Head & Neck Flashcards

1
Q

Discuss EGFR in the context of H&N SCC

A

EGFR = Transmembrane/cell surface glycoprotein, part of receptor tyrosine kinase family; role in signal transduction

Structure - consists of 3 domains:
- Extracellular Ligand-Binding Domain: binds epidermal growth factor and other ligands. Binding of the ligand to domain causes a conformational change that activates the receptor
- Transmembrane Domain: single alpha-helix segment spans across the cell membrane - role in the receptor’s dimersiation with ligand binding
- Intracellular Tyrosine Kinase Domain: within the cell, this domain has tyrosine kinase activity. Upon activation, it autophosphorylates specific tyrosine residues, triggering downstream signalling pathways.

EGFR in H&N SCC
- Overexpression:
□ EGFR often overexpressed in head and neck SCC → associated with poor prognosis and aggressive tumour behaviour.
□ Overexpression leads to enhanced signalling that promotes cell proliferation, survival, and migration
- Target for Therapy:
□ eg. EGFR inhibitors which aim to block signalling (or its down stream signalling molecules); thereby inhibiting tumour growth and progression
□ Eg. cetuximab (a monoclonal antibody)
□ Eg. Erlotinib (tyrosine kinase inhibitor)
- Predictive Biomarker:
□ EGFR expression levels and mutations can serve as biomarkers to predict the response to targeted therapies.
□ High EGFR expression may indicate a better response to EGFR inhibitors.
- Resistance Mechanisms:
□ Despite efficacy of EGFR-targetted therapioes, resistance can develop.
□ includes mutations in the EGFR gene, activation of alternative signalling pathways, and phenotypic changes in cells.

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

Describe the epidemiology and biological behaviour of HPV associated H&N cancers and compare to HPV negative cancers

A
  • Aetiology
    * Mainly mediated by HPV 16
    * E6 inactivates p53, E7 inactivates RB
    • Microscopic appearance
      • Usually non-keratinising SCC (basaloid type), predominantly oropharynx (BoT, tonsil)
      • IHC - p16 positive (surrogate marker)
      • HPV detection through ISH or PCR
    • Biologic behaviour
      • Earlier T stage, but more advanced N stage than non-HPV cancer
      • Favourable prognosis
        • 5yr OS St 3-4 (AJCC 7th edition) 80% vs. 40% cf. non-HPV associated oropharyngeal cancer
      • Responds better to treatment
    • Epidemiology
      • Increased incidence of BoT and tonsil cancer, despite decreased incidence of smoking and H&N cancer at other site
      • Incidence: Increasing, 70% of oropharyngeal SCC in Australia
      • Age: Middle-aged (younger than non-HPV)
      • Sex: M>F
      • Often higher socioeconomic status, non-smokers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Describe HPV mediated carcinogenesis

A

HPV has high-risk and low-risk subtypes
* HPV 16, 18, 31, 33 are high-risk
* Remainder are low risk (e.g. 6, 11)

HPV replicates by entering a cell and incorporating its own DNA into the cellular DNA. In this way, the cellular machinery is manipulated to create many copies of the virus, which is then released for further spread and infection.
 
The E6 and E7 oncoproteins of the high-risk HPV subtypes manipulate cellular machinery in such a way to cause carcinogenesis
	* E6 associates with the p53 protein and leads to the ubiquitination, thereby inhibiting its function. E6 also causes other more minor protease-mediated cellular damage
	* E7 associates with the Rb (retinoblastoma) protein and causes its proteasome-mediated degradation.
E5 is also oncogenic, but less potent
	* E5 can activate EGFR signalling and thus promote cellular growth
 
This in turn, causes carcinogenesis in the following ways
	* Cellular immortality (disabled apoptotic pathways)
		* E6 disables p53
	* Genomic instability (impaired cellular DNA repair pathways)
		* Disabled apoptosis allows accumulation of genetic damage without an endpoint
	* Unregulated cellular proliferation (removal of cell-cycle arrest mechanisms)
		* E6 disables p53
		* E7 disables Rb
	* Sustained proliferative signalling
		* E5 can activate EGFR pathway
	* Impaired immunity (ongoing infection and inflammation)
		* E6/7 inhibit interferon signalling (impair innate immunity)
		* E5 down-regulates MHC-I expression
		* This leads to persistence of infection (which is required for carcinogenesis --> accumulate genetic damage)
	* Other
		* E7 induces angiogenesis
		* E7 induces Warburg effect (altered metabolism)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Describe the pathological features of leukoplakia and erythroplakia

A
  • Leukoplakia
    * Definition / clinical features: Clinical descriptive term. White patch or plaque on mucosa (solitary or multiple) that cannot be scraped off and cannot be characterised clinically or pathologically as any other disease
    * I.e. if a white lesion can be given a specific diagnosis, it is not leukoplakia
    * Location: Commonly buccal mucosa, floor of mouth, ventral surface of tongue, hard palate
    * Microscopic appearance:
    * Spectrum of epithelial changes ranging from:
    * Hyperkeratosis overlying a thickened, acanthotic but orderly mucosal epithelium; to
    * Markedly dysplastic changes, sometimes merging into carcinoma in situ
    * With increasing dysplasia there is an increasing frequency of subepithelial inflammatory infiltrate of lymphocytes and macrophages
    * Biologic behaviour:
    * 5-10% progress to invasive disease
    * Considered pre-malignant
    * Management:
    * Options: observe or resect
    * Decision based on compliance with follow up, location (ease of resection) and presence of erythroplakia (higher malignant transformation potential)
    * All patients should encouraged to cease smoking and alcohol
    • Erythroplakia
      • Clinial appearance: Red velvety, possibly eroded lesion
      • Epidemiology: Less common but more ominous
      • Microscopic: ]
        • Only rarely consist of orderly epidermal maturation
        • Virtually all (~90%) contain superficial erosions, with dysplasia, carcinoma in situ or already developed carcinoma in the surrounding margins
        • Often an intense subepithelial inflammatory reaction with vascular dilatation is seen that likely contributes to the reddish appearance
      • Biologic behaviour: 30-50% progress to invasive carcinoma
      • Management: Should be resected due to high risk of malignant transformation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Describe the incidence and RF for oral cavity cancers

A

Incidence (Australian statistics)
- ALL = 4500 cases annually
- ORAL = 2000 cases annually
- Approx 8 per 100000 people

Rising incidence of young people < 40yo (especially females)
- Likely HPV related

Median age is 60 years
Male predominance (up to 4:1)

Aetiology

1) Smoking (RR is up to 10x)
	a. ALSO - cigar use, chewing tobacco, betel nut
2) Alcohol use (RR is up to 5x)
	a. Synergistic with smoking
3) Leukoplakia
	a. Up to 10-25% of these lesions will progress to cancer
4) Occupational exposure
	a. Asbestos
	b. Mustard Gas
	c. Nickel
	d. Tar
5) Viral
	a. HPV is implicated (less strongly than for oropharynx)
6) Immunosuppression
7) Nutritional deficits
	a. Fruits and vegetables are protective
8) Poor oral hygiene & periodontal disease
9) Previous radiation
10) Genetic factors
	a. Fanconi anaemia
	b. Ataxia telangiectasias
	c. Xeroderma pigmentosum

Metachronous lesions
- Second H+N primary = 20% risk
- Lung = 5-10%
- Oesophagus = <5%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Describe the pathological features of hyperplasia and dysplasia

A
  • Squamous hyperplasia
    * Response to carcinogen exposure, mechanical irritation or other noxious stimuli. Tobacco is the most common cause
    * Favoured locations
    * Buccal mucosa
    * Floor of mouth
    * Ventral surface of tongue
    • Squamous dysplasia
      • Explanatory notes:
        • In normal squamous mucosa, mitotic activity is confined to the basal layer of cells (small cuboidal cells with dark nuclei and scant cytoplsam) and immediately adjacent cells (para-basal)
          • As cells mature -> move up, acquiring more cytoplasm -> become progressively keratinised
          • Near surface, cells flatten out and nucleus disappears
        • In squamous dysplasia, normal architectural pattern of maturation is disturbed, indicative of neoplastic transformation
        • Progressive disorganisation and loss of maturation (polarity) of the squamous epithelium as cells move upward from the basal layer to the nonkeratinised surface
        • Dyskeratosis (out of sequence individual cell keratinisation) often accompanies loss of polarity
      • Morphology
        • Loss of polarity, often accompanied by dyskeratosis extending from the basal layer of the epithelium
        • Mitotic figures
        • Nuclear pleomorphism may be present in all grades but is not a conspicious feature
      • Degree of dysplasia is graded based on extent of thickness of eptihelial layer involved with dysplastic basal type cells
        • Mild dysplasia: Abnormal architectural and cytological features largely confined to hte lower thrid of the epithelium
        • Moderate dysplasia: Dysplastic process extends to middle thirdof epithelium
        • Severe dysplasia: Extension of preoces sinto the upper third of the epithelium
        • Carcinoma in situ: Full thickness invovlement is present in the absence of invasion (for practical purposes severe dysplasia and carcinoma in situ are synonymous
      • Biologic behaviour
        • May regress spontaneously
        • May progress to invasive malignancy
          • Relationship less well defined than for cervix SCC
          • Moderate dysplasia: 4-11% risk
          • Severe dysplasia: 20-35% risk
        • Invasive malignancy may develop in the absence of surface dysplasia
    • Clinical appearance
      • Both squamous hyperplasia and dysplasia may present as leukoplakia
      • Erythroplakia usually represents squamous dysplasia, carcinoma in situ or invasive carcinoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Describe the mechanisms for developing synchronous tumours in the Head and neck.

A

Field cancerisation
- Due to equal carcinogenic distribution, all areas are likely to have mutational damage
- Ie. Entire mucosal surface of upper aerodigestive tract exposed to carcinogens (smoking, ETOH, HPV, chronic inflammation), leading to widespread genetic alterations and a higher risk of developing multiple primary tumours

Monoclonal seeding:
- multiple primary tumours may arise from the clonal expansion of a single mutated cell that has spread across different sites aerodigestive tract
- through intraepithelial migration or dissemination (and re-implantation) of malignant cells within the mucosal field
- Seemingly new tumour deposit proliferates

Genetic predisposition
- Mutations in tumor suppressor genes (e.g., TP53) or DNA repair genes increase the risk of multiple primary cancers.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are the histological subtypes for oral cavity cancers?

A
  • Squamous Cell Carcinoma (95%)
    ○ Verrucous carcinoma (5%)
    ○ Spindle cell
    ○ Basaloid
    • Salivary gland
    • Neuroendocrine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Describe the pathological features of oral cavity SCC.

A
  • By far, the most common histological subtype of oral cavity cancer
    • Macroscopic
      ○ Pink/grey area of mucosal thickening
      § Ulcerated mass when late/advanced
      ○ May be superimposed on leukoplakia
    • Microscopic
      ○ Nests of keratinising squamous cells which invade the basement membrane
      § Keratin pearls and intercellular bridges
      ○ Variable cellular anaplasia
    • Variants
      ○ Verrucous –> white, warty, cauliflower like lesion
      § Low metastatic potential
      ○ Sarcomatoid/spindle cell –> biphasic mixed carcinoma with spindle component
      § Aggressive disease course
      ○ Basaloid –> small basaloid cells with peripheral palisading and high-grade features (high N;C ratio, pleomorphism, hyperchromatic nuclei, mitoses)
      § Aggressive disease course
      § o Positive stains: 34beta E12, EMA, AE1/3, CAM 5.2, neuron specific enolase, CEA, S100
      ○ o Negative: chromogranin, syanptophysin, muscle specific actin, GFAP
      ○ adenosquamous
    • Immunohistochemistry
      ○ POS = AE1/AE3, HMWCK
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What are the subsites of the oral cavity and what is their respective lymphatic drainage routes?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What are the prognostic factors for oral cavity cancers?

A

Patient Factors
- Age and performance status
- Nutritional status
- Immunosuppression
- Smoking (local control and toxicity)
- Anaemia
○ Non-reversible –> transfusion does not improve outcomes

Tumour Factors
- TNM stage
- Tumour site
○ Tongue is worse
- Depth of Invasion
- Pattern of invasion (discohesive cells at tumour front)
○ Worst is pattern 5 = satellites >1mm from main tumour, or PNI, LVI outside tumour
- Grade (of limited value)
- LVI
- PNI
- ECE

Treatment Factors
- High-volume experienced centre
- Surgical margins
- Addition of adjuvant radiotherapy
○ Overall treatment time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What features on history and examination for oral cavity cancers are important?

A
  • History
    ○ HPI
    § Observation
    □ Non-healing ulcer (painful or painless)
    □ Ill-fitting dentures
    □ Exophytic mass or leukoplakia
    □ Red area
    § Odynophagia
    § Speech deficits
    § Bleeding
    § Otalgia (referred)
    § Trismus
    Neck lumps (30% LN mets at diagnosis. ○ ant tongue common 50-60%
    ○ Hard palate/aveolar ridge uncommon
    ○ lip <10%, but more common in T3/4 or with commisure involvement. Rare to have distant mets
    ).
    ○ PMHx:
    § Previous H+N cancer
    § Previous radiotherapy
    § Immunosuppression
    ○ Meds:
    § Immunosuppression
    ○ Social:
    § Smoking
    § Alcohol
    § Other (e.g. betel nut)
    • Examination
      ○ Thorough oral cavity examination
      § Ulcerated lesions
      § Fixation to underlying structures
      ○ Should inspect remaining mucosa for synchronous lesions
      ○ Mental nerve function
      ○ Cervical lymphadenopathy
      ○ Flexible Nasendoscopy to exclude synchronous lesions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What investigations would you order for oral cavity cancers?

A
  • Biopsy
    ○ EUA
    ○ Biopsy any equivocal neck nodes (unless already proceeding with neck dissection)
    § USS guided FNA
    § If unresectable or metastatic disease then core bx for immune-genomic testing
    • MR Head and Neck
      ○ Surgical evaluation
    • FDG-PET
      ○ Nodal and distant staging
    • Diagnostic CT Head and Neck
      ○ Evaluate bony invasion
    • Pre-chemotherapy bloods
      ○ FBC
      ○ EUC, CMP
      ○ LFT and coags
      ○ HepB sAg, sAb, cAb
      ○ Hep C serology
      ○ HIV serology
    • Dental assessment
    • OPG
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is the TNM staging for oral cavity cancers?

A
  • Stage I
    ○ T1N0
    • Stage II
      ○ T2N0
    • Stage III
      ○ T3 OR N1
    • Stage IV
      T4+ OR N2+
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are the general principles when managing oral cavity cancers?

A
  • Always aim to use as few modalities as is feasible (reduce morbidity)
    • Always establish relevant supportive care early
      ○ Smoking cessation
      ○ Dietician and speech pathology input
      § Consideration of prophylactic gastrostomy
      ○ Dental input
      § Pre-RT OPG
      ○ Social & psychological supports as appropriate
    • In general, options may include
      ○ Early Stage (stages I-II)
      § Surgical resection alone (+/- adjuvant RT)
      § Radiotherapy alone (uncommon)
      ○ Advanced Stage (stages III-IV)
      § Surgical resection + adjuvant RT
      § Chemoradiotherapy (less common)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

In general compare surgery vs RT, for oral cavity cancers.

A

Surgery preferred
· Less morbidity à ± adjuvant RT or CRT
· Allows pathological stagin
· Control rates poorer in early stage dis
· Easily accessible to sx, often good fu’nal and cosmetic results
· RT can have signif tox
· Elective neck for T1-2 (D’Cruz, 2015, demonstrated OS benefit)
RT
· Can’t tolerate sx or sx resection would result in severe fu’nal impair
· Uncommon in stage I/II oral tongue & FOM
· May be applicable to early tumors of lip or retromolar trigone
· Absolute contraind: bone involve, airway compromise
Relative contraind: cartilage involve, poor organ fu’n, connective tissue (CT) dis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

In general what is the management for Stage I-II oral cavity cancers?

A
  • Surgery to the primary forms the basis of management
    ○ Aim for macroscopic margin of 10mm (for path margin of 5mm)
    § In the case of positive margins, re-resection should be attempted
    ○ Always consider the functional deficit with surgery
    ○ Associated with less morbidity (even with adjuvant RT)
    ○ Allows for pathological staging (including depth of invasion)
    • Definitive radiotherapy should be considered if medically or technically inoperable
      ○ Indications for Definitive RT over surgery
      § Lip: oral commissure involved, >2cm, >50% of lip, upper lip
      § Buccal mucosa - T1-2N (Tx ipsilateral Ib-II)
      § RMT with tonsillar pillar involvement
    • Most patients should have elective neck management (except for early tongue DOI <1mm or T1 lower lip, otherwise all need elective neck management)
      ○ Laterality is dependent on primary site
      ○ In most cases, should include levels I-III
      § Consider level IV in oral tongue (skip lesions)
      ○ Exceptions to elective nodal management
      § T1 SCC of the lower lip
      § Small T1 SCC of limited DOI (<1mm)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Describe management options for oral tongue and floor of mouth cancers.

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Describe management options for Retromolar trigone and buccal mucosa cancers.

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What are the indications for adjuvant RT in oral cavity cancers?

A
  • Consider adjuvant post-operative radiotherapy alone if (INTERMEDIATE RISK):
    ○ Close/positive margins
    ○ ENE on histopathology
    ○ T3/T4 disease
    ○ 2+ nodes involved (i.e. N2+ disease)
    ○ DOI >4mm (specific for oral cavity)
    ○ LVI
    ○ PNI
    • Add concurrent cisplatin (CRT): positive margins and ENE
      ○ Improved LRC and DFS. No difference in OS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Describe management options for oral lip and hard palate cancers.

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is the management for stage III/IV oral cavity cancers?

A
  • Surgery to the primary forms the basis of management
    ○ Aim for macroscopic margin of 10mm (for path margin of 5mm)
    § In the case of positive margins, re-resection should be attempted
    ○ Always consider the functional deficit with surgery
    • Definitive radiotherapy should be considered if medically or technically inoperable
    • Adjuvant radiotherapy is typically required for all patients
      ○ Criteria remain as above
    • Elective nodal management is typically required for all patients
      ○ Bilateral nodal management is required if
      § Approaching midline (<10mm)
      § Multiple ipsilateral nodes
      § Ventral tongue or floor of mouth (always bilateral)
      ○ The cN0 contralateral neck can be managed with RT
      § Minimises toxicity compared with bilateral neck dissections
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Describe the types of mandibular resections

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is the evidence for use of radiotherapy in oral cavity cancers?

A

Retrospective NCDB study (Ellis, 2018)
○ 20779 patients with stage I-II oral cavity SCC were retrospectively reviewed
§ Note that 95% had surgical management
○ Outcomes
§ Primary radiotherapy was associated with increased risk of mortality (HR 1.97)
§ Probability of receiving RT was associated with
□ Age >70; black; Medicare only (no insurance); clinical stage II; low-volume treatment centre
□ HIGHLY BIASED DATA

Retrospective single-institution review (Hinerman, 2004)
	○ 226 patients treated with post-operative RT for oral cavity SCC
		§ More than half were T3-T4
		§ More than half were cN positive
	○ Outcomes
		§ Variable impacting locoregional control include
			□ T-stage and N-stage
			□ ENE
			□ Surgical margins
			□ PNI and LVI
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is your definitive radiotherapy technique for oral cavity tumours?

A

Patients

1) Stage I-II (no chemotherapy)
1. Consider brachytherapy boost
2) Stage III-IV (concurrent chemotherapy)

Pre-simulation

MDT Discussion (review path)
Dietician + speech path involvement
- Consider prophylactic PEG
OPG + Dental review
Audiometry
Smoking cessation

Simulation

Supine with thermoplastic mask
- Handgrips
- Vacbag and ankle/knee-blocks

Wire any surgical scars

CT (2mm with IV contrast)
- Vertex to mid chest

Fusion

PET-CT and MR fusion

Dose prescription

SIB technique
- 70Gy/35F to the involved region
- 63Gy/35F to the high-risk region
- 56Gy/35F to the elective nodal regions

Concurrent chemotherapy (if stage III-IV)
- Cisplatin 100mg/m2 q3wk
- Cisplatin 40mg/m2 q1wk

VMAT technique

10 days per fortnight
- Accelerate to 6F per week if no chemotherapy

Volumes

GTVp = macroscopic primary disease
GTVn = macroscopic nodal disease

CTV70
- (GTVp + GTVn) + 5mm
CTV63
- GTVp + 10mm
- If BOT or soft palate, include entire organ
CTV56
- Elective nodal regions

PTV
- CTV + 5mm

Nodal regions

- Highest risk = levels I-III
- If oral tongue = include level IV
- If bulky level II = include VII
- If multiple nodes = include contralateral neck

- Bilateral inclusion dependent on location
	○ FOM and ventral tongue
	○ <1cm from midline

Target Verification

Daily CBCT

OARs

Brainstem + Optic Nerves/Chiasm
- Dmax < 54Gy

Cochlear
- Mean < 45Gy

Lacrimal Gland
- Dmax < 30Gy

Eye
- Mean < 35Gy
- Dmax < 50Gy

Lens
- Dmax < 7Gy

Parotid (spare 1 gland)
- Mean < 26Gy (optimal)

Oral cavity
- Mean < 30Gy

Mandible
- Dmax < 70Gy

Larynx
- Mean < 45Gy

Oesophagus
- Mean < 35Gy

Pharyngeal constrictors
- Mean < 50Gy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is your adjuvant radiotherapy technique for oral cavity tumours?

A

Adjuvant Therapy
Patients 1) Intermediate risk (RT alone)
* Close margins
* T3/T4 disease
* 2 or more LN involved (i.e. N2+ disease)
* PNI/LVI
* DOI >4mm

2) High Risk (concurrent chemotherapy)
        * Positive margins
        * ECE

Pre-simulation MDT Discussion (review path)
Dietician + speech path involvement
- Consider prophylactic PEG
OPG + Dental review
Audiometry
Smoking cessation
Simulation Supine with thermoplastic mask
- Handgrips
- Vacbag and ankle/knee-blocks

Consider intra-oral appliances
        - Bite-block (immobilise or displace tongue)
        - Wax buccal spacer (for buccal primary)
Wire any surgical scars

CT (2mm with IV contrast)
        - Vertex to mid chest

Fusion Pre-op PET-CT and MR fusion

Dose prescription
SIB technique

        * 60Gy/30F to tumour bed and surgical bed
                ○ If gross macro residual --> Treat as Definitive [alternatively boost to 66Gy]
                ○ If pos margin or close margin --> 63Gy
        * 54Gy to elective nodal regions


VMAT technique

10 days per fortnight

Concurrent chemotherapy (if positive margin or ECE)
        - Cisplatin 100mg/m2 q3wk
        - Cisplatin 40mg/m2 q1wk

Volumes Pre-op GTVp = based on original MR
GTVp = residual macroscopic disease

CTV63 = pre-op GTVp + 5mm
        - If gross residual --> CTV66 = GTVp + 5mm
CTV60= Tumour bed (GTVp+10mm )
        Entire surgical bed
        GTVn+10mm

CTV54 = elective nodal regions
        - Generally I, II, III
        - Consider level IV
        - Consider 5a/5b for N2, 7b if bulky II

PTV = CTV + 5mm Target Verification	Daily CBCT

OARs Brainstem + Optic Nerves/Chiasm
- Dmax < 54Gy

Cochlear
        - Mean < 45Gy

Lacrimal Gland
        - Dmax < 30Gy
        
Eye
        - Mean < 35Gy
        - Dmax < 50Gy
        
Lens
        - Dmax < 7Gy

Parotid (spare 1 gland)
        - Mean < 26Gy (optimal)

Oral cavity
        - Mean < 30Gy
        
Mandible
        - Dmax < 70Gy

Larynx
        - Mean < 45Gy

Oesophagus
        - Mean < 35Gy

Pharyngeal constrictors
        - Mean < 50Gy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is your follow up for oral cavity cancers. What is the expected 5yr OS (by stage) and 2 year LC (by T stage) for oral cavity cancers

A
  • Clinical review every two months for the first two years (shared care)
    ○ Clinical examination
    ○ Consider CT NCAP once at 12mo
    • Clinical review every three months for years 3-5
      ○ Clinical examination
    • Annual surveillance thereafter for late effects
    • Single FDG-PET at 12 weeks post RT
    • Consider TFTs if neck irradiated
    • Regular dental review (every six months)
    • Supportive care
      ○ Smoking cessation
      ○ Dietician and speech path review PRN
      ○ Lymphoedema review PRN
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What is the evidence for 6# a week in head and neck RT?

A

Summary:
- Altered fractionation improves oncological outcomes
○ Most benefit found with larynx SCC
- Increase in early toxicity, but this appears tolerable
Concurrent chemotherapy is superior to altered fractionation

DAHANCA 6 & 7 (Overgaard, 2003)
○ 1476 patients with H+N SCC were randomised to 5F vs 6F per week
§ DAHANCA 6 = glottic SCC
§ DAHANCA 7 = all other SCC
○ Up to 68Gy in 2Gy/F was administered (without chemotherapy)
§ Nimorazole was randomized in DAHANCA 7 only
○ Outcomes
§ Acceleration improved LRC (HR 0.66) and CSS (HR 0.71)
□ Larynx SCC benefited most
§ No improvement in OS (HR 0.98)
○ Significant increase in early toxicity in the accelerated arm
§ Confluent mucositis = 55% vs 33%
○ No difference in late toxicity between arms

IAEA-ACC study (Overgaard, 2010)
	○ 908 patients with H&N SCC were randomised to 5F vs 6F per week
		§ 66-70Gy/33-35F without systemic therapy
	○ Outcomes
		§ 6F per week was associated with improved LRC (42% vs 30%)
			□ Biggest benefit gained for larynx SCC
		§ Only a borderline OS benefit was seen (HR 0.70; p=0.07)
		§ Significant increase in early skin and mucositis toxicity
			□ No difference in late toxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What is the evidence for chemotherapy with H&N RT

A

MACH-HN (Lacas, 2017)
○ 107 trials including 19805 patients were included in the analysis
§ Trials investigated the integration of chemotherapy into H+N SCC management
○ Outcomes
§ Concurrent chemotherapy
□ Associated with improved OS (HR 0.83), CSS (HR 0.79) and LRC (HR 0.71)
□ Absolute OS benefit is 6.5% at 5 years
§ Induction chemotherapy
□ Benefit seen for distant failure (HR 0.76)
□ No benefit seen with respect to LRC, CSS or OS
§ Adjuvant chemotherapy
□ No benefit seen with respect to OS
§ Concurrent chemotherapy is superior to induction chemotherapy (OS = HR 0.84)
○ Age is an important factor
§ Decreased impact of concurrent chemotherapy with increasing age
Marked increase in non-cancer death when age > 70 years

MARCH meta-analysis (Lacas, 2017)
	○ 34 trials including 11969 patients were included
		§ SCC of the oral cavity, oropharynx, hypopharynx and larynx
		§ Trials investigated altered fractionations (acceleration or hyperfractionation)
	○ Outcomes
		§ Altered fractionation was associated with improved OS compared with RT alone (HR 0.94; 3.1% at 5 years)
			□ OS benefit isolated to the hyperfractionated group
			□ Acceleration associated with improvement in CSS and LRC Altered fractionation resulted in worse OS compared with chemoradiotherapy (HR 1.22)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What is the evidence for Cetuximab vs cisplatin in H&N ChemoRT?

A

Summary
- Cetuximab initially proposed as a form of treatment de-escalation in the setting of reducing cisplatin toxicity
- Cetuximab is inferior to cisplatin as a concurrent agent (In terms of Tumour control, OS, PFS)
However, there is efficacy and it can be considered in patients where cisplatin is contraindicated

- Randomised Bonner trial (Bonner, 2006)
	○ 424 patients with stage III-IV H+N SCC were randomised to
		§ 70Gy with cetuximab vs placebo
	○ Outcomes
		§ Improved OS with cetuximab (HR 0.03)
		§ Improved LRC with cetuximab (HR 0.68)
	○ Increase in acneiform rash and infusional reactions seen
		§ All other toxicity unchanged
		
- ARTSCAN III trial (Gebre-Medhin, 2021)
	○ 298 patients with stage III-IV H+N SCC were randomised to
		§ 68-73Gy of RT with either weekly cisplatin vs cetuximab
	○ Outcomes
		§ Trial ceased early due to safety concerns
		§ Cisplatin resulted in improved OS at 3 years (88% vs 78%)
		§ Cisplatin resulted in improved LRF rates at 3 years (23% vs 9%)
	○ Cetuximab is inferior to cisplatin

- RTOG 1016 (2020) and DeESCALTE (2018) trials 
	○ RTOG cetuximab inferior for OS and PFS compared with cisplatin + RT for HPV positive OPSCC DeEcalate cetuximab no benefit in terms of reduced toxicity, but detriment in terms of tumour control
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What is the evidence for adjuvant RT in H&N treatment?

A

Summary:
- Adjuvant radiotherapy at doses of 60Gy effectively control intermediate risk factors
○ No dose response relationship is seen
- Positive margins and ECE are independently associated with worse prognosis
○ Proven long-term benefit with concurrent chemotherapy

* Peters (IJROBP 1993)
○ RCT from MDACC
○ 302 pts randomised to dose of PORT
○ Found that pts with high risk for LR recurrence had either ECE or 2 of: oral cavity, close/+ve margin, 2+ LNs, LN >3cm (N2+), PNI (or treatment delay >6 weeks)
* Ang (IJROBP 2001)
○ Prospective trial validating risk factors and need for PORT
* Low risk patients (no RFs)- 5yr LRC 90% without RT
* Intermediate risk patients (1 RF, not ECE) -5yr LRC 94% with PORT, 57.6Gy.
▪ As pts with intermediate risk did as well as patients with low risk if got PORT- validates the use of PORT
* High risk pts (ECE or 2+ of others) -> 5yr LRC 68% with 63Gy PORT
○ Indications for primary and nodal PORT should be considered separately on their merits
* Interpretation:
○ Peters detected certain factors predicting for worse LRC. These factors were validated by Ang, who found that patients with intermediate risk had same level of LRC as low risk patients if had PORT, hence this trial validates the risk factors and shows PORT decreases LR risk

MDACC Adjuvant Radiotherapy Trial (Rosenthal, 2017)
○ 301 patients with H+N SCC of any primary site and any stage underwent surgical management
§ Randomised on the basis of risk factors
□ Primary = T-stage, margins, PNI
□ Nodes = number of nodes, size and ECE
§ Randomised to one of three dose levels for adjuvant RT
□ 57.6Gy/32F
□ 63Gy/35F
□ 68.4Gy/38F
○ Outcomes
§ No dose response seen between cohorts (i.e. above 57.6Gy)
§ Positive margins and ECE were the only major high-risk factors associated with prognosis
□ Intermediate risk factors were not associated with increase in LRC (i.e. adjuvant RT was effective for these)
§ Overall treatment time was associated with prognosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is the evidence for adjuvant ChemoRT in H&N Treatment.

A

2x RCT Improved LRC and DFS. No difference in OS

RTOG 9501 trial (Cooper, 2012)
	○ 410 patients with locally advanced H+N SCC underwent surgical resection and had high-risk features
		§ High-risk = positive margins, ECE or number of LN (>2)
		§ Randomised to adjuvant RT 60Gy +/- cisplatin chemotherapy
	○ Outcomes
		§ At long-term follow-up for the overall cohort, no persistent LRC benefit seen with cisplatin (HR 0.73)
		§ For the "ECE or positive margin" group, there is persistent benefit
			□ LRC (HR 0.56; p=0.02)
			□ OS (HR 0.76; p=0.07)
		§ There is no benefit by number of LN involved

EORTC 22931 (Bernier, NEJM 2004)
	High risk: N2-3, positive margin, PNI, vascular tumour embolism, involved nodes at level IV/V from oral cavity/oropharynx primary
	RT +- 3 weekly cisplatin
	
	Improved LRC and DFS. No difference in OS
	Worse acute gr 3-4 toxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What is the evidence for definitive ChemoRT vs RT alone?

A

Intergroup trial RCT –stopped early. CRT superior to RT alone for LRC and OS

* Intergroup trial  (Aldelstein JCO 2003)
	○ Stopped due to slow accrual, 295/463 pts, Stage III-IV OC, OPx, Larynx, HypoPx
	○ RT alone 70Gy/35F vs CRT 70Gy/35F weekly Cis vs Split course CRT (30Gy, break, 30-40Gy) with 3 weekly Cis+5FU.
	○ CRT is superior to RT alone, but benefit is mitigated with split course.
	○ 3yr DSS - CRT 51%, RT alone 33%, Split CRT 41%
	○ 3yr OS - CRT 37%, RT alone 23%, Split CRT 27%
	 
		* GORTEC trial (Calais JNCI 1999)
			○ RT 70Gy/35F +/- Chemo (3 weekly carbo/5FU) CRT better than RT alone in 5yr LRC ( 25-> 48%), DSS (17-> 27%) and OS (16 -> 22%)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is the epidemiology for Oropharyngeal Ca?

A

Incidence (Australian statistics)
- ALL = 4500 cases annually
- OROPHARYNX = 1500 cases annually
- Approx 4 per 100000 people

HPV considered distinct clinical disease w regard to risk factor profiles, treatment response and prognosis
- Incidence of HPV positive OPSCC rising
○ Frequently male, young (<50) and good performance status
○ More likely to present in the tonsils, PD, early T stage and advanced N stage
○ Better cure rates then alcohol or tobaccco OPSCC
- Incidence of HPV negative OPSCC decreasing

Rising incidence of young people < 50yo (both males and females)
- Likely HPV related

Median age is 60 years
- Younger for HPV positive
Male predominance (up to 4:1)
- Approaches parity in HPV positive

Aetiology

1) Smoking (RR is up to 10x), both HPV and nonHPV
	a. ALSO - cigar use, chewing tobacco, betel nut
2) Alcohol use (RR is up to 5x)
	a. Synergistic with smoking
3) Viral (HPV)
	a. HPV is implicated in up to 60-80% of cases
	b. Behavioural risk factors --> number of sexual partners, oral sex, age at first intercourse
4) Occupational exposure
	a. Asbestos
	b. Mustard Gas
	c. Nickel
	d. Tar
5) Immunosuppression
6) Nutritional deficits
	a. Fruits and vegetables are protective
7) Poor oral hygiene & periodontal disease
8) Previous radiation
9) Genetic factors
	a. Fanconi anaemia

Metachronous lesions
- Second H+N primary = 20% risk
- Lung = 5-10%
- Oesophagus = <5%

HPV-related cancers have a lower risk of metachronous lesions
- Lack of field effect change
- Other RF:
○ Number of sex partners
○ Not alcohol
○ No precancerous lesions
○ More nonkeratinising

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Describe SCC HPV mediated carcinogenesis.

A

HPV-mediated carcinogenesis

HPV has high-risk and low-risk subtypes
* HPV 16, 18, 31, 33 are high-risk
* Remainder are low risk (e.g. 6, 11)

HPV replicates by entering a cell and incorporating its own DNA into the cellular DNA. In this way, the cellular machinery is manipulated to create many copies of the virus, which is then released for further spread and infection.

The E6 and E7 oncoproteins of the high-risk HPV subtypes manipulate cellular machinery in such a way to cause carcinogenesis
* E6 associates with the p53 protein and leads to the ubiquitination, thereby inhibiting its function. E6 also causes other more minor protease-mediated cellular damage
* E7 associates with the Rb (retinoblastoma) protein and causes its proteasome-mediated degradation.
E5 is also oncogenic, but less potent
* E5 can activate EGFR signalling and thus promote cellular growth

This in turn, causes carcinogenesis in the following ways
* Cellular immortality (disabled apoptotic pathways)
○ E6 disables p53
* Genomic instability (impaired cellular DNA repair pathways)
○ Disabled apoptosis allows accumulation of genetic damage without an endpoint
* Unregulated cellular proliferation (removal of cell-cycle arrest mechanisms)
○ E6 disables p53
○ E7 disables Rb
* Sustained proliferative signalling
○ E5 can activate EGFR pathway
* Impaired immunity (ongoing infection and inflammation)
○ E6/7 inhibit interferon signalling (impair innate immunity)
○ E5 down-regulates MHC-I expression
○ This leads to persistence of infection (which is required for carcinogenesis –> accumulate genetic damage)
* Other
○ E7 induces angiogenesis
○ E7 induces Warburg effect (altered metabolism)
○ P16 induced but doesn’t slow cell cycle due to E7/RB

Can test for via P16 IHC –80% specific in oropharynx, DNA or RNA ISH
On cytology p16 maybe patchy (done on unknown primary)

Better prognosis, more treatment sensitive, lower mutation rates/genetic mutations.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Describe non-HPV related H&N SCC carcinogenesis.

A

Dysplastic spectrum within the epithelium –occurs on surface epithelium (rather than palatine/lingual tonsil crypts)
- Mild atypia
- High-grade dysplasia
- Carcinoma in-situ
- Invasive carcinoma

Driven by chronic inflammation or chronic carcinogen exposure
Note that withdrawal of smoking can result in regression of dysplasia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What are the subtypes for H&N SCC. Describe the Macroscopic and Microscopic features.

A

Squamous Cell Carcinoma
- By far, the most common histological subtype of oral cavity cancer

- Macroscopic
	○ Pink/grey area of mucosal thickening
		§ Ulcerated mass when late/advanced
	○ Verrucous and spindle cell variants may be more exophytic or polypoid in appearance
- Microscopic
	○ Nests of keratinising squamous cells which invade the basement membrane
		§ Keratin pearls and intercellular bridges
		§ Grade/differentiation is dependent on keratinising features
	○ Large polygonal cells with variable cellular anaplasia
	○ For nonkeratinising (HPV):
		§ Nodules/sheets of cells –often arising from crypts
		§ Focal areas of sqaumous/keratin <10%
		§ Comednecrosis frequent
		§ Permeated lymphocyes
		§ No grading for HPV, no insitu disease
- Variants
	○ Verrucous 
		§ White, warty, cauliflower like lesion
		§ Low metastatic potential
	○ Sarcomatoid/spindle cell 
		§ Biphasic mixed carcinoma with spindle component
		§ Aggressive disease course
	○ Basaloid (HPV associated)
		§ Small basaloid cells with peripheral palisading and high-grade features (high N:C ratio, pleomorphism, hyperchromatic nuclei, mitoses)
		§ Aggressive disease course (but not when HPV associated)
	○ Papillary
	○ Lymphoepithelial like –syncytial appearance –indistinct cell borders, plasma and lymphocytes+
- Immunohistochemistry
	○ POS = AE1/AE3, HMWCK, p40, p63
- Molecular
	○ P16 overexpression is used as a surrogate for HPV status
		§ Can be IHC or ISH
		§ There is still a reasonable false negative (20%) rate
	○ HPV PCR is the most accurate 
		§ Also gives information on the particular subtype present --> prognostic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What are the subsites of oropharynx cancers?

A
  • Tonsils
    • BOT
    • Soft palate
      • Upper lateral and posterior pharyngeal walls
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What are the prognostic factors for oropharyngeal cancers?

A

Patient Factors
- Age and performance status
- Nutritional status
- Immunosuppression
- Smoking (local control and toxicity)
- Anaemia
○ Non-reversible –> transfusion does not improve outcomes

Tumour Factors
- TNM stage
- HPV status (see risk groups below)

nonHPV
- Depth of Invasion (oral cavity)
○ Measured from basement to depth of tumour
- Grade (of limited value)
- LVI
- PNI
- ECE >2mm –often seen in LN >3cm

Treatment Factors
- High-volume experienced centre
- Overall treatment time (if chemoRT)
- Surgical margins
○ Addition of adjuvant radiotherapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

How would you work up a patient with an oropharyngeal lesion?

A

Consultation

- History
	○ HPI
		§ Dysphagia, aspiration or odynophagia
		§ Otalgia/ear effusion (Eustachian obstruction or referred pain from CN 9 + 10)
		§ Sensation of pharyngeal mass
		§ Palpable cervical node
	○ PMHx:
		§ Immunosuppression
		§ HPV infection
		§ Other metachronous H+N/lung malignancy
	○ Meds:
		§ Immunosuppressants
		§ Radiosensitisers
	○ Social
		§ Smoking
		§ EtOH
		§ Social supports
	○ Family History

- Examination
	○ General examination
		§ Fitness and performance status
		§ Nutrition
		§ Dental condition
	○ H+N examination
		§ Palpable lymphadenopathy
		§ Oral cavity examination (including trismus)
		§ Nasoendoscopy
			□ Examine primary site
			□ Examine remaining mucosa
			□ Examine cords

Work-Up

- Biopsy
	○ MUST include p16 staining
	○ Biopsy any equivocal neck nodes (unless already proceeding with neck dissection)

- MR Head and Neck
	○ Surgical evaluation

- FDG-PET
	○ Nodal and distant staging
	
- Diagnostic CT Head and Neck
	○ Evaluate bony invasion

- Pre-chemotherapy bloods
	○ FBC
	○ EUC, CMP
	○ LFT and coags
	○ HepB sAg, sAb, cAb
	○ Hep C serology
	○ HIV serology
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What is the staging for Oropharynx cancers?

A

FYI P16+ve staging

N-Staging
N1 Any ipsilateral nodes (<6cm)
N2 Any contralateral nodes (<6cm)
N3 Any node >6cm in size

M-Staging
M1 Distant metastasis

Stages
* Stage I
○ T1-2 and N0-1
* Stage II
○ T1-3 and N2

* Stage III
	○ Tany and N3
	○ T4 and Nany
* Stage IV
	○ M1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

In general how would you manage Oropharynx cancers?

A

General Principles

- Always aim to use as few modalities as is feasible (reduce morbidity)

- In general, options may include
	○ Early Stage (stages I-II)
		§ Surgical resection alone
		§ Radiotherapy alone
	○ Advanced Stage (stages III-IV)
		§ Chemoradiotherapy

- Always establish relevant supportive care early
	○ Smoking cessation
	○ Dietician and speech pathology input
		§ Consideration of prophylactic gastrostomy
	○ Dental input
		§ Pre-RT OPG
	○ Social & psychological supports as appropriate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What would be your management for Stage I-II Oropharynx cancers?

A
  • Definitive options include
    1) Definitive radiotherapy
    § 70Gy/35F alone (no concurrent chemotherapy)
    § Aim to accelerate where feasible (6 fractions per week)
    § Ipsilateral RT if tonsil (less than 1cm extention to soft palate/BOT and N0-1 (single <3cm)
    2) Surgical resection
    § TORS + neck dissection
    □ Aim for small functional deficit (T1-2 tonsillar fossa, lateral BOT, L pharyngeal wall)
    □ Contraindication: soft palate, posterior arch (affects speech), central BOT (remove less than half of tongue), trismus, fixed tumour, carotid invasion
    □ If TORS is not feasible, opt for radiation instead (i.e. do not jaw split)
    □ Still risk of needing adjuvant RT
    □ PH2 TORS (3mm margin) + 50Gy vs 60Gy similar
    § Ipsilateral neck dissection is reasonable if well lateralised primary (e.g. tonsil)
    □ Bilateral neck dissection if BOT, soft palate or posterior wall primary
    • In the absence of high-level evidence, decision making is based on functional outcome and treatment morbidity
    • Consider adjuvant post-operative radiotherapy alone if (INTERMEDIATE RISK):
      ○ Close/positive margins
      ○ ENE on histopathology -hard to predict on MRI/CT due to inflammatory reaction
      ○ T3/T4 disease
      ○ 2+ nodes involved (N2+ disease)
      ○ LVI
      ○ PNI
    • Only positive margins and ENE justify adjuvant concurrent chemoradiotherapy (HIGH RISK)
    • There is no role for adjuvant chemotherapy
      ○ MACH meta-analysis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What would be your management for Stage III-IV Oropharynx cancers?

A
  • General approach
    ○ Aim for single modality therapy to minimise toxicity
    ○ In this way, TORS has a limited role
    § If adjuvant radiotherapy is likely, prefer to proceed with definitive chemoradiotherapy
    ○ Extensive surgery (including mandibulectomy) should be performed if gross bony invasion is present
    § Adjuvant RT will always follow
    • Standard therapy is concurrent chemoradiotherapy (OS and LRC better than RT alone)
      ○ 70Gy/35F (5F per week)
      ○ High dose Cisplatin 100mg/m2 every 3 weeks
      § Alternative is weekly cisplatin at 40mg/m2
      § Carbo/5FU
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

How would management change for HPV related oropharyngeal cancers?

A
  • In general, the approach is very similar to the HPV-negative cohort
    • Key differences
      ○ T1-2 and N1 (single node < 3cm) can be considered for single agent RT
      § Only if low-risk patients (i.e. non-smokers)
    • Unknown primary node positive
      ○ PET will detect 25-50%, most common tongue base or tonsil
      ○ Unilateral tonsillectomy (lateral oropharyngectomy) and lingual tonsil (no tongue muscle)
    • Treatment de-intensification should not be considered routine at this time
      ○ Data are inconclusive and immature
      ○ Trials:
      § Surgical resection
      § Reduce radiation dose or omit chemotherapy
      □ Reduced RT dose 50 vs 60Gy –similar outcomes
      □ Omit chemotheray = higher locoregional failure
      □ Alternative chemo: cetuximab worse survival
      Induction chemo then deescalate responders
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What is the evidence for oropharynx cancer management with RT vs Surgery?

A

ORATOR (Nichols, 2022)
○ 68 patients with T1-2 N0-2 SCC of the oropharynx were randomised to
§ Definitive radiotherapy (concurrent chemotherapy if N1-2)
§ TORS with neck dissection (adjuvant RT as indicated)
○ Outcomes
§ RT had improved dysphagia scores (MD Anderson Dysphagia Inventory)
□ Improvement at 1 year (uncertain clinical significance of small change)
□ Declining absolute difference at years 2 and 3
§ TORS had worse pain and dental concerns in the first year
□ Resolved by year 2-3
§ Increased xerostomia in the RT arm

MA—similar DSS and OR w/ TORS vs. RT for early-stage tumour. 
	○ PORT indicated in ~70% and post-op chemoRT in ~30%. 
	○ RT tox: oseophageal stenosis, ORN, gastrostomy tube. 
	○ TORS tox: haemorr, fistula, 12% needs trachy but mostly decannulated prior to DC.
MA 2002 Parsons—51studies OPX tx w/ surg vs RT. Similar 5yr LC, LRC, OS and CSS. However,  Poor functional outcomes/complications w/ surg, Thus, Def CRT was adopted as SOC.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What is your radiotherapy technique for definitive and adjuvant oropharynx cancers?

A

Adjuvant Therapy
1) Stage I-II (no chemotherapy)
2) Stage III-IV (concurrent chemotherapy)

Definitive Therapy
1) Intermediate risk (RT alone)
* Close margins
* T3/T4 disease
* 2 or more LN involved (i.e. N2+ disease)
* PNI/LVI
2) High Risk (concurrent chemotherapy)
* Positive margins
* ECE

MDT Discussion (review path)
Dietician + speech path involvement
- Consider prophylactic PEG
OPG + Dental review
Audiometry
Smoking cessation

Supine with thermoplastic mask
- Handgrips
- Vacbag and ankle/knee-blocks
Wire any surgical scars
CT (2mm with IV contrast)
- Vertex to mid chest
Supine with thermoplastic mask
- Handgrips
- Vacbag and ankle/knee-blocks
Wire any surgical scars
CT (2mm with IV contrast)
- Vertex to mid chest
PET-CT and MR fusion
Pre-op PET-CT and MR fusion

Target Verification
- Daily CBCT

OARs
Brainstem + Optic Nerves/Chiasm
- Dmax < 54Gy
Cochlear
- Mean < 45Gy
Lacrimal Gland
- Dmax < 30Gy
Eye
- Mean < 35Gy
- Dmax < 50Gy
Lens
- Dmax < 7Gy

Parotid (spare 1 gland)
- Mean < 26Gy (optimal)
Oral cavity
- Mean < 30Gy

Mandible
- Dmax < 70Gy
Larynx
- Mean < 45Gy
Oesophagus
- Mean < 35Gy
Pharyngeal constrictors
- Mean < 50Gy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What is the elective nodal volumes for p16-ve oropharynx cancers?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What is the prognosis and follow up for oropharynx cancers?

A

Follow-Up

- Clinical review every two months for the first two years (shared care)
	○ Clinical examination
	○ Consider CT NCAP once at 12mo
- Clinical review every three months for years 3-5
	○ Clinical examination
- Annual surveillance thereafter for late effects

- Single FDG-PET at 12 weeks post RT
	○ SUV decrease by 50% = 50% predictor for complete response, especially SUV<2.5
		§ Negative predictive value 92-97%
	○ HPV nodes take longer to go. 
		§ Mild uptake -repeat PET
		§ Enlarged but no activity -clinical followup
	○ Also surgery relatively easy -less fibrosis than later

- Consider TFTs if neck irradiated
- Regular dental review (every six months)
- Supportive care
	○ Smoking cessation
	○ Dietician and speech path review PRN
	○ Lymphoedema review PRN

Ongoing research in to PET at 2-3 weeks during RT -less tumour uptake is reponse. Before toxicity/inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What is the epidemiology and risk factors for Larynx and hypopharynx cancers?

A

Incidence (Australian statistics)
- LARYNX = 650 cases annually
- Approx 2 per 100000 people

Median age is 65 years
Male predominance (up to 4:1)

Aetiology

1) Smoking (RR is up to 10x)
2) Alcohol use (RR is up to 5x)
	a. Synergistic with smoking
3) Occupational exposure
	a. Asbestos
	b. Mustard Gas
	c. Nickel
	d. Tar
4) GORD
	a. Presume chronic inflammation
5) Viral
	a. HPV is implicated (less strongly than for oropharynx)
6) Immunosuppression
7) Nutritional deficits
	a. Fruits and vegetables are protective
8) Previous radiation
9) Genetic factors
	a. Fanconi anaemia

Metachronous lesions
- Second H+N primary = 20% risk
- Lung = 5-10%
- Oesophagus = <5%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

List the subsites of the larynx and hypopharynx and their respective LN Drainage

A

Supra-glottis: AVISA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What are the prognostic factors for Larynx and Hypopharynx cancers?

A

Patient Factors
- Age and performance status
- Functional larynx
- Nutritional status
- Immunosuppression
- Smoking (local control and toxicity)
- Anaemia
○ Non-reversible –> transfusion does not improve outcomes

Tumour Factors
- TNM stage
- Tumour site
○ Hypopharynx is worse than larynx
- Grade (of limited value)
- LVI
- PNI
- ECE

Treatment Factors
- High-volume experienced centre
- Surgical margins
Overall treatment time (if chemoradiotherapy)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

How would you work up a patient with a larynx or hypopharynx cancer?

A
  • History
    ○ HPI
    § Dysphonia or impaired cough
    § Dysphagia, aspiration or odynophagia
    § Otalgia/ear effusion (Eustachian obstruction or referred pain from CN 9 + 10)
    § Sensation of pharyngeal mass
    § Palpable cervical node
    ○ PMHx:
    § Immunosuppression
    § Other metachronous H+N/lung malignancy
    ○ Meds:
    § Immunosuppressants
    § Radiosensitisers
    ○ Social
    § Smoking
    § EtOH
    § Social supports
    ○ Family History
    • Examination
      ○ General examination
      § Fitness and performance status
      § Nutrition
      § Dental condition
      ○ H+N examination
      § Palpable lymphadenopathy
      § Oral cavity examination (including trismus)
      § Nasoendoscopy
      □ Examine primary site
      □ Examine remaining mucosa
      □ Examine cords

Work-Up

- Biopsy
	○ EUA + microlaryngoscopy
	○ Biopsy any equivocal neck nodes (unless already proceeding with neck dissection)

- MR Head and Neck
	○ Surgical evaluation

- FDG-PET
	○ Nodal and distant staging
	
- Diagnostic CT Head and Neck
	○ Evaluate bony invasion

- Pre-chemotherapy bloods
	○ FBC
	○ EUC, CMP
	○ LFT and coags
	○ HepB sAg, sAb, cAb
	○ Hep C serology
	○ HIV serology
- Dental Ax + OPG; dietician and SP review; prophylactic enteral feeding.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What is the staging for Larynx cancers?

A

Stages
* Stage I
○ T1N0
* Stage II
○ T2N0

* Stage III
	○ T3 OR N1
* Stage IV T4+ OR N2+
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What is the staging for Hypopharynx cancers?

A

Subsites:
Postcricoid, hypopharyngeal wall, pyriform sinus

Stages
* Stage I
○ T1N0
* Stage II
○ T2N0

* Stage III
	○ T3 OR N1
* Stage IV
	○ T4+ OR N2+
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

How would you manage a stage I-II supraglottic cancer?

A

§ Either RT or laser surgery
All patients need elective bilateral nodal therapy (either surgical or RT)

  • prospective data only no RCT for Sx Vs. RTx
    Supraglottis
    - Rich lymphatics: T1-2 (30-40% N+) and T3-4 (55-65% N+)
    1. Definitive RTx alone to 70/56Gy primary + bilateral neck nodes (preferred dt voice preservation, less morbid, surgical salvage option of recurrence).
    ○ LC T1 90%, T2 80-90%
    2. Surgery - Laser excision or Supraglottic laryngectomy + bilateral ELND (retention of vocal and swallowing function, but high aspiration risk)- not suitable if poor lung function
    ○ LC T1 100%, T2 81%
    ○ Alternative - Total laryngectomy + bilateral ELND
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

How would you manage a stage I-II Glottic cancer?

A

§ Either RT or laser surgery
No elective nodal therapy required

Glottis
- Definitive small field RT (Hypofn 2.25Gy/#) –> 63Gy/28F (T1) or 65.25Gy/29 (T2) ↑ LC (15%) vs. 2 Gy/F. NO neck ENI needed
○ LC: T1 90%, T2 75-80%.
○ Organ preservation - 80-90%. Surgery salvageable in 90-95%
○ LVH uses 60Gy/25F for T1
○ Stage carefully as T3 chemorads may do better
- Uk guideline rt 63gy/28, 50gy/16, 55/20
- Surgery- Laser excision - T1 or T2 non-bulky.
○ LC: same as RT
○ Contraindication for laser: deep pre-epiglottic space extension, infiltration of the cricoarytenoid joint leading to vocal cord fixation, paraglottic space encroachment, anterior commissure infiltration and inadequate exposure of the larynx.
○ Don’t disrupt vocal ligament (deeper layer of vocal cord) -will affect voice quality
○ Alternative- Partial/hemilaryngectomy or total laryngectomy (based on anatomic considerations, poor voice quality).
- Note:
○ Data suggest similar oncological and vocal preservation outcomes between options
○ Decision making is as per institutional experience and patient preference
Salvage laryngectomy is the ultimate treatment for failure after either approach

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What are the advantages and disadvantages of managing a T1-2 Glottic cancer with EBRT vs Laser resection vs Surgery?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

How would you manage a stage I-II Subglottic cancer?

A

Subglottis
○ Rare, no consensus
○ Definitive 70/56/35# RTx alone (preferred) to primary and bilateral neck nodes (lower neck and upper mediastinal nodes)
§ LC: 75%
§ 5yrs DFS Stage I. 70%, Stage II 40%
§ 5yrs OS Stage I 85%, Stage II 50%
Surgery - Total laryngectomy + bilateral ELND (usually Stage III-IV and usually presents with airway obstruction)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

How would you manage a stage I-II Hypopharynx cancer?

A
  • Rare for T1, as usually late presentation
    • Definitive options include
      1) Definitive radiotherapy
      § Due to limited organ-preserving surgical approaches, definitive radiotherapy is the standard of care. In the absence of high-level evidence, decision making is based on functional outcome and treatment morbidity
      § Definitive RTx alone to 70/56Gy primary + bilateral neck nodes- better functional outcomes.
      ○ LC: T1 89%, T2 77%, pyriform sinus worse
      § Aim to accelerate where feasible (6 fractions per week)
      2) Surgical resection (very limited role)
      § TORS + neck dissection
      ○ Partial or total pharyngolaryngectomy+ bilateral ELND
      ○ If TORS/minimally invasive surgery is not feasible, opt for radiation instead
      § All require bilateral neck dissection
    • Due to limited organ-preserving surgical approaches, definitive radiotherapy is the standard of care
      ○ In the absence of high-level evidence, decision making is based on functional outcome and treatment morbidity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

When would you consider adjuvant radiotherapy and adjuvant chemoradiotherapy in head and neck cancers?

A
  • Consider adjuvant post-operative radiotherapy alone if (INTERMEDIATE RISK):
    ○ Close/positive margins
    ○ ENE on histopathology
    ○ T3/T4 disease
    ○ 2+ nodes involved (N2+ disease)
    ○ LVI
    ○ PNI
    • Only positive margins and ENE justify adjuvant concurrent chemoradiotherapy (HIGH RISK)
    • There is no role for adjuvant chemotherapy
      ○ MACH meta-analysis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

How would you manage Stage III-IV Hypopharynx and Larynx cancers?

A
  • General approach
    ○ Aim for single modality therapy to minimise toxicity
    § In this way, TORS has a limited role
    § If adjuvant radiotherapy is likely, prefer to proceed with definitive chemoradiotherapy
    ○ Aim for organ-preserving therapy where feasible
    ○ Laryngectomy if poor voice, immobile cords, poor swallow/aspiration.
    • Concurrent chemoRTx 70Gy/35F (5F/week) w/ 3 weekly (100mg/m2, RCT) or weekly (40mg/m2) cisplatin (expert consensus EVIQ)- SOC with organ preservation
      ○ LC 50-60%, larynx preservation 60%
      ○ NOT IF T4b or extending into BOT (not eligible for RTOG 91-11)
      ○ <T4 pts 28% salvage laryngectomy Vs T4 pts 58% (SS –> VA larynx trial)
      ○ T4 disease:
      § 60% pf patients may require salvage laryngectomy due to poor post chemoRTx larynx function
      § 20-25% of patients will need long term PEG/tracheostomy due to treatment toxicity.
      ○ OS same at 10 yrs (as per RTOG trial, but better LC with concurrent treatment)
      ○ Concurrent ChemoRT RTOG 91-11 (concurrent just as good as induction chemo/sx + RTx or RTx alone)
      § If not well enough for CRTx, consider RTx only with altered fractionation
      ○ Only worthwhile if larynx is salvageable and likely to have adequate function after Rx
      § Mainly in regards to aspiration/swallowing/airway rather than voice quality
    • Standard therapy is concurrent chemoradiotherapy =50-60% local control, larynx preserved 60%, permanent PEG 25%
    • Laryngectomy + bilat LND should be considered if:
      ○ T4 disease
      ○ Non-functional larynx (trache dependent/PEG dependent/Recurrent aspiration)
      ○ Elderly unable to tolerate chemoradiotherapy
    • Adjuvant radiotherapy almost always follows laryngectomy
    • Consider stomal boost if:
      ○ Pre-operative tracheostomy (especially if emergent)
      ○ Locally advanced disease (involvement of anterior neck
      ○ Subglottic disease location
      ○ Positive surgical margins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What is the evidence for chemotherapy for head and neck cancer in elderly patients?

A
  • Elderly
    ○ MACH-HN meta-analysis provides some guidance for management
    § Concurrent chemoRT has limited benefit if > 70years
    § Likely to be harmful if >80 years (increase in non-cancer mortality)
    ○ Alternatives to cisplatin-based concurrent chemoradiotherapy
    § Sequential induction chemotherapy –> RT alone
    § Definitive RT alone
    § Concurrent carboplatin (single-agent) or cetuximab
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What is the evidence for radiotherapy in early stage larynx cancers?

A

Summary:
- Data are inconclusive and somewhat conflicting
○ Radiotherapy is likely an appropriate alternative to surgical management
§ Similar oncological outcomes
§ Similar or better vocal outcomes
○ Small chance of slightly inferior laryngeal preservation with RT

Seminal Russian RCT (Ogol'tsova, 1990)
	○ 269 patients with stage I-II laryngeal cancer were randomised to
		§ Surgery alone (partial laryngectomy)
		§ Radiotherapy alone
		§ Chemoradiotherapy
	○ Outcomes
		§ With respect to 5 year OS
			□ Surgery --> T1 = 100% & T2 = 97%
			□ RT alone --> T1 = 92% & T2 = 89%
		§ No benefit to chemoradiotherapy

Meta-analyses (multiple)
	○ T1 glottic SCC (Mo, 2017)
		§ Laser surgery improves laryngeal preservation and OS
		§ No difference in LRC
	○ T1 glottic SCC (Yoo, 2014)
		§ No difference in LRC or OS
		§ Suggestion of superior vocal outcomes with RT
	○ Tis/T1a glottic SCC (Guimaraes, 2018)
		§ Laser surgery improves laryngeal preservation and OS
		§ No difference in LRC
		§ RT delivers possible benefit with respect to vocal quality 
	○ T2 glottic SCC (Warner, 2017)
		§ No difference in 5yr OS between groups
		§ Increase in local failure with T2b (irrespective of cohort)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What is the evidence for hypofractionation in early glottic SCC

A

Osaka Yamazaki 2006 (IJROBP)
○ 180pt, T1 RCT. 60-66Gy/30-33F vs 56.25Gy/25F (<2/3 of VC) or 63Gy/28F (>2/3 of VC)
○ 5yr LC better in hypofractioanted arm 92% vs 77%
○ 5yr CSS 97 vs 100% (NS)

KROG 0201
	○ 156pts,  60-66Gy/30-33F vs 63Gy/28F for T1 and 70Gy/35F vs 67.5/30F for T2
	○ 5yr LPFS was better for the hypofractionated arm ( 77.8% for CR arm vs 88.5% in HP arm), No significant difference in toxicity profile. 

- Mendenhall- U Florida Lit Review- hypoFx provide Comparable LC, voice preservation, LC and OS compared to pe with transoral last excision, open partial layrngectomy, and RT.
	○ pt with anterior comminsure involvement may have a higher risk of LC recurrence after partial laryngectomy but doesnt no influence LC after RT

- 2017 NCD analysis showed that HypoFn (2.25Gy/F to 63-65.25Gy) in T1-T2 N0 glottic ca, improve OS when compared to conventional Fx
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What is the evidence for Stage III/IV larynx cancer chemoradiotherapy?

A

Summary:
- Chemoradiotherapy is a reasonable approach for T1-T3 disease
○ No OS detriment compared with laryngectomy+Adj RT, but better LC with ChemoRT
○ Caution should be used for T4 disease (not included in major RTOG 9111 trial)
- Larynx preservation rate is approximately 60%
- May opt for surgery if elderly, poor baseline larynx function (trache dependent/PEG dependent/Recurrent aspiration) and cannot cope with chemoRTx.

VA trial (Wolf, 1991)
	○ 332 patients with stage III or IV SCC of the larynx were prospectively randomised to
		§ Arm 1: Induction chemotherapy (3x cisplatin + 5-FU) followed by RT (66-76Gy), at risk areas at least 50-50.4Gy (all at 1.8-2Gy/#)
			□ ±salvage sx if no response/recur vs sx + XRT
		§ Arm 2: Laryngectomy with adjuvant RT
	○ Outcomes
		§ After 2c response rate: CR = 31%, PR = 54%
		§ No difference in OS between arms: 2yr OS: 68% both grps
		§ RFS: CRT grp: ↑ local recurrence (12% vs 2%) but ↓ distant mets, 36% req total laryngect (64% did NOT req laryngect!)
		§ Larynx preservation in 64% of patient

RTOG 9111 trial (Forastiere, 2013)
	○ 547 patients with stage III or IV SCC of the larynx were randomised to
		§ Radiotherapy (70Gy/35F)
		§ Concurrent chemoradiotherapy (cisplatin q3 weekly)
		§ Induction chemotherapy (cisplatin/5-FU) followed by radiotherapy
	○ Note that T4 patients were excluded
	○ Outcomes
	○ 5yr results: signif improve in LFS for both I+RT & CRT; for laryngeal preservation & LRC, CRT is still superior.
	○ Both chemotherapy arms improved OS compared with RT alone (HR 0.75 and 0.78)
		§ No significant difference between chemotherapy arms
	○ Concurrent chemotherapy improved larynx preservation rates (HR 0.58)
		§ 5 year larynx preservation rate of 45%
	○ Better locoregional control but same overall survival. ?laryngeal function, silent aspiration
	○ 10yr results
		§ Both chemo regiments incr LFS: HR = 0.75
		§ No diff OS
		§ Concomitant cisplatin + XRT: incr larynx preserve rate over induction (HR = 0.58) & RT alone
		§ No diff late effects – but deaths d/t other causes hgher w/ CRT (31%) vs chemo  RT (20%), RT (17%)

EORTC 24891 trial (Lefebvre, 2012)
	○ 194 patients with stage III-IV SCC or the hypopharynx were randomised to
		§ Induction chemotherapy (cisplatin/5-FU) followed by radiotherapy 70Gy/35F
		§ Laryngectomy + partial pharyngectomy with adjuvant radiotherapy
	○ Outcomes
		§ No difference in OS or failure patterns
		§ Larynx preservation in survivors is seen in 60% of patients
	○ Toxicity 15-20% g3 laryngeal tox. 5-10% feeding tube
66
Q

What would be your radiotherapy technique for Hypofractionated stage 1 glottic larynx SCC?

A

Definitive Radiotherapy (hypofractionated)

Patients
1) Stage I-II glottic larynx SCC

Pre-simulation
MDT Discussion (review path)
Dietician + speech path involvement
- Consider prophylactic PEG
OPG + Dental review
Audiometry
Smoking cessation
Thorough endoscopy to ensure lack of extension to lymphatic rich areas

Simulation
Supine with thermoplastic mask
- Head slightly extended
- Handgrips
- Vacbag and ankle/knee-blocks
Consider bolus (especially if anterior commissure)
CT (2mm without contrast)
- Vertex to mid chest

Fusion
N/A

Dose prescription
Hypofractionated technique
- T1N0 = 60Gy/25F
- T2N0 = 65.25Gy/29F
VMAT technique
10 days per fortnight

Vs fields: less carotid dose
Control same,

Volumes
GTV
- Gross disease
CTV
- Entire larynx at level of GTV
- Extend superior and inferior by 10mm
PTV
- CTV + 5mm

No nodes for t1, t2 glottis <5% nodal risk
Supraglottis includes nodes bilateral

Traditional Fields 4-6cm box, opposed lateral with wedges with bolus if ant commisure

- Ant = >10mm overshoot
- Post = at least at anterior vertebral edge
- Sup = Superior edge of thyroid cartilage
- Inferior = Inferior border of cricoid

SIngle vocal cord field: reduced arytenoid dose for vocal function. Reduced carotid dose

Target Verification
Daily CBCT

OARs
Spinal Cord
- Dmax < 45Gy
Carotid -alarm
Oesophagus
- Mean < 35Gy
Pharyngeal constrictors
- Mean < 50Gy

67
Q

What would be your radiotherapy technique for Definitive Stage I-IV larynx/hypopharynx cancer (Excluding glottic)

A

Definitive Radiotherapy

Patients
1) Stage I-II hypopharynx (no chemotherapy)
2) Stage I-II supra-/sub-glottic larynx (no chemotherapy)
3) Stage III-IV hypopharynx and larynx (chemotherapy)

Pre-simulation
MDT Discussion (review path)
Dietician + speech path involvement
- Consider prophylactic PEG
OPG + Dental review
Audiometry
Smoking cessation

Simulation
Supine with thermoplastic mask
- Handgrips
- Vacbag and ankle/knee-blocks
Wire any surgical scars
CT (2mm with IV contrast)
- Vertex to mid chest

Fusion
PET-CT and MR fusion

Dose prescription
SIB technique
- 70Gy/35F to the involved region
- 63Gy/35F to the high-risk region
- 56Gy/35F to the elective nodal regions

Concurrent chemotherapy (if stage III-IV)
- Cisplatin 100mg/m2 q3wk
- Cisplatin 40mg/m2 q1wk

VMAT technique
10 days per fortnight
- Accelerate to 6F per week if no chemotherapy

Volumes
GTVp = macroscopic primary disease
GTVn = macroscopic nodal disease

CTV70
- (GTVp + GTVn) + 5mm
CTV63
- GTVp + 10mm
- Include entire larynx/hypopharynx
CTV56
- Elective nodal regions
PTV
- CTV + 5mm

Nodal regions
- Highest risk = levels II-IV & VII bilaterally
- If LN pos = include levels Ib and V
- If bulky level II = include Ib and VII
- If piriform apex/oesophagus/subglottic = level VI

Target Verification
Daily CBCT

OARs
Brainstem + Optic Nerves/Chiasm
- Dmax < 54Gy
Cochlear
- Mean < 45Gy
Lacrimal Gland
- Dmax < 30Gy
Eye
- Mean < 35Gy
- Dmax < 50Gy
Lens
- Dmax < 7Gy
Parotid (spare 1 gland)
- Mean < 26Gy (optimal)
Oral cavity
- Mean < 30Gy
Mandible
- Dmax < 70Gy
Larynx
- Mean < 45Gy
Oesophagus
- Mean < 35Gy
Pharyngeal constrictors
- Mean < 50Gy

68
Q

What would be your radiotherapy technique for Adjuvant Intermediate risk and high risk larynx/hypopharynx cancer?

A

Adjuvant Therapy
Patients
1) Intermediate risk (RT alone)
* Close margins
* T3/T4 disease
* 2 or more LN involved (i.e. N2+ disease)
* PNI/LVI
2) High Risk (concurrent chemotherapy)
* Positive margins
* ECE
Pre-simulation MDT Discussion (review path)
Dietician + speech path involvement
- Consider prophylactic PEG
OPG + Dental review
Audiometry
Smoking cessation
Simulation Supine with thermoplastic mask
- Handgrips
- Vacbag and ankle/knee-blocks
Wire any surgical scars
CT (2mm with IV contrast)
- Vertex to mid chest
Fusion Pre-op PET-CT and MR fusion

Dose prescription
SIB technique
- 60Gy/30F to tumour bed
○ If gross residual –> 66Gy/33#
- 54Gy to the surgical bed
- 52Gy to the elective nodal regions

VMAT technique
10 days per fortnight
Concurrent chemotherapy (if positive margin or ECE)
        - Cisplatin 100mg/m2 q3wk
        - Cisplatin 40mg/m2 q1wk

Volumes
Pre-op GTVp = based on original MR
GTVp = residual macroscopic disease
CTV60* = pre-op GTVp + 5mm
- If gross residual –> CTV66 = GTVp + 5mm
CTV54 = surgical bed
CTV52 = elective nodal regions
- Generally II, III, IV
- Consider levels Ib, VI and VII
PTV = CTV + 5mm

*Treat tracheostomy site if trach was placed through tumour or when tumour involves subglottis. In the IMRT era, contour the trach site and use surface dosimeters to ensure surface dose 60 Gy (does not require donut bolus). Target Verification	Daily CBCT

OARs
Brainstem + Optic Nerves/Chiasm
- Dmax < 54Gy
Cochlear
- Mean < 45Gy
Lacrimal Gland
- Dmax < 30Gy
Eye
- Mean < 35Gy
- Dmax < 50Gy
Lens
- Dmax < 7Gy
Parotid (spare 1 gland)
- Mean < 26Gy (optimal)
Oral cavity
- Mean < 30Gy
Mandible
- Dmax < 70Gy
Larynx
- Mean < 45Gy
Oesophagus
- Mean < 35Gy
Pharyngeal constrictors
- Mean < 50Gy

69
Q

What low risk elective nodes would you include in your larynx treatment for different nodal stages?

A
70
Q

What low risk elective nodes would you include in your hypopharynx treatment for different nodal stages?

A
71
Q

What is the local control and OS for larynx and hypopharynx cancers by T-stage?

A
72
Q

What are the different methods of voice production post laryngectomy?

A

1) Tracheo-oesophageal Speech àpulmonary driven –
à By inhaling air and then occluding the stoma (with thumb or hands free speaking valve), the air is diverted through the voice prosthesis and into the oesophagus. This allows for voice to occur via a new source of vibration – the neoglottis.
à The voice prosthesis incorporates a one way valve, and retaining flanges at each end. It is sized by a Speech Pathologist according to the length (mm) & diameter (Fr.) required. 2 types – pt changeable non-indwelling or semi-permanent (3-6m) indwelling fitted by speech path.
2) Electronic Devices à held against the neck (or intra-orally with small tube) and the mechanically generated sound is transmitted through the tissue and sets the static air within the vocal tract into vibration. This sound is then shaped into speech by the lips, teeth and tongue. The Servox Digital and NuVois are two commonly used hand held devices
3) Oesophageal Speech à produced by inhaling or injecting air into the oesophagus. When the air pressure in the oesophagus is great enough to overcome the pharyngo-esophageal segment (neoglottis) resistance, the PE tissue is set into vibration. Air passing through the vibrating PE segment is then shaped into intelligible speech by the lips, teeth and tongue

73
Q

What is the Epidemiology, risk factors and biological behaviour for Nasopharynx cancers?

A

Incidence (Australian statistics)
- NASOPHARYNX = 200 cases annually (mostly Asian migrants)
- Incidence is <1 per 100000 people

Median age is 50 years
- May be younger in endemic areas
Male predominance (up to 3:1)
Marked geographic variation
- High = China and South-East Asia
- Intermediate = North Africa, Middle East
- Low = USA and Europe

Aetiology

1) Viral (EBV) --> Endemic form
	a. Major risk factor in Endemic regions
2) Dietary
	a. Salted/cured foods --> inhalation of volatile nitrosamines in steam
3) Smoking
4) Alcohol use
	a. Synergistic with smoking
5) Previous radiation
6) Genetic factors Genetic variation in HLA --> ? Susceptibility to EBV

Natural history
* 90% have subclinical LN disease therefore electively treat bilat neck
* Sporadic type - propensity to locoregional spread, see large primary, no clinically involved LN. 70% have LN involved
* Endemic type - tend to have small primary but large clinical LN or DM. 90% have LN involvement
* Usually from posterolateral wall, fossa of Rosenmuller.
* Locoregional spread
○ Superior- Sphenoid sinus, CN 3-6, V2 + middle cranial fossa (via foramen lacerum)
○ Anterior- Nasal cavity, ethmoid sinus, cribriform plate, maxillary sinus, orbit.
○ Inferior- Palate, oropharynx, C1
○ Posterior- Retropharyngeal space, carotid sheath, base of skull (CN 9-12), clivus
○ Lateral- Parapharyngeal space, then up into pterygoid m (trismus), Eustachian tube -> middle ear
* LN spread at diagnosis- 80% ipsilateral, 50% bilateral. RP, Upper deep Cx LN +/- post triangle LN
○ The N staging correlated with mets
○ N1 ~10%, N3 ~70%
○ 1st echelon node- Level II and RP
○ 2nd station node - Level III and V
* RP (VIIa), upper deep cervical LN (II&III), +/- posterior triangle LN (V) (post triangle node DDx skin, NPC
* Haematogenous spread bone, lung, liver. 10% at diagnosis, more in endemic type compared to sporadic.

74
Q

Describe the pathogenesis for Nasopharynx cancer and pattern of spread.

A

EBV carcinogenesis

EBV virus infection is endemic in Asia and parts of Africa

Mucosal disruption allows virus to invade and infect basal layer cells
- Limited change in the short period –> latent period
EBV integrates into host DNA and promotes expression of LMP1 and LMP2 proteins
- LMP2 –> EMT and suppression of differentiation
- LMP1 –> increased expression of pro-inflammatory molecules
-
There is also upregulation of a number of growth pathways
- EGFR
- PI3K/mTOR
- JAK-STAT

Pattern of spread
* Starts at fossa Rosenmuller
○ Easy spread to nasal cavity, palatal muscle
▪ Including levator veli palatini (up to base of skull, between medial and lateral pteryoid plates)
▪ Tensor veli palatini –hooks around hamulus (medial pterygoid)
* Both immediately adjacent to rosenmuller either side of auditory canal
* Base of skull (no anatomical barrier to bottom of sphenoid) = don’t skimp on sphenoid
* Into bone, perineural

75
Q

Describe the pathology for nasopharynx cancer.

A

Histological types
- WHO subtypes of SCC (95%)
○ Non-keratinising SCC –> endemic
§ WHO 2 Differentiated type (30%)
§ WHO 3 Undifferentiated type (>50%)
○ WHO 1 Keratinising SCC (20%) –> sporadic
○ Basaloid SCC –more aggressive
- Lymphoma (5%)

Macroscopic
- Mass within the nasopharyngeal space
○ May have associated ulceration or bleeding
- Distortion of normal anatomy (e.g. Fossa of Rosenmuller, Torus tubarus)

Microscopic
- Non-keratinising undifferentiated (50%) (Type 3)
○ Two key patterns
§ Syncytial arrangement of cells with indistinct margins
§ Diffuse infiltrate of non-cohesive cells
○ Moderate cytoplasm, round nuclei
○ No keratinising features
○ Apoptosis and mitosis common
○ No necrosis
○ Dense lymphocytic stroma infiltrate
○ Strong EBV
- Non-keratinising differentiated (Type 2)
○ Interconnecting cords of tumour with little keratinisation
○ Cells with well-defined cell borders
○ Variable stroma lymphoplasmacytic infiltrate
○ No desmoplastic stroma
○ EBV 50/50
- Keratinising (Type 1)
○ As per keratinising SCC of the remaining pharynx
§ Keratin pearls, intercellular bridges
§EBV negative
- Basaloid (Non-WHO - Rare)
○ As per basaloid SCC of the remaining pharynx
§ Sheets of cells with peripheral palisading
§ High-grade features (high N:C ratio, scant cytoplasm, nuclear pleomorphism)
○ Poor prognosis

Immunohistochemistry
- POS = EBV LMP, PanCK, HMWCK, p40, p63
- NEG = CK7, CK20

Molecular
- EBER ISH
- EBV PCR

76
Q

What are the prognostic features of Nasopharynx cancers?

A

Patient Factors
- Age and performance status
- Nutritional status
- Immunosuppression
- Smoking (local control and toxicity)
- Anaemia
○ Non-reversible –> transfusion does not improve outcomes

Tumour Factors
- TNM stage
○ Advanced T stage predicts worse local control and overall survival
○ Advanced N stage predicts distant metastasis and worse survival
○ Presence of distant metastasis usually indicates poor prognosis and treatment palliative
- Histological subtype
○ Non-keratinising has best prognosis
- EBV viral load
○ Both pre-and post treatment
- LVI
- PNI
- ECE

Treatment Factors
- High-volume experienced centre
- Delivery of chemoRT
○ Addition of chemoradiotherapy
○ Overall treatment time
○ Sequence (induction chemotherapy)

- Whilst serum EBV DNA is prognostic, there is insufficient information for how to utilise this information
	○ Baseline pre-treatment --> prognostic
	○ Post-chemoRT to predict benefit of adjuvant chemotherapy --> no supporting evidence, currently being investigated in trial by NRG-HN001 Ongoing long-term surveillance biomarker --> insufficient evidence
77
Q

How would you work up a patient with nasopharynx cancer?

A

Consultation

- History
	○ HPI
		§ Most common presentation = painless neck mass.
		§ Features of local disease
			□ Nasal- postnasal obstruction, loss of smell, epistaxis, blood stained nasal discharge
			□ Aural- otitis media, deafness, otalgia, tinnitus.
			□ Orbital- proptosis, restricted eye movement, diplopia, impaired vision.
			□ Pharyngeal- difficulty speaking, dysphagia
			□ Trismus dt masticator space extension.
			□ Neurologic- CN involvement- 2-6 superior invasion, 9-12 posterior.
		 
		§ Secondary spread-
			□ Neck mass, unilat or bilat, upper deep Cx LN. Most commonly RPLN (LN of Rouvier ant to C1), post Cx LN + jugular chain LN.
		§ Other- pain, headache, nausea, Vomiting, anorexia, loss of weight.
	○ PMHx:
		§ Immunosuppression
		§ Previous H+N malignancy
		§ Previous RT
	○ Meds
		§ Immunosuppressants
	○ Social:
		§ Ethnicity
		§ Smoking

- Examination
	○ General inspection
	○ Cervical nodal palpation
	○ Nasoendoscopy
		§ Examine entire pharynx (synchronous lesions)
	○ Cranial nerve examination

Work-Up

- Biopsy
	○ MUST include EBV staining (or EBER-ISH)
	○ Biopsy any equivocal neck nodes (unless already proceeding with neck dissection)

- MR Head and Neck (must include base of skull)
	○ Surgical evaluation

- FDG-PET
	○ Nodal and distant staging
	
- Diagnostic CT Head and Neck
	○ Evaluate bony invasion

- Serum EBV PCR
	○ Prognostic (pre-treatment and post-treatment kinetics)

- Pre-chemotherapy bloods
	○ FBC
	○ EUC, CMP
	○ LFT and coags
	○ HepB sAg, sAb, cAb
	○ Hep C serology
	○ HIV serology
78
Q

What is the staging for Nasopharynx cancer?

A

Stages:
- Stage I
○ T1N0
- Stage II
○ T1-2 N0-1

- Stage 3
	○ T3+ OR N2+
- Stage IV
	○ T4 OR N3
79
Q

In general what are the management options for different stages of nasopharynx cancer?

A
  • Radiotherapy-based management is the mainstay for nasopharyngeal cancer
    • In general, options may include
      ○ Stage I Disease (?low ebv)
      § Radiotherapy alone
      ○ Stage II Disease
      § Concurrent chemoradiotherapy
      § For non-bulky T2N0, can consider RT alone
      ○ Advanced Stage (stages III-IV)
      § Induction chemotherapy followed by concurrent chemoRT (preferred)
      □ Consider omitting induction chemo if T3N0
      § Concurrent chemoRT followed by adjuvant chemo
    • Some experts choose induction therapy for:
      ○ Large primary tumours (T4) lesions (high risk of DM)
      ○ Advanced nodal disease (N3, large burden or supraclavicular location) (high risk of DM)
      ○ When full dose RT not deliverable because the tumour abuts critical structures (e.g. optic apparatus, brainstem, temporal lobes)
    • Always establish relevant supportive care early
      ○ Smoking cessation
      ○ Dietician and speech pathology input
      § Consideration of prophylactic gastrostomy
      ○ Dental input
      § Pre-RT OPG
      ○ Social & psychological supports as appropriate
    • Note that much of the data is established in endemic regions (Asia)
      ○ Uncertain relevance for non-Asian populations
80
Q

What is the role of surgery in management of nasopharynx cancer?

A

Role of surgery
- Role limited due to anatomical constraints, technically difficult with poor functional outcome, cannot reach RPLN. (Essentially not curable)
- EUA + Bx for diagnosis
- LND for residual LN or recurrence in neck after RT
- Salvage nasopharyngectomy- select cases (T1-T2) w recurrence in NPX (good PS, small)
○ Highly morbid surgery- complications: meningitis, ORN, free flap necrosis, aspiration pneumonia, middle ear effusion, hypernasality, nasal regurgitation, CSF rhinorrhoea, ectropium, impaired swallowing, trismus, orbital numbness.

81
Q

How would you manage a locoregional recurrence of nasopharynx cancer?

A

○ Local recurrence rate up to 10% especially for larger tumours. Difficult surgery as prev IR tissue, deep in skull
○ A carefully selected subset of patients with locoregionally recurrent NPC may achieve long-term survival when managed with salvage surgery and/or re-irradiation
○ Workup
§ Hx + PE
§ MRI H&N
§ PET/CT to exclude distant metastasis
○ Management options
§ Salvage chemo -cisplatin/gemcitabine
□ Alternative: other single agent chemo
□ Immunotherapy trials second line (not PBS)
§ Salvage surgery only for select cases, T1-2N0M0
□ RE-RT is indicated if close or pos margin
HOWEVER, Re-irradiation NOT suitable IF: disease interval < 12mo, toxicity >G1 in critical organs and > G3 in non-critical organs
□ potential long term OS in select cases
□ Options
® IMRT (60-66Gy/33#)
® Intracavitary and interstitial brachytherapy
® SBRT
® Proton beam RT
□ Choice depends on local expertise and technical considerations including location and extent of recurrent disease
® Local recur: prefer endoscopic > Re-RT
® Re-RT (No Chemo) 65Gy/54F bid (Guangzhou), 3yOS 70 vs 50% favour bid (d/t late TRM, not DRM), G3 Tox and death rate were significantly lower with BiD regimen
◊ Volume: GTV, CTV 5mm, PTV 3mm
◊ Constraints:
Spinal Cord: max 60Gy
B’stem optic: max 70Gy
Temporal lobe: 90Gy
Carotid artery: 125Gy

82
Q

Discuss induction, concurrent and adjuvant chemotherapy in nasopharynx cancer.

A
  • Chemoradiotherapy is superior to radiotherapy alone
    ○ MAC-NPC meta-analysis
    § any combination of Chemo- CCRT alone, Induction chemo> CCRT or CRT> Adj Chemo. Addition of induction Adj Chemo improved OD benefit to CCRT, but which one is better? Still being debated although CCRT > Adj Chemo yielded the most benefit.
    ○ Concurrent cisplatin (as per standard H+N)
    • For Stage III-IV NPC, induction chemotherapy improves OS compared with chemoRT alone
      ○ Should be considered a standard of care
      ○ Cisplatin + 5-FU (or capecitabine)
    • Role of adjuvant chemotherapy is controversial
      ○ Data suggests that concurrent chemoRT + adjuvant chemotherapy is superior to RT alone
      § Intergroup 099 trial - both concurrent and adjuvant CT provides 30% benefit. however, unsure the contribution of adjuvant rather than concurrent Ctx. Also only 55% of pts completed adjuvant CT
      § Limited due to poor design (uncertainty regarding which component of chemotherapy derives benefit)
      ○ Uncertain benefit of adding adjuvant chemotherapy to chemoRT- might improve 3yr OS and FFS based on P3 RCT comparing Adj Capecitabine vs obsessed post Def CRT
      § Consider addition of metronomic capecitabine as adjuvant therapy for 12mo
      □ Metronomic = low-dose long-term dosing (aims to target angiogenesis and limit toxicity)
      □ 650mg/m2 BD for 12mo

Summary
- Concurrent chemoradiotherapy is superior to RT alone
- Addition of extra chemotherapy is superior to chemoRT alone
No clear difference between induction vs adjuvant

83
Q

What is the evidence for management of early stage nasopharyngeal cancer?

A

Early stage RT alone noninferior:
Tang et al RCT JAMA 2022 - CRT vs RT alone
○ 341pts with low-risk NPC, AJCC 7 Stage II or T3N0 without any of the following RFs:
§ cervical node >3cm
§ Lvl 4/5 nodes
§ ENE
§ EBV DNA >4000 copies/ml
○ RT alone results in 3-year LR control, DFS, OS, DMFS and LRFS comparable to CRT, with significantly less toxicity for patients with stage II and T3N0, low-risk, endemic NPC
○ RT alone IMRT demonstrated non inferiority 3yr FFS (91vs 92%) and OS (98 vs 99%)
○ DMFS and LRFS were also similar for the 2 treatment
○ IMRT had lower toxicities and better QoL

84
Q

What is the evidence for induction or adjuvant chemo in nasopharynx cancer?

A

CRT/chemo improves overall survival, LRC:
MAC-NPC meta-analysis (Ribassin-Majed, 2017)
○ 5144 patients from 20 trials
○ Outcomes
§ Compared with RT alone
□ ChemoRT alone improved OS (HR 0.77)
□ Induction chemo + chemoRT improved OS (HR 0.81)
□ ChemoRT + adjuvant chemo improved OS (HR 0.65)
§ No significant difference between any of these arms
§ Induction chemo + chemoRT had the highest probability for superiority for (not a true direct comparison)
□ Distant control
□ Locoregional control
○ Conclusion:
§ Additional chemotherapy, whether induction or adjuvant, is necessary to improve OS (except, perhaps, in T1-2N0; addition of any chemo OS HR 0.98). There is heated debate as to whether induction or adjuvant chemo is standard of care, as CCRT followed by adjuvant chemotherapy has been the historical standard.
§ 5y OS benefit for all chemo/RT combinations ~6%.
□ OS benefit is most pronounced for WHO type I (Keratinising SCC is most radioresistant)
□ 5y OS benefit of 12% for concurrent and adjuvant chemo, or 8% for CCRT alone
§ 2017 update demonstrates CCRT→ CTX > CTX→ CCRT, but each with benefit. Induction was associated with the best distant control. Chemo benefit decreases with age
§ 2022 update: confirms CCRT-> CTX is superior to CTX-> CCRT in absolute 10yr OS improvement 14% vs 10%
□ HR and 5yr OS for chemo is better if 40- <60yo

85
Q

What is the evidence for adjuvant capecitebine in nasopharynx cancer?

A

Phase III metronomic capecitabine trial (Chen, 2021)
○ 406 patients with stage III-IV nasopharynx cancer were randomised to
§ Concurrent chemoradiotherapy alone
§ Addition of adjuvant metronomic capecitabine (650mg/m2 for 12mo)
○ Outcomes
§ Improved 3 year OS (93% vs 89%)
§ Improved FFS (85% vs 76%)

86
Q

What is the evidence for local treatment in metastatic nasopharynx cancer?

A

Phase III RCT (You, 2020)
- 126 patients with de-novo metastatic disease were treated with
○ 3x cisplatin/5-FU and if CR or PR, randomised to
§ Chemoradiotherapy
§ Chemo alone (complete 6 cycles)
- Outcomes
○ Locally aggressive therapy improved 2 year OS (76% vs 54%)
○ Improved PFS

87
Q

Is there evidence for acceleration in Nasopharynx cancer?

A

PC-0501 trial (Lee, 2015)
- 706 patients with stage III to IV NPC were randomised to a 3x2 randomisation (6 arms)
○ Concurrent chemoRT (conventional vs accelerated)
§ 70Gy at 5 vs 6F per week
○ Induction vs adjuvant chemotherapy
○ Cisplatin + 5-FU vs capecitabine
- Outcomes
○ No benefit to acceleration
○ Cisplatin/capecitabine was superior for PFS (HR 0.54) and OS (HR 0.42)
○ No difference between induction vs adjuvant chemotherapy

88
Q

For nasopharynx cancer, what is your radiotherapy technique?

A

Definitive Therapy

Patients
1) Stage I (no chemotherapy)
2) Stage II (concurrent chemoradiotherapy)
1. Consider RT alone if non-bulky T2N0
3) Stage III-IV (induction chemotherapy + concurrent chemoradiotherapy)

Pre-simulation
MDT Discussion (review path)
Dietician + speech path involvement
- Consider prophylactic PEG
OPG + Dental review
Audiometry
Smoking cessation
Thorough baseline cranial nerve exam

Simulation
Supine with thermoplastic mask
- Handgrips
- Vacbag and ankle/knee-blocks
CT (2mm with IV contrast)
- Vertex to mid chest

Fusion
Pre-chemotherapy PET-CT and MR fusion
- Treat on pre-induction volume

Dose prescription
SIB technique
- 70Gy/35F to the involved region
- 63Gy/35F to the high-risk region
- 56Gy/35F to the elective nodal regions
Concurrent chemotherapy (if stage II-IV)
- Cisplatin 100mg/m2 q3wk
- Cisplatin 40mg/m2 q1wk

VMAT technique
10 days per fortnight
- Accelerate to 6F per week if no chemotherapy

Volumes
GTVp = macroscopic primary disease
(often include involved retropharnygeal extension/nodes)
GTVn = macroscopic nodal disease

CTV70
- (GTVp + GTVn) + 5mm (min 1mm if critical OAR)
+-remaing nasopharynx to depth of 5mm to C1
CTV63
- GTVp + 10mm (min 2mm if critical)
- Include pre-chemo GTV if they have had induction chemo
- GTVn + 10mm (if ENE eg into SCM)
- Include all of:
○ Nasopharynx
○ Nasal cavity (posterior 5mm), maxillary sinus
○ Ethmoid (posteroinferior), vomer
▪ Half of sphenoid sinuses
▪ T3/4 +whole of sphenoid sinus (if involved)
○ Skull base (cover foramen ovale/rotundum/lacerum/petrous tip)
○ Pterygoid plates and fossa (and hook of hamulus)
○ Parapharyngeal space bilat
○ Clivus (anterior 1/3) -all of it if involved
○ Ipsilateral Cavernous sinus (if T3-4 only)
- Include air
- Don’t edit medial and lat pterygoid muscle
- If high Jugular nodes –include jugular foramen

CTV56
- Elective nodal regions
PTV
- CTV + 5mm
Nodal regions
- Highest risk = bilateral II, III, IVa, Va/b, VIIa, VIIb
○ Often cover 1b (if adjacent 2cm lvl 2 node or ECE) and Vc
- If cN0 on that side = omit IV and Vb
- If level IVa involved = include IVb

After Induction Chemo:
[As per EVIQ]
Fuse pre-Chemo PETCT
CTV70: Residual GTVp+GTVn (as per POST-chemo images) + 5mm
CTV63:
* CTVp70+10mm
* All CTVp63 at risk site as above
* Include GTVp (pre-chemo)+5mm and GTVn+5mm
* CTVn63= CTVn70+5mm

Target Verification
Daily CBCT

OARs
Brainstem + Optic Nerves/Chiasm
- Dmax < 54Gy
Temporal Lobe
- Dmax < 60Gy
Spinal Cord
- Dmax < 45Gy
Cochlear
- Mean < 45Gy
Lacrimal Gland
- Dmax < 30Gy
Eye
- Mean < 35Gy
- Dmax < 50Gy
Lens
- Dmax < 7Gy
Parotid (spare 1 gland)
- Mean < 26Gy (optimal)
Oral cavity
- Mean < 30Gy
Mandible
- Dmax < 70Gy
Larynx
- Mean < 45Gy
Oesophagus
- Mean < 35Gy
Pharyngeal constrictors
- Mean < 50Gy

89
Q

What would be your elective neck lymph node targets in nasopharynx cancer?

A
90
Q

What would be the expected 5year overall survival for nasopharynx cancer patients?

A
91
Q

What is the epidemiology and risk factors for Salivary cancers?

A

Incidence (Australian statistics)
- 265 cases annually
- Less than 1 per 100000 people

Slight female predominance (1.5:1)
Broad age range (median 50 years)
- Children can be impacted also (mucoepidermoid)

Clinicopathological Features

Bilateral tumours * Warthin’s tumour
* Pleomorphic adenoma
Children * Pleomorphic adenoma
* Mucoepidermoid carcinoma
* Adenoid cystic carcinoma
Nodal * Generally unilateral
Metastases * Generally to lungs

Inverse relationship between gland size and likelihood of malignancy
1) Parotid
a. 80% benign | 20% malignant
2) Submandibular
a. 60% benign | 40% malignant
3) Sublingual
a. 40% benign | 60% malignant
4) Minor salivary
a. 50% benign | 50% malignant

Aetiology

1) Smoking
	a. Especially Warthin's tumour
2) Previous radiation

Minor risk factors
1) Viral infections
a. EBV –> lymphoepithelial carcinoma
b. HPV –> mucoepidermoid (controversial)

Postulated (insufficient evidence)
- Alcohol use
- Mobile phone use
- Nutritional deficiencies
- Use of hair dye

92
Q

Describe the pathological features of salivary pleomorphic adenomas.

A

Pleomorphic Adenoma
- Represent 60% of all tumours of the parotid gland
- Most common salivary gland tumour in children and adults. All ages 30-60. F>M
- Commonly parotid gland.
- Slow growing, painless
- Small chance of malignant transformation (10% risk if lesion is present for 15 years)
- Can be bilateral or multifocal

- Macroscopic
	○ Moderate (<6cm), rounded and well-demarcated tumours
	○ Cut surface = grey-white with myxoid areas of chondroid stroma
- Microscopic
	○ Tri-phasic tumour with marked intratumoural heterogeneity
		§ Epithelial/ductal = inner layer of tubules
		§ Myoepithelial = outer layer of cysts and tubules
		§ Stromal = myxoid or chondroid
	○ Bosselated outer surface with tongue-like protrusions (pseudopods)
- Immunohistochemistry
	○ POS
		§ Epithelial = AE1/AE3, CAM5.2
		§ Myoepithelial = S100, GFAP, p40, p63
93
Q

Describe the pathological features of Warthin’s tumours

A

Warthin’s Tumour (5%)
- Almost always within the parotid gland
- Most common aetiology of bilateral parotid tumours
- Associated with smoking

- Macroscopic
	○ Encapsulated and lobulated tumour
	○ Cut surface = grey with multiple cysts containing mucin/serous secretion)
- Microscopic
	○ Double layer of epithelial cells resting on a dense lymphoid stroma
		§ Clefts are narrowed by lymphoepithelial projections
	○ No myoepithelial component
94
Q

List the benign and malignant histopathological types of salivary gland tumours.

A

Histopathology (Benign)
Subtypes
- Pleomorphic adenoma (50%)
- Warthin’s tumour (5%)
- Oncocytoma

Histopathology (Malignant)

Subtypes
- High-grade
○ Adenoid Cystic Carcinoma (25%)
○ High-grade Mucoepidermoid Carcinoma (17%)
○ Adenocarcinoma (12%)
○ Carcinoma Ex Pleomorphic Adenoma (8%)
○ Squamous Cell Carcinoma (5%)
- Low-Grade
○ Low-grade Mucoepidermoid Carcinoma
○ Acinic Cell Carcinoma (10%)
Lymphoepithelial Carcinoma (2.6%)

95
Q

Describe the pathological features of Mucoepidermoid carcinoma

A
  • Most common malignant tumour (15% of all salivary gland tumours) in children + adults
    • Generally present with a slow growth pattern
    • Macroscopic
      ○ Can grow as large as 8cm in diameter
      ○ Appear well circumscribed, there is no true capsule and infiltrative margins are common
      ○ Cut section = pale, grey-white with small mucinous cysts
    • Microscopic
      ○ Cells range from monotonous and bland to highly anaplastic
      § Mixture of mucin-producing and squamous cells
      § Keratinisation rare
      ○ Growth in sheets or cystic formations
      ○ Grading can be very subjective (high interobserver variation)
      § Armed Forces Institute of Pathology
      § MSKCC
    • Immunohistochemistry
      ○ POS = PanCK, CK5/6, p40, p63, MUC1/MUC2, CK7
      ○ NEG = S100, GFAP, AR, HER2, CK20, SOX10, GATA3
    • Molecular
      >50% will have a CRTC1-MAML2 fusion gene
96
Q

Describe the pathological features of Adenoid cystic

A

Adenoid Cystic Carcinoma
- Larger proportion of ACC occur in the minor salivary glands
- Known for its high local recurrence rate (even 10-15 years later)
- PNI

- Macroscopic
	○ Small, poorly encapsulated and infiltrative lesions
- Microscopic
	○ Biphasic ductal and myoepithelial differentiation
	○ Monotonous basaloid cells with scant cytoplasm and bland nuclei
		§ Appears low-grade despite behaviour
		§ Whilst graded histologically, all are considered 'high-grade' for management decisions
	○ Cribriform pattern (Swiss cheese) --> extracellular hyaline material fills spaces
	○ Solid pattern is associated with poor prognosis
	○ PNI is very common
	○ Can have blue and pink secretions (H+E)
- Immunohistochemistry
	○ POS = CK7, CAM5.2, p40, p63, S100 & GFPA (myoepithelial component)
- Molecular
	○ MYB-NFIB rearrangements are present in a large proportion (>60%)
97
Q

Describe the pathological features of Carcinoma Ex- Pleomorphic adenoma

A

Carcinoma Ex Pleomorphic Adenoma
- Malignant transformation of an existing pleomorphic adenoma
○ Typically a rapid increase in size in a long-term mass
- Prognosis is much poorer than denovo malignant tumours

- Note that this is not a histopathological diagnosis
	○ Ductal  and myoepithelial carcinomas are the most common
- Identification of the surrounding benign adenoma is essential for diagnosis

- Histopathology and IHC is dependent on the histopathological diagnosis
98
Q

What are the prognostic factors for salivary gland cancers?

A

Prognostic factors for LRC
* T stage ie size (benefit for RT for >4cm tumours)
* Grade
* Histology
* Good prognosis: acinic cell, , LG mucoepidermoid carcinoma (MEC)
* Poorer prognosis: HG MEC, adenocarcinoma, SCC, cancer ex pleomorphic adenoma, undiff or poorly differentiated, adenoid cystic carcinoma (late relapses PNI and metastatic)
* PNI esp ‘named’ nn involvement, size, length of involvement
* Facial n palsy
* Cx LN +ve
* Surgical margins +ve (clear = >5mm, close = 1-5mm)
* Extracapsular spread
* Ki67 (investigational)
Site: submandibular worse than parotid

99
Q

How would you work up a patient with a salivary gland tumour?

A

Consultation

- History
	○ HPI
		§ Mass in the salivary gland
			□ Parotid most common
		§ Typically painless
		§ May have facial nerve symptoms (paralysis)
	○ PMHx:
		§ Previous skin cancer of H+N
		§ Previous radiotherapy
	○ Social
		§ Smoking history
		§ Functional state
		
- Examination
	○ Local lesion
		§ Location and depth of lesion
		§ Tenderness
		§ Facial nerve involvement
	○ Contralateral gland
		§ Warthin's tumours are commonly bilateral
	○ H+N examination
		§ Thorough nodal palpation
		§ Skin examination
		§ Cranial examination esp CN V and VII
		§ Consider endoscopy if indicated

Work-Up

- CT HNCAP
	○ Completion of local staging
	○ Distant staging (lung and liver are most likely metastatic sites)

- MR H+N
	○ Local staging
	○ Assess for radiological PNI

- Biopsy
	○ FNAB is usually performed
		§ Generally sufficient to distinguish benign vs malignant
		§ Unable to determine histological subtype
100
Q

What is the staging for salivary gland cancer?

A
101
Q

Discuss surgical management of salivary gland tumours.

A
  • Surgery remains the mainstay of treatment
    • Resection of the Primary
      ○ Parotid
      § Types of surgery
      □ Benign –> superficial parotidectomy
      □ Low-grade malignant –> consider superficial parotidectomy
      □ High-grade malignant –> must have total parotidectomy
      § Always dissect out the plane of the facial nerve –risk facial nerve palsy –inability to close eye
      § Risk of Frey syndrome without recon
      □ Gustatory sweating
      § Recon options:
      □ Fat graft
      □ Locoregional flap
      □ Free flap
      ○ Submandibular gland
      § Benign –> simple excision (spare lingual, hypoglossal and marginal mandibular nerves)
      § Malignant –> en bloc excision (only spare nerves if clearly uninvolved)
      ○ Lower facial muscle weakness, tongue sensation/power
    • Management of the Neck
      ○ If cN+ disease
      § Ipsilateral neck dissection (levels Ib to IV)
      ○ If cN0 disease
      § high risk of occult met in 12-45% of clinically negative neck
      § Considerprophylactic neck dissection only if high-risk features
      □ T3/T4 disease
      □ High-grade histology
      □ Clinical facial nerve involvement
      § If radiotherapy is definitely planned, elective neck dissection can be omitted
      □ RT is equivalent for microscopic disease
102
Q

Discuss the role of adjuvant radiotherapy in benign salivary gland tumours.

A
  • Benign tumours
    • Generally no role for adjuvant RT. LR post op <2%.
      ○ If Warthin’s- no Adj RT is needed as LRR <5%
      ○ For Pleomorphic adenoma- LR <2% if parotidectomy, but higher RR if tumour spill >50% or inadequate enucleation 25%
      § Consider Adj RT IF multiply recurrent tumours, pos margin which cannot be resected due to CN7 compromise or tumour spill (lower dose - 50Gy/25F).
      LC for Sx+Adj RT 90-95%, but LC is lower for recurrent tumour ~80%
103
Q

Discuss the role of adjuvant radiotherapy in malignant salivary gland tumours.

A

Limited RCT and good prospective studies - mostly retrospective, small studies and poor quality. BUT SEER analysis and several retrospective analysis comparing Surg vs Surg+PORT support the use of PORT to improve 5yr LC and OS

- Low-grade malignant tumours
	○ Adjuvant RT indicated if:
		§ Positive margins
		§ Consider if multiply recurrent
		§ High-risk features (see below)

- High-grade malignant tumours
	○ Nearly all receive adjuvant radiotherapy (on basis of grade alone)
	○ Adjuvant RT indicated if:
		§ Positive margins
		§ High-grade (all adenoid cystic carcinomas)
		§ T2+
		§ Node positive
		§ Nerve invasion
		§ Tumour spill
		§ Bone/skin involvement
		§ LVI

- Radiotherapy technique
	○ Dose
		§ R0 resection = 60/54Gy/30#
		§ R1 resection = 63/60/54Gy/30#
		§ R2 resection = 70/63/56Gy/35#
		§ Accelerated regimen  6 F per week
	○ Volume
		§ Consider extension along facial nerve
		§ Must treat facial nerve for adenoid cystic
	○ No role for concurrent chemotherapy in adjuvant setting
		§ An analysis of over 2000 patients from the National Cancer Database failed to demonstrate any improvement in overall survival with the addition of chemotherapy to RT compared with RT alone (JAMA 2016)
104
Q

Discuss definitive radiotherapy for salivary gland tumours.

A
  • Only indicated when medically or technically inoperable
    ○ Surgery is preferred
    • Aim for 66-70Gy
      Concurrent cisplatin for definitive CCRT- extrapolated from the mucosal H&N
105
Q

What is the evidence for adjuvant radiotherapy in salivary gland tumours?

A
  • Data available is scant
    ○ Poor quality
    ○ Predominantly retrospective
    ○ Small numbers per series
    ○ Improved LRC and OS
    • There is no randomised data available to guide practice
    • Mahmood 2011 SEER database multivariate analysis- Adj RT improved OS HR 0.76
    • Terhaard 2004 Retrospective analysis
      ○ 538 pt, major salivary gland tumour, Surg+ PORT vs Surg alone. LRC was better for Surg+PORT.
      ○ Relative risk for Surg alone for Local Recurrence 9.7 and regional recurrence 2.3
    • Mendehall 2005
      ○ 224 pt, Surg+PORT vs Surg alone.
      ○ 10 LC for Surg+PORT vs Surg alone- 96% vs 75% for T1-3; 70% vs 21% for T4
      ○ 10yr OS for T4 27% in Surg+PORT vs 21% in Surg alone
    • Safdieh 2017- retro data of NCDB= significant OS associated with PORT
      ○ 5yr and median OS- 56% and 73mo in PORT vs 50.6% and 61mo in Surg alone. HR 0.79 (SS)
106
Q

Describe your adjuvant radiotherapy technique for salivary gland tumours.

A

Adjuvant Therapy

Patients
1) High-grade disease
2) Low-grade disease with high-risk features
3) Recurrent benign

Pre-simulation
MDT Discussion (review path)
Dietician + speech path involvement
OPG + Dental review

Simulation
Supine with thermoplastic mask
- Handgrips

Wire any surgical scars
Consider 1cm bolus (if skin invasion)
Consider intra-oral appliances.
- Bite block/mouth bung—to immobilise/displace tongue.
- Cheek spacer/wax buccal spacer held against inside of cheek to help separate from remainder of OC—for buccal primary.

CT (2mm with IV contrast)
- Vertex to mid chest

Fusion
Pre-op MR fusion

Dose prescription
Microscopic disease ONLY
- 63Gy/30 to tumour bed
- 60Gy/30F to tumour bed+ surgical bed
- 54Gy to the elective nodal regions

Macroscopic disease
- 70Gy/35F to residual macroscopic disease
- 63Gy to the tumour bed+ surgical bed
- 56Gy to elective nodal regions

VMAT technique
Accelerate to 6F per week

Volumes
GTVp= gross disease preop and residual gross disease post op
CTV63: tumour bed (GTVp+5mm) [IF parotid use 1.5cm instead]
CTV60: tumour bed + 5mm, Surgical bed include clips, flap, and surgically disturbed area

-If parotid, include parotid bed= parapharyngeal space, infra temporal fossa, masseter, pterygoids, mandibular ramus
- Include mandible if bone invasion.

CTV54: elective region as per below

[Parotid]
- Only parotid bed if pN0 on LND.
- Ipsi Lv 2-4 IF cN0, NO LND done.
- Ipsi Lv 1b-5 IF N+.

[Salivary gland]
- Ipsi Lv 1b-4. Include Ipsi Lv 5 if node pos.
- Consider contrala 1-2 if level 1 involved
- Include Contralateral if primary close to midline

[Micro PNI]
- Include named nerve to BOS if micro PNI.
- IF CNVII Extend to stylomastoid foramen + IAM
- IF CNXII to hypoglossal canal
- IF CNV3 to foramen ovale
* If gross PNI, include elective neural coverage and branches in CTV60 instead
PTV= CTV +5mm

Target Verification
Daily CBCT

OARs
Brainstem
- Dmax < 54Gy
Cochlear
- Mean < 45Gy
Lacrimal Gland
- Dmax < 30Gy
Eye
- Mean < 35Gy
Lens
- Dmax < 7Gy
Parotid (spare 1 gland)
- Mean < 20Gy (optimal)
Mandible
- Dmax < 70Gy
Larynx
- Mean < 45Gy

107
Q

Describe your definitive radiotherapy technique for salivary gland tumours.

A

Definitive Therapy
Patients Unresected disease
Pre-simulation MDT Discussion (review path)
Dietician + speech path involvement
OPG + Dental review

Simulation
Supine with thermoplastic mask
- Handgrips
Wire any surgical scars
Consider 1cm bolus (if skin invasion)
CT (2mm with IV contrast)
- Vertex to mid chest
Fusion MR fusion

Dose prescription Macroscopic disease
- 70Gy/35F to the macroscopic tumour
- 63Gy/35F to the intermediate volume
- 56Gy/35F to the elective nodal regions

VMAT technique
10 days per fortnight
Consider concurrent cisplatin (controversial)

Volumes
GTVp = macroscopic primary disease
GTVn = macroscopic nodal disease
CTV70
- GTVp + 5mm
- GTVn + 5mm
CTV63
- GTVp + 10mm
- Try to include entire parotid gland
CTV56
- Elective nodal regions
○ Generally II, III, IV
PTV = CTV + 5mm

Target Verification
Daily CBCT

OARs
Brainstem
- Dmax < 54Gy
Cochlear
- Mean < 45Gy
Lacrimal Gland
- Dmax < 30Gy
Eye
- Mean < 35Gy
Lens
- Dmax < 7Gy
Parotid (spare 1 gland)
- Mean < 20Gy (optimal)
Mandible
- Dmax < 70Gy
Larynx
- Mean < 45Gy

108
Q

What is the epidemiology and risk factors for Sinonasal tumours?

A

Incidence (international statistics)
- Very rare cancers (as an entire group)
- Up to 5-10 per million people

Male predominance (2:1)
Mean age > 60 years
No race predilection

Aetiology

Overall
1) Smoking
2) Alcohol consumption
3) Occupational exposures
a. Chromium
b. Mustard Gas
c. Radium

Adenocarcinoma
1) Woodworkers (inhalation of saw-dust)
2) Formaldehyde exposure
3) Industrial glue/adhesive exposure

Squamous Cell Carcinoma
1) Woodworkers (inhalation of saw-dust)
2) Construction workers
3) Bakers, pastry chefs and grain millers
4) Nickel workers

109
Q

List the benign and malignant tumours that can arise in the sinonasal region.

A

Histopathology (Benign)

Subtypes
- Nasopharyngeal angiofibroma
- Schneiderian Papilloma
○ Inverted papilloma (most common)- 50-80%. 10% risk of malignant transformation
○ Exophytic Papilloma - 20-50%. No malignant potential
○ Oncocytic papilloma - 5%

Histopathology (malignant)

Subtypes
- Squamous Cell (32%)
- Sarcoma (13%)
- Esthesioneuroblastoma (11%)
- Lymphoma (10%)
- Adenoid cystic carcinoma (9%)
- Undifferentiated - SNUC (8%)
- Adenocarcinoma (6%)

110
Q

Describe the pathology for Nasopharyngeal angiofibroma

A
  • Almost exclusively occurs in adolescent males
    • Benign, but locally aggressive tumour
    • main treatment is surgical removal, might need preop embolisation to minimise bleeding.
    • RT if tumour is unresectable
    • Macroscopic
      ○ Firm, polypoid mass most commonly from posterolateral wall of nasal cavity
      ○ Can be yellow to dark red/black depending on haemorrhage
    • Microscopic
      ○ Fibrovascular lesion with two components
      § Vascular spaces with vessels of varying calibre
      § Fibrous/collagenous stroma
      ○ Mitoses are absent
111
Q

Describe the pathology for sinonasal inverted papilloma

A

nverted papilloma (Schneiderian)
- Benign neoplasm (but locally aggressive) most commonly arising in the lateral nasal wall
- Male pre-dominance and typically arise >40 years old
- Possible implication of HPV in development (remains controversial)
- At risk of malignant transformation - 10% risk (carcinoma ex sinonasal papilloma)

- Macroscopic
	○ Pink-grey polypoid growth with a convoluted or wrinkled surface
- Microscopic
	○ Prominent downward endophytic growth of epithelial nests with a smooth outer contour Hyperplastic epithelium Oedematous stroma
112
Q

Describe the pathology for sinonasal SCC

A
  • Most common subtype of sinonasal origin
    • Mean age of 60 years
    • Most frequently in maxillary sinus > nasal cavity > ethmoid sinus > sphenoid / frontal sinuses
    • Nasal origin has better prognosis than maxillary sinus
    • Macroscopic
      ○ Highly variable appearance
      § Exophytic or fungating
      § Haemorrhagic, necrotic or friable
    • Microscopic
      ○ 85% of cases are keratinising (intercellular bridging, keratin pearls)
    • Immunohistochemistry
      ○ POS = CK5/6, CK7
113
Q

Describe the pathology for sinonasal Adenocarcinoma

A
  • Woodworking is the primary risk factor
    • Poor overall prognosis
    • Macroscopic
      ○ Irregular exophytic pink mass
      ○ May have gelatinous appearance
    • Microscopic
      ○ Three key types
      § Intestinal type
      □ Resembles colonic mucosa
      § Salivary type (most common)
      □ Can present as adenoid cystic carcinoma
      § Non-salivary non-intestinal type
      Best prognosis
114
Q

Describe the pathology for Sinonasal Undifferentiated carcinoma (SNUC)

A
  • Poorly differentiated epithelial tumours
    • Rare tumours with extremely poor prognosis
    • Median age 55 years
    • Macroscopic
      ○ Ill-defined fungating tumours, very rapid
      ○ Extensively invades adjacent structures
    • Microscopic
      ○ Nests, lobules or sheets of medium sized epithelial cells
      § Scant cytoplasm, large hyperchromatic nuclei, indistinct membranes
      § High mitosis and necrosis rates
      ○ No specific lineage is obvious (no keratinisation, no glandular, no neuroendocrine)
      ○ LVI, PNI
    • Immunohistochemistry
      ○ POS = AE1/AE3, CAM5.2
      ○ NEG = neuroendocrine markers (CD56, chromogranin, synaptophysin), squamous markers (CK5/6, p40, p63),
    • Molecular
      ○ IDH2 mutations are highly common
      ○ EBER is negative (EBV neg)
115
Q

Describe the pathology for Esthesioneuroblastoma

A

Esthesioneuroblastoma (olfactory neuroblastoma)
- Arise from the neuroectodermal olfactory cells
- Bimodal incidence peaks (15yo and 50yo)
- Very rare tumour

- Macroscopic
	○ Red-grey vascularised polypoid mass
	○ Typically in the superior nasal cavity (olfactory cells)
- Microscopic
	○ Lobular arrangement of neuroblastoma cells
	○ Small blue round cell tumour, pseudorosettes or true rosettes
		§ Lower grade tumours grow in nests/lobules with a fibrovascular stroma
		§ Higher grade tumours grow in sheets/solid pattern and may have marked mitoses + necrosis
	○ Fibrillary neural matric may be present
- Immunohistochemistry
	○ POS = CD56, synaptophysin, chromogranin, S100
	○ NEG = FLI-1 (Ewing's), EBER, NUT, lymphoma markers (e.g. CD45)
116
Q

Describe the pathology, clinical presentation, and work up of NUT tumours

A

NUT midline carcinoma, often referred to as NUT tumors, is a rare and aggressive type of cancer.
Genetic Mutation and Pathogenesis:
○ NMC is characterized by chromosomal rearrangements involving the NUT gene on chromosome 15. The most common rearrangement is a translocation between chromosomes 15 and 19, resulting in a fusion gene combining NUT with BRD4 (BRD4-NUT).
○ This fusion gene disrupts the normal regulation of cell growth and differentiation, leading to uncontrolled proliferation of cells and tumor formation.
Clinical Presentation:
○ NMC typically arises in midline structures of the body, including the head, neck, and thoracic regions (such as the lungs and mediastinum).
○ Symptoms are related to the location of the tumor and can include swelling, pain, respiratory or swallowing difficulties, and other organ-specific issues.
Diagnosis:
○ Diagnosis is challenging due to the rarity of the disease and its nonspecific symptoms.
○ Histologically, NMCs are poorly differentiated carcinomas.
○ Definitive diagnosis requires identification of the NUT gene rearrangement, usually through fluorescence in situ hybridization (FISH) or other molecular diagnostic techniques.
Age of Onset and Demographics:
○ NMC can affect individuals of any age, including children and adults, but is more commonly diagnosed in adolescents and young adults.
○ There is no significant gender predilection for NMC.
○ No particular RF, no lifestyle/ environmental factors, familial predisposition

Prognosis:
○ The prognosis of NMC is generally poor, with a high rate of metastasis and recurrence.
○ The median survival time from diagnosis is typically less than a year.
Prognosis depends on several factors, including the extent of disease at diagnosis, response to treatment, and overall health of the patient.

117
Q

Discuss the treatment options for NUT tumours.

A

○ Treatment options are limited due to the aggressive nature of the disease and resistance to conventional chemotherapy.
○ Management typically involves a multimodal approach including surgery, radiation therapy, and chemotherapy, but the overall response to these treatments is often poor.
○ Emerging therapies, including targeted therapies that specifically inhibit the function of the BRD4-NUT fusion protein, are under investigation.
○ Surgical Resection:
□ When feasible, surgical removal of the tumor is a preferred option. The goal of surgery is to achieve clear margins, meaning no residual cancer cells are left at the edges of the removed tissue.
□ However, due to the aggressive nature of NMC and its tendency to occur in anatomically complex regions, complete surgical resection is not always possible.
Radiation Therapy:
□ Radiation therapy is often used as part of the treatment plan, either postoperatively (adjuvant therapy) to target residual disease or as a primary treatment when surgery is not feasible.
□ It can help control local tumor growth and provide symptom relief, especially in cases where the tumor is causing pain or compressing vital structures.
Chemotherapy:
□ Chemotherapy regimens used for other aggressive carcinomas are often employed in treating NMC. This might include combinations of drugs like cisplatin or carboplatin with etoposide, or doxorubicin and cyclophosphamide.
□ Chemotherapy can be used before surgery to shrink the tumor (neoadjuvant therapy), after surgery to eliminate residual disease (adjuvant therapy), or as the main treatment when surgery isn’t an option.
Targeted Therapy and Emerging Treatments:
□ Given the role of the BRD4-NUT fusion protein in NMC, targeted therapies that inhibit this fusion protein are being explored.
□ BET (bromodomain and extra-terminal motif) inhibitors, which target proteins like BRD4, have shown promise in preclinical studies and early-phase clinical trials.
□ Other emerging treatments are being investigated, including new drug combinations and immunotherapy.
Clinical Trials:
□ Participation in clinical trials can be a crucial option for patients, providing access to novel therapies that are not yet widely available.
□ Clinical trials for NMC often explore new drugs or drug combinations, targeted therapies, and other innovative treatment strategies.
Palliative Care:
□ In advanced stages, palliative care becomes an important aspect of treatment, focusing on managing symptoms and maintaining quality of life.
□ This includes pain management, nutritional support, and addressing psychological and social needs.

118
Q

How would you work up a Sinonasal tumour?

A

Consultation

- History
	○ HPI
		§ Headache and facial pain
		§ Epistaxis
		§ Nasal obstruction
		§ Chronic sinusitis
		§ Cranial nerve invasion (diplopia, facial sensory change)
		§ Ear effusion
		§ Palpable neck mass
	○ PMHx:
		§ Immunosuppression
		§ Previous H+N malignancy
		§ Previous RT
	○ Meds
		§ Immunosuppressants
	○ Social:
		§ Employment (construction/carpenters)
		§ Smoking
		§ Alcohol

- Examination
	○ General inspection
	○ Cervical nodal palpation
	○ Nasoendoscopy
		§ Examine entire pharynx (synchronous lesions)
	○ Cranial nerve examination

Work-Up

- Biopsy
	○ Biopsy any equivocal neck nodes (unless already proceeding with neck dissection)

- MR Head and Neck (must include base of skull)
	○ Surgical evaluation

- FDG-PET
	○ Nodal and distant staging
	
- Diagnostic CT Head and Neck
	○ Evaluate bony invasion

- Pre-chemotherapy bloods
	○ FBC
	○ EUC, CMP
	○ LFT and coags
	○ HepB sAg, sAb, cAb
	○ Hep C serology
	○ HIV serology
119
Q

What is the staging for sinonasal tumours?

A
120
Q

What are the general management options for early and advanced stage sinonasal tumours?

A
  • In general, options may include
    ○ Observation for nasal cavity T1 g1 SM-
    ○ Early Stage (stages I-II)
    § Surgical resection alone (+/- adjuvant RT)
    § Radiotherapy alone (uncommon)
    ○ Advanced Stage (stages III-IV)
    § Surgical resection + adjuvant RT
    § Chemoradiotherapy
    • Always establish relevant supportive care early
      ○ Smoking cessation
      ○ Dietician and speech pathology input
      § Consideration of prophylactic gastrostomy
      ○ Dental input
      § Pre-RT OPG
      ○ Social & psychological supports as appropriate
121
Q

When managing sinonasal cancer, what are the indications for adjuvant radiotherapy, adjuvant chemoRT and definitive chemoRT?

A
  • Indication for adjuvant RT
    ○ Ethmoid sinus: ALL except for T1N0, completely excised with no adverse features
    ○ Maxillary sinus:
    § T3/T4
    § N+
    § pos Margin (*Always ask surgeons to re-resect positive margins.)
    § Adenoid cystic
    § PNI/LVSI+
  • Adj concurrent chemoRT for pt with close/pos margin and ENE
  • [Bible] Elective LN RTx due to high rate LN relapse if LNs not electively treated (controversial).
    • Coverage of node-negative neck is controversial: Consider if G3, ENB, pT3-4, maxillary sinus or invasion of palate, NPX, skin of cheek or maxillary alveolar ridge.
    • Volume:
      § Cover RP and IB-IV, IF node positive.
      § Consider elective coverage of RP and IB-II, IF maxillary T3-4 or undifferentiated histology.
    • If adenoid cystic, you do not need to cover neck due to a low probability of nodal spread.
  • Definitive chemoradiotherapy
    • T4b Intracranial extension,
    • or T3-4a high grade
122
Q

Discuss management of Sinonasal undifferentiated carcinoma and small cell carcinoma.

A
  • Both surgical resection and definitive chemoradiotherapy give similar survival outcomes
    • Role of surgery, therefore, is:
      ○ If resectable with endoscopic approach only
      ○ Aim to reduce RT dose to adjuvant
    • Do not proceed with any radical disfiguring surgery
    • IF disease is resectable:
      ○ Maximal safe surgery -> RT +/- CDDP (Sx aim to cytoreduce to decr dose of Adj RT)
      OR
      ○ Induction CDDP-based chemo-> surg-> PORT
      § MDACC retro study showed that induction chemo improved 5yr DSS compared to no induction chemo, esp those who had good response
    • If disease is unresectable:
      1. Upfront induction chemotherapy
        § 3x cisplatin/etoposide
      2. If excellent response
        § Chemoradiotherapy
      3. If poor response
        § Surgical management
        § Adjuvant radiotherapy
    Note: RT should cover bilateral 1b and 2 nodes
123
Q

What is the general management of non-sinonasal undifferentiated tumours, in the sinonasal region?

A
  • Preference is for surgical resection of primary tumour
    • Most patients will require adjuvant radiotherapy
      ○ Consider surgery alone if low-risk T1N0 (clear margins, no PNI and low-grade histology)
    • Almost all patients will require bilateral elective neck management
      ○ Consider omission if low-grade T1-T2 N0
    • If unresectable, proceed with concurrent chemoradiotherapy (as per normal H+N SCC)
      ○ 70Gy/35F with cisplatin
    • Can consider induction chemotherapy (however limited data to support)
      ○ Cisplatin/5-FU
124
Q

Describe the management of Esthesineuroblastoma/ olfactory neuroblastoma.

A
  • These tumours are quite chemo and radiosensitive
    ○ Benefit is especially seen for higher-grade lesion
    • Despite this, without surgery these tumours have high recurrence rates
    • General treatment paradigm is dependent on local extent of disease
      ○ Early-Stage (Kadish A)
      § Endoscopic surgical resection (preferred)
      § Definitive RT alone (70Gy)
      ○ Intermediate-Stage (Kadish B)
      § Endoscopic surgical resection
      □ Adjuvant RT may be required (60Gy)
      § Definitive RT alone is an alternative
      ○ Advanced-Stage (Kadish C+D)
      § Neoadjuvant chemoradiotherapy
      □ 45-50Gy with concurrent Cisplatin + Etoposide
      § Surgical resection + Adj RT (for Kadish C)
125
Q

Describe management of nasal vestibule cancer

A

NASAL VESTIBULE CANCER
- Covered with skin (nostrils to the internal nasal valve) → mostly SCC w occasional BCC, melanoma, NHL.
- LN spread→ ipsilateral facial nodes and Level Ib
- Can invade cartilage, skin, lip, nasal cavity, premaxillary space
- Without elective LN Rx → 15% nodal relapse.
- Usually present as nodules, may ulcerate, locally invade and cause pain.
- RT usually preferred → better cosmesis.
- Occult LN spread rare if < 2cm, but up to 40% if larger.
○ Primary <2cm → Rx primary alone
○ Primary > 2cm→ Rx primary + elective LN RT.

RT technique
- Will need to bolus primary area, facial node and upper neck.
- Mark CTV border on skin with wire.
- Head and shoulder mask, chin neutral and both nasal cavities filled with wax bolus for heterogeneity. Sim with bite block to displace tongue posteriorly and lead shield anterior to the maxillary alveolar ridge.
- CT scan with IV contrast, fuse with MRI if available.
- Target volumes
○ GTV = clinically/radiologically evident dx
○ CTVp = nasal vestibule (+ 2cm margin if PNI)
○ CTVn = GTVn+5mm
○ CTVn0 = facial LN, Lv 1b+2-4
○ PTV70 = CTVp+n +3mm
○ PTV56 = CTVn0 +3mm
Dose prescription- 70Gy/35F,63Gy/35Fr, 56Gy/35F to clinically N0 areas.

126
Q

What would be you definitive radiotherapy technique for a sinonasal cancer?

A

Definitive Therapy

Patients
1) Stage I-II (no chemotherapy)
2) Stage III-IV (concurrent chemotherapy)

Pre-simulation
MDT Discussion (review path)
Dietician + speech path involvement
- Consider prophylactic PEG
OPG + Dental review
Audiometry
Smoking cessation

Simulation
Supine with thermoplastic mask
- Handgrips
- Vacbag and ankle/knee-blocks

Bolus in nasal cavity
Consider intra-oral tongue device

CT (2mm with IV contrast)
- Vertex to mid chest

Fusion
PET-CT and MR fusion

Dose prescription
SIB technique
- 70Gy/35F to the involved region
- 63Gy/35F to the high-risk region
- 56Gy/35F to the elective nodal regions

Concurrent chemotherapy (if stage III-IV)
- Cisplatin 100mg/m2 q3wk
- Cisplatin 40mg/m2 q1wk

VMAT technique
10 days per fortnight
- Accelerate to 6F per week if no chemotherapy

Volumes
GTVp = macroscopic primary disease
GTVn = macroscopic nodal disease
CTV70
- (GTVp + GTVn) + 10mm
CTV63
- GTVp + 20mm
- Entire involved sinus
- GTVn+10mm
CTV56
- Elective nodal regions
- Nerve pathways e.g. V2 to BOS (especially if adenoid cystic)
PTV
- CTV + 5mm

Nodal regions
For nasal cavity:
- 1st echelon: Lv 1b-2, 7a
- 2nd echelon: Lv3-4
- include Lv9 if involving anterior subsites
For Paranasal sinus:
- 1st echelon: Lv 1b-2a, 7a
- 2nd echelon: 3, 4

Unilateral nodal coverage unless lesion is crossing midline, SNUC or ENB [bible]

Target Verification
Daily CBCT

OARs
Brainstem + Optic Nerves/Chiasm
- Dmax < 54Gy
Temporal Lobe
- Dmax < 60Gy
Spinal Cord
- Dmax < 45Gy
Cochlear
- Mean < 45Gy
Lacrimal Gland
- Dmax < 30Gy
Eye
- Mean < 35Gy
- Dmax < 50Gy
Lens
- Dmax < 7Gy
Parotid (spare 1 gland)
- Mean < 26Gy (optimal)
Oral cavity
- Mean < 30Gy
Mandible
- Dmax < 70Gy
Larynx
- Mean < 45Gy
Oesophagus
- Mean < 35Gy
Pharyngeal constrictors
- Mean < 50Gy

127
Q

What would be your adjuvant radiotherapy technique for sinonasal cancer?

A

Adjuvant Therapy
Patients
1) Intermediate risk (RT alone)
* Close margins
* T3/T4 disease
* 2 or more LN involved (i.e. N2+ disease)
* PNI/LVI
2) High Risk (concurrent chemotherapy)
* Positive margins
* ECE

Pre-simulation
MDT Discussion (review path)
Dietician + speech path involvement
- Consider prophylactic PEG
OPG + Dental review
Audiometry
Smoking cessation
Simulation Supine with thermoplastic mask
- Handgrips
- Vacbag and ankle/knee-blocks

Wire any surgical scars
Bolus in nasal cavity
Consider intra-oral tongue device

CT (2mm with IV contrast)
        - Vertex to mid chest

Fusion
Pre-op PET-CT and MR fusion

Dose prescription
SIB technique
- 66Gy/33F to gross disease/positive margin
- 62Gy/33F to Tumour bed
- 58Gy/33F to Surgical bed
- 54Gy/33F to the elective nodal regions + elective nerves
VMAT technique
10 days per fortnight

Concurrent chemotherapy (if positive margin or ECE)
- Cisplatin 100mg/m2 q3wk
- Cisplatin 40mg/m2 q1wk

Volumes Pre-op GTVp = based on original MR
GTVp = residual macroscopic disease

CTV60 = pre-op GTVp + 10mm
- If gross residual –> CTV66 = GTVp + 10mm
CTV54
- Surgical bed
- Include entire involved sinus
CTV50
- Elective nodal regions (generally ipsilateral Ib, II, III, VIIa, IX)
- Nerve pathways to BOS (e.g. V2)
*treat contralateral neck if crossing midline or N+
PTV = CTV + 5mm
Target Verification Daily CBCT

OARs
Brainstem + Optic Nerves/Chiasm
- Dmax < 54Gy
Temporal Lobe
- Dmax < 60Gy
Spinal Cord
- Dmax < 45Gy
Cochlear
- Mean < 45Gy
Lacrimal Gland
- Dmax < 30Gy
Eye
- Mean < 35Gy
- Dmax < 50Gy
Lens
- Dmax < 7Gy
Parotid (spare 1 gland)
- Mean < 26Gy (optimal)
Oral cavity
- Mean < 30Gy
Mandible
- Dmax < 70Gy
Larynx
- Mean < 45Gy
Oesophagus
- Mean < 35Gy
Pharyngeal constrictors
- Mean < 50Gy

128
Q

What elective lymph node regions would you cover for sinonasal cancers?

A
129
Q

What is the expected 5 year OS by stage and by histology

A
130
Q

Describe the importance of oral care in radiotherapy and ways to prevent ORN.

A
  • Relevant RT S/E:
    Impaired vascularity of mandible/maxilla and risk of osteoradionecrosis
    Direct damage to teeth in high dose region of field (>60Gy) esp 70gy
    Xerostomia, which increases the risk of dental caries
    • ORN patient risk factors:
      ○ Poor dentition, alcohol, smoking
      ○ Immunodefeciency
      ○ Inciting trauma
    OPG + dental review
    * Removal of bad teeth in high dose region
    * Allow 2 weeks to heal
    * Rationale: To decrease risk of ORN, teeth with risk factors should be removed prior to receiving doses of 60Gy
    * Patients compliance with daily ora hygiene, regular dental appointments
    * Post-RT dental care
    Fluoride tray treatment
    Artificial saliva to counter carious effects of xerostomia
    Regular dental reviewORN prevention
    Wait 2 weeks after extraction before starting RT
    Stop smoking
    Wait 9 months post RT to make dentures
    Prophylactic HBO pre- and post tooth extraction (30% vs 5%)
130
Q

Describe the pathogenesis of osetoradionecrosis (4 marks)

A

Definition: exposed non-vital bone in previously irradiated area with no evidence of residual or recurrent disease.

Vascular obliteration with endarteritis, fibrosis and thrombosis of inferior alveolar artery, decreased vascular supply of bone, leading to tissue hypoxia and fibrosis
Direct damage to osteoclasts/osteoblasts

Often precipitated by injury, infection, tooth extraction
Symptoms: pain, hallitosis, dysgeusia, trismus, fistula
Grade 1 = exposed alveolar bone
2 = exposed not responding to HBO
Gr 3 = full thickness or fracture
If there is osteomyelitis on imaging, assume this is overlying ORN.

Risk 4 months to 3 years ~3% risk. Doses <60Gy did not occur, 60-70Gy 1.8% and >70Gy 9%.

131
Q

Discuss the management of osteoradionecrosis

A

Asssesment
Examination with dentist + surgeon, OPG
CT -soft tissue thickening, bony changes -cortical bone erosion, fragmentation
Specific to ORN -bone sclerosis, intraosseus gas
Recurrence -solid or cystic mass
MRI -soft tissue thickening
Can tell viable vs nonviable bone via marrow signal on T1

Chondronecrosis
	CT fragmentation/collapse of cartilage
	MRI loss of high cartilage T1 signal.

Management of ORN

o Conservative measures can include:
o analgesia and anti-inflammatory medication for pain
o antibiotics for acute infections
o good oral hygiene
o removal of devitalised tissue (sequestrectomy)
o anti-radiation fibrosis drugs may be useful in the management of established ORN
o Pentoxifylline + Vitamin D
o Surgery
o When conservative management fails and there is progressive ORN (Notani Stage III) resulting in pathological fractures and draining fistula, surgery is typically used to manage the condition.
* Debridement via bone curettage -also excludes malignancy
* Sequestrectomy, fistula closure,
* Graft reconstruction -high risk of complications, fistulas, irradiated vessels less viable for flap attachement.
o Medical: Pentoxyfylline and vitamin E (multiple small prospective/retrospective trials
400 mg of PTX twice a day with 1000 IU of tocopherol for at least 6 months

o  o Hyperbaric oxygen therapy
o Widespread adoption of HBOT as an addition to surgery for ORN has evolved, however the validity of the supporting data is still undetermined. A 2022 study combined data from two randomised clinical trials (DAHANCA-21 and NWHHT2009-1) and found that complementary HBOT improved ORN healing (70%) when compared to surgical debridement alone (51%), but not significantly, as the studies were underpowered. 
o statpearls: 30 sessions prior to tooth extraction/ORN debridement and 10 sessions after
132
Q

What are the periorbital complications from radiotherapy?

A
133
Q

A 48 year old female presents with a six month history of severe oral pain radiating to her jaw. Examination demonstrates an ulcerated lesion on the left lateral edge of the anterior two thirds of the tongue.
a. List the key investigations required to work up this patient. (1 mark)

A

· Bloods: FBC, EUC, CMP, Coags
· Biopsy of tongue +/- equivocal neck lymph nodes
· CT H&N
· OPG
· PET
· MRI Head
· Audiometry

134
Q

A 48 year old female presents with a six month history of severe oral pain radiating to her jaw. Examination demonstrates an ulcerated lesion on the left lateral edge of the anterior two thirds of the tongue.
Pathology from a left hemi glossectomy and ipsilateral neck dissection confirms a 28mm poorly differentiated discohesive SCC with a depth of invasion 11mm, medial margin 1mm, perineural invasion of lingual nerve and 2/16 nodes, both at level 2, with no extra nodal extension. The decision is made to treat the patient with radical radiation therapy. b. Describe in detail your: (4 marks) i. clinical target volumes, ii. dose fractionation schedule, and ili. organs at risk dose constraints.

A

T3N2M0 (Stage IV)
· Clinical target volumes
o CTV60: Tumour bed: Pre-op GTV +5mm. V3 to foramen ovale if margin positive
o CTV 54: Pre-op-GTV +10mm + Surgical bed and nodal bed, including flaps, scar. Ipsilateral undissected neck and Right neck level 1-5b, Path of lingual nerve/V3 back to foramen ovale. Cover inferior alveolar nerve and its Anterograde path (Given jaw pain)
o PTV: 5mm expansions of above
· Dose fractionation
o 60Gy/30# to Surgical bed
o 54Gy/30# to bilateral neck and anterograde/retrograde coverage of V3 to foramen Ovale
o 10 days per fortnight, VMAT technique, complete treatment within 6 weeks
· OARs
o Brainstem + Optic Nerves/Chiasm
o Dmax < 54Gy

o Cochlear
		o Mean < 45Gy
 
o Lacrimal Gland
		o Dmax < 30Gy
 
o Eye
		o Mean < 35Gy
		o Dmax < 50Gy
 
o Lens
		o Dmax < 7Gy
 
o Parotid (spare 1 gland)
		o Mean < 26Gy (optimal)
		o Submandibular gland: mean dose <39Gy
		o  
o Mandible
		o Dmax < 70Gy
 
o Larynx
		o Mean < 45Gy
 
o Oesophagus
		o Mean < 35Gy
 
o Pharyngeal constrictors
		o Mean < 50Gy
135
Q

A 48 year old female presents with a six month history of severe oral pain radiating to her jaw. Examination demonstrates an ulcerated lesion on the left lateral edge of the anterior two thirds of the tongue.
After three weeks of Adjuvant radiation treatment the patient requires a replan for weight loss. This results in a treatment break of 4 treatment days. C. What are the management options for this patient? Justify your answer. (2 marks)

A

Extending the overall treatment time can result in tumour repopulation and decrease local control. Management options to account for this would be to:
o If dose overlay has tumour within ptv and critical oar within tolerance, keep using old plan until new plan ready.
o Keep the same over all treatment time by accelerating the remaining treatment
o BD treatments (6 hrs apart)
o 6 fractions a week
o Increase the overall dose by 3Gy to account for delay
o Accept the potentially reduced local control and continue with the treatment as planned
o Other
o Dietitian review, consider enteral feeding
o Regular treatment reviews
o Manage pain

136
Q

Discuss the optimal management of ORN (3 marks)

A

o Conservative measures can include:
o analgesia and anti-inflammatory medication for pain
o antibiotics for acute infections
o good oral hygiene
o removal of devitalised tissue (sequestrectomy)
o anti-radiation fibrosis drugs may be useful in the management of established ORN
o Pentoxifylline + Vitamin E
o Surgery
o When conservative management fails and there is progressive ORN (Notani Stage III) resulting in pathological fractures and draining fistula, surgery is typically used to manage the condition.
o Hyperbaric oxygen therapy
Widespread adoption of HBOT as an addition to surgery for ORN has evolved, however the validity of the supporting data is still undetermined. A 2022 study combined data from two randomised clinical trials (DAHANCA-21 and NWHHT2009-1) and found that complementary HBOT improved ORN healing (70%) when compared to surgical debridement alone (51%), but not significantly, as the studies were underpowered.

137
Q

A fit 60-year-old male is diagnosed with a locally advanced squamous cell carcinoma of the glottic larynx with bilateral cervical lymph nodes. There is no evidence of distant metastatic disease. (Stage T3N2cM0)
Discuss the advantages and disadvantages of the different treatment strategies available for locoregionally advanced laryngeal carcinoma (Stages IlI and IV). (5 marks)

A
138
Q

List the factors which increase the risk of stomal recurrence in patients receiving post-operative radiotherapy after total laryngectomy for laryngeal cancer. (3 marks)

A
  • Pre-operative tracheostomy (especially if urgent)
    • Advanced T stage/ Locally advanced disease (involvement of anterior neck)
    • Subglottic disease location
    • Positive surgical margins
    • Pre and paratracheal LN involvement
    • PNI
    • Previous tracheostomy
    • Tumour seeding
      Delay with starting adjuvant treatment
139
Q

List the factors which increase the risk of laryngeal necrosis with definitive radiotherapy/chemoradiotherapy for larynx cancer. (2 marks)

A
  • Alcohol and smoking
    • Tumour invasion
    • Post-operative infection
    • Trauma/ infection
    • Radiation Technique
    • Laryngeal dose/ hot spots
    • Chemotherapy
    • Weight loss during treatment
140
Q

A fit 55 year old man is diagnosed with nasopharyngeal carcinoma. PET/CT shows intracranial extension of the tumour and right sided level 2-4 lymphadenopathy. He has no distant metastases (cT4N3M0).

a. In general, what treatment strategies are available for locoregionally advanced nasopharyngeal carcinoma? Include in your answer the rationale of each approach. (2 marks)

A

· Induction chemotherapy -> Concurrent Chemo/Radiotherapy
o Larger primary and more advanced nodal disease both increase the risk of distant metastasis – upfront chemotherapy can assist with decreasing this risk
o May decrease bulky disease and reduce dose to critical OARs by the time radiotherapy is scheduled
o Options for induction chemo –gem/cis, cis/5FU/docetaxel
o Studies show improved OS, DMFS and LRC compared to ChemoRT alone
o Treat micrometastatic disease

· Concurrent chemo/radiotherapy -> Adjuvant chemotherapy
o Improves OS compared to concurrent chemoRT alone
o Still not clear if this has any different outcomes to induction chemotherapy approach
o The additional chemotherapy improves distant and locoregional control
· ChemoRT alone
o Not recommended due to the inferior OS, DMFS and DFS
· RT alone -inferior
· Surgery -inferior as high surgical morbidity and unable to dissect retropharyngeal nodes. Unlikely clear margins

141
Q

A fit 55 year old man is diagnosed with nasopharyngeal carcinoma. PET/CT shows intracranial extension of the tumour and right sided level 2-4 lymphadenopathy. He has no distant metastases (cT4N3M0).

The recommendation from the MDT is to treat the patient with definitive chemoradiotherapy. Describe your target volumes, radiation therapy technique and dose fractionation schedule. (3 marks)

A

· Pre-SIM
o Induction chemotherapy would have been recommended, but assumed this was not given in this case
o MDT discussion
o Dietitian/speech pathologist
o Dental review
o Audiometry
o Smoking cessation
o Prophylactic PEG
· Simulation
o Supine with thermoplastic mask
o Vacbag, knee block, ankle stocks
o CT from vertex to mid chest with 2mm slices + IV contrast
o Fusion with PET and MRI
· Dose prescription
o 70Gy/35# to Involved disease on PET/MRI/CT
o 63Gy/35# to high risk region
o 56Gy/35# to elective nodal region
o 10 fractions a fortnight, VMAT technique, 6MV photons
o Concurrent cisplatin chemotherapy 40mg/m2 weekly
o Daily CBCT for target verification
· Volumes
o GTVp= macroscopic primary disease
o GTVn= macroscopic nodal disease
o CTV70= GTVp + GTVn + 5mm margin + remaining nasopharynx to 5mm depth
o CTV63= GTVp + GTVn + 10mm. Including all of nasopharynx, nasal cavity (posterior 5mm), maxillary, ethmoid, sphenoid sinuses, skull base, pterygoid plates and fossa, parapharyngeal space, Clivus, ipsilateral cavernous sinus
§ Clip to anatomical structures
o CTV56= bilateral neck lvl II, III, IVa, Va/b, VIIa/b. On ipsilateral side also cover 1b and Ivb, Vc
o PTV= CTV + 5mm

142
Q

A fit 55 year old man is diagnosed with nasopharyngeal carcinoma. PET/CT shows intracranial extension of the tumour and right sided level 2-4 lymphadenopathy. He has no distant metastases (cT4N3M0).
This is managed with chemoRT.

Two years later the patient is found to have a 1.5cm recurrence in the lateral wall of the nasopharynx. What further information do you require to formulate a management plan for this patient? (2 marks)

There are no distant metastases. The decision is made to offer this patient re-irradiation with radical intent. Describe a suitable radiation therapy treatment and dose fractionation schedule, including critical normal tissue tolerances. (3 marks)

A

· History and physical examination
o Any Grade 1+ toxicity of critical organs or Grade 3+ toxicity of non-critical organs
· Symptoms, cranial nerve involvement
· MRI to determine local extent
· PET to assess locoregional/ distant metastasis
· Fitness/ suitability for surgery
· Performance status
· Biopsy confirmation
· Previous dose to OARs

· Pre-SIM
o MDT discussion
o Dietitian/speech pathologist
o Dental review
o Audiometry
o Smoking cessation
· Simulation
o Supine with thermoplastic mask
o Vacbag, knee block, ankle stocks
o CT from vertex to mid chest with 2mm slices + IV contrast
o Fusion with PET and MRI
· Dose prescription
o 65Gy/54# BD to Involved disease on PET/MRI/CT
o 10 fractions a week (6hrs apart), VMAT technique, 6MV photons
o Daily CBCT for target verification
· Volumes
o GTVp= macroscopic primary disease
o CTV65= GTVp 5mm margin
o PTV65= CTV65 + 3mm
· OARs
o Spinal cord= Dmax 60Gy
o Brain stem/ Optic nerves= Dmax 70Gy
o Temporal lobe= Dmax 90Gy
o Carotid artery= Dmax 125Gy

142
Q

A 56 year old man presents with proptosis and opthalmoplegia of the left eye. A CT scan demonstrates a tumour of the left maxillary antrum involving the floor of the orbit. Biopsy confirms a poorly differentiated squamous carcinoma. There is no lymphadenopathy.

a) What further investigations would you recommend? (1 mark)

b) In general, what are the relative contraindications to surgery when treating maxillary antral tumours? (2 marks)

A

o Nasoendoscopy of entire pharynx to exclude synchronous lesions.
o Bloods – FBC, EUC, LFT, CMP, hep B/C serology, HIV
o Visual assessment
o CT Neck
o PET
o MRI Head
o OPG
o EUA

General factors that preclude surgery include poor performance status, life expectancy, co-morbidities and patient preference. Brain invasion and Orbital invasion are relative contraindications to maxillary surgery. However open, disfiguring surgery may still be an option.

143
Q

A 56 year old man presents with proptosis and opthalmoplegia of the left eye. A CT scan demonstrates a tumour of the left maxillary antrum involving the floor of the orbit. Biopsy confirms a poorly differentiated squamous carcinoma. There is no lymphadenopathy.

The patient undergoes maxillectomy and orbital exenteration. The histological margins are clear with the exception of focal involvement of the posterior margin of the maxilla, and extensive perineural infiltration of the infraorbital nerve. Post-operative radiation therapy is recommended.

c) Describe a suitable radiation technique and dose prescription for this man’s treatment. (4 marks)

e) What is the likely five year survival for this patient? (1 mark)

A

· 63/60/54gy in 30# chemoRT 10/fortnight vmat 6mv with concurrent cisplatin
· Pre-SIM
o MDT discussion
o Post op MRI
· SIM
o Supine in thermoplastic mask
o Bite block to shift mandible inferiorly
o Wire scars and bolus
o CT scan with IV contrast, 2mm slices from vertex to mid chest
o Fusion with pre-op/post-op MRI and PET
· Prescription
o 63Gy/30# to positive margin
o 60Gy/30# to Tumour bed
o 56Gy/30# to surgical bed
o 54Gy/30# to elective ipsilateral neck nodes + V2 nerve -chasing back to foramen ovals
o Concurrent cisplatin 40mg/m2 weekly
o VMAT technique 10#/fortnight, 6MV photons
· Volumes
o Pre-op GTV + Residual GTV= GTVp
o CTV63= GTVp+5mm
o CTV60= Tumour bed
o CTV56= Surgical bed, including entire maxillary sinus and surgical scars
o CTV54= Ipsilateral 1b, II, III, VIIa, IX + V2 back to BOS, ppf, vidian canal.
o PTV= CTV + 5mm

5yr OS of 40%

144
Q

A 60 year old man has a macroscopic endoscopic resection of a well differentiated SCC of the upper right nasal cavity.

The tumour is 18mm and has been resected with microscopically clear margins. There are no involved nodes (T1N0M0). The decision has been made to treat with adjuvant radiotherapy. b. Describe a suitable radiation therapy technique and dose fractionation schedule. (3 marks)

A

60/54Gy/30F 5 fractions/week VMAT 6MV photons with daily CBCT
(63Gy if positive margin or ECE)
Presim: dental + OPG
Sim: supine, head mask, consider bolus and nasal packing if tumour anterior. Biteblock to move tongue inferior, CT vertex to mid thoracic,
Fuse preop imaging to define tumour -MRI, PET
Fuse postop imaging -MRI
Volumes: CTV60 = surgical bed + 1.5cm
CTV 54: Nodes -If T3-T4 or LVSI cover bilateral 1b and 2, 7a
Cover nerve to base of skull (VII, V1, V2) if microscopic PNI

144
Q

A 60 year old man has a macroscopic endoscopic resection of a well differentiated SCC of the upper right nasal cavity.
a. How would you further evaluate this patient and what further information do you require? (2 marks)

A

H+e
Bleeding, pain, post op healing, smell, obstruction, hearing, vision/cataracts
Comorbidities, previous cancers/treatments, smoking, social, access, age performance status, immunosuppression,
Cranial nerve function: v1, v2, v3, vii , iii, iv, vi

Tumour:
Margins, PNI, invasion of sinuses/bone, EBV, HPV
FNE for other mucosal tumours/dysplasia
Staging -nodes, tumour size, ENE: PET, MRI

Treatment -contraindications to RT
Need for bolus if superficial
Dental review, OPG
Nutrition

145
Q

A 60 year old man has a macroscopic endoscopic resection of a well differentiated SCC of the upper right nasal cavity. He has Adjuvant RT.

Four years later the patients re-presents with a local recurrence limited to the upper nasal cavity. d. What options are available for further management? (2 marks)

A

Surgery
Chemo/immuno therapy
Reirradiation (with concurrent chemotherapy)-30Gy/10# - 60Gy/30# depending on evidence of late toxicity, and critical OAR doses from the initial irradiation. Must accept risks of late toxicity eg visual loss
Can also consider hyperfraction 60Gy/40# BD RT which has limited data but less late toxicity
Clinical trial - Could use brachytherapy, proton therapy or SRS
Palliative Systemic therapy –
If TPS>50% Pembrolizumab +- platinum/5FU
Chemo +- cetuximab
Taxane
Best supportive care

146
Q

A 70 year woman. old woman presents with a hard 3cm x 4cm x 4cm mass in the right parotid

a. What are the differential diagnoses? (2 marks)

A

Malignant:
SCC -cutanous met
adenoidcystic carcinoma
Mucoepidermoid carcinoma
Carcinoma ex pleomorphic adenoma
Lymphoepithelilal carcinoma
Acinic cell carcinoma
adenocarcinoma
Lymphoma, sarcoma, melanoma (met)
Benign:
Warthin tumour, Pleomorphic Adenoma, oncocytoma
Infection/sialdenitis/abscess

147
Q

A 70 year woman. old woman presents with a hard 3cm x 4cm x 4cm mass in the right parotid.

a. An FNA indicates metastatic Squamous Cell Carcinoma. What factors should be considered in determining her management? Justify your answer. (3 marks)

A

Pt -smoking, comorbidities, contraindications for surgery/radiotherapy/chemotherapy
a. Previous skin cancers (likely primary) and previous surgery
b. Immunosuppression
c. Social, access, comorbidity
Tumour -size of primary, location, intradermal lymphatics/satellite, PNI, hair bearing areas, bone/cartilage invasion, suitability for flap reconstruction
Grade and thickness
Subtype of SCC

PNI esp facial nerve. determine extent of surgery/RT
	Facial Numbness, pain, weakness, ear pain
Nodes: number, size, ECE -determine adjuvant treatment Rx: resectability re relation to facial nerve and anticipated postop deficits, need for flap and assess donor sites.
148
Q

A 70 year woman. old woman presents with a hard 3cm x 4cm x 4cm SCC in the right parotid.

The patient declines surgery and the decision is made to treat with curative radiation therapy alone. C. Describe a suitable radiation therapy technique and dose fractionation schedule. Justify your treatment volumes. (3 marks)

A

60Gy/60/56Gy in 30# 5-6 fractions per week with VMAT 6MV with CBCT.
Dose as per ICRU 83 to 95% of PTV
Fuse MRI and PET
CTV63 = GTVp and GTVn + 5mm = high dose to macroscopic disease
CTV 60 = GTV+10mm, and include entire parotid gland
CTV 56 = elective nodes = ipsilateral level 2, 3,4 . If microscopic PNI also elective irradiation of CNVII to stylomastoid foramen, or if macroscopic then include internal auditory meatus.

149
Q

A 70 year woman. old woman presents with a hard 3cm x 4cm x 4cm SCC in the right parotid.
She has RT alone.

What is the: (2 marks) i. likely acute and late toxicity? ii. what is the likelihood of locoregional control and how would this compare to other treatment options?

A

Acute: dermatitis, mucositis, pain, dysphagia, xerostomia, conjunctivitis, dry eye
Late: dry eye, xerostomia, cataract, trismus, ORN, cosmetic skin changes
hypopituitarism, neck lymphoedema fibrosis, hypothyroidism, alopicea, temporal lobe necrosis

LRC: 60-70%.
Surgery + adjuvant RT superior ~80%
Surgery alone 40%

150
Q

A 64 year old woman presents with an enlarged right tonsil. Biopsy shows moderately differentiated, p16 negative squamous cell carcinoma. Examination, CT scan and PET/CT show a 20mm primary confined to the tonsillar fossa, no lymphadenopathy and no distant metastases (T1N0MO).

a. The decision is made to treat with definitive radiation therapy. Describe a suitable radiation therapy technique and dose fractionation schedule. (3 marks)

A

Prescription: 3 dose levels 70 Gy/ 63 Gy/56 Gy in 35#
VMAT technique. 6 MV photons.
Presim: dental
SIM: Supine in thermoplastic mask with hands by sides in a vac bag. CT with IV contrast.
Fusion: PET + MRI if available.
Volumes:
GTVp- macroscopic disease as defined by clinical examination, CT and PET +/- MRI imaging
CTV70: GTVp + 5mm
CTV63: GTVp + 10 mm
CTV low risk 56 Gy: elective nodes ipsilateral neck Level II, III, IV

151
Q

A 64 year old woman presents with an enlarged right tonsil. Biopsy shows moderately differentiated, p16 negative squamous cell carcinoma. Examination, CT scan and PET/CT show a 20mm primary confined to the tonsillar fossa, no lymphadenopathy and no distant metastases (T1N0MO).

She is treated with RT alone.
What acute toxicities would you expect and how would you manage these? (3 marks)

A

Mucositis: Regular monitoring during treatment. Supportive management with xylocaine viscus and analgesia (paracetamol and opioid based). May require hospital admission for supportive management and alternative nutritional optimisation for PO intake (PEG or NG).
Oral Thrush: Regular monitoring during treatment. Nilstat drops or amphotericin lozenges if needing to escalate anti-fungal treatment.
Radiation dermatitis: Erythema, itch with dry or moist desquamation. Start with topical sorbolene cream. With desquamation of skin change to solosite gel + dressings for comfort. Monitor for signs of infection when evidence of desquamation. Optimise analgesia with simple analgesia and opioid based.
Xerostomia: Supportive strategies including chewing gum and encouraging water intake.
Dysgeusia: Supportive management, reassurance that is an expected side effect and can last months after treatment.
Odynophagia/dysphagia: Regular speech pathology review. Monitor for signs of aspiration and monitor chest for evidence of pneumonia.

152
Q

A 64 year old woman presents with an enlarged right tonsil. Biopsy shows moderately differentiated, p16 negative squamous cell carcinoma. Examination, CT scan and PET/CT show a 20mm primary confined to the tonsillar fossa, no lymphadenopathy and no distant metastases (T1N0MO). She is treated with RT alone.

The patient has a complete response to treatment but two years develops a 35mm Level lI right sided neck mass. Biopsy confirms squamous cell carcinoma. This is an isolated local nodal recurrence.
c. What are treatment options for this patient? Include in your answer a discussion of the aim, the risks and limitations of each treatment option. (4 marks)

A

· Neck dissection
o Surgical management would be preferred salvage treatment option in this patient, to preserve anatomical function and avoid effects of re-irradation.
o Aim for R0 resection for involved lymph node, in addition to analysis of draining lymph nodes stations for the detection of micrometastatic disease
o Risks: Surgical complications: Infection, air leak, post-operative bleeding, chylous fistula (<1%), carotid artery rupture (often lethal), nerve injury- spinal accessory nerve with pain/weakness of shoulder
o Limitations: Performance status, age and co morbidities can be contraindications for surgery. Neck dissection more difficult to perform with post-Rt tissues due to fibrosis. Tumour encasement of carotid artery or deep infiltration or the prevertebral space/deep neck muscles- unresectable disease. Should be performed in a high-volume, specialist centre due to the significant morbidity and mortality risks association with procedure.
· Systemic Therapy
o Chemo-RT in the setting of re-irradiation based on patient factors
o Immunotherapy: emerging as a monotherapy (pembrolizumab or nivolumab) in the setting of recurrent head and neck SCC with high PDL 1
o Risks: Immunotherapy toxicity: Cardiotoxicity, colitis, hepatotoxicity, arthralgias, pneumonitis
o Limitations: If lymph node does not respond to systemic therapy, would need to consider palliative management or resection/RT, which may be difficult to attempt if patient has developed side effects related to systemic therapy.
· Re-irradiation
o Would not advocate in the MDT setting without specific consideration of patient factors and consent.
o Assess degree of late toxicity g1 for critical oar, g3 for no critical
o If not resectable or a surgical candidate. Re-irradiation with or without chemotherapy is an option.
o Risks: Fatal haemorrhage (carotid artery blow out), dysphagia and neck fibrosis with long term swallowing and functional difficulties high, osteoradionecrosis of the jaw
o Limitations: Re-irradiation more feasible using modern techniques. Caution in patients treated with older fields. High volume centre with MDT support. Patient must be consented appropriately on the risks.
· Best supportive care
Would not advocate for in this patient, who is 64 with no additional co morbidities mentioned.

153
Q

A 72-year-old male presents with moderate odynophagia and dysphagia, along with 6 kg of unintentional weight loss. He is found to have a right pyriform fossa squamous cell carcinoma measuring 3 cm and involved ipsilateral lymph nodes, staged cT2N2bM0. He has several comorbidities and a long smoking history and is ECOG 2.

Discuss the potentially curative treatment options available, including the advantages and disadvantages of each. (3 marks)

A

He has Stage IVa Hypopharynx cancer. His options depend on his pre-existing larynx/hypopharynx function. They would include:
· Definitive concurrent chemoradiotherapy - Conventional 70Gy/35# + 3 weekly cisplatin
o Advantages
§ Non-invasive
§ Moderate local control and chance of larynx preservation
o Disadvantages
§ Protracted course of treatment
§ May not be suitable/ gain much benefit from chemotherapy given age, PS
§ Will need prophylactic enteral feeding
o Permanent PEG in 25% of patients
· Induction chemotherapy followed by radiotherapy
o Advantages:
§ Non-invasive
§ May be more tolerable compared to concurrent chemoRT
§ Moderate chance of local control
o Disadvantages
§ Further Protracted course of treatment
§ Lower chance of larynx preservation compared to concurrent chemoRT
§ May not be suitable/ gain much benefit from chemotherapy given age PS
§ Will need prophylactic enteral feeding
o Permanent PEG in 25% of patients
· Accelerated Radiotherapy alone
o Advantages
§ Non-invasive
§ May be more appropriate if not able to tolerate chemotherapy
o Disadvantages
§ Worse local control and larynx preservation
§ Low chance of cure
· Laryngectomy + unilateral LND
o Advantages
§ Single procedure
§ Best option if patient is not suitable for chemotherapy
§ Best option if non-functioning larynx, to reduce risk of aspiration
§ Best chance of returning to oral diet
o Disadvantages
§ Morbid/ mortality risks with surgery
§ Loss of larynx function
§ Need to care for trache
§ Post-op RT is often required

154
Q

A 72-year-old male presents with moderate odynophagia and dysphagia, along with 6 kg of unintentional weight loss. He is found to have a right pyriform fossa squamous cell carcinoma measuring 3 cm and involved ipsilateral lymph nodes, staged cT2N2bM0. He has several comorbidities and a long smoking history and is ECOG 2.

b. Which pre-treatment assessments and investigations would you require in order to recommend a treatment plan? (2 marks)

A

· Focused history
o Assessment of larynx function
§ Speech quality
§ Poor swallow/ Aspiration
§ Immobile cords
o FNE to assess remaining mucosa
o Co-morbidities
o Contraindications to Radiotherapy, chemotherapy, surgery

· Pre-chemo bloods
· MRI Head/neck
· PET
· Dental review/ OPG
· Dietitian/ speech path review

· Medical Oncology review for suitability for chemotherapy
· MDT discussion

155
Q

A 72-year-old male presents with moderate odynophagia and dysphagia, along with 6 kg of unintentional weight loss. He is found to have a right pyriform fossa squamous cell carcinoma measuring 3 cm and involved ipsilateral lymph nodes, staged cT2N2bM0. He has several comorbidities and a long smoking history and is ECOG 2.

The patient has a pharyngolaryngectomy and right neck dissection and makes an uneventful recovery. Histopathology reveals a 25 mm primary tumour of the right hypopharynx posterior wall, and two lymph nodes involved in right level Ill measuring 20 and 10 mm respectively, with no extracapsular spread. All margins are clear. The patient presents for postoperative radiation therapy. Describe a suitable radiation therapy technique and dose fractionation schedule. Note: No organ at risk constraints are required for this section. (4 marks)

A

I would prescribe 60Gy/30# (Tumour bed), 56Gy/30# to surgical bed and 54Gy/30# (Elective nodes) to the PTV as per ICRU 83. 10# a week using a VMAT technique. Daily CBCT for target verification. Fuse PET/MRI

· Pre-sim
o Ensure wound healing
o Treat 4-8 weeks post operatively
o MDT discussion and pathology review
o Dietitian speech path
o OPG, Dental review
o Smoking cessation
· SIM
o Supine in thermoplastic mask
o Vacbag, Knee block, ankle stock
o Wire scars
o CT from vertex to mid chest. IV Contrast
· Volumes
o CTV60: GTVp+ 5mm (Tumour bed) -> +5mm to PTV 60
o CTV56: GTVp + 10mm + clips (Surgical bed) -> +5mm to PTV56
o CTV54: Elective LNs (Right neck level VIIa/b, Va/b, Ib. Left neck level II-Iva, VIIa)
o CTV54 + 5mm = PTV54

156
Q

A 45 year old male presents with a mass in his neck. Biopsy shows the presence of poorly differentiated malignant cells. (3 marks)

i. List the possible differential diagnoses (primary head and neck cancers) in this setting.

A

o Mucosal head and neck SCC to Lymph node
o Oral cavity, nasopharynx, oropharynx, larynx, hypopharynx
o Cutaneous SCC to LN
o Cutaneous or mucosal Melanoma to LN
o Salivary gland cancer
o Submandibular
o Parotid to LN
o Thyroid cancer
o Lymphoma
o Plasmacytoma
o Sarcoma
o Neuroendocrine

157
Q

Describe the implications of EGFR over-expression in head and neck mucosal squamous cell carcinoma. (2 marks)

A

o EGFR is over-expressed in over 90% of head and neck cancers and is a poor prognostic indicator
o HPV+ve oropharyngeal SCCs have lower chance of EGFR mutation
o EGFR influences gene expression, proliferation, angiogenesis, apoptosis inhibition, cell motility, metastasis
o Target for therapy: Cetuximab is a monoclonal antibody targeting the extracellular domain of EGFR.
o Most of the data for the use of Cetuximab is in HPV +ve population, which are less likely to have EGFR mutations. As such it has unclear benefit in EGFR mutated head and neck cancers

158
Q

What are the diagnostic criteria for p16 positivity?

A

o Positive staining is defined as ‘block staining’
o Strong nuclear and cytoplasmic expression in continuous segment of cells (at least 10-20 cells) in squamous epithelium
o Block positivity needs to involve basal and parabasal layers
o Require >70% of cells positive

158
Q

A 45 year old male presents with a mass in his neck. Biopsy shows the presence of poorly differentiated malignant cells.

ii. How may immunohistochemistry assist in identifying the potential primary tumour?

A

o Many head and neck cancers when poorly differentiated can have a similar histological appearance, regardless of primary origin
o IHC can further characterise cancers based on both positive and negative staining of specific markers
o Eg p16 positive may make a oropharyngeal primary more likely.

159
Q

Discuss the pros and cons of fine needle aspiration in the diagnosis of epithelial parotid neoplasms and how it can guide surgical management. (2 marks)

A

· Pros
o Rapid, Simple procedure
o Cost effective
o Minimally invasive
o Generally sufficient to distinguish benign from malignant
§ Guide if superficial/ complete parotidectomy needed or just observation
· Cons
o May not be able to determine histological subtype
o May not provide enough tissue for diagnosis
o Sensitivity and specificity is dependent on operator
o May not give tissue representative of whole tumour