2012;48(14):2192-2202.3. PR Newswire. Novocure EF-14 PhIII. www.prnewswire.com/

news-releases/novocure-announces-the-ef-14-phase-iii-clinical-trial-of-tumor-treating-fields-in-patients-with-newly-diagnosed-glioblastoma-has-been-terminated-at-the-interim-analysis-due-to-early-success-282808841.html. Accessed December 17, 2015.

Trial[4] Study Arms N mPFS[3] mOS[3] 2-yr Survival[3]

EF-14NCT00916409Phase III

SOC + TTF*vs

SOC

315[3]

(interim analysis)

7.1 vs 4 mo

HR=0.63; P=0.001

19.6 vs 16.6 mo

HR=0.75; P=0.034

43% vs 29%

HR, hazard ratio; mOS, median overall survival; mPFS, median progression-free survival; SOC, standard of care; TTF, tumor-treating fields.

* Administered as 4 insulated electrode arrays placed on scalp.[4]

Page 33: Glioblastoma vibhay

33

• Virtually all patients eventually relapse[1]

• There is no standard of care for relapsed patients[2]

Overview of Current Treatment Options for Recurrent Glioblastoma

• Chemotherapy− Temozolomide− Nitrosoureas− PCV− Cyclophosphamide− Cisplatin/Carboplatin

• Targeted therapies− Bevacizumab* ±

chemotherapy− Erlotinib/Imatinib†

• Re-resection ± carmustine wafer

• Alternating electric field therapy

• Re-irradiation‡

• Clinical trials (NCCN and ESMO/EANO)

BEV, Bevacizumab; EANO, European Association for Neuro-Oncology; ESMO, European Society for Medical Oncology; EMA, European Medicines Agency; FDA, Food and Drug Administration; NCCN, National Comprehensive Cancer Network; PCV, procarbazine/lomustine/vincristine.

1. Felsberg J et al. Int J Cancer. 2011;129(3):659-670.2. Gil-Gil MJ et al. Clin Med Insights Oncol. 2013;7:123-135.3. NCCN Guidelines. Central Nervous System Cancers. V1. 20154. Stupp R et al. Ann Oncol. 2014;25(Suppl3):iii93-iii101.

* Currently approved by FDA but not EMA. BEV + chemo considered if BEV monotherapy fails (NCCN); BEV ± Irinotecan is Category 3C in ESMO.

† Recommended in ESMO guidelines (Category 2C) but not in NCCN guidelines.‡ Data are lacking on re-irradiation of recurrent glioblastomas, and its use is controversial.

Recurrent Disease[3,4]

Page 34: Glioblastoma vibhay

34

• Even before the 2009 accelerated approval in US, bevacizumab has been used for management of recurrent glioblastoma in US and parts of Europe[1]

• In Japan, BEV use has been increasing since its approval[2]

• Despite PFS benefit, no significant effect of bevacizumab on mOS has been reported in phase III trials[3,4]

Bevacizumab: Registrational Trial Data

Trial Study Arms Ph Study Setting N ORR, % mPFS, mo mOS, mo

BRAIN[5]BEV

II Recurrent glioblastoma 167

28 4.2 9.2BEV + irinotecan 38 5.6 8.7

NCI[6] BEV II Recurrent glioblastoma 48 35 16 wks 31 wks

JO22506[7] BEV IIRecurrent

glioblastoma (Japan)

31 28 3.3 10.5

AVAglio[3] BEV + RT + TMZ vsPlacebo + RT + TMZ III Newly diagnosed

glioblastoma 921 NA 10.6 vs 6.2* 16.8 vs 16.7†

* Statistically significant. † Not statistically significant (P=0.10).

BEV, bevacizumab; mOS, median overall survival; mPFS, median PFS; NA, not applicable; NCI, National Cancer Institute; ORR, objective response rate; PFS, progression-free survival; Ph, phase; RT, radiotherapy; TMZ, temozolomide.1. Wick W et al. J Clin Oncol. 2010;28(12):e188-189.

2. CancerMPact® Japan, 2013. April 2014, v1.1. 3. Chinot OL et al. N Engl J Med. 2014;370(8):709-722.4. Gilbert MR et al. N Engl J Med. 2014;370(8):699-708.5. Friedman HS et al. J Clin Oncol. 2009;27(28):4733-4740. 6. Kreisl TN et al. J Clin Oncol. 2009;25(5):740-745. 7. Nagane M et al. Jpn J Clin Oncol. 2012;42(10):887-895.

Page 35: Glioblastoma vibhay

35

• The FDA conferred accelerated approval for bevacizumab (2009) based on nonrandomized phase II trials[1]

– Approval based on durable objective responses observed in two single-arm trials, AVF3708g and NCI 06-C-0064E.

Regulatory Approval of Bevacizumab

3. ESMO Oncology News. Available at http://www.esmo.org/Oncology-News/European-Medicines-Agency-Adopts-a-Final-Negative-Opinion-for-an-Extension-of-Indications-for-Bevacizumab. Accessed December 17, 2015

4. ESMO Oncology News. Available at http://www.esmo.org/Oncology-News/European-Medicines-Agency-Recommends-to-Refuse-a-Change-to-the-Marketing-Authorisation-for-Bevacizumab. Accessed December 17, 2015.

5. Roche Media Release, 2013. Available at http://www.roche.com/media/media_releases/med-cor-2013-06-17.htm Accessed December 17, 2015.

• The EMA rejected regulatory approval for bevacizumab in 2009 and 2014[2,3]

– Doubtful of ORR and PFS as endpoints and wary of lack of OS benefit[4]

• ORR is highly variable in independent assessments[2]

• Anti-angiogenic agents affect brain permeability and therefore imaging responses[2]

• The Japanese MHLW approved bevacizumab for both lines of therapy[5]

– Approval in the newly diagnosed setting was based on improved mPFS results in the AVAglio study[5]

EMA, European Medicines Agency; FDA, Food and Drug Administration; MHLW, Ministry of Health/Labor/and Welfare; ORR, objective response rate; OS, overall survival; mPFS, median progression-free survival; PFS, progression-free survival.

1. Cohen MH et al. Oncologist. 2009;14(11):1131-1138.2. Wick W et al. J Clin Oncol. 2010;28(12):e188-e189.

Page 36: Glioblastoma vibhay

36

• Despite PFS improvement, no OS benefit observed in either trial[1,2]

• Due to discrepancy in QOL, it has been proposed that the investigators of these studies should share their raw data with the US FDA[5]

Conflicting Quality of Life Outcomes From RTOG 0825 Trial and AVAGlio

Trial N Primary Endpoint(s) Key Efficacy Data Key Safety Data

RTOG 0825NCT00884741[1,3] 637 OS, PFS

BEV vs PBO• mOS: 15.7 vs 16.1 mo• mPFS*: 10.7 vs 7.3

• Increased AE profile in BEV group• Increased symptom burden,

worse QOL, declined neurocognition in BEV group

AVAglioNCT00943826[2,4] 921 OS, PFS

BEV vs PBO• mOS: 16.8 vs 16.7 mo• mPFS†: 10.6 vs 6.2 mo

• More frequent grade >3 AE in BEV group

• QOL and PS maintained longer in BEV group

1. Gilbert MR et al. N Engl J Med. 2014;370(8):699-708.2. Chinot OL et al. N Engl J Med. 2014;370(8):709-722.3. Clinicaltrials.gov. NCT00884741 4. Clinicaltrials.gov. NCT009438265. Fine HA. N Engl J Med. 2014;370(8):764-765.

* Did not meet prespecified cutoff for outcome benefit (P=0.007).† Statistically significant (P<0.001).

Phase III RTOG 0825 and AVAglio trials compared SOC + BEV vs placebo in newly diagnosed glioblastoma patients[1,2]

AE, adverse event; BEV, bevacizumab; OS, overall survival; mPFS, median progression-free survival; PBO, placebo; PFS, progression-free survival; PS, performance status; QOL, quality of life; RTOG, Radiation Therapy Oncology Group; SOC, standard of care.

Page 37: Glioblastoma vibhay

37

• Recently reported results from a randomized trial evaluating BEV ± lomustine demonstrated potentially meaningful OS benefit for the combination[1]:Prespecified criteria for further phase III studies were met for the combination[2]

Bevacizumab in Combination With Lomustine: BELOB and EORTC 26101

Trial[1] Experimental Arms Ph Study Setting N ORR, % 6-mo PFS, % 9-mo OS, %

BELOBNTR1929

LomustineII Recurrent

glioblastoma 1485 13 43

BEV 38 16 38 Lomustine* + BEV 34 41 59

1. Taal W et al. Lancet Oncol. 2014;15(9):943-953.2. Taal W et al. Presentation at SNO 2014. AT-55.3. Clinicaltrials.gov : NCT01290939.4. Wick W. et al SNO 2015 Abstract

1L, first line; 2L, second line; BEV, bevacizumab; EORTC, European Organisation for Research and Treatment of Cancer; NTR, Netherlands Trial Register; ORR, objective response rate; OS, overall survival; PFS, progression-free survival.

* 90 mg/m2 dose, determined after interim safety analysis.

• EORTC 26101: Phase III Trial Exploring the Combination of Bevacizumab and Lomustine vs Lomustine in Patients With First Recurrence of Glioblastoma3,4

Trial Experimental Arms Study design Ph N Primary Endpoint mPFS* mOS**

EORTC 26101 NCT01290939

Lomustine ± BEV Recurrent III 433 OS

1.54 mos (LOM; n=149) vs.

4.17 mos (BEV+LOM; n=288)

mOS 8.64 mos (LOM; n=149) vs.

9.10 mos (BEV+LOM;

n=288) * Locally assess PFS was longer with the addition of BEV to LOM: HR 0.49 (95% CI 0.39, 0.61) P< .0001

** With 329 OS events (75.3%) OS was not superior in the combination therapy arm (HR: 0.95, CI: 0.74-1.21, p=0.650)

Toxicity was in the expected range with more events in the combination arm being also longer on treatment

Page 38: Glioblastoma vibhay

38

Key Adverse Events by Class of Therapy

VEGF-Targeted Agents[4-9]

• Hypertension and diarrhea occur in all VEGF/VEGFR inhibitors

• Other AEs include gastrointestinal AEs (diarrhea, nausea, vomiting), dermatologic AEs (HFS), and fatigue

• Bevacizumab has a black box warning in the US for GI perforation and serious bleeding[9]

Alkylating Agents(TMZ, BCNU)[2-3]

• Alopecia• Fatigue• Nausea/vomiting• Thrombocytopenia• Neutropenia

RT[1]

• Hair loss• Nausea• Fatigue• Increased ICP• Other AEs include

memory loss and radiation necrosis

• Adverse events associated with TMZ are generally milder than other alkylating agents[2]

AE, adverse event; BCNU, carmustine; GI, gastrointestinal; HFS, hand-foot syndrome; ICP, intracranial pressure; RT, radiotherapy; TMZ, temozolomide; VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor.

1. Lawrence YR et al. Br J Cancer. 2011;104(9):1365-1371.2. Chang L et al. Onco Targets Ther. 2014;7:235-244.3. Reithmeier T et al. BMC Cancer. 2010;10:30.

4. Eisen T et al. J Natl Cancer Inst. 2012;104(2):93-113. 5. SUTENT [package insert]. New York, NY: Pfizer Labs; 2014. 6. VOTRIENT [package insert]. Middlesex, UK: GlaxoSmithKline; 2014.7. NEXAVAR [package insert]. Berlin, Germany: Bayer HealthCare

Pharmaceuticals Inc; 2013. 8. INLYTA [package insert]. New York, NY: Pfizer Labs; 2013. 9. AVASTIN [package insert]. San Francisco, CA: Genentech, Inc; 2013.

Page 39: Glioblastoma vibhay

39

Summary of Currently Available Agents Approved for Glioblastoma

Agent[1-4] MOA[1,3,5] Line[1,2] Admin[1,2,6-9] Guideline Recommendation[1-4]

Temozolomide Alkylating agent ND, R PO/IV NCCN, ESMO

Carmustine (wafer) Alkylating agent ND, R Implant NCCN, ESMO

Carmustine (systemic) Alkylating agent R IV NCCN, ESMO*

Lomustine Alkylating agent R PO NCCN, ESMO

Bevacizumab VEGF inhibitorND

IVJapan only

R NCCN, JapanTTF device Anti-mitotic R Electrodes NCCN†, Japan‡

* Single agent nitrosourea therapy.[2]

† Not all panelists recommended TTF.[1]‡ TTF has been made available on a restricted case basis for compassionate use in Japan.[4]

Admin, administration; ESMO, European Society for Medical Oncology; IV, intravenous; NCCN, National Comprehensive Cancer Network; MOA, mechanism of action; ND, newly diagnosed; PO, by mouth; R, recurrent; TTF, tumor-treating fields; VEGF, vascular endothelial growth factor.

1. NCCN Guidelines ®. Central Nervous System Cancers. V1. 2015.2. Stupp R et al. Ann Oncol. 2014;25(Suppl3):iii93-iii101.3. Roche Press Release, June 17, 2013. Available at:

http://www.roche.com/media/store/releases/med-cor-2013-06-17.htm. Accessed February 25, 2015.

4. Muragaki Y et al. Presentation at SNO 2014. NT-25.5. Decision Resources: Glioblastoma Multiforme. September 2013.6. TEMODAR [package insert]. Whitehouse Station, NJ: Merck & Co, Inc;

2014. 7. BiCNU [package insert]. Princeton, NJ: Bristol-Myers Squibb Company;

2011.8. CeeNU [package insert]. Princeton, NJ: Bristol-Myers Squibb

Company; 2012.9. AVASTIN [package insert]. South San Francisco, CA: Genentech, Inc;

2014.

Page 40: Glioblastoma vibhay

40

Additional Recommended Agents for Glioblastoma

Agent[1,2] MOA[2-4] Line[1,2] Admin[3,4] Guideline Rec[1,2] Key Efficacy[2,4-6]

Cyclophosphamide Alkylating agent R PO NCCN Modest mPFS increase

PCVProcarbazine Alkylating

agents RPO

NCCN, ESMO Comparable efficacy with TMZLomustine PO

Vincristine IV

Carboplatin DNA crosslinking agents

R IV NCCNModest activity

Cisplatin R IV NCCN

Bevacizumab VEGF inhibitor R IV ESMO Steroid-sparing effect†

Irinotecan Topoisomerase inhibitor R IV NCCN, ESMO Modest mPFS

increase (+BEV)†

Erlotinib EGFR inhibitor R PO ESMO Anti-tumor efficacy not shown‡Imatinib PDGFR inhibitor R PO ESMO

Admin, administration; bev, bevacizumab; DNA, deoxynucleic acid; EGFR, epidermal growth factor receptor; ESMO, European Society for Medical Oncology; IV, intravenous; mPFS, median progression-free survival; NCCN, National Comprehensive Cancer Network; PCV, procarbazine/ lomustine/vincristine; PDGFR, platelet-derived growth factor receptor; PO, by mouth; R, recurrent; rec, recommendation; RR, response rate; TKI, tyrosine kinase inhibitor; TMZ, temozolomide,

VEGF, vascular endothelial growth factor. 1. NCCN Guidelines®. Central Nervous System Cancers. V1. 20152. Stupp R et al. Ann Oncol. 2014;25(Suppl3):iii93-iii101.3. Decision Resources: Glioblastoma Multiforme. September 2013.4. Chamberlain MC, Tsao-Wei DD. Cancer. 2004; 100(6):1213-20.5. Roci E et al. Med Arch. 2014;68(2):140-143.6. Friedman HS et al. J Clin Oncol. 2009;27(28):4733-4740.

* As single agents in recurrent high grade gliomas[5]; † In a study of bevacizumab±irinotecan, there was a trend toward stabilization/decrease in steroid use, in patients taking corticosteroids at baseline[6]; ‡ In unselected patient populations.[2]

Page 41: Glioblastoma vibhay

41

Limitations of Current Therapies

BBB, blood-brain barrier; mOS, median overall survival; mPFS, median progression-free survival; RR, response rate; tx, treatment.

1. Patel MA et al. Cancers. 2014;6(4):1953-1985. 2. SEER Stat Fact Sheets: Brain and Other Neurons System

Cancer. Available at: http://seer.cancer.gov/statfacts/html/brain.htmlAccessed December 17, 2015.

Treatment Outcomes

• Over the past 4 decades, treatment advances have only modestly impacted overall patient survival[1,2]

• Recurrence rate remains ~100%[3]

• Prognosis for patients with relapsed glioblastoma remains extremely poor[4]

− RR <4%–16% − mPFS ~2.3 months− mOS 3–9 months

Challenges and Unmet Needs

• Imaging challenges due to tx-related changes in brain permeability[5]

• Fewer tx options for elderly population despite higher incidence[6]

• Drug delivery past the BBB[7]

• Neurotoxicity of treatments[7]

3. NCCN Guidelines. Central Nervous System Cancers. V1. 2015.4. Gil-Gil MJ et al. Clin Med Insights Oncol. 2013;7:123-135. 5. Hygino da Cruz LC et al. AJNR Am J Neuroradiol.

2011;32(11):1978-1985.6. Arvold ND, Reardon DA. Clin Interv Aging. 2014;9:357-367.7. Chamberlain MC. Neurosurg Focus. 2006;20(4):E19.

Page 42: Glioblastoma vibhay

Investigational Agents for Glioblastoma

Module 4

Page 43: Glioblastoma vibhay

43

Investigational Agents:Chemotherapies (cont’d)

VAL-083 (DelMar) is a novel alkylating agent under phase I/II development for recurrent glioblastoma[1,2]

• Unaffected by MGMT methylation status in vitro[1]

Has orphan drug designation in US (2012) and Europe (2013) for gliomas[3]

Preliminary results of phase I/II dose escalation trial in newly diagnosed pts*[2] have recently been reported[4]:

• Safety: Well-tolerated; MTD not yet reached• Efficacy: Improved clinical signs in 3 pts reaching SD or PR†

1. Steino A et al. Mol Cancer Ther. 2013;12(11 suppl):B252.2. Clinicaltrials.gov. NCT01478178.3. DelMar Press Release. Available at:

http://www.delmarpharma.com/DelMarPharmaVAL-083EUORPHANDRUG130107.pdf. Accessed February 17, 2015.

4. Shih KC et al. Poster presentation at ASCO 2014. 2093.

AE, adverse event; MGMT, O6-methylguanine DNA methyltransferase; MTD, maximum tolerated dose; PR, partial response; SD, stable disease; TMZ, temozolomide.

* Also enrolling patients previously treated with surgery and/or radiation who have failed on both TMZ and bevacizumab.[4]

† Maximum response of 28 cycles (84 weeks) prior to discontinuing due to unrelated AE.[4]

Page 44: Glioblastoma vibhay

44

Although approved in 1L and 2L settings, OS benefit has not been observed[1]

Investigational Agents: Antiangiogenics

Other Antiangiogenic Agents• TKIs targeting the VEGFR pathway have

shown limited or no clinical benefit in glioblastoma, with some trials demonstrating high toxicity rates[1,2]

• VEGF-targeting next-generation antibody pegdinetanib is in phase II development for glioblastoma[3]

Adapted from Oudard S et al 2012.[4]

PI3K

mTOR

AKT

Angiogenesis

VEGFR

VEGF

Tumorcell

Bevacizumab*

*Bevacizumab is further discussed in Module 3

1. Wilson TA et al. Surg Neurol Int. 2014;5:64.2. Reardon DA, Wen PY. Oncologist. 2006;11(2):152-164.3. Evans JB, Syed BA. Nat Rev Drug Discov. 2014;13(6):413-4144. Oudard S et al. Cancer Treat Rev. 2012;38(8):981-987.

1L, first line; 2L, second line; AKT, protein kinase B; mAb, monoclonal antibody; mTOR, mammalian target of rapamycin; OS, overall survival; PI3K, phosphoinositide-3 kinase; TKI, tyrosine kinase inhibitor; VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor.

Roche

Page 45: Glioblastoma vibhay

45

• DNA-histone H1 complex*

• Radioactive Iodine

Early Phase Targeted Agents for Glioblastoma: Cytotoxic Antibody Conjugates• Antibody-drug conjugates are designed to deliver cytotoxic drug activity

specifically to tumor cells[1]

• LRP-1†

• Anti-MT agent paclitaxel

• EGFR or mut EGFRvIII

• Anti-MT agent MMAF

131I-chTNT-1/B MAb[2]

ANG-1005[3]

ABT414[4]

Adapted from http://www.seattlegenetics.com/adc_technology.

1 ADC bindsto target

ADC receptor complex is

internalized

2

Apoptosis4

Cytotoxic agent is released

3

1. Seattle Genetics. Advancing ADC technology. Available at: http://www.seattlegenetics.com/adc_technology. Accessed December 17, 2015

2. Shapiro WR et al. J Clin Oncol. 2010;28:15s. Abstract 2039.3. Drappatz J et al. Poster presentation at ASCO 2010. 2009.4. Gan HK et al. Poster presentation at ASCO 2014. 2021.

* DNA-histone H1 complex is exposed in tumor necrotic core[2].† LRP1 is expressed in BBB endothelium and in tumor cells[3].

ADC, antibody-drug conjugate; EGFR, epidermal growth factor receptor; EGFRvIII, EGFR variant III; LRP-1, low-density lipoprotein receptor-related protein 1; MMAF, monomethyl auristatin F; MT, microtubule.; mut, mutated.

Peregrine

Angiochem

AbbVie

Page 46: Glioblastoma vibhay

46

Early Phase Targeted Agents for Glioblastoma: Cytotoxic Antibody Conjugates (cont’d)

Study N Efficacy Safety Status

131I-chTNT-1/B MAb[1-3]

Phase II

Dose-confirmatory in pts at 1st relapse

41 • mOS: 9.3 mo• 2-yr OS: 19%

Grade 3/4 AE in 22% patients

• Orphan drug status, FDA and EMA

• Fast-track status, FDA

• No currently ongoing trials*

ANG1005[4,5]

Phase I (NCT00539344)

Safety and preliminary efficacy in recurrent glioblastoma and AG pts

63 2 CR and 2 PR in glioblastoma pts

• Well-tolerated• No CNS toxicity• Minimal systemic

toxicity

• Orphan drug status, FDA[6]

• Fast-track status, FDA[6]

• Phase II trial currently ongoing[4]

ABT414[4,7]†

Phase I +TMZ(NCT01800695)

Safety and preliminary efficacy in newly diagnosed or recurrent pts

21 1 CR and 3 PR in TMZ-refractory pts

MMAF-induced corneal epithelial microcysts

• Orphan drug status, FDA and EMA

• Trial still ongoing

1. Peregrine Press Release. 2012. http://ir.peregrineinc.com/releasedetail.cfm?ReleaseID=725023 Accesed December 21, 2015

2. Peregrine Press Release. 2011. http://ir.peregrineinc.com/releasedetail.cfm?ReleaseID=579150 Accesed December 21, 2015

3. Shapiro WR et al. J Clin Oncol. 2011;29. Abstract 2035.4. Clinicaltrials.gov.5. Drappatz J et al. Poster presentation at ASCO 2010. Abstract 2009.6. Angiochem. Angiochem’s ANG1005 received orphan drug designation from FDA for

the treatment of glioblastoma multiform. http://angiochem.com/angiochem%E2%80%99s-ang1005-received-orphan-drug-designation-fda-treatment-glioblastoma-multiform. Accessed February 15, 2015.

7. Gan HK et al. Poster presentation at ASCO 2014. 2021.

AE, adverse events; AG, angioblastoma; CNS, central nervous system; CR, complete response; EMA, European Medicines Agency; FDA, Food and Drug Administration; mAb, monoclonal antibody; MMAF, monomethyl auristatin F; mOS, median OS; OS, overall survival; TMZ, temozolomide.

* Plans for phase III trial have been approved by FDA as of 2012.[5]

† Phase II trial is planned.[1]

Page 47: Glioblastoma vibhay

47

Phase III Failures of Targeted Agents Highlight Need for New Treatment Modalities

Agent MOA Patient Population Results (Primary Endpoint)

Enzastaurin[1] Serine/threonine kinase inhibitor

Recurrent Terminated following interim futility analysis (no PFS benefit)

Cediranib[2] (REGAL trial) VEGF Inhibitor Recurrent No PFS, OS benefit demonstrated

Imatinib[3] TKI Recurrent No PFS benefit demonstratedCintredekin besudotox (IL13-PE38)[4]

Cytotoxin Recurrent No OS benefit demonstrated

TransMID (Tf-CRM107)[5,6]

Cytotoxin Progressive or Recurrent (unresectable)

Terminated following interim analysis (unlikely to meet trial criteria for efficacy)

Trabedersen (AP12009)[7] TGF-βoligonucleotide

Progressive or Recurrent Grade III and IV gliomas

Serious adverse events

Cilengitide[8,9] Integrin inhibitor Newly diagnosed, MGMT-methylated

No OS, PFS benefit demonstrated

Nimotuzumab[10] EGFR mAb Newly diagnosed No PFS, OS benefit demonstrated

4. Kunwar S et al. Neuro Oncol. 2010;12(8):871-881. 5. Clinicaltrials.gov. NCT00083447. 6. Drugs.com. Celtic terminates Transmid trial. Available at: http://www.drugs.com/news/celtic-pharma-terminates-transmid-trial-ksb311r-ciii-001-5246.html. Accessed December 17, 2015. 7. Clinicaltrials.gov. NCT00761280. 8. Tactical Therapeutics. Glioblastoma—overcoming resistance in malignant brain cancer. Available at: http://www.tacticaltherapeutics.com/ glioblastoma/. Accessed December 17, 2015. 9. Stupp R et al. Lancet Oncol. 2014;15(10):1100-1108. 10. Westphal M, Bach F. J Clin Oncol. 2012 (suppl):30. Abstract 2033.

EGFR, epidermal growth factor receptor; mAb, monoclonal antibody; MGMT, O6-methylguanine DNA methyltransferase; MOA, mechanism of action; mOS, median OS; OS, overall survival; PFS, progression-free survival; TGF-β, transforming growth factor-beta; Tf, transferrin; TKI, tyrosine kinase inhibitor; VEGF, vascular endothelial growth factor.

1. Wick W et al. J Clin Oncol. 2010;28(7):1168-1174.2. Batchelor TT et al. J Clin Oncol. 2013;31(26):3212-3218.3. Dresemann G et al. J Neurooncol. 2010;96(3):393-402.

Page 48: Glioblastoma vibhay

48

PD-1/PD-L1 in Glioblastoma

PD-1 was found to be expressed in 29% of tumor-infiltrating lymphocytes in glioblastoma tumor samples[1]*

• Expressed in 88% of newly diagnosed and 72% of recurrent glioblastomas†

PD-L1 was highly expressed in glioblastoma tumors[1]*:

• Low expression associated with proneural/G-CIMP glioblastoma subtypes[1]

• High expression associated with mesenchymal subtype[1]

• Immune suppressive heterogeneity has been observed between molecular subtypes[2]

PD-L1 expression correlated with glioblastoma molecular subtypes:

1. Berghoff AS et al. Neuro Oncol. 2014;pii:nou307. 2. Gilbert MR. Oral presentation at EANO 2014.

* Expression assayed by IHC in retrospective cohort.[1]

† Expression pattern of variable extent.[1]

G-CIMP, glioma CpG island methylator phenotype; IHC, immunohistochemistry; PD-1, programmed death-1; PD-L1, PD ligand-1; TIL, tumor-infiltrating lymphocyte.

Page 49: Glioblastoma vibhay

49

PD-1/PD-L1 in Glioblastoma (cont’d)

*Retrospective analysis (N=563).[1]

• Correlation between PD-L1 expression and response to PD-1 pathway inhibitors has been considerably variable across tumor types[1,2] − Predictive value of PD-L1 expression remains under investigation in

ongoing trials, including trials in glioblastoma1

− Positivity cutoffs used in PD-L1 IHC assays can also affect correlation[3]

PD-L1 expression in tumor samples did not correlate with patient survival*[1]

1. Berghoff AS et al. Neuro Oncol. 2014;pii:nou307. 2. Gettinger SN. Oral presentation at WCLC 2013. MO19.09.3. Goodman A. The ASCO Post. 2014;5(7).

IHC, immunohistochemistry; PD-1, programmed death-1; PD-L1, PD ligand-1.

Page 50: Glioblastoma vibhay

50

Emerging Biomarkers: PD-L1

Expression in[1,2]

Primary glioblastoma(membranous)

16.7%–38.3%

Primary glioblastoma(diffuse/fibrillary)

72.2%–88%

Prognostic Value

• Tumor cell expression of PD-L1 (and PD-1) associated with poor outcomes in glioblastoma patients[2]

Clinical Significance• Lower expression of PD-L1 on monocytes

was predictive for improved survival in glioblastoma patients treated with an autologous peptide vaccine[3]

• Tumor-expressed PD-L1 is being evaluated for value as a biomarker across multiple tumor types[4]

PD-1, programmed death-1; PD-L1, PD ligand-1. 1. Berghoff AS et al. Neuro Oncol. 2015;17(8):1064-1075.

2. Nduom EK et al. Neuro Oncol. 2016; 18(2): 195-205 3. Bloch O et al. Oral presentation at ASCO 2015. 2011. 4. Preusser M et al. Nat Rev Neurol. 2015;11(9):504-514. doi: 10.1038/nrneurol.2015.139.

Page 51: Glioblastoma vibhay

Immunotherapeutic Approaches for Glioblastoma

Page 52: Glioblastoma vibhay

52

• Out of all ongoing trials, 19 are Phase III trials*,†[1]

– All novel agents in Phase III trials are immunotherapies[1]

– Immunotherapies are the most likely adjuvant treatments to effectively target glioblastoma with minimal toxicity[2]

Immunotherapies in Phase III Trials for Glioblastoma

Trial[1] MOA Study Arms Study EligibilityNCT01765088(China)

Chemotherapy + cytokine RT Adjuvant TMZ ± IFN-α Newly diagnosed grade III

or IV glioma

NCT00045968 Vaccine DCVax-L vs placebo with standard treatment‡

Newly diagnosed, resectable glioblastoma

NCT01480479 Vaccine Rindopepimut +TMZ vs KLH+TMZ with standard treatment‡

Newly diagnosed, resectable EGFRvIII+ glioblastoma

NCT02017717 Checkpoint Inhibition

Nivolumab vs nivolumab+ipilimumab vs bevacizumab after RT+TMZ

Recurrent glioblastoma

1. Clinicaltrials.gov. “Glioblastoma” + “Interventional” + “Adult” + “Phase III” + “Recruiting” or “Active, not recruiting” search results. Accessed December 22, 2015.

2. Bloch O, Parsa AT. Neuro Oncol. 2014;16(5):758-759.

* Criteria for Phase III: Open trials for agents that are intended to directly treat tumors and that have not yet failed.

† 8 are academic/ISRs and 7 are industry-sponsored.[3]

‡ Standard therapy, involving (attempted) surgical resection and chemoradiation.[3]

DCVax-L, dendritic cell vaccine L; EGFRvIII, epidermal growth factor receptor variant III; IFN-α, interferon alfa; ISR, industry-sponsored research; KLH, keyhole limpet hemocyanin; MOA, mechanism of action; RT, radiotherapy; TMZ, temozolomide.

Page 53: Glioblastoma vibhay

53

Cell-based vaccines[1,2]:DCs pulsed with tumor cells or TAAs, or tumor cell–derived vaccines transferred back to body to induce immune response

Immunotherapeutic Approaches in Glioblastoma: Vaccines

Tumor cells DCs

Peptide-based vaccine[1,2]:Mimic TAAs or tumor-targeting peptides to induce immune response (± adjuvant)

TAAs

1. Reardon DA et al. Expert Rev Vaccines. 2013;12(6):597-615.2. Hegde M et al. Discov Med. 2014;17(93):145-154.3. Mohme M et al. Cancer Treat Rev. 2014;40(2):248-258.DC, dendritic cell; TAAs, tumor-associated antigens.

Page 54: Glioblastoma vibhay

54

Phase III, randomized, double-blind study evaluating addition of DCVax-L to the current standard of care in newly diagnosed glioblastoma[1]

DCVax-L: Phase III Trial in Newly Diagnosed Glioblastoma

N=300

SOC + DCVax-L

SOC + Placebo

Key Inclusion Criteria• Newly diagnosed, resectable

glioblastoma• Age 18–70 years• KPS ≥70• No PD at the end of RT course• Adequate BM and liver function

R2:1

Start Date: December 2006Estimated Primary Completion Date: September 2016 (Final data collection date for primary outcome measure)Status: Ongoing, but not recruitingStudy Director: Northwest Biotherapeutics

• Primary Outcome Measure: PFS• Secondary Outcome Measure:

OS and TTP

Crossover option upon PD

Cell-based vaccine

1. Clinicaltrials.gov. NCT00045968.

BM, bone marrow; DCVax-L, dendritic cell vaccine-L; KPS, Karnofsky performance status; OS, overall survival; PD, progressive disease; PFS, progression-free survival; R, randomized; RT, radiotherapy; SOC, standard of care; TTP, time to progression.

Page 55: Glioblastoma vibhay

55

ICT-107: Phase II Clinical Data

Key Efficacy and Safety Data[3,4,6] • PFS (ICT-107 vs placebo): 11.4 vs 10.1 mo• QoL (FACT-BR): Similar between groups• OS: Benefit not significant• Strongest activity observed in HLA-A2 subgroup

• No treatment-related Grade > 4 AE observed

• Trend for less steroid use in ICT-107 arm (30% vs. 44%)

• KPS maintained longer in those receiving ICT-107

• ICT-107 (ImmunoCellular Therapeutics, Ltd): Autologous DC vaccine targeting six different antigens associated with glioblastoma[1,2]

ICT-107

PlaceboR

Newly diagnosed glioblastoma

RT+TMZ TMZ or ICT-107 or placebo

Induction MaintenanceN=124

Phase II, randomized, double-blind study evaluating addition of ICT-107 to the current standard of care in newly diagnosed glioblastoma[2]

• Phase III trial design currently being finalized[4]

• Orphan drug designation in US and Europe[5]

MGMT-stratified

Cell-based vaccine

1. Reardon DA et al. Expert Rev Vaccines. 2013;12(6):597-615.2. Clinicaltrials.gov. NCT01280552. 3. Wen P et al. Oral presentation at ASCO 2014. 2005. 4. Market Watch. ICT-107. Available at: http://www.marketwatch.com/story/immunocellular-

therapeutics-presents-updated-ict-107-phase-ii-data-in-patients-with-newly-diagnosed-glioblastoma-at-the-2014-asco-annual-meeting-2014-06-01. Accessed December 17, 2015.

5. ImmunoCellular Therapeutics, Ltd. Overiew of ICT-107. Available at: http://www.imuc.com/pipeline/ict-107. Accessed December 17, 2015.

6. Wen P et al. Poster presented at ASCO 2015. 2036.

AE, adverse event; DC, dendritic cell; HLA-A2, human leukocyte antigen A2; MGMT, O6-methylguanine DNA methyltransferase; FACT-BR, Functional Assessment of Cancer Therapy –Brain ; KPS, karnofsky Performance Status; OS, overall survival; PFS, progression-free survival; QoL, quality of life; R, randomized; RT, radiotherapy; TMZ, temozolomide.

Page 56: Glioblastoma vibhay

56

• Rindopepimut (Celldex Therapeutics): EGFRvIII-targeted peptide vaccine[1]

– Three Phase II clinical trials demonstrated consistent improvement in mPFS and mOS over historical controls:

Rindopepimut: Phase II Clinical Data

Trial N Study Design Patients mPFS (mo) mOS (mo)ACTIVATE[2] 18 RT+TMZ rindopepimut

• Newly diagnosed• EGFRvIII+• GTR• No PD through CRT

14.2 26

ACT II[3,5] 22RT+TMZ rindopepimut standard or dose-intensified TMZ

Overall: 15.2

Overall: 23.6

ACT III[4,5] 65 RT+TMZ rindopepimut TMZ 12.3 24.6

Peptide-based vaccine

– Safety: Across clinical trials, rindopepimut toxicity has been generally mild and limited to skin reactions at injection site, fatigue, rash, nausea, and pruritus[2-4]

• Rindopepimut is currently being investigated in Phase III and II trials[1,5]

1. Babu R, Adamson DC. Core Evidence. 2012;7:93-103.2. Sampson JH et al. J Clin Oncol. 2010;28(31):4722-4729.3. Sampson JH et al. Neuro Oncol. 2011;13(3):324-333.4. Lai RK et al. Presentation at SNO 2011.5. Clinicaltrials.gov. NCT00643097.

CRT, chemoradiotherapy; EGFRvIII, epidermal growth factor receptor variant III; GTR, gross total resection; mo, months; mOS, median overall survival; mPFS, median progression-free survival; PD, progressive disease; RT, radiotherapy; TMZ, temozolomide.

Page 57: Glioblastoma vibhay

57

HSPPC-96: Overview and Key Data

HSPPC, heat shock protein peptide-complex; SOC, standard of care; mOS, median overall survival; mPFS, median progression free survival; MGMT, Hypermethylation of the O6-methylguanine-DNA-methyltransferase

1. Bloch O. Poster presentation at ASCO 2015. Abstract #2011.2. Clinicaltrials.gov. NCT00905060.

HSPPC-96: Autologous tumor-derived heat shock protein peptide vaccineA phase II clinical trial was designed to evaluate the addition of HSPPC-96 to SOC for newly diagnosed glioblastoma

Agenus Inc

Trial Design and Key Data[1,2]

Trial Ph Arm Patients N Key Data

Single arm[1] II

•HSPPC-96 administered at 25 µg per dose once weekly, then monthly with SOC

• Newly diagnosed GBM• ≥ 90 % tumor resection 46

• mOS = 23.8mo • mPFS = 17.8mo• PD-L1 High mOS = 18mo • PD-L1 Low mOS =

44.7mo• Adverse events = No

severe (grade 3/4) adverse events; vaccine well tolerated

Protein peptide vaccine

• Expression of PD-L1 on peripheral monocytes has been shown to be elevated in glioblastoma patients and was evaluated as a predictor of survival [1]

• A multivariate proportional hazards model revealed MGMT methylation status and PD-L1 expression as the greatest independent predictors of survival [1]

Page 58: Glioblastoma vibhay

58

Summary of Investigational Therapies and Unmet Needs

Glioblastoma Pipeline

• Most targeted agents yielding promising results in Phase I and II trials have failed Phase III trials[1,2]

• 19 Phase III trials are currently ongoing[3]

Immunotherapy for

Glioblastoma

• Immune escape mechanisms have emerged as a therapeutic target[4]

• All four novel agents in Phase III are immunotherapies[3]

• Combining treatment modalities may result in increased effectiveness[5,6]

Survival • No treatment has improved mOS over TMZ in the past 10 years[1,2]

mOS, median overall survival; TMZ, temozolomide.

1. Anton K et al. Hematol Oncol Clin N Am. 2012;26(4):825-853.2. Ohka F et al. Neurol Res Int. 2012; 2012:878425. doi:

10.1155/2012/878425.

3. Clinicaltrials.gov. “Glioblastoma” + “Interventional” + “Adult” + “Phase III” + “Recruiting” or “Active, not recruiting” search results. December 17, 2015.

4. Jackson CM et al. Clin Cancer Res. 2014;20(14):3651-3659.5. Zitvogel L et al. J Clin Invest. 2008;118(6):1991-2001.6. Drake CG. Ann Oncol. 2012;23(suppl 8):viii41-viii46.