SlideShare a Scribd company logo
1 of 118
PRESENTER- DR. JASMEET SINGH TUTEJA
MODERATOR- PROF. SEEMA GUPTA
 Gliomas arise from supportive cells (glial cells) that surround nerve cells.
 Most common type of primary brain tumours.
 4 types of glial cells-
 Astrocytes- balance of chemical in brain, wound healing & repair.
 Oligodendrocytes- myelin synthesis
 Ependymal cells- CSF (ventricular lining)
 Microglial cells- defend brain from disease- causing factor.
Different type of gliomas vary in treatment management & prognosis.
EPIDEMIOLOGY
Incidence - 17th position
Death – 12th position
 Incidence – 10th position
 Death – 10th position
Environmental
 Ionizing radiation (meningioma, gliomas, sarcomas) - 2.3% incidence in
prophylactic cranial irradiation in children
 Petrochemicals (formaldehyde, vinyl chloride, acrylonitrite)
Hereditary
 Cowden, Turcot , Lynch and Li-Fraumeni (gliomas)
 Gorlin (PNET),
 Neurofibromatosis I & II (meningiomas, optic nerve gliomas, schwannoma)
 Von Hippel Lindau syndrome (hemangioblastoma)
first-degree family member shown to have increase risk approximately two-fold
WHO classification Lyon 2007 –
 Histological criteria
 Morphological criteria
 In 2014, a meeting held in Haarlem, the Netherlands, under the auspices of the
International Society of Neuropathology, 2016 WHO Classification was formed
attended by 117 contributors from 20 countries to formulate guidelines
Why new classification ?
 Inter/ Intra observer variation
 Traditional histologic grading does not provide good prognostic power.
 Within the same tumour subtype and grade – a lot of variation in prognosis
Primary focus was on:
• Molecular markers
• Hence, Integrating Genotypic & Phenotypic parameters to make final
diagnosis
NEW CHANGES:
 Molecular parameters were introduced
 More objective and more precisely defined entities
 Improved tailoring of patient therapy
 Better classification for research purpose
 Wastebasket categories: NOS
 Need of more focused study of these less defined group => clarification of their status
Can we rely on molecular parameters alone? & skip
histology?
9
10
Brainstem
• Difficulty swallowing, speaking
• Drowsiness
• Headaches
• Hearing loss
• Muscle weakness
• Hemiparesis
• Uncoordinated gait
• Vision loss, ptosis, strabismus
• Vomiting
Frontal lobe
 Behavioural and emotional changes
 Impaired judgement , Impaired sense of smell
 Memory loss
 Hemiplegia
 Cognitive dysfunction
 Vision loss
 Papilledema Temporal lobe
• Impaired speech
• Seizures
• Homonymous superior
quandrantanopsia
• Auditory hallucinations
• Abnormal behaviour
Occipital Lobe
• Visual field deficits
• Visual hallucinations
Parietal lobe
 Impaired speech
 Inability to write
 Lack of recognition
 Seizures
 Spatial disorder
 Imaging – MRI, CT , PET
 Biopsy (craniotomy/ stereotactic)
 CSF
 IHC
 Cytogenetics
18
 Investigation of choice
MRI sequences :
 T1 weighted
 T2 weighted
 T2 weighted FLAIR
 T1 post contrast
 Diffusion weighted imaging
 Magnetic resonance spectroscopy
 Magnetic resonance perfusion study
Anatomical details
 CSF – dark
 Gray matter – gray
 White matter - white
 Most Pathologies – dark
Anatomical details of T2W
 Pathological details
 Gray matter – bright
 White matter – dark
 CSF- bright
FLAIR
 Similar to T2 except
free water
suppression
(inversion recovery)
 Pathology – bright
 Good for lesions near
ventricles or sulci
 Diagnosis - made on axial images.
 T2Wt- and FLAIR - display the margins of a tumor and its surrounding edema or a
direct tumor infiltration.
 The sagittal & coronal images - confirm exact location
 Non-invasive physiologic imaging that
measure relative levels of tissue
metabolites
 Complements MRIs
 Guidance for stereotactic biopsy.
 Differentiates between Psuedoprogression &
Recurrent gliomas.
 11C Methionine (MET-PET)- uptake is
correlated with tumour proliferation  superior
contrast & tumour delineation.
 FET-PET (18F- fluoro-ethyl-L-tyrosine)-
longer half life
 Stereotactic frame or scalp fiducials
 CT or MRI is performed and the data are loaded into an image guidance
system.
 A target and entry points are selected, and the trajectory is visualized on a
workstation.
 The entry point - patient’s scalp, and a small burr hole or twist drill hole is
made.
 The biopsy needle is passed to the appropriate depth, and tissue samples are
obtained.
 Essential for staging tumors with a propensity for CSF spread (medulloblastoma,
germ cell tumors, and CNS lymphoma).
 best done before surgery or more than 3 weeks after surgery, as long as intracranial
pressure is not elevated.
 Tumor markers in the CSF may help in making the diagnosis.
Important exception is pilocytic
Astrocytoma which enhances with
contrast because the blood vessels
go degenerative changes and in
contrast the HGGs enhance
because because of microvascular
proliferation.
How to recognise blood in
the MRI?
– look for severe hyper
intensity with a surrounding
black border in the FLAIR
sequence
Cerebral metastases can often be confusing with HGG.
There are some useful characteristics when trying to differentiate
these radiologically.
1. cerebral mets usually occur in the gray/white junction,and
less common in deeper brain,
2. do not involve the periventricular white matter and
3. rarely ever goes to the corpus callosum.
4. cerebral mets are well circumscribed usually and they do not
have multiple areas of heterogenous contrast enhancement –
i.e. looks like a single round bright rim with a single dark
centre in T1w image with Contrast.
In contrast GBM – centers on the sub-cortical white matter, with
subependymal and corpus callosum extension seen frequently. There
can be multifocal GBM that looks like many small GBMs but they are
all embedded in a single unit / area of FLAIR enhancement.
31
WHO
Grade
Definition Gliomas
I Circumscribed tumors of low proliferative
potential associated with the possibility of cure
following resection (Low proliferation)
Pilocytic astrocytoma, Pleomorphic
xanthoastrocytoma, Ganglioglioma,
Subependymal giant cell astrocytoma
II Infiltrative tumors with low proliferative
potential with increased risk of recurrence
(Nuclear atypia)
Diffuse astrocytomas (IDHmut/IDHwt) and
oligodendrogliomas (IDH mut, 1p19q codel)
III Tumors with histologic evidence of malignancy,
including nuclear atypia and mitotic activity,
associated with an aggressive clinical course
(PLUS Mitoses)
Anaplastic astrocytomas (IDHmut/IDHwt) and
anaplastic oligodendrogliomas (IDH mut,
1p19q codel)
IV Tumors that are cytologically malignant,
mitotically active, and associated with rapid
clinical progression and potential for
dissemination (PLUS Necrosis Vascular
proliferation)
Glioblastoma (IDH mut/wt)
CLINICALLY RELEVANT MOLECULAR MARKERS
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
Prognostic 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 grade II
Diffuse astrocytoma 70%–80% 15% 40%–50%
Oligodendroglioma/
oligoastrocytoma
70%–80% 30%–60% 60%–80%
WHO grade III
Anaplastic astrocytoma 50%–70% 15% 50%
Anaplastic
Oligodendroglioma/
oligoastrocytoma
50%–80% 50%–80% 70%
WHO grade IV
Glioblastoma 5%–10% <5% 35%
 Neuroradiologists
 Neurosurgeons
 Neurologists
 Neuropathologist
 Radiation oncologists
 Medical oncologists
 Neuro-rehabilitation
3/7/2022 33
LOW GRADE
GLIOMA
 Grow relatively slowly
 Diffuse infiltrative pattern
 Most transform in high grade gliomas- with aggressive clinical course &
shortened OS
 Treatment modalities include: surgery, radiotherapy, & chemotherapy
 Management of LGG is complex & controversial- MDT is essential.
 Estimated risks & benefits of adjuvant therapies (PFS, OS & neurocognitive
preservation) should be discussed with the patient whenever possible.
36
37
T2-FLAIR
MISMATC
H SIGN
 The T2-FLAIR mismatch sign- highly specific
radiogenomic signature for DIFFUSE
ASTROCYTOMA (IDH-mutant, 1p/19q-non-
codeleted molecular status)
 Helps in distinguishing a diffuse astrocytoma from
an oligodendroglioma.
 On T2 weighted- tumours have extensive areas of
fairly homogeneous and strikingly high signal.
 On T2-FLAIR - majority of these areas become
relatively hypointense in signal due to incomplete
suppression.
 At the margins of the tumour, a rim of hyperintensity is
usually seen.
38
39
41
 CONSIDER MAXIMAL SAFE RESECTION
 AIM TO:
1. Obtain a histological & molecular diagnosis
2. Remove as much as tumor as safely as possible
3. To preserve or improve QOL especially by controlling seizures.
 Why?
-Greater extent of resection can improve OS
 How?
-Consider intraoperatively MRI & U/S, awake craniotomy with
language & other appropriate functional monitoring.
 Extent of resection & post surgical residual volume are independent prognostic
factor significantly associated with longer OS.
 Surgical excision is also independent prognostic factor correlating with increased
malignant PFS*
Advantage of extensive resection:
 Across all molecular subtypes.
 Even min. residual volume negatively affects OS- most relevant for IDH-mut
astrocytomas (in oligodendroglioma – trend towards better OS)
 Consider 2nd look op to remove minor residuum if safely possible.
 Post-operative radiotherapy
 Decisions largely based on risk factors and extent of resection
• No overall survival benefit from immediate RT vs delayed RT (7.4vs 7.2 yrs)
• Improved progression free survival with immediate RT by ~24 months but at expense of
toxicity (e.g. neuro-cognitive) median 5.4 vs 3.7 years
• Better seizure control (25% vs 41% at 1 yr)
Absence of OS benefit for early RT suggest that RT slows
progression of LGG’s but does not prevent malignant
transformation & that RT given at time of radiological PD is
equally effective.
• Grade 2 glioma (any subtype).. High risk
• <40 AND subtotal resection
• >40 years (any biopsy/surgery)
• 54Gy in 30 fractions +/- 6 x PCV q8 weekly
• Improved overall survival with addition of PCV
• Median: 13.3 vs 7.8 years
• 10 year OS: 60% vs 40%
• Largest benefit in oligodendroglioma
• Use for oligodendrogliomas as above
• Not significant for astrocytoma….. In practice, tend not to use adjuvant chemotherapy here.....
Wait & watch.
• Improved progression free survival with PCV
• 10 year PFS: 21% vs 51%
Prognostic factors for survival in adult patients with cerebral low-grade glioma.
IMAGING SUGGESTIVE OF LOW GRADE
GLIOMA
>80-90% resection
possible?
Resection (often awake craniotomy)
Biops
y
Observation to
determine
behaviour
Ye
s
N
o
Integrated diagnosis (inc cytogenetics IDH, 1p 19q, MGMT, TERT) confirms
G2
Assess risk factors for progression: Age >40, sub-total resection / biopsy, size >6cm,
crosses midline, poor cytogenetics, symptomatic (but not seizures), adverse radiology (eg
enhancement)
High Low
Decide with patient whether to use immediate non-surgical treatment or
observe
(influenced by level of risk and patient’s wishes)
Observation
(Consider more surgery
or non-surgical
treatment if progresses
Q: Is it high grade
now?)
Non-surgical
treatment
Olig
o
RT +
Astrocytom
a
R
?treat as GBM
if G2
astrocytoma
?chemo
alone if
gliomatosis
HIGH GRADE GLIOMA
56
Anaplastic
Astrocytoma
Anaplastic
Oligodendroglioma
T1 T2 T1W + Gad
GLIOBLASTOMA
 Stereotactic image guided biopsy is accurate & safe way to get to even relatively
inaccessible areas of the brain such as basal ganglia & thalamus.
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.
Simpson et al , IJROBP 1993
 Patients with grade III (=anaplastic) oligodendroglial tumours
 RT vs RT (59.4Gy) + adjuvant PCV
 Median OS: 42.3 vs 30.6 months
 PFS also improved (Median PFS: 24.3 VS 13.2 Months)
 Improved outcomes also in patients with IDH mutations and methylated tumours
 Therefore: consider adjuvant PCV for grade III oligos (by modern classification these are all co-
deleted)
More benefit in patients with 1p19q co-deletion: trend towards improved OS here, not
in non-co-deleted.
Overall survival in both treatment arms for (A) the patients with 1p/19q-codeleted tumours
(B) the patients with non–1p/19q- codeleted tumours
Progression Free survival in both treatment arms for (A) the patients with 1p/19q-
codeleted tumours (B) the patients with non–1p/19q- codeleted tumours
CO-DELETED TUMOUR- OS was not
reached in RT+PCV vs 112 months in RT group
NON-CODELETED TUMOUR- OS 25 vs 21
months
CO-DELETED TUMOUR- PFS- 157
VS 50 MONTHS (RT + PCV VS RT)
NON CO-DELETED TUMOUR- 15
VS 9 MONTHS (RT + PCV VS RT)
59.4 Gy/33# of
RT
PCV → 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 2.7 y 2.6 y
PFS - 1p/19q non codeleted AO/AOA 1 y 1.2 y
Adults (>18) with newly diagnosed 1p/19q World
Health Organization (WHO) grade III
oligodendroglioma were randomized in 3 arms:
• Radiotherapy (RT-59.40Gy) alone;
• RT with concomitant and adjuvant temozolomide; or
• TMZ alone
INTERIM RESULT OF CODEL TRIAL:
 TMZ-alone patients had significantly shorter PFS than patients treated on
Radiotherapy arms.
 The ongoing CODEL trial trial has been redesigned to compare RT + PCV versus
RT + TMZ.
Design- Patients with histologically confirmed Anaplastic Astrocytoma were randomized to 2 arms-
• RT+TMZ
• RT+NU
Results. Median survival time was 3.9 (RT/TMZ arm) vs 3.8 years (RT/NU ARM).
Conclusions. No significant improvement in OS , PFS & TTP in both arms
RT+TMZ was better tolerated.
IDH1-R132H mutation was associated with longer survival.
oSlightly better PFS with TMZ
oHigher toxicity and discontinuation rates with nitrosureas
IDH positive with a better OS in IDH-positive patients
(median survival time 7.9 vs 2.8 y)
• Grade III gliomas which were NOT 1p19q co-
deleted
• Improved overall survival in arms with adjuvant
temozolomide
• Larger advantage in MGMT methylated (70%)
• Therefore, consider adjuvant temozolomide for
non-co-deleted Grade III gliomas (now =
anaplastic astrocytoma).
The 2nd interim analysis declared futility of concurrent TMZ
(median OS was 66.9 months with concurrent TMZ vs 60.6 months
without concurrent TMZ)
By contrast, adjuvant TMZ improved improved OS compared with
no adjuvant TMZ (median OS 82.3 months vs 46.9 months)
Adjuvant TMZ therapy, but not concurrent TMZ chemotherapy, was
associated with a survival benefit in patients with 1p/19q non co-
deleted anaplastic glioma
https://doi.org/10.1016/S1470-2045(21)00090-5
Design- Patients with anaplastic gliomas randomized 2:1:1 In 3 arms:
• standard radiotherapy (RT) (arm A),
• Procarbazine, lomustine and vincristine (PCV) (arm B1), or
• Temozolomide (TMZ) (arm B2).
There is no differential activity of primary chemotherapy versus RT in any subgroup of
anaplastic glioma. Molecular diagnosis is superior to histology.
http://www.jnccn.org/content/12/5/665.full
Patient were randomized in 2 arms:
• RT alone (60Gy) vs
• RT+TMZ  TMZ (6cycles)
Median PFS 6.9 vs 5 months
Median Survival- 14.6 vs 12.1 months
2- year survival rate- 26.6 vs 10.4 months
• GBM: 60Gy in 30 fractions RT +/- concurrent temozolomide 75mg/m2 then 4 week
break then adjuvant temozolomide 150-200mg/m2 x 6 months
• Given with prophylactic oral Septrin (co-trimoxazole) 960mg during RT
• Addition of temozolomide improved overall survival:
• Median 12.1 months vs 14.6 months
• 2 year OS: 10.4% vs 26.5% (unheard of in GBM before)
• Well tolerated
• 90% completed at least 90% of planned temozolomide
• 7% grade 3 or 4 toxicity
• Patients with complete resection did better
• 2 year OS: 37%
• Patients with methylated MGMT did better
• 2 year OS: 46%
MGMT promoter methylation has been
demonstrated as the strongest prognostic
& predictive marker for outcome, and the
added benefit of Temozolomide.
MGMT
gene(10q26)
Encodes DNA
repair protein
Removes alkyl
groups from
the DNA
(DNA repair)
TMA alkylates
O6 position of
guanine
Resistance to
alkylating
agents
(removal of
alkyl group by
MGMT)
Methylation of
the MGMT
promoter
silences
expression of
the protein
Cytotoxicity
and apoptosis
by alkylating
agent (TMZ)
 Retrospective study (morning vs evening intake)
 Survival benefit in median OS when patients take TMZ in
morning versus in evening. (mOS of 2.13 years in AM vs
1.63yrs in PM)
Morning TMZ administration improves survival with patients with GBM.
https://doi.org/10.1093/noajnl/vdab041
82
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 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.
83
Role of Bevacizumab in newly diagnosed GBM
Citation Arms Median OS
(months)
RTOG 0825
Gilbert NEJM 2014
60 Gy + TMZ → TMZ 16.1
60 Gy + TMZ → TMZ + BEV 15.7
AVAglio
Chinot NEJM 2014
60 Gy + TMZ → TMZ 16.7
60 Gy + TMZ → TMZ + BEV 16.8
No significant benefit in median OS on addition of Bevacizumab.
ELDERLY GBM
• Nordic and German trial have randomized patients into 3 arms- conventional RT,
hypofractionated RT (34GY in 10#) and temozolomide alone
• Interpretation- Standard Radiotherapy was associated with poor outcome in patients
more than 70 years.
• Both TMZ & hypofractionated RT were associated with longer survival & can be
considered as standard treatment options in elderly GBM.
https://doi.org/10.1016/s1470-2045(12)70265-6
 Age >70 is a dreadful prognostic factor in
GBM
 Perry et al NEJM 2017 patient were
randomized in 2 arms:
 RT 40Gy in 15 fractions +/-
temozolomide concurrent
75mg/m2 and adjuvant 150-
200mg/m2 for up to 12 cycles
 Patients ≥65 years, PS 0-2
 Addition of TMZ improved overall
survival
 9.3 months vs 7.6 months
 Addition of TMZ improved PFS
 5.3 months vs 3.9 months
 Consider TMZ-RT 40Gy in 15 fractions
and adjuvant TMZ if >70 and PS 0-2
• Patients with Butterfly Glioma have almost poor performance status.
• Poor PS patients have dreadful outcome
• PS 3 and <70 years: consider 30Gy in 6 fractions over 2 weeks (or 25Gy in 5
fractions over one week (Roa et al JCO 2015))
• <70 years, GBM
• PS 0/1: chemoRT 60Gy in 30 fractions + adjuvant temozolomide x 6
• PS 2 (not butterfly): 60Gy in 30 fractions
• PS 3 (or PS 2 butterfly glioma): 30Gy in 6 fractions over 2 weeks
• PS 4: steroids and best supportive care
• >70 years, GBM (?top age)
• PS0-1: chemo RT 40Gy in 15 fractions + adjuvant temo up to 12 cycles
• PS 2: possibly temozolomide alone or hypofractionated RT (need MGMT) status.
Consider BSC.
• PS3/4: steroids and best supportive care
• <70 years, grade III
• PS 0-1: RT 60Gy in 30 fractions
• 1p19q co-deleted oligo: adjuvant PCV
• Non-1p19q: adjuvant temo
https://www.nice.org.uk/guidance/ng99/resources/visual-summary-for-glioblastoma-management-pdf-
90
Tumor-treating fields involve
• Transducer arrays are placed on scalp. These are connected to electric field generator. Light weight
battery pack is carried by patient and worn for atleast 18 hours a day. Transducer arrays delivers low
intensity, intermediate frequency, alternating electric field which acts by disrupting mitosis and
inducing 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
91
oKPS > 70; median age 56
oRandomized 2:1 after 60Gy, TMZ
Adj TMZ + TTF
Adjuvant TMZ
oImproved median survival from time
of randomization
16→20.9 months
oToxicity – dermatitis in 52%
oImproved OS:
>18 hours/day
 Corticosteroids, preferably dexamethasone reduce symptomatic peritumoral vasogenic
edema, neurological deficits and signs of increased intracranial pressure
 Lowest dose of steroids – shortest time possible
 No need for prolonged steroid therapy after tumour resection or for prophylaxis during
radiotherapy in asymptomatic patients
 Downward titration of dose – whenever possible
 Extensive mass effect – receive steroids for at least 24 hours before RT
 Monitor for side effects
 Chronic corticosteroids (≥ 20 mg prednisone equivalents daily for ≥ 1 month) - consider
prophylaxis for osteoporosis and pneumocystis jerovecii pneumonia should be considered
94
Antiepileptic Drugs
▪ Increased risk of thromboembolic events due to a tumour-induced hyper-coagulable state, as a
consequence of neurological deficits, immobilisation and steroid use.
▪ Prophylactic anticoagulation is not recommended
▪ No contraindication for the use of standard anticoagulants in patients with proven thrombosis.
Anticoagulants
▪ Non Enzyme inducing anti seizure medications should be used eg: levetiracetam, topiramate,
valproic acid
▪ Current guidelines recommend tapering AEDs 1-2 weeks after surgery and avoiding long-term
prophylaxis.
▪ No role for primary perioperative prophylaxis (i.e. in patients who have never had a seizure).
 SUPINE, HEAD IN NEUTRAL POSITION
 HEAD IMMOBILISED- Perspex or thermoplastic shell.
More rigorous immobilization with a stereotactic frame.
 I.V. contrast 1mg/kg most of time 50mg i.v.
 Thin-slice CT imaging is recommended (at 1–2 mm
slices) from the vertex to the C3-C7 and, for ease of
fusion, ideally should match the MR image slice
thickness.
 Multimodal MR fusion, using at least the contrast
enhanced T1 and FLAIR sequences
 PET/CT fusion can be helpful distinguish between
postradiation changes and disease recurrence on MR
imaging.
 3D-CRT remains standard for the majority of
GBM
 IMRT/ VMAT is - volumetrically or spatially
challenging tumours.
 Proton beam radiation therapy due to its
“Bragg peak effect, safer than traditional photon
radiation therapy
 Boron neutron capture therapy (BNCT),
increases the efficacy in destruction of tumor
• Superior: skin fall off
• Anterior: skin fall off
• Posterior: skin fall off
• Inferior: cranial base
2 opposed lateral fields.
 A rectangular treatment field over the
cranial vault with the collimator angled so
as to place the inferior border 1–2 cm
below a line drawn from the medial
canthus to the mastoid tips.
 A small block is carefully placed to shield
the globes while still allowing adequate
coverage of the anterior–inferior extent of
the cranial contents
Whole-brain RT
Anaplastic astrocytoma Phase 1 Phase 2
60 Gy in 30# 46 Gy in 23# 14 Gy in 7#
59.4 Gy in 33# 45 to 50.4 Gy in 1.8 Gy/# 9 to 14.4 Gy in 1.8 Gy/#
Anaplastic
oligodendroglioma
Phase 1 Phase 2
60 Gy in 30# 50.4 Gy in 28# 9 Gy in 5#
Studies interrogating dose escalation beyond 60 Gy using standard
fractionation have not demonstrated any survival benefit.
LGG- 54Gy in 30 fractions or 50.4Gy in 28 fractions (EORTC 22844)
 GTV- non-enhancing tumour extent (T2/FLAIR)
 CTV- GTV+1-1.5cm
 PTV- CTV + 0.5cm
EORTC treatment volumes 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)
GT
V
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)
GT
V2
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 PTV
2
CTV2 plus a margin of
3–5 mm
*Margins up to 3 cm were allowed in 22981/22961 trial, and 1.5 cm in 26981–22981 trial.
TARGET DELINEATION OF GLIOBLASTOMA
Stupp et al.(2005) Chinot et al (2014) Gilbert (2014)
RTOG- 2 phases of target volume delineation.
(A) The initial GTV includes postoperative peritumoral edema based on the
axial T2 fluid-attenuated inversion recovery sequence (red line); the initial
CTV (green line) includes postoperative peritumoral edema plus a 2 cm
expansion in all directions.
(B) The boost GTV includes the surgical cavity and residual enhancement
based on the axial T1 sequence with gadolinium (red line) and the boost
CTV is a 2 cm expansion in all directions (green line)
EORTC-1 phase of target volume
delineation.
GTV: surgical cavity and residual
enhancement based on the axial T1
sequence with gadolinium (red line) and
CTV is a 2 cm expansion in all directions
(green line).
Zhao et al Onco Targets Ther 2016
 Principles of intensity-modulated
radiotherapy (IMRT)-based planning for
dose-escalated HFRT.
 The two regions are prescribed differential
doses using IMRT, with the high-dose area
receiving higher tumour biological
effective dose from increased dose and
higher fraction size.
 This technique is inappropriate for
tumours in close proximity to critical
structures.
Hingorani et al 2012 BJR
104
From left to right: transition from 2D RT to 3D RT to intensity modulated
radiotherapy to intensity modulated proton therapy harnesses the potential
for sparing normal, uninvolved brain substructures from unnecessary RT
dose; whether this produces meaningful patient clinical benefit is a subject of
current clinical trial testing.
2D 3D IMRT
IM Proton
Therapy
105
OAR OBJECTIVE(S)
Brainstem D <54 Gy; 1–10 cc < 59 Gy (periphery)
Chiasm Dmax <55 Gy
Cochlea Ideally one side mean <45 Gy ( Most protocols allow
ipsilateral cochlea to receive 60 Gy rather than compromise
dose )
Eyes Macula <45 Gy
Lacrimal glands Dmax <40 Gy
Lens Ideally <6 Gy Max 10 Gy
dose limits should never compromise PTV dose
Optic nerves and chiasma Dmax <55 Gy or 1 % of PTV cannot exceed 60 Gy
Pituitary Dmax <50 Gy
Hippocampus Max ≤16 Gy and no more than 9 Gy to 100 % volume
OAR definitions and dose limits in GBM patients
(individual adaptation necessary according to the clinical situation)
M. Niyazi et al. / Radiotherapy and Oncology 118 (2016)
Based on the University of Maryland treatment planning guidelines
ACUTE
 Fatigue
 Erythema
 Alopecia
 Headache
 Nausea
 Vomiting
LATE
 Somnolence
 Neurocognitive impairment –
hippocampal sparing)
 Brain Necrosis
FOLLOW UP
 Brain MRI every 3-6 month followed by serial imaging up to 3-5 years & then
atleast annually is recommended according to NCCN followed with clinical
correlation
• Occurs in about 30% of patients
• Most within 3 months of chemo-RT
• Represents endothelial damage and subsequent hypoxia
• Scan looks worse, patient often well, but not always
• Increased enhancing lesion
• Increased FLAIR
• Tricky to differentiate from recurrence
• Advanced imaging/ Radiologist may help
• Reduced relative CBV (cerebral blood volume) supports diagnosis of pseudo-progression
(increased in recurrence)
• MR-spectroscopy: Low choline:NAA ratio (≤1.4) or low choline (=low levels of metabolites)
or high lactate supports diagnosis of pseudo- progression
• Diffusion weighted imaging: Elevated ADC (apparent diffusion coefficient) supports diagnosis
of pseudo-progression= less impedance of water motion (in recurrences have lots of cells
impede water motion)
First progression Definition
Progressive disease < 12
weeks after completion of
chemoradiotherapy
Progression can only be defined using diagnostic imaging if there is new enhancement outside of
the radiation field (beyond the high-dose region or 80% isodose line) or if there is unequivocal
evidence of viable tumor on histopathologic sampling
Note: Given the difficulty of differentiating true progression from pseudoprogression, clinical
decline alone, in the absence of radiographic or histologic confirmation of progression, will not be
sufficient for definition of progressive disease in the first 12 weeks after completion of concurrent
chemoradiotherapy.
Progressivedisease≥ 12
weeks after
chemoradiotherapy
completion
1.New contrast-enhancing lesion outside of radiation field on decreasing, stable, or increasing
doses of corticosteroids.
2.Increase by ≥ 25%in the sum of the products of perpendicular diameters between the first
postradiotherapy scan, or a subsequent scan with smaller tumor size, and the scan at 12 weeks or
later on stable or increasing doses of corticosteroids.
3.Clinical deterioration not attributable to concurrent medication or comorbid conditions is sufficient
to declare progression on current treatment but not for entry onto a clinical trial for recurrence.
 Surgical resection
 Chemotherapy: TMZ/ Carmustine/ Lomustine
 Locally delivered CARMUSTINE OR GLIADEL wafers:
o Implanted into the surgical cavity →modest survival advantage
o It offers statistically significant but marginal improvement (31 vs 23
weeks) in OS after re-operation of HGG.
o Significant peritumoral edema is a recognized side-effect.
o Requires careful patient selection and a gross total resection.
o Specialist centres only
 Trial: Westphal et al
 HGG: maximum surgical resection +/- gliadel wafers, followed by RT
 2.3 month increase in median survival with gliadel
 11.3 months to 13.9 months
 Greater gain (4.2 months) if ≥90% resection 110
 Bevacizumab-FDA approved
 Alternating Electrical Field Therapy
 Re-Irradiation
 Best Supportive Care
111
▪ Cognitive deficits, personality changes, and mood disturbances are major comorbidities.
▪ “Chemobrain” - severe cognitive dysfunction that can persist after the cessation of
treatment↔ chemotherapeutic agent-induced inflammation in the hippocampus, which is
involved in learning and memory
▪ Endocrinopathies are common – surgery, steroid, RT – monitor and manage
▪ Psychiatric comorbidity treated with psychotherapy and pharmacotherapy
▪ Early discussion of goals of care and involvement of palliative care should be considered
▪ Diverse support - nurse specialists in neuro-oncology, social workers, counsellors,
clinical psychologists, palliative-medicine professionals, and patient focus groups.
▪ Rehabilitative measures should be explored during and after tumour-specific therapy
 Externally generated ionizing radiation is used to eradicate a
define target without the need to make an incision.
 Target is defined by high resolution sterotactic imaging.
 Usually delivered in single session (maximum of 5) using
stereotactic guiding device. ( strict immobilization)
 Multiple non opposed radiation beams converge on target in
the brain avoiding normal tissues.
 Highly conformal
 Can be delivered using a conventional or modified LINAC, a
Gamma Knife or a robotically controlled miniaturized linear
accelerator (CyberKnife), particle beam accelerator or
multisource Co60 unit
Leksell Stereotactic system
trUpoint ARCH SRS/SRT Syste
113
 Used 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:
 Small target/treatment volume
 Sharply defined target
 Accurate radiation delivery
 High conformality
 Sensitive structures excluded from target
Radiobiology: achieving increased cell kill from a higher dose per fraction → 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) and reducing the effect
from accelerated tumour cell repopulation by shortening the overall treatment time
114
 4 phases:
1. placement of the head frame,
2. imaging of the tumor location,
3. computerized dose planning, and
4. radiation delivery
SRS dosing: RTOG 9005
results
< 2 cm 24 Gy
2.1 -3 cm 18 Gy
3.1 – 4 cm 15 Gy
115
BEVACIZUMAB
o Monoclonal antibody that binds to VEGF and inhibits the growth of tumor blood
vessels
o Prolongs OS either alone or in combination with a cytotoxic agent by about 4
months in recurrent glioblastoma but not in primary Glioblastoma
CILENGITIDE- no established role (CORE trial)
ANTIANGIOGENIC THERAPY
Vaccines for Glioblastoma
o DCVax- a dendritic cell (DC)-based personalized cancer vaccine with purified tumor-
specific antigens or tumor cell extracts derived from tumor at the time of resection.
o Epidermal Growth Factor Receptor Variant III as a Vaccine Target for Glioblastoma
o Heat Shock Protein-Based Vaccine
o Recombinant nonpathogenic polio-rhinovirus chimera (PVSRIPO) targets the
neurotropic poliovirus receptor CD155, which is abundantly expressed on glioblastoma
cells.
116
Biomarkers
• EGFRvIII amplification is targeted by EGFR vaccine rindopepimut.
• KIT amplification or mutation is target is targeted by KIT inhibitor imatinib
• PDGFRA amplification is targeted by PDGFR inhibitor dasatinib
• PTEN deletion or mutation is targeted by AKT inhibitor or mTOR inhibitor voxtalisib
• MDM2 amplification is targeted by a MDM2 inhibitor such as AMG232
• TP53 wild-type is targeted by a MDM2 inhibitor such as AMG232
• RB1wild-type is targeted by CDK4/6 inhibitor ribociclib
MicroRNAs→ Dysregulation → pathogenesis of glioblastoma
Targeting miRNA(s) could alter multiple genes simultaneously and may prove more effective
than targeted focused at targeting single gene or pathway
CAR-T cell therapy
• Multiple infusions into the ventricular system of CAR-T cells targeting the tumor-associated
antigen IL-13 receptor alpha 2 (IL13Ra2) in a patient with recurrent multifocal
• Intravenous delivery of a single dose of autologous CAR-T cells targeting EGFRvIII
mutation in patients with recurrent GBM → feasible and safe
117
Convection-Enhanced Delivery: use of intracerebrally implanted catheters to deliver a drug
into the brain parenchyma or tumor, at a slow but continuous rate of flow.
• Topoisomerase-I inhibitor, topotecan
• Thermosensitive liposomes can encapsulate drugs to release them at the target site
• Angiopep-2 (An2) for BBB transcytosis and anti-CD133 MAb for specific delivery to glioma
stem cells have been incorporated in a dual-targeting immunoliposome encapsulating
• Targeting molecule transferrin helps the transport of drug to glioblastoma which contains
abundant transferrin receptors on the surface
• Genetically engineered bacteria that selectively destroys tumor cells while sparing the
normal brain tissue
Direct Delivery of Therapeutic Agents
Gene Therapy
• Viral vector mediated insertion of drug sensitivity genes
• Insertion of tumor suppressor genes: Transfer of wild type p53 or p27, Retinoblastoma
gene transfer
Management of gliomas

More Related Content

What's hot

Recent advances in Glioblastoma Multiforme Management
Recent advances in Glioblastoma Multiforme ManagementRecent advances in Glioblastoma Multiforme Management
Recent advances in Glioblastoma Multiforme ManagementRajesh Balakrishnan
 
astro guideline on brain mets
 astro guideline on brain mets astro guideline on brain mets
astro guideline on brain metsradiation oncology
 
Low Grade Gliomas
Low  Grade  GliomasLow  Grade  Gliomas
Low Grade GliomasArnab Bose
 
Adjuvant therapy of Glioblastoma in 2020: Marching ahead.
Adjuvant therapy of Glioblastoma in 2020:  Marching ahead.Adjuvant therapy of Glioblastoma in 2020:  Marching ahead.
Adjuvant therapy of Glioblastoma in 2020: Marching ahead.subhas123
 
RADIOTHERAPY FOR OPHTHALMOLOGISTS
RADIOTHERAPY FOR OPHTHALMOLOGISTSRADIOTHERAPY FOR OPHTHALMOLOGISTS
RADIOTHERAPY FOR OPHTHALMOLOGISTSKanhu Charan
 
MANAGEMENT OF MENINGIOMA
MANAGEMENT OF MENINGIOMAMANAGEMENT OF MENINGIOMA
MANAGEMENT OF MENINGIOMAKanhu Charan
 
Glioblastoma multiforme (GBM) Radiotherapy planning and management principles
Glioblastoma multiforme (GBM) Radiotherapy planning and management principlesGlioblastoma multiforme (GBM) Radiotherapy planning and management principles
Glioblastoma multiforme (GBM) Radiotherapy planning and management principlesGebrekirstos Hagos Gebrekirstos, MD
 
Classification of brain tumors AND MANAGEMENT OG LOW GRADE GLIOMA
Classification of brain tumors AND MANAGEMENT OG LOW GRADE GLIOMAClassification of brain tumors AND MANAGEMENT OG LOW GRADE GLIOMA
Classification of brain tumors AND MANAGEMENT OG LOW GRADE GLIOMAradiation oncology
 
2D PLANNING IN BRAIN TUMOR
2D PLANNING IN BRAIN TUMOR2D PLANNING IN BRAIN TUMOR
2D PLANNING IN BRAIN TUMORKanhu Charan
 
PENTEC GUIDELINES FOR PAEDIATRIC RADIOTHERAPY
PENTEC GUIDELINES FOR PAEDIATRIC RADIOTHERAPYPENTEC GUIDELINES FOR PAEDIATRIC RADIOTHERAPY
PENTEC GUIDELINES FOR PAEDIATRIC RADIOTHERAPYKanhu Charan
 
HIPPOCAMPUS TARGET DELINEATION
HIPPOCAMPUS TARGET DELINEATIONHIPPOCAMPUS TARGET DELINEATION
HIPPOCAMPUS TARGET DELINEATIONKanhu Charan
 
ASCO AND SNO GUIDELINES FOR GLIOMA MANAGEMENT
ASCO AND SNO GUIDELINES FOR GLIOMA MANAGEMENTASCO AND SNO GUIDELINES FOR GLIOMA MANAGEMENT
ASCO AND SNO GUIDELINES FOR GLIOMA MANAGEMENTKanhu Charan
 
Radiosurgery for brain metastases
Radiosurgery for brain metastasesRadiosurgery for brain metastases
Radiosurgery for brain metastasesRobert J Miller MD
 
Radiation Therapy in the Management of Lung Cancer
Radiation Therapy in the Management of Lung CancerRadiation Therapy in the Management of Lung Cancer
Radiation Therapy in the Management of Lung Cancerflasco_org
 
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
 
HIGH GRADE GLIOMA MANAGEMENT
HIGH GRADE GLIOMA MANAGEMENTHIGH GRADE GLIOMA MANAGEMENT
HIGH GRADE GLIOMA MANAGEMENTNabeel Yahiya
 
Primary cns lymphoma ppt
Primary cns lymphoma pptPrimary cns lymphoma ppt
Primary cns lymphoma pptShashank Bansal
 

What's hot (20)

Recent advances in Glioblastoma Multiforme Management
Recent advances in Glioblastoma Multiforme ManagementRecent advances in Glioblastoma Multiforme Management
Recent advances in Glioblastoma Multiforme Management
 
astro guideline on brain mets
 astro guideline on brain mets astro guideline on brain mets
astro guideline on brain mets
 
Low Grade Gliomas
Low  Grade  GliomasLow  Grade  Gliomas
Low Grade Gliomas
 
Adjuvant therapy of Glioblastoma in 2020: Marching ahead.
Adjuvant therapy of Glioblastoma in 2020:  Marching ahead.Adjuvant therapy of Glioblastoma in 2020:  Marching ahead.
Adjuvant therapy of Glioblastoma in 2020: Marching ahead.
 
RADIOTHERAPY FOR OPHTHALMOLOGISTS
RADIOTHERAPY FOR OPHTHALMOLOGISTSRADIOTHERAPY FOR OPHTHALMOLOGISTS
RADIOTHERAPY FOR OPHTHALMOLOGISTS
 
MANAGEMENT OF MENINGIOMA
MANAGEMENT OF MENINGIOMAMANAGEMENT OF MENINGIOMA
MANAGEMENT OF MENINGIOMA
 
Glioblastoma multiforme (GBM) Radiotherapy planning and management principles
Glioblastoma multiforme (GBM) Radiotherapy planning and management principlesGlioblastoma multiforme (GBM) Radiotherapy planning and management principles
Glioblastoma multiforme (GBM) Radiotherapy planning and management principles
 
Classification of brain tumors AND MANAGEMENT OG LOW GRADE GLIOMA
Classification of brain tumors AND MANAGEMENT OG LOW GRADE GLIOMAClassification of brain tumors AND MANAGEMENT OG LOW GRADE GLIOMA
Classification of brain tumors AND MANAGEMENT OG LOW GRADE GLIOMA
 
2D PLANNING IN BRAIN TUMOR
2D PLANNING IN BRAIN TUMOR2D PLANNING IN BRAIN TUMOR
2D PLANNING IN BRAIN TUMOR
 
PENTEC GUIDELINES FOR PAEDIATRIC RADIOTHERAPY
PENTEC GUIDELINES FOR PAEDIATRIC RADIOTHERAPYPENTEC GUIDELINES FOR PAEDIATRIC RADIOTHERAPY
PENTEC GUIDELINES FOR PAEDIATRIC RADIOTHERAPY
 
HIPPOCAMPUS TARGET DELINEATION
HIPPOCAMPUS TARGET DELINEATIONHIPPOCAMPUS TARGET DELINEATION
HIPPOCAMPUS TARGET DELINEATION
 
ASCO AND SNO GUIDELINES FOR GLIOMA MANAGEMENT
ASCO AND SNO GUIDELINES FOR GLIOMA MANAGEMENTASCO AND SNO GUIDELINES FOR GLIOMA MANAGEMENT
ASCO AND SNO GUIDELINES FOR GLIOMA MANAGEMENT
 
Medulloblastoma n csi kiran
Medulloblastoma n csi kiranMedulloblastoma n csi kiran
Medulloblastoma n csi kiran
 
Radiosurgery for brain metastases
Radiosurgery for brain metastasesRadiosurgery for brain metastases
Radiosurgery for brain metastases
 
Radiation Therapy in the Management of Lung Cancer
Radiation Therapy in the Management of Lung CancerRadiation Therapy in the Management of Lung Cancer
Radiation Therapy in the Management of Lung Cancer
 
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!
 
HIGH GRADE GLIOMA MANAGEMENT
HIGH GRADE GLIOMA MANAGEMENTHIGH GRADE GLIOMA MANAGEMENT
HIGH GRADE GLIOMA MANAGEMENT
 
Primary cns lymphoma ppt
Primary cns lymphoma pptPrimary cns lymphoma ppt
Primary cns lymphoma ppt
 
Brain metastasis
Brain metastasis Brain metastasis
Brain metastasis
 
Radiation for Glioblastoma
Radiation for GlioblastomaRadiation for Glioblastoma
Radiation for Glioblastoma
 

Similar to Management of gliomas

Overview of brain tumors
Overview of brain tumorsOverview of brain tumors
Overview of brain tumorsDrAyush Garg
 
Who intracranial tumour classification 2016 update- Dr S. Kiranmai
Who intracranial tumour classification 2016 update- Dr S. KiranmaiWho intracranial tumour classification 2016 update- Dr S. Kiranmai
Who intracranial tumour classification 2016 update- Dr S. Kiranmaibenedictsyndrome
 
Lab diagnosis of pituitary tumors
Lab diagnosis of pituitary tumorsLab diagnosis of pituitary tumors
Lab diagnosis of pituitary tumorsgargitignath12
 
Primitive Neuroectodermal Tumor.pptx
Primitive Neuroectodermal Tumor.pptxPrimitive Neuroectodermal Tumor.pptx
Primitive Neuroectodermal Tumor.pptxMedhatMoustafa3
 
Essentials of gliomas
Essentials of gliomas Essentials of gliomas
Essentials of gliomas NeurologyKota
 
Spinal cord lesions and its radiological imaging finding.
Spinal cord lesions and its radiological imaging finding.Spinal cord lesions and its radiological imaging finding.
Spinal cord lesions and its radiological imaging finding.Navneet Ranjan
 
Medulloblastoma - A Closer Look
Medulloblastoma - A Closer LookMedulloblastoma - A Closer Look
Medulloblastoma - A Closer LookHerbert Engelhard
 
Brain tumors Imaging
Brain tumors Imaging Brain tumors Imaging
Brain tumors Imaging MiadAlsulami
 
Presentation1.pptx. radiological imaging of epilepsy.
Presentation1.pptx. radiological imaging of epilepsy.Presentation1.pptx. radiological imaging of epilepsy.
Presentation1.pptx. radiological imaging of epilepsy.Abdellah Nazeer
 

Similar to Management of gliomas (20)

Overview of brain tumors
Overview of brain tumorsOverview of brain tumors
Overview of brain tumors
 
A Case of CNS Tumour
A Case of CNS TumourA Case of CNS Tumour
A Case of CNS Tumour
 
Who intracranial tumour classification 2016 update- Dr S. Kiranmai
Who intracranial tumour classification 2016 update- Dr S. KiranmaiWho intracranial tumour classification 2016 update- Dr S. Kiranmai
Who intracranial tumour classification 2016 update- Dr S. Kiranmai
 
Glioblastoma Multiforme
Glioblastoma MultiformeGlioblastoma Multiforme
Glioblastoma Multiforme
 
Glioblastoma Multiforme
Glioblastoma MultiformeGlioblastoma Multiforme
Glioblastoma Multiforme
 
Glioblastoma multiforme
Glioblastoma multiformeGlioblastoma multiforme
Glioblastoma multiforme
 
Harbor UCLA Neuro-Radiology Case 6
Harbor UCLA Neuro-Radiology Case 6Harbor UCLA Neuro-Radiology Case 6
Harbor UCLA Neuro-Radiology Case 6
 
Lab diagnosis of pituitary tumors
Lab diagnosis of pituitary tumorsLab diagnosis of pituitary tumors
Lab diagnosis of pituitary tumors
 
MEDULLOBLASTOMA
MEDULLOBLASTOMAMEDULLOBLASTOMA
MEDULLOBLASTOMA
 
Brain stem gliomas
Brain stem gliomasBrain stem gliomas
Brain stem gliomas
 
Primitive Neuroectodermal Tumor.pptx
Primitive Neuroectodermal Tumor.pptxPrimitive Neuroectodermal Tumor.pptx
Primitive Neuroectodermal Tumor.pptx
 
Essentials of gliomas
Essentials of gliomas Essentials of gliomas
Essentials of gliomas
 
Spinal cord lesions and its radiological imaging finding.
Spinal cord lesions and its radiological imaging finding.Spinal cord lesions and its radiological imaging finding.
Spinal cord lesions and its radiological imaging finding.
 
Neuro oncology 1
Neuro oncology 1Neuro oncology 1
Neuro oncology 1
 
Medulloblastoma - A Closer Look
Medulloblastoma - A Closer LookMedulloblastoma - A Closer Look
Medulloblastoma - A Closer Look
 
BRAIN-TUMORS.pptx
BRAIN-TUMORS.pptxBRAIN-TUMORS.pptx
BRAIN-TUMORS.pptx
 
BRAIN-TUMORS.pptx
BRAIN-TUMORS.pptxBRAIN-TUMORS.pptx
BRAIN-TUMORS.pptx
 
Brain tumors Imaging
Brain tumors Imaging Brain tumors Imaging
Brain tumors Imaging
 
Brain tumor in children
Brain tumor in childrenBrain tumor in children
Brain tumor in children
 
Presentation1.pptx. radiological imaging of epilepsy.
Presentation1.pptx. radiological imaging of epilepsy.Presentation1.pptx. radiological imaging of epilepsy.
Presentation1.pptx. radiological imaging of epilepsy.
 

Recently uploaded

Presiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsPresiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsanshu789521
 
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdfBASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdfSoniaTolstoy
 
Crayon Activity Handout For the Crayon A
Crayon Activity Handout For the Crayon ACrayon Activity Handout For the Crayon A
Crayon Activity Handout For the Crayon AUnboundStockton
 
Class 11 Legal Studies Ch-1 Concept of State .pdf
Class 11 Legal Studies Ch-1 Concept of State .pdfClass 11 Legal Studies Ch-1 Concept of State .pdf
Class 11 Legal Studies Ch-1 Concept of State .pdfakmcokerachita
 
MENTAL STATUS EXAMINATION format.docx
MENTAL     STATUS EXAMINATION format.docxMENTAL     STATUS EXAMINATION format.docx
MENTAL STATUS EXAMINATION format.docxPoojaSen20
 
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxSOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxiammrhaywood
 
How to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxHow to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxmanuelaromero2013
 
A Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy ReformA Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy ReformChameera Dedduwage
 
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxPOINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxSayali Powar
 
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTiammrhaywood
 
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...M56BOOKSTORE PRODUCT/SERVICE
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityGeoBlogs
 
microwave assisted reaction. General introduction
microwave assisted reaction. General introductionmicrowave assisted reaction. General introduction
microwave assisted reaction. General introductionMaksud Ahmed
 
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxRoyAbrique
 
CARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxCARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxGaneshChakor2
 

Recently uploaded (20)

Presiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsPresiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha elections
 
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdfBASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
 
Crayon Activity Handout For the Crayon A
Crayon Activity Handout For the Crayon ACrayon Activity Handout For the Crayon A
Crayon Activity Handout For the Crayon A
 
Class 11 Legal Studies Ch-1 Concept of State .pdf
Class 11 Legal Studies Ch-1 Concept of State .pdfClass 11 Legal Studies Ch-1 Concept of State .pdf
Class 11 Legal Studies Ch-1 Concept of State .pdf
 
MENTAL STATUS EXAMINATION format.docx
MENTAL     STATUS EXAMINATION format.docxMENTAL     STATUS EXAMINATION format.docx
MENTAL STATUS EXAMINATION format.docx
 
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxSOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
 
How to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxHow to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptx
 
Model Call Girl in Bikash Puri Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Bikash Puri  Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Bikash Puri  Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Bikash Puri Delhi reach out to us at 🔝9953056974🔝
 
A Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy ReformA Critique of the Proposed National Education Policy Reform
A Critique of the Proposed National Education Policy Reform
 
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxPOINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
 
TataKelola dan KamSiber Kecerdasan Buatan v022.pdf
TataKelola dan KamSiber Kecerdasan Buatan v022.pdfTataKelola dan KamSiber Kecerdasan Buatan v022.pdf
TataKelola dan KamSiber Kecerdasan Buatan v022.pdf
 
Staff of Color (SOC) Retention Efforts DDSD
Staff of Color (SOC) Retention Efforts DDSDStaff of Color (SOC) Retention Efforts DDSD
Staff of Color (SOC) Retention Efforts DDSD
 
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPTECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
ECONOMIC CONTEXT - LONG FORM TV DRAMA - PPT
 
9953330565 Low Rate Call Girls In Rohini Delhi NCR
9953330565 Low Rate Call Girls In Rohini  Delhi NCR9953330565 Low Rate Call Girls In Rohini  Delhi NCR
9953330565 Low Rate Call Girls In Rohini Delhi NCR
 
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activity
 
microwave assisted reaction. General introduction
microwave assisted reaction. General introductionmicrowave assisted reaction. General introduction
microwave assisted reaction. General introduction
 
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
 
Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1
 
CARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxCARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptx
 

Management of gliomas

  • 1. PRESENTER- DR. JASMEET SINGH TUTEJA MODERATOR- PROF. SEEMA GUPTA
  • 2.  Gliomas arise from supportive cells (glial cells) that surround nerve cells.  Most common type of primary brain tumours.  4 types of glial cells-  Astrocytes- balance of chemical in brain, wound healing & repair.  Oligodendrocytes- myelin synthesis  Ependymal cells- CSF (ventricular lining)  Microglial cells- defend brain from disease- causing factor. Different type of gliomas vary in treatment management & prognosis.
  • 3. EPIDEMIOLOGY Incidence - 17th position Death – 12th position  Incidence – 10th position  Death – 10th position
  • 4. Environmental  Ionizing radiation (meningioma, gliomas, sarcomas) - 2.3% incidence in prophylactic cranial irradiation in children  Petrochemicals (formaldehyde, vinyl chloride, acrylonitrite) Hereditary  Cowden, Turcot , Lynch and Li-Fraumeni (gliomas)  Gorlin (PNET),  Neurofibromatosis I & II (meningiomas, optic nerve gliomas, schwannoma)  Von Hippel Lindau syndrome (hemangioblastoma) first-degree family member shown to have increase risk approximately two-fold
  • 5.
  • 6. WHO classification Lyon 2007 –  Histological criteria  Morphological criteria  In 2014, a meeting held in Haarlem, the Netherlands, under the auspices of the International Society of Neuropathology, 2016 WHO Classification was formed attended by 117 contributors from 20 countries to formulate guidelines Why new classification ?  Inter/ Intra observer variation  Traditional histologic grading does not provide good prognostic power.  Within the same tumour subtype and grade – a lot of variation in prognosis
  • 7. Primary focus was on: • Molecular markers • Hence, Integrating Genotypic & Phenotypic parameters to make final diagnosis
  • 8. NEW CHANGES:  Molecular parameters were introduced  More objective and more precisely defined entities  Improved tailoring of patient therapy  Better classification for research purpose  Wastebasket categories: NOS  Need of more focused study of these less defined group => clarification of their status Can we rely on molecular parameters alone? & skip histology?
  • 9. 9
  • 10. 10
  • 11.
  • 12.
  • 13.
  • 14.
  • 15. Brainstem • Difficulty swallowing, speaking • Drowsiness • Headaches • Hearing loss • Muscle weakness • Hemiparesis • Uncoordinated gait • Vision loss, ptosis, strabismus • Vomiting Frontal lobe  Behavioural and emotional changes  Impaired judgement , Impaired sense of smell  Memory loss  Hemiplegia  Cognitive dysfunction  Vision loss  Papilledema Temporal lobe • Impaired speech • Seizures • Homonymous superior quandrantanopsia • Auditory hallucinations • Abnormal behaviour Occipital Lobe • Visual field deficits • Visual hallucinations Parietal lobe  Impaired speech  Inability to write  Lack of recognition  Seizures  Spatial disorder
  • 16.
  • 17.  Imaging – MRI, CT , PET  Biopsy (craniotomy/ stereotactic)  CSF  IHC  Cytogenetics
  • 18. 18
  • 19.  Investigation of choice MRI sequences :  T1 weighted  T2 weighted  T2 weighted FLAIR  T1 post contrast  Diffusion weighted imaging  Magnetic resonance spectroscopy  Magnetic resonance perfusion study
  • 20. Anatomical details  CSF – dark  Gray matter – gray  White matter - white  Most Pathologies – dark
  • 21. Anatomical details of T2W  Pathological details  Gray matter – bright  White matter – dark  CSF- bright
  • 22. FLAIR  Similar to T2 except free water suppression (inversion recovery)  Pathology – bright  Good for lesions near ventricles or sulci
  • 23.  Diagnosis - made on axial images.  T2Wt- and FLAIR - display the margins of a tumor and its surrounding edema or a direct tumor infiltration.  The sagittal & coronal images - confirm exact location
  • 24.
  • 25.  Non-invasive physiologic imaging that measure relative levels of tissue metabolites  Complements MRIs
  • 26.  Guidance for stereotactic biopsy.  Differentiates between Psuedoprogression & Recurrent gliomas.  11C Methionine (MET-PET)- uptake is correlated with tumour proliferation  superior contrast & tumour delineation.  FET-PET (18F- fluoro-ethyl-L-tyrosine)- longer half life
  • 27.  Stereotactic frame or scalp fiducials  CT or MRI is performed and the data are loaded into an image guidance system.  A target and entry points are selected, and the trajectory is visualized on a workstation.  The entry point - patient’s scalp, and a small burr hole or twist drill hole is made.  The biopsy needle is passed to the appropriate depth, and tissue samples are obtained.
  • 28.  Essential for staging tumors with a propensity for CSF spread (medulloblastoma, germ cell tumors, and CNS lymphoma).  best done before surgery or more than 3 weeks after surgery, as long as intracranial pressure is not elevated.  Tumor markers in the CSF may help in making the diagnosis.
  • 29.
  • 30. Important exception is pilocytic Astrocytoma which enhances with contrast because the blood vessels go degenerative changes and in contrast the HGGs enhance because because of microvascular proliferation. How to recognise blood in the MRI? – look for severe hyper intensity with a surrounding black border in the FLAIR sequence Cerebral metastases can often be confusing with HGG. There are some useful characteristics when trying to differentiate these radiologically. 1. cerebral mets usually occur in the gray/white junction,and less common in deeper brain, 2. do not involve the periventricular white matter and 3. rarely ever goes to the corpus callosum. 4. cerebral mets are well circumscribed usually and they do not have multiple areas of heterogenous contrast enhancement – i.e. looks like a single round bright rim with a single dark centre in T1w image with Contrast. In contrast GBM – centers on the sub-cortical white matter, with subependymal and corpus callosum extension seen frequently. There can be multifocal GBM that looks like many small GBMs but they are all embedded in a single unit / area of FLAIR enhancement.
  • 31. 31 WHO Grade Definition Gliomas I Circumscribed tumors of low proliferative potential associated with the possibility of cure following resection (Low proliferation) Pilocytic astrocytoma, Pleomorphic xanthoastrocytoma, Ganglioglioma, Subependymal giant cell astrocytoma II Infiltrative tumors with low proliferative potential with increased risk of recurrence (Nuclear atypia) Diffuse astrocytomas (IDHmut/IDHwt) and oligodendrogliomas (IDH mut, 1p19q codel) III Tumors with histologic evidence of malignancy, including nuclear atypia and mitotic activity, associated with an aggressive clinical course (PLUS Mitoses) Anaplastic astrocytomas (IDHmut/IDHwt) and anaplastic oligodendrogliomas (IDH mut, 1p19q codel) IV Tumors that are cytologically malignant, mitotically active, and associated with rapid clinical progression and potential for dissemination (PLUS Necrosis Vascular proliferation) Glioblastoma (IDH mut/wt)
  • 32. CLINICALLY RELEVANT MOLECULAR MARKERS 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 Prognostic 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 grade II Diffuse astrocytoma 70%–80% 15% 40%–50% Oligodendroglioma/ oligoastrocytoma 70%–80% 30%–60% 60%–80% WHO grade III Anaplastic astrocytoma 50%–70% 15% 50% Anaplastic Oligodendroglioma/ oligoastrocytoma 50%–80% 50%–80% 70% WHO grade IV Glioblastoma 5%–10% <5% 35%
  • 33.  Neuroradiologists  Neurosurgeons  Neurologists  Neuropathologist  Radiation oncologists  Medical oncologists  Neuro-rehabilitation 3/7/2022 33
  • 35.  Grow relatively slowly  Diffuse infiltrative pattern  Most transform in high grade gliomas- with aggressive clinical course & shortened OS  Treatment modalities include: surgery, radiotherapy, & chemotherapy  Management of LGG is complex & controversial- MDT is essential.  Estimated risks & benefits of adjuvant therapies (PFS, OS & neurocognitive preservation) should be discussed with the patient whenever possible.
  • 36. 36
  • 37. 37
  • 38. T2-FLAIR MISMATC H SIGN  The T2-FLAIR mismatch sign- highly specific radiogenomic signature for DIFFUSE ASTROCYTOMA (IDH-mutant, 1p/19q-non- codeleted molecular status)  Helps in distinguishing a diffuse astrocytoma from an oligodendroglioma.  On T2 weighted- tumours have extensive areas of fairly homogeneous and strikingly high signal.  On T2-FLAIR - majority of these areas become relatively hypointense in signal due to incomplete suppression.  At the margins of the tumour, a rim of hyperintensity is usually seen. 38
  • 39. 39
  • 40.
  • 41. 41
  • 42.
  • 43.
  • 44.  CONSIDER MAXIMAL SAFE RESECTION  AIM TO: 1. Obtain a histological & molecular diagnosis 2. Remove as much as tumor as safely as possible 3. To preserve or improve QOL especially by controlling seizures.  Why? -Greater extent of resection can improve OS  How? -Consider intraoperatively MRI & U/S, awake craniotomy with language & other appropriate functional monitoring.
  • 45.  Extent of resection & post surgical residual volume are independent prognostic factor significantly associated with longer OS.  Surgical excision is also independent prognostic factor correlating with increased malignant PFS* Advantage of extensive resection:  Across all molecular subtypes.  Even min. residual volume negatively affects OS- most relevant for IDH-mut astrocytomas (in oligodendroglioma – trend towards better OS)  Consider 2nd look op to remove minor residuum if safely possible.
  • 46.  Post-operative radiotherapy  Decisions largely based on risk factors and extent of resection
  • 47. • No overall survival benefit from immediate RT vs delayed RT (7.4vs 7.2 yrs) • Improved progression free survival with immediate RT by ~24 months but at expense of toxicity (e.g. neuro-cognitive) median 5.4 vs 3.7 years • Better seizure control (25% vs 41% at 1 yr)
  • 48. Absence of OS benefit for early RT suggest that RT slows progression of LGG’s but does not prevent malignant transformation & that RT given at time of radiological PD is equally effective.
  • 49. • Grade 2 glioma (any subtype).. High risk • <40 AND subtotal resection • >40 years (any biopsy/surgery) • 54Gy in 30 fractions +/- 6 x PCV q8 weekly
  • 50. • Improved overall survival with addition of PCV • Median: 13.3 vs 7.8 years • 10 year OS: 60% vs 40% • Largest benefit in oligodendroglioma • Use for oligodendrogliomas as above • Not significant for astrocytoma….. In practice, tend not to use adjuvant chemotherapy here..... Wait & watch. • Improved progression free survival with PCV • 10 year PFS: 21% vs 51%
  • 51. Prognostic factors for survival in adult patients with cerebral low-grade glioma.
  • 52.
  • 53.
  • 54. IMAGING SUGGESTIVE OF LOW GRADE GLIOMA >80-90% resection possible? Resection (often awake craniotomy) Biops y Observation to determine behaviour Ye s N o Integrated diagnosis (inc cytogenetics IDH, 1p 19q, MGMT, TERT) confirms G2 Assess risk factors for progression: Age >40, sub-total resection / biopsy, size >6cm, crosses midline, poor cytogenetics, symptomatic (but not seizures), adverse radiology (eg enhancement) High Low Decide with patient whether to use immediate non-surgical treatment or observe (influenced by level of risk and patient’s wishes) Observation (Consider more surgery or non-surgical treatment if progresses Q: Is it high grade now?) Non-surgical treatment Olig o RT + Astrocytom a R ?treat as GBM if G2 astrocytoma ?chemo alone if gliomatosis
  • 57. T1 T2 T1W + Gad GLIOBLASTOMA
  • 58.  Stereotactic image guided biopsy is accurate & safe way to get to even relatively inaccessible areas of the brain such as basal ganglia & thalamus. 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. Simpson et al , IJROBP 1993
  • 59.
  • 60.  Patients with grade III (=anaplastic) oligodendroglial tumours  RT vs RT (59.4Gy) + adjuvant PCV  Median OS: 42.3 vs 30.6 months  PFS also improved (Median PFS: 24.3 VS 13.2 Months)  Improved outcomes also in patients with IDH mutations and methylated tumours  Therefore: consider adjuvant PCV for grade III oligos (by modern classification these are all co- deleted)
  • 61. More benefit in patients with 1p19q co-deletion: trend towards improved OS here, not in non-co-deleted. Overall survival in both treatment arms for (A) the patients with 1p/19q-codeleted tumours (B) the patients with non–1p/19q- codeleted tumours Progression Free survival in both treatment arms for (A) the patients with 1p/19q- codeleted tumours (B) the patients with non–1p/19q- codeleted tumours CO-DELETED TUMOUR- OS was not reached in RT+PCV vs 112 months in RT group NON-CODELETED TUMOUR- OS 25 vs 21 months CO-DELETED TUMOUR- PFS- 157 VS 50 MONTHS (RT + PCV VS RT) NON CO-DELETED TUMOUR- 15 VS 9 MONTHS (RT + PCV VS RT)
  • 62. 59.4 Gy/33# of RT PCV → 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 2.7 y 2.6 y PFS - 1p/19q non codeleted AO/AOA 1 y 1.2 y
  • 63. Adults (>18) with newly diagnosed 1p/19q World Health Organization (WHO) grade III oligodendroglioma were randomized in 3 arms: • Radiotherapy (RT-59.40Gy) alone; • RT with concomitant and adjuvant temozolomide; or • TMZ alone
  • 64. INTERIM RESULT OF CODEL TRIAL:  TMZ-alone patients had significantly shorter PFS than patients treated on Radiotherapy arms.  The ongoing CODEL trial trial has been redesigned to compare RT + PCV versus RT + TMZ.
  • 65.
  • 66. Design- Patients with histologically confirmed Anaplastic Astrocytoma were randomized to 2 arms- • RT+TMZ • RT+NU Results. Median survival time was 3.9 (RT/TMZ arm) vs 3.8 years (RT/NU ARM). Conclusions. No significant improvement in OS , PFS & TTP in both arms RT+TMZ was better tolerated. IDH1-R132H mutation was associated with longer survival.
  • 67. oSlightly better PFS with TMZ oHigher toxicity and discontinuation rates with nitrosureas IDH positive with a better OS in IDH-positive patients (median survival time 7.9 vs 2.8 y)
  • 68. • Grade III gliomas which were NOT 1p19q co- deleted • Improved overall survival in arms with adjuvant temozolomide • Larger advantage in MGMT methylated (70%) • Therefore, consider adjuvant temozolomide for non-co-deleted Grade III gliomas (now = anaplastic astrocytoma).
  • 69.
  • 70. The 2nd interim analysis declared futility of concurrent TMZ (median OS was 66.9 months with concurrent TMZ vs 60.6 months without concurrent TMZ) By contrast, adjuvant TMZ improved improved OS compared with no adjuvant TMZ (median OS 82.3 months vs 46.9 months) Adjuvant TMZ therapy, but not concurrent TMZ chemotherapy, was associated with a survival benefit in patients with 1p/19q non co- deleted anaplastic glioma https://doi.org/10.1016/S1470-2045(21)00090-5
  • 71. Design- Patients with anaplastic gliomas randomized 2:1:1 In 3 arms: • standard radiotherapy (RT) (arm A), • Procarbazine, lomustine and vincristine (PCV) (arm B1), or • Temozolomide (TMZ) (arm B2).
  • 72. There is no differential activity of primary chemotherapy versus RT in any subgroup of anaplastic glioma. Molecular diagnosis is superior to histology.
  • 74.
  • 75. Patient were randomized in 2 arms: • RT alone (60Gy) vs • RT+TMZ  TMZ (6cycles)
  • 76. Median PFS 6.9 vs 5 months Median Survival- 14.6 vs 12.1 months 2- year survival rate- 26.6 vs 10.4 months
  • 77. • GBM: 60Gy in 30 fractions RT +/- concurrent temozolomide 75mg/m2 then 4 week break then adjuvant temozolomide 150-200mg/m2 x 6 months • Given with prophylactic oral Septrin (co-trimoxazole) 960mg during RT • Addition of temozolomide improved overall survival: • Median 12.1 months vs 14.6 months • 2 year OS: 10.4% vs 26.5% (unheard of in GBM before) • Well tolerated • 90% completed at least 90% of planned temozolomide • 7% grade 3 or 4 toxicity • Patients with complete resection did better • 2 year OS: 37% • Patients with methylated MGMT did better • 2 year OS: 46%
  • 78.
  • 79. MGMT promoter methylation has been demonstrated as the strongest prognostic & predictive marker for outcome, and the added benefit of Temozolomide.
  • 80. MGMT gene(10q26) Encodes DNA repair protein Removes alkyl groups from the DNA (DNA repair) TMA alkylates O6 position of guanine Resistance to alkylating agents (removal of alkyl group by MGMT) Methylation of the MGMT promoter silences expression of the protein Cytotoxicity and apoptosis by alkylating agent (TMZ)
  • 81.  Retrospective study (morning vs evening intake)  Survival benefit in median OS when patients take TMZ in morning versus in evening. (mOS of 2.13 years in AM vs 1.63yrs in PM) Morning TMZ administration improves survival with patients with GBM. https://doi.org/10.1093/noajnl/vdab041
  • 82. 82 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 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.
  • 83. 83 Role of Bevacizumab in newly diagnosed GBM Citation Arms Median OS (months) RTOG 0825 Gilbert NEJM 2014 60 Gy + TMZ → TMZ 16.1 60 Gy + TMZ → TMZ + BEV 15.7 AVAglio Chinot NEJM 2014 60 Gy + TMZ → TMZ 16.7 60 Gy + TMZ → TMZ + BEV 16.8 No significant benefit in median OS on addition of Bevacizumab.
  • 85. • Nordic and German trial have randomized patients into 3 arms- conventional RT, hypofractionated RT (34GY in 10#) and temozolomide alone • Interpretation- Standard Radiotherapy was associated with poor outcome in patients more than 70 years. • Both TMZ & hypofractionated RT were associated with longer survival & can be considered as standard treatment options in elderly GBM. https://doi.org/10.1016/s1470-2045(12)70265-6
  • 86.  Age >70 is a dreadful prognostic factor in GBM  Perry et al NEJM 2017 patient were randomized in 2 arms:  RT 40Gy in 15 fractions +/- temozolomide concurrent 75mg/m2 and adjuvant 150- 200mg/m2 for up to 12 cycles  Patients ≥65 years, PS 0-2  Addition of TMZ improved overall survival  9.3 months vs 7.6 months  Addition of TMZ improved PFS  5.3 months vs 3.9 months  Consider TMZ-RT 40Gy in 15 fractions and adjuvant TMZ if >70 and PS 0-2
  • 87. • Patients with Butterfly Glioma have almost poor performance status. • Poor PS patients have dreadful outcome • PS 3 and <70 years: consider 30Gy in 6 fractions over 2 weeks (or 25Gy in 5 fractions over one week (Roa et al JCO 2015))
  • 88. • <70 years, GBM • PS 0/1: chemoRT 60Gy in 30 fractions + adjuvant temozolomide x 6 • PS 2 (not butterfly): 60Gy in 30 fractions • PS 3 (or PS 2 butterfly glioma): 30Gy in 6 fractions over 2 weeks • PS 4: steroids and best supportive care • >70 years, GBM (?top age) • PS0-1: chemo RT 40Gy in 15 fractions + adjuvant temo up to 12 cycles • PS 2: possibly temozolomide alone or hypofractionated RT (need MGMT) status. Consider BSC. • PS3/4: steroids and best supportive care • <70 years, grade III • PS 0-1: RT 60Gy in 30 fractions • 1p19q co-deleted oligo: adjuvant PCV • Non-1p19q: adjuvant temo
  • 90. 90 Tumor-treating fields involve • Transducer arrays are placed on scalp. These are connected to electric field generator. Light weight battery pack is carried by patient and worn for atleast 18 hours a day. Transducer arrays delivers low intensity, intermediate frequency, alternating electric field which acts by disrupting mitosis and inducing 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
  • 91. 91 oKPS > 70; median age 56 oRandomized 2:1 after 60Gy, TMZ Adj TMZ + TTF Adjuvant TMZ oImproved median survival from time of randomization 16→20.9 months oToxicity – dermatitis in 52% oImproved OS: >18 hours/day
  • 92.
  • 93.  Corticosteroids, preferably dexamethasone reduce symptomatic peritumoral vasogenic edema, neurological deficits and signs of increased intracranial pressure  Lowest dose of steroids – shortest time possible  No need for prolonged steroid therapy after tumour resection or for prophylaxis during radiotherapy in asymptomatic patients  Downward titration of dose – whenever possible  Extensive mass effect – receive steroids for at least 24 hours before RT  Monitor for side effects  Chronic corticosteroids (≥ 20 mg prednisone equivalents daily for ≥ 1 month) - consider prophylaxis for osteoporosis and pneumocystis jerovecii pneumonia should be considered
  • 94. 94 Antiepileptic Drugs ▪ Increased risk of thromboembolic events due to a tumour-induced hyper-coagulable state, as a consequence of neurological deficits, immobilisation and steroid use. ▪ Prophylactic anticoagulation is not recommended ▪ No contraindication for the use of standard anticoagulants in patients with proven thrombosis. Anticoagulants ▪ Non Enzyme inducing anti seizure medications should be used eg: levetiracetam, topiramate, valproic acid ▪ Current guidelines recommend tapering AEDs 1-2 weeks after surgery and avoiding long-term prophylaxis. ▪ No role for primary perioperative prophylaxis (i.e. in patients who have never had a seizure).
  • 95.
  • 96.  SUPINE, HEAD IN NEUTRAL POSITION  HEAD IMMOBILISED- Perspex or thermoplastic shell. More rigorous immobilization with a stereotactic frame.  I.V. contrast 1mg/kg most of time 50mg i.v.  Thin-slice CT imaging is recommended (at 1–2 mm slices) from the vertex to the C3-C7 and, for ease of fusion, ideally should match the MR image slice thickness.  Multimodal MR fusion, using at least the contrast enhanced T1 and FLAIR sequences  PET/CT fusion can be helpful distinguish between postradiation changes and disease recurrence on MR imaging.
  • 97.  3D-CRT remains standard for the majority of GBM  IMRT/ VMAT is - volumetrically or spatially challenging tumours.  Proton beam radiation therapy due to its “Bragg peak effect, safer than traditional photon radiation therapy  Boron neutron capture therapy (BNCT), increases the efficacy in destruction of tumor
  • 98. • Superior: skin fall off • Anterior: skin fall off • Posterior: skin fall off • Inferior: cranial base 2 opposed lateral fields.  A rectangular treatment field over the cranial vault with the collimator angled so as to place the inferior border 1–2 cm below a line drawn from the medial canthus to the mastoid tips.  A small block is carefully placed to shield the globes while still allowing adequate coverage of the anterior–inferior extent of the cranial contents Whole-brain RT
  • 99. Anaplastic astrocytoma Phase 1 Phase 2 60 Gy in 30# 46 Gy in 23# 14 Gy in 7# 59.4 Gy in 33# 45 to 50.4 Gy in 1.8 Gy/# 9 to 14.4 Gy in 1.8 Gy/# Anaplastic oligodendroglioma Phase 1 Phase 2 60 Gy in 30# 50.4 Gy in 28# 9 Gy in 5# Studies interrogating dose escalation beyond 60 Gy using standard fractionation have not demonstrated any survival benefit. LGG- 54Gy in 30 fractions or 50.4Gy in 28 fractions (EORTC 22844)
  • 100.  GTV- non-enhancing tumour extent (T2/FLAIR)  CTV- GTV+1-1.5cm  PTV- CTV + 0.5cm
  • 101. EORTC treatment volumes 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) GT V 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) GT V2 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 PTV 2 CTV2 plus a margin of 3–5 mm *Margins up to 3 cm were allowed in 22981/22961 trial, and 1.5 cm in 26981–22981 trial. TARGET DELINEATION OF GLIOBLASTOMA Stupp et al.(2005) Chinot et al (2014) Gilbert (2014)
  • 102. RTOG- 2 phases of target volume delineation. (A) The initial GTV includes postoperative peritumoral edema based on the axial T2 fluid-attenuated inversion recovery sequence (red line); the initial CTV (green line) includes postoperative peritumoral edema plus a 2 cm expansion in all directions. (B) The boost GTV includes the surgical cavity and residual enhancement based on the axial T1 sequence with gadolinium (red line) and the boost CTV is a 2 cm expansion in all directions (green line) EORTC-1 phase of target volume delineation. GTV: surgical cavity and residual enhancement based on the axial T1 sequence with gadolinium (red line) and CTV is a 2 cm expansion in all directions (green line). Zhao et al Onco Targets Ther 2016
  • 103.  Principles of intensity-modulated radiotherapy (IMRT)-based planning for dose-escalated HFRT.  The two regions are prescribed differential doses using IMRT, with the high-dose area receiving higher tumour biological effective dose from increased dose and higher fraction size.  This technique is inappropriate for tumours in close proximity to critical structures. Hingorani et al 2012 BJR
  • 104. 104 From left to right: transition from 2D RT to 3D RT to intensity modulated radiotherapy to intensity modulated proton therapy harnesses the potential for sparing normal, uninvolved brain substructures from unnecessary RT dose; whether this produces meaningful patient clinical benefit is a subject of current clinical trial testing. 2D 3D IMRT IM Proton Therapy
  • 105. 105 OAR OBJECTIVE(S) Brainstem D <54 Gy; 1–10 cc < 59 Gy (periphery) Chiasm Dmax <55 Gy Cochlea Ideally one side mean <45 Gy ( Most protocols allow ipsilateral cochlea to receive 60 Gy rather than compromise dose ) Eyes Macula <45 Gy Lacrimal glands Dmax <40 Gy Lens Ideally <6 Gy Max 10 Gy dose limits should never compromise PTV dose Optic nerves and chiasma Dmax <55 Gy or 1 % of PTV cannot exceed 60 Gy Pituitary Dmax <50 Gy Hippocampus Max ≤16 Gy and no more than 9 Gy to 100 % volume OAR definitions and dose limits in GBM patients (individual adaptation necessary according to the clinical situation) M. Niyazi et al. / Radiotherapy and Oncology 118 (2016) Based on the University of Maryland treatment planning guidelines
  • 106. ACUTE  Fatigue  Erythema  Alopecia  Headache  Nausea  Vomiting LATE  Somnolence  Neurocognitive impairment – hippocampal sparing)  Brain Necrosis
  • 107. FOLLOW UP  Brain MRI every 3-6 month followed by serial imaging up to 3-5 years & then atleast annually is recommended according to NCCN followed with clinical correlation
  • 108. • Occurs in about 30% of patients • Most within 3 months of chemo-RT • Represents endothelial damage and subsequent hypoxia • Scan looks worse, patient often well, but not always • Increased enhancing lesion • Increased FLAIR • Tricky to differentiate from recurrence • Advanced imaging/ Radiologist may help • Reduced relative CBV (cerebral blood volume) supports diagnosis of pseudo-progression (increased in recurrence) • MR-spectroscopy: Low choline:NAA ratio (≤1.4) or low choline (=low levels of metabolites) or high lactate supports diagnosis of pseudo- progression • Diffusion weighted imaging: Elevated ADC (apparent diffusion coefficient) supports diagnosis of pseudo-progression= less impedance of water motion (in recurrences have lots of cells impede water motion)
  • 109. First progression Definition Progressive disease < 12 weeks after completion of chemoradiotherapy Progression can only be defined using diagnostic imaging if there is new enhancement outside of the radiation field (beyond the high-dose region or 80% isodose line) or if there is unequivocal evidence of viable tumor on histopathologic sampling Note: Given the difficulty of differentiating true progression from pseudoprogression, clinical decline alone, in the absence of radiographic or histologic confirmation of progression, will not be sufficient for definition of progressive disease in the first 12 weeks after completion of concurrent chemoradiotherapy. Progressivedisease≥ 12 weeks after chemoradiotherapy completion 1.New contrast-enhancing lesion outside of radiation field on decreasing, stable, or increasing doses of corticosteroids. 2.Increase by ≥ 25%in the sum of the products of perpendicular diameters between the first postradiotherapy scan, or a subsequent scan with smaller tumor size, and the scan at 12 weeks or later on stable or increasing doses of corticosteroids. 3.Clinical deterioration not attributable to concurrent medication or comorbid conditions is sufficient to declare progression on current treatment but not for entry onto a clinical trial for recurrence.
  • 110.  Surgical resection  Chemotherapy: TMZ/ Carmustine/ Lomustine  Locally delivered CARMUSTINE OR GLIADEL wafers: o Implanted into the surgical cavity →modest survival advantage o It offers statistically significant but marginal improvement (31 vs 23 weeks) in OS after re-operation of HGG. o Significant peritumoral edema is a recognized side-effect. o Requires careful patient selection and a gross total resection. o Specialist centres only  Trial: Westphal et al  HGG: maximum surgical resection +/- gliadel wafers, followed by RT  2.3 month increase in median survival with gliadel  11.3 months to 13.9 months  Greater gain (4.2 months) if ≥90% resection 110  Bevacizumab-FDA approved  Alternating Electrical Field Therapy  Re-Irradiation  Best Supportive Care
  • 111. 111 ▪ Cognitive deficits, personality changes, and mood disturbances are major comorbidities. ▪ “Chemobrain” - severe cognitive dysfunction that can persist after the cessation of treatment↔ chemotherapeutic agent-induced inflammation in the hippocampus, which is involved in learning and memory ▪ Endocrinopathies are common – surgery, steroid, RT – monitor and manage ▪ Psychiatric comorbidity treated with psychotherapy and pharmacotherapy ▪ Early discussion of goals of care and involvement of palliative care should be considered ▪ Diverse support - nurse specialists in neuro-oncology, social workers, counsellors, clinical psychologists, palliative-medicine professionals, and patient focus groups. ▪ Rehabilitative measures should be explored during and after tumour-specific therapy
  • 112.  Externally generated ionizing radiation is used to eradicate a define target without the need to make an incision.  Target is defined by high resolution sterotactic imaging.  Usually delivered in single session (maximum of 5) using stereotactic guiding device. ( strict immobilization)  Multiple non opposed radiation beams converge on target in the brain avoiding normal tissues.  Highly conformal  Can be delivered using a conventional or modified LINAC, a Gamma Knife or a robotically controlled miniaturized linear accelerator (CyberKnife), particle beam accelerator or multisource Co60 unit Leksell Stereotactic system trUpoint ARCH SRS/SRT Syste
  • 113. 113  Used 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:  Small target/treatment volume  Sharply defined target  Accurate radiation delivery  High conformality  Sensitive structures excluded from target Radiobiology: achieving increased cell kill from a higher dose per fraction → 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) and reducing the effect from accelerated tumour cell repopulation by shortening the overall treatment time
  • 114. 114  4 phases: 1. placement of the head frame, 2. imaging of the tumor location, 3. computerized dose planning, and 4. radiation delivery SRS dosing: RTOG 9005 results < 2 cm 24 Gy 2.1 -3 cm 18 Gy 3.1 – 4 cm 15 Gy
  • 115. 115 BEVACIZUMAB o Monoclonal antibody that binds to VEGF and inhibits the growth of tumor blood vessels o Prolongs OS either alone or in combination with a cytotoxic agent by about 4 months in recurrent glioblastoma but not in primary Glioblastoma CILENGITIDE- no established role (CORE trial) ANTIANGIOGENIC THERAPY Vaccines for Glioblastoma o DCVax- a dendritic cell (DC)-based personalized cancer vaccine with purified tumor- specific antigens or tumor cell extracts derived from tumor at the time of resection. o Epidermal Growth Factor Receptor Variant III as a Vaccine Target for Glioblastoma o Heat Shock Protein-Based Vaccine o Recombinant nonpathogenic polio-rhinovirus chimera (PVSRIPO) targets the neurotropic poliovirus receptor CD155, which is abundantly expressed on glioblastoma cells.
  • 116. 116 Biomarkers • EGFRvIII amplification is targeted by EGFR vaccine rindopepimut. • KIT amplification or mutation is target is targeted by KIT inhibitor imatinib • PDGFRA amplification is targeted by PDGFR inhibitor dasatinib • PTEN deletion or mutation is targeted by AKT inhibitor or mTOR inhibitor voxtalisib • MDM2 amplification is targeted by a MDM2 inhibitor such as AMG232 • TP53 wild-type is targeted by a MDM2 inhibitor such as AMG232 • RB1wild-type is targeted by CDK4/6 inhibitor ribociclib MicroRNAs→ Dysregulation → pathogenesis of glioblastoma Targeting miRNA(s) could alter multiple genes simultaneously and may prove more effective than targeted focused at targeting single gene or pathway CAR-T cell therapy • Multiple infusions into the ventricular system of CAR-T cells targeting the tumor-associated antigen IL-13 receptor alpha 2 (IL13Ra2) in a patient with recurrent multifocal • Intravenous delivery of a single dose of autologous CAR-T cells targeting EGFRvIII mutation in patients with recurrent GBM → feasible and safe
  • 117. 117 Convection-Enhanced Delivery: use of intracerebrally implanted catheters to deliver a drug into the brain parenchyma or tumor, at a slow but continuous rate of flow. • Topoisomerase-I inhibitor, topotecan • Thermosensitive liposomes can encapsulate drugs to release them at the target site • Angiopep-2 (An2) for BBB transcytosis and anti-CD133 MAb for specific delivery to glioma stem cells have been incorporated in a dual-targeting immunoliposome encapsulating • Targeting molecule transferrin helps the transport of drug to glioblastoma which contains abundant transferrin receptors on the surface • Genetically engineered bacteria that selectively destroys tumor cells while sparing the normal brain tissue Direct Delivery of Therapeutic Agents Gene Therapy • Viral vector mediated insertion of drug sensitivity genes • Insertion of tumor suppressor genes: Transfer of wild type p53 or p27, Retinoblastoma gene transfer

Editor's Notes

  1. Neurons transmit message through chemical & electric signals. Glial cells surround and support neuron cells.
  2. Atypia , cellularity. Pleomorphism, mitotic activity, endothelial prolfration, necrosis
  3. WHO CLASSIFICATION- First time ever, molecular markers along with histology have been used in classification of any tumor Major changes are seen in glioma and medulloblastoma groups. Few entities have been added such as diffuse midline glioma, H3 K27M-mutant, RELA fusion-positive ependymoma, embryonal tumor with multilayered rosettes, C19MC-altered, and hybrid nerve sheath tumors. Few variants and patterns that no longer have diagnostic and/or biological relevance and have been deleted such as glioblastoma cerebri, protoplasmic and fibrillary astrocytoma, and cellular ependymoma. Other changes include deletion of term “primitive neuroectodermal tumor,” addition of criterion of brain invasion in atypical meningioma, separation of melanotic schwannoma from other schwannoma, and combination of solitary fibrous tumors and hemangiopericytoma as one entity. There is also expansion of entities in nerve sheath tumors and hematopoietic/lymphoid tumors of the CNS.
  4. Provides prognostic or predictive data within diagnostic categories established by conventional histology which allow effective targeted treatment 1. Who grade determination are made on the basis of histologic criterias. 2. There are individual tumours that do not need the more narrowly defined phenotype & genotype criteria. Examole- the rare phenotypically classical diffuse astrocytoma that lacks signature genetic characterstics of IDH & ATRX mutations.
  5. A key concept was that diagnoses should be “layered” in order to provide a format for displaying multiple types of information
  6. The Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy—Not Official WHO (cIMPACT-NOW)
  7. The symtoms partly depend on where the tumour is & how fast it grows. 50% headaches – Early morning due to venous congestion A/w nausea and vomiting poorly localized and worsened on Valsalva Seizures 50-80%
  8. FRONTAL- GBM-MC GD 1 - CEREBELLUM
  9. DUE TO INFILTRATIVE NATURE OF DIFFUSE LGG, THE CT APPEARANCE OF LGG IS NON ENHANCING ,POORLY DELINEATED AREA OF LOW ATTENUATION. GRADE 2- isodense or hypodense, no enhancement GRADE 3- low attenuation, variable enhancement GRADE 4- hyperintense with contrast, surrounding vasogenic edema
  10. Bright on T1: Fat , Haemorrhage, Melanin ,Early calcification,Proteins (colloid cyst/ rathke cyst), Posterior pituitary ,Gadolinium Tumor on T1- hypointense
  11. T1 – subacute hmg , fat , anatomy T2 – edema , tumour, infarction, hmg FLAIR – edema , tumour , periventricular lesions
  12. Most of the time lesion is identified on T2/Flair.
  13. Perfusion images measures blood flow in tumours & helps to guide sterotactic biopsy. Increased rCBV in LGG is a marker of malignant transformation prior to development of enhancement. Conventional MR imaging is very limited in making the distinction from primary tumor versus metastatic. Contrast enhancement on T1w images reflect areas of BBB breakdown regardless of pathology. FLAIR images can depict large portion of tumour but is not specific.
  14. A) Low grade astrocytoma (WHO grade I), B) Low grade astrocytoma (WHO grade II), C) Anaplastic astrocytoma (WHO grade III) and D) Glioblastoma multiforme (WHO grade IV). LGG- Increased choline- increase membrane turnover, decreased aspartate- reflecting neuronal losss & increase myoinositol- glial proliferation. HGG_ Increased lactates & lipids is A/W more aggressive behaviour.
  15. FDG is actively transported across the intact BBB. FDG uptake - information on tumor aggressiveness and can act as an adjunct to prognostication markers. Suspicious lesions on MR imaging, that show increased FDG uptake are likely to represent tumour recurrence , rather than radiation necrosis MET uptake by normal brain cells is lower than FDG uptake.
  16. Open & sterotactic biopsy Are done to guide resection & used when most or all the tumour can’t be removed ex- spinal cord, midline structures.
  17. CSF spread of tumor - CSF pressure above 150 mm H2O at the lumbar level in a laterally positioned patient, elevated protein level (>40 mg/dL in the lumbar cistern), a reduced glucose level (below 50 mg/mL), and the finding of tumor cells by cytologic examination.
  18. Kernohan Grading System
  19. IDH mutated tumours are A/W with a more favourable prognosis
  20. 70-90% have calcifications & tend to have cortical involvement.
  21. The T2-FLAIR mismatch sign describes the MRI appearance considered a highly specific radiogenomic signature for diffuse astrocytoma (IDH-mutant, 1p/19q-non-codeleted molecular status), as opposed to other lower-grade gliomas. It is particularly helpful in distinguishing a diffuse astrocytoma from an oligodendroglioma that will not demonstrate T2-FLAIR mismatch.  On T2 weighted images, these tumours have extensive areas of fairly homogeneous and strikingly high signal. On T2-FLAIR, instead, the majority of these areas become relatively hypointense in signal due to incomplete suppression. At the margins of the tumour, a rim of hyperintensity is usually seen. 
  22. Genetic loss of 1p/19q codeletion describes distinct tumour identity characterized by prolonged natural history irrespective of treatment
  23. 3. QOL especially in preop intractable epilepsy & in insular gliomas.
  24. *Suggesting that the surgical impact on OS may be explained by the fact that surgery delays histological upgrading i.e., malignant transformation. Interestingly, if resection is not complete in 1st go due to functional reasons, then the residual tumour can be removed in 2nd surgery while preserving QOL owing to mechanism of neuroplasticity. Allowing multistage therapeutic approach In IDH-mut astrocytoma: In univariate analysis – no diff in OS was seen between 5.1-15 cm3 residue. Post havoc analysis- post op volume of up to 25 cm3 still showed a significant longer OS vs with >25 cm3
  25. >3 markers- give adjuvant RT
  26. Anaplastic Astrocytoma- Relative hypointensity except hyperintense rim (t2-mismatch sign) Anaplastic oligodendroglioma- temporal lobe tumour- with hypointensity
  27. T1- temporal lobe tumour with hypointense with central heterogenous signal T2- Hyperintense T1w+gadolinium- Enhancing, reduced diffusion, usually sorrounds the necrotic part.
  28. Can be used at time of second surgery
  29. The IDH status is once again THE MAIN PROGNOSTIC FACTOR. As the IDH status was added retrospectively only about 50% got the tests done. This shows that RT plus adjuvant PCV gave a OS of 19 months With a median follow-up of 140 months, OS in the RT/PCV arm was significantly longer (42.3 v 30.6 months in the RT arm,
  30. PFS was significantly shorter for the TMZ-alone arm, which persisted after adjusting for IDH status. Among patients with WHO grade III oligodendroglioma with 1p/19q codeletion, TMZ monotherapy resulted in significantly shorted PFS compared with RT with or without TMZ. The analysis of 36 patients pooled the RT arms & compared them with the TMZ-alone. After a median follow up of 7.5 years, 83.3% of patients in the TMZ-alone progressed compared with 37.5% in the Radiotherapy arms.
  31. The study closed early because the target accrual rate was not met. The differences in progression-free survival (PFS) andTTP (Time to Progression) between the 2 arms were not statistically significant. Patients in the RT+NU arm experienced more grade ≥3 toxicity (75.8% vs 47.9%, P < .001), mainly related to myelosuppression
  32. In the entire study population, 70% of patients had an IDH-mutated tumor, & 70% of tumors showed MGMT methylation. how to manage the IDH wild type subgroup- the clear answer is not known. Also in the IDH wt group particularly below the age of 60yr additional testing should be done for TERT, Chr. 7 loss/ 10 gain, EGFR mutation, H3F3A G34M mutation should be done to confirm whether the molecular the molecular signature is similar to GBM. The full significance of the tests in relation to individualizing the treatment is still being understood.
  33. This is the summary slide of all Grade-III gliomas in which the rold of TMZ is clear in Anaplastic Astrocytoma & the role of PCV in oligodendroglioma. The role of TMZ in OLIGO is still awaited.
  34. Optimum time for Temozolomide capsules is 1 hour prior to radiotherapy on an empty stomach. Anti-emetics to be taken at least 30 minutes prior to the Temozolomide. Consider Co-Trimoxazole (Septrin) prophylaxis for PCP 480mg twice daily on Monday, Wednesday and Fr IV temozolomide (TEMODAR) is used as the same dose as oral capsule over 90 mins infusion
  35. MGMT is a DNA repair enzyme that repairs damage caused by alkylating agents such as TMZ. MGMT promoter methylation results in epigentic silencing & decreased expression of MGMT which increases tumour sensitivity to alkylating agents. TMZ cause methylation of the Guanine base at the O6 position and thus damaging the Base. This damaged base is unable to pair with Cytosine and instead pairs with thymine. The Mismatch Repair enzymes try to correct it but often this is not possible and the double strand breaks ensues which results in cell death.
  36. MGMT gene is present at 10q26 & codes for MGMT enzyme which is a suicide DNA repair enzyme responsible for removing alkylating agents from DNA caused by alkylating agents such as TMZ thus helpomg in DNA repair. TMZ causes alkylation of DNA at 6 th position generating O6-methyl guanine. Normally, This MGMT- O6-methyl guanine methyl transferase enzyme removes the alkyl group from DNA and in turn cause resistance to TMZ. Coming to MGMT methylated scernio, there exist CpG islands on MGMT genome in the promoter region. In neoplastic condition there is methylation in the promoter region so the MGMT synthesis is suppressed leading to action of TMZ ias DNA repair is not possible in GBM.
  37. Till now we have seen that there is established role of Concurrent and adjuvant TMZ in GBM. Now further trials were done which evaluated if there was any benefit in OS by the addition of Bevacizumab in newly diagnosed GBM
  38. Thus Hypofractionated RT was established as standard treatment option in elderly GBM patients.
  39. One of the short coming of stupp et al was that is excluded patients of age >70 years.
  40. Butterfly glioma Almost always poor performance status Eloquent area of brain and therefore not resectable Can treat with concurrent chemoradiotherapy if PS1 but this is very unusual. Palliative radiotherapy if performance status adequate.
  41. Radiotherapy dosing in HGG depends on 2 most important factor i.e., Age & performance status. Here I have mentioned the doses of HGG tumours of different age group and performance status & will try to explain it in tabular form in next slide.
  42. Peri op pts, prior h/o gi bleed/ulcer, receiving NSAIDs / anti coagulants- H2/proton pump blockers
  43. should be fused to the planning CT. Although the entire anatomic volume should be matched, the primary matching focus is the tumor
  44. Historically, GBM patients - treated with whole-brain radiotherapy (WBRT) alone or followed by a cone down boost to the tumour, but high doses were needed to maximise local control. For smaller, spherical frontal and/or parietal tumours 3D-CRT is often sufficient, whereas IMRT/VMAT can provide superior solutions for tumours (e. g. temporal, insular) that are in close proximity to the brainstem or orbit, or which have irregular shapes . VMAT > IMRT → similar conformality and faster planning and delivery
  45. (EORTC 22981/22961, 26071/22072 (Centric), 26981–22981, and AVAglio trials)
  46. CTV expansion margins should not traverse anatomically discontiguous structures or include areas unlikely to be infiltrated by tumor. Inclusion of the bony skull is unnecessary unless direct tumor extension is suspected. The falx is a true anatomical boundary between lobes. With some exceptions, “compartmental crossing” to the contralateral hemisphere or, for example, into the posterior fossa or the brainstem for a supratentorial cortical tumor, is not necessary, but because many infiltrating gliomas “cross” through the anterior and/or posterior corpus callosal tracts, these should be adequately included in CTV margin selection.
  47. ≥95 % of the PTV should receive 100 % of the prescribed dose. 100 % of the GTV should be covered by 100 % of the dose. The maximum dose to any point should be limited to 105 % of the prescription dose and ideally should be located in the GTV
  48. Fatigue- m/c in 40-70% exacerbated during EBRT Brain necrosis- rare and serious, bevacizumab
  49. Follow up consists of clinical evaluation with neurological function, seizures & corticosteroid use.
  50. RANO- radiological assessment in neurooncology RANO recommendation: avoiding enrolling patients within 3 months of completion of radiochemotherapy into clinical trials for recurrent disease, unless the recurrence is mainly outside the radiotherapy field or there is tissue confirmation of progression.
  51.  favorable performance status (KPS ≥70) and younger age, smaller tumors, non-eloquent brain location, greater interval from initial treatment to recurrence, and corticosteroid dependence. Gliadel or Carmustine wafers implantation- it offers statistically significant but marginal improvement (31 vs 23 weeks) in OS after re-operation of HGG. Significant peritumoral edema is a recognized side-effect. Option for patients with high grade glioma and ≥90% resection Biodegradable wafers containing BCNU Gliadel wafers have been tried in newly diagnosed GBM but there was no statistically significant benefit.
  52. Strict immobisation is achieved by either strict head frame or reliable internal fiducial markers (bony landmarks/implanted markers).
  53. In LINAC radiosurgery - spherical target of <4 cm in diameter, miniaturized multileaf collimators allow beam shaping. For lesions larger than 4 cm and/or located in critical regions, the delivery of a single large fraction treatment as in SRS is not desirable because of a high risk of CNS toxicity. Perfexion device, the use of the “Extend” frame permits FSRT as well as targeting of lower cranial lesions. On-board volumetric imaging, modern LINACS and the most recent version of the Gamma Knife (Icon) also permit fractionation, using noninvasive mask-fixation systems.
  54. Edema managed by high dose corticosteroids and bevacizumab Single dose radiosurgical treatment of recurrent previously irradiated primary brain tumors and brain metastases: final report of RTOG protocol 90-05
  55. CORE- Cilengitide in patients with newly diagnosed GliOblastoma multifoRme and Unmethylated MGMT genE promoter. Human telomerase reverse transcriptase (hTERT) transcripts; survival is worse in high Receptor tyrosine kinases as a signal blocker to hinder the growth of gliomas
  56. Chimeric antigen receptor T cells are T cells that have been genetically engineered to produce an artificial T-cell receptor for use in immunotherapy.   small non-coding RNA molecule-RNA silencing and post-transcriptional regulation of gene expression