CNS II General Management & GBMs Flashcards

1
Q

how is cerebral oedema managed

A

glucocorticoids such as dexamethasone, considered the ‘gold standard’.
-> side effects of glucocorticoids, such as dexamethasone, include Cushing’s syndrome, increased risk of pneumonitis, long term osteoporosis and compression fractures.
- Pitter et al (2016) reported that corticosteroid use during RT with or without chemotherapy is an independent predictor of poor outcome in GBM (if you have to continue corticosteroid use during RT it is usually a poor prognostic outcome)
- suggest replace dexamethasone with short term VEGF antagonists instead. this is not the gold standard

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

problems with using corticosteroids in RT practice?

A

swelling - causes problems with masks
in working environment, when booking patients, pre-empt the swelling that the patient might have and CT re-scan that they may need

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

seizure control

A
  • risk of seizures varies between 60-100% in LGGs and between 40% and 60% in GBMs
  • seizures as a symptom are a favourable prognostic factor for survival ( a seizure is showing that where the patient has a tumour, they still have some functioning brain parenchyma )
  • recurrence or worsening of seizures during the disease trajectory may indicate tumour progression
  • one third of patients with brain tumour-related epilepsy show pharmacoresistance to antiepileptic drugs
  • levetiracetam is the drug of choice, followed by valproic acid, based on the evidence ( these drugs can be very nauseating )
  • if seizures are recurring, these can be combined
  • other options are: lacosamide, lamotrigine or zonisamide if the others are not tolerable to the patient
  • valproic acid is of interest as it may prolong survival in patients with GBM (EORTC study, 2011)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

surgical management

A

best prognosis with maximal safe surgery typically using a craniotomy
- CNS tissue is intolerant of trauma ( if a brain surgeon removes some neurones, they are gone and can no longer function. you cannot come in with the margins that you would in other sites)
- CNS tissue is not capable of regeneration
- CNS tissue is critical to normal body functioning
- surgical debulking is most frequent procedure, maximising brain function

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

medical oncology management

A
  • chemotherapy drugs as radiosensitisers for treatment of gliomas
  • BCNU (Carmustine) can be directly implanted into the brain (Glial wafer) and CCNU (Lomustine) ( this negates any issues with blood brain barrier. work best at rest phase of cell cycle, therefore cell cycle non-specific )
  • procarbazine
  • vincristine ( inhibit microtubule formation and hence prevent cell division, cell cycle specific )
  • temezolomide
  • lipid soluble drugs so can cross blood-brain barrier
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

general guidelines for target volumes

A
  • in general the GTV is individualised for each tumour volume based on the likelihood of the tumour to infiltrate ( grade 1 tumour a lot less likely to infiltrate surrounding brain parenchyma than a grade 4. you must follow the disease and know the disease tract of the tumour )
  • the disease must be followed along with the white matter tracts and a non-uniform (isotropic) margin used
  • volumes should pay respect to anatomical borders ( strange shapes. awkward for tx planning )
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

field arrangements CNS tumours

A
  • mainly modulated techniques especially for hippocampal sparing in whole brain treatments ( hippocampus is responsible for memory. QoL of patients. you must be able to taper the dose –> modulated techniques )
  • be careful of exit doses, vertex fields are problematic in some scenarios with photons
    -> high dose to thyroid, carotid artery and jugular vein
    ( vertex fields means you are shooting straight through the patient. couch is turned and gantry is turned to either 90deg or 270deg. vertex field goes through the head of the patient and hits everything through the patient on exit dose. if you raise the couch and turn the gantry slightly - this is called a superior anterior oblique - the gantry is at 60deg and the couch is at 90deg or the gantry is at 300deg and the couch is at 90deg. **juvenile patients - you don’t want to cause any issues to thyroid / carotid artery / jugular vein )
  • location of hot spots critical, remember ‘double trouble’
    ( ‘double trouble’ - if you are treating a patient’s brain to 60Gy/30# but there is a hot spot of 110% near the optic chiasm. This would mean that this hot spot is getting 2Gy x 110% = 2.2Gy/# and it is also getting 60Gy x 110% = 66Gy. something like this would render the patient blind )
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

OARs CNS tumours

A

when contouring target volume and OARs, fuse planning CT and pre- and post-op MRIs
OARs:
- brainstem
- hippocampus
- hypothalamus
- lens
- orbits
- pituitary gland
- lacrimal glands
- middle and inner ear
- optic apparatus (optic nerve & optic chiasm)
- spinal cord
- cochlea
( there is a hierarchy of OARs. Lens is always bottom of the list - we would never compromise target coverage to spare the lens / optic apparatus )
(Tolerance of normal tissues: dependence on TD and fractionation, in context of histology, site, PS and expected OS.
Brain parenchyma: 54-60 Gy in 30 fractions
Risk of necrosis 5% with a dose of 60 Gy in 30 but volume dependant.
A volume effect also exists for intellectual damage., in theory ok with doses up to 54Gy in 30 fractions but not well understood.
Brain stem: 54Gy in 30 or 55GY in 33 (same)
Optic nerves/chiasm: 45-50 Gy in 1.67 or 1.8 Gy fractions
Pituitary and hypothalamus: Little long term effect for doses under 20-24 Gy. In children, doses of 40-60Gy damage hormone secretion. Adults who receive 50-60Gy have significant long-term pituitart-hypothalamus dysfunction.
Middle and inner ears: doses up to 60Gy recover function
Lacrimal glands: 20Gy
Lens: 5-6Gy over 30 fractions no cataracts.
Permanent alopecia: Depends on dose to hair follicles in the dermis however 50% of patients can experience alopecia with a dose of 43Gy. )

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

GBM

A
  • ** GBMs are derived form neural stem cells, neural stem cell-derived astrocytes and oligodendrocytes precursor cells
  • ** GBM is extremely challenging to treat with 5-year overall survival being 7.2%, according to US data
  • ** almost all GBMs recur even after aggressive management due to:
    -> incomplete resection and high infiltrative nature of GBMs ( maximal safe surgery. can cross cerebral hemispheres )
    -> high degree of genetic heterogeneity within the tumour ( one part of the GBM could be completely different to the other parts )
    -> immunosuppressive tumour microenvironment ( unless you have a hot tumour microenvironment you won’t be suitable for immunotherapy)
  • GBMs are highly hypoxic and permit the development of glioma initiating cells, which are self-renewing ( for RT to work, it must be in a highly oxygenated environment to create the free radicals that will damage the cells. There is also a highly necrotic cell volume with GBM, RT will not work in necrotic environment )
    -> this can lead to potentially more aggressive recurrent tumours that are radio and chemo resistant
  • genetic heterogeneity makes target therapy development problematic as there are 4 genomic subtypes of GBM known:
    -> Mesenchymal (Neurofibromatosis (NF1, PTEN TP53 gene mutation)
    -> Classical (EGFR amplication, no TP53 mutation)
    -> Proneural (TP53, IDH1, PDGFRA mutations)
    -> neural (contamination of original tumour with non-tumour cells)
  • issue is that all of these subtypes can vary spatially and temporally within the same tumour, according to RNA analysis
  • immunosuppressive microenvironment of GBMs is inhospitable as the tumours lack pre-existing tumour T cell infiltration, making them resistant to immune checkpoint inhibitors (known as ‘cold’ tumours). RT can modulate this microenvironment and turn these ‘cold’ tumours to ‘hot’.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

presentation of GBM

A
  • majority of the malignant gliomas are GBMs ( stage 3 and 4. most are stage 4 )
  • arise in adults, with a peak incidence at 45-65 years
  • usually found in frontal and temporal lobes
  • extremely necrotic and haemorrhagic
  • may be multi-focal ( could arise in more than one location )
  • main presentation is headaches, followed by seizures
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

surgical oncology management of GBM

A
  • extent of surgical resection is positively correlated with survival time
  • aim for gross total resection but problematic:
    -> successful identification of tumour margins, microscopic finger-like projectile growth of GBMs
    -> avoiding eloquent areas ( pt typically awake during procedure )
    -> ‘brain shift’ - movement of brain during surgery relative to pre-operative imaging (gravity, pt position, tissue/fluid loss during surgery, drugs used to manage intracranial pressure)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

medical oncology management GBM

A

standard is the Stupp protocol post-operatively
- 6 weeks of concomitant Temezolomide with RT (75mg/m^2), followed by adjuvant Temezolomide (150-200mg/m^2) for 5 days every 28 days for 6 cycles. ( = Temadol / Tremadol. Pts can be very nauseous with Temezolomide. Taken in tablet form chemo )
- Temexolomide is a small molecular alkylating agent that methylates the purine bases of DNA and thereby damages its structure
- Its main cytotoxic action is via O^6-methyguanine lesions that lead to cellular senescence, apoptosis and autophagy ( cellular senescence = cells getting old. apoptosis = cell death by cell suicide. autophagy = cells consuming themselves )
- used with RT to increase the likelihood of double strand breaks
- side effects of Temezolomide are haematologic primarily
-> thrombocytopaenia presents in 10-20% of patients
- non haematologic side effects are less common and include nausea, fatigue, anorexia and hepatotoxicity
- BCNU in the form of Gliadel wafters can also be considered for GBM as can CCNU but Temezolomide is the current gold standard

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Methyl Guanine Methyl Transferase (MGMT)

A
  • Temezolomide kills tumour cells by transferring methyl groups to DNA - it alters the DNA configuration by attaching a thymine rather than a cytosine during DNA replication
    -> this damage can be reversed by the DNA repair enzyme MGMT, hence the methylation status of the MGMT gene promotor has very relevant clinical significance
    -> MGMT inactivation or ‘silencing’ is associated with improved OS and PFS and is a very important prognostic biomarker in the Stupp protocol
    -> MGMT status is determined using assays such as Methylation-Specific PCR, but there are others and consensus on which assay is optimal as yet.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

medical oncology management GBM

A

Bevacizumab is used predominantly in the treatment of recurrent GBM ( recurrent - pts who started with a lower grade glioma that then changed into GBM )
-> monoclonal antibody administered intravenously
-> has anti-angiogenic properties and inhibits VEGF-A to stop angiogenesis
-> levels of VEGF-A are 30 times higher in GBM than in low grade glioma, hence its use mainly in GBM
-> can improve PFS in newly diagnosed GBM but not OS (unlike Temexolomide), therefore mainly used in recurrent disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Indications for RT and systemic therapies (ASTRO Delphi methodology)

A
  • RT improves OS compared with best supportive care or chemo alone
  • use of fractionated RT and TMZ as standard of care in patients up to 70 years of age
  • addition of Bevacizumab does not improve OS but may prolong PFS
  • addition of other systemic therapies to RT and TMZ remains investigational
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Dose/fractionation and variance in this (ASTRO Delphi methodology)

A
  • no benefit to anything other than 60Gy in 2Gy fractions over 6 weeks in patients <70 years with KPS >= 60
  • patients >= 70 years with fair-good PS (KPS >= 50), RT following biopsy or surgery instead of supportive care alone
    -> TMZ has not been investigated in an RCT in this group alone but can be considered
  • for older patients, hypofractionated treatments (40Gy/15#) are as effective as 60Gy/30#. Superior survival and less requirement for steroids using hypofractionated regimen and is recommend for older patients with fair-good PS
  • Temezolomide can be an effective alternative in older patients with MGMT promoter methylation but not in those with unmethylated promoters
  • poor performance status patients: hypofractionated RT alone, TMZ alone or best supportive care
17
Q

Target volumes (ASTRO Delphi methodology)

A
  • ** partial brain RT does not have inferior survival to WBRT. Partial brain RT is the SoC. ( ** GBM are volumed partial brain treatments )
  • can use 2-phase or a single-phase approach
  • in the 2 phase approach: phase I covering oedema on T2 or FLAIR and gross residual tumour/resection cavity. Phase II encompasses the gross residual tumour or resection cavity only
  • single phase approach includes gross residual tumour/resection cavity with wide margins, without specifically targeting oedema
18
Q

Re-irradiation (ASTRO Delphi methodology)

A
  • need to consider tumour size and location, but in younger patients with good PS, focal re-irradiation (stereotactic or brachytherapy approach) may improve outcomes compared to best supportive care or systemic therapy alone
19
Q

key recommendations from ESTRO

A
  • single phase treatment, based on T1/T2 FLAIR abnormalities with isotropic margins ( ESTRO more specific than ASTRO )
  • GTV = surgical resection cavity plus any residual enhancing tumour on postcontrast T1 MRI ( Gadolinium is usual contrast used )
  • CTV = GTV + 2cm margin edited for anatomical barriers to tumour spread
  • PTV = institution-dependent, typically CTV + 3-5mm
20
Q

DVC: Brainstem (ESTRO)

A

Dmax <= 54Gy
1-10cc < 59Gy (periphery)
( 2 objectives. maximum constraint demonstrating serial architecture. it also has volumetric constraint )

21
Q

DVC: Optic Chiasm (ESTRO)

A

Dmax < 55Gy

22
Q

DVC: Cochlea (ESTRO)

A

Ideally one side mean < 45Gy

( mean dose indicates parallel architecture )

23
Q

DVC: Eyes (ESTRO)

A

Macula <= 45Gy

24
Q

DVC: Lacrimal glands (ESTRO)

A

Dmax < 40Gy

25
Q

DVC: Lens (ESTRO)

A

Ideally Dmax < 6Gy but up to Dmax <10Gy acceptable
( this DVC comes from RTOG. you would never compromise your plan according to lens dose. For GBM lens dose is kind of irrelevant, patient doesn’t live long enough to have effects )

26
Q

DVC: Optic nerves (ESTRO)

A

Dmax < 54-55Gy

27
Q

DVC: Pituitary (ESTRO)

A

Dmax < 50Gy
( we do not compromise plan according to pituitary )

28
Q

dose recommendations from ESTRO

A
  • 60Gy in 30# over 6 weeks is standard
  • hypofractionated regimens in those > 70 years or those with poor PS (KPS <70). can be 40Gy in 15# over 3 weeks ( trial by Roa et al. ) or 34Gy/10# over 2 weeks ( Danish group )
  • phase II data illustrates that 30Gy in 5-6Gy per # can also be utilised ( Cohort study not a RCT )
29
Q

RT Pathway GBM frontal lobe: Positioning & Immobilisation

A

Supine 3 point thermoplastic mask. Chin in line with feet (neutral position). Paediatric for medulla blastoma is typically flexed neck. Most important stability points on mask: bridge of nose, forehead, chin. Open face masks must be very stable on shin and part of forehead. Open face mask with SGRT. SGRT is compensating for the open immobilisation

30
Q

RT Pathway GBM frontal lobe: Simulation/CT: borders? slice thickness? contrast?

A

Borders: clear of the vertex / top of skull to around C2. Slice thickness: 1-2.5mm. 2mm is optimal. Contrast: YES enhances tumour volume and blood vessels. Must be Gadolinium - lipid soluble contrast
DLP = dose length products
You must include full length of organ for OARs
1mm slices give great detail (cochlea) and no issue of partial volume effect on smaller slices
When it comes to planning, 1mm slices are very time consuming - delineation and calculations take a lot longer
Anything higher than 2.5mm -> partial volume effect

31
Q

RT Pathway GBM frontal lobe: Target volume and OAR delineation

A

T1 T2 FLAIR for target volume
OAR - must have full length of organs

32
Q

RT Pathway GBM frontal lobe: Treatment Planning

A

Modulated techniques mostly standard of care. Sometimes get better heterogeneity within target is better with 3DCRT but better OAR sparing with modulated techniques. If you have a tumour very close to a critical structure, you may have to be more strict. VMAT typically 2 arcs. Avoidance sectors within the arc when going over critical structures. Inverse optimisation. Sometimes conflict between target and dose to OAR. May present multiple plans to RO and decide on most suitable

33
Q

RT Pathway GBM frontal lobe: Treatment Verification

A

IGRT. Daily online imaging CBCT. You can optimise the imaging settings to minimise dose to patient. Motion within the brain is a lot less in comparison to other sites. Offline imaging can be okay for GBMs because of the lack of motion. Offline review is different to offline imaging. Offline imaging - no action level protocol (NAL) not really done anymore. Image on days 1-3 or 1-4 if they are in tolerance, no more imaging // extended no action level protocol (eNAL) image days 1-4 or 1-4 and then calculate shifts and weekly image. If image is OOT on weekly image, you must go back to imaging day 1-3. offline imaging looking for systematic error. If you don’t reduce systematic errors you may not achieve the plan that you have aimed for. Not a lot of motion but patient can be swelling up and down day to day

34
Q

RT Pathway GBM frontal lobe: psychosocial and patient care

A

Terminal diagnosis. Be more cautious and aware of what you are saying to these patients. These patients may have a lot of guilt because of impact on their family

35
Q

RT Pathway GBM frontal lobe: side effect management

A

Acute side effects - headaches go up and down. Usually get better with RT but the headaches can get worse at the beginning. Pain management is very important. FATIGUE can be very extreme in these patients. Advice for fatigue - light exercise if they are able to. Light exercise for these patients could be walking to their gate and back. Plenty of rest and plenty of fluids. If patient has loss of short term memory, make family members aware. Alopecia - if you can look at their plan in advance and let patient know at their first day chat what areas they are likely to loose their hair.
If you treat hair follicles to more than 46Gy (Dmax >46Gy) the hair is not likely to grow back. Ensure patient is aware of this. How people can look when they are dead can be very important to them. shaving head and skin toxicity - be aware of this Using blow drier/hair straightener not recommended. Baby shampoo. Do not wash hair too often. Ask them to make you aware they will be shaving head - mask
Make patient as aware of side effects as possible. Very unique way of giving care to patient CASE STUDIES

36
Q

future treatments for GBM

A

Inhibitor therapies: PARP (Ploy (ADP ribose) polymerase) is a family of 17 nuclear enzymes. PARP inhibitors can have a synergistic effect together with Temezolomide and RT
- protein kinase inhibitors
- chimeric antigen receptor T (CAR T) cells ( CAR T cells have cured people with lymphomas and some melanomas )

The future of RT in the management of GBM will be in priming and activating immune cells to tumour-derived antigens, and improving the immuno-suppressive environment (turning from ‘cold’ to ‘hot’) to sensitise GBMs to immunotherapies