SlideShare a Scribd company logo
PATHOLOGY, MOLECULAR BIOLOGY &
MANAGEMENT OF HIGH GRADE GLIOMA
PRESENTER- Dr HARDIK SHARMA
MODERATOR- Dr RENU MADAN
EPIDEMIOLOGY & RISK FACTORS
• Yearly incidence of malignant glioma ∼6/100 000
• Gliomas - 80% of malignant brain tumors; Glioblastoma(GBM) – most common primary malignant brain
tumor
• Slight predominance in males (1.6times)
• In a report of the Hospital based Cancer registries, 2021, CNS tumors constituted 1.6 % of all malignancies
• Exposure to ionizing irradiation
• Rare hereditary syndromes carry an increased risk for glioma: Cowden-, Turcot-, Lynch-, Li-Fraumeni
syndrome and neurofibromatosis type 1.
• Viruses like RNA ,DNA virus- HPV, Adenoviruses
• Chemical agents a. Polycyclic hydrocarbons b. Alkylating agents
EANO GUIDELINES
WHO
GRADE
DEFINITION GLIOMAS
I low proliferative potential (hypercellular) Pilocytic astrocytoma, Pleomorphic
xanthoastrocytoma, Ganglioglioma,
Subependymal giant cell astrocytoma
II +/- nuclear atypia Diffuse astrocytomas and
oligodendrogliomas
III Nuclear atypia + brisk miotic activity Anaplastic astrocytomas and
oligodendrogliomas
IV Nuclear atypia + mitotic activity + necrosis and/or
microvascular proliferation
Glioblastoma
High Grade Glioma
▪ Grade 3 and 4 astrocytoma
▪ Grade 3 oligodendroglioma
▪ Glioblastoma (IDH wild type-4)
TUMOR TYPE CNS WHO
GRADE
CHARACTERSTIC MOLECULAR
ALTERATIONS
ADULT TYPE DIFFUSE GLIOMAS
• Astrocytoma IDH Mutant 2,3,4 IDH1,IDH2
• Oligodendroglioma IDH Mutant,1p/19q
Codeleted
2,3 IDH1,IDH2,1p/19q
• Glioblastoma IDH Wildtype 4 IDH-wildtype ,Ch-7 &10,TERT,EGFR
PEDIATRIC HIGH GRADE GLIOMAS
• Diffuse Midline Glioma, H3 K27 4 H3 K27,EGFR,EZHIP
• Diffuse Hemispheric Glioma, H3 G34 4 H3 G34
• Diffuse Pediatric HGG,H3-Wildtype & IDH
wildtype
4 IDH-wildtype, EGFR, PDGFRA, MYCN
• Infant Type Hemispheric Glioma NA RTK genes
WHO HIGH GRADE GLIOMA
Pediatric Type Vs Adult Type Gliomas
• Although, both share overlying histology
• But the genetic & biology are distinctively different
• Generally pediatric type - indolent despite “anaplastic” features & lack IDH mutation & 1p/19q
codeletion i.e genetic hallmark of adult type
• They harbor genetic profiling such as MAPK pathway alteration (Pediatric type)
• Do not depend upon the patient age , instead defined based on representative molecular
alterations, implicating that pediatric-type gliomas may occur in adults & vice versa
Molecular Biology-Adult High Grade Glioma
WHO 2016 2021
Molecular marker Biological function Diagnostic roles
IDH 1 OR IDH2 Gain of function mutation Distinguishes diffuse gliomas with IDH
mutation from IDH-wild-type glioblastomas
and other IDH-wild-type gliomas
1p/19q codeletion Inactivation of TSP on 1p and 19q Distinguishes oligodendroglioma, IDH-
mutant and 1p/19q-codeleted from
astrocytoma, IDH-mutant
Loss of nuclear ATRX Cellular proliferation & promotion of
cellular longevity
Loss of nuclear ATRX in an IDH-mutant
glioma is diagnostic for astrocytic lineage
tumors
Histone H3 K27 altered Histone H3.3 missense mutation affecting
epigenetics
Defining molecular feature of diffuse midline
glioma, H3.3 G34-mutant
Histone H3.3G34 altered Histone mutation affecting epigenetic
regulation
Defining molecular feature of diffuse
hemispheric glioma, H3.3 G34-mutant
MGMT promoter
methylation
DNA repair None, but is a predictive biomarker of benefit
from alkylating chemotherapy in patients
with IDH-wild-type glioblastoma
CDKN2A/2B homozygous
deletion
Encode CDKNA2A &2B ,regulators of
RB17 p-53 dependent signaling
A marker of poor outcome and WHO grade 4
disease in IDH-mutant astrocytomas
EGFR amplification Cell proliferation ; invasion & resistance to
induction of apoptosis
EGFR amplification occurs in ~40–50% of
glioblastoma, IDH wild type Molecular
marker of glioblastoma, IDH wild type,
WHO grade 4
TERT promotor mutation Cell proliferation; promotes cellular
longevity by increasing TERT expression
OCCUR IN -70% OF Glioblastoma,IDH
wild-type & >95% oligodendroglioma,IDH-
mutant
Molecular marker of glioblastoma wild-4
+7/-10 cytogenetic
signature
Gain of ch7 combined with loss of ch10 Molecular marker of glioblastoma, IDH wild
type, WHO grade 4 (
BRAFv600E mutation Oncogenic driver mutation leading to
MAPK pathway activation
Rare in adult diffuse gliomas but amenable to
pharmacological intervention
IDH1/2 mutation 1p/19q co-deletion MGMT promoter
methylation
Diagnostic Role DD glioma versus gliosis
Typical for transformed low-grade glioma
Pathognomonic for oligodendroglioma None
Prpgnostic Role Protracted natural history in IDH-mutated
tumours
Protracted natural history in 1p/19q
codeleted tumours
Prognostic for anaplastic glioma patients
(possibly with IDH mutations) treated with
RT or alkylating drugs
Predictive Role Absence of mutation suggests predictive role
for MGMT promoter methylation
Prolongation of survival with early
chemotherapy in 1p/19q-co-deleted
oligodendrogliomas
Predictive in GBM for benefit from
alkylating chemotherapy.Elderly GBM:
MGMT-methylated→TMZ; MGMT
unmethylated→ RT
Frequency:WHO-II
Diffuse Astrocytoma 70-80% 15% 40-50%
Oligodendroglioma 70-80% 30-60% 60-80%
Who G-III
Anaplastic
Astrocytoma
50-70% 15% WHO G-II
Anaplastic
Oligodendroglioma
50-80% 50-80% 70%
Who G-IV
Glioblastoma 5-10% <5% 35%
CLINICAL RELEVANT MOLECULAR MARKERS
Location & Spread Of Glioma
• Approximately 60% of gliomas are located in one of the cerebral hemispheres
 Frontal lobe(25.6%)>temporal lobe (19.6%)> parietal lobe (12.6%)
Glioblastoma multiforme is uncommon in the region of third ventricle(<1%) and rarely occurs
in posterior fossa
Although most of the Glioblastoma are centered in the deep white matter
Epicenter at the gray-white matter junction-10%
• Malignant gliomas tends to spread or recur in adjacent brain region along white matter tracts
• Spread outside the central nervous system is extremely rare
• Rarely , gliomas may spread systemically
History and Clinical Examination
• Clinical presentation: new onset epilepsy, focal deficits, including neurocognitive impairment,
and indicators of intracranial mass effect, such as headache, vomiting, and altered consciousness.
• History: duration of neurological symptoms and signs, familial risk or exogenous risk factors,
including exposure to radiation
• Physical examination: focuses on the detection of systemic cancer and contraindications for
neurosurgical procedures. Neurocognitive assessment, performance status should be documented.
▪ MMSE is widely used as a screening instrument to detect neurocognitive impairment
Imaging Of Brain
▪ MRI – gold standard
T2-weighted and fluid-attenuated inversion recovery sequences, and T1-weighted sequences before and after
gadolinium use, in at least one plane, is the standard method for the detection of a glioma
▪ MR Spectroscopy: differentiating tumor from radiation necrosis, grading tumors, assessing response; area
most abnormal → biopsy
▪ MR Perfusion: differentiating grade of tumor or tumor versus radiation necrosis; area of highest perfusion
→ biopsy
▪ CT: patients who cannot have MRI, shows calcification
▪ Amino acid PET helps define metabolic hotspots for biopsy
▪ Intra-arterial angiography can aid the surgical strategy
Diagnostic Characteristics
Tumor Imaging Characteristics
Glioblastoma
Multiforme
• Enhancing MRI or CT lesion, hypodense interior, often with associated
edema & necrosis
• Angiography: highly abnormal vasculature with arteriovenous shunting and
early venous drainage.
Anaplastic
Astrocytoma
• CT- inhomogeneous density
• T2-weighted FLAIR MRI - hyperintense, space-occupying lesions, patchy
enhancement after contrast administration and often have peritumoral
edema.
• Up to 30% can show no enhancement on CT or MRI.
• Abnormal vessels can be visualised by MRI angiography
Oligodendroglioma • Heterogeneous lesion that may or may not enhance on MRI or CT,
frequently
with calcification, cystic regions, or hemorrhage
58-year-old with a glioblastoma who presented with gait disturbance
ADC
FLAIR T1C DWI
MRSPECTROSCOPY
Increase in choline & decrease NAA level in glioma Lactate level indicate necrosis but non specific
ANAPLASTIC
ASTROCTOMA
OLIGODENDROGLIOM
A
TREATMENT
• Surgery
• Radiotherapy
• Chemotherapy and Targeted Therapy
• Symptomatic management
Standard of care
Maximal surgical resection
+
Postoperative radiotherapy
and
Chemotherapy
Surgical Management
• Histological diagnosis for all patients - biopsy or decompression surgery unless there is a
specific contraindication
• Use of neuronavigation systems, Laser Interstitial Thermal Therapy, use of fluorescent
tumor localizing dyes such as 5-aminolevulinec acid
Type of resection Extent of resection
Biopsy <10%
Partial 10-50%
Subtotal 50-90%
Near-total >90%-94%
Gros -total >95%
▪ Microsurgical resection of a right-sided recurrent IDHwt glioblastoma WHO grad IV using intraoperative
neuronavigation, neuromonitoring and 5-ALA fluorescence techniques.
▪ A: T1CE axial, sagittal and coronal planes including DTI fiber tracking (blue fibers). The green trajectories /
red points represent the pointer for intraoperative neuronavigation.
▪ B: Upper image: corresponding intraoperative 5-ALA fluorescence image taken from the area as depicted
by neuronavigation. Lower image: opening of the right ventricle due to critical involvement by tumor
formation.
▪ C: Postoperative MRI confirms gross total resection without residual contrast enhancement, no
perilesional ischemia (diffusion-weighted image upper right).
Influence of location and extent of surgical resection on survival of patients with
glioblastoma multiforme: results of three consecutive Radiation Therapy
Oncology Group (RTOG) clinical trials. Simpson et al , IJROBP 1993
o Significant correlation of age, Karnofsky Performance Status, extent of surgery, and primary site with survival.
There was no statistical difference in survival based on tumor size.
o The best survival rates occurred in patients who had at least three of the following features:
• < 40 years of age
• high Karnofsky Performance Status(80-100)
• frontal tumors, and
• total resection (17 months median)
Procedure Median survival
Total resection 11.3 months
Partial resection 10.4 months
biopsy 6.6 months
Goals of surgery: maximal safe resection
Diagnosis Alleviate symptoms
Decrease need for steroids Increase survival
• Early postoperative MRI (within 48 hours) -
confirms the degree of resection and can help with
radiotherapy planning ( to assess for the revision
surgery)
• Patients should be re-discussed at the specialist MDT
with histology and general performance status
details available.
• Patients should be staged using the WHO system.
• Adjuvant treatment should be commenced within 2
weeks of biopsy or 4 weeks of decompression
surgery.
MEDICAL MANAGEMENT
• ANTIEPILEPTIC DRUGS
Non Enzyme inducing anti seizure medications should be used eg: levetiracetam, topiramate,
valproic acid, lacosamide
Close monitoring for side effects
No role for primary perioperative prophylaxis (i.e. in patients who have never had a seizure)
Important to know about prior history of seizure
• STEROIDS
Corticosteroids, preferably dexamethasone (in conjunction with gastric protection if used at high doses) -
reduce symptomatic peritumoral vasogenic edema, neurological deficits and signs of increased
intracranial pressure
Lowest dose of steroids – shortest time possible
Downward titration of dose – whenever possible
Extensive mass effect – receive steroids for at least 24 hours before RT
Chronic corticosteroids (≥ 20 mg prednisone equivalents daily for ≥ 1 month) - consider prophylaxis for
osteoporosis and pneumocystis jerovecii pneumonia should be considered
RPA Score Based Prognosis
Class Patient Characterstics Median Survival
(Months)
I, II Astrocytoma
Age ≤50 yr, normal mental status or
age >50 yr
• KPS >70, symptoms >3 mo
40-60
III, IV Astrocytoma
• Age ≤50 yr, abnormal mental status
• Age >50 yr, symptoms <3 mo
Glioblastoma
• Age <50 yr
• Age >50 yr, KPS ≥70
11-18
V, VI Glioblastoma
• Age >50 yr, KPS <70 or abnormal
mental status
5-9
• Evaluated protein biomarkers + clinical variable (age, KPS,
extent of resection, neurologic function)
• Ki-67, c-Met and MGMT found to be significant biomarkers
Class Median OS (months)
I 21
II 16.6
III 9.4
High Grade Glioma
Adjuvant treatment Preferred Regime Other Recommended
Regime
Oligodendroglioma (3)
(KPS > 70)
• RT with adjuvant PCV
• RT with neoadjuvant PCV
• RT with adjuvant and
concurrent TMZ
• RT with adjuvant TMZ
Astrocytoma (3,4)
(KPS > 70)
• RT with adjuvant and
concurrent TMZ
• RT followed by adjuvant
TMZ
• RT with adjuvant PCV
• RT with neoadjuvant PCV
High Grade glioma (KPS < 70) Single modality:
• RT (hypofractionation
preferred) or
• TMZ (if tumor is MGMT
promoter methylated)
EANO GUIDELINES
Glioblastoma
Preferred Regime Other Recommended
Regime
Useful In Certain
Circumstances
Adjuvant
treatment
RT with concurrent and
adjuvant TMZ + TTF
• RT with concurrent and
adjuvant TMZ ( age < 70y and
KPS <60)
• TMZ (if tumor is MGMT
promoter methylated and KPS
<60 or age >70 and KPS > 60)
• RT with conc and adj lomustine
and TMZ (if tumor is MGMT
promoter methylated, KPS >
60, and age < 70y)
Survival Curves Of Patients Who Received: Median Survival
A) Best conventional care but no radiotherapy
or chemotherapy
14 weeks
B) BCNU 18.5 weeks
C) Radiotherapy 36 weeks
D) BCNU and radiotherapy 34.5 weeks
Walker et al 1978
• Radiotherapy significant influence on the
survival of patients
• The specific relationship between 50 & 60Gy
indicates a 1.3 times increase in median life
span associated with the higher dose (p =
.004)
RT Dose Median survival
No 18 weeks
45Gy 13.5 weeks
50Gy 28 weeks
55Gy 36 weeks
60Gy 42 weeks
1979
• 1.Age 18 - 70 y
• 2.Randomized 1:1
• Partial brain RT 60 Gy
• Partial brain RT 60 Gy / TMZ → TMZ
• 3.Improved Median survival
• 12.1 → 14.6 months
• P < 0.001
• 4.Improved 5 yr Survival
• 1.9% → 9.8%
• 5.More Grade 3-4 haematological toxicity
with TMZ i.e 7% only
2005
MGMT methylation status- EORTC PHASE III
• Regardless of treatment, MGMT methylation was a favorable
prognostic factor (p<0.0001)
• MGMT methylation strongest predictor outcome: 23 mo vs 13
mo
• MGMT methylated cases
• MS was 21.7 vs 15.3 mo for chemoRT vs RT alone
• MGMT un-methylated cases:
• Difference in overall survival favoring the temozolomide-plus-
radiotherapy group was only marginally significant (p=0.06 )
• MS was 12.7 vs 11.8 mo for chemoRT vs RT alone
2009
2019
• GBM with methylated MGMT
• 60Gy + con & adj TMZ Vs 60Gy + TMZ+Lomustine
• Median OS 31mo Vs 48 mo (p=0.0492)
• Median PFS 16.7mo, not different
• Arm 1 XRT with TMZ & Bevacizumab Vs Arm 2 XRT with placebo & TMZ
• Addition of BEVACIZUMAB significantly prolonged PFS survival (p<0.001)
• Median PFS 10.6 months vs 6.2 months
• However, the median OS was not significantly different in both arms
• Rates of adverse events was higher with bevacizumab than placebo
2014
TUMOR TREATING FIELDS
• the placement of noninvasive transducer arrays on the scalp and the delivery of low intensity (1 to 3 V/cm),
intermediate frequency (100 to 300 kHz), alternating electric fields to the tumor.
• Lead to tumor cell kill during
• 1. metaphase - disrupting the formation of the mitotic spindle, and
• 2. cytokinesis - dielectrophoretic dislocation of intracellular constituents resulting in
apoptosis.
• Most effective - applied in the direction of the division axis of the dividing cell, and therefore, two sequential
field directions are applied to tumors by using two perpendicular pairs of transducer arrays in order to increase
the efficacy of TTFields.
TUMOR TREATING FIELDS
o 1. KPS > 70; median age 56
o 2. Randomized 2:1 after 60Gy, TMZ
Adj TMZ + TTF
Adjuvant TMZ
o 3. Improved median survival from time
of randomization
16→20.9 mth P<0.001
o 4. Toxicity – dermatitis in 52%
o 5. Improved OS: >18 hours/day
ELDERLY GLIOBLASTOMA
NCIC (Roa JCO
2004)
• Age>60, KPS >50
• 60Gy/30# vs 40
Gy/15#
• No diff in MS:
5.1 vs 5.6 mth
Nordic Trial (Malmstrom
Lancet 2012)
• Age > 60y, GBM
• 34Gy/10# was not
inferior to 60Gy/30#
• Methylated MGMT
associated with better
survival for TMZ for
age > 70y
NOA-08 (Wick, lancet, 2012)
• Age >65y, KPS > 60, AA,
GBM
• TMZ(100mg/m2days 1-7 of
1 week on, 1 week off
cycles) VS 60 Gy/30# RT
• 8.6 vs 9.6 month median
survival
• MGMT Unmethylated – EFS
longer with RT
• MGMT methylated – EFS
longer with TMZ
CCTG/EORTC/TROG (Perry
NEJM 2017)
• Age > 65 y, ECOG PS 0-2
• 40 Gy/15# RT/TMZ → TMZ
better than 40 Gy/15# RT alone
• 9.3 vs 7.6 months median survival
• MGMT Unmethylated – 10 mth vs
7.9 mth
• PFS – 5.3 mth vs3.9 mth
• 1 yr survival rate 37.8% vs 22.2%.
• Arm 1 (25Gy/5#/5days) Vs Arm 2
(40Gy/15#/3.3weeks)
• Median OS (7.9 mo) Vs (6.4 mo)
• PFS- 4.2 mo Vs 4.2 mo
2015
Treatment Scheme For Elderly HGG
• RT with conc Temozolamide vs RT alone
• OS was 146 days vs 121 days (p=0.146)
• PFS was 109.5 days vs 77 days (p=0.028)
• Dose- 25GY/5#/5days
Minimizing the uncertainties regarding the effects of delaying radiotherapy for
Glioblastoma: A systematic review and meta-analysis Loureiro et al Radiother Oncol 2016
No evidence of a true effect on OS by delaying RT in GBM patients ( range 20-86 days)
Postoperative neoadjuvant temozolomide before radiotherapy versus standard
radiotherapy in patients 60 years or younger with anaplastic astrocytoma or
glioblastoma: a randomized trial
Malmstorm et al ActaOncologia 2017
GBM or AA, age < 60 y,
PS 0-2
2-3 cycles TMZ → RT RT only
Median
OS
GBM + AA 17.7 months 20.3 months
AA 95.1 months 35.2 months
Astrocytoma Grade 3-4
• Anaplastic astrocytomas, IDH-mutant, are the most common WHO grade III astrocytomas.
• Standard treatment includes maximal safe removal followed by radiotherapy and chemo
• RT with concurrent TMZ ~ RT with concurrent nitrosureas
• Slightly better PFS with TMZ
Grade-III GLIOMA (noncodeleted): Interim CATNON
1:1:1: randomization
• RT(59.4Gy)
• RT with concTMZ
• RT TMZ
• RT/TMZ 12 TMZ
Adjuvant TMZ improved OS
• OS- (116mo) (21mo)
IDH mutant but not in IDH wild
• Benefit from adj TMZ OS(82.3 mo) Vs
(47mo)
• No OS benefit conc TMZ
• Hematological toxicity- 8-12% but reversible
Oligodendroglioma Grade 3
• Relatively rare
• 1p19q codeleted and IDH1-mut or IDH2-mut
• Better prognosis than other high grade gliomas
• PCV added to RT
• CODEL trial ongoing: RT (59.4 Gy) + adj PCV vs RT + conc TMZ + adj TMZ ( result
pending)
59.4 Gy/33# of RT 59.4 Gy/33# of RT → six cycles of adjuvant PCV
Median OS 30.6 months 42.3 months
Median PFS 13.21 months 24.3 months
Improved OS in pts with MGMT methylation and IDH mutations.
Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402
Cairncross et al JCO 2013
59.4 Gy/33# of RT PCV(4cycles) → RT
Median Survival 4.7 years 4.6 years
OS - 1p/19q codeleted AO/AOA 7.3 years 14.7 yrs
PFS - 1p/19q codeleted AO/AOA 2.9 y 8.4 y
OS - 1p/19q non codeleted AO/AOA 3.3 y 5.5 y
PFS - 1p/19q non codeleted AO/AOA 1 y 1.2 y
Long-term follow-up results of EORTC 26951: A randomized phase III study on adjuvant PCV chemotherapy in
anaplastic oligodendroglial tumors (AOD).
Van Den Bent et al JCO 2012
Temozolomide
• oral DNA alkylating agent
• good blood–brain barrier penetration
• favourable safety profile - myelosuppression,
notably thrombocytopenia dose-limiting toxicity
Nitrosoureas
• second choice after temozolomide for
glioma treatment
• higher efficacy of procarbazine, lomustine,
and vincristine over temozolomide in
patients with anaplastic glioma with a good
prognosis.
• Side effects: prolonged leukopenia and
thrombocytopenia
• Pulmonary fibrosis with carmustine
• Higher rates of discontinuation due to
toxicity
• Temzolamide is an alkylating agent &
result in DNA damage with displacing
alkyl group in different DNA positions
• MGMT plays an important role in DNA
repair enzymes by synthesizing repair
proteins
• MGMT promoter methylation results in
reduced expression of this gene &
increase response to alkylating agents
• In patients with GBM, OS & prognosis remains
dismal
• Use concurrent & adjuvant TMZ to
maximize the benefit of post-op
radiotherapy
• Patient with poor KPS score , considered for
hypofractionated RT, have similar median
survival rates as conventional RT
2013
RTOG 0525: A randomized phase III trial comparing standard adjuvant temozolomide
(TMZ) with a dose-dense (dd) schedule in newly diagnosed glioblastoma (GBM). Gilbert et al,
JCO, 2006
• Arm 1: standard TMZ (150-200 mg/m2 body surface area days 1 to 5 of 28)
• Arm 2: intensified regimen (75-100 mg/m2 body surface area days 1 to 21 of 28) for 6-12 cycles.
• Conclusion:
o Validated MGMT-promoter methylation as a favorable prognostic factor regardless of treatment.
o MGMT methylation was associated with improved OS (21.2 v 14 months), PFS (8.7 v 5.7
months)
o Dose-dense temozolomide was more toxic than standard dosing and did not significantly alter
PFS or OS regardless of MGMT status.
Is more better? The impact of extended adjuvant temozolomide in
newly diagnosed glioblastoma: a secondary analysis of EORTC and
NRG Oncology/RTOG
Blumentha et al, NeuroOncol, 2017
Conclusion:
• Treatment with more than 6 cycles of TMZ was associated with a somewhat improved PFS
in particular for patients with methylated MGMT
• OS was not affected by the number of TMZ cycles including the MGMT methylated
subgroup(p=0.52)
• Treatment with 6 cycles of TMZ was associated with improved PFS (p=0.03)
• Better in MGMT methylated patients
• Overall survival was not impacted by the number of TEMOZOLAMIDE
• C-TMZ Vs E-
TMZ(15.4mo Vs
23.8mo) p=0.044
• PFS (12.8mo
Vs16.8mo)
• 5% Vs 15%
2017
Radiotherapy Techniques
Stereotactic
Radiosurgery
Conventional
2D
3DCRT
Brachytherapy
Proton Beam
Therapy
Conventional 2D Approach- Whole Brain RT
• Position- supine
• Ask to patient to stare up straight to the roof
• Draw a line along the pupillary line on the
forehead
• Draw a line joining the lobule of the ear & the
lateral canthus of the eye & extend it to the
former line
• Rest of the two borders are flashed 1cm in air
Fluoroscopic simulation
▪ Left and right parallel oppose lateral field
▪ Anterior temporal lobe ,cribriform plate, posterior aspects
of eyes included
▪ Inferior field edge passing through C1-C2 junction &
base of skull
▪ Safety margin of at least 1cm ( flashed in air)
▪ Placing a radiopaque marker on both lateral canthus
Limitation Of Conventional Planning
▪ Irradiation of large volumes of brain with normal tissue also
▪ Higher toxicity and side effects
▪ Lack of 3D visualization of tumor
▪ 2D planning of 3D tumor
Radiotherapy for newly diagnosed malignant glioma in adults: a systematic review
The Cancer Care Ontario Practice Guidelines Initiative Neuro-Oncology Disease Site Group1
Laparriere et al, 2001
No significant difference in survival rates for whole brain radiation versus more local fields that
encompass the enhancing primary plus a 2 cm margin
• 80-90% of recurrence is local ( within 2cm of enhancing tumor on CT)
• Radionecrosis- 50% of patients treated with WBRT had grade3-4 white matter changes as
compared with 14% of patients treated only to local field
• 3DCRT makes partial brain irradiation for glioma possible & reduces neurotoxicity
• It may be recommended for multifocal disease , but it is rare and failure occur typically within
original disease
SIMULATION – CONFORMAL RT
▪ CT simulation supine with the head in a neutral
position
▪ Head immobilised in an individual Perspex or
thermoplastic shell
▪ Slice thickness: 2.5-3mm
▪ Postop MR fusion for better tumor delineation
▪ RT should start within 4-6 weeks of surgery
• Mean GTV increased by 46%
between early CECT and MRI,
the difference was only 4%
between delayed CT & MRI
• Delayed CECT is superior to
early CECT with MRI for target
delineation
EARLY CT DELAYED CT
POST OP MRI
Contouring
guidelines
MDACC
RTOG
EORTC
NRG
ORGAN AT RISK
▪ LENS
▪ B/L OPTIC NERVE
▪ B/L EYE
▪ OPTIC CHIASM
▪ BRAINSTEM
▪ B/L TEMPORAL LOBE
▪ SPINAL CORD
EORTC treatment volumes
(EORTC 22981/22961,
RTOG treatment volumes (RTOG 0525, 0825, 0913, and AVAglio
trials)
Phase 1 (to 60 Gy in 30 fractions) Phase 1 (to 46 Gy in 23 fractions) Phase 2 (14 Gy boost in 7 fractions)
GTV surgical resection cavity
plus any residual
enhancing
tumour (postcontrast T1
weighted MRI scans)
GTV1 surgical resection cavity plus any residual
enhancing tumour (postcontrast T1
weighted MRI scans) plus surrounding
oedema (hyperintensity on T2 or FLAIR
MRI scans)
GTV
2
surgical resection cavity
plus any residual
enhancing tumour
(postcontrast T1 weighted
MRI
scans)
CTV GTV plus a margin of 2
cm*
CTV1 GTV1 plus a margin of 2 cm (if no
surrounding oedema is present, the CTV is
the contrast enhancing tumour plus 2.5
cm.
CTV
2
GTV2 plus a margin of 2
cm
PTV CTV plus a margin of 3–
5 mm
PTV1 CTV1 plus a margin of 3–5 mm PTV2 CTV2 plus a margin of 3–
5 mm
TARGET DELINEATION OF GLIOBLASTOMA
*Margins up to 3 cm were allowed in 22981/22961 trial, and 1.5 cm in 26981–22981 trial.
RTOG EORTC
MDACC GUIDELINES
• INITIAL PHASE (50 Gy):
GTV = Resection cavity and any residual
contrast-enhancing tumor(T1) but ignoring any
edema
CTV = GTV + 2 cm isotropic margin
Peritumoral edema was not intentionally
included in the CTV except for edema existing
within the 2-cm margin
PTV = CTV + 0.5 cm margin
• BOOST PHASE (10 Gy): PTV = GTV + 0.5cm
margin
NRG – Contouring Guidelines
• Careful delineation
of anatomic
pathways and
barriers to spread
can spare radiation
to uninvolved tissue
without
compromising target
coverage
GTV & GTV BOOST CTV & CTV BOOST PTV & PTV BOOST
GTV- RED & GTV BOOST- PINK CTV- RED & CTV BOOST- PINK PTV- RED & PTV BOOST- PINK
PGI PROTOCOL
Dose constraint of conventional 3D-CRT
OAR OBJECTIVE(S) End point
Brainstem Dmax<54 Gy; 1–10 cc < 59 Gy (periphery) Neuropathy (<5%) or necrosis
Optic Chiasm Dmax <55 Gy Optic neuropathy (<3%)
Cochlea Ideally one side mean <45 Gy ( Most protocols allow ipsilateral cochlea to
receive 60 Gy rather than compromise dose )
Sensorineural hearing loss
(<30%)
Eyes Macula <45 Gy Retinopathy
Lens Ideally <6 Gy Max 10 Gy
dose limits should never compromise PTV dose
Cataract
Optic nerves and
chiasma
Dmax <55 Gy or 1 % of PTV cannot exceed 60 Gy Optic neuropathy (<3%)
Brain Dmax <60Gy Symptomatic necrosis
(<3%)
Hippocampus Dmax ≤6 Gy & V3Gy ≤ 20% Cognitive impairment
Spinal cord Dmax <50Gy Grade >2 myelopathy (<1%)
• No significant difference in the mean PTV(initial) among the groups (p = .24), whereas a
significant reduction in mean PTV boost was observed with the MDACC protocol compared
with the RTOG protocol
• No statistically significant differences were observed in treatment-related acute toxicities and
steroid requirements between both arms
• Central recurrences: commonest in both groups
• Statistically significant differences in QoL favoring those treated with MDACC protocol
RTOG VS EORTC
• Perfusion- and diffusion weighted MRI potentially add information about the regional blood
volume and microstructural architecture of the high-grade glioma.
• No difference in progression-free or overall survival was observed
• Larger PTVs failed to produce a significant reduction in marginal or distant recurrences
3DCRT VS IMRT/VMAT
• Sparing of normal brain
• Local boosting of tumor alone to higher doses
• IMRT especially useful in presence of critical structures close to- optic nerve, brainstem, dose
escalation , etc
• IMRT did not significantly improve target coverage compared with 3D-CRT
• However, it resulted in a decreased Dmax to the spinal cord, optic nerves, and eye by 16%, 7%,
and 15%
• It is unlikely that IMRT will improve local recurrence without dose escalation However, it might
result in decreased late toxicities associated with radiotherapy
• VMAT achieved equal or better PTV coverage and OAR sparing while using fewer monitor units
• A-68Ga-Pentixafor fused PET increased uptake in LT
F-T region with GBM
• B- IHC evidence of CXCR-4 expression on the tumor
• The affinity of this tracer help in developing
CXCR4 targeting radiotherapeutics
• A- 38 yr male pt with GBM radiological
evidence of residual disease
• B- Repeat scan post 3month (post-op) showed
disease progression
• GBM patients 60Gy/30# Vs 67Gy/30# ( boost 21Gy/7#)
• Slightly higher median OS as compared to historical data that dose escalation is feasible
First image- PENTIXAFOR PET show active disease Third image- Phase 1 dose color wash
Second image- Target volume delineation Fourth image- dose color wash including SIB
• Pt diagnosed with grade (1-4) glioma
• Proton vs Photon therapy
• PBT associated with improved OS (45.9mo vs
29.7 mo ) to XRT
• Retrospective analysis supports ongoing
prospective trials comparing the two modalities
2018
EXPECTED SURVIVAL OF GLIOMA
Nat Rev Neurol. Author manuscript; available in PMC 2020 June 10
HIGH GRADE GLIOMA MEDIAN SURVIVAL
Oligodendroglioma - IDH mutant ( 1p/19q codeletion) 17.5 years
Oligodendroglioma - IDH mutant ( no 1p/19q codeletion) 6.3 years
Oligodendroglioma – IDH wild 1.6 years
Astrocytoma - IDH mutant 9.3 years
Astrocytoma - IDH wild 1.9 years
Glioblastoma - IDH mutant 3.6 years
Glioblastoma - IDH wild 1.2 years
Glioblastoma MGMT methylated 21.2 mo
Glioblastoma MGMT unmethylated 14 mo
PSEUDOPROGRESSION
• Radiochemotherapy and immunotherapy - transient worsening of contrast enhancement on MRI for
several months in approximately 10-30% of patients, sometimes associated with symptoms of intracranial
mass-effect
• Diagnosis - challenging
 Dynamic susceptibility contrast–enhanced MRI: neoangiogenesis → accompanies neoplastic growth;
treatment-related changes increase vascular permeability without neoangiogenesis.
 Amino-acid PET imaging may be helpful: brain tumours - increased protein synthesis; amino acid
tracers →imaging modality.
A 31-year-old male patient with prior
resection of a right frontal
glioblastoma multiforme presented
later with a 1-cm enhancing nodule
concerning for recurrence compared
with pseudoprogression
A. Magnetic resonance image of the
new nodule.
B. Map of relative cerebral blood
volume demonstrates increased
flow in the nodule (arrow).
C. Spectroscopic imaging
demonstrates an elevated choline
peak compared with
D. Normal contralateral brain
parenchyma in the same patient
DSC perfusion in pseudoprogression & progressive disease
A. T1C & rCBV map show enhancing lesion in surgical bed after RT in a patient of GBM , white arrow
show rCBV 0.5 in comparison with c/l lobe indicating (Pseudoprogression)
B. T1C & rCBV map show enhancing lesion close to the margin of radiation field. White circle show rCBV
2.8 in comparison with c/l lobe indicating predominance malignant cells ( Progressive disease)
56 year old male – GBM – Pseudoprogression
A. T1 contrast MRI 1 week after an operation show ring enhancement
B. After RT, marked enhancement in the margins following which he developed rt sided hemiparesis
C. At that time T1 MRI showed ^se in mass effect , midline shift which turned to soap bubble
appearance- radiation necrosis
D. Pt improved clinically & after 4months, fading of the enhancing lesion & decrease in mass effect
RANO Criteria
▪ 12 weeks post-RT : progression can only be determined if the majority of the new enhancement is
outside 80% Isodose line or with pathologic confirmation
RANO criteria CR PR SD PD
T1 enhancing disease None >50% decrease <50% if but < 25%
if
> 25%
T2/FLAIR Stable/decreased Stable/decreased Stable/decreased increase
NEW LESION None None None Present
CORTICOSTEROID
USE
None Stable or Stable or NA
CLINICAL STATUS Stable/improved Stable/improved Stable/improved Declined
Management of Recurrence
• Surgical excision
• Chemotherapy- Temozolamide/Carmustine/Lomustine
• Bevacizumab-FDA approved
• Alternating Electrical field therapy
• Re-irradiation
• Locally delivered Carmustine (Gliadel) Wafers
Implanted into surgical cavity – modest survival advantage with newly diagnosed ¾
Gliomas, or recuurent glioblastoma
Patients without systemic treatment options
Requires careful patient selection & a gross total resection
Studies in GBM have shown only marginal benefit
REIRRADIATION
▪ Young patient
▪ Good KPS score
▪ Long interval since previous RT > 6months
▪ Previous dose should be know
▪ Tumor size and location should be taken into
account when deciding whether re-irradiation
would be safe
• ReRT(35GY/10#) and conc bevacizumab Vs bevacizumab(10mg/kg) alone
• First prospective randomized multi-institutional study
• Median PFS 7.1 Vs 3.8 months (p=0.05)
• Median OS was similar
• Overall, ReRT was shown to be safe and well tolerated
• Re-irradiation of brain tumors – increasingly
considered because of developments in
radiation technology & imaging make high
accurate targeting of recurrent tumors
▪ Further, exploration of the role of re-irradiation
on its own or in combination with novel agents
is needed
Systematic review 2016
Stereotactic Radiosurgery
• SRS: delivered using a conventional or modified LINAC, a Gamma Knife (Elekta Corp., Stockholm,
Sweden) or a robotically controlled miniaturized linear accelerator (CyberKnife).
• Radiosurgery appears to be a reasonable option for small well-circumscribed high-grade gliomas that
recur after prior conventional large-field radiotherapy and chemotherapy
• For lesions larger than 4 cm and/or located in critical regions - Fractionated Stereotactic
Radiotherapy
• KEY REQUIREMENTS FOR OPTIMAL STEREOTACTIC IRRADIATION:
i. Small target/treatment volume
ii Sharply defined target
▪ Accurate radiation delivery
▪ High conformality
▪ Sensitive structures excluded from target
Properties
Brachytherapy & Radiocolloid Solutions
• Selection criteria for brachytherapy:
i. small size (<5 to 6 cm),
ii. tumor confined to one hemisphere,
iii. no transcallosal or subependymal spread,
iv. well circumscribed on CT or MRI, and
v. accessible location for the implant.
• A balloon based system, GliaSite, placed into the cavity at the time of surgery - recurrent malignant gliomas
whose largest spatial dimension is <4 cm and are roughly spherical.
• After treatment planning, the balloon is filled with a liquid that contains organically bound I125 and treatment is
completed within 3 to 7 days.
• 3D Ir192 HDR BT (Ir Knife) allows intratarget dose escalation with superior conformity.
• Stereotactic 125I brachytherapy with TMZ chemotherapy was shown to be effective for treatment of
thalamic glioblastoma
Targeted Therapies For Recurrent Glioblastoma
EORTC 26101 phase III trial exploring the combination of bevacizumab and
lomustine in patients with first progression of a glioblastoma
• Lomustine (90mg/m2) every six weeks vs lomustine & bevacizumab(10mg/m2) every two
weeks f/b best investigation
• Median OS 9.1 months vs 8.6 months (p =0.625)
• PFS was longer with addition of bevacizumab 4.2 vs 1.5 months
• Bevacizumab with progressive glioblastoma despite PFS does not confer a survival advantage
• Methlyated group showed a longer median OS 13.5 mo vs unmethylated group was 8 mo
Vaccine & immune checkpoint inhibtors for glioblastoma
• DCVax- a dendritic cell (DC)-based personalized cancer vaccinewith purified tumor-specific antigens
or tumor cell extracts derived from tumor at the time of resection.
• Epidermal Growth Factor Receptor Variant III as a Vaccine Target for Glioblastoma
• Heat Shock Protein-Based Vaccine
• Recombinant nonpathogenic polio-rhinovirus chimera (PVSRIPO) targets the neurotropic
poliovirus receptor CD155, which is abundantly expressed on glioblastoma cells.
• Phase-III trial of NIVOLUMAB vs BEVACIZUMAB failed to show any survival difference between
the two arms
CONCLUSION
• Molecular markers are well established in adult and pediatric type gliomas
• IDH1 mutation is a strongest prognostic marker
• There is no definite treatment guidelines for the pediatric type high grade glioma til
• Despite of molecular markers, standard of treatment remains same
• PCV regimen is more sensitive for oligodendroglial tumor as compared to TMZ
• Role of bevacizumab is established in reccurent glioblastoma
THANK YOU

More Related Content

What's hot

MANAGEMENT OF MENINGIOMA
MANAGEMENT OF MENINGIOMAMANAGEMENT OF MENINGIOMA
MANAGEMENT OF MENINGIOMA
Kanhu Charan
 
Embrace ii protocol
Embrace ii protocolEmbrace ii protocol
Embrace ii protocol
GloribelBolvar1
 
Radiotherapy planning for rectal cancer ,2D updates!
Radiotherapy planning for rectal cancer ,2D   updates!Radiotherapy planning for rectal cancer ,2D   updates!
Radiotherapy planning for rectal cancer ,2D updates!
Gebrekirstos Hagos Gebrekirstos, MD
 
Radiosurgery for brain metastases
Radiosurgery for brain metastasesRadiosurgery for brain metastases
Radiosurgery for brain metastases
Robert J Miller MD
 
High grade glioma kiran
High grade glioma  kiranHigh grade glioma  kiran
High grade glioma kiran
Kiran Ramakrishna
 
Contouring guidelines Cervix IMRT
Contouring guidelines Cervix IMRTContouring guidelines Cervix IMRT
Contouring guidelines Cervix IMRT
Debarshi Lahiri
 
Hippocampal sparing whole brain radiation therapy- Making a case!
Hippocampal sparing  whole brain radiation therapy- Making a case!Hippocampal sparing  whole brain radiation therapy- Making a case!
Hippocampal sparing whole brain radiation therapy- Making a case!
VIMOJ JANARDANAN NAIR
 
LUNG SBRT A LITERATURE REVIEW
LUNG SBRT A LITERATURE REVIEWLUNG SBRT A LITERATURE REVIEW
LUNG SBRT A LITERATURE REVIEW
Kanhu Charan
 
Plan evaluation in Radiotherapy- Dr Kiran
Plan evaluation in Radiotherapy- Dr KiranPlan evaluation in Radiotherapy- Dr Kiran
Plan evaluation in Radiotherapy- Dr Kiran
Kiran Ramakrishna
 
EWINGS SARCOMA & RADIOTHERAPY
EWINGS SARCOMA & RADIOTHERAPYEWINGS SARCOMA & RADIOTHERAPY
EWINGS SARCOMA & RADIOTHERAPY
Paul George
 
Management of brain metastases
Management of brain metastasesManagement of brain metastases
Management of brain metastases
Shreya Singh
 
RADIOTHERAPY IN CARCINOMA BREAST (EARLY AND LOCALLY ADVANCED)
RADIOTHERAPY IN CARCINOMA BREAST (EARLY AND LOCALLY ADVANCED)RADIOTHERAPY IN CARCINOMA BREAST (EARLY AND LOCALLY ADVANCED)
RADIOTHERAPY IN CARCINOMA BREAST (EARLY AND LOCALLY ADVANCED)
DrAnkitaPatel
 
Brain metastasis
Brain metastasis Brain metastasis
Brain metastasis
Kiran Ramakrishna
 
APBI-Dr Kiran
APBI-Dr Kiran APBI-Dr Kiran
APBI-Dr Kiran
Kiran Ramakrishna
 
Radiotherapy lymphoma
Radiotherapy lymphoma Radiotherapy lymphoma
Radiotherapy lymphoma
vrinda singla
 
Prostate ca
Prostate caProstate ca
Radiotherapy sarcomas
Radiotherapy sarcomas Radiotherapy sarcomas
Radiotherapy sarcomas
Ashutosh Mukherji
 
2D PLANNING IN BRAIN TUMOR
2D PLANNING IN BRAIN TUMOR2D PLANNING IN BRAIN TUMOR
2D PLANNING IN BRAIN TUMOR
Kanhu Charan
 
HYPOFRACTIONATION IN RADIOTHERAPY
HYPOFRACTIONATION IN RADIOTHERAPYHYPOFRACTIONATION IN RADIOTHERAPY
HYPOFRACTIONATION IN RADIOTHERAPY
Rejil Rajan
 

What's hot (20)

MANAGEMENT OF MENINGIOMA
MANAGEMENT OF MENINGIOMAMANAGEMENT OF MENINGIOMA
MANAGEMENT OF MENINGIOMA
 
Embrace ii protocol
Embrace ii protocolEmbrace ii protocol
Embrace ii protocol
 
Radiotherapy planning for rectal cancer ,2D updates!
Radiotherapy planning for rectal cancer ,2D   updates!Radiotherapy planning for rectal cancer ,2D   updates!
Radiotherapy planning for rectal cancer ,2D updates!
 
Radiation for Lung Cancer
Radiation for Lung CancerRadiation for Lung Cancer
Radiation for Lung Cancer
 
Radiosurgery for brain metastases
Radiosurgery for brain metastasesRadiosurgery for brain metastases
Radiosurgery for brain metastases
 
High grade glioma kiran
High grade glioma  kiranHigh grade glioma  kiran
High grade glioma kiran
 
Contouring guidelines Cervix IMRT
Contouring guidelines Cervix IMRTContouring guidelines Cervix IMRT
Contouring guidelines Cervix IMRT
 
Hippocampal sparing whole brain radiation therapy- Making a case!
Hippocampal sparing  whole brain radiation therapy- Making a case!Hippocampal sparing  whole brain radiation therapy- Making a case!
Hippocampal sparing whole brain radiation therapy- Making a case!
 
LUNG SBRT A LITERATURE REVIEW
LUNG SBRT A LITERATURE REVIEWLUNG SBRT A LITERATURE REVIEW
LUNG SBRT A LITERATURE REVIEW
 
Plan evaluation in Radiotherapy- Dr Kiran
Plan evaluation in Radiotherapy- Dr KiranPlan evaluation in Radiotherapy- Dr Kiran
Plan evaluation in Radiotherapy- Dr Kiran
 
EWINGS SARCOMA & RADIOTHERAPY
EWINGS SARCOMA & RADIOTHERAPYEWINGS SARCOMA & RADIOTHERAPY
EWINGS SARCOMA & RADIOTHERAPY
 
Management of brain metastases
Management of brain metastasesManagement of brain metastases
Management of brain metastases
 
RADIOTHERAPY IN CARCINOMA BREAST (EARLY AND LOCALLY ADVANCED)
RADIOTHERAPY IN CARCINOMA BREAST (EARLY AND LOCALLY ADVANCED)RADIOTHERAPY IN CARCINOMA BREAST (EARLY AND LOCALLY ADVANCED)
RADIOTHERAPY IN CARCINOMA BREAST (EARLY AND LOCALLY ADVANCED)
 
Brain metastasis
Brain metastasis Brain metastasis
Brain metastasis
 
APBI-Dr Kiran
APBI-Dr Kiran APBI-Dr Kiran
APBI-Dr Kiran
 
Radiotherapy lymphoma
Radiotherapy lymphoma Radiotherapy lymphoma
Radiotherapy lymphoma
 
Prostate ca
Prostate caProstate ca
Prostate ca
 
Radiotherapy sarcomas
Radiotherapy sarcomas Radiotherapy sarcomas
Radiotherapy sarcomas
 
2D PLANNING IN BRAIN TUMOR
2D PLANNING IN BRAIN TUMOR2D PLANNING IN BRAIN TUMOR
2D PLANNING IN BRAIN TUMOR
 
HYPOFRACTIONATION IN RADIOTHERAPY
HYPOFRACTIONATION IN RADIOTHERAPYHYPOFRACTIONATION IN RADIOTHERAPY
HYPOFRACTIONATION IN RADIOTHERAPY
 

Similar to high grade glioma

LOW GRADE GLIOMA.pptx
LOW GRADE GLIOMA.pptxLOW GRADE GLIOMA.pptx
LOW GRADE GLIOMA.pptx
Brijesh Maheshwari
 
Glioma markers in neurosurgery
Glioma markers in neurosurgeryGlioma markers in neurosurgery
Glioma markers in neurosurgery
Dr. Shahnawaz Alam
 
Molecular pathogenesis of CNS tumors
Molecular pathogenesis of  CNS tumorsMolecular pathogenesis of  CNS tumors
Molecular pathogenesis of CNS tumors
MD Patholgoy, AFMC
 
Brain tumour patient forum Michael Buckland Tests related to diagnosis pathol...
Brain tumour patient forum Michael Buckland Tests related to diagnosis pathol...Brain tumour patient forum Michael Buckland Tests related to diagnosis pathol...
Brain tumour patient forum Michael Buckland Tests related to diagnosis pathol...
Cure Brain Cancer Foundation
 
Management of Low Grade Glioma
Management of Low Grade GliomaManagement of Low Grade Glioma
Management of Low Grade Glioma
Shreya Singh
 
Pre-managment of Low Grade Glioma- low grade glioma
Pre-managment of Low Grade Glioma- low grade gliomaPre-managment of Low Grade Glioma- low grade glioma
Pre-managment of Low Grade Glioma- low grade glioma
7pwys74fgg
 
Low grade gliomas
Low grade gliomasLow grade gliomas
Low grade gliomas
Erion Junior de Andrade
 
Low grade gliomas kiran
Low grade gliomas   kiranLow grade gliomas   kiran
Low grade gliomas kiran
Kiran Ramakrishna
 
Role of radiation in benign conditions
Role of radiation in benign conditionsRole of radiation in benign conditions
Role of radiation in benign conditions
Purvi Rathod
 
Molecular studies in cns tumors
Molecular studies in cns tumorsMolecular studies in cns tumors
Molecular studies in cns tumors
Dr.Amrita Rakesh
 
low grade glioma.pptx
low grade glioma.pptxlow grade glioma.pptx
low grade glioma.pptx
MedhatMoustafa3
 
WHO CNS Classification of Tumors 2021-2.pptx
WHO CNS Classification of Tumors 2021-2.pptxWHO CNS Classification of Tumors 2021-2.pptx
WHO CNS Classification of Tumors 2021-2.pptx
RabiyyahBashir1
 
RT in Benign diseases.pptx
RT in Benign diseases.pptxRT in Benign diseases.pptx
RT in Benign diseases.pptx
AtulGupta369
 
High Grade Glioma
High Grade GliomaHigh Grade Glioma
High Grade Glioma
DrAyush Garg
 
Brain tumor dr. abeer elsayed
Brain tumor dr. abeer elsayedBrain tumor dr. abeer elsayed
Brain tumor dr. abeer elsayed
Abeer Ibrahim
 
Multiple Myeloma
Multiple MyelomaMultiple Myeloma
Multiple Myeloma
Sadia Sadiq
 
low grade NHL- final.pdf
low grade NHL- final.pdflow grade NHL- final.pdf
low grade NHL- final.pdf
HardikSharma590779
 
LOW GRADE GLIOMA management in radiotherapy
LOW GRADE GLIOMA management in radiotherapyLOW GRADE GLIOMA management in radiotherapy
LOW GRADE GLIOMA management in radiotherapy
srinivasreddy200927
 
Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...
Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...
Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...
CrimsonPublishersAICS
 

Similar to high grade glioma (20)

LOW GRADE GLIOMA.pptx
LOW GRADE GLIOMA.pptxLOW GRADE GLIOMA.pptx
LOW GRADE GLIOMA.pptx
 
Glioma markers in neurosurgery
Glioma markers in neurosurgeryGlioma markers in neurosurgery
Glioma markers in neurosurgery
 
Molecular pathogenesis of CNS tumors
Molecular pathogenesis of  CNS tumorsMolecular pathogenesis of  CNS tumors
Molecular pathogenesis of CNS tumors
 
Brain tumour patient forum Michael Buckland Tests related to diagnosis pathol...
Brain tumour patient forum Michael Buckland Tests related to diagnosis pathol...Brain tumour patient forum Michael Buckland Tests related to diagnosis pathol...
Brain tumour patient forum Michael Buckland Tests related to diagnosis pathol...
 
Management of Low Grade Glioma
Management of Low Grade GliomaManagement of Low Grade Glioma
Management of Low Grade Glioma
 
Multiple myeloma
Multiple myelomaMultiple myeloma
Multiple myeloma
 
Pre-managment of Low Grade Glioma- low grade glioma
Pre-managment of Low Grade Glioma- low grade gliomaPre-managment of Low Grade Glioma- low grade glioma
Pre-managment of Low Grade Glioma- low grade glioma
 
Low grade gliomas
Low grade gliomasLow grade gliomas
Low grade gliomas
 
Low grade gliomas kiran
Low grade gliomas   kiranLow grade gliomas   kiran
Low grade gliomas kiran
 
Role of radiation in benign conditions
Role of radiation in benign conditionsRole of radiation in benign conditions
Role of radiation in benign conditions
 
Molecular studies in cns tumors
Molecular studies in cns tumorsMolecular studies in cns tumors
Molecular studies in cns tumors
 
low grade glioma.pptx
low grade glioma.pptxlow grade glioma.pptx
low grade glioma.pptx
 
WHO CNS Classification of Tumors 2021-2.pptx
WHO CNS Classification of Tumors 2021-2.pptxWHO CNS Classification of Tumors 2021-2.pptx
WHO CNS Classification of Tumors 2021-2.pptx
 
RT in Benign diseases.pptx
RT in Benign diseases.pptxRT in Benign diseases.pptx
RT in Benign diseases.pptx
 
High Grade Glioma
High Grade GliomaHigh Grade Glioma
High Grade Glioma
 
Brain tumor dr. abeer elsayed
Brain tumor dr. abeer elsayedBrain tumor dr. abeer elsayed
Brain tumor dr. abeer elsayed
 
Multiple Myeloma
Multiple MyelomaMultiple Myeloma
Multiple Myeloma
 
low grade NHL- final.pdf
low grade NHL- final.pdflow grade NHL- final.pdf
low grade NHL- final.pdf
 
LOW GRADE GLIOMA management in radiotherapy
LOW GRADE GLIOMA management in radiotherapyLOW GRADE GLIOMA management in radiotherapy
LOW GRADE GLIOMA management in radiotherapy
 
Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...
Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...
Giant Glioblastoma in a Patient with Previous Prostate Adenocarcinoma_Crimson...
 

Recently uploaded

HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
GL Anaacs
 
How STIs Influence the Development of Pelvic Inflammatory Disease.pptx
How STIs Influence the Development of Pelvic Inflammatory Disease.pptxHow STIs Influence the Development of Pelvic Inflammatory Disease.pptx
How STIs Influence the Development of Pelvic Inflammatory Disease.pptx
FFragrant
 
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists  Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Saeid Safari
 
Hemodialysis: Chapter 3, Dialysis Water Unit - Dr.Gawad
Hemodialysis: Chapter 3, Dialysis Water Unit - Dr.GawadHemodialysis: Chapter 3, Dialysis Water Unit - Dr.Gawad
Hemodialysis: Chapter 3, Dialysis Water Unit - Dr.Gawad
NephroTube - Dr.Gawad
 
ANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptxANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptx
Swetaba Besh
 
Superficial & Deep Fascia of the NECK.pptx
Superficial & Deep Fascia of the NECK.pptxSuperficial & Deep Fascia of the NECK.pptx
Superficial & Deep Fascia of the NECK.pptx
Dr. Rabia Inam Gandapore
 
Physiology of Chemical Sensation of smell.pdf
Physiology of Chemical Sensation of smell.pdfPhysiology of Chemical Sensation of smell.pdf
Physiology of Chemical Sensation of smell.pdf
MedicoseAcademics
 
Physiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of TastePhysiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of Taste
MedicoseAcademics
 
Prix Galien International 2024 Forum Program
Prix Galien International 2024 Forum ProgramPrix Galien International 2024 Forum Program
Prix Galien International 2024 Forum Program
Levi Shapiro
 
BRACHYTHERAPY OVERVIEW AND APPLICATORS
BRACHYTHERAPY OVERVIEW  AND  APPLICATORSBRACHYTHERAPY OVERVIEW  AND  APPLICATORS
BRACHYTHERAPY OVERVIEW AND APPLICATORS
Krishan Murari
 
24 Upakrama.pptx class ppt useful in all
24 Upakrama.pptx class ppt useful in all24 Upakrama.pptx class ppt useful in all
24 Upakrama.pptx class ppt useful in all
DrSathishMS1
 
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 UpakalpaniyaadhyayaCharaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Dr KHALID B.M
 
KDIGO 2024 guidelines for diabetologists
KDIGO 2024 guidelines for diabetologistsKDIGO 2024 guidelines for diabetologists
KDIGO 2024 guidelines for diabetologists
د.محمود نجيب
 
POST OPERATIVE OLIGURIA and its management
POST OPERATIVE OLIGURIA and its managementPOST OPERATIVE OLIGURIA and its management
POST OPERATIVE OLIGURIA and its management
touseefaziz1
 
For Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #Girls
For Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #GirlsFor Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #Girls
For Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #Girls
Savita Shen $i11
 
Triangles of Neck and Clinical Correlation by Dr. RIG.pptx
Triangles of Neck and Clinical Correlation by Dr. RIG.pptxTriangles of Neck and Clinical Correlation by Dr. RIG.pptx
Triangles of Neck and Clinical Correlation by Dr. RIG.pptx
Dr. Rabia Inam Gandapore
 
NVBDCP.pptx Nation vector borne disease control program
NVBDCP.pptx Nation vector borne disease control programNVBDCP.pptx Nation vector borne disease control program
NVBDCP.pptx Nation vector borne disease control program
Sapna Thakur
 
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdfAlcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Dr Jeenal Mistry
 
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdfBENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
DR SETH JOTHAM
 
TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...
TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...
TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...
kevinkariuki227
 

Recently uploaded (20)

HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
 
How STIs Influence the Development of Pelvic Inflammatory Disease.pptx
How STIs Influence the Development of Pelvic Inflammatory Disease.pptxHow STIs Influence the Development of Pelvic Inflammatory Disease.pptx
How STIs Influence the Development of Pelvic Inflammatory Disease.pptx
 
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists  Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
 
Hemodialysis: Chapter 3, Dialysis Water Unit - Dr.Gawad
Hemodialysis: Chapter 3, Dialysis Water Unit - Dr.GawadHemodialysis: Chapter 3, Dialysis Water Unit - Dr.Gawad
Hemodialysis: Chapter 3, Dialysis Water Unit - Dr.Gawad
 
ANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptxANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptx
 
Superficial & Deep Fascia of the NECK.pptx
Superficial & Deep Fascia of the NECK.pptxSuperficial & Deep Fascia of the NECK.pptx
Superficial & Deep Fascia of the NECK.pptx
 
Physiology of Chemical Sensation of smell.pdf
Physiology of Chemical Sensation of smell.pdfPhysiology of Chemical Sensation of smell.pdf
Physiology of Chemical Sensation of smell.pdf
 
Physiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of TastePhysiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of Taste
 
Prix Galien International 2024 Forum Program
Prix Galien International 2024 Forum ProgramPrix Galien International 2024 Forum Program
Prix Galien International 2024 Forum Program
 
BRACHYTHERAPY OVERVIEW AND APPLICATORS
BRACHYTHERAPY OVERVIEW  AND  APPLICATORSBRACHYTHERAPY OVERVIEW  AND  APPLICATORS
BRACHYTHERAPY OVERVIEW AND APPLICATORS
 
24 Upakrama.pptx class ppt useful in all
24 Upakrama.pptx class ppt useful in all24 Upakrama.pptx class ppt useful in all
24 Upakrama.pptx class ppt useful in all
 
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 UpakalpaniyaadhyayaCharaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
 
KDIGO 2024 guidelines for diabetologists
KDIGO 2024 guidelines for diabetologistsKDIGO 2024 guidelines for diabetologists
KDIGO 2024 guidelines for diabetologists
 
POST OPERATIVE OLIGURIA and its management
POST OPERATIVE OLIGURIA and its managementPOST OPERATIVE OLIGURIA and its management
POST OPERATIVE OLIGURIA and its management
 
For Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #Girls
For Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #GirlsFor Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #Girls
For Better Surat #ℂall #Girl Service ❤85270-49040❤ Surat #ℂall #Girls
 
Triangles of Neck and Clinical Correlation by Dr. RIG.pptx
Triangles of Neck and Clinical Correlation by Dr. RIG.pptxTriangles of Neck and Clinical Correlation by Dr. RIG.pptx
Triangles of Neck and Clinical Correlation by Dr. RIG.pptx
 
NVBDCP.pptx Nation vector borne disease control program
NVBDCP.pptx Nation vector borne disease control programNVBDCP.pptx Nation vector borne disease control program
NVBDCP.pptx Nation vector borne disease control program
 
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdfAlcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
 
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdfBENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
 
TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...
TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...
TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...
 

high grade glioma

  • 1. PATHOLOGY, MOLECULAR BIOLOGY & MANAGEMENT OF HIGH GRADE GLIOMA PRESENTER- Dr HARDIK SHARMA MODERATOR- Dr RENU MADAN
  • 2. EPIDEMIOLOGY & RISK FACTORS • Yearly incidence of malignant glioma ∼6/100 000 • Gliomas - 80% of malignant brain tumors; Glioblastoma(GBM) – most common primary malignant brain tumor • Slight predominance in males (1.6times) • In a report of the Hospital based Cancer registries, 2021, CNS tumors constituted 1.6 % of all malignancies • Exposure to ionizing irradiation • Rare hereditary syndromes carry an increased risk for glioma: Cowden-, Turcot-, Lynch-, Li-Fraumeni syndrome and neurofibromatosis type 1. • Viruses like RNA ,DNA virus- HPV, Adenoviruses • Chemical agents a. Polycyclic hydrocarbons b. Alkylating agents EANO GUIDELINES
  • 3. WHO GRADE DEFINITION GLIOMAS I low proliferative potential (hypercellular) Pilocytic astrocytoma, Pleomorphic xanthoastrocytoma, Ganglioglioma, Subependymal giant cell astrocytoma II +/- nuclear atypia Diffuse astrocytomas and oligodendrogliomas III Nuclear atypia + brisk miotic activity Anaplastic astrocytomas and oligodendrogliomas IV Nuclear atypia + mitotic activity + necrosis and/or microvascular proliferation Glioblastoma
  • 4. High Grade Glioma ▪ Grade 3 and 4 astrocytoma ▪ Grade 3 oligodendroglioma ▪ Glioblastoma (IDH wild type-4)
  • 5. TUMOR TYPE CNS WHO GRADE CHARACTERSTIC MOLECULAR ALTERATIONS ADULT TYPE DIFFUSE GLIOMAS • Astrocytoma IDH Mutant 2,3,4 IDH1,IDH2 • Oligodendroglioma IDH Mutant,1p/19q Codeleted 2,3 IDH1,IDH2,1p/19q • Glioblastoma IDH Wildtype 4 IDH-wildtype ,Ch-7 &10,TERT,EGFR PEDIATRIC HIGH GRADE GLIOMAS • Diffuse Midline Glioma, H3 K27 4 H3 K27,EGFR,EZHIP • Diffuse Hemispheric Glioma, H3 G34 4 H3 G34 • Diffuse Pediatric HGG,H3-Wildtype & IDH wildtype 4 IDH-wildtype, EGFR, PDGFRA, MYCN • Infant Type Hemispheric Glioma NA RTK genes WHO HIGH GRADE GLIOMA
  • 6. Pediatric Type Vs Adult Type Gliomas • Although, both share overlying histology • But the genetic & biology are distinctively different • Generally pediatric type - indolent despite “anaplastic” features & lack IDH mutation & 1p/19q codeletion i.e genetic hallmark of adult type • They harbor genetic profiling such as MAPK pathway alteration (Pediatric type) • Do not depend upon the patient age , instead defined based on representative molecular alterations, implicating that pediatric-type gliomas may occur in adults & vice versa
  • 9. Molecular marker Biological function Diagnostic roles IDH 1 OR IDH2 Gain of function mutation Distinguishes diffuse gliomas with IDH mutation from IDH-wild-type glioblastomas and other IDH-wild-type gliomas 1p/19q codeletion Inactivation of TSP on 1p and 19q Distinguishes oligodendroglioma, IDH- mutant and 1p/19q-codeleted from astrocytoma, IDH-mutant Loss of nuclear ATRX Cellular proliferation & promotion of cellular longevity Loss of nuclear ATRX in an IDH-mutant glioma is diagnostic for astrocytic lineage tumors Histone H3 K27 altered Histone H3.3 missense mutation affecting epigenetics Defining molecular feature of diffuse midline glioma, H3.3 G34-mutant Histone H3.3G34 altered Histone mutation affecting epigenetic regulation Defining molecular feature of diffuse hemispheric glioma, H3.3 G34-mutant MGMT promoter methylation DNA repair None, but is a predictive biomarker of benefit from alkylating chemotherapy in patients with IDH-wild-type glioblastoma
  • 10. CDKN2A/2B homozygous deletion Encode CDKNA2A &2B ,regulators of RB17 p-53 dependent signaling A marker of poor outcome and WHO grade 4 disease in IDH-mutant astrocytomas EGFR amplification Cell proliferation ; invasion & resistance to induction of apoptosis EGFR amplification occurs in ~40–50% of glioblastoma, IDH wild type Molecular marker of glioblastoma, IDH wild type, WHO grade 4 TERT promotor mutation Cell proliferation; promotes cellular longevity by increasing TERT expression OCCUR IN -70% OF Glioblastoma,IDH wild-type & >95% oligodendroglioma,IDH- mutant Molecular marker of glioblastoma wild-4 +7/-10 cytogenetic signature Gain of ch7 combined with loss of ch10 Molecular marker of glioblastoma, IDH wild type, WHO grade 4 ( BRAFv600E mutation Oncogenic driver mutation leading to MAPK pathway activation Rare in adult diffuse gliomas but amenable to pharmacological intervention
  • 11. IDH1/2 mutation 1p/19q co-deletion MGMT promoter methylation Diagnostic Role DD glioma versus gliosis Typical for transformed low-grade glioma Pathognomonic for oligodendroglioma None Prpgnostic Role Protracted natural history in IDH-mutated tumours Protracted natural history in 1p/19q codeleted tumours Prognostic for anaplastic glioma patients (possibly with IDH mutations) treated with RT or alkylating drugs Predictive Role Absence of mutation suggests predictive role for MGMT promoter methylation Prolongation of survival with early chemotherapy in 1p/19q-co-deleted oligodendrogliomas Predictive in GBM for benefit from alkylating chemotherapy.Elderly GBM: MGMT-methylated→TMZ; MGMT unmethylated→ RT Frequency:WHO-II Diffuse Astrocytoma 70-80% 15% 40-50% Oligodendroglioma 70-80% 30-60% 60-80% Who G-III Anaplastic Astrocytoma 50-70% 15% WHO G-II Anaplastic Oligodendroglioma 50-80% 50-80% 70% Who G-IV Glioblastoma 5-10% <5% 35% CLINICAL RELEVANT MOLECULAR MARKERS
  • 12. Location & Spread Of Glioma • Approximately 60% of gliomas are located in one of the cerebral hemispheres  Frontal lobe(25.6%)>temporal lobe (19.6%)> parietal lobe (12.6%) Glioblastoma multiforme is uncommon in the region of third ventricle(<1%) and rarely occurs in posterior fossa Although most of the Glioblastoma are centered in the deep white matter Epicenter at the gray-white matter junction-10% • Malignant gliomas tends to spread or recur in adjacent brain region along white matter tracts • Spread outside the central nervous system is extremely rare • Rarely , gliomas may spread systemically
  • 13. History and Clinical Examination • Clinical presentation: new onset epilepsy, focal deficits, including neurocognitive impairment, and indicators of intracranial mass effect, such as headache, vomiting, and altered consciousness. • History: duration of neurological symptoms and signs, familial risk or exogenous risk factors, including exposure to radiation • Physical examination: focuses on the detection of systemic cancer and contraindications for neurosurgical procedures. Neurocognitive assessment, performance status should be documented. ▪ MMSE is widely used as a screening instrument to detect neurocognitive impairment
  • 14. Imaging Of Brain ▪ MRI – gold standard T2-weighted and fluid-attenuated inversion recovery sequences, and T1-weighted sequences before and after gadolinium use, in at least one plane, is the standard method for the detection of a glioma ▪ MR Spectroscopy: differentiating tumor from radiation necrosis, grading tumors, assessing response; area most abnormal → biopsy ▪ MR Perfusion: differentiating grade of tumor or tumor versus radiation necrosis; area of highest perfusion → biopsy ▪ CT: patients who cannot have MRI, shows calcification ▪ Amino acid PET helps define metabolic hotspots for biopsy ▪ Intra-arterial angiography can aid the surgical strategy
  • 15. Diagnostic Characteristics Tumor Imaging Characteristics Glioblastoma Multiforme • Enhancing MRI or CT lesion, hypodense interior, often with associated edema & necrosis • Angiography: highly abnormal vasculature with arteriovenous shunting and early venous drainage. Anaplastic Astrocytoma • CT- inhomogeneous density • T2-weighted FLAIR MRI - hyperintense, space-occupying lesions, patchy enhancement after contrast administration and often have peritumoral edema. • Up to 30% can show no enhancement on CT or MRI. • Abnormal vessels can be visualised by MRI angiography Oligodendroglioma • Heterogeneous lesion that may or may not enhance on MRI or CT, frequently with calcification, cystic regions, or hemorrhage
  • 16. 58-year-old with a glioblastoma who presented with gait disturbance ADC FLAIR T1C DWI
  • 17. MRSPECTROSCOPY Increase in choline & decrease NAA level in glioma Lactate level indicate necrosis but non specific
  • 19. TREATMENT • Surgery • Radiotherapy • Chemotherapy and Targeted Therapy • Symptomatic management Standard of care Maximal surgical resection + Postoperative radiotherapy and Chemotherapy
  • 20. Surgical Management • Histological diagnosis for all patients - biopsy or decompression surgery unless there is a specific contraindication • Use of neuronavigation systems, Laser Interstitial Thermal Therapy, use of fluorescent tumor localizing dyes such as 5-aminolevulinec acid Type of resection Extent of resection Biopsy <10% Partial 10-50% Subtotal 50-90% Near-total >90%-94% Gros -total >95%
  • 21. ▪ Microsurgical resection of a right-sided recurrent IDHwt glioblastoma WHO grad IV using intraoperative neuronavigation, neuromonitoring and 5-ALA fluorescence techniques. ▪ A: T1CE axial, sagittal and coronal planes including DTI fiber tracking (blue fibers). The green trajectories / red points represent the pointer for intraoperative neuronavigation. ▪ B: Upper image: corresponding intraoperative 5-ALA fluorescence image taken from the area as depicted by neuronavigation. Lower image: opening of the right ventricle due to critical involvement by tumor formation. ▪ C: Postoperative MRI confirms gross total resection without residual contrast enhancement, no perilesional ischemia (diffusion-weighted image upper right).
  • 22. Influence of location and extent of surgical resection on survival of patients with glioblastoma multiforme: results of three consecutive Radiation Therapy Oncology Group (RTOG) clinical trials. Simpson et al , IJROBP 1993 o Significant correlation of age, Karnofsky Performance Status, extent of surgery, and primary site with survival. There was no statistical difference in survival based on tumor size. o The best survival rates occurred in patients who had at least three of the following features: • < 40 years of age • high Karnofsky Performance Status(80-100) • frontal tumors, and • total resection (17 months median) Procedure Median survival Total resection 11.3 months Partial resection 10.4 months biopsy 6.6 months
  • 23. Goals of surgery: maximal safe resection Diagnosis Alleviate symptoms Decrease need for steroids Increase survival • Early postoperative MRI (within 48 hours) - confirms the degree of resection and can help with radiotherapy planning ( to assess for the revision surgery) • Patients should be re-discussed at the specialist MDT with histology and general performance status details available. • Patients should be staged using the WHO system. • Adjuvant treatment should be commenced within 2 weeks of biopsy or 4 weeks of decompression surgery.
  • 24. MEDICAL MANAGEMENT • ANTIEPILEPTIC DRUGS Non Enzyme inducing anti seizure medications should be used eg: levetiracetam, topiramate, valproic acid, lacosamide Close monitoring for side effects No role for primary perioperative prophylaxis (i.e. in patients who have never had a seizure) Important to know about prior history of seizure
  • 25. • STEROIDS Corticosteroids, preferably dexamethasone (in conjunction with gastric protection if used at high doses) - reduce symptomatic peritumoral vasogenic edema, neurological deficits and signs of increased intracranial pressure Lowest dose of steroids – shortest time possible Downward titration of dose – whenever possible Extensive mass effect – receive steroids for at least 24 hours before RT Chronic corticosteroids (≥ 20 mg prednisone equivalents daily for ≥ 1 month) - consider prophylaxis for osteoporosis and pneumocystis jerovecii pneumonia should be considered
  • 26. RPA Score Based Prognosis Class Patient Characterstics Median Survival (Months) I, II Astrocytoma Age ≤50 yr, normal mental status or age >50 yr • KPS >70, symptoms >3 mo 40-60 III, IV Astrocytoma • Age ≤50 yr, abnormal mental status • Age >50 yr, symptoms <3 mo Glioblastoma • Age <50 yr • Age >50 yr, KPS ≥70 11-18 V, VI Glioblastoma • Age >50 yr, KPS <70 or abnormal mental status 5-9
  • 27. • Evaluated protein biomarkers + clinical variable (age, KPS, extent of resection, neurologic function) • Ki-67, c-Met and MGMT found to be significant biomarkers Class Median OS (months) I 21 II 16.6 III 9.4
  • 28. High Grade Glioma Adjuvant treatment Preferred Regime Other Recommended Regime Oligodendroglioma (3) (KPS > 70) • RT with adjuvant PCV • RT with neoadjuvant PCV • RT with adjuvant and concurrent TMZ • RT with adjuvant TMZ Astrocytoma (3,4) (KPS > 70) • RT with adjuvant and concurrent TMZ • RT followed by adjuvant TMZ • RT with adjuvant PCV • RT with neoadjuvant PCV High Grade glioma (KPS < 70) Single modality: • RT (hypofractionation preferred) or • TMZ (if tumor is MGMT promoter methylated) EANO GUIDELINES
  • 29. Glioblastoma Preferred Regime Other Recommended Regime Useful In Certain Circumstances Adjuvant treatment RT with concurrent and adjuvant TMZ + TTF • RT with concurrent and adjuvant TMZ ( age < 70y and KPS <60) • TMZ (if tumor is MGMT promoter methylated and KPS <60 or age >70 and KPS > 60) • RT with conc and adj lomustine and TMZ (if tumor is MGMT promoter methylated, KPS > 60, and age < 70y)
  • 30. Survival Curves Of Patients Who Received: Median Survival A) Best conventional care but no radiotherapy or chemotherapy 14 weeks B) BCNU 18.5 weeks C) Radiotherapy 36 weeks D) BCNU and radiotherapy 34.5 weeks Walker et al 1978
  • 31. • Radiotherapy significant influence on the survival of patients • The specific relationship between 50 & 60Gy indicates a 1.3 times increase in median life span associated with the higher dose (p = .004) RT Dose Median survival No 18 weeks 45Gy 13.5 weeks 50Gy 28 weeks 55Gy 36 weeks 60Gy 42 weeks 1979
  • 32. • 1.Age 18 - 70 y • 2.Randomized 1:1 • Partial brain RT 60 Gy • Partial brain RT 60 Gy / TMZ → TMZ • 3.Improved Median survival • 12.1 → 14.6 months • P < 0.001 • 4.Improved 5 yr Survival • 1.9% → 9.8% • 5.More Grade 3-4 haematological toxicity with TMZ i.e 7% only 2005
  • 33. MGMT methylation status- EORTC PHASE III • Regardless of treatment, MGMT methylation was a favorable prognostic factor (p<0.0001) • MGMT methylation strongest predictor outcome: 23 mo vs 13 mo • MGMT methylated cases • MS was 21.7 vs 15.3 mo for chemoRT vs RT alone • MGMT un-methylated cases: • Difference in overall survival favoring the temozolomide-plus- radiotherapy group was only marginally significant (p=0.06 ) • MS was 12.7 vs 11.8 mo for chemoRT vs RT alone 2009
  • 34. 2019 • GBM with methylated MGMT • 60Gy + con & adj TMZ Vs 60Gy + TMZ+Lomustine • Median OS 31mo Vs 48 mo (p=0.0492) • Median PFS 16.7mo, not different
  • 35. • Arm 1 XRT with TMZ & Bevacizumab Vs Arm 2 XRT with placebo & TMZ • Addition of BEVACIZUMAB significantly prolonged PFS survival (p<0.001) • Median PFS 10.6 months vs 6.2 months • However, the median OS was not significantly different in both arms • Rates of adverse events was higher with bevacizumab than placebo 2014
  • 36. TUMOR TREATING FIELDS • the placement of noninvasive transducer arrays on the scalp and the delivery of low intensity (1 to 3 V/cm), intermediate frequency (100 to 300 kHz), alternating electric fields to the tumor. • Lead to tumor cell kill during • 1. metaphase - disrupting the formation of the mitotic spindle, and • 2. cytokinesis - dielectrophoretic dislocation of intracellular constituents resulting in apoptosis. • Most effective - applied in the direction of the division axis of the dividing cell, and therefore, two sequential field directions are applied to tumors by using two perpendicular pairs of transducer arrays in order to increase the efficacy of TTFields.
  • 37. TUMOR TREATING FIELDS o 1. KPS > 70; median age 56 o 2. Randomized 2:1 after 60Gy, TMZ Adj TMZ + TTF Adjuvant TMZ o 3. Improved median survival from time of randomization 16→20.9 mth P<0.001 o 4. Toxicity – dermatitis in 52% o 5. Improved OS: >18 hours/day
  • 38. ELDERLY GLIOBLASTOMA NCIC (Roa JCO 2004) • Age>60, KPS >50 • 60Gy/30# vs 40 Gy/15# • No diff in MS: 5.1 vs 5.6 mth Nordic Trial (Malmstrom Lancet 2012) • Age > 60y, GBM • 34Gy/10# was not inferior to 60Gy/30# • Methylated MGMT associated with better survival for TMZ for age > 70y NOA-08 (Wick, lancet, 2012) • Age >65y, KPS > 60, AA, GBM • TMZ(100mg/m2days 1-7 of 1 week on, 1 week off cycles) VS 60 Gy/30# RT • 8.6 vs 9.6 month median survival • MGMT Unmethylated – EFS longer with RT • MGMT methylated – EFS longer with TMZ
  • 39. CCTG/EORTC/TROG (Perry NEJM 2017) • Age > 65 y, ECOG PS 0-2 • 40 Gy/15# RT/TMZ → TMZ better than 40 Gy/15# RT alone • 9.3 vs 7.6 months median survival • MGMT Unmethylated – 10 mth vs 7.9 mth • PFS – 5.3 mth vs3.9 mth • 1 yr survival rate 37.8% vs 22.2%. • Arm 1 (25Gy/5#/5days) Vs Arm 2 (40Gy/15#/3.3weeks) • Median OS (7.9 mo) Vs (6.4 mo) • PFS- 4.2 mo Vs 4.2 mo 2015
  • 40. Treatment Scheme For Elderly HGG
  • 41. • RT with conc Temozolamide vs RT alone • OS was 146 days vs 121 days (p=0.146) • PFS was 109.5 days vs 77 days (p=0.028) • Dose- 25GY/5#/5days
  • 42. Minimizing the uncertainties regarding the effects of delaying radiotherapy for Glioblastoma: A systematic review and meta-analysis Loureiro et al Radiother Oncol 2016 No evidence of a true effect on OS by delaying RT in GBM patients ( range 20-86 days) Postoperative neoadjuvant temozolomide before radiotherapy versus standard radiotherapy in patients 60 years or younger with anaplastic astrocytoma or glioblastoma: a randomized trial Malmstorm et al ActaOncologia 2017 GBM or AA, age < 60 y, PS 0-2 2-3 cycles TMZ → RT RT only Median OS GBM + AA 17.7 months 20.3 months AA 95.1 months 35.2 months
  • 43. Astrocytoma Grade 3-4 • Anaplastic astrocytomas, IDH-mutant, are the most common WHO grade III astrocytomas. • Standard treatment includes maximal safe removal followed by radiotherapy and chemo • RT with concurrent TMZ ~ RT with concurrent nitrosureas • Slightly better PFS with TMZ
  • 44. Grade-III GLIOMA (noncodeleted): Interim CATNON 1:1:1: randomization • RT(59.4Gy) • RT with concTMZ • RT TMZ • RT/TMZ 12 TMZ Adjuvant TMZ improved OS • OS- (116mo) (21mo) IDH mutant but not in IDH wild • Benefit from adj TMZ OS(82.3 mo) Vs (47mo) • No OS benefit conc TMZ • Hematological toxicity- 8-12% but reversible
  • 45. Oligodendroglioma Grade 3 • Relatively rare • 1p19q codeleted and IDH1-mut or IDH2-mut • Better prognosis than other high grade gliomas • PCV added to RT • CODEL trial ongoing: RT (59.4 Gy) + adj PCV vs RT + conc TMZ + adj TMZ ( result pending)
  • 46. 59.4 Gy/33# of RT 59.4 Gy/33# of RT → six cycles of adjuvant PCV Median OS 30.6 months 42.3 months Median PFS 13.21 months 24.3 months Improved OS in pts with MGMT methylation and IDH mutations. Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402 Cairncross et al JCO 2013 59.4 Gy/33# of RT PCV(4cycles) → RT Median Survival 4.7 years 4.6 years OS - 1p/19q codeleted AO/AOA 7.3 years 14.7 yrs PFS - 1p/19q codeleted AO/AOA 2.9 y 8.4 y OS - 1p/19q non codeleted AO/AOA 3.3 y 5.5 y PFS - 1p/19q non codeleted AO/AOA 1 y 1.2 y Long-term follow-up results of EORTC 26951: A randomized phase III study on adjuvant PCV chemotherapy in anaplastic oligodendroglial tumors (AOD). Van Den Bent et al JCO 2012
  • 47. Temozolomide • oral DNA alkylating agent • good blood–brain barrier penetration • favourable safety profile - myelosuppression, notably thrombocytopenia dose-limiting toxicity Nitrosoureas • second choice after temozolomide for glioma treatment • higher efficacy of procarbazine, lomustine, and vincristine over temozolomide in patients with anaplastic glioma with a good prognosis. • Side effects: prolonged leukopenia and thrombocytopenia • Pulmonary fibrosis with carmustine • Higher rates of discontinuation due to toxicity • Temzolamide is an alkylating agent & result in DNA damage with displacing alkyl group in different DNA positions • MGMT plays an important role in DNA repair enzymes by synthesizing repair proteins • MGMT promoter methylation results in reduced expression of this gene & increase response to alkylating agents
  • 48. • In patients with GBM, OS & prognosis remains dismal • Use concurrent & adjuvant TMZ to maximize the benefit of post-op radiotherapy • Patient with poor KPS score , considered for hypofractionated RT, have similar median survival rates as conventional RT 2013
  • 49. RTOG 0525: A randomized phase III trial comparing standard adjuvant temozolomide (TMZ) with a dose-dense (dd) schedule in newly diagnosed glioblastoma (GBM). Gilbert et al, JCO, 2006 • Arm 1: standard TMZ (150-200 mg/m2 body surface area days 1 to 5 of 28) • Arm 2: intensified regimen (75-100 mg/m2 body surface area days 1 to 21 of 28) for 6-12 cycles. • Conclusion: o Validated MGMT-promoter methylation as a favorable prognostic factor regardless of treatment. o MGMT methylation was associated with improved OS (21.2 v 14 months), PFS (8.7 v 5.7 months) o Dose-dense temozolomide was more toxic than standard dosing and did not significantly alter PFS or OS regardless of MGMT status.
  • 50. Is more better? The impact of extended adjuvant temozolomide in newly diagnosed glioblastoma: a secondary analysis of EORTC and NRG Oncology/RTOG Blumentha et al, NeuroOncol, 2017 Conclusion: • Treatment with more than 6 cycles of TMZ was associated with a somewhat improved PFS in particular for patients with methylated MGMT • OS was not affected by the number of TMZ cycles including the MGMT methylated subgroup(p=0.52) • Treatment with 6 cycles of TMZ was associated with improved PFS (p=0.03) • Better in MGMT methylated patients • Overall survival was not impacted by the number of TEMOZOLAMIDE
  • 51. • C-TMZ Vs E- TMZ(15.4mo Vs 23.8mo) p=0.044 • PFS (12.8mo Vs16.8mo) • 5% Vs 15% 2017
  • 53. Conventional 2D Approach- Whole Brain RT • Position- supine • Ask to patient to stare up straight to the roof • Draw a line along the pupillary line on the forehead • Draw a line joining the lobule of the ear & the lateral canthus of the eye & extend it to the former line • Rest of the two borders are flashed 1cm in air
  • 54. Fluoroscopic simulation ▪ Left and right parallel oppose lateral field ▪ Anterior temporal lobe ,cribriform plate, posterior aspects of eyes included ▪ Inferior field edge passing through C1-C2 junction & base of skull ▪ Safety margin of at least 1cm ( flashed in air) ▪ Placing a radiopaque marker on both lateral canthus
  • 55. Limitation Of Conventional Planning ▪ Irradiation of large volumes of brain with normal tissue also ▪ Higher toxicity and side effects ▪ Lack of 3D visualization of tumor ▪ 2D planning of 3D tumor Radiotherapy for newly diagnosed malignant glioma in adults: a systematic review The Cancer Care Ontario Practice Guidelines Initiative Neuro-Oncology Disease Site Group1 Laparriere et al, 2001 No significant difference in survival rates for whole brain radiation versus more local fields that encompass the enhancing primary plus a 2 cm margin
  • 56. • 80-90% of recurrence is local ( within 2cm of enhancing tumor on CT) • Radionecrosis- 50% of patients treated with WBRT had grade3-4 white matter changes as compared with 14% of patients treated only to local field • 3DCRT makes partial brain irradiation for glioma possible & reduces neurotoxicity • It may be recommended for multifocal disease , but it is rare and failure occur typically within original disease
  • 57. SIMULATION – CONFORMAL RT ▪ CT simulation supine with the head in a neutral position ▪ Head immobilised in an individual Perspex or thermoplastic shell ▪ Slice thickness: 2.5-3mm ▪ Postop MR fusion for better tumor delineation ▪ RT should start within 4-6 weeks of surgery
  • 58. • Mean GTV increased by 46% between early CECT and MRI, the difference was only 4% between delayed CT & MRI • Delayed CECT is superior to early CECT with MRI for target delineation EARLY CT DELAYED CT POST OP MRI
  • 60. ORGAN AT RISK ▪ LENS ▪ B/L OPTIC NERVE ▪ B/L EYE ▪ OPTIC CHIASM ▪ BRAINSTEM ▪ B/L TEMPORAL LOBE ▪ SPINAL CORD
  • 61. EORTC treatment volumes (EORTC 22981/22961, RTOG treatment volumes (RTOG 0525, 0825, 0913, and AVAglio trials) Phase 1 (to 60 Gy in 30 fractions) Phase 1 (to 46 Gy in 23 fractions) Phase 2 (14 Gy boost in 7 fractions) GTV surgical resection cavity plus any residual enhancing tumour (postcontrast T1 weighted MRI scans) GTV1 surgical resection cavity plus any residual enhancing tumour (postcontrast T1 weighted MRI scans) plus surrounding oedema (hyperintensity on T2 or FLAIR MRI scans) GTV 2 surgical resection cavity plus any residual enhancing tumour (postcontrast T1 weighted MRI scans) CTV GTV plus a margin of 2 cm* CTV1 GTV1 plus a margin of 2 cm (if no surrounding oedema is present, the CTV is the contrast enhancing tumour plus 2.5 cm. CTV 2 GTV2 plus a margin of 2 cm PTV CTV plus a margin of 3– 5 mm PTV1 CTV1 plus a margin of 3–5 mm PTV2 CTV2 plus a margin of 3– 5 mm TARGET DELINEATION OF GLIOBLASTOMA *Margins up to 3 cm were allowed in 22981/22961 trial, and 1.5 cm in 26981–22981 trial.
  • 63. MDACC GUIDELINES • INITIAL PHASE (50 Gy): GTV = Resection cavity and any residual contrast-enhancing tumor(T1) but ignoring any edema CTV = GTV + 2 cm isotropic margin Peritumoral edema was not intentionally included in the CTV except for edema existing within the 2-cm margin PTV = CTV + 0.5 cm margin • BOOST PHASE (10 Gy): PTV = GTV + 0.5cm margin
  • 64. NRG – Contouring Guidelines • Careful delineation of anatomic pathways and barriers to spread can spare radiation to uninvolved tissue without compromising target coverage
  • 65. GTV & GTV BOOST CTV & CTV BOOST PTV & PTV BOOST GTV- RED & GTV BOOST- PINK CTV- RED & CTV BOOST- PINK PTV- RED & PTV BOOST- PINK PGI PROTOCOL
  • 66. Dose constraint of conventional 3D-CRT OAR OBJECTIVE(S) End point Brainstem Dmax<54 Gy; 1–10 cc < 59 Gy (periphery) Neuropathy (<5%) or necrosis Optic Chiasm Dmax <55 Gy Optic neuropathy (<3%) Cochlea Ideally one side mean <45 Gy ( Most protocols allow ipsilateral cochlea to receive 60 Gy rather than compromise dose ) Sensorineural hearing loss (<30%) Eyes Macula <45 Gy Retinopathy Lens Ideally <6 Gy Max 10 Gy dose limits should never compromise PTV dose Cataract Optic nerves and chiasma Dmax <55 Gy or 1 % of PTV cannot exceed 60 Gy Optic neuropathy (<3%) Brain Dmax <60Gy Symptomatic necrosis (<3%) Hippocampus Dmax ≤6 Gy & V3Gy ≤ 20% Cognitive impairment Spinal cord Dmax <50Gy Grade >2 myelopathy (<1%)
  • 67. • No significant difference in the mean PTV(initial) among the groups (p = .24), whereas a significant reduction in mean PTV boost was observed with the MDACC protocol compared with the RTOG protocol • No statistically significant differences were observed in treatment-related acute toxicities and steroid requirements between both arms • Central recurrences: commonest in both groups • Statistically significant differences in QoL favoring those treated with MDACC protocol
  • 68. RTOG VS EORTC • Perfusion- and diffusion weighted MRI potentially add information about the regional blood volume and microstructural architecture of the high-grade glioma. • No difference in progression-free or overall survival was observed • Larger PTVs failed to produce a significant reduction in marginal or distant recurrences
  • 69. 3DCRT VS IMRT/VMAT • Sparing of normal brain • Local boosting of tumor alone to higher doses • IMRT especially useful in presence of critical structures close to- optic nerve, brainstem, dose escalation , etc • IMRT did not significantly improve target coverage compared with 3D-CRT • However, it resulted in a decreased Dmax to the spinal cord, optic nerves, and eye by 16%, 7%, and 15% • It is unlikely that IMRT will improve local recurrence without dose escalation However, it might result in decreased late toxicities associated with radiotherapy • VMAT achieved equal or better PTV coverage and OAR sparing while using fewer monitor units
  • 70. • A-68Ga-Pentixafor fused PET increased uptake in LT F-T region with GBM • B- IHC evidence of CXCR-4 expression on the tumor • The affinity of this tracer help in developing CXCR4 targeting radiotherapeutics • A- 38 yr male pt with GBM radiological evidence of residual disease • B- Repeat scan post 3month (post-op) showed disease progression
  • 71. • GBM patients 60Gy/30# Vs 67Gy/30# ( boost 21Gy/7#) • Slightly higher median OS as compared to historical data that dose escalation is feasible First image- PENTIXAFOR PET show active disease Third image- Phase 1 dose color wash Second image- Target volume delineation Fourth image- dose color wash including SIB
  • 72. • Pt diagnosed with grade (1-4) glioma • Proton vs Photon therapy • PBT associated with improved OS (45.9mo vs 29.7 mo ) to XRT • Retrospective analysis supports ongoing prospective trials comparing the two modalities 2018
  • 73.
  • 74.
  • 75. EXPECTED SURVIVAL OF GLIOMA Nat Rev Neurol. Author manuscript; available in PMC 2020 June 10 HIGH GRADE GLIOMA MEDIAN SURVIVAL Oligodendroglioma - IDH mutant ( 1p/19q codeletion) 17.5 years Oligodendroglioma - IDH mutant ( no 1p/19q codeletion) 6.3 years Oligodendroglioma – IDH wild 1.6 years Astrocytoma - IDH mutant 9.3 years Astrocytoma - IDH wild 1.9 years Glioblastoma - IDH mutant 3.6 years Glioblastoma - IDH wild 1.2 years Glioblastoma MGMT methylated 21.2 mo Glioblastoma MGMT unmethylated 14 mo
  • 76. PSEUDOPROGRESSION • Radiochemotherapy and immunotherapy - transient worsening of contrast enhancement on MRI for several months in approximately 10-30% of patients, sometimes associated with symptoms of intracranial mass-effect • Diagnosis - challenging  Dynamic susceptibility contrast–enhanced MRI: neoangiogenesis → accompanies neoplastic growth; treatment-related changes increase vascular permeability without neoangiogenesis.  Amino-acid PET imaging may be helpful: brain tumours - increased protein synthesis; amino acid tracers →imaging modality.
  • 77. A 31-year-old male patient with prior resection of a right frontal glioblastoma multiforme presented later with a 1-cm enhancing nodule concerning for recurrence compared with pseudoprogression A. Magnetic resonance image of the new nodule. B. Map of relative cerebral blood volume demonstrates increased flow in the nodule (arrow). C. Spectroscopic imaging demonstrates an elevated choline peak compared with D. Normal contralateral brain parenchyma in the same patient
  • 78. DSC perfusion in pseudoprogression & progressive disease A. T1C & rCBV map show enhancing lesion in surgical bed after RT in a patient of GBM , white arrow show rCBV 0.5 in comparison with c/l lobe indicating (Pseudoprogression) B. T1C & rCBV map show enhancing lesion close to the margin of radiation field. White circle show rCBV 2.8 in comparison with c/l lobe indicating predominance malignant cells ( Progressive disease)
  • 79. 56 year old male – GBM – Pseudoprogression A. T1 contrast MRI 1 week after an operation show ring enhancement B. After RT, marked enhancement in the margins following which he developed rt sided hemiparesis C. At that time T1 MRI showed ^se in mass effect , midline shift which turned to soap bubble appearance- radiation necrosis D. Pt improved clinically & after 4months, fading of the enhancing lesion & decrease in mass effect
  • 80. RANO Criteria ▪ 12 weeks post-RT : progression can only be determined if the majority of the new enhancement is outside 80% Isodose line or with pathologic confirmation RANO criteria CR PR SD PD T1 enhancing disease None >50% decrease <50% if but < 25% if > 25% T2/FLAIR Stable/decreased Stable/decreased Stable/decreased increase NEW LESION None None None Present CORTICOSTEROID USE None Stable or Stable or NA CLINICAL STATUS Stable/improved Stable/improved Stable/improved Declined
  • 81. Management of Recurrence • Surgical excision • Chemotherapy- Temozolamide/Carmustine/Lomustine • Bevacizumab-FDA approved • Alternating Electrical field therapy • Re-irradiation • Locally delivered Carmustine (Gliadel) Wafers Implanted into surgical cavity – modest survival advantage with newly diagnosed ¾ Gliomas, or recuurent glioblastoma Patients without systemic treatment options Requires careful patient selection & a gross total resection Studies in GBM have shown only marginal benefit
  • 82. REIRRADIATION ▪ Young patient ▪ Good KPS score ▪ Long interval since previous RT > 6months ▪ Previous dose should be know ▪ Tumor size and location should be taken into account when deciding whether re-irradiation would be safe
  • 83. • ReRT(35GY/10#) and conc bevacizumab Vs bevacizumab(10mg/kg) alone • First prospective randomized multi-institutional study • Median PFS 7.1 Vs 3.8 months (p=0.05) • Median OS was similar • Overall, ReRT was shown to be safe and well tolerated
  • 84. • Re-irradiation of brain tumors – increasingly considered because of developments in radiation technology & imaging make high accurate targeting of recurrent tumors ▪ Further, exploration of the role of re-irradiation on its own or in combination with novel agents is needed Systematic review 2016
  • 85. Stereotactic Radiosurgery • SRS: delivered using a conventional or modified LINAC, a Gamma Knife (Elekta Corp., Stockholm, Sweden) or a robotically controlled miniaturized linear accelerator (CyberKnife). • Radiosurgery appears to be a reasonable option for small well-circumscribed high-grade gliomas that recur after prior conventional large-field radiotherapy and chemotherapy • For lesions larger than 4 cm and/or located in critical regions - Fractionated Stereotactic Radiotherapy • KEY REQUIREMENTS FOR OPTIMAL STEREOTACTIC IRRADIATION: i. Small target/treatment volume ii Sharply defined target ▪ Accurate radiation delivery ▪ High conformality ▪ Sensitive structures excluded from target Properties
  • 86. Brachytherapy & Radiocolloid Solutions • Selection criteria for brachytherapy: i. small size (<5 to 6 cm), ii. tumor confined to one hemisphere, iii. no transcallosal or subependymal spread, iv. well circumscribed on CT or MRI, and v. accessible location for the implant. • A balloon based system, GliaSite, placed into the cavity at the time of surgery - recurrent malignant gliomas whose largest spatial dimension is <4 cm and are roughly spherical. • After treatment planning, the balloon is filled with a liquid that contains organically bound I125 and treatment is completed within 3 to 7 days. • 3D Ir192 HDR BT (Ir Knife) allows intratarget dose escalation with superior conformity. • Stereotactic 125I brachytherapy with TMZ chemotherapy was shown to be effective for treatment of thalamic glioblastoma
  • 87. Targeted Therapies For Recurrent Glioblastoma EORTC 26101 phase III trial exploring the combination of bevacizumab and lomustine in patients with first progression of a glioblastoma • Lomustine (90mg/m2) every six weeks vs lomustine & bevacizumab(10mg/m2) every two weeks f/b best investigation • Median OS 9.1 months vs 8.6 months (p =0.625) • PFS was longer with addition of bevacizumab 4.2 vs 1.5 months • Bevacizumab with progressive glioblastoma despite PFS does not confer a survival advantage • Methlyated group showed a longer median OS 13.5 mo vs unmethylated group was 8 mo
  • 88. Vaccine & immune checkpoint inhibtors for glioblastoma • DCVax- a dendritic cell (DC)-based personalized cancer vaccinewith purified tumor-specific antigens or tumor cell extracts derived from tumor at the time of resection. • Epidermal Growth Factor Receptor Variant III as a Vaccine Target for Glioblastoma • Heat Shock Protein-Based Vaccine • Recombinant nonpathogenic polio-rhinovirus chimera (PVSRIPO) targets the neurotropic poliovirus receptor CD155, which is abundantly expressed on glioblastoma cells. • Phase-III trial of NIVOLUMAB vs BEVACIZUMAB failed to show any survival difference between the two arms
  • 89. CONCLUSION • Molecular markers are well established in adult and pediatric type gliomas • IDH1 mutation is a strongest prognostic marker • There is no definite treatment guidelines for the pediatric type high grade glioma til • Despite of molecular markers, standard of treatment remains same • PCV regimen is more sensitive for oligodendroglial tumor as compared to TMZ • Role of bevacizumab is established in reccurent glioblastoma

Editor's Notes

  1. Point
  2. Oligodendroglioma AND GLIOBLASTOMA- PALIISADING pattern of tumor cells with increased vascularity Rounded nuclei- Perinuclear halo Calcification Acutely branched capillary sized vessels- chicken wire calcifications ASTROCYTOMA Hypercellular tumor Marked nuclear pleomorphism Mitotic figures Lack of endothelial vascular proliferation or necrosis
  3. H3.1g34 padna hai
  4. Mapk- mitogen activated protein kinase Nos- not otherwise specified, can give general diagnosis but not complete diagnosis Nec- not elsewhere specified, mismatch between clinical , histo ,ihc or genetic features
  5. Role f h3.3 nd h3.1
  6. BRAF Role in pilocytic astrocytoma targeted therapy murine sarcoma viral oncogene) Tert- telomerase reverse transcriptase
  7. Study MMSE- orientation, registration ie name three objects, attention, recall, language 30 questionnaire based examination 25-30 no , 20-24 mild, 10-19 mod, <10 severe
  8. There is evidence of hypercellularity as evident by low diffusivity on ADC map
  9. Naa decreased in glioma & choline and lactate(necrosis) increased in glioma
  10. Realtime navigation is use with intra op usd, inta op mri and awake craniotomy
  11. that redosurgery can be done, after 24 hrs hemoragic components and hemosiderin component predominant
  12. Anti epilptics first steroids duration
  13. MGMT gene(10q26) Encodes DNA repair proteinRemoves alkyl groups from the O6 position of guanineResistance to alkylating agentsMethylation of the MGMT promoter silences expression of the protein Cytotoxicity and apoptosis by alkylating agent
  14. Role in reccurent settings
  15. TMZ was not inferior to RT 25/5 WAS NOT INFERIOR TO 40/15
  16. Pfs is better for odg tgan astrocytoma
  17. Procarbazine - 75mg/m2 per oral daily D8-D21 Lomustine - 110mg/m2 per oral D1 Vincristine - 1.4mg/m2 i.v. D8 & d29
  18. Partial brain radiation
  19. GLIOBLASTOMA WE WONT DO WHOLE BRAIN IRRADIATION & wbrt recommended for multifocal disease
  20. Hypervascular retains contrast
  21. EORTC and MDACC guidelines differ from RTOG guidelines in their approach toward inclusion of peritumoral edema in the treatment volume, whereas RTOG and MDACC guidelines differ from EORTC in the concept of boost phase
  22. PTV margins account for variations in set-up and reproducibility. Reducing PTV margins to modify organ at risk (OAR) dose(s) is not generally permissible. However, OAR must be defined, along with a planning risk volume (PRV) for each OAR. Each PRV will be its OAR plus 3 mm The rationale for peritumoral edema inclusion is that histologically identified microscopic tumor cells have been found in these areas A potential disadvantage to such an approach is that this may lead to larger radiation treatment fields that irradiate a larger volume of brain than is necessary when there is significant edema Basis: most recurrence within 2cm of primary tumor. Firstly,T2/FLAIR signals can substantially fluctuate depending on tumour mass-effect and postoperative oedema. Secondly, using the entirety of T2/FLAIR hyper-intensity signals to define the CTV (if not using a sequential decreased boost volume), will often translate into a target volume associated with an irradiation dose/volume beyond the tolerance of the normal brain
  23. Trim karna hai isko yaa change krna hai
  24. OS comparable to that of conv 60gy with tmz 14.1 months No added survival benefit- in dose escalation
  25. ADD ANY DATA
  26. Table update krni hai
  27. DUE TO THE LEAKY VASCULATURE, psuedoprogression occur in 6months & radiation necrosis occur in 1 year
  28. Tumor r CBV from 0.5 to 4.84 DSC
  29. 2d dimensional method- bi- perpendicular dimension , biopsy is confirmative
  30. Advantage – Minimal systemic toxicity,no limitation posed by the BBB,delivery of very high local concentrations of chemotherapy
  31. Data- AND CHANGE KRNA HAI PHOTO ISKI 1 SRS- 18-25Gy , 1 HYPO- 30-43Gy (5to18#) 3 conventional- 45 Gy in 25# Criteria- favourable location, ability to spare oar , no previous use of bev failure, consider srs pr hypo
  32. Small in field recurrence – T1 enhancing volume+ 0.5-1cm margin depending on the grade of tumor Marginal recurrence- T1 enhancing volume + 1cm margin
  33. 57 yr pt with gbm volume is 12.3ml treated with srs 18gy single# and scnd one pt of gbm with vol 123ml with nearby critical structure fractionated srt 36Gy /6 #
  34. Radiobiology: higher proportion of tumor cells killed with ablative doses of radiation → expose more tumor antigens to stimulate immune responses both against the irradiated tumor and also possibly against more distant nonirradiated tumor (referred to as the abscopal response)
  35. MAb that binds to VEGF and inhibits the growth of tumor blood vessels