Thoracic Flashcards

1
Q

What is the aetiology for NSCLC?

A

1) Smoking
a. Strong link with dose-response relationship
i. Stonger link to SCC
b. Associated with 80+% cases of NSCLC
c. Lifetime risk of 30% in smokers
d. Increased risk with increased pack years
e. Risk decreases with time since quitting, approaches but never equals risk in non-smoker
f. Small increased risk with passive nosmokers

2) Environmental exposures
	a. Heavy metals 
	b. Asbestos (in addition to mesothelioma risk)
		i. X5 fold risk of lung cancer
		ii. Synergistic with smoking to give x50 fold risk
		iii. Latent period of 10-30years
	c. Air pollution
		i. Eg wood smoke. Developing countries
3) Ionising radiation
	a. Therapeutic/diagnostic radiotherapy
		i. Younger patient population at risk
	b. Radon gas eg. uranium miners
4) Familial/genetic
	a. First degree relative increases risk (RR ~1.8)
	b. Likely complex with multiple susceptibility loci
	c. No clear single inherited gene

Note that the biological, molecular and clinical characteristics of NSCLC in non-smokers is markedly different to that of smokers

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

What are the Differentials for a lung lesion (including histological subtypes)?

A

Adenocarcinoma (50%); LAMPS
○ Favourable = Lepidic,
○ Intermediate = Papillary, Acinar
○ Poor = Micropapillary, Solid
○ Other: Minimally invasive adenocarcinoma, Pre invasive (Adenocarcinoma insitu, Atypical adenomatous hyperplasia), mucinous, colloid, fetal
* Squamous cell carcinoma (20%)
○ Keratinising
○ Non-keratinising
○ Basaloid
○ Pre-invasive: SCC in situ
* Large cell carcinoma (5%)
○ Sarcomatoid
○ Pleomorphic
* Neuroendocrine
○ SCLC
○ LCNEC
○ Carcinoid
▪ Typical
▪ Atypical
* Other:
* Adenosquamous carcinoma
* Mesenchymal tumours – sarcoma
* Mesothelioma
* Salivary gland – mucoepidermoid, adenoid cystic
* Lymphoproliferative tumours – MALT, DLBCL
* Melanoma
* Germ cell tumours
* Metastatic Tumours
* Benign Epithelial Tumours
▪ - Adenoma
▪ - Papillomas
▪ - Harmatomas

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

Desccribe the pathology of lung adenocarcinoma.

A

Adenocarcinoma
- Most common subtype of NSCLC (40%)
○ Increasing incidence
- Slightly higher prevalence in females
- Presentation
○ More common in peripheral regions of lung, upper lobe> lower lobe
○ Most common metastases (brain –> bone –> liver –> adrenal), metastasise early
- Macroscopic
○ Well defined but unencapsulated lesion
○ White-tan cut surface
○ May have central scar or necrosis
○ Pleural invasion common
- Microscopic
○ Characterised by glandular formation and presence of intracytoplasmic mucin
○ Architecture can be one of (LAMPS):
- Lepidic = neoplastic cells resemble type 2 pneumocytes and proliferate to line alveolar wall (without stromal invasion)
- Acinar = gland forming cells which invade stroma
- Papillary = malignant cuboidal cells replace alveolar lining; papillary projections (fronds) with fibrovascular core
- Micropapillary = ill-defined projections which lack fibrovascular core
- Solid = sheets of neoplastic cells
○ Regardless of subtype, tend to have lepidic growth at margin
- Immunohistochemistry
○ POS = TTF1 (positive Adenocarcinonma), Napsin A, CK7, AE1/AE3 (anti-cytokeratin antibiodies)
○ NEG = CK20 (only positive in non-lung adenocarcinoma), p40 (positive squamous carcinoma
), p63

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

Describe the pathology for lung SCC and basosquamous cell carcinoma

A

Squamous Cell Carcinoma
- Second most-common subtype (30%)
○ Decreasing incidence (previously most common) –> reduction in smoking
- More common in men and is highly associated with smoking
- Well-defined pre-cursor lesions (dysplasia –> in-situ carcinoma)
- Tends to have central growth (bronchial/peri-hilar)

- Macroscopic
	○ Hard, white-gray tumours with central necrosis, endobronchial growth
- Microscopic
	○ Usually defined by presence of keratin 
		○ Histological grade determined by degree of keratinisation
		○ Non-keratinising SCC requires IHC for diagnosis
	○ Intracellular bridges if well differentiated 
	○ Graded keratin pearls
	○ Large polygonal malignant cells
- Immunohistochemistry
	○ POS = CK5/6,  p40, p63
	○ NEG = TTF1 & Napsin A (usually), p16, CK7, CK20
	
	Basaloid-squamous Cell Carcinoma
- Rare subtype of SCC
- Unfavourable prognosis

- Microscopic
	○ Typically has features of both non-keratinising SCC and BCC
		§ BCC = peripheral palisading of nuclei, organoid/lobular structures
	○ High grade cytology
- IHC (as per non-keratinising SCC)
	○ POS = p40, p63 NEG = CK20, TTF1, Napsin A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Describe the pathology for lung large cell carcinoma.

A

Large cell –Diagnosis of exclusion
Sarcomatoid Carcinoma
○ Rare subtype involving mixed epithelial and spindle cell populations
○ May be termed carcinosarcoma or pleomorphic sarcomatoid carcinoma
○ Poorer prognosis in general

	○ Macroscopic
		- As per other large cell carcinomas
		- Necrosis +/- haemorrhage is common
	○ Microscopic
		- Component with epithelial cells is unchanged
			* Carcinosarcoma may have heterologous sarcoma components (e.g. osteo, chondro, rhabdomyosarcoma)
		- Frequent mitoses, extensive necrosis
Pleomorphic carcinoma
	- Presumed epithelial origin
	- Contains at least 10% with spindle or giant cells
		* Spindle cells generally resemble fibrosarcoma or peripheral nerve sheath tumour 
	○ IHC: 
		* POS = Surfactant protein A (in 30%), CK7 , TTF1 NEG= CK20, Chomogranin A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Describe the features of Lung cancer driver mutations.

A

PD-L1 expression (30% NSCLC)
- Ligand for the PD1 receptor on immune cells
○ Overexpressed by cancer cells
○ Results in T-cell exhaustion –> immune evasion
- More likely to be over-expressed in smokers
○ More complex mutational burdens
- Proportion of cells over-expressing PDL1 is prognostic and predictive
○ Higher scores (i.e. >50%) indicate higher chance of success for immunotherapy (ICIs)
- 30% NSCLC have 1% or more PD-L1

EGFR (10-15% adenocarcinoma)
- Cell-membrane receptor which triggers RAS-RAF-MEK-ERK pathway for cell cycle stimulation
- 10-20% of adenocarcinomas (esp non-smokers, 50% in Asians, females)

- Mutations can include copy number gain, autonomous activation
	○ Increased downstream transcription
- Key mutations
	○ Favourable EGFR-TKI response
		- Exon 19 deletion
		- Exon 21 missense mutation (L858R)
	○ Unfavourable EGFR-TKI response
		- Most exon 20 mutations = T790M (can develop on treatment)

ALK (5%)
- Uncommon mutation (incidence is 5%) –> Fusion of the EML4 and ALK
○ Occur in younger patients who are non-smokers/ light smokers
- Tend to be exclusive of EGFR/KRAS mutations
- ALK TKIs such as crizotinib/alectinib are active

ROS1 (<2%)
- Rare mutation/fusion (<2% of all adenocarcinomas) –> multiple fusion partners
○ Tends to occur in non-smokers
- Tends to respond to first-generation ALK TKIs (e.g. crizotinib)

RAS (20% adenocarcinoma)
- KRAS is the most frequently mutated (20% of adenocarcinomas)
○ Mutually exclusive of EGFR or ALK re-arrangement
○ Most common mutation is KRAS G12C
- Impart poorer prognosis
○ Sotorasib (selective KRAS G12C inhibitor) is now available for use
○ Results in inefficacy of EGFR TKIs (mutation further downstream)

BRAF (<5% lung adenocarcinomas)
- Activator of downstream MAPK signalling
- Present in <5% of lung adenocarcinomas
- May involve mutation of the V600 locus or others
If V600E, can use dabrafenib and trametinib doublet therapy (as per melanoma)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are th prognostic factors for NSCLC?

A

Key Prognostic Factors

Patient Factors
- Age >65, male, and performance status >1
- Weight loss >5%, Anorexia
- Ongoing smoking
- Lung function/co-morbidity
- High neutrophil/lymphocyte ratio>4

Tumour Factors
- TNM stage
○ Larger size
○ Oligometastatic disease does better than extensive metastases
○ Pleural effusion is adverse
- Histological subtype
○ Unfavourable = sarcomatoid, basaloid, micropapillary
- Mutation status
○ EGFR mutation have better prognosis (irrespective of TKI therapy)
○ EGFR ≅ ALK > KRAS > Wild type.

- LVI
- High SUV on PET-CT
- Hb, Albumin, LDH
- Pancoast (better prognosis)

Treatment Factors
- Complete resection (if early stage)
- Concurrent chemoradiotherapy
- Suitable for immunotherapy
- High volume centre
- Access to treatment

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

How does NSCLC and SCLC present?

A

Asymptomatic and incidental finding on imaging.
* Finger clubbing
○ 35% of NSCLC and 5% SCLC
○ Lung cancer one of the most common cause of finger nail clubbing
* Vocal cord paralysis
○ Secondary to recurrent laryngeal nerve injury
○ Assessed on laryngoscopy
* SVC obstruction
○ Chest pain, cough, dyspnoea, blurred vision, plethora, headache, confusion
* Lobar collapse
○ Follows obstruction of bronchus
○ Acute presentations- dyspnoea, hypoxia, chest pain
○ Chronic collapse- may be asymptomatic, chronic cough, weight loss
○ Radiologically volume loss and displacement of fissure towards collapsed lobe, collapsed lobe is triangular or pyramidal shape
* Pleural effusion
○ Dyspnoea, hypoxia, cough, pleuritic chest pain
○ Transudate (CCF, cirrhosis): Low protein and low LDH
○ Exudate (Carcinoma, PE, Pneumonia): High protein, high LDH

* Pericardial effusion
	○ Clinical presentation relates to rate at which the fluid accumulates, gradual accumulation allows the pericardium to stretch 
	○ Dyspnoea on exertion, chest pain , syncope
	○ Beck's triad: Low BP, distension of jugular veins, muffled heart sounds/pericardial rub
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What features on history are important for someone with a lung lesion?

A

Consultation
History
- Symptoms related to primary
○ Dyspnoea
○ Cough
○ Haemoptysis
○ Chest Pain
○ Weight loss
○ Voice hoarseness (RLN palsy)
- SVCO
- Pancoast
○ Brachial plexopathy
§ Should/ arm pain/ paresthesia/ weakness
○ Horner’s syndrome (due to involvement of paravertebral sympathetic chain/ inferior cervical ganglion)
§ Ptosis, miosis and anhidrosis
- Paraneoplastic
○ Hypercalcaemia (PTHrP) –> SCC (squamous)
○ SIADH -Small cell

- PMHx -
	○ Interstitial lung disease 
		§ ILD-GAP score -predicts mortality of ILD at 1,2,3 years
			* Score 0-1: 3yr mortality of 10%
			* Score >5: 3yr mortality of 75%
	○ COPD
- Social History
	○ Smoking -> council on smoking cessation
	○ Occupational exposures (e.g. asbestos)
	○ Functional status
	○ Exercise tolerance

Examination
- Observations (including saturations)
- Chest auscultation
- UL neurology exam (if suspicious for brachial plexopathy)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What investigations are important for someone with a lung lesion?

A

Tissue diagnosis
○ Primary tumour:
- Sputum cytology - esp. central, large tumours, +ve in 70% central airway tumours, only 5% peripheral
- Bronchoscopy (to 2nd/3rd segmental divisions) + biopsy, brushings or lavage. Bronchial findings of proximity to carina relevant to staging.
- CT-guided percutaneous biopsy (core or FNA): 1/3 develop pneumothorax, 5% need ICU. ↑risk if bullous emphysema.
- Thoracoscopy- in rare cases where difficulty in establishing histological diagnosis by other means. Usually due to inflammation associated with small focus of ca.
○ Mediastinal LNs
- EBUS-TBNA (endobronchial US-guided transbronchial needle aspiration) guided bx- good for sampling mediastinal LN too.
* Can access 2R/2L, 4R/4L, 7, 10R/10L
- EUS-FNA (oesophageal endoscopic US-guided FNA)
* Can access 5, 7, 8 and 9
- Mediastinoscopy - Bx LN, can determine whether surgery is an option. Not superseded by PET.
○ Nodal Sampling
- PET/CT has a ~5% false negative rate for peripheral disease, while upwards of 25% for central disease.
○ Peripheral tumours with negative PET/CT in N2 may not require EBUS if frail or elderly.
○ Realistic to avoid mediastinal sampling if ≤ 2 cm peripherally located lesion with negative PET.
○ Peripheral tumours with positive PET/CT in N2 are encouraged to have EBUS for confirmation given false positives.
○ Central tumours with negative PET/CT in N2 or hilar are encouraged to have confirmatory EBUS.
○ Sample at least 3 stations.
○ Sample all abnormal LN.

		- Cervical mediastinoscopy: Gold standard (Sn 44-92%, Sp 100%) but risk of complication and operator dependent. Incision 1 cm above suprasternal notch. ?can only be done once due to scarring.
	○ Molecular panel for Adenocarcinoma 
	
	○ Metastatic disease
		- Pleural tap (+Bx) cytology- low yield (recommended if a pleural effusion is present in patients who would otherwise have curable disease)
		- Imaging-guided biopsy of other metastatic sites
		
	○ Important to biopsy any equivocal LN on PET-CT
	○ VATS - Video assisted thoracic surgery
	

- Bloods (pre-chemotherapy)
	○ FBC, EUC, CMP, LFT
	○ Current evidence suggests that neutrophil to lymphocyte ratio may be a biomarker for poor prognosis in lung cancer. Values >4 in a metanalysis found to have associated poorer prognosis
	○ Coags
	○ HepB sAg, sAb and cAb
	○ Hep C serology
	○ HIV serology
	○ TSH, T4, cortisol (immunotherapy)

- Lung Function Tests
	○ FEV1
		- Correlates well with the degree of respiratory impairment in patients with COPD, and it provides an indirect measure of pulmonary reserve. 
		- FEV1 <60% predicted was the strongest predictor of postop complications. 
		- Suggested thresholds:
			* Lobectomy: >0.8L (or >40% predicted); 
			* Pneumonectomy: >2L (>60% pred) for pneumonectomy. 
			* Radical radiotherapy (esp. stage III): 1.5L (or >50% predicted)
	○ DLCO (diffusing capacity for carbon monoxide)
		- Indirect assessment of gas exchange capacity of the lung.  
		- Need
			*  >40% of predicted for surgery or XRT (as DLCO drops during XRT).
			*  >60% low risk for post-op complications.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What’s the differential diagnosis for a lung lesion?

A

Benign
○ Infection: pneumonia, abscess, TB
○ Lung hamartoma (overgrowth of mature lung tissue)
○ Benign mesenchymal tumour (eg fibroma, lipoma, haemangioma)
Malignant
○ Primary lung cancer
○ Metastasis
○ Carcinoid
○ Lymphoma
○ Malignant mesenchymal tumour (eg sarcoma)

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

What’s the TNM staging for lung cancer?

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

What is the general management for Stage I and Stage II NSCLC?

A
  • Standard of care is a lobectomy with mediastinal sampling in medically operable
    ○ (lobectomy >segmentectomy>wedge)
    ○ Advantages: mediastinal nodal staging, complete histopath/margins
    ○ Disadvantage: GA and surgical risks, loss of lung volume
    • Reasonable alternative is SABR in patients who are medically inoperable/declines surgery
      ○ No comparative RCT
      ○ Local control data are similar
    • If SABR is not feasible for whatever reason (e.g. ultracentral), consider EBRT alone
      ○ Definitive hypofractionated RT alone (55Gy/20F without chemo)
      ○ No concurrent chemoRT
    • Medically inoperable/declines surgery:
      ○ N0
      - Peripheral, <5cm: SBRT 48Gy/4# or 54Gy/3# CHISEL (improved LC/OS cf. conventional - CHISEL)
      - Central, <5cm: Consider SBRT 50Gy/5# vs. conventional RT
      - Ultra-central / >5cm: Conventional/hypofractionated RT (60Gy/15#, 55Gy/20# or 60Gy/30#) Single institution data – excessive toxicity (0-22% gr5 toxicity) with SBRT, esp if on blood thinners
      ○ N1:
      - 60Gy/30# with concurrent chemotherapy, adjuvant durvalumab Extrapolation from data for concurrent CRT for stage III disease
    • Adjuvant treatment
      ○ Stage IA, R0 resection: observation
      ○ Stage II (+/- stage IB): benefit from adjuvant cisplatin double chemotherapy (5 yr OS benefit 5%) LACE MA, MA Collaborative group MA (5yr OS benefit 5%)
      - Cisplatin-based regimen
      □ +Adenocarcinoma = pemetrexed
      □ + Squamous = gemcitabine
      ○ pN2: chemo > PORT 50-54Gy/25-27Fx - PORT MA (LC), ANITA analysis (OS), NCDB population studies (OS)
      ○ R1: PORT 60Gy/30Fx - NCDB studies (OS)
      ○ R2: concurrent chemoRT 60Gy/30Fx - NCDB studies (OS)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Discuss indications for surgical management for Stage IIIa NSCLC and adjuvant radiotherapy/adjuvant chemotherapy.

A

Surgery
- Should carefully consider establishing pathological mediastinal nodal status
○ Only single station, non-bulky N2
- Two main options
○ Lobectomy with hilar dissection and mediastinal sampling
○ Definitive chemoradiotherapy

Adjuvant RT
- Adjuvant radiotherapy is indicated after surgery if:
○ Positive surgical margin (usually bronchial)
- Adjuvant RT for incidental pN2 disease is controversial
○ Post-operative finding of pN2 disease is common (20% of surgical specimens are upstaged)
○ No clear benefit seen, with possible survival disadvantage demonstrated
- LungART & PORT-C trials were negative
- Dose is 54Gy/27F

Adjuvant chemotherapy
- Adjuvant cisplatin-based chemotherapy is standard of care for all stage II-III patients
○ Cisplatin-based regimen
- Adenocarcinoma = pemetrexed
- Squamous = Gemcitabine

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

What is the general management for Stage III NSCLC?

A
  • Standard of care is definitive chemoradiotherapy
    ○ 60Gy/30F to primary and involved nodal disease (no ENI)
    ○ ○ Concurrent cisplatin/etoposide
    § Carboplatin/paclitaxel may be an alternative
    ○ Concurrent CT/XRT has superior survival.
    XRT alone: median survival 1yr, 2yr OS 10-20%, 5yr OS 5%.
    • Two options if chemotherapy is to be omitted
      ○ Boost to 66Gy/33F
      ○ Hypofractionation (55Gy/20F)
    • Adjuvant durvalumab to be given for 12 months
      ○ PACIFIC trial
      ○ Following definitive chemoradiotherapy, adjuvant durvalumab 10mg/kg every 2 weeks for up to 12 months, started within 6 weeks of completing chemoradiotherapy improves overall survival by 10% at 2 years (66% vs. 56%)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What is the general management for a Pancoast tumour?

A
  • Two key options
    ○ Neoadjuvant chemoradiotherapy followed by resection (standard of care) swog and jcog ph2 trials 5year overall survival 50%
    - 45Gy/25F with concurrent cisplatin/etoposide
    - Surgical resection
    - Adjuvant chemotherapy (adjuvant cisplatin/etoposide)
    ○ Definitive chemoradiotherapy alone
    - 60Gy/30F with concurrent cisplatin/etoposideUnresectable tumour.
    * Proposed criteria for unresectability may include:
    ○ the involvement of ipsilateral or contralateral mediastinal nodes (N2/N3)
    ○ involvement of the brachial plexus above the T1 nerve root
    ○ involvement of more than 50% vertebral bodies
    ○ invasion of the oesophagus and/or trachea
    ○ metastatic disease.
    • Note the difference in radiotherapy dose (difficult to compensate at the end of treatment)
      ○ Ideally decide surgical or non-surgical pathway at the beginning of treatment paradigm
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is the evidence for SABR in early stage NSCLC?

A

SABR
For medically inoperable peripheral lung tumours with a maximum diameter less than 5 cm, SABR with BED >100 Gy should be considered as standard therapy, and achieves 5-year local control rates of >90% and 3-year overall survival (OS) rates of 60%-70%.

· RTOG 0236 (Phase II, Timmerman, 2010, 2014, 2018 )
o SABR in medically inoperable early stage (T1-3,N0) non- central <5cm ,
o 54Gy/3Fx,
o 3y OS > 50% with most ppl dying of other comorbidities. MS 48 mo.
o 3y LC 98%, involved lobar control 91%, 3y DM 22%, 3y DFS 48%. 3y LRR 13%, 3y OS 56%. MS 48 mo.
o 5y DFS only 26%.
o 5y LR 7%, 5y involved lobar recurrence 20%, 5y LRF 26%, 5y DM 24%, 5y DFS 25%. 5y OS 40%.
o Grade 3+ toxicity of 30%, twice as high as RTOG 06-18 (operable pt) likely due to non-operable candidates in this study.
· CHISEL TROG 09.02 trial (Phase 3 RCT, Ball, 2019)
o 101 patients with Stage I (T1-T2a) NSCLC were randomised to SABR vs conventional RT
- PET-CT mandated
- SABR (54Gy/3F, 48Gy/4F - if <2cm from CW) or EBRT-3DCRT (66Gy/33F or 50Gy/20F)
- Inoperable pt or refusing surgery
o At 2 years SABR had less local recurrence (65% vs 89%, HR 0.32) and improved OS (77% vs 59%).
o Similar toxicity between groups (low, only 1 grade 4 tox)
· LUSTRE (Canada)(Phase 3 RCT, Clin Lung Ca, Swaminath, 2017; Astro 2022)
o 60/15 (BED = 84) vs. SBRT 48/4 or 60/8 (for central).
o Underpowered non-significant trend to improved LC with SABR.
o LC was numerically better with SBRT w/ no difference in toxicity compared to HFRT
o 324 pts expected to detect 3y LC improvement from 75% with CRT to 87.5% with SBRT. 233 patients with ≤ 5 cm stage I NSCLC or a suspicious growing PET avid lesion were accrued from 2014-2020. MFU 3y.
o 3y LC ~81→ 88% (p=0.15).
o Only one patient in each arm experienced acute G3 toxicity, no G4-5 toxicities were observed.
o Late G3-4 in 7 patients: SABR central (7%), peripheral (2%). Moderate hypofrac central (5%), peripheral (2%).
o One patient who received 60/8 for a central NSCLC experienced possible G5 toxicity (haemoptysis).
· SEER 2001-2001 (Shirvani, Red Journal, 2012)
o 10923pts, age ≥66, stage Ia-Ib NSCLC: lobectomy, sublobar resection, conventional RT, SABR, observe
- Lobectomy:59%, sublobar resect: 12%, conventional RT: 15%, SABR: 1%, observe: 13%
- Median f/up: 3.2yrs
o SABR: lowest risk of death w/in 6mo of dx (HR = 0.48)
o After 6mo: lobectomy assoc w/ best OS, DFS
o In propensity score matched analysis: OS after SABR similar to lobectomy (no stat diff)
o Worst outcomes: conventional RT, observe

SABR vs surgery
- Multiple small randomised trials, but all closed early due to accrual
- Pooled analysis of STARS and ROSEL trials (Chang, 2015)
○ 58 patients in total
○ 3 year OS was better with SABR (95% vs 79%; p=0.037)
○ Toxicity better with SABR

- Retrospective VA database study (Bryant, 2018)
	○ 4000 patients identified
	○ CSS mortality is worse with SABR compared with lobectomy (HR 1.45)
	○ Immediate short-term mortality is worse with surgery

	○ Conclusion:
		- Lobectomy remains the standard of care, but SABR is a reasonable alternative if
			* Not medically or technically fit for surgery
			* Patient preference
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What is the evidence for adjuavant RT in NSCLC with positive margins?

A
  • Retrospective NCDB data (Wang, 2015)
    Increased median OS (33.5mo vs 23.7mo; mvHR 0.8; 95% CI 0.70-0.92) when patients with positive surgical margins following resection of stage II/III NSCLC are given PORT to 50-74Gy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What is the evidence for adjuavant RT in NSCLC with incidental pN2?

A

PORT for incidental N2 should not be routinely given
- LungART RCT (Le Pechoux, 2022)
○ 501 patients with pN2 positive disease after surgery (had to be cN0-1 at diagnosis)
- FDG-PET staging (91%)
- 3D-CRT mandated, but IMRT allowed (only 11%)
○ Randomised to PORT or observation
- 54Gy/27F
○ 3 year disease free-survival was unchanged (HR 0.86; p=0.18)
○ No difference in OS (HR 0.97)

- PORT-C (2021). Phase III. Single institution. R0N2→ CTX x4c→ ± PORT 50/25. 
	○ 3y OS ~83 obs→ 78% PORT (p=0.93). 
	○ mDFS 18.6mo Obst vs 22mo PORT
	○ 3y LR-only 18% obs→ 10% PORT.
	○ 3y LRFS 60→ 67% (HR 0.71; 95% CI 0.51-.097). 
	○ 3y DMFS 38→ 42%. 
	○ Conclusion: For resected, pN2 NSCLC, PORT halves the rate of locoregional recurrence, but that does not appear to significantly improve survival outcomes. Although PORT halves the rate of mediastinal relapse for resected N2 NSCLC, the benefit is insufficient to overcome the effect of metastatic progression and the potential adverse cardiopulmonary effects on overall survival.

- ANITA trial (Douillard, 2008) 
	○ A randomised trial of cisplatin + vinorelbine vs observation in completely resected NSCLC 
	○ Post-hoc analysis of those who received PORT (non-randomised) demonstrated a reduction in OS in the overall cohort 
		- pN2 disease subgroup had improved OS with PORT in both chemotherapy and observation arms
		- The survival detriments were isolated to the pN1 cohort who received chemotherapy

- SEER analysis (Lally, 2006) 
	○ 7465 patients from the SEER database 
	○ For pN2 disease, improved OS (HR 0.85; p=0.007)  OS detriment for pN0 (HR 1.176; p=0.04) and pN1 disease (HR 1.097; p=0.019)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What is the evidence for adjuavant chemotherapy in NSCLC, post surgery?

A
  • LACE meta-analysis (Pignon, 2008)
    ○ Meta-analysis of 5 trials and 4600 patients
    ○ Cisplatin-based chemotherapy was associated with improved OS (HR 0.89; p=0.005)
    ○ Subgroup analyses found benefit was isolated to stage II/III patients
    • Cochrane MA NSCLC collaborative group (Burdett, 2015)
      ○ MA 47 trials (trials surg vs. surg + adj chemo; and surg +RT vs. Surg +RT +chemo)
      Improvement in recurrence and 4% absolute OS benefit
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is the evidence for neoadjuvant ChemoRT in NSCLC, with resectable disease?

A

Resectable N2 disease
- Phase III randomised trial (Albain, 2009)
○ 396 patients with T1-3 pN2 NSCLC were given 2x cisplatin/etoposide + radiotherapy 45Gy/25F
§ If no progression, randomisation to surgical resection OR ongoing radiotherapy to 61Gy (uninterrupted)
○ There was no difference in median OS (HR 0.87; p=0.24)
Surgery provided a PFS benefit (HR 0.77; p=0.017)

NA Chemo-> Sx vs NA ChemoRT -> Sx?
- No diff in EFS or OS between NA Chemo vs NA CRT, hence, no benefit to the addition of neoadjuvant radiotherapy to neoadjuvant chemotherapy, except for increase pCR and mediastinal downstaging
○ The Swiss SAKK16/00 (Lancet, 2015) provides the most straightforward answer to this question. In patients with stage IIIA (N2) NSCLC, sequential induction chemotherapy then radiotherapy (to 44Gy/22#) followed by surgery was compared to induction chemotherapy followed by surgery. There was no difference in event free survival or overall survival.
○ The German GLCCG RCT (Lancet Oncol, 2008) answers a similar question. Neoadjuvant chemotherapy -> radiotherapy -> surgery was compared to neoadjuvant chemotherapy -> surgery -> adjuvant radiotherapy. Neoadjuvant RT improved pathological response and mediastinal down-staging. However, there was no difference in PFS between the two groups (5-year PFS 16% vs. 14%).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is the evidence for efficacy and timaing of definitive chemoradiotherapy in stage III NSCLC?

A

Definitive Chemoradiotherapy - Efficacy

- Phase III randomised trial (EORTC 08941, van Meerbeeck, 2007) 
	○ 579 enrolled and 321 proceeded to randomisation following clinical response from 3x platinum-based induction chemotherapy
		- If clinical response, randomised to RT 60-62.5Gy/30-32F or surgical resection 
	○ 5yr OS did not differ between surgery or RT (HR 1.06; 95% CI 0.84 – 1.35)

Definitive Chemoradiotherapy - Timing

- Sequential vs concurrent definitive chemoradiotherapy 
	○ Auperin meta-analysis (2010) 
		- 1205 patients across six trials comparing concurrent vs sequential chemotherapy 
			□ Key trials included (CALGB 8831, WJLCG, RTOG 94-10) 
		- Concurrent therapy improved OS (HR 0.84; p=0.004) and LRC (HR 0.77; p=0.01) 
		- Increased G3-4 oesophageal toxicity (RR 4.9; p<0.001)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is the evidence not supporting elective nodal irradiation in Stage III NSCLC?

A
  • PET-PLAN (Nestle, 2020)
    ○ RT to PET avid sites + 50Gy elective nodal irradiation vs RT to PET avid sites only
    ○ Concurrent chemo in both
    ○ Treating avid disease only resulted in improved LRR (14% vs 29%).
    ○ Slightly higher average dose (~2Gy) able to be given in PET only (less strain on OARs)
    Elective nodal irradiation leads to impaired LRR which is mirrored in Jinan, China Trial (2007)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What evidence does not supporting dose escalation in Stage III NSCLC?

A
  • Dose-escalation to 60Gy provides OS benefit
    ○ RTOG 73-01 trial (Perez, 1987)
    - 551 patients with unresectable NSCLC were given definitive RT alone
    □ Randomised to 40Gy, 50Gy or 60Gy
    - Intrathoracic failure rates were improved with increasing doses (27% with 60Gy vs 48% with 40Gy)
    • Dose-escalation to 74Gy reduces OS
      ○ RTOG 06-17 trial (Bradley, 2015)
      - 544 patients with unresectable NSCLC were randomised in a 2x2 trial design
      □ 60Gy vs 74Gy with concurrent carboplatin/paclitaxel and +/- concurrent cetuximab
      - Median OS was inferior in the 74Gy arm (28.7mo vs 20.3mo; p=0.004)
      Cetuximab did not improve OS and increased toxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is the evidence for immunotherapy post chemoRT, in Stage III NSCLC?

A
  • New standard of care for locally advanced NSCLC
    ○ PACIFIC trial (Phase 3, Antonia, 2017, 2022)
    - ChemoRT (60-66Gy in 30-33#, platinum based chemo)
    □ Consolidation Durvalumab q2w for 1 year vs placebo
    - Improved OS, PFS, RR and DM.
    - 5yr OS 43% vs 33%. Median OS 47.5months vs 29.1months
    - Effective in all PDL1 ≥1%
    Grade 3-4 toxicity 30% vs 26%
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is the evidence for management of pancoast tumours?

A
  • Phase II Intergroup 0160 trial (Rusch, 2007)
    ○ 111 patients with T3-4 N0-1 NSCLC underwent 2x cisplatin/etoposide with concurrent 45Gy/25F of radiotherapy
    - If disease response, proceeded to thoracotomy (3-5 weeks post chemoRT) 2x cisplatin/etoposide
    ○ 80% of patients proceeded to surgery
    - 92% of patients proceeding to surgery had R0 resection, with 65% showing pCR
    ○ 5-year OS was 54% if R0 resection (44% for whole cohort)
    pCR was associated with improved OS (p=0.02)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Describe a radiotherapy technique for NSCLC concurrent chemoRT

A

Patients
Stage III NSCLC
Good performance status

Pre-simulation
MDT discussion
Lung function tests
Smoking cessation

Simulation
Supine in vacbag with arms up
- Knee supports and ankle stocks
CT from mid neck to below diaphragm
- 2mm with IV contrast
- 4D-CT

Fusion
FDG-PET

Dose prescription
60Gy/30F (prescribed to PTV as per ICRU 83)
- If no chemo, consider boost to 66Gy/33F
VMAT technique
10 days per fortnight
Concurrent chemotherapy
- Cisplatin (50mg/m2 on D1, D8 & D29, D36)
- Etoposide (50mg/m2 on D1-5 & D29-33)
Alternatively:
- Carboplatin (2 AUC)
- Paclitaxel (45mg/m2)

Volumes
GTVp = lung disease on CT/PET
iGTVp = GTV on all respiratory phases
ITVp = iGTV + 7mm
PTVp = ITV + 7mm
GTVn = nodal disease on CT/PET
iGTVn = GTV on all respiratory phases
ITVn = iGTV + 5mm
PTVn = ITV + 7mm

Target Verification
Daily CBCT

OARs
- Bilat Lung (minus iGTV)
○ Mean < 20Gy
○ V20 < 35%
○ V5 < 60%
- Spinal Cord
○ Dmax < 45Gy
- Oesophagus
○ Mean < 28Gy
○ Avoid hotspots
- Heart
○ Mean < 20Gy
○ V50 < 25%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Describe a radiotherapy technique for NSCLC hypofractionated lung RT

A

Stage III NSCLC
Reasonable performance status
Ineligible for concurrent chemoRT

Pre-SIM
MDT discussion
Lung function tests
Smoking cessation

SIM
Supine in vacbag with arms up
- Knee supports and ankle stocks
CT from mid neck to below diaphragm
- 2mm with IV contrast
- 4D-CT with respiratory motion management

Fusion
FDG-PET

Prescription
55Gy/20F (prescribed to PTV as per ICRU 83)
VMAT technique
10 days per fortnight

Volumes
GTVp = lung disease on CT/PET
iGTVp = GTV on all respiratory phases
ITVp = iGTV + 7mm
PTVp = ITV + 7mm
GTVn = nodal disease on CT/PET
iGTVn = GTV on all respiratory phases
ITVn = iGTV + 5mm
PTVn = ITV + 7mm

Target varification
Daily CBCT

OARs
- Bilat Lung (minus iGTV)
○ Mean < 16Gy
○ V20 < 30%
○ V5 < 60%
- Spinal Cord
○ Dmax < 44Gy
- Oesophagus
○ Avoid hotspots
- Heart
○ Mean < 18Gy
○ V46 < 25%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Describe a radiotherapy technique for NSCLC with SABR

A

Stage I-II (cN0) NSCLC
- <5cm dimension
Not suitable for lobectomy (or patient preference)
Not appropriate for ultracentral tumours (PTV overlaps central structures)

Pre-SIM
MDT discussion
Lung function tests
Smoking cessation

SIM
Supine on a SBRT board, on a vacbag with arms up
CT from mid neck to below diaphragm
- 2mm with IV contrast
- Motion management
* 4D-CT with respiratory motion management (gating)
* Breath hold (insp/exp)
* Abdominal compression

Fusion
FDG-PET

Prescription
Prescribed as per ICRU 91 (D95-99% ≥100% of prescribed dose)
Peripheral tumours
- 54Gy/3F
- 48Gy/4F (when GTV <1cm from CW)
- 30Gy/1#
Central tumours
- 50Gy/5F
- 60Gy/8Fx
VMAT-based SABR technique
Alternate days
Motion management is mandatory

Volumes
GTV = lung disease on CT
iGTV/ITV = GTV on all respiratory phases
PTV = iGTV + 5mm

NOTE: no CTV expansion

Target varification
Daily CBCT
- RO present for day 1

OARs
For 48Gy/4F (SABR)
- Bilat Lung (minus iGTV)
○ Mean < 6Gy
- Heart
○ Dmax = 34Gy
- Oesophagus
○ Dmax = 30Gy
- Spinal Cord
○ Dmax = 20.8Gy
- Brachial Plexus
○ Dmax = 24Gy
- Chest wall
- D30cc < 30Gy
- Great vessels
- Dmax < 49Gy
- Trachea + PBT
- D0.5cc < 30Gy

Plan objectives
PTV: Ensure that 98% of the PTV receives ≥100% of the prescription dose
- D99% = >90% PD
- Dmax = 125-143% PD
R100= Conformity index (Vol 100% PD / Vol PTV)
- Aim <1.2 (<1.5 acceptable)
R50= Gradient Index (Vol 50% PD / Vol PTV)
- AKA intermediate dose spillage
- Tumour size dependent 6-12
D2cm = maximum dose 2cm from the PTV / prescription dose

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

When treating metastatic NSCLC, what factors should be taken into account?

A
  • Patient
    ○ Age, performance status
    ○ Comorbidities inclluding contraindications to systemic therapy, radiotherapy
    ○ Preference & goals
    ○ Symptoms
    • Tumour
      ○ Histopathology/molecular
      - Squamous vs. non-squamous histopathology
      - PDL1 status
      - Driver mutation eg. EGFR, ALK, ROS1 BRAF for which a specific targeted therapy is available
      ○ Burden of disease
      - Number and sites of mets
      - Presence of symptoms
    • Treatment
      ○ Geographical location

Molecular testing
- Lung panel - tumour assessment for presence of a driver mutation
- Routine PDL1 in all pt with newly diagnosed met for consideration of immunotherapy

Symptom management

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

For a patient with oligometastatic NSCLC, how would you manage?

A
  • If definitive therapy for thoracic disease is feasible
    ○ Systemic therapy and restaging to confirm non-progression (ESTRO/ASTRO = 3 months)
    ○ Treat thoracic disease as per T/N stage
    - Surgery
    -SBRT
    - Or conventional/hypofx +/- concurrent chemo
    Then maintenance therapy or observation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What is the systemic management of metastatic NSCLC?

A
  • No driver mutation present, or unknown
    ○ PDL1 >50%: pembro or atezo monotherapy
    - Improvement in OS compared with double chemo alone in this population
    - Combination chemoimmunotherapy preferred in rapidly progressive disease
    - CI to immunotherapy: connective disease, rheumatological, ILD
    ○ PDL1 <50%
    - Non-squamous:
    □ Doublet chemotherpay + pembro
    □ Chemo: no singler optimal combination
    □ Cisplatin>carboplatin
    □ Pemetrexed only in non-squamous histo
    - Squamous:
    □ Carboplatin + paclitaxel or nab paclitaxel + pembro
    □ OS benefit over chemo alone
    ○ 4-6 cycles, then consider maintenance
    • Driver mutation present
      ○ EGFR positive
      - Treatment with EGFR TKI: osemertinib, erlotinib, geritinib, afatinib as single agents
      - Preferred over chemo and immuno
      - Improved outcomes compared to standard platinum basedchemo
      - Impact on OS less evident, as frequently used as second line therapy after chemo in trials
      - Osemertinib > erlotinib, gefitinib
      - Continued until progression, or toxicity
      □ Oligo progressive disease > consider MDT
      ○ ALK
      - Treatment with ALK TKI preferred
      • Alectinib first line
        • Continued until disease progression or tox
        • Ceritinib
          ○ Others:
          c-ROS oncogene 1 (ROS1), BRAF, RET, TRK, MET, and Kirsten rat sarcoma viral oncogene homolog (KRAS).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is the evidence for agressive treatment of oligometastic NSCLC?

A

o Gomez et al lancet oncol 2016, 2019
Phase II, Gomez, 2016, 2019
74pts, ≤ 3mets, after 1st line sytemics w/o progress: aggressive therapy vs chemo
mPFS: 12mo (aggressive) vs 4mo
MS: 41mo (aggressive) vs 17mo
- Design: Phase II RCT (multicentre)
- Population (n=74): Stage IV NSCLC, <=3 metastases, ECOG PS <=2, no disease progression with standard first line systemic therapy
- Initial systemic therapy (all patients):
- 4 or more cycles of platinum dowblet chemotherapy, OR
- 3 or more months of EGFR or ALK TKI
- Arm 1: Local consolidative therapy (chemoRT or resection of all lesions) +/- subsequent maintenance systemic therapy
- Arm 2: Maintenance treatment alone (which could include observation only)
- Outcomes:
- Consolidative local therapy improved PFS (primary endpoint) 12 months vs. 4 months (SS)
- Updated results presented at ASCO 2018 additionally demonstrated improved median OS 41 vs. 17 months (SS)
- Commentary:
- First randomised trial to find a survival benefit for local consolidative therapy
- Limitations:
- Limited sample size (n=76)
- Molecular heterogeneity (including EGFR and ALK patients)
- Definition of oligometastatic disease (which was defined after systemic therapy)
o Lyengar et al. JAMA oncology 2018
- Design: Phase II RCT (single centre)
- Population (n=29): Stage IV NSCLC, <=5 metastases without driver mutation, PR or SD after induction chemotherapy
- Arm 1: SABR to all sites of gross disease (including SABR or hypofractionated RT to the primary) followed by maintenance chemotherapy
- Arm 2: Maintenance chemotherapy
- Outcomes:
- Consolidative SABR associated with improved PFS (10 months vs. 4 months)
- Toxicity similar between arms

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

What patient factors affect lung surgery?

A

○ Age. Perioperative mortality increases with age.
- Age is not a contraindication alone for lobectomy or wedge resection. But age should stronlgly be considered when deciding fitness of pneumonectomy
○ Pulmonary reserve
- FEV 1 >1.5 L for lobectomy and >2L for pneumonectomy for
- Predict post-op FEV1 and DLCO and if <40% considered high risk
○ Cardiovascular fitness
- Optimise IHD risk factors + stents prior to being considered for lung resection
○ Weight loss, PFS and nutrition
- Pre-op weight loss >10% or ECOG 2 are more likely to have advanced disease and poorer post op outcomes

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

WHat are the options for lung surgery and their indications?

A

○ Operations available
- Sublobar resection (wedge resection)
□ May be suitable for peripheral tumours <1cm
□ Useful for patients with impaired pulmonary reserve - higher local recurrence rate than a full lobectomy
- Lobectomy
□ Standard of care compared with wedge resection (1995 trial)
□ Mortality rates for surgeon after a surgical lobectomy should be <4% as per BMJ
- Pneumectomy
□ If cancer involved >1 lobe, the pleura/pericardium or diaphragm (as portions if this can be dissected with the procedure
□ Mortality rates for a surgeon after pneumectomy should be <8% as per BMJ
- Systematic lymph node dissection at time of lung resection is essential for lung resection to achieve accurate staging

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

What are the indications for Bronchoscopic laser resection and stents?

A

Brochoscope (rigid or flexible) to laser resect endobronchial lesions
○ Primary indication: Obstructive lesions of the trachea, main bronchi and bronchus intermedius
- Contraindication: Extrinsic compression
- Relative contraindications: Broncho-oesophageal fistula, coagulopathy, hypoxaemia
○ Laser resection of segmental bronchi does not significantly improve ventillation
○ Goal is often palliation with endobronchial manifestations of inoperable lung cancer
○ If used for curative intent- needs to be combined with other treatment modalities such as endobronchial brachytherapy

Stents
Stents are devices for internal splinting of luminal structures
4 major indications
1) Extrinsic compression from tumour or nodes
2) Stabilising airway patency after endoscopic intraluminal resection of a tumour or prior to EBRT
3) Sealing malignant fistulas
4) Benign strictures

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Describe the management options for pleural effusion.

A
  • Thoracentesis
    ○ Drainage of pleural effusion with local anaesethetic, often performed on the ward
    ○ May need an indwelling pleural catheter
    • VATs
      ○ Video assisted thorascopic surgery is done under moderate or GA
      ○ Triangular placement of ports in the chest wall, considered minimally invasive.
      ○ Pleurodesis by either chemical or mechanical methods
      - Chemical: Sterile talc in the pleural space to cause if to adhere
      - Mechanical: Brushing with dry gauze and mechanical irritaion
    • Pleurodesis
      • Sterile talc powder in the plerual space, causing the layers to adhere together and prevent accumulation of pleural fluid/effusion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Discuss the evidence for neoadjuvant chemo/immuno in resectable NSCLC

A
  • Evidence CHECKMATE
    ○ Phase 2 Stage Ib-IIIa resectable NSCLC randomised to nivolumab + platinum based chemotherapy and resection or platinum based chemotherapy alone and resection
    ○ Primary end point event-free survival, pathological response
    ○ Median event free survival 31.6 months vs 20.8 months and pathological response favoured for most subgroups with nivolumab
    The addition of nivolumab did not increased grade 3-4 toxcities
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Discuss the evidence for adjuvant chemotherapy in NSCLC

A

Cisplatin and pemetrexed
* Stage ll or lllA Adenocarcinoma - non small cell lung cancer following complete resection, in patients with performance status 0 or 1
○ JIPANG study only randomised pateints with non squamous NSCLC and hence cisplatin/pemetrexed remains standard of care for adenocarcinoma

* LACE MA (JCO 2008) (Lung Adjuvant Cisplatin Evaluation)
		- MA of 4584 patients. Individual patient data from 5 large RCTs:
				- International adjuvnat lung trial (IALT) - NEJM 2004
				- JBR.10 (NEJM 2005, JCO 2010)
				- ANITA (Lancet Oncol 2005)
				- Big Lung Trial
				- Adjuvant Lung Project Italy
		- Included patients with completely resected stage I-III disease
		- All-comers: Postop chemotherapy improved 5 yr OS by 5.4% (absolute benefit)
		- Subgroup analysis by stage:
				- IA - no benefit, trend to detriment
				- IB - HR for death 0.92 (95% CI 0.8-1.1)
				- Greatest benefit in patietns with stage II and III disease (particularly if good PS)
		- Beneficial for ECOG 0-1, may be detrimental for ECOG 2
		- Conclusion: Adjuvant chemotherapy indicated for stage II, III with good PS (0-1), improves OS by 5% at 5 years
				- Controversial for Stage IB
				- Not indicated for stage IA
* MA collaborative group MA (Lancet 2015)
		- MA of 26 RCTs, 8450 patients
		- Adjuvant chemotherapy associated with 4% improvement in OS at 5 years
		- Largest benefit (5%) for stage II and III
		- Benefit not SS for stage IA
		- Stage IB benefit 3% at 5 years
		- Conclusion: consider adjuvant chemotherapy for stage IB, II and III
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

List the common side effects for NSCLC chemotherapy, immunotherapy and targeted therapy.

A

Chemotherapy:
* Haematological (Neutropenia, thrombocytopenia, anemia)
* Electrolyte derangement
* Peripheral neuropathy
* Ototoxcitiy
* Mucositis
* Nausea and vomiting
* Fatigue
* Hair loss
Immunotherapy

Immune related events:
* GI toxcitiy (Colitis, oesophagitis, hepatits, pancreatitis)
* Cardiotoxcitiy (Myocarditis)
* Haematological (AIHA, TTP, Aplastic anaemia, acquired haemophilia
* Hepatotoxcitiy (Bilirubin elevation and transaminitis)
* MSK (Arthralgia, inflammatory arthritis)
* Neurological (Encephalitis, meningitis, GBS, myasthenia gravis)
* Ocular (Eye pain, uveitis/blepharitis, watery eyes)
* Pulmonary (Pneumonitits)
* Renal
* Skin (rash, blisters)
* Thyroid (Thyroid toxicity common with immune check point inhibitors. Typically HYPER in acute state and then progress to HYPO thyroidism)
* Endocrinology (Diabetes)
Non immune related events
* Nausea and vomiting
* Fatigue

Targeted Therapy
* Acneiform rash to face and upper body
* Anorexia
* Diarrhoea
* Fatigue
* Occular changes
* Oral mucositis
* QT prolongation
* Paronychia
* Pulomonary toxicity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Describe radiation induced lung injury

A
  • Risk factors:
    ○ Lung comorbidities e.g ILD
    ○ prior lung RT, mean lung dose, and V20 are important predictors of symptomatic pneumonitis after lung SBRT, higher w/ chemo
    • Pathogenesis
      Immediate phase (Hours to days – Asymptomatic)
      · Driven by cascade of damage associated molecular patterns (DAMPs), pro-inflammatory cytokines (TGFb, TNFa, PDGF, IL1, IL6) and chemokines released from dying cells.
      · Pulmonary oedema
      o Hyperaemic congested mucosa with leukocyte infiltration
      o Increased capillary permeability
      · Exudative alveolitis
      · Tracheal bronchial hypersecretion
      · Degenerative changes in the alveolar epithelium/endothelium
      · Type 1 pneumocytes are sloughed and surfactant levels are increased.
      Latent phase (Weeks – cough/ poor sputum clearance)
      · Thick secretions
      o Increased number of goblet cells
      · Ciliary dysfunction
      Acute exudative phase (6 weeks to 6 months – Dyspnoea, cough – from impaired gas exchange)
      · Sloughing of endothelial and epithelial cells, with capillary narrowing
      · Hyaline membrane formation – due to alveolar pneumocyte desquamation
      · Fibrin rich exudate leakage into alveoli.
      · Type 2 pneumocytes become hyperplastic, with marked atypia
      o Alveoli collapse
      Intermediate phase
      · Resolution of alveolar exudate and dissolution of hyaline membranes
      · Collagen deposition by fibroblasts
      ii. radiation fibrosis.
      Fibrosis (6 months to years – Dyspnoea) – Driven by TGF-b produced by activated immune cells as well as by alveolar pneumocytes and fibroblasts.
      · Increase in the number of Myofibroblasts within interstitium and alveolar spaces
      · Increase collagen deposition
      · Anatomic narrowing of alveolar spaces, resulting in diminished lung volume
      o Vascular subintimal fibrosis causes loss of capillaries
      o Traction bronchiectasis
    • Incidence:
      ○ Lung cancer - conventional
      - V20 >22-31 = 7%
      - V20>31-40 = 13%
      ○ Breast
      - 1-9%
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What is the 5yr OS for NSCLC, stage I, Stage II, Stage III, Stage IV

A

5 year OS
Stage I 65%
Stage II 50%
Stage III 43%
Stage IV 10%

Local control with SABR (2 years)
- T1 = 80%
- T2 = 70%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What is the epidemiology of SCLC?

A

Incidence (Australian statistics) –> lung cancer overall
- 13800 cases annually
- 5th most common malignancy
- Leading cause of cancer death

Small cell lung cancer represents about 15-20% of all lung cancers
- (adeno 40%, SCC 30%, SCLC 20%, ULCC 10%)
- Therefore, incidence is approx 2000 cases annually in Australia
- Incidence declining

Male predominance (1.2:1)
Reduction in incidence secondary to reduction in smoking
- Very strong link
- 99% smokers
- <1% in non-smokers

Natural History
- Most aggressive form of lung ca- rapid progression, early dissemination, often advanced disease at presentation.
- 67% of patients present with metastatic disease outside of chest (extensive stage).
- Local, regional (lymphatic) or distant (haematogenous) spread, usually to lung, liver, adrenal, brain and bone.
- Median survival time without treatment 2-4 months.
- Treated with CT, median survival 10-16 months.
- 2 year survival 10%; 5 year survival 5%
- Local relapse following CT alone 60%, CT+RT 30%.
- Commonly relapses <2years.

Aetiology/Risk Factors

1) Cigarette smoking (dominant risk - very strong link)
	a. Suggestion of no cases arising in men who are never-smokers
	
2) Environmental exposures
	a. Heavy metals
	b. Asbestos (in addition to mesothelioma risk)
	c. Air pollution esp industrialised countries
3) Ionising radiation
	a. Therapeutic radiotherapy
	b. Radon gas
	c. Uranium (in miners)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

Describe the pathology of SCLC

A

Histological Subtypes

Typically arise from the neuroendocrine cells of the lung/bronchus epithelium
Neuroendocrine carcinomas of lung have 3 key subtypes
1) Small cell lung carcinoma (SCLC) (aggressive end of spectrum)
2) Large cell neuroendocrine carcinoma (LCNEC)
3) Carcinoid (benign end of spectrum)

Small Cell Lung Carcinoma
- Accounts for 15% of all lung cancers
- Exclusively related to smoking
- Highly aggressive nature
○ Tends to have early metastases (may present with bulky nodes and reasonably small primary) 20% of patients present de novo metastatic disease
○ Paraneoplastic syndromes common (SIADH, Lambert-Eaton)

- Macroscopic
	○ Typically a centrally-located or hilar mass
	○ White-tan soft and friable tumour
	○ Necrosis is common
- Microscopic 'sheets small round blue cells, with little cytoplasm, no distinct nucleoli and high mitotic rates'
	○ Small blue round cells with minimal "scanty" cytoplasm (high N:C ratio, 2x size of lymphocyte)
		- Cells may grow in sheets, clusters or rosettes, ribbons, peripheral palisading
		- Delicate fibrovascular stroma often separates clusters
		- Neither glandular nor squamous organisation
	○ Cells may demonstrate finely dispersed chromatin (chromatin pattern: salt & pepper) without discrete nucleoli
	○ Frequent mitoses/High mitotic rate and accordingly, marked necrosis/apoptosis seen
	○ Nuclear moulding prominent feature
- Immunohistochemistry
- KEY: Positive TTF1, synaptophysin and chromogranin A
	○ POS = 
		- For at least one NE marker: chromogranin A, synaptophysin, INSM1, CD56 (NE markers),  NSE
		- CK7 (low molecular weight cytokeratin), 
		- Adeno markers: TTF1 ( neg in 15-20%), 
		- p53, EMA (positive in most carcinomas)
	○ NEG = CK20 (high molecular weight cytokeratin), PAX8, p63, 
		- Adeno marker: Napsin A (positive in most Lung Adeno, neg in non-lung adeno mets & SCC)
		- Squamous markers Neg : p40, p63
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Describe the pathology for Large cell neuroendocrine carcinoma (LCNEC)

A
  • Quite uncommon (~3% of all lung cancers)
    ○ More common as a mixed pattern rather than pure
    • Large cell variant of SCLC
    • Remains aggressive with poor prognosis
    • Macroscopic
      ○ Well-circumscribed mass with necrosis and a tan-red colour
    • Microscopic
      ○ Appears similar to NSCLC, IHC stains positive for NE
      ○ Larger cells (3x the size of SCLC) with:
      - Abundant cytoplasm
      - Coarse granular chromatin, nuclear pleomorphism and prominent nucleoli
      ○ Necrosis and mitoses frequent (>10 per 10hpf)
      ○ Multiple patterns (organoid, nesting, solid)
    • Immunohistochemistry
      ○ POS = chromogranin A, synaptophysin, CD56, CK7, TTF1, AE1/AE3
      NEG = CK20, p40, p63, Napsin A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Describe the pathology for lung carcinoid

A

Carcinoid (well-differentiated neuroendocrine tumours)
- Exists on a spectrum of neuroendocrine tumours (i.e. LCNEC are poorly-differentiated)
○ Typical = well-differentiated
○ Atypical = moderately-differentiated, poorer prognosis
- Uncommon tumour (1% of all lung cancers)
- Not related to smoking, unclear aetiology
○ Related to MEN1 syndrome
- Highly curable (usually surgery alone)

- Macroscopic
	○ Well-circumscribed polypoid tan coloured tumours
	○ Typically located centrally (frequently in bronchial lumen with obstruction)
- Microscopic
	○ Can be arranged as organoid, trabecular, rosette
	○ Typical
		- Cytologically bland large cells with round nuclei, finely dispersed chromatin and inconspicuous nucleoli
		- Moderate to abundant cytoplasm
		- Few mitoses (<2 per 10hpf) and necrosis is absent
	○ Atypical
		- More mitoses (2-10 per 10hpf) or presence of necrosis
		- Generally begin to have more cytological atypia
	○ Delicate fibrovascular stroma
	○ Ki67 <20%
- Immunohistochemistry
	○ POS = chromogranin A, synaptophysin, CD56, TTF1, AE1/AE3
	○ NEG = CDX2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Describe the prognostic features for SCLC

A

Patient Factors
- Age and performance status (tolerability of treatment)
- Ongoing smoking during treatment
- Weight loss
- Females have better survival (unclear cause)

Tumour Factors
- Stage (typically LS vs ES)
○ TNM is also prognostic however
○ Number of metastatic sites
○ CNS, marrow and liver mets (unfavourable cf. other sites)
- Paraneoplastic syndromes are unfavourable (e.g. SIADH) ie. ¯Na
- Elevated LDH

Treatment Factors
- Overall treatment time for RT: Start of chemo > end of RT (OS decreases by 2% for every 1
week of prolonged treatment time)
- Early chemotherapy
Early radiotherapy (cycle 2 at latest)

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

Describe the features to check on history for SCLC

A

History
- Primary lesion
○ Dyspnoea
○ Chest pain
○ Cough
○ Haemoptysis
○ Hoarse voice - recurrent laryngeal n involvement
○ Dysphagia
○ Weight loss
- Symptoms of SVCO
○ Mediastinal lesion grow to obstruct blood flow from SVC
▪ Either extrinsic compression or direct tumour invasion
○ Facial plethora
○ Arm swelling
○ Dyspnoea and headaches
○ Collateral vessels on skin
- Paraneoplastic syndromes
○ Lambert-Eaton (proximal muscle weakness, anti voltage-gated ca channel Ab)
▪ Classic association
▪ <5% of patients SCLC
▪ Proximal weakness leading to difficulty walking/difficult to perform ADLs
▪ Develop antibodies to the pre-synaptic calcium channels in the neuromuscular junction leading to weakness. Characteristically improves with repeated contraction (unlike myasthenia gravis)
○ SIADH (5-10%)
- Malaise, weakness, confusion, volume depletion, nausea
- Hyponatremia, euvolemia, low serum osmolality, inappropriately concentrated (high) urine osmolality
○ Cushing Sx
- Weight gain, moon facies, HTN, hyperglycemia, generalised weakness
- High serum cortisol and ACTH, hypernatremia, hypokalaemia, alkalosos
○ Hypercalcaemia (calcitonin)
- Constitutional symptoms
- Other
○ Oesophageal obstruction
○ CNS metastases (seizures, ICP symptoms)
○ Leptomeningeal disease, adrenal mets, liver mets, bone mets and associated sx
- PMHx
- Social History
○ Smoking –Counsel on cessation

Examination
- Observations (including saturations)
- Chest auscultation
- Pemberton’s sign

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

What investigations are needed to work up SCLC?

A
  • Bloods (pre-chemotherapy)
    ○ FBC, EUC, CMP, LFT, LDH (LDH non-specific marker, elevated levels in SCLC associated with greater disease burden and worse OS)
    ○ Coags
    ○ HepB sAg, sAb and cAb
    ○ Hep C serology
    ○ HIV serology
    ○ TSH, T4, cortisol (immunotherapy)
    • Histopathology
      ○ Percutaneous core-biopsy lung/node
      ○ EBUS + core biopsy (often endobronchial approach)
      ○ Sputum cytology
    • Staging imaging
      ○ CT CAP + WBBS
      § Alternative is FDG-PET-CT (recently funded)
      □ Stage change in 5-10% of patients
      ○ MR Brain (↑ sensitivity, particularly if Sx and neg CT)
      § BM positive in 15% at Dx, 5-8% asymptomatic
    • Lung function testing
    • Other (as indicated)
      ○ Pleural cytology
      ○ Lumbar puncture
      ○ BMAT if features suggestive of marrow infiltration (cytopenias, nucleated RBCs on smear)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

Describe the surgical option for management of Limited stage SCLC.

A
  • A potential option for cT1-2 N0M0 disease
    ○ SCLC is rarely diagnosed so early (can be an incidental finding in resection for a lung nodule/ open biopsy)
    ○ <5% of patients
    • Requires histological mediastinal staging prior to proceeding
    • Lobectomy with mediastinal lymphadenectomy
    • Should proceed to adjuvant chemotherapy with 4x cisplatin/etoposide
    • +/- mediastinal RT (sequential or concurrent if incidentally node positive)
    • If refusing surgery then this group could also be managed with SABR + adjuvant chemotherapy or Concurrent chemoRT
      Can consider omission of PCI in stage 1, especially for elderly patients (ASTRO)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

Describe the general management of Limited stage SCLC.

A

Chemoradiotherapy

- Patients: Good performance status (ECOG 0-2) or poor performance status due to SCLC
- Chemo and RT to commence ASAP
- Ideally aim for concurrent chemoradiotherapy
	○ Concurrent is superior to sequential
	○ Evidence shows that early radiotherapy is better (With C1 or C2)
		- Commencing with C2 is usually more feasible and may be preferred if bulky disease pre-chemo
	○ Can consider sequential in poor performance status (ECOG 3-4) due to SCLC

- Aim for four cycles of cisplatin/etoposide (preferred)
	○ Carboplatin (poor kidney function) can be swapped if toxicity is a concern
	○ Aim 2 cycles with concurrent RT, 2 cycles post 
	○ Cisplatin 60mg/m2 IV infusion, day 1 (carbo in ES-SCLC)
	○ Etoposide 120mg/m2 IV infusion, days 1-3
		- Higher response rates with cisplatin/etoposide (therefore preferred for limited stage)
		- Better toxicity profile with carboplatin/etoposide (therefore preferred for extensive stage)
			□ Less risk of nephrotoxicity, neuropathy, neurotoxicity and emesis 
			□ Higher risk of mucosal toxicity, interstitial pneumonitis and haematologic toxicity
	○ Toxicity:  
		- Immediate: hypersen, N/V, taste/ smell
		- Early: neutropenia, TCP, hypo Mg, Ca, K; mucosiits, oesophagitiis, anorexia, fatigue, peripheral neuropathy (platinum), nephrotoxiciry, ototoxicity 
		- Late: anaemia, alopecia, infertility

- Thoracic radiotherapy should commence ASAP
	○ Liaise with med onc: ideally concurrent with C1; often not feasible (planning), so aim for C2

- Options for thoracic radiotherapy; optimal not established
	○ Turrisi --> 45Gy/30F/1.5GY BD over 3 weeks preferred
		- Preferred, but logistically difficult
		- NEJM 1999 accelerated RT 45Gy in 1.5 fractions BID (6 hours apart) to take advantage of rapid turnover/high mitotic rate.  
	○ CONVERT --> 66Gy/33F daily
		- Phase 3 trial unsuccessful at demonstrating superiority compared with Turrisi
	○ EviQ - allows a range if concerns re tolerability, preference for higher doses when achievable
		- 50Gy/25Fx
		- 40Gy/15Fx
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Describe PCI in the SCLC LS setting

A
  • PCI is associated with OS improvement and reduction in symptomatic brain metastases in LS-SCLC
    ○ Commenced in all pt with LS-SCLC (with complete or partial response) with good performance status
    ○ Dose = 25Gy/10F
    ○ Higher doses increase risk of neurotoxicity
    ○ Rationale: IC mets occur in >50% of pt with SCLC; sanctuary site
    • Patients should be restaged after completion of chemotherapy prior to proceeding
    • Administer after resolution of acute toxicities
    • Consider adding memantine (not PBS funded, off label)
      ○ Improve neurocognitive toxicity with WBRT
      ○ Schedule
      - Commence on D1 of WBRT
      - 5mg mane for 1 week, then escalate to 5mg BD for 1 week
      - Continue weekly escalation until 10mg BD
      - Continue for 6 months
    • At this stage, use of HA-WBRT should be considered experimental
      ○ Only one trial, with previous contradictory data
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

Describe the initial general management of ES SCLC

A

MDT discussion
–> Upfront systemic treatment is priority
-Radiotherapy can be used initially for symptomatic brain mets
-> radiation for symptomatic bone mets if not respond to upfront chemotherapy or in rare cases of concern for impending cord compression with no response to systemic therapy.

Upfront chemotherapy + immunotherapy
- For Good PS patients (ECOG 0 -2) or poor PS due to SCLC
- 4-6 cycles of carboplatin/etoposide/atezolizumab (IMpower133)
○ Number of cycles depending on tolerability
- Carboplatin AUC 5 IV infusion day 1 (as opposed to cisplatin with limited stage, although both are reasonable - no combination has established superiority)
□ Both combinations were equally effective in tumour response and survival (EviQ)
- Etoposide 100mg/m2 infusion days 1-3
○ Common toxicities: (Carbo/Etop has better toxicity profile compared to Cis/Etop)
- Immediate: hypersensitivity, N/V, taste and smell alteration
- Early: neutropenia, thrombocytopenia, oral mucosiits, anorexia, fatigue
- Late: anaemia, alopecia

- Alternative options 
	○ Platinum/etoposide + durvalumab x 4 cycles  until progression/toxicity (CASPIAN)
	○ Carboplatin/etoposide x 4-6 cycles 
	○ Cisplatin/etoposide x 4-6 cycles

- Complete restaging upon completion of chemotherapy
	○ CT CAP
	○ MR Brain

- Maintenance immunotherapy to continue until disease progression/toxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What is the role of Radiotherapy in ES SCLC?

A

Consolidation Thoracic Radiotherapy
- Consider in patients with a good partial response (NCCN guidelines; CREST)
○ Residual thoracic disease
○ Minimal residual extrathoracic disease
○ No benefit if no residual disease
○ Volumes similar to LS-SCLC
○ Treat post chemotherapy volume, but include all mediastinal nodal stations that were invovled prechemo
- Thoracic RT has been shown to improve overall survival
○ NOTE: these trials did not allow immunotherapy (uncertain if benefit persists)
- Typical dose would be 30Gy/10F (CREST)
○ Withhold immunotherapy for this period (concerns for thoracic toxicity)

Symptomatic Lesions
- Consider upfront management of symptomatic lesions prior to systemic therapy
○ SVC obstruction
- For most patients, intiially chemo is treatment of choice, as chemosensitive and response is usually rapid
- Consider endovascular or RT if potentially life threatening, or in those who don’t respond to chemo
○ Significant bronchial obstruction
○ Significant pain/bone metastases
- Stablisation/fixation > RT
○ Cord compression
- Initiate steroids + RT before systemic therapy, unless immediate systemic therapy required

- Brain metastases as a special case
	○ If asymptomatic, can consider upfront systemic therapy
		- Need close MR surveillance during chemotherapy
			□ Brain MRI every 2 cycles and for RT if progression before finishing systemic treatment
		- Then WBRT following completion of induction chemo
	○ If symptomatic, but be treated with surgery/RT upfront
		- Steroids + WBRT first 
		- Systemic therapy following
	○ Recommended dose:
		- 30Gy/10Fx 
	○ WBRT vs. SRS
		- NCCN guidelines: WBRT rather than SRS
			□ If recur, SRS preferred; re-WBRT can be consider
		- Rationale: patients tend to develop multiple CNS mets
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Discuss PCI in ES SCLC

A

PCI

- May be considered if excellent response (complete or partial) to chemoimmunotherapy (and good performance status)
	○ No clear OS benefit
	○ Reduce incidence of symptomatic brain metastases
- Note that close MR surveillance is an appropriate alternative in patients suitable
	○ 3 monthly for year 1 and then 6 monthly there after
	○ In pt with no brain mets on MRI, PCI reduces incidence of BM (40 vs. 65% at 18 months), but does not improve OS cf MRI surveillance

- Dose is typically 25Gy/10F (though 20Gy/5F is reasonable given poor prognosis)
	○ Consider withholding immunotherapy for this period
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What is the evidence for efficacy of chemoradiotherapy in LS-SCLC?

A

Efficacy in LS-SCLC
- Thoracic RT improves (below)
- Local relapse following Chemo alone 60%, CRT 30%
- Demonstrated in 2 x Meta-analysis
- (Pignon, 1992)
○ 13 trials and 2140 patients with LS-SCLC
○ Population LS-SCLC
○ Comparison of chemoradiotherapy vs chemotherapy alone
○ Improved OS with chemoradiotherapy (HR 0.86; p=0.001)
- 14% mortality reduction
- + (Warde, JCO 1992)
○ MA 11 RCTs
○ 5% improvement in OS at 2 years
○ Improvement in 2-year LC by 23 % (47% vs. 24%)

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

What is the evidence for timing of chemoradiotherapy in LS-SCLC?

A
  • Japanese trial - JCOG 9104 (Takada, 2002)
    ○ 231 patients with LS-SCLC
    ○ Randomised to concurrent or sequential chemoRT (45Gy/30F BD +4x cisplat/etop)
    ○ Concurrent treatment lead to improved OS (median OS 27mo vs 19mo, NS)
    ○ 5% gain in OS at 5 years with concurrent CRT (24 vs. 18%)
    ○ More frequent G3 oseophagitis 9 vs. 4%
    • Systematic review (De Ruysscher, 2006)
      ○ 4 RCTs with 1056 patients
      ○ Time from start of any therapy to completion of radiotherapy was predictive for OS (RR 0.63)
      Optimal if <30 days
58
Q

What is the evidence for the different dose fractionation scheduled in LS-SCLC?

A
  • Turrisi RCT (Turrisi, 1999)
    ○ 417 patients with LS-SCLC
    ○ Randomised to 4x cisplatin/etoposide and radiotherapy (45Gy with BD vs 45 Gy daily fractionation 1.8 Gy/# over 5 weeks)
    ○ Improved OS with BD fractionation (mean OS 23mo vs 19mo)
    ○ Turrisi study included elective nodal coverage, however recent studies suggest isolated nodal failures are uncommon
    • CONVERT trial (Faivre-Finn, 2017)
      ○ 547 patients with LS-SCLC
      ○ Randomised to 45Gy/30F BD vs 66Gy/33F daily (with concurrent cisplatin/etoposide)
      - Trail failed to show superiority of OD regimen
      - Superiority trial with BD fractionation deemed standard
      ○ No difference in OS between the cohorts (median OS 30mo vs 25mo)
      - Thus, BD fractionation remains the gold-standard
      - Daily fractionation can only be considered a reasonable alternative
      ○ G3-4 oesophagitis = 20% in both arms
      ○ Primary end point was OS difference of 12% to demonstrate superiority of once-daily radiation therapy. Study not powered to show equivalence, the implication is that twice-daily radiation therapy should continue to be considered the standard of care in this setting.
    • CALGB trial/RTOG (Bogart et al 2021)
      ○ Majority of patients in clinical practice are treated with high dose once daily thoracic RT vs standard BID treatment
      ○ Randomised thoracic RT, all started with C1 or 2 of cisplatin/etoposide
      - 45Gy/30# BID over 3 weeks
      - 70Gy/35# OD over 7 weeks
      - 61.2 GY concomitant boost over 5 weeks (this arm was discontinued due to a planned interim toxicity analysis)
      No significant difference in OS and PFS and most G3 toxicity were similar. Although did not improve superiority, favourable outcomes were still seen in the once daily arm
59
Q

What is the evidence for consolidation RT in ES-SCLC?

A
  • CREST RCT (Slotman, 2015)
    ○ Phase III RCT
    ○ Population :498 patients with ES-SCLC with good response to chemotherapy
    ○ Intervention/ Control: Randomised to thoracic RT (30Gy/10F) or observation after completion of chemotherapy
    - All pt received PCI
    ○ Trial did not meet primary end point of significant OS improvement at 1 year
    ○ Thoracic RT associated with improved 2-year OS (13% vs 3%)
    - And 6 month PFS (24% vs. 7%)
    - Unplanned subgroup analysis:
    □ Only patients with residual disease benefit (no elective adjuvant therapy)
    o Residual disease in chest after chemo present (434 of 495 patients)
    - Improved 1-year OS, 33% vs. 26%, HR 0.81, SS p=0.044
    - Improved 2-year OS 12% vs. 3%
    o Residual disease in chest after chemo absent (61 of 495 patients):
    - No significant benefit to thoracic RT
    - Ie no residual disease = no benefit
    ○ Toxicity: minimal difference
    • Earlier phase III RCT (Jeremic, JCO, 1999)
      ○ Improved PFS and OS with addition of thoracic RT, concuurent with further chemo
      - For reponders to 3 cycles of platinum/etop
      - Accelerated hyperfx 54Gy/36Fx BID
60
Q

What is the evidence for PCI in LS-SCLC?

A
  • Multiple RCTS show PCI effective decreasing incidence of brain mets
    • Most unpowered to show meaningful survival advantage
    • OS advantage demonstrated in 2x meta-analysis with similar findings
    • Auperin Meta-analysis (1999)
      ○ PCI improves 3year OS by over 5% for those with complete response
      ○ Some trials defined complete response with CXR and did not image brain
      - 50% imaged brain with CT prior to PCI (No MRI)
    • Meta-analysis (Meert, 2001)
      ○ 12 RCTs with 1547 patients
      ○ CT-staging only
      ○ Outcomes
      - Improved OS in PCI (HR 0.82)
      - Reduced incidence of brain metastases (HR 0.48)
    • January 2024 metanalysis for MRI confirmed no metastasis
      ○ No overall survival benefit from PCI
61
Q

What is the evidence for PCI in ES-SCLC?

A
  • 2x RCTs with opposing results
    ○ EORTC trial (Slotman, 2007)
    - 286 patients with ES-SCLC were randomised to PCI or observation
    □ CT or MRI staging allowed (imaging was not required prior tho)
    □ Multiple fractionations (20Gy/5F, 24Gy/12F, 25Gy/10F, 30Gy/10F)
    - Decreased symptomatic brain mets (15 v2. 40%), improved 1yr OS (27 vs. 13%)
    - PCI associated with improved median OS (6.7mo vs 5.4mo)
    ○ Japanese trial (Takayashi, 2017)
    - 224 patients with ES-SCLC randomised to PCI or observation
    □ MR mandated, and no brain mets vs.
    □ 25Gy/10F mandated
    - Higher BM in surveillance group (33% vs 59%); but
    - PCI no OS advantage (median OS 11.6mo vs 13.7mo)
    • It is contended that in the Slotman trial, a number of patients were having existing macroscopic brain metastases treated (CT occult, but MR would have shown)
      ○ Strict q3mo MR surveillance is an appropriate option
      PCI should be given if patient unable to comply with surveillance
62
Q

What is the evidence for HA-WBRT in LS-SCLC?

A

Can be considered in the PCI setting (no suggestion of oncological deficit)
Data is not transferrable to the brain metastases setting

- PREMER trial (Rodriguez de Dios, 2021)
	○ 150 patients with SCLC undergoing PCI were randomised to
		- HA-WBRT vs WBRT alone (25Gy/10F)
	○ Outcomes
		- HA-WBRT associated with improved cognitive outcomes
		- No difference in oncological outcomes (OS, incidence of brain mets, QoL)
- Belderbos ph3 RCT 2021
	○  168 pt
	○ -no increased mets, no benefit in neurocognitive decline with HA
63
Q

What is the evidence for immunotherapy in ES-SCLC?

A
  • Impower 133 trial (Liu, 2021)
    ○ 403 patients with ES-SCLC randomised to carboplatin/etoposide +/- atezolizumab
    - 4x induction cycles
    - Atezolizumab maintenance to continue until progression/toxicity
    ○ NOTE: thoracic RT was NOT allowed
    - PCI was allowed
    ○ Improved median OS (12.3mo vs 10.3mo)
    • CASPIAN trial
      ○ Platinum/etoposide + durva x 4 cycles > maintenance durva until progression/toxicity
      Improved median OS 13m vs. 10m cf. chemo alone
64
Q

Describe a radiothearpy technique for LS-SCLC with concurrent chemoRT

A

Patients
LS-SCLC
ECOG 0-2

Pre-simulation
MDT discussion
Lung function tests
Aim to commence ASAP (C2 at latest)

Simulation
Supine in vacbag with arms up

CT from mid neck to below diaphragm
- 2mm with IV contrast (Delineation of pulmonary vessels vs tumour)
- 4D-CT with respiratory motion management

Fusion
FDG-PET
- use as guide only
- treat as per residual disease on CT (after C1 chemo)

Dose prescription
1) 45Gy/30F BD (prescribed to PTV as per ICRU 83)
2) 66Gy/33F daily
VMAT technique
10 days per fortnight
Concurrent chemotherapy (q21 days)
- Cisplatin (60mg/m2 on D1)
- Etoposide (120mg/m2 on D1-3)

Volumes*
*Depend on timing with chemotherapy
GTVp = lung disease on CT/PET
iGTVp = GTV on all respiratory phases
ITVp = iGTV + 7mm
PTVp = ITV + 7mm
GTVn = nodal disease on CT/PET
iGTVn = GTV on all respiratory phases
ITVn = iGTV + 5mm
PTVn = ITV + 7mm
*If patient has already received induction chemotherapy. The primary can use post induction chemo volume, but include the pre-chemo nodal volumes in GTVn

Target Verification
Daily CBCT

OARs
For 66Gy/33F
- Bilat Lung (minus iGTV)
○ Mean < 20Gy
○ V20 < 35%
○ V5 < 60%
- Spinal Cord
○ Dmax < 45Gy
- Oesophagus
○ Mean < 28Gy
○ Avoid hotspots
- Heart
○ Mean < 20Gy
○ V50 < 25%

For 45Gy/30F BD
- Bilat Lung (minus iGTV)
○ Mean < 18Gy
○ V18 < 35%
- Spinal Cord
○ Dmax < 41Gy
- Oesophagus
○ Avoid hotspots
- Heart
○ Mean < 20Gy
○ V50 < 25%

65
Q

Describe a radiothearpy technique for ES-SCLC with consolidation RT

A

ES-SCLC with residual disease following chemotherapy (ie. Initial response)
Good response to chemotherapy and minimal extrathoracic disease

Pre-SIM
MDT discussion
Review imaging >ILD
Lung function tests - FEV1, DLCO

SIM
Supine in vacbag with arms up
CT from mid neck to below diaphragm
- 2mm with IV contrast
- 4D-CT with respiratory motion management

Fusion
FDG-PET
- use as guide only
- treat as per residual disease on CT (residual disease after completion of chemo)

Prescription
30Gy/10F (prescribed to PTV as per ICRU 83)
Over 2 weeks, 10Fx/14
VMAT technique
10 days per fortnight

Volumes
GTVp = lung disease on CT/PET
iGTVp = GTV on all respiratory phases (or MIP)
ITVp = iGTV + 7mm
PTVp = ITV + 7mm
GTVn = nodal disease on CT/PET
iGTVn = GTV on all respiratory phases
ITVn = iGTV + 5mm
PTVn = ITV + 7mm

Target varification
Daily CBCT

OAR
For 30Gy/10F
- Bilat Lung (minus iGTV)
○ Mean < 18Gy
○ V18 < 35%
- Spinal Cord
○ Dmax < prescription
- Oesophagus
○ Avoid hotspots

66
Q

Describe a radiothearpy technique for PCI in LS-SCLC

A

No evidence of brain metastases following completion of chemotherapy
- need to restage

Pre-SIM
MDT discussion
MRI brain prior

SIM
Supine with thermoplastic mask
CT from vertex to mid neck
- 2mm with IV contrast

Prescription
25Gy/10F
3D-CRT or VMAT technique
10 days per fortnight

Volumes
CTV = entire brain
- use helmet field (cribriform plate + down to C2)
PTV = 5mm
Hippocampal avoidance

Target varification
Daily CBCT

OARs
Lens
- Dmax < 6Gy
Hippocampus
* Minimum dose 9Gy
* Acceptable D100% ≤10 Gy
* Acceptable maximum dose ≤17 Gy

67
Q

What is the prognosis for LS and ES SCLC?

A

Prognosis:
Untreated, extensive stage SCLC has a median OS of 1-2 months

Median OS 5-year OS
Limited Stage 24mo 15%
Extensive Stage 12mo 5%

68
Q

What are the primary causes for SVCO?

A
69
Q

What are the signs and symptoms of SVCO and those that indicate acute vs chronic development?

A

○ Mediastinal lesion grow to obstruct blood flow from SVC
▪ Either extrinsic compression or direct tumour invasion
○ Chest pain
○ Facial plethora (typically worse in the morning)
○ Arm swelling
○ Dyspnoea
○ Collateral vessels on skin/dilated chest veins
○ Headaches, Confusion, dizziness, syncope
○ Haemoptysis
○ Pemberton’s sign (Patients with SVC obstruction will often become plethoric and/or breathless when both arms are raised above their head for over 60 seconds).

Underlying malignancy:
○ Lung cancer sx: Cough, haemoptyisis, dyspnoea, chest pain, weight loss
B symptoms: Fevers, night sweats, weight loss

Rapid vs slower development of SVCO
Extrinsic compression SVC either by primary or enlarged LN; or via direct invasion
Can form collaterals, however
Rapid tumour grown does not allow adequate time
Cardiac output may be diminished transienty by acute SVCO

70
Q

Describe the grading for SVCO

A

If Grade 3-4 –> Emergency management and stenting
If not suitable for stenting steroids and RT is reasonable alternative

Then aim for work up and tissue diagnosis once patient is stabilised

71
Q

Describe the urgent management of SVCO

A

Urgent Intervention (Grade 3-4)
○ Immediate supportive management
▪ Head elevation
▪ Oxygen
▪ If no contraindications, IV/PO dexamethasone 8mg BD and PPI cover
▪ Anxiolytics/morphine
▪ Consider diuretics (not at cost of intravascular depletion)
○ Stenting
▪ Stenting safe and effective (95%), often with immediate relief of symptoms
- Preferred over RT for following reasons:
○ Objective measurement in change in obstruction may not necessarily parallel symptomatic improvement
- May have some improvement due to collateralisation
○ Relief may not be achieved for up to 4 weeks
- Most do improve within 3-7 days
- Disadvantages of stenting
○ Stent failure: Through stent/SVC thrombus vs extrinsic compression of tumour around the stent
○ Stent migration
○ Infection/perforation/bleeding
○ PE
○ Invasive procedure

*Ideally tissue biopsy post stenting**

○ Radiotherapy
▪ As an initial intervention or adjuvant after stenting
- Most malignancies causing SVCO are radiosensitive
- RT provides relief by reducing tumour burden
- If sx severe consider stenting, followed by RT
- RT effective for symptoms in 75% of patients (vs 95% for stenting) in 3–7 days, but can be weeks
- Single 8 Gy, 20Gy/5 fractions
○ Chemotherapy
▪ NHL, germ cell tumours and SCLC–> chemo sensitive –> first line, especially if treatment naïve
▪ Clinical response usually rapid
- Response to RT is poorer, yields poorer long term results
○ Surgery
- May be considered to bypass of resect tumours (eg. Thymic)
- Thymoma or germ cell

○ Thrombolytics and anticoagulation
▪ As per ESMO guidelines benefit of anticoagulation is unclear, aspirin often recommended after stent placement in the absence of thrombus
▪ If thrombus is present –> therapeutic anticoagulation

72
Q

Describe the epidemiology for Mesothelioma

A

Incidence (Australian statistics)
- Uncommon/rare malignancy
- Age 60-70
- M>F
Incidence is increased with age with previous asbestos exposure (25-50 year latent period)
Decreasing in young people

Marked male predominance
- Occupational exposure –building, shipping, car brakes

Asbestos also causes pleural plaques, lung fibrosis/interstitial disease (pulmonary asbestosis), lung cancer (more commonly than mesothelioma)
Synergistic relationship of smoking with asbestos (RR 60) for lung cancer (but not for mesothelioma)

Biological behaviour:
- Poor prognosis > 5 year OS 15%
○ MS even with treatment 6-18 months
- Local spread
○ Encases lungs –> respiratory failure (restrictive)
○ Pleural effusion
○ Seeds along bx tracts
○ Local invasion
- LN
○ 50% spread, usually to levels 8 + 9
- Mets to contralateral lung, liver, bone, peritoneum
- Paraneoplastic syndromes

73
Q

What are the risk factors for Mesothelioma?

A

1) Asbestos exposure (80% of cases)
a. 10% lifetime risk of mesothelioma amongst asbestos workers or their partners
2) Ionising radiation
a. e.g. Hodgkin Lymphoma survivors following mantle field treatment/ Post testicular cancer radiotherapy
3) Carbon nanotubes
a. Similar function to asbestos
4) Erionite from gravel roads
5) Viral oncogenes
a. Simian virus 40 (SV40) is a polyomavirus with oncogenic potential
i. act as a co-carcinogen via p53 suppression (controversial)
b. (polio vaccine contaminant prior to 1963)
6) Genetic/familial clustering
a. Possible inactivation of BAP1 gene
i. BAP1 germline mutation increases cancer risk if exposed to asbestos
ii. Tumour suppressor gene –helps apoptosis

74
Q

What are the two types of asbestos particles and describe the pathogenesis for mesothelioma.

A

Two types of asbestos
- Amphibole (most carcinogenic)
○ Crocidolite is the most carcinogenic
○ Brittle, fragments, reaches terminal airwars
- Serpentine (limited carcinogenesis)
As longer, and caught in the upper airways and cleared

Poorly understood
* Toxic oxygen radical generation
* Chronic pleural irritation
* Persistent kinase mediated signalling

Fibres of asbestos are thought to trigger a chronic inflammatory response
- Activation of NF-kB pathway is a major event towards carcinogenesis
- Upregulation of PD-L1 is seen
- Mutations in p53 and BAP1 are also commonly seen

Differentiating reactive mesothelial proliferation from mesothelioma
- Loss of expression of BRCA1 associated protein (BAP1) or
- methylthioadenosine phosphorylase (MTAP) or
- homozygous deletion of cyclin dependent kinase inhibitor 2A (CDKN2A) (p16)
- by FISH helps to distinguish

This is a prolonged process and latent period is more than 15 years and up to 50 years

75
Q

What are the histological subtypes for mesothelioma? Describe their macroscopic/ macroscopic appearance and IHC

A

Pleural mesothelioma; Three histological subtypes
- Epithelioid (70%)
- Sarcomatoid (10%)
- Biphasic (20%) - At least 10% of each of the epithelioid and sarcomatoid types

Macroscopic (same for all subtypes)
○ Diffuse lesion arising from visceral/parietal pleural layers
○ Firm grey tumour forms nodules that coalesce in a thick pleural rind
○ Multiple pleural nodules/ studded pleura
○ Can locally infiltrate diaphragm, chest wall, mediastinum

Epithelioid Mesothelioma
- Most common type of mesothelioma 60%
- Best prognosis also
- Microscopic
○ Cuboidal or flattened cells forming sheets, clusters, morules or papillary structures (resembles adenocarcinoma)
§ IHC to distinguish from lung adenocarcinoma
○ Other subtypes = Acinar, adenomatoid and solid
○ Stromal invasion distinguishes from benign hyperplasia
○ Psammoma bodies
- Immunohistochemistry
○ POS = Keratin AE1/AE3, CAM 5.2,
§ CK5/6 (75-100% in meso, 2-20% in lung adeno focally positive),
§ WT1 (70-95%- cf adeno neg),
§ D2-40 (podoplanin, 90-100%), calretinin
§ Surface can be positive in reactive, but full thickness staining in mesothelioma
○ NEG =
§ BAP 1, MTAP, BerEP4
§ Lung adeno markers CEA TTF1, Napsin A
§ Gyane adeno PAX8,
§ Breast :GATA3
§ Serosal mets: claudin 4
- Epithelioid can look like adeno, may need EM to differentiate
○ Meso have long slender microvilli with tonofilaments
§ Adeno have short stubby micovili
Sarcomatoid Mesothelioma
- Least common 10% but worst prognosis
- Defined as >90% spindle cells
○ If 50-90%, then usually termed desmoplastic
- Microscopic
○ Apparent spindle cells arranged in either fascicles or in a haphazard distribution
○ Storiform patterm
○ Can have heterologous differentiation (osteosarcoma, chondrosarcoma, etc.)
- Immunohistochemistry
○ Similar panel as above
§ CK5/6 negative
§ Need to use broader CK stains (e.g. AE1/AE3 and CAM 5.2) as staining may be weaker
§ Exclude other sarcoma diagnoses (CD31 = angiosarcoma, CD34 = solitary fibrous tumour)

76
Q

What are the prognostic factors for mesothelioma?

A

Patient Factors
- Age and performance status
- Female better than male
- Weight loss
- Respiratory function, nonsmoker
- Brief symptomatic period (<6mo)
○ Absence of chest pain is positive
- Bloods
○ Hb
○ Albumin
○ Leukocytosis (is bad)
○ Thrombocytosis and PDGF levels

Tumour Factors
- Histological subtype
○ Epithelioid has best
○ Sarcomatoid is worst
- Limited volume
- TNM staging
○ Especially nodal metastases
○ Limited invasion
- Normal WCC

Treatment Factors
- Complete surgical resection (resectability)
- Chemotherapy (tolerability)

Brims Model (Brims, et al. 2018)
4 prognostic groups based on:
- Weight loss
- Hb
- Performance status
- Histology
- Albumin

77
Q

What is the work up for mesothelioma?

A

History
- Slow-onset of multiple non-specific symptoms
○ Dyspnoea
○ Chest pain
○ Cough
○ Hoarseness
- PMHx
○ Previous radiotherapy
- Social History
○ Smoking is not a risk factor
○ Occupational exposures (e.g. asbestos)
- Note tendency to grow along biopsy/chest tube sites

Examination
- Observations (including saturations)
- Chest auscultation
- Palpate for SCF or axillary lymphadenopathy
- Palpate drain/biopsy tracks for seeding

Investigation

- Imaging
	○ CXR: recurrent pleural effusions 
	○ CT Chest with contrast
		§ Thickening, effusions, contraction of hemithorax
	○ Consider PET-CT
		§ Increased sensitivity and specificity
		§ Helps delineate benign pleural plaques
		§ Nodal or distant mets
	○ MRI if potentially resectable  -  diaphragm, fascia, chest wall invasion 

- Histopathology
	○ FNA –can't make diagnosis
	○ Pleural biopsy
		§ Thoracoscopic (VATS) is preferred
		§ CT-guided core is alternative
		§ Open biopsy
	○ Diagnostic pleurocentesis (50% sensitive)
		§ Pleural effusion is very common
	○ Needs fat invasion to be mesothelioma (so not effusion or FNA)
- Mediastinal staging
	○ EBUS/EUS-FNA or mediastinoscopy 
- Cardiac stress test
- Extended surgical staging (if considering extrapleural pneumonectomy)
	○ Consider mediastinoscopy or EBUS if suspicious mediastinal LN
	○ Consider laparoscopy and peritoneal lavage (exclude peritoneal seeding)
		§ Generally only done if imaging indicates diaphragmatic invasion
		§ To ensure no infra-diaphragmatic surface involvement 

- Lung function test
78
Q

What is the differential diagnosis for a pleural tumour?

A

Differential for pleural tumour
Mesothelioma, lunng adenocarcinoma
Lymphoma, solitary fibrous tumour, sarcoma –synovial or angiosarcoma, metastasis (lung, breast, ovary, thymoma, grastic), benign (infection, inflammation, chronic empyema, reactive pleural hyperplasia, connective tissue disorder)

79
Q

What is the TNM staging for pleural mesothelioma?

A
80
Q

What are the general management options for resectable mesothelioma?

A

Stage I to IIIa (i.e. resectable disease)

- NOTE: only epithelioid mesothelioma should proceed along with curative-intent treatment
	○ Prognosis of sarcomatoid mesothelioma is too grim to justify the toxicity
	○ Can be considered for biphasic tumours
- <5% are resectable at diagnosis
- Considerations
	○ Highly selected 
	○ Good lung and cardiac function 
	○ Aim post op FEV1 >30%, 1L 
	○ Negative mediastinoscopy and laparoscopy pre surgery

Treatment schema

- Surgery -> chemotherapy -> hemithorax radiotherapy
- NACT -> surgery -> Hemithorax radiotherapy 
1. Consider induction chemotherapy
	a. Cisplatin/pemetrexed
	b. Re-stage prior to proceeding with surgery

2. Surgical resection - goal of surgery is macroscopic complete resection, which in reality is an R1 resection in the majority of cases
	a. Optimal is an extrapleural pneumonectomy (EPP) + mediastinal LN sampling
		i. En-bloc oncological resection of parietal/visceral pleurae, ipsilateral lung,  mediastinal LN, hemi-diaphragm and ipsilateral pericardium
		ii. Considerable operative mortality rate (5-30%)
		iii. Local failure ~40%
	b. Alternative is a pleurectomy + decortication + mediastinal LN sampling
		i. Resection of parietal and visceral pleura
			1) Extended pleurectomy includes ipsilateral pericardium + hemidiaphragm
			2) Preserves lung 
		ii. Less-invasive, but generally considered to have reduced chance of cure
		iii. 2.9% peri-operative mortality and 27% morbidity
		iv. Local failure ~70%

10% risk of seeding biopsy tract - this should be resected at time of surgery
	○ Prophylactic biopsy tract RT no longer recommended 

3. Consider adjuvant chemotherapy
	a. Typically only required if neoadjuvant was not given

4) Consider adjuvant radiotherapy
	a. Type of surgery
		i. EPP --> hemi-thoracic RT (no randomised data to support)
		ii. Pleurectomy  --> hemi-thoracic pleural RT
			1) (Phase 3 trial increased OS)
	b. Dose
		i. Adjuvant = 54Gy/27F (limited by OARs)
		ii. Macroscopic residual = 60-66Gy (as able - limited by OARs)
81
Q

What are the general management options for unresectable mesothelioma?

A

Advanced unresectable or Biphasic/sarcomatoid mesothelioma
* NO role for definitive radiotherapy to hemithorax
* Studies have shown no diff in OS and +++ toxicity

1) Palliative chemotherapy
	a. Cisplatin/pemetrexed
	b. Consider addition of immunotherapy (ipilimumab/nivolumab)
		i. Especially for sarcomatoid

2) Palliative Radiotherapy
	a. Typically only given for chest wall pain, etc.
		i. 20Gy/5F or similar
	b. Due to incurability, no rationale for definitive high-dose RT
	
3) Best supportive care
82
Q

What are the advantages and disadvantages for management options of pleural effusions?

A
83
Q

What is the evidence for surgery in mesothelioma?

A
  • EPP vs PD controversial -> P/d associated with less mortality
    • No RCT. Phase 2 series data shows long term OS with highly selected patients
    • MSKCC (Flores et al. J Thoracic Cardio Surg 2008): retrospective review, 663 patients treated with EPP or P/D
      ○ P/D had lower mortality 4 vs 7%
      ○ P/D assoc with improved survival 16 vs 12 months
    • Cao et al (Lung Cancer 2014): MA ot series comparing EPP vs P/D
      ○ P/D had lower peri-op mortality P/D had lower mortality 4 vs 7%
      ○ P/D assoc with improved survival 28 vs 62%
    • MARS (Treasure et al. Lancet Oncol 2011): 50 pts randomised after induction chemo to EPP and hemithoracic RT vs no EPP
      ○ Survival was worse in EPP group  MS 19.5 vs 14.4 months with 3 peri-operative deaths
      Study criticised since primary outcome was feasibility and not powered to detect survival difference
84
Q

What is the evidence for adjuvant radiotherapy post pleurectomy/decortication in mesothelioma?

A
  • IMPRINT prospective phase II trial (Rimner, 2016)
    ○ 45 patients with mesothelioma were enrolled
    § Neoadjuvant chemotherapy (cisplatin/pemetrexed)
    § P/D surgery
    § Adjuvant RT 50.4Gy/28F
    ○ Median PFS = 12mo
    ○ Median OS = 24mo
    ○ Toxicity
    § 2 patients with G3 pneumonitis
    § No G4+ toxicity
    • Italy Phase III RCT - Hemithorax RT improves OS
      ○ 108 patients, any histology
      ○ Lung-sparing surgery included extended P/D, P/D, and partial pleurectomy.
      ○ Adjuvant hemithorax RT (50Gy to pleural cavity +60Gy to gross disease/25#) vs pall RT to surgical scar or gross residual (21Gy/3# to 30Gy/10#)
      ○ 2 year OS 58% vs 28%
      ○ Median OS 25.6mo vs 12.4mo
      ○ In hemithorax arm: 20% acute G3 toxicity, 30% late G3/4 toxicity, 1 patient with G5 toxicity
85
Q

Describe a suitable radiation technique, dose fraction, volumes and OARs for pleural mesothelioma, after EPP or P/D.

A

Simulation
Supine in vacbag with arms up
- Knee supports and ankle stocks
Wire biopsy site, drain sites and surgical scar
0.5cm bolus over these areas (generous coverage)
CT from mid neck to below kidneys
- 2mm with IV contrast
- 4D-CT with respiratory motion management

Fusion - FDG-PET

Dose prescription
R0/R1 resection
- 54Gy/27F (prescribed to the PTV as per ICRU 83)
- If not safe, dose reduce to 50.4Gy/28F

R2 resection
- Boost to 60-66Gy

IMRT/VMAT technique
10 days per fortnight

Volumes
GTV = high-risk or macroscopically evident residual disease
- Often demarcated by surgical clips
CTV54
- Entire surgically violated ipsilateral pleural space
- Note that this includes
○ The apex of the lung
○ The posterior recess (usually extends to lower pole of kidney)
○ The ipsilateral mediastinum
- Encompass surgical/drain sites
- Add 7mm CTV expansion (i.e. chest wall - does not go to skin)
CTV60 = GTV + 7mm
ITV = ensure CTV encompassed on all phases of respiratory cycle (4DCT)
PTV = CTV + 7mm

Target Verification
Daily CBCT
Respiratory motion management (RPM)

OARs
* Contralateral Lung
○ Preferentially spare the remaining lung
○ Mean < 8Gy
○ V20 < 7%
* Spinal Cord
○ Dmax < 45Gy
* Brachial Plexus
○ Dmax < 66Gy
* Oesophagus
○ Mean < 34Gy
○ Avoid hotspots
* Heart
○ V40 < 35%
* Contralateral kidney
○ V15 < 33%
* Liver
○ V30 < 50%
○ Mean < 30Gy

86
Q

Describe the epidemiology for Thymus tumours.

A

Uncommon, but most common anterior mediastinal tumour (50%)
33-50% diagnosed incidentally on CXR

Incidence (International statistics)
- Thymoma = 0.25 per 100000 annually (i.e. 62 people in Australia)
- Thymic Carcinoma = 0.05 per 100000 annually (i.e. 12 people in Australia)

Median age = 52 years
Equal gender distribution
Higher incidence in asians, african americans, pacific islanders
?related to EBV infection

Natural History
- Primary: slow growing, potential for local invasion, locally recurrent
- Nodal: uncommon
Mets: rarely as nodules on pleural or pericardial surfaces, rare to have extrathoracic mets

87
Q

What is the OS by stage and histology, for mesothelioma?

A
88
Q

What are the risk factors for thymic tumours?

A

No clearly defined risk factors, however some suggestions include:

1) EBV infection
	a. Correlation between high-rates of thymoma/carcinoma and endemic EBV in Asia
2) Mediastinal irradiation
	a. Relative increase in risk in childhood leukaemia/lymphoma survivors Family history
89
Q

Describe the WHO classification system for thymic tumours

A
  • Three key classifications
    ○ A = cytologically benign with benign/non-invasive natural history
    ○ B = cytologically benign with malignant/invasive natural history (e.g. capsular invasion)
    ○ C = cytologically malignant
    • Subtypes (6 subtypes)
      ○ Type A = medullary thymoma, spindle cell thymoma○ Type AB = mixed thymoma○ Type B
      § Type B1 = lymphocyte-rich, lymphocytic, predominantly cortical, organoid
      § Type B2 = cortical
      § Type B3 = epithelial, atypical, squamoid, WD thymic carcinoma

Type C = thymic carcinoma

90
Q

Describe the pathological features for Type A,B and C thymic tumours.

A

Thymoma (Types A-B)
- Whilst invasive, tend to be locally recurrent predominantly
○ Rarely can metastasise and very rarely lead to death
- Heterogenous histologies, but grouped and treated based on clinicopathological behaviour
- Tumour cells originate from thymic epithelium (not T-cells)

- Macroscopic
	○ Large anterior mediastinal mass
	○ Nodular, lobulated and encapsulated (fibrous capsule)
	○ Cut surface = grey-tan colour and can be cystic in appearance

- Microscopic
	○ Lobulated architecture with lobules separated by fibrous bands
		§ Can have cystic or necrotic changes
	○ Either spindled/polygonal/epithelioid neoplastic cells with non-malignant T-lymphocytes/thymocytes spaced between
	○ For classification
		§ A = spindled cells; B = polygonal cells
		§ Increasing numbers suggest increasing cytological atypia
- Distinguishing subtypes (WHO histological classification)
	○ Type A - Medullary thymoma
		§ Bland oval to spindle cells
		§ Few thymocytes/lymphocytes
	○ Type AB - Mixed thymoma
		§ As above
		§ Abundance of immature T cells
	○ Type B1 - Predominantly cortical thymoma
		§ Presence of medullary islands (Hassall corpuscle-like elements)
		§ Predominantly thymocytes with scattered neoplastic cells (<3 contiguous epithelial cells)
			□ Ie. Lymphocyte rich
	○ Type B2 - Cortical thymoma
		§ More polygonal/dendritic epithelial cells
		§ Mixed neoplastic cells and thymocytes
		§ Occasional medullary islands
	○ Type B3 - Well differentiated thymic carcinoma
		§ Neoplastic polygonal cells with no thymocytes (lymphocyte poor)
		§ May show considerable cytologic atypia

- Immunohistochemistry
	○ Epithelial cells
		§ POS = PanCK, CK7, EMA, p40, p63 (squamous epithelial origin), PAX8, CEA
			□ Epithelial markers useful to distinguish from lymphoma
		§ NEG = CK20
	○ Thymocytes
		§ POS = TdT, CD1a, CD99

Thymic Carcinoma (Type C)
- Most aggressive subtype
- By definition, has cellular anaplasia

- Macroscopic
	○ Unencapsulated tumour without fibrous septations
	○ Cut surface = firm, hard and gritty surface of grey colour
	○ Necrosis and haemorrhage may be present
- Microscopic
	○ Generally cohesive cellular growth with geographic necrosis
		§ Stromal invasion with desmplastic pattern
	○ Profound nuclear atypia with mitoses and necrosis (much more marked than in B2/B3)
		§ Round/oval nuclei
	○ Hassall corpuscles and medullary differentiation are rare or not present
	○ Multiple subtypes
		§ SCC (keratinising or non-keratinising)
		§ Sarcomatoid
		§ Basaloid
		§ Clear Cell
		§ Adenocarcinoma
		§ Undifferentiated
- Immunohistochemistry
	○ As above
	○ Add CD5, which is common in thymic carcinoma but rare in thymoma
91
Q

What are the prognostic factors for Thymic tumours?

A

Key Prognostic Factors

Patient Factors
- Age and performance status
- Weight loss
- Comorbiditiies
- Fitness for surgery

Tumour Factors
- Tumour histology/type (WHO classification)
○ Carcinoma 5year os 30%
○ Epithelioid (B) worse than spindle (A)
- Stage (TNM or Masaoka)
○ Depth of invasion
○ LN involvement
- Tumour size (>7cm)

Treatment Factors
- Completeness of resection (most important preditor of survival)
- R2 worse
- Adjuvant therapy (limited evidence)

Uncertain significance
- Presence of autoimmune sequelae (e.g. myasthenia gravis)

92
Q

Describe work up for a thymic tumour.

A

History
- Mass effect
○ Chest pain
○ Dyspnoea
○ Cough
○ SVCO
○ Phrenic nerve palsy
- Gradual onset of symptoms cf. lymphoma, small cell, NSCLC
- Paraneoplastic syndrome (very common - 70%)
○ Myaesthenia gravis (diplopia, lid lag, fatigue and proximal weakness)
§ In 45% of Thymoma
§ 15% of pt with MG associated with thymoma
§ Anti-Ach receptor Ab > Ach receptor deficiency at motor end plate > rapi exhaustion of voluntary muscle contractions, and slow return to normal state
○ SIADH
○ Hypercalcaemia
○ Red-cell aplasia
○ Hypogammahlobulinaemia
- PMHx
○ Previous radiation exposure
○ Previous EBV

Examination
- Chest auscultation
- Sternal palpation (tenderness –> invasion)
- Pemberton’s sign
- Palpate neck and axilla for lymphadenopathy

Investigations

- Local imaging
	○ CT Chest +/- MRI
		§ Assess local invasion and resectability
		§ Necrosis, cystic or calcified foci --> thymic carcinoma
		§ Thymic epithelial tumour likely if
			□ Well defined anterior mediastinal mass, not contiguous with thyroid
			□ No other adenopathy

- Bloods
	○ Germ-cell markers (exclude, negative)
		§ AFP, bHCG, LDH
	○ Serum ACh-r Ab (check pre-op to avoid resp failure during surgery)
		§ 50% need Ach inhibition pre-operatively (neostigmine)
	
- Staging imaging
	○ PET/CT
		§ Useful if high-grade (highly sensitive)
		§ Often negative for lower grade
	○ CT NCAP is alternative
		§ With contrast
	○ MRI for local staging (if unable to tolerate contrast for CT)

- Histopathology
	○ Discuss above results within context of MDT
		§ If highly suspicious for thymoma --> proceed to surgical resection
		§ If uncertain --> percutaneous or open core biopsy
		§ If unresectable --> percutaneous or open core biopsy

- PFTs
93
Q

What is the differential diagnosis for mediastinal tumours?

A

Differential Dx of anterior mediastinal mass
- Thymoma
- Thyroid -retrosternal goiter
- Teratoma/ Germ cell tumour
- Terrible lymphoma
- Carcinoid
- Metastasis
Others: reactive lymph nodes, thymic hyperplasia, thoracic Ao Aneursym

Middle mediastinum - M’s
1. Metastses to LNs
o Lung, H&N, oesophageal, breast, melanoma, other
o DDx: Infection, sacroid, castleman
2. Lymphoma
3. Mesenchymal
o Benign - atrial myxoma, lipoma, haemangioma
o Malignant - sarcoma
o Cysts - pericardial

Posterior mediastinum - N
· Neurogenic
o Benign - schqannoma, neurofibroma, gangioneuroma
Malignant - Malignant peripheral nerve sheath tumour (MPNST), malignant schwannoma

94
Q

Describe the staging system for Thymic tumours

A
95
Q

What are the indications for Radiotherapy in management of thymic tumours?

A

1) Adjuvant radiotherapy
a. All Masaoka stage III thymoma (macroscopic invasion of adjacent organs (e.g. lung, pericardium))
b. All thymic carcinomas (R0 or R1)
c. R1 resection (microscopic margins) for any stage

2) Definitive radiotherapy
	a. Unresectable
	b. R2 resection (macroscopic residual)
	c. Can be 'curative' in oligomet

Histological grade does not impact decision for adjuvant radiotherapy (exception is carcinoma)
- Treat B3 as you would B1

95
Q

Discuss in general the management of Thymic tumours.

A

Resectable Disease:
- Completely encapsulated tumours, or tumours invading readily resectable adjacent structures (mediastinal pleura, pericardium, adjacent lung)
- If myaesthenia gravis present, this must be managed prior
○ Anaesthetic risk
○ Refer neurologist
- If thymoma suspected (Well defined mass, no nodes, negative tumour markers, thyroid not involved)
○ Proceed with upfront resection
§ Median sternotomy with local lymph node dissection. Check for pleural mets
§ Total thymectomy

Potentially Resectable Disease
- Biopsy (percutaneous or open)
- Neoadjuvant chemotherapy should be given
○ Thymoma = Cisplatin/etoposide (as per NSCLC or SCLC)
○ Carcinoma = Carboplatin/Paclitaxel
- Re-stage and assess prior to surgery
○ If resectable, proceed with surgery –> consider adjuvant radiotherapy
○ If unresectable, proceed with safe de-bulking –> then consider radiotherapy

Unresectable Disease
- Safe debulking surgery should be considered
- Should then be treated with chemoradiotherapy
○ As per NSCLC or SCLC protocols
§ 60Gy/30F with concurrent cisplatin/etoposide

96
Q

Discuss in general the management of thymic neuroendocrine tumorus.

A

Localized disease (Stage I-II)
* Surgery: Total resection is preferred over partial resection. Open thymectomy is recommended as standard of care.

Resectable locoregional disease (Stage IIIA/B)
* Surgery: See Stage I-II above.
* Incomplete resection and/or positive margins with low grade (typical carcinoid): Consider observation or RT.
* Incomplete resection and/or positive margins with intermediate grade (atypical carcinoid): Consider observation or RT ± cytotoxic chemotherapy.
○ Chemoradiation is thought to have the most efficacy for tumours with atypical histology or tumours with higher mitotic and proliferative indices.
○ Cytotoxic chemotherapy option include cisplatin + etoposide or carboplatin + etoposide.

Unresectable locoregional disease (Stage IIIA/B)
* For symptom control, consider addition of focal therapy (i.e., endobronchial therapy debulking/ablation).
* Primary therapy, low grade (typical carcinoid):
○ Observation (if asymptomatic)
○ Octreotide or lanreotide (if SSR+ and/or hormonal symptoms) or everolimus, or TMZ ± capecitabine or RT.
* Primary therapy, intermediate grade (atypical carcinoid):
○ Observation (if asymptomatic and non-progressive)
○ RT ± concurrent cisplatin + etoposide or carboplatin + etoposide (CCRT is thought to have most efficacy for tumours with atypical histology or tumours with higher mitotic and proliferative indices)
○ Cytotoxic chemotherapy with cisplatin + etoposide or TMZ ± capecitabine or octreotide or lanreotide (if SSR+ and/or hormonal symptoms) or Everolimus
* Subsequent therapy: If disease progression, treatment with octreotide or lanreotide should be discontinued for non-functional tumours and continued in patients with functional tumours; those regimens may be used in combination with any of the subsequent options.

97
Q

What is the evidence to support management of Thymomas?

A

Thymoma

- Multiple retrospective series have suggested no clear benefit to adjuvant RT for stage I-II disease
	○ Somewhat contradictory results for stage II (unclear benefit)
- Clear benefit for Stage III

2022 metaanalysis falkson (nonrandomised trials)
Rt benefit in stage 3+
Rt benefit in r0 ?
R1/r2 benefit improved overall survival
For stage 1/2 complete resection no benefit.
Stage 3/4 complete resection not statistically significant

- Meta-analysis (Lim, 2016)
	○ 7 retrospective series with 1724 patients (thymoma only - no carcinoma)
	○ With respect to OS benefit with PORT
		§ Overall cohort --> no benefit (HR 0.79)
		§ Stage II --> no benefit (HR 1.45)
		§ Stage III + IV --> OS benefit seen (HR 0.63)	
- Retrospective NCDB analysis (Jackson, 2017)
	○ 4056 patients (either thymoma or carcinoma)
	○ PORT was associated with
		§ Improved OS in IIb & III thymoma
		§ No significant difference for I-IIa thymoma
- Retrospective SEER analysis (Forquer, 2010)
	○ 901 patients (either thymoma or carcinoma)
	○ PORT was found to have
		§ No significant benefit in Stage I disease
		§ Borderline in stage II and III (OS benefit, but no CSS benefit)
			□ This benefit was seen particularly after incomplete resection

B3 Thymoma

- Retrospective series (Gao, 2013)
	○ 188 patients all with B3 thymoma
		§ 30% with Masaoka I-II
	○ In stage I-II, adjuvant RT was not associated with OS or PFS benefit
		§ Benefit was isolated to stage III-IV patients
98
Q

What is the evidence to support management in Thymic carcinoma?

A

Thymic Carcinoma

- Retrospective series suggest OS advantage with RFS and OS
	○ It is unclear which subgroups/stages will benefit most
- Meta-analysis (Hamaji, 2017)
	○ 973 patients with thymic carcinoma only (including SEER data)
	○ PORT resulted in
		§ Improved OS (HR 0.66)
		§ Improved PFS (HR 0.54)
- Japanese retrospective series (Omasa, 2015)
	○ 1265 patients with II or III thymoma or carcinoma
	○ PORT associated with
		§ Improved RFS in carcinoma
		§ No OS benefit in carcinoma
		§ No RFS or OS benefit in thymoma
99
Q

Describe a suitable Radiation technique, dose fractionation, volumes and OARs for Adjuvant thymic tumour treatment.

A

Patients
1) Masaoka Stage III
2) Incomplete resection (R1)
3) All thymic carcinoma (R0 or R1)

Pre-simulation
MDT discussion
Lung function tests

Simulation
Supine in vacbag
Arms up
CT from mid neck to below diaphragm
- 2mm with IV contrast
- 4D-CT with respiratory motion management

Fusion
FDG-PET
Pre-op MR chest (if performed)

Dose prescription
R0 = 50Gy/25F
R1 or carcinoma = 54Gy/27F
VMAT technique
10 days per fortnight

Volumes
GTV = gross residual disease
CTV
- GTV + 7mm
- Entire surgical bed
- Use pre-op imaging to help guide “at risk” region
ITV = CTV on all respiratory phases
PTV = ITV + 7mm

Target Verification
Daily CBCT
Respiratory motion management (RPM)

OARs
As per definitive lung
- Bilat Lung (minus iGTV)
○ Mean < 20Gy
○ V20 < 35%
○ V5 < 60%
- Spinal Cord
○ Dmax < 45Gy
- Oesophagus
○ Mean < 28Gy
○ Avoid hotspots
- Heart
○ Mean < 20Gy
○ V50 < 25%

100
Q

Describe a suitable Radiation technique, dose fractionation, volumes and OARs for Definitive thymic tumour treatment.

A

Patients
1) Unresectable disease
2) All R2 disease (thymoma or carcinoma)

Pre-SIM
MDT discussion
Lung function tests
Supine in vacbag
Arms up

SIM
CT from mid neck to below diaphragm
- 2mm with IV contrast
- 4D-CT with respiratory motion management
FDG-PET
Pre-op MR chest (if performed)

Prescription
Standard = 60Gy/30F
- Consider boost to 66Gy/33F if definitive (especially if carcinoma)
VMAT technique
10 days per fortnight
Concurrent chemotherapy (q21 days)
- Cisplatin (60mg/m2 on D1)
- Etoposide (120mg/m2 on D1-3)

Volumes
GTV = gross residual disease
CTV
- GTV + 7mm
- Entire surgical bed (if applicable)
ITV = CTV on all respiratory phases
PTV = ITV + 7mm

Target verification
Daily CBCT
Respiratory motion management (RPM)

OARs
As per definitive lung
- Bilat Lung (minus iGTV)
○ Mean < 20Gy
○ V20 < 35%
○ V5 < 60%
- Spinal Cord
○ Dmax < 45Gy
- Oesophagus
○ Mean < 28Gy
○ Avoid hotspots
- Heart
○ Mean < 20Gy
○ V50 < 25%

101
Q

What is the 10 year OS and LF rates for the different Masaoka stages of thymic tumours?

A

10 year OS Local Failure
Masaoka I 100% 1-3%
Masaoka II >95% 27%
Masaoka III 80%
Masaoka IV 50%

102
Q

Describe the advantages and disadvantages of the different methods of obtaining a tissue diagnosis for lung cancer

A
103
Q

What are the local treatment options for endobronchial progression with near occlusion. Include advantages and disadvantages.

A

Endobronchial stenting:
- Adv: quick and immediate relief of occlusion, flexible, active expansion, one stage procedure, improve QoL
- Disad: limited to the size and central tumours/location of the occlusion, temporary would still require Radiotherapy to prolong duration of stent, risk of bleeding + infection, stent tumour ingrowth, and stent migration, invasive procedure and limited to operator skills, tumour ingrowth, risk of fistula

Bronchoscopic Laser diathermy or mechanical debridement
- Adv: quick relief of symptoms, haemostasis, recanalisation prior to brachytherapy, cheap, available in many centres
- Disad: limited to truly central lesions, GA risk, risk of perforation, expensive equipment

Palliative radiotherapy EBRT
- Adv: quick and easy to plan, non invasive approach, no risk of bleeding, infection or perforation, not limited by location and extend of narrowing
- Disadv: limited to people who had previous high dose RT, risk of worsening occlusion due to RT related oedema of the mass, slow to response to RT, side effect of RT eg pneumonitis

Palliative endobronchial brachytherapy
- Adv: local RT dose delivery, high dose delivery to minimise dose to surrounding OAR
- Disav: limited availability, limited to location and size of occlusion, dependent on the operator skills and experience, risk of radiation bronchitis, fistula and stenosis, occupational radiation exposure

Dexamethasone:
- Adv: no re-irradiation toxicitiy risk non invasive, simple daily medication orally or via IV
- Disadv: temporary decrease oedema contributing to narrowing, only symptomatic relief, no anti-tumour effect, metabolic side effect from prolonged course of steroids.

104
Q

How would you manage M1a NSCLC?

A

M1a (intrathoracic metastasis)
* Contralateral lung nodule: Treat as two primary lung tumours if both curable
* Malignant pleural/pericardial effusion: Local therapy if necessary (e.g. pleurodesis, small catheter drainage, pericardial window) then as for M1b/M1c
○ **Treat as M1c disseminated disease. Pleural effusions excluded from oligomets phase II trials and the ongoing phase III SARON trial.
Pleural nodule: As for M1b

105
Q

How would you manage M1b NSCLC?

A

M1b (solitary extrathoracic metastasis)
* Brain metastasis (manage brain met first):
○ See management of brain metastases- see below
○ May involve surgical resection +/- cavity FSRS or SRS +/- WBRT
* If definitive therapy for thoracic disease feasible:
○ Systemic therapy and restaging to confirm non-progression
(OR proceed directly to definitive therapy) in NCCN guidelines, but not done in the phase II trials
○ Treat thoracic disease as per T and N stage
2x Phase II RCTS Gomez 2016 (n=76), Lyengar 2018 (n=29); improved PFS (12 vs 4mo) and median OS (41 vs 17mo) cf. maintenance systemic therapy alone
SARON Phase III RCT in progress
○ Then maintenance systemic therapy or observation
* Duration of therapy and maintenance therapy (for patients without a driver mutation)
▪ For patients treated with TKIs - generally continue until progression or significant toxicity
▪ For patients initially treated with combination chemotherapy, treatment is generally limited to 4-6 cycles
▪ In the absence of disease progression, subsequent maintenance systemic therapy has been shown to prolong PFS and OS (the alternative is close observation)
▪ Options for maintenance therapy are in the table below

				* If definitive treatment of thoracic disease not feasible, treat as per M1c (systemic therapy)
106
Q

How would you manage M1c NSCLC?

A

Stage IVB (M1c)
* Palliation depending on PS, systemic or local sx.
* Management of local problems:
○ Thoracic: XRT, laser, stent, pleural tap, pleurodesis
○ Extra-thoracic: CT, biological agents, XRT (brain mets, bone mets), fixation
* Systemic therapy (first line):
○ Adenocarcinoma, large cell carcinoma
* EGFR mutation positive: erlotinib or gefitinib or osimertinib
* ALK positive: alectinib (preferred) or crizotinib
* ROS1 positive: crizotinib
* BRAF V600E positive: dabrafenib + trametinib
* No driver mutation:
▪ PD-L1 >=1%: pembrolizumab (preferred if PD-L1>=50%) OR platinum + pemetrexed + pembrolizumab OR carboplatin + paclitaxel + bevacizumab + atezolizumab
▪ PD-L1 <1%: platinum-based doublet chemotherapy +/- pembrolizumab or bevacizumab (or a number of other options)

			○ Squamous cell carcinoma
				* PD-L1 >=1%: pembrolizumab (preferred if PD-L1 >=50%); OR carboplatin + paclitaxel + pembrolizumab OR Cemiplimab
				* PD-L1 <1%: Platinum-based doublet chemotherapy +/- pembrolizumab (or a number of other options)
 
Note: 
	○ Patients with high tumour burden or rapidly progressing disease, give doublet chemotherapy with pembrolizumab (as per KEYNOTE-189 [non-squamous] or KEYNOTE 407 [Squamous])-> improve mOS, mPFS regardless of the PD-L1 status cf to chemo alone.
	○  Contraindications to immunotherapy include: Connective tissue, rheumatologic or interstitial lung disease
107
Q

What is the evidence for management of Oligometastatic NSCLC?

A
  • Guidelines (NCCN):
    ○ Well-selected pts with good PS
    ○ If brain mets present -> treat these first
    ○ If definitive therapy for thoracic disease feasible:
    * Consider systemic therapy and re-staging to confirm non-progression; OR
    * Proceed directly to definitive therapy
    * Treat thoracic disease as per T and N stage (i.e. with surgery, SBRT, or conventional RT +/- concurrent chemo)
    ○ If definitive treatment of thoracic disease not feasible, treat as per M1c
    • Evidence:
      ○ The best evidence comes from 2x randomised phase II trials which demonstrate improved PFS and OS in patients with synchronous oligometastatic disease who respond to initial systemic therapy
      ○ Further supported by several non-randomised studies
      * Gomez et al lancet oncol 2016
      ▪ Design: Phase II RCT (multicentre)
      ▪ Population (n=74): Stage IV NSCLC, <=3 metastases, ECOG PS <=2, no disease progression with standard first line systemic therapy
      ▪ Initial systemic therapy (all patients):
      * 4 or more cycles of platinum doublet chemotherapy, OR
      * 3 or more months of EGFR or ALK TKI
      ▪ Arm 1: Local consolidative therapy (chemoRT or resection of all lesions) +/- subsequent maintenance systemic therapy
      ▪ Arm 2: Maintenance treatment alone (which could include observation only)
      ▪ Outcomes:
      * Consolidative local therapy improved PFS (primary endpoint) 12 months vs. 4 months (SS)
      * Updated results presented at ASCO 2018 additionally demonstrated improved median OS 41 vs. 17 months (SS)
      ▪ Commentary:
      * First randomised trial to find a survival benefit for local consolidative therapy
      * Limitations:
      * Limited sample size (n=76)
      * Molecular heterogeneity (including EGFR and ALK patients)
      * Definition of oligometastatic disease (which was defined after systemic therapy)
      * Lyengar et al. JAMA oncology 2018
      ▪ Design: Phase II RCT (single centre)
      ▪ Population (n=29): Stage IV NSCLC, <=5 metastases without driver mutation, PR or SD after induction chemotherapy
      ▪ Arm 1: SABR to all sites of gross disease (including SABR or hypofractionated RT to the primary) followed by maintenance chemotherapy
      ▪ Arm 2: Maintenance chemotherapy
      ▪ Outcomes:
      * Consolidative SABR associated with improved PFS (10 months vs. 4 months)
      * Toxicity similar between arms
108
Q

What is the evidence for adjuvant immunotherapy for completely resected NSCLC?

A

Suitable for COMPLETED RESECTED NSCLC Stage IIIA, PDL1>1%, or EGFR mutant-> Improve DFS significantly. No OS benefit data as inmature

- Adj Atezolizumab :
	○ Indication: completely resected stage II to IIIA NSCLC after at least 1 (up to 4) cycles of cisplatin-based doublet adjuvant chemotherapy in patients with tumour PD-L1 expression ≥50%, without EGFR or ALK genomic tumour aberrations
	○ Evidence: IMPOWER: StII-III resected NSCLC-> Atezo vs BSC.
		§ improved 3yr DFS for Stage II-IIIA pt with PD-L1 >1% 60 vs 56%, benefit most noticeable if PDL-1 > 50%
		§ OS inmature
- Adj Osimertinib:
	○ Indication: completely resected stage IB to IIIA NSCLC with EGFR mutation (exon 19 deletion or L858R)
	○ Evidence ADAURA: Stage IB-IIIA EGFR mutant NSCLC-> Adj Chemo-> Adj osimertinib vs placebo
		§ Improved 4yr DFS38 -> 73%, HR 0.27
		§ OS inmature
		§ Lowered disease recurrence 27 vs 60% and CNS events
	○ Caveat: not everyone had PETCT and MRI brain for staging. Patients with occult metastatic disease were likely included in this trial, strengthening the experimental arm
109
Q

What is the evidence to support neoadjuvant chemotherapy prior to surgery?

A

NA chemo-> Sx vs Sx alone?
- This question was addressed (Burdett et al. JTO 2007) by a meta-analysis (not individual patient data) of 12 RCTs which compared neoadjuvant chemotherapy + surgery vs. surgery alone across all stages of NSCLC.
- Induction chemotherapy was associated with a 6% improvement in 5-year OS (from 14% to 20%).
- Because the MA did not use individual patient data, it was unclear which subgroups achieved the greatest benefit, or if some subgroups did not benefit.

NA Chemo-> Sx vs Sx-> Adj Chemo?
- NAC vs. adjuvant chemo yields no difference in EFS or OS [NATCH RCT].
Critique: Lots of stage I patients who are not thought to benefit from chemo.
- Nearly 100% compliance with preop chemo, only 66% compliance with post op chemo.

NA Chemo-> Sx vs NA ChemoRT -> Sx?
- No diff in EFS or OS between NA Chemo vs NA CRT, hence, no benefit to the addition of neoadjuvant radiotherapy to neoadjuvant chemotherapy, except for increase pCR and mediastinal downstaging
○ The Swiss SAKK16/00 (Lancet, 2015) provides the most straightforward answer to this question. In patients with stage IIIA (N2) NSCLC, sequential induction chemotherapy then radiotherapy (to 44Gy/22#) followed by surgery was compared to induction chemotherapy followed by surgery. There was no difference in event free survival or overall survival.
The German GLCCG RCT (Lancet Oncol, 2008) answers a similar question. Neoadjuvant chemotherapy -> radiotherapy -> surgery was compared to neoadjuvant chemotherapy -> surgery -> adjuvant radiotherapy. Neoadjuvant RT improved pathological response and mediastinal down-staging. However, there was no difference in PFS between the two groups (5-year PFS 16% vs. 14%).

110
Q

What is the evidence for neoadjuvant chemoRT compared to definitive chemoRT?

A

NA ChemoRT-> Sx vs Def ChemoRT?
- NA ChemoRT = Def CRT
- No diff in OS, although ChemoRT->Surgery may offer improved PFS (5-yr 22% vs. 11% in INT-0139)
- Caveat: risk of repopulation and Tumour regrowth in the 4-6weeks post CRT until surgery
- Note that there is also RCT data that induction chemo -> surgery is equivalent to induction chemoRT -> surgery

	○ Phase III Intergroup-0139 RCT (Albain, Lancet 2009).
	Patients with T1-3N2M0 NSCLC were all treated with chemoRT to 45Gy then randomly assigned to completion of chemoRT to 61Gy vs. surgical resection. There was no difference in OS. Surgery improved PFS (5yr PFS 22% vs. 11%).
	 
	○ ESPATUE was a similar phase III RCT which did not meet accrual. There were excellent 5-yr OS (~40%) and PFS (~35%) outcomes with no difference between the arms. Both considered acceptable treatment options.
111
Q

What is the evidence for Neoadjuvant immunotherapy, prior to surgery?

A

NA 3x Nivo+doublet chemo improve PCR rate and EFS preferentially in Stage IIIA, PD-L1 pos, non-squamous histo.
mOS not reached
Critics:
- Need to determine surgical resectability UPFRONT and to not count of disease regression to allow for surgery, as if not a candidate, then lack of receipt of surgery would create a “data-free zone” where there is essentially no data to support CCRT after neoadjuvant chemo-IO therapy.

Checkmate  816 Ph3 RCT - 3x NA ICI-> Sx vs 3x NA CT -> Sx
	○ 358 adults with stage IB (≥ 4 cm) to IIIA (7th edition; 64% were IIIA) resectable NSCLC. No known EGFR/ALK alterations. 
	○ 3x Nivo+doublet chemo -> Surg vs 3x doublet chemo -> Surg
	○ Chemo regimen:
		§ Carboplatin / paclitaxel for any histo
		§ Cis / Gem for Squamous
		§ Cis/ permetrexed for non-squamous 
	○ Definitive surgery was performed within 6w of treatment. MFU 30 mo.
	○ Definitive surgery in 75→ 83%. R0 in ~80%. 
		§ Aggressive surgery performed: Roughly 20% received sleeves, bi-lobectomies, or pneumonectomies in the experimental arm, c.f. around 35% in the control arm
	○ Nivo+CT improved pCR and EFS. pCR 2→ 24%. mEFS 21→ 32 mo. 2y EFS 45→ 64%. 
	○ Subgroup analysis favoured Stage IIIA, PDL1>1%, and Non-squamous group-> Hence this pt are more likely to benefit from this approach
	○ mOS not reached
	○ G3/4 toxicity 5.7% of pt cf 3.4% in chemo arm alone arm. Slight increase of adverse event leading to cancellation or surgery in 1.1% vs 0.6%.
112
Q

Discuss heart constraints in lung radiotherapy

A

IF MHD <4-4.5Gy, then risk is insignificant. However each increase in MHD >5Gy, yields the same excess risk of cardiac event as an increase in Anthracycline dose of 50mg/m2 (eg. 1x cycle of ABVD or CHOP)

Latent toxicities of Cardiac dx include Arrhythmias, MI, pericarditis, myocarditis, pericardial effusion, cardiac death.
· Stanford data (1998) 3.9% survivors died from cardiac dx at 10yrs. RR death 3.1, but only in pts getting >30Gy to mediastinum.
· Related to XRT field size, risk lingers even 30+ yrs after Rx.

113
Q

List and briefly describe the paraneoplastic syndromes associated with SCLC.

A
  • Endocrine (due to ectopic hormone production; usually reversible with successful anticancer therapy)
    * Syndrome of inappropriate antidiuretic hormone secretion (SIADH)
    * Mechanism: Ectopic vasopressin (ADH) secretion
    * Clinically significant hyponatremia in 5-10% of SCLC
    * Malaise, weakness, confusion, obtundation, volume depletion, nausea
    * Hyponatremia, euvolemia, low serum osmolality, inappropriately concentrated urine osmolality, normal thyroid and adrenal function
    * Cushing’s syndrome
    * Mechanism: Ectopic ACTH secretion
    * Weight gain, moon facies, hypertension, hyperglycaemia, generalised weakness
    * High serum cortisol and ACTH, hypernatremia, hypokalemia, alkalosis
    • Neurologic: All specific syndromes rare
      • If paraneoplastic syndrome suspected, consider obtaining a comprehensive paraneoplastic antibody panel
      • Subacute cerebellar degeneration
        • Mechanism: anti-Yo antibody
        • Sx: Ataxia, dysarthria
      • Encephalomyelitis
        • Mechanism: ANNA-1 (anti-Hu) antibody
        • Sx: Ataxia, dysarthria
      • Sensory neuropathy
        • Mechanism: Anti-dorsal root ganglion antibody
        • Sx: Pain, sensory loss
      • Lambert-Eaton Myaesthenic Syndrome (LEMS)
        • Mechanism: Anti-voltage-gated calcium channel antibody
        • Sx: Weakness, autonomic dysfunction
      • Cancer-associated retinopathy
        • Mechanism: Anti-recoverin antibody
        • Sx: Visual loss, photosensitivity
    • Haematologic
      • Anaemia of chronic disease
      • Leukemoid reaction - leukocytosis
      • Trosseau’s phenomenon - migratory thrombophlebitis

Note: The paraneoplastic syndrome particularly associated with squamous cell lung cancer is hypercalcaemia secondary to PTHrP secretion

114
Q

What are the advantages and disadvantages to the VA staging of SCLC compared to TNM?

A

Advantages
Simplicity and Ease of Use: The VALCS was simpler than the TNM system, making it easier for clinicians to quickly assess a patient’s disease stage and likely prognosis. This can be particularly beneficial in settings where quick decisions need to be made or when detailed imaging and diagnostic resources are limited.

Focus on Treatment Planning: For its time, the VALCS provided a straightforward approach to categorizing patients in a manner that was directly relevant to the treatment options available, including considerations for surgery, radiation, and chemotherapy.

Historical Data and Research: Studies and treatment outcomes based on this system can offer valuable historical insights, especially for longitudinal studies or when comparing the evolution of treatment efficacy over time.

Disadvantages
Less Precision Compared to TNM: The TNM staging system offers a more detailed and nuanced approach to staging, which can lead to more tailored and potentially effective treatment plans. The VALCS’s simpler approach might not capture the full complexity of a patient’s cancer, especially with SCLC, which has a very aggressive and variable clinical course.

Limited Prognostic Value for Modern Therapies: As new therapies and diagnostic techniques (like PET scans and advanced molecular testing) have become standard, the VALCS might not offer the same prognostic value as the TNM system, which has been updated to incorporate these advances.

Reduced Relevance in Current Clinical Practice: With the widespread adoption of the TNM staging system and its regular updates to reflect advancements in cancer diagnosis and treatment, the VALCS might not align well with current clinical guidelines and practices, potentially leading to confusion or misalignment with standard care pathways.

Lack of International Standardization: The TNM system is used internationally, which facilitates collaboration, research, and comparison of treatment outcomes across different countries and healthcare systems. The VALCS, being less commonly used, might not offer the same level of international standardization

115
Q

What is the evidence for adjuvant immunotherapy/ Targeted therapy after surgery in NSCLC?

A

Adjuvant Immunotherapy (Start after completion of Adj Chemo)
Suitable for COMPLETED RESECTED NSCLC Stage IIIA, PDL1>1%, or EGFR mutant-> Improve DFS significantly. No OS benefit data as inmature

- Adj Atezolizumab :
	○ Indication: completely resected stage II to IIIA NSCLC after at least 1 (up to 4) cycles of cisplatin-based doublet adjuvant chemotherapy in patients with tumour PD-L1 expression ≥50%, without EGFR or ALK genomic tumour aberrations
	○ Evidence: IMPOWER: StII-III resected NSCLC-> Atezo vs BSC.
		§ improved 3yr DFS for Stage II-IIIA pt with PD-L1 >1% 60 vs 56%, benefit most noticeable if PDL-1 > 50%
		§ OS inmature
- Adj Osimertinib:
	○ Indication: completely resected stage IB to IIIA NSCLC with EGFR mutation (exon 19 deletion or L858R)
	○ Evidence ADAURA: Stage IB-IIIA EGFR mutant NSCLC-> Adj Chemo-> Adj osimertinib vs placebo
		§ Improved 4yr DFS38 -> 73%, HR 0.27
		§ OS inmature
		§ Lowered disease recurrence 27 vs 60% and CNS events
	○ Caveat: not everyone had PETCT and MRI brain for staging. Patients with occult metastatic disease were likely included in this trial, strengthening the experimental arm
116
Q

What is the evidence for neoadjuvant chemotherapy prior to surgery for NSCLC and how does this compare to adjuvant chemo?

A

NA chemo-> Sx vs Sx alone?
- This question was addressed (Burdett et al. JTO 2007) by a meta-analysis (not individual patient data) of 12 RCTs which compared neoadjuvant chemotherapy + surgery vs. surgery alone across all stages of NSCLC.
- Induction chemotherapy was associated with a 6% improvement in 5-year OS (from 14% to 20%).
- Because the MA did not use individual patient data, it was unclear which subgroups achieved the greatest benefit, or if some subgroups did not benefit.

NA Chemo-> Sx vs Sx-> Adj Chemo?
- NAC vs. adjuvant chemo yields no difference in EFS or OS [NATCH RCT].
Critique: Lots of stage I patients who are not thought to benefit from chemo.
- Nearly 100% compliance with preop chemo, only 66% compliance with post op chemo.

117
Q

What is the evidence for neoadjuvant chemo/immunotherapy prior to surgery for NSCLC?

A

NA 3x Nivo+doublet chemo improve PCR rate and EFS preferentially in Stage IIIA, PD-L1 pos, non-squamous histo.
mOS not reached
Critics:
- Need to determine surgical resectability UPFRONT and to not count of disease regression to allow for surgery, as if not a candidate, then lack of receipt of surgery would create a “data-free zone” where there is essentially no data to support CCRT after neoadjuvant chemo-IO therapy.

Checkmate  816 Ph3 RCT - 3x NA ICI-> Sx vs 3x NA CT -> Sx
	○ 358 adults with stage IB (≥ 4 cm) to IIIA (7th edition; 64% were IIIA) resectable NSCLC. No known EGFR/ALK alterations. 
	○ 3x Nivo+doublet chemo -> Surg vs 3x doublet chemo -> Surg
	○ Chemo regimen:
		§ Carboplatin / paclitaxel for any histo
		§ Cis / Gem for Squamous
		§ Cis/ permetrexed for non-squamous 
	○ Definitive surgery was performed within 6w of treatment. MFU 30 mo.
	○ Definitive surgery in 75→ 83%. R0 in ~80%. 
		§ Aggressive surgery performed: Roughly 20% received sleeves, bi-lobectomies, or pneumonectomies in the experimental arm, c.f. around 35% in the control arm
	○ Nivo+CT improved pCR and EFS. pCR 2→ 24%. mEFS 21→ 32 mo. 2y EFS 45→ 64%. 
	○ Subgroup analysis favoured Stage IIIA, PDL1>1%, and Non-squamous group-> Hence this pt are more likely to benefit from this approach
	○ mOS not reached
	○ G3/4 toxicity 5.7% of pt cf 3.4% in chemo arm alone arm. Slight increase of adverse event leading to cancellation or surgery in 1.1% vs 0.6%.
118
Q

What is the evidence for hypofractionation in NSCLC?

A
  • Accelerated hypofractionated image-guided vs conventional radiotherapy for pts with stage I/II NSCLC – Lyengar et al, JAMA 2021.
    ○ 103 patients, RCT. NSCLC stage II-III, PS ≥2
    ○ Hypofractionated RT 60Gy/15# Vs Conventional fractionated RT 60Gy/30#
    ○ Accelerated hypofractionated RT is not superior to conventional fractionation in locally advanced lung cancer. But may represent a viable treatment alternative for those unable to have chemotherapy.
    • SOCCAR trial – Maguire et al, 2014
      ○ 130 patients, Phase II RCT. Inoperable stage III NSCLC
      ○ 55Gy/20# RT with:
      § Sequential chemo vs concurrent chemo
      ○ Hypofractionation with 55Gy/20# for lung cancer is safe
    • 55Gy/20# is a recommended fractionation schedule as per the NICE guidelines
    • Common regimen used in the UK - Din et al
      ○ Retrospective data from 4 UK centre RT alone 55gy/20Gy vs concurrent/sequential ChemoRT
      ○ 2yr median OS 24mo, 2yr OS 50%
    • GRIN trial - T1-2 N0-1 NSCLC RT 55Gy/20F with vs without gembitabine.
      ○ 5yr EFS 20% vs 31% NS; 2y OS 56% vs 52% NS; 5yr OS 20% vs 33% NS
    • Pemberton et al. 2009 (55 Gy in 20 fractions) and Lester et al. 2004 (50-55 Gy in 15-20 fractions), reporting 2yr OS of 45% and 44.4%, respectively.
119
Q

What are the advantages and disadvantages for induction chemo/immunotherapy then surgery for NSCLC?

A

Induction immunochemo –> surgery
Upfront resectable, PDL>1%, no mutations
- Advantages
○ Downstaging to make surgery easier
○ Can assess tumour biology via response
○ Treatment of micrometastatic disease
○ All patients can commence systemic therapy (more tolerable than post op)
- Disadvantages
○ Possibility of tumour progression precluding surgery
○ Surgery may be more difficult due to IO effects
○ Significant toxicity from chemo/IO may preclude surgery
○ Surgical toxicities

120
Q

What are the advantages and disadvantages for adjuvant chemo/immunotherapy post surgery for NSCLC?

A

Improve DFS significantly. No OS benefit data as immature
- Advantages
○ Fastest removal of cancer
○ Full pathology including from mediastinal LN dissection to determine adjuvant Rx
- Disadvantages
○ Surgical complications
○ Proportion of patients do not recover well enough to have adjuvant systemic therapy or are delayed in having this
○ R1/R2 resection if patients not selected appropriately or delay between staging scans and operation

121
Q

What is the evidence for neoadjuvant chemo/immunotherapy prior to surgery for NSCLC? and also for maintenance immunotherapy?

A

Neoadjuvant chemo/immunotherapy

- Checkmate 816 –doublet chemo + surgery vs chemo+immuno + surgery = improved PFS and pCR
	○ Nivolumab
	○ Pcr 22% vs 2% (pcr has 95% survival)
	○ 2 year OS benefit 82 vs 70%
	○ 4 year OS benefit 71% vs 58% (2024 update)
	○ Most benefit -nonsmoker, PDL1>50%, stage 3a, adenocarcinoma 
- Significant proportion of patients did not proceed to surgery (17-25% )and did not achieve an R0 resection (17% had R1).

Induction immuno -> surgery -> adjuvant immuno
○ Checkmate, keynote, Aegean
Improve 2 year event free survival by 10-20% 50-60% vs 40%

122
Q

In carcinogenesis, what are the typical characteristics of a driver mutation? (1 mark)

A

Driver mutations are mutations that cause uncontrolled growth and proliferation, often leading to cancer. EG: EGFR, ALK. Sustained growth through producing own growth signals, insensitivity to anti-growth signals , limitless replicative potential and evasion of apoptosis.

123
Q

For lung adenocarcinoma: (1.5 marks)

i. What is the clinical significance of detecting an Epidermal Growth Factor Receptor (EGFR) mutation?

A
  • EGFR triggers the RAS-RAF-MEK-ERK pathway for cell cycle growth. Acquired mutations can be driver mutations leading to unregulated growth and over expression
    • Significance:
      ○ Allows for the use of targeted therapy in the form of EGFR inhibitors and predicts patients response to treatment
      ○ Prognostic information: Exon 19 deletion and point mutation E21 more favourable prognosis due to sensitivity to tyrosine kinase inhibitors
      ○ Response to treatment: EGFR exon 20 insertions less responsive to tyrosine kinase inhibitors
      ○ PBS funding of medications for patients with EGFR mutation
      ○ Presence of driver mutation ?higher propensity to metastasise
124
Q

What are the potential mechanisms of acquired treatment resistance to 1st generation EGFR tyrosine kinase inhibitors?

A

1st generation reversibly bind to EGFR and inhibit the binding of ATP to stop cell proliferation and result in apoptosis. Mechanisms of resistance
* Point mutations that decrease the binding affinity of TKIs to EGFR
○ E.g ‘gatekeeper’ T790M mutation (60% of patients with disease progression on 1st generation of TKI found to have the T790M mutation) in EGFR exon 20 which hinders the binding of TKI to the ATP binding site of EGFR (3rd generation TKI irreversible block the T790M mutant)
* Activation of alternative signalling pathways e.g. HER 2 and MET amplification
* Epithelial mesenchymal transformation which confers resistance to chemotherapy and targeted therapy
* Histological transformation to small cell carcinoma

125
Q

ili. Besides EGFR, what are two other examples of driver mutations? Include their frequency of occurrence.

A

BRAF
- Activator of downstream MAPK signalling
- Present in <5% of lung adenocarcinomas
RAS
- KRAS is the most frequently mutated (20% of adenocarcinomas)
○ Mutually exclusive of EGFR or ALK re-arrangement
ROS1
- Rare mutation/fusion (<2% of all adenocarcinomas) –> multiple fusion partners
ALK
Uncommon mutation (incidence is 5%) –> EML4-ALK fusion

126
Q

What are the potential acute and late effects of radiation therapy on the structures of the heart? (1 mark)

A

Acute: Pericarditis
Late: Accelerated atherosclerosis/MI risk, arrythmia, valvulopathy, heart failure

127
Q

What are the aetiology/risk factors that increase the risk of cardiac effects after radiation therapy? (1 mark)

A
  • Total dose and dose per fraction
  • Mean heart dose
  • V20 Gy of heart (volume of heart radiated)
  • Use of radiosensitisers and/or systemic therapy especially anthracyclines
  • Anatomical location of heart and distance from field
  • Technique – higher integral dose with VMAT technique vs 3D CRT
  • Use of shielding
  • Use of heart dose minimising strategies such as DIBH, prone treatment
  • Comorbidities especially hypercholesterolaemia, coronary artery disease, smoking, obesity and physical inactivity and pre-existing heart disease
  • Age- younger patients higher life time risk
128
Q

Describe the pathogenesis of cardiac complications following radiation therapy. (2 marks)

A

Acute
* Radiation induced endothelial injury and oxidative damage from free radical formation
· Inflammation generates reactive oxygen specifies and proinflammatory cytokines e.g. TNF alpha/beta and IL 1/6
· Inflammation can damage endothelial vessels, increase thrombosis and cause inflammation of pericardium
Late
* Chronic changes after radiation large quantities of collage and foam cell deposition
○ Accelerated vessel stenosis, intimal fibrosis and atherosclerosisà leading to CAD (most common complication)
· Valve cusps fibrotic changes and thickening à valvular disease
· Fibrosis from endothelial cell injury and restricted blood flow to myocytesà Cardiomyopathy
· Fibrosis and pro inflammatory state can result in conduction abnormalities and arrhythmias (higher incidence of QT prolongation, AV blocks and bundle branch blocks)

129
Q

How do the radiologic findings in RILI differ in the acute and late settings? (2 marks)

A

Pneumonitis
* Ground glass opacities
* Airspace consolidation

Fibrosis
* Consolidation
* Volume loss
* Linear scarring
* Pleural thickening
* Reticulation and interlobular septal thickening

130
Q

Tumour Proportion Score (TPS) IHC is often performed on Non Small Cell Lung Cancers. (1.5)
i. What is the TPS?

A

a measure of the percentage of viable tumour cells showing partial or complete membrane staining for PD-L1 on IHC

Steps to Calculate the PD-L1 TPS:
- Bx prepared, stained using Ab bind to PDL1, highlights PDL1 expression
- Microscopic evaluation- identification and counts no. of tumour cells that show any positivity (partial or complete)
- TPS calced as % of viable tumour cells that show PDL1 positivity out of total no. of viable tumour cells

PD⁠-⁠L1 expression level in advanced NSCLC is determined by the TPS, which is reported as a percentage on a scale of 0% to 100%.
A minimum of 100 viable tumor cells in the PD⁠-⁠L1–stained slide is required for the specimen to be considered adequate for PD⁠-⁠L1 evaluation.

TPS < 1%: PD-L1 negative
TPS 1-49%:low PD-L1 expression
TPS ≥ 50%: high PD-L1 expression

131
Q

You have been asked to provide an opinion about the value of setting up a population screening program for lung cancer.
a. What advice would you give? Provide relevant evidence for population screening for lung cancer. (2 marks)

A

Screen high risk populations -20 pack year history (active or last 15 years), aged 50-80
Has risks to pt and health system -high false positives (27% of population vs 1-5% true positive), overdiagosis (uncertain, but 25% of tumours were indolent/slow)

Benefit
vs harm -false positive / toxicity from biopsy/ radiation exposure, overdiagnosis,
anxiety
Cost effectiveness
Participation and FU protocols

Annual Low dose CT is the choice modality for screening and for size followup
PET may be helpful when risks discordant

Evidence:
2 RCT
Screening vs no: 24% reduction in lung cancer mortality
CT vs CXR: -reduced lung cancer mortality 20% and 6.7% all cause mortality
Number screened per death prevented 303
$700,000 per death avoided
Trials have had poor compliance

132
Q

A 79-year-old man has an incidental finding of a 18 mm nodule in the right middle lobe. He has a 60-pack-year smoking history and known ischaemic heart disease and peripheral vascular disease.
i. What are the differential diagnoses for this nodule? (1 mark)

A
  • NSCLC (adenocarcinoma, squamous carcinoma) small cell carcinoma, neuroendcrine
    • Preinvasive neoplasm: Adenocarinoma in situ,
    • Metastasis, lymphoma
    • Benign:
    • Hamartoma
    • Granuloma
      ○ Inflammatory
      ○ Infectious -aspergillous, crypytococcus, coccidiomycosis, histoplasmosis, TB
    • Malignant characteristics: >10mm, irregular/spiculated, ground glass, noncalcified, doubling time 1-12months (benign can be quicker or slower, mostly solid,) (30% change in diameter = a doubling)
133
Q

Briefly discuss the factors that may influence potential late side effects when treating lung cancer.

A

PATIENT
* Age (younger patients greater life expectancy in developing late effects)
* Radiation insensitivity
* Use of radio sensitising medications e.g. methotrexate
* Previous radiation therapy
* Smoking
* Connective tissue disorders

TREATMENT
* Dose per fraction
* Total dose
* Volume irradiated
* Plan homogeneity and presence of hot spots in OARS
* Radiation type
* Radiosensitisers such as chemotherapy

ORGAN
* Pre-existing organ compromise i.e. poor PFTs
* Tissue irradiated e.g serial or parallel organ

134
Q

A. Describe the pathogenesis related to Late spinal cord effects

A

Radiation myelopathy or radionecrosis of the spinal cord resulting in myelopathy
Death of progenitors for oligodendrocytes (glial cells) → failure to replace oligodendrocytes → loss of myelin → altered nerve conduction
Death of endothelial cells → increased permeability → oedema, inflammation
Abnormal / uncoordinated endothelial proliferation, fibrosis, vessel lumen thickening→haemorrhage, thrombosis, hypoxia
Death of starved neuronal cells → necrosis → neuropathy.

135
Q

An asymptomatic 55-year-old man is diagnosed with non-small cell lung cancer (NSCLC). Staging investigations at diagnosis reveal a right peri-hilar 4 cm primary tumour with right hilar lymphadenopathy, and a solitary 13 mm left cerebellar metastasis on MRI (T2aN1M1b). Lung function tests are within normal limits.

a.
i. What are the management options for this patient? Which would you recommend and justify your answer. (6 marks)
A

This patient has denovo oligometastatic non-small cell lung cancer with a solitary intracranial metastasis (stage IV)
Management should be discussed at MDT
Options depend on factors including

Patient:
- Comorbidities, performance status – impacting overall survival and treatment options
- Fitness and contraindications to chemotherapy and radiotherapy 
- Disease symptoms 
- Patient preference 
Tumour:
- Histopathology + molecular profile/PDL1 status, Targetable mutation/driver mutation
- Location of brain met and impending complication eg. Bronchial obstruction
Treatment:
- Proposed systemic therapy and CNS activity
 
Assuming asymptomatic and not at risk of a short-term complication 
Treatment should be discussed at MDT 
However
Reasonable approach would be to commence systemic therapy up front as per management of stage IV NSCLC if proposed systemic therapy has reasonable CNS activity 
Reassessment/restaging after 3-6 months of systemic therapy and subsequent consolidation and local treatment of oligomets (brain SRS)
 
Another reasonable approach would to offer SRS to the brain met up front 
Followed by standard of care systemic therapy 
Restaging 
If no progression of disease then offer definitive local therapy to the primary 
Ie. 55Gy in 20 fractions to the primary and nodal disease 
 
This is supported by estro/Astro guidelines for oligometastatic disease 

Gomez (2019) demonstrate improved PFS  and MS with local consolidation therapy compared to maintenance systemic therapy alone  in patients with <=3 mets
 
Lastly, best supportive care is also an option in this patient with metastatic NSCLC 
 
In this young patient with a solitary intracranial metastasis, assuming a targetable mutation and appropriate systemic therapy I would offer up front systemic therapy with restaging and consolidation/treatment of the intracranial met
136
Q

A patient is planned for VMAT lung treatment for their SCLC to commence with the fisrt cycle of chemotherapy, but the lung DVH constraints cannot be met. What are the options for managing this situation and justify your answer.

A

Check that with 4D CT that the GTV is subtracted from both lungs
Discuss with planners if lung dose can be improved
Review the V20 and V5 constraints, prioritise the V20 over the V5

Consider other motion management, such as DIBH or gated treatment, to reduce the ITV volume.

If unable to meet constraints, consider if alternative treatment option such as IMRT/3D CRT could be utilise to reduce bilateral lung dose in the case of large encompassing fields

Wait for reduced tumour volume with chemo and commence with second cycle. -if issue is bulky disease.

137
Q

b. With regard to Stereotactic Ablative Body Radiation Therapy (SABR) for lung cancer, what are the general principles of SABR for lung tumours? (2 marks)

A

· SABR aims to deliver an ablative radiation dose (typically ≥8Gy/#) to tumours, over a few treatments
· Aims for high conformality with a high dose gradient and rapid dose fall off
· Heterogenous dose (125Gy -145Gy)
· Requires high accuracy via image guided radiotherapy, motion management and precision to achieve small margins
· Is typically used in lung treatment for patients with stage I to IIa cancer that are not suitable for/ have declined surgery.

138
Q

In general, discuss the advantages and disadvantages of radical surgery compared with SABR in early stage non-small cell lung cancer. (2 marks)

A
139
Q

Twelve months after treatment a patient presents with symptoms of late radiation oesophagitis. Discuss the possible presentation, investigation and management options for late oesophageal toxicity. (3 marks)

A

· Presentation
o Odynophagia
o Dysphagia to solids +/- liquids
o Regurgitation
o Haematemesis
o Food bolus
· Investigations
o Bloods: FBC, EUC, Albumin,
o CT chest + C
o Endoscopy
· Management
o Stricture
§ Oesophageal dilatation -multiple procedures with aim of increasing aperture of oesophagus from strictures
§ Oesophageal stent
§ NGT for nutrition vs PEG/RIG if unable to get past oesophageal obstruction
§ Referral to speech pathologist/ dietitian
o Bleeding
§ Argon laser/ cauterisation
§ haemospray
o Perforation
§ Surgery - oesophagectomy
o Ulceration
§ Hyperbaric oxygen
o Oesophageal dysmotility
§ Referral to speech pathologist/ dietitian
§ NGT for nutrition vs PEG/RIG if unable to get past oesophageal obstruction