Skin Flashcards

1
Q

What is the epidemiology of NMSC?

A

BCC

I: Common, BCC comprises the majority of all NMSCs (75%)
900000 new diagnoses annually

A: Incidence increased with age

G: Male predominance up to 50, in older years incidence is similar.

R: White/Caucasian. Compared with people born in Aus, immigrants from UK show lower incidence rate (strong positive association between childhood sun exposure and lifetime risk of BCC). Rare in dark-skinned population.

Geographical location: Higher in areas with highest UV exposure

cSCC

I: Common, 271 per 10 000

A : Increasing age

G : Male predominance
R: White/Caucasian. Higher in people born in Aus.

Geographical: Higher in QLD, lowest in TAS

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

What is the aetiology for NMSC?

A

1) UV-B exposure–> Sunlight, artificial radiation (Tanning beds 75% increase lifetime risk of melanoma)
a. Photo damaged skin
b. Actinic keratosis
c. Telangiectasia
2) Advancing age
3) Ethnicity (fair-skinned) and skin type
a. Fitzpatrick phenotype
b. Light skin that burns or prone to freckles
c. Red hair and blue eyes
4) Immunosuppression
a. Iatrogenic (transplant)
b. Infective (e.g. HIV)
c. NHL, CLL
5) Other benign skin lesions
a. Chronic ulceration/trauma (Marjolin’s ulcer)
b. Bowen’s
c. Actinic keratosis
6) Environmental exposures
a. Arsenic
b. Smoking (MA evdience)
7) Personal history of NMSC
8) Genetic syndromes
a. Gorlin’s (basal cell naevus syndrome)
-Rare autosomal dominant disorder characterised by the development of multiple early-onset BCCs (mutations in PTCH1 TSG–> key regulator of the hedgehog signalling pathway)
b. Xeroderma pigmentosum
9) Chronic inflammation
a. Draining sinus 10 years Marjolin ulcer

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

What is Gorlin syndrome?

A

-‘Basal cell nauevus syndrome’
-<1 % of skin cancers attributed to Gorlins (1/30 000 incidence)
-Mutations in PTCH1 gene. Sonic hedgehog is the ligand for patched-1 receptor. Patched-1 blocks cell growth and division until Sonic Hedgehog is activated.
-PTCH1 is TSG, mutations result in uncontrolled/unchecked replication
-Autosomal dominant
-Second hit to develop BCC

Macrocephaly, skeletal abnormalities, early onset BCC

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

How does UV light induce carcinogenesis

A

Multi-step carcinogenic process which is driven by UV-mediated DNA damage
- Melanocytes are derived from the neural crest cells and can be found in dermis/epidermis. Melanin important role in giving skin pigment + filtering UV radiation
- Fitzpatrick scale is s semi-quantitative scale to describe skin colour, melanin level and sensitivity to UV
- UV falls between light and gamma rays on the spectrum
- UV C shortest wavelength, highest energy. UV A longest wavelength, lowers energy

- Sunburn/Acute UV exposure: Inflammatory response and increase cytokines. Keratinocytes are damaged--> activation of DNA repair mechanisms (p53 activation) and apoptosis
- Protective/Sub-acute: Epidermal hyperkeratosis (epidermal growth factors to increase the thickness of the epidermis) _ adaptive melanisation 'tanning'
- Oxidative damage: UV damage results in ROS, the base-excision repair pathway is the main pathway to repair free-radical DNA damage.
- Nucleotide excision repair important pathway in repairing photodamage and bulky DNA lesions. In xeroderma pigmentosum (Rare UV hypersensitivity syndrome with homozygous defect in multiple common pathways that execute NER)

Tumour uv signature dna damage, c to t transitions on dna.

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

How does immunosuppression impact on skin cancer pathogenesis?

A
  • NK cells and macrophages are cytotoxic, part of the innate immune system
    • NK cells bind to cancer cells or virus infected cells and release cytotoxic granules that kill the cell
    • T cells are part of the adaptive immune system, cytotoxic T cells can destroy cancer cells
    • Prednisone bind to glucocorticoid receptors in cells, move to nucleus and block pro-inflammatory genes (e.g. NF kappa-B, A2) and inhibitors macrophages
    • Azathioprine (solid organ transplants) inhibits pure synthesis, which is necessary for NK cells, T cells and B cells. Azothioprine inhibits the immune systems ability to repair UV DNA damage (Transplant patients that take azathioprine RR x2 SCC and 1.3x melanoma, compared with transplant patients who don’t take azathioprine therapy)
      -TNF alpha inhibitors for rheumatological conditions, increased risk for non-melanoma skin cancers
    • In immunosuppressed patients:
      Skin cancers grow more quickly, more likely to recur, metastasize and cause death
      Oral B3 (nicotinamide) 500 mg BD reduces rate of skin cancer and acitinic keratosis by 23%
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What genes are involved in cSCC and BCC carcinogenesis?

A

SCC
- Large number of genes with a large mutational burden
○ P53 mutation is an early event
○ NOTCH mutations facilitate HRAS carcinogenesis
- Chronic inflammation can also contribute to this
○ Trauma (e.g. burn)
○ HPV infection

BCC
- Multiple genes are implicated
○ SHH family are implicated in 90% of cases –> PTCH1 receptor (Gorlin’s)
§ PTCH TSG that blocks activation, until SHH signalling
○ P53 is also frequently implicated

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

Describe the histopathological features of Keratoacanthoma and SCC.

A

Ketatocanthoma
- Rapidly developing neoplasm that clinically & histologically mimics well diff SCC, but can regress spontaenously
- Macro: sun-exposed areas, flesh colour nodules with central keratin filled plug
- Micro: keratin filled cater surrounded by proiliferating epithelial cells, extend upward as a lip around the plug and downward into dermis

Squamous Cell Carcinoma

- Macroscopic
	○ Pre-malignant in-situ lesions may appear as well-circumscribed erythematous and scaling plaques
	○ Malignant lesions may be nodular with variable ulceration
		§ Often raised nodular border with central ulceration
	○ Variable amount of hyperkeratosis 
- Microscopic
	○ Atypical keratinocytes, penetrating basement membrane → dermis,
		§ Large cellular size, nuclear atypia, mitotic figures, intercellular bridges
	○ Tumours with sheets of polygonal epithelial cells arranged in nests
		§ Penetrates into the dermis
	○ Evidence of keratinisation (keratin pearls, intercellular bridges)
	○ Differentiation varies, such that no keratinisation may be evident
		§ Well diff SCC→ atypical cells but with keratinisation present (keratin pearls)
		§ Poorly diff SCC → keratinocytes anaplastic with little no no differentiation or keratinisation. Focal necrosis 
		§ Nuclear pleomorphism and mitoses also contribute to grading
	○ May have PNI ~5-10% of excised SCCs but often asymptomatic 
	○ Variants
		§ Low-risk
			□ Keratoacanthoma (WD)
			□ Verrucous carcinoma (minimal atypia)
		§ High-risk
			□ Acantholytic (pseudoglandular, loss of intercellular adhesion)
			□ Spindle cell
			□ Desmoplastic
		i. Nb. Basaloid not skin,  mucosal h+n or anal
- Immunohistochemistry
	○ POS = AE1/AE3, CK5/6, p40, p63
	○ NEG = CAM 5.2, CK20, S100, MelanA, SOX10
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

List the Subtypes of BCC and the macro, microscopic features of each. List the IHC for BCC

A

Immunohistochemistry
- POS = AE1/AE3 (epithelial origin- antibody cocktail for cytokeratin’s), p63 (Transcription factor, master regular of epidermal keratinocyte proliferation), BerEP4 (Epithelial cell adhesion molecule vs SCC), CAM5.2 (variable, low molecular weight keratin)
NEG = CK20, CK7, S100, MelanA, SOX10, CEA

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

What are the prognostic factors for NMSC?

A

Patient Factors
- Age and performance status
- Fitness for surgery
- Immunosuppression: Transplant, HIV, CLL
- Immunosuppressive Medications: Prednisone, azathioprine, TNF alpha inhibtors

Tumour Factors
- Histological subtypes
○ High risk BCC = morphoeic, infiltrative
- TNM stage
○ Size of primary
- Tumour location (H-zone of face & ear)
- Grade
- DOI (>5mm)
- PNI (especially >0.1mm diameter nerve OR symptomatic)
- LVI
- Multiplicity or recurrence
Treatment Factors
- R0 resection

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

Describe specific features of the work up for NMSC.

A

Full history, including:
- Lesion
○ Duration and rate of growth
○ Pain/bleeding/discharge
○ Neuropathic symptoms (incl paraesthesia)
- PMHx including contra-indications to RT
○ Immunosuppression
○ Consider Gorlin’s syndrome –> no RT
- Medications including contraindications to RT
- Social
○ Sun exposure
○ Particularly to establish ability to attend for fractionated therapy
○ Performance status

Examination:
- Characterise the primary lesion
○ Location and size
○ Depth and fixation
○ Ulceration
○ Borders
- Palpate for lymphadenopathy
- CN examination:
○ Paresis of facial muscles (motor CN7). Potential to spread to origin in midbrain base of skull destruction, meningeal carcinomatosis, parenchymal brain invasion.
○ CN V - sensory to scalp, face + tongue via 3 divisions. Emerges lateral to pons trigem ganglion 3 divisions
○ cavernous sinus Superior orbital fissure.
○ cavernous sinus foramen rotundum pterygopalatine fossa inferior orbital fissure infraorbital forarnen.
○ V3 foramen ovale.
○ CN Vll - motor to facial muscles, taste ant 2/3 tongue. Emerges from pons→ Internal auditory meatus→ bony facial canal→ stylomastoid foramen→parotid

Investigations

Need a biopsy prior to any treatment

Staging
- For cT1 cN0
○ No staging indicated
- For all others (i.e. cT2+ or cN+)
○ Minimum CT NCAP
○ Consider PET-CT (only funded if H+N primary)

If PNI is clinically suspected, consider MR

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

Describe the TNM Staging for NMSC

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

What factors guide your management decisions for NMSC?

A

I. Patient Factors:
○ Age: young should be treated with surgery due to longer life expectancy for RT late effects ie 2nd Ca, cosmesis
○ Co-morbidities: ability to undergo anaesthesia, although most are performed under LA, anti-coagulation. Skin specific: psoriasis, scleroderma, atropic skin (age, steroids), extensive contiguous actinic keratoses
○ Prior treatment history: prior RT may rule out re-irradiation
○ SHx: ability to care for wounds, dressings, apply topical treatments or compliance with therapy, daily attendance
○ Patient preferences

II. Tumour Factors:
	○ Histologic Features: treat advanced lesions more aggressively & irregular borders with Moh’s to achieve clear margin
	○ Site: CI to RT are hair-bearing areas or prone to trauma and poor healing- below knee, pretibial- esp. if PVD, varicosities, oedema. Ideal- surgery + graft+ strict bed rest.
	○ CI to Sx include lip, nose, canthus for cosmesis
	○ Size and depth of invasion: bone and cartilage invasion may be better served by surgery
	○ Primary vs Recurrence: recurrence has worse prognosis, treat with surgery when possible & adjuvant RT
 
III. Treatment Factors:
	○ Cure & recurrence rates, SEs, cosmesis (younger pt, facial), cost, availability, physician expertise
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Describe the non-radiotherapy options for NMSC.

A

I. Surgical Excision Preferred in young pts, and generally, where possible
○ A: margin control, one step procedure, good cosmesis if adequate skin for flap
○ D: invasive, may require GA, 2-step if margin+, disfiguring in functional or cosmetic sensitive areas (lip, eyelid, nose)
○ Surgical Margins: well-circumscribed BCC: 3mm; small (T1) & well-diff SCC: 4mm; larger lesions or poorly diff: 1cm

II. Moh’s Micrographic Surgery: Under LA, resect tumour + small rim of adjacent tissue; 100% of the margin is examined microscopically + mapped. Any +ve margins re-excised till clear.
	○ A: achieves negative margin, high cure rate, removes minimum of normal tissue
	○ D: expensive, prolonged, requires expertise
	○ Indications: lesions with features that make clear margin difficult to obtain eg morpheic, recurrent, sensitive areas
 
III. Curettage: lesion excised by curetting with 3-4mm margin from apparent tumour under LA
	○ A: quick, single visit, good cosmesis, no sedation. Suitable for <1cm, superficial or nodular BCC, SCC-in-situ
	○ D: margin not controlled, recurrence rates high with large lesions (>5mm) in high risk sites
 
IV. Cryotherapy: compressed liquid nitrogen sprayed onto lesion until frozen then allowed to thaw & re-sprayed. Ice crystal formation and cell membrane disruption. Immediate erythema + oedema. Exudation (24h) + sloughing (7d). Healing 6w (face), 12w (trunk + extremities).
	○ A: quick, cheap, single visit, non-invasive, excellent cosmesis, good for multiple lesions. Useful for AK or small/superficial BCC
	○ D: not margin controlled, tendency to recur, some discomfort associated with procedure
 
V. Topical Chemotherapy
	○ Suitable for actinic keratoses, SCC in situ, superficial BCC. NOT for nodular BCC, deeply invasive or thick (b/c affects epidermis)
		§ 5-FU (Efudix): Pyrimidine analogue, interferes with DNA synthesis by inhibiting thymidylate synthetase
			□ applied twice daily 6wks depending on clinical response (increase if no inflamm r/n)
		§ Imiquimod (Aldara): immune response modifier, promotes apoptosis, circumvents anti-apoptotic pathways in Ca cells, activates langheran’s cells
			□ applied daily (BCC) or 2/week (AK) for 6-12 weeks.
	○ A: non-invasive, good cosmesis. Clearance rates 70-80%
	○ D: margin not controlled, may penetrate to depth required, SEs: burning, erythema, irritation, scabbing, (imiquimod – fatigue, flu-like, angioedema)
 
VI. Photodynamic Therapy: contraindicated in thick lesions
	○ Photosensitising porphyrin applied topically to lesion for hours, higher concentration & half-life within cancer cells
	○ Area is then exposed to light (630nm), porphyrin becomes unstable & reactive causing cell death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are the advantages, disadvantages and contraindications when comparing radiotherapy to surgery for NMSC?

A
  1. Radiotherapy vs. Surgery
    Choice of Modality
    • Results of RT comparable to surgery
    • Surgery is Rx of choice when lesions small + amenable to excision w/o sig morbidity.
    • RT reserved minority of BCCs + SCCs w Mx problems for surgery where:
      i) Surgery is not feasible
      - Patient unfit for surgery
      - Technically difficult to obtain margin clearance
      - Patient refuses
      - Anticoagulation problems
      ii) Surgery will result in worse toxicity
      - Functional/cosmetic morbidity unacceptable to pt + RT would have better cosmesis (e.g. loss of function lips/eyelids, rhinectomy, large tissue deficit, multiple lesions)
    • CI/relative CI to RT
      ○ CI: prior RT in field, RT hypersensitivity syndrome
      ○ Relative:
      § Young patients < 50-60yrs- 2nd malignancy
      § Poor blood supply, esp. pretibial, dorsum of hands (prone to trauma)- fully fractionate to aid healing
      § Sites- Hair bearing areas, overlying lacrimal gland, upper eyelid.
      § Advanced lesions invading bone/tendons. Def RT provide LC 50-75%, better LC if Surg+Adj RT
      § Cartilage involvement not a CI, but ↑ risk chondroradionecrosis. Avoided in large pinna lesions (or fully #ate for max cartilage repair)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are the indications for treating BCC with Radiotherapy?

A

Radiotherapy for BCC:
I. Primary: limited role as surgery preferred but can be used for reasons mentioned above
II. Residual:
○ After RT: clinical resolution can take up to 4mths, confirm clinical persistence with biopsy & treat with excision
○ After Sx: Local recurrence after margin+ 33%-> re-excision or RT -> LR 9%
§ Other features high risk for recurrence: PNI, morphoeic, basosquamous, invasion of skeletal m/cartilage/bone
○ Review at MDT and consider further excision or adjuvant RT
III. Recurrent BCC
○ After definitive RT: 90% occur within 5y, treat with excision, recurrence after salvage Sx is 15%
○ After other treatment modalities: treat as for primary, may require wider resection or more aggressive RT

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are the indications for treating cSCC with Radiotherapy?

A

Primary:
○ solar keratosis & SCC in situ-> more readily treated by Sx, topical 5-FU or cryo
○ keratocanthoma: difficult to distinguish from SCC, excise if in doubt, if not in doubt RT can hasten resolution
○ invasive indications as above
II. Adjuvant RT to primary after surgical excision:
○ Absolute indications:
§ pT4
§ margin+ or close (if <2mm re-excision preferred otherwise 60Gy should achieve equivalent LC)
§ PNI (symptomatic or named nerve)
§ recurrent disease (when irradiating nodes for local failure within 12mths & primary is within 5cm from field)
○ Relative indications: ≥2 of:
§ pT2/3
§ >5mm depth
§ margin<5mm
§ poorly diff
§ extratumoural PNI of unnamed nerve
§ extratumoural LVSI
§ Immunosuppression
Adjuvant skin radiotherapy
○ Need a higher dose in general to compensate for tumour bed hypoxia.

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

When would you consider elective lymph node treatment in cutaneous SCC?

A

Incompletely excised SCC are at risk of higher LR and regional mets, up to 25-45% depending on the site. Adequate radiation margin 10-15mm

	I. Elective Treatment of Regional Lymph Nodes in Primary Cutaneous SCC, N-
	○ <5% of pt’s with primary skin SCC will develop LN mets
	○ 5yr OS following nodal disease is markedly reduced to 40%
	○ Pt’s are curable but are at increased risk of developing distant mets
	○ Pt’s at high risk of LN mets due to features of primary should have elective nodal treatment (ENT)
	○ Elective nodal treatment can be with lymphadenectomy or RT 
		§ Dose: 50Gy (undisrupted tissue) or 54Gy (surgically disrupted) IF Non-IMRT technique. IF IMRT technique, then 56Gy to disrupted and 54Gy (undisrupted) 
 
	Risk factors for subclinical nodal mets: ENT may be of benefit when 1 or a combo of these are present:
		§ DOI/thickness: >4-5mm or Clark 4+ (invasion of reticular dermis or subcutaneous tissues)
		§ Size: tumours >2cm
		§ Recurrent: 25-45% risk of LN spread
		§ Site: ear & lip
		§ Histology: poorly-diff, desmoplastic (aggressive variant with high propensity for regional mets)
		§ PNI: increases regional & distant mets, ?LVI
		§ Immunosuppression: higher risk of recurrent & metastatic SCC

* Elective nodal irradiation based on location of the primary as per Porceddu below. Some centre opt to observe the nodal basin. In area where the first echelon node may be unclear, it may be more appropriate to observe the nodal regions rather than risk unnecessary toxicity from ENI.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

How would you manage high risk NMSC?

A

Treatment of High-Risk NMSC

1) Surgical resection (preferred)
	a. Aim margins > 2mm
	b. Most will require adjuvant RT 
		i. Absolute indications
			1) T4 disease
			2) N+ disease
			3) Positive margins
			4) Recurrent lesion (if no previous RT)
		ii. Usually if >/=2 high-risk features
			1) T2/T3
			2) DOI > 6mm
			3) Poorly differentiated
			4) Location (H-zone of face)
			5) PNI (>0.1mm)
			6) Multifocality or satellite lesions
			7) LVI
	
2) Definitive RT
	a. Not the preferred approach May be considered in elderly patients not suitable for resection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

How would you manage LN +ve cSCC?

A

1) Surgical resection of primary + nodal disease
a. Including parotidectomy or selective neck dissection if appropriate
i. ~25% regional failure without RT
a. Will require adjuvant RT to 60Gy to parotid bed
i. Virtually all cases (consider exclusion if single ipsilateral LN <3cm without ENE)
ii. If neck dissection negative no RT
iii. If no neck dissection then elective radiation (15% risk)
iv. No contralateral neck treatment.

Cervical Lymph Node Mets
* Surgery should be used if patient is fit and disease is resectable
* Post-op RT to 60Gy indicated IF >1LN+, ECE+, LN>3cm, SM+ or close <2mm, tumour spill, OR recurrence
* If there are LNs and RT is being used definitively, treat to 70Gy
* TROG 0501 POST trial: randomised to concurent carboplatin: no difference in 5 year recurrence or survival
* There is evidence in mucosal H&N SCC that adding chemo to RT in post-op RT is beneficial for DFS & OS
*

Axilla/Groin mets: Sx -> PORT IF >2LN+, ECE or LN>3cm

There is no evidence to support elective nodal irradiation

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

What are the systemic therapy options in cSCC?

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

What is the evidence to support definitive radiotherapy in NMSC?

A
  • 2021 meta-analysis (Krauss, 2021)
    ○ Multiple retrospective series
    ○ 5 yr LC = 85-90%
    • Office-based radiation therapy for cutaneous carcinoma, 2007 -Hernandez-machin et al
      ○ Retrospective analysis of 604 BCCs and 106 SCC
      ○ Definitive radiotherapy with KV Photons
      ○ 5 year local control 94% BCC and 93% for SCC
      ○ 15 year local control 85% BCC and 78% for SCC
      Predictors for local recurrence were size ≥10mm and tumour location in the nasolabial fold
    • Hypofractionated radiation treatment for BCC and SCC: A meta-analysis, 2018 -Zaorsky et al
      ○ Analysis of 21 studies assessing treatment with definitive RT, with primary endpoint of cosmesis
      ○ EBRT (9255 pts) and Brachytherapy (474 pts)
      ○ Median dose of 45Gy/11 fractions and median follow up of 3 years
      Hypofractionated schedules produce similar cosmesis compared to conventional fractionation, for the same BED (alpha/beta 3)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is the evidence for adjuvant radiotherapy for NMSC?

A
  • TROG 05.01 RCT (Porceddu, 2018)
    ○ Randomised high-risk cSCC to adjuvant RT +/- concurrent chemotherapy
    ○ 5-year LC = 85%
    ○ No benefit with chemotherapy (p=0.58)
    • LN + –> 2019 meta-analysis (Sahovaler, 2019)
      ○ 20 retrospective series + 1 RCT
      Adjuvant RT improves OS and CSS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Is there a role for concurrent chemotherapy when treating cSCC?

A
  • TROG 05.01 RCT (Porceddu, 2018)
    ○ Randomised high-risk cSCC to adjuvant RT +/- concurrent chemotherapy
    ○ 5-year LC = 85%
    ○ No benefit with chemotherapy (p=0.58)
    ○ R0/1→ PORT 60-66 Gy ± Concurrent carboplatin.
    ○ This is the only reported prospective trial with a pre-defined RT protocol and a RT QA program.
    ○ There is no role for CCRT in SqCC. There is excellent FFLR in both arms.
    Critique: Carboplatin was used, not cisplatin.
    - 321 SqCC pts. ECE, ~50% SM+ 5-10%. PNI not in inclusion criteria. Mostly HR nodal.
    ○ HR SqCC: T3-4 H&N primary excluding nose, lip or in-transit nodal mets or HR nodal.
    ○ HR nodal: ECE, intra-parotid nodal dz, 2+ cervical nodes or >3 cm.
    ○ RT: PORT 60-66 Gy ± carboplatin AUC 2 q1w x6c.
    - 2y FFLR ~88%, 5y FFLR ~85%.
    - 5y DFS ~70%, 5y OS ~78%.
    - Cetuximab may sometimes produce tumor regression in unresectable/metastatic SCC. May also consider H&N chemo such as CDDP ± 5-FU.
    - SqCC: Typically no role for systemic therapy, but consider cetuximab or CDDP ± 5-FU.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is the evidence for systemic management of BCC?

A

Unresectable BCC
Vismodegib and sonidegib - small molecule inhibitors of hedgehog pathway. Approved for non-surgery/RT candidates. ~30-65% response rate with median duration 7-10 mo.
* ERIVANCE [Sekulic JAAD ‘15, BMC ‘17]: Single arm phase II. Vismodegib.
○ 33 pts metastatic BCC with 30% response, 62 pts locally advanced BCC with 43% response.
§ Very low pCR 0-5%.
Response = decrease of at least 30% in externally visible or radiographic dimension of the lesion or complete resolution of ulceration.

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

What is the evidence for systemic treatment in metastatic cSCC?

A

Cemiplimab demonstrated to have 65% disease control and is approved in metastatic SCC
Cemiplimab is a highly protenant PD1 antibody
Approved on PBS for patients with metastatic or locally advanced cutaneous SCC in patients who are not candidates for curative surgery or RT

  • Metastatic SqCC [Migden NEJM ‘18, Lanc Onc ‘20]: Phase I/II. Cemiplimab 3 mg/kg q2w (PD-1).
    TBL: The PD-1 inhibitor cemiplimab is active in advanced cSCC.
    Methodology: Most metastatic SqCC’s are immunosuppressed and often hypermutated.
    ○ 78 pts. Locally advanced or metastatic cutaneous SqCC. MFU 9 mo.
    ○ ORR nearly 50% with over half of these having a duration of response exceeding 6 mo.
    ○ Complete response in 13%, partial response in 31%.
    ○ G3-4 in 44%. Serious treatment emergent AEs in 29% of patients.
    ○ Durable disease response in 63% of patients (at least stable disease for at least 3.5 mo).
    ○ MPFS and MS have not been reached at 9 mo of followup.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Describe your radiotherapy technique for definitive management of NMSC with EBRT.

A

Alternative:
Brachytherapy
-Flat surface
-Alternative to SXRT in superficial tumours
-Selection of appropriate sized Valencia applicator at simulation
-36Gy/12F prescribed to 5mm depth

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Describe your radiotherapy technique for adjuvant management of NMSC with VMAT.

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

Describe your radiotherapy technique for adjuvant management of NMSC with KV photons.

A

EviQ
* For a well-defined nodular BCC, a field margin of 0.5-1 cm beyond macroscopic disease or surgical site is usually adequate.
* For infiltrative/morphoeic BCC, a wider field margin of 1-1.5 cm is required to encompass surrounding subclinical spread.

There are numerous variations in defining an appropriate field margin, however for SCC, a field margin of 1-1.5 cm is required to encompass surrounding subclinical spread.

T1/T2 SCC- 1-1.5cm radially and 0.5cm deep margin
T3/T4 SCC- 1-1.5cm radially and 0.5-1cm deep margin
* IF RF for LR, then use 2-3cm margin 

* Small electron fields (<4 cm) are associated with certain disadvantages (isodose constriction) when compared with superficial and orthovoltage treatment. Bowing of isodoses due to lateral electron scatter means this modality is less useful with tumours close to the eyes.

Margins
* Recent article in R+O (12) - looked at extent of microscopic disease after resection and made recommendations for margins:
○ < 2cm size BCC/SCC- use 1cm margin
○ > 2cm size BCC/SCC- use 1.5cm margin.
○ Consider 2cm for poorly diff/large (>3cm) SCC.

* Margins:
	○ 0.5cm for small, well-defined BCC
	○ 1cm for larger, superficial or ill-defined BCC & well-diff SCC 1.5-2cm for poorly diff or large (>3cm) SCC

1) Consider wax-coated lead for:
	a. Internal eye-shields (SXRT only - consider if 6meV electrons)
	b. Intraoral mandibular shields (SXRT and electrons)
2) Consider custom lead cut-out (collimation on skin)
	a. Especially if on face near eyes Minimum size of 4x4cm
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What factors do you need to take into account with different radiation treatment modalities for treatment of skin cancers?

A

Modality: Factors to consider:
* Skin dose
* Dose at depth ie adequate coverage of tumour
* Dose to critical structures at depth: ie rate of dose drop-off
* Dose to critical structures lateral to PTV ie margins required for penumbra
* Dose to bone: higher in orthovoltage due to photoelectric effect

I. Superficial: 100-150kV
* suitable up to palpable tumour thickness of 5mm, max field diameter 10cm
* margin: 5mm for BCC, 10mm for large/superficial/ill-defined BCC, 15mm for SCC
* HVL (range 1-4mm Al) chosen to deliver 90% of dose to base of tumour

II. Orthovoltage: 150-300kV, can treat to max depth of 1.5cm
* Preferred when require narrow penumbra due to adjacent critical structure, or if irregular surface E inhomogeneity
* A: high skin dose, narrow penumbra, ease of collimation with lead cut-out on skin, ease of eye, alveolus and mucosal shielding. Shield is lead with wax to absorb backscatter
* D: high dose to underlying bone due to PEE, max depth 1.5cm, significant dose at depth
* HVL 0.5-1mm Cu to achieve 90% of surface dose at depth of tumour

III. Electrons: 6-15meV, max field size 25cm
* Preferred when require sparing of deeper structures due to rapid dose fall-off, more deeply infiltrating lesions (>1.5cm)
* Require wider margin due to penumbra (extra 1cm on above margins) and electron cut-out & bolus to ensure skin dose. Field size must be >4cm, need flat surface (obliquity < 30 degrees). Inhomogeneities (bone, air spaces) can be problematic although usually distal clinically. Eye protection more difficult than orthovoltage
-MeV: Dmax = E/4, D90% = E/3.2, D80% = E/2.8, D50% = E/2

IV. Megavoltage Photons: use for bony infiltration or where difficult to use KV/e- due to curvature of pt, penumbra issues. IMRT w 1cm bolus

Additional notes:
* Use bolus to bring skin dose up to 100%, ie 0.5-1.5cm depending on energy
* TLD: use when adjacent to critical structure (eg lens/cornea) or uncertainty re/ dosimetry (uneven surface, tissue inhomogeneity, junction)
* Do not hypofractionate >3Gy/#, esp for large lesions
* Use external eyeshield where required and internal eye shield when treating eyelid
* Pack the ear and nostril with vaz-gauze to flatten ala nasi & to provide shielding for underlying structures and from backscatter eg internal mouth shield wax coated to protect gingiva and minimize mucositis of lip
Keratin horns should be de-bulked

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

What are factors to consider when treating skin lesions on the nose and hands?

A
  • Nose/ear- MV photons if bone/cartilage involved (generally larger tumour, KV ↑bone absorption). Wax coated lead shield in nasal cavity protects nose for DXRT/electrons. Tip of nose- consider E or parallel-opposed MV photons if unacceptable standoff.
    • Dorsum hand- fully # as little subcut tissue (↑risk necrosis + ↓funcn). SXRT→ ↑ absorption in bone. Avoid exit dose on palmar aspect + no ↑absorption in bone.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

How does PNI affect treatment of cSCC?

A

Perineural spread
* PNI is assoc with higher LR and worse OS, increase morbidities and mortality. Rarely salvageable if BOS recurrence
* Multiple retrospective studies showed Adj RT improve LC and decrease lethal BOS recurrence
* Incidental PNI has better outcome than symptomatic/gross PNI; 5yr LC 85 vs 55%
○ Microscopic PNI= histological finding of PNI > 0.1mm
○ Gross PNI= clinical or radiological evidence of PNI
* Dose:
○ IF Micro PNI, resected with neg margin -> 54Gy to the CN at risk (in column 2)
○ IF Gross PNI, resected with neg margin -> 60Gy to the CN at risk and inter nerve connections (in column 2 and 5)
IF Gross PNI, resected with pos margin on the nerve-> 66Gy in tumour bed and remaining course to 60Gy

**IF Micro PNI- cover the CN by anatomic proximity to BOS + relevant branches
**
IF Gross PNI- cover the CN by anatomic proximity AND the elective CN via internerve connections
** Ensure to over 5mm CTV from the nerve
**
For Malar Cheek, in the case of incidental PNI, they do not routinely cover the course of the nerve to the foramen rotundum

* PNI of CN should be considered for widefield RT to encompass potential neural pathway back to brainstem
* Gross evidence of PNI on MRI should receive BoS resection + PORT to 50-60Gy Definitive RT may be attempted but is morbid & difficult to achieve doses of 70Gy close to brainstem
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What are the side effects of skin radiotherapy?

A

a) Acute Effects
* Erythema (10-20Gy)
* Dry desquamation >20Gy
* Patchy then confluent Moist desquamation – epi cell death, exposing underlying dermis.

b) Late Effects
* Atrophy/fibrosis, telangiectasia
* Hypo/hyperpigmentation (larger #size→ ↑hypopigmentation)
* Skin appendage loss: epilation (>25Gy permanent), ↓sweating, ↓wax prodn (ear)
* Necrosis – <5% dt inability of residual epi to remain intact→ ulceration, commonly painful. Small may heal, but larger lesions →plastics repair w skin graft.
* ORN underlying bone, chondronecrosis (nose, ears)
Second malignancy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is the expected local control after skin radiotherapy, with different T stages?

A

T1 LC = 95%
T2 LC = 85%
T3 LC = 60%
T4 LC = 50%
N+ OS = 40%

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

Describe the epidemiology for Melanoma.

A

Incidence (Australian statistics)
- 16900 cases in 2021
- 3rd most common malignancy

Median age 59
Slight male pre-dominance (1.5:1)
Geographical and ethnic disparity (increased in white skinned populations)

Immunosuppression
Moles>50

Presentation:
* Localised 84%
* Nodal 9%
Mets 4%

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

What are the risk factors for melanoma?

A

1) UV exposure
a. In addition to pale skin
2) Other benign skin lesions
a. Giant congenital naevus
b. Dysplastic naevi
3) Immunosuppression
4) Familial melanoma
a. CDKN2A gene (familial atypical multiple mole melanoma syndrome - FAMMMS)
b. CDK4 gene
5) Xeroderma pigmentosum
Impaired DNA repair

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

Describe the pathogenesis for melanoma.

A

Majority of cases are related to UV-induced carcinogenesis
- Can occur de-novo or within an existing naevus

Main oncogenic signalling pathways:
1) Mitogen activated protein kinase (MAPK) pathway (RAS / RAF / MEK / ERK)
2) PI3K / AKT / mTOR/ WNT / beta catenin signalling pathway

4 step model of carcinogenesis:
- Mitogenic driver mutation (BRAF)
- Escapes senescence (CDKN2A mutation)
- Overcoming apoptosis (p53)
- Immortalisation (TERT promoter mutation)

Growth typically occurs in two main phases
- Radial growth
Deep/vertical growth (potential for metastasis)

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

Describe the histopathological features of the following melanoma subtypes:
-Superficial spreading
-Lentigo maligna

A

Classic melanoma markers
- S100
- SOX10 (neural crest)
- MelanA (may be negative in desmoplastic)
- HMB45
Negative
- CKs
- P16 (loss of p16)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Describe the histopathological features of the following melanoma subtypes:
-Nodular melanoma
-Acral lentiginous
-Desmoplastic

A

Classic melanoma markers
- S100
- SOX10 (neural crest)
- MelanA (may be negative in desmoplastic)
- HMB45
Negative
- CKs
- P16 (loss of p16)

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

In relation to melanoma, describe the importance of BRAF, NRAS and cKIT.

A

BRAF
- 50% of melanomas with BRAF mutation, BRAF is a proto-oncogene
- Most common mutation in the BRAF gene is V600E (90%)
- Non-BRAF V600 mutations account 3-5% of melanomas
- BRAF critical role in the MAPK (RAS-RAF-MEK-ERK) pathway. Majority of BRAF mutations result in constitutive phosphorylation of MEK, independent of upstream activation. Results in promotion of cell growth and evasion of apoptosis
- Prior to targeted therapy, BRAF was a poor prognostic biomarker, patients tended to be younger, lack sun damage, trunk associated lesions and have worse OS compared with wild-type BRAF.
- BRAF V600 mutation is a predictive biomarker to targeted therapy
- daBRAFenib (BRAF inhibitor) and tRAMetinib (MEK inhibitor) in unresectable or metastatic melanoma
- Resistance: NRAS mutations and activation of the PI3K pathway
-BRAF also involved in -colorectal, NSCLC (adeno) and papillary thyroid cancers

NRAS
- RAS important role signal transduction pathway
- NRAS mutation in 20% of melanomas
- Poorer prognosis: Shorter time to progression, higher risk of CNS involvement
- Higher prevelance on arms and legs

cKIT
- KIT is a proto-oncogene encoding got the transmembrane receptor tyrosine kinase
- 3% of melanomas (Higher proportion in acral, mucosal and chronically sun damaged skin)
- C-kit activates downstream signalling pathways: MAPK and Pi3K pathways
- Often mutations in KIT almost never occur if BRAF or NRAS mutations present
- GIST, leukaemia, melanoma and lung cancer
- Tyrosine kinase inhibtors (TKIs) e.g. imatinib

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What are the 2 ways to describe depth in melanoma?

A

Breslow depth = simple depth in mm

Clark level = discrete levels
- 1 = epidermis
- 2 = papillary dermis
- 3 = dermis
- 4 = reticular dermis
- 5 = subcut fat

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

What are the prognostic factors important in melanoma?

A

Patient Factors
- Age and performance status
- Gender (females are favourable)
- LDH high- worse prognosis

Tumour Factors
- Clinicopathological Type
○ Nodular has worst prognosis
- Pathological Features
○ DOI (Breslow) - higher risk of nodal+
○ Mitoses
○ Ulceration
○ Neurotropism
○ LVI
○ TILs & tumour regression (host immune reactivity)
○ Molecular mutations: BRAF, NRAS, cKIT
○ Microstatelitosis
- TNM stage
○ Nodal and distant metastases
○ Nodes: number, burden and size, ENE
- Location
○ Extremity is favourable
○ H+N or trunk is unfavourable
- In-transit metastases

Treatment Factors
- Margin of resection
- Sentinel node negative

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What’s important in history and clinical examination for melanoma?

A
  • Can occur anywhere on the skin surface, but predominate in sun-exposed areas
  • Anatomic distribution:
    • Caucasian male: Most common on face and back
    • Caucasian female: Most common on face, shoulders and lower leg (knee to ankle)
    • Lentigo maligna melanoma - sun-damaged areas, e.g. H&N
    • Acatal lentiginous melanoma - palms, soles, nailbeds of darker skinned people
  • 10% of patients present with LNs and no evidence of primary
    • → better prognosis
    • May have arisen from naevus cells in LN and when resected → nil residual disease
  • Clinical features
    • A: Asymmetry
    • B: Border irregularities
    • C: Colour variegation
    • D: Diameter >5mm
    • E: Enlargement
    • F: Clinical features (itch / pain)
    • G: Grown-ups / adults - new lesions in later life
    • Changes in size, colour, shape, +/- inflammation, bleeding/crusting, sensory change
  • Hx, exam, + dermoscopy in experienced hands
  • Dermatoscopy (x10 magnification - 90% sensitive) evaluation
    ABCD (asymmetric lesion, irregular border, irregular colour and differential structure - invading other structures)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

How would you investigate melanoma?

A

Always biopsy the primary lesion.

Histopathology should have:
* Histological subtype
* Depth of invasion/ Breslow thickness
* PNI/neurotrophism
* LVI/ angiolymphatic invasion
* Molecular status (BRAF, PD-L1)
* Microsatellitosis (present or absent)
* Ulceration status (present or absent)
* Dermal mitotic rate (#/mm^2)
* Margins: deep and peripheral
pure desmoplasia if present

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

How would you stage melanoma?

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Describe the management of Stage I+II Melanoma.

A

Management of primary:
* Surgical excision with adequate margins
○ Stage 2b consider adjuvant immunotherapy or observation (improved recurrence free survival)
Microscopic margins:
LM: for >3mm = 2-3% local recurrence, if <3mm =30%

	* Definitive radiotherapy (60Gy/30F or 50Gy/20F) - considered non-curative 
	○ Indications:
		§ Unresectable
		§ Medically inoperable
		§ Mucosal site
		§ Lentigo maligna or lentigo maligna melanoma (50Gy/25F IF >5cm or 45Gy/10F if <5cm field size)
* Adjuvant immunotherapy for locally advanced

Adjuvant RT to primary IF: (48Gy/20F ***** or 30Gy/5 twice weekly)
* Desmoplastic histology, esp. neurotropic
* Inadequate surgical margins, where further surgery difficult (e.g. Face, H&N area)
* Poor pathological response
* PNI/Extensive LVSI 90% relapse with in-transit / satellite lesions
* Recurrence
* Unresectable positive/close margin
* DOI >4mm, ulceration, satellosis
Evidence: No RCT data, but Ph2 TROG and MDACC showed improved LC with Adj PORT to High risk melanoma (>1 node, ENE, recurrent, tumour spill, Clark 4). For desmoplastic, 2 retrospective study (MIA and NCDB) showed improve OS and LC with Adj PORT

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Describe the management for Stage III Melanoma.

A

Stage III (Clinically node positive) (PBS funded for Stage IIIB/C/D)
* Biopsy of node (FNA/excision), BRAF test.
- If node positive on biopsy and resectable then
○ -> Neoadjuvant immunotherapy (Pembrolizumab, Ipi/Nivo) improves EFS
§ Only neoadjuvant pembro on PBS in 2024
○ Can also proceed without neoadjuvant treatment (then would require adjuvant immunotherapy).
- WLE + nodal dissection
○ Overall 30% local recurrence, 50% metastasis
- Lymphoedema risk
○ Axilla 10%
○ Groin 10-15%
○ Pelvic +10%

- Adjuvant systemic therapy 
	○ (if had neoadjuvant doublet immuno and complete response or major response then no adjuvant therapy)
	○ ONE YEAR DURATION then observation
	○ PBS funded
	○ Drug choice
		® If BRAF mutant --> Dabrafenib / trametinib COMBI-AD: Improved OS/RFS cf. placebo. Use only if contraindications to immunotherapy and BRAFV600E mutant.
		§ If BRAF wildtype --> single agent nivolumab, pembrolizumab –improved 3 year RFS 63 vs 50%
			□ Nivolumab x12 months CheckMate 238, improved 2yr RFS 63 vs. 50% cf. ipilimumab
			□ Pembrolizumab x 12 months KEYNOTE-054. Improved 3yr RFS 64 vs. 44% cf. placebo
			□ OS data immature but more durable response than BRAF
		
		Note: Note: There are no head to head comparison between BRAF/MEK therapy and anti-PDL1. However, BRAF/MEK inhib are more toxic than anti-PD1 therapy  (G3 tox 40% vs 14%) and has higher discontinuation rate (26 vs <10%).

- Satellite lesions(<2cm from primary)/ intransit mets (>2cm from primary) (stage IIIb/c/d):
	○ Resectable
		§ Neoadjuvant systemic therapy + Surgical resection + adjuvant therapy as per stage IIIb/c disease
		§ Surgical resection + adjuvant therapy as per stage IIIb/c disease
	○ Unresectable
		§ Systemic therapy - preferred
			□ Immunotherapy
			□ BRAF/MEK inhibitors
		□ Topical DPCP (Diphencyprone) 40% overall response rate
			□ Pt sensitised (made allergic) to DPCP by applying to arm (contact dermatitis)
			□ Then applied to tumour field weekly for up to 3months. Cream strength titrated to g2 skin reaction.
		§ Regional chemotherapy  Should be followed by systemic immunotherapy 
			□ Isolated limb perfusion  ORR 56%, CR 22%; or
			□ Isolated limb infusion ORR 75%, CR 33%
				* RPA and Peter Mac units only 
				* Intensive salvage  treatment
				* 30min chemo in theatre. 1 week in hospital with swollen painful leg. Risk oedema, dead muscle, compartment syndrome. 
		§ Intralesional treatment (T-VEC) reserved for patients with injectable lesions in the setting of distant metastatic disease, ORR 17%
			□ Only IL2 and bleomycin available in Australia
		§ Consider palliative RT if above not feasible (used infrequently)
	
- Consider adjuvant RT to nodal basin after dissection 
	○ Regional control benefit 21% vs 36%. No survival benefit
	○ 48Gy/20F (TROG trial)
	○ Consider when risk factors
		§ Extracapsular extension
		§ Large nodal burden
			□ Parotid = 1+ LN
			□ Cervical = 2+ LN or >3cm size
			□ Axillary = 2+ LN or >4cm size
			□ Inguinofemoral = 3+ LN or >4cm size
	○ Note this is questionable in the context of adjuvant immunotherapy/BRAF inhibition –use if patient has medical contraindications

- If not well enough for a LN dissection
	○ Ideally, aim for selective resection of macroscopic nodes
	○ Completion RT to entire LN basin
		§ 30Gy/6F twice per week
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What is the management of Oligometastatic Melanoma?

A
  • Oligometastatic disease (one or a very limited number of sites)
    ○ Resectable
    § Complete surgical resection of all disease
    § Adjuvant immunotherapy with induction ipi/nivo → maintenance nivolumab
    ○ Unresectable
    § Initial immunotherapy
    § Re-evaluate to determine suitability for resection of sites of residual disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What is the management of Stage IV melanoma?

A

Stage IV
- Oligometastatic disease (one or a very limited number of sites)
○ Resectable
§ Complete surgical resection of all disease
§ Adjuvant immunotherapy with induction ipi/nivo → maintenance nivolumab
○ Unresectable
§ Initial immunotherapy
□ Palliative surg esp heart, gut (bleeding), brain surg/SRS if these mets are symptomatic and interfering with sysyemic
§ Re-evaluate to determine suitability for resection of sites of residual disease

- All other patients
	○ Immunotherapy (preferred in all cases, even if BRAF mutants)
		§ Combination immunotherapy 
			□ Induction ipi/nivo (3-weekly x 4 doses)→ maintenance nivolumab (3-weekly) CheckMate 067 exploratory analysis: superior response rate and PFS (5yr PFS 36 vs. 29% SS) cf. nivolumab (5yr OS 52 vs. 44%, NS)
				* ORR 60%
				* Subgroup –particular benefit effective for BRAF mutants.
			□ Nivo + Relatlimab (LAG3 inhib)
				* Better than nivo alone,  less toxic than ipi/nivo
				* G3 toxicity 20% (vs ipi/novo 60%)
		§ PD-L1 inhibitor monotherapy: less toxicity 
			□ Nivolumab CheckMate 066 improved OS cf. dacarbazine (mOS 38 vs, 11 months, 3yr OS 51 vs. 22%); or 
				* ORR 40%
			□ Pembrolizumab for up to 2 years Keynote-006: improved OS (mOS 33 vs. 16 months; 5yr OS 39 vs. 31%) and mPFS (8.4 vs. 3.4 months) cf. ipilimumab. G≥3 toxicity 17%
		Ipilimumab (anti-CTLA-4 inhibitor) monotherapy →  inferior to PD-L1: 
			
	○ Targeted therapy:
		* BRAFV600E mutation:  
			□ Sequencing trial ipi/Nivo before BRAF had better  overall survival
		§ 
			□ Dabrafenib + trametinib COMBI-D: cf. dabrafenib alone improved mPFS 11 vs. 9 months; improved mOS 25 vs. 19 months; improved objective response rate (68 vs. 55%)
				* Quicker onset of response
				* Not as durable as immunotherapy but still 20% long term response
			□ Encorafenib + Binimetinib
			□ Venmurafenib + cobimetinib
		§ KIT mutation: Imatinib

- Systemic therapy
	○ Chemo has <10-15% response rate
		§  Chemotherapy- minimal role now due to small response rate and median duration of effect 4-6mo. No OS benefit
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What is the management of mucosal head and neck melanoma?

A
  • Commonly nasal cavity, paranasal sinus, oral (hard palate, upper alveolus). Oral cavity often node positive 25%
    * AJCC 8th edition staging - begins at stage III to reflect poor prognosis
    * Resectable:
    ▪ WLE - LR rates high (30-80%) despite aggressive surgery
    ▪ Neck dissection for oral cavity (not sinonasal)
    ▪ + adjuvant RT as high rates of local recurrence.
    ® Improved local control with RT in retrospective case series
    ◊ No impact on OS
    * Unresectable:
    ▪ Definitive RT
    ® Tumour control rates 60-85% in case series
    ® Optimal dose/fractionation not established
    * Prognosis
    ○ Poor
    ○ 5yr OS 12-30%
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What is the management of ocular melanoma?

A

○ ○ Uveal: ~95% of ocular melanomas
* Most common primary intraocular malignancy in adults, but ocular mets are more likely. GNAQ mutation –growth. BAP 1 mutation -malignant
* Can arise from any part of the uveal tract (iris, ciliary body, choroid)
□ Plaque brachytherapy with 125Iodine
® Dose rate 0.7-1Gy/h (Tx 4-7 days)
® Dose: 85Gy to the tumour apex or 5mm from internal surface of the sclera with 2mm margin around the tumour
® Not suitable if Thickness >5mm, blind painful eye, no light perception
□ SBRT
§ Dose/fractionation: Typically 50Gy/5#, PTV= GTV+1mm, have the pt focus on a red LED dot on the mounted camera, which tracks the eye movement and is gated to the treatment machine.. If the eye movement certain amount, the camera software stop the beam
□ Enucleation
§ Retrospective studies did not show a survival advantage over RT
§ QOL outcomes appear similar to RT
§ Generally reserved for patients who would not be expected to have a favourable outcome with RT
□ Large tumours
□ Marked extrascleral extension
□ Neovascular glaucoma
□ Extensive retinal detachment
□ Poor visual potential
□ Select patients who prefer enucleation
* Prognosis
* LC 95% with local treatment; BUT
* DM in 50%

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

What is the evidence for SLNBx in Melanoma?

A
  • MSLT-I
    ○ 2001 patients randomised to SLNB or observation
    ○ SLNB is prognostic
    § Intermediate (1.2-3.5mm) and thick melanomas had worse survival if SNB positive
    § SLN management improves DFS in all melanoma patients. DMFS and CSS are also improved in intermediate thickness.
    § In thin melanoma, too few events to make conclusions
    ○ Did not show significant survival benefit with ∴ not considered therapeutic
    • MSLT-II
      ○ 1934 patients with positive SLNB
      § Randomised to LND or observation (US surveillance)
      ○ Outcomes
      § No change to melanoma-specific survival with upfront dissection
      § LND improves regional control and provides prognostic information, but has no benefit in CSS
      § Higher rate of lymphoedema if proceeding with dissection
    • DeCOG-SLT (2006-2014) [Leiter JCO ‘19]: SLNB(+)→ ± Completion LND.
      TBL: Long-term results of the German DeCOG-SLT trial confirm that there is no improvement in OS, DMFS, or RFS with a completion lymph node dissection for melanoma.
      There is an approximate 25% incidence of positive NSLNs after (+) SLNB. There is an improvement in regional recurrence.
      ○ 483 pts. Cutaneous melanoma of the trunk and extremities
      ○ MVA demonstrated tumor thickness > 2mm most impactful on DMFS, OS and RFS. 1 mm SLN mattered less.
      ○ G3+ toxicity 13% (mostly lymphedema).

In a Nutshell:
- Everyone with intermediate (>1mm) or thick melanoma (>4mm) should get SLNbx as MLTS I showed that SLNBx provided important prognostication and help decide on nodal mx. Improves DMFS and DSS esp in those with intermediate risk
- Once the SLNbx is Pos, The SOC is LND. However if SLNB <1mm then the option is for either immediate LND or obs with USS surveillance as per MSLTII trial and DeCOG. As immediate LND only provide LC benefit but no CSS, and increase risk of lymphaedema significantly

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

What is the evidence for Neoadjuvant immunotherapy for melanoma?

A
  • SWOG1801 trial
    ○ Randomized 313 patients with resectable stage III–IV melanoma to 3 doses of neoadjuvant pembrolizumab 200 mg every 3 weeks followed by adjuvant pembrolizumab versus adjuvant pembrolizumab; both groups received surgical excision.
    ○ The neoadjuvant arm was associated with improved event-free survival (EFS) at 2 years (72% vs. 49%, P < .01).
    ○ Additional studies with 1–3 doses of anti-PD-1–based regimens given prior to surgery (either monotherapy or in combination with CTLA-4 or LAG-3 blockade) have also demonstrated high pathologic response rates and toxicities largely consistent with their use in the metastatic setting
    • NADINA trial
      ○ Stage III
      ○ Neoadjuvant IPI and NIVO prior to surgery improved EFS compared to adjuvant NIVO
      If PCR –no adjuvant immuno
53
Q

What is the evidence for adjuvant RT in melanoma?

A
  • TROG 96.06 [Burmeister RTO ‘06]: Phase II. Post-op 48/20.
    ○ 234 pts. HR: >1 LN, ENE, locally recurrent after surgery, tumor spill at time of surgery. MFU over 3y.
    ○ Regional in-field first site of relapse in 7%. 5y OS 36%. 5y PFS 27%. 5y LRC 91%.
    ○ G3 lymphedema 19% groin, 9% axilla.
    ○ 5y RR 33→ 18%, more lymphedema, no OS benefit.
    • MDACC H&N [Ang IJROBP ‘94]: Phase II. WLE→ PORT 30/5 twice weekly.
      ○ Five fraction adjuvant RT led to favorable LRC and low toxicity.
      ○ The fairly low 5y OS is likely attributable to distant disease.
      § 174 pts. HR melanoma of H&N: size ≥ 1 mm or Clark’s level IV, palpable nodes, nodal relapse. Some got LND.
      § 5y LRC 88%, 5y OS 47%.
54
Q

What is the evidence for Adjuvant RT for desmoplastic melanoma?

A
  • Retrospective study at Melanoma Institute of Australia (2017)
    ○ Retrospective analysis of 671 patients with desmoplastic neurotropic melanoma
    ○ Adjuvant RT was associated with reduction in local recurrence (HR 0.48)
    § Only if surgical margins <8mm
    • Retrospective NCDB study (2019)
      ○ 2390 patients with early stage desmoplastic melanoma
      Adjuvant RT after WLE was associated with improved OS
55
Q

In melanoma management, what is the evidence for adjuvant RT to the nodal basin?

A

Adjuvant RT for cN+ lymph nodes has small improvements in regional relapse but no confirmed benefit for DM or OS. RT is likely appropriate for patients with high risk of regional recurrence, such as ECE. It may be reasonable to advocate for RT if ECE or high nodal burden, but otherwise consider observing (especially if LE) due to lymphedema risk and high risk of distant failure. Adjuvant RT after LND is NCCN category 2B.
In the modern era, immunotherapy, BRAFi, and observation are reasonable treatment options as well.
In the context of (+)SLNB, there appears to be no DMFS, CSS or OS advantage with completion LND

- Randomised TROG 02.01 trial (2015)
	○ 250 patients with HR features were randomised to adjuvant RT vs observation to nodal basin following LN dissection
		§ 48Gy/20F
		§ High-risk features = extra-nodal extension and nodal burden
		§ Eligibility: 1 parotid, 2 neck, 2 axilla, 3 groin, ≥ 3 cm neck, ≥ 4 cm axillary/inguinal ("1, 22, 33, 4").
	○ Improved local relapse in the adjuvant RT group (HR 0.52)
		§ 3y LRC 68→ 82% (HR 0.52). 6y relapse 36→ 21% without improved RFS or OS. 5y OS ~35% (not powered).
		§ No adjuvant systemic therapy used
	○ No difference in OS
56
Q

What would be your adjuvant radiotherapy technique in melanoma management for the primary and nodes?

A
57
Q

What would be your definitive radiotherapy technique in upfront melanoma management?

A

Definitive setting (primary)

  • MDT discussion
    • Establish plan for any systemic therapy
  • Consider OPG + dental review (if H+N site)

-Wire CTV –> lesion + 3-4cm
1cm bolus

-If H+N, needs thermoplastic mask

-FDG-PET fusion

-66Gy/33F prescribed to the PTV (as per ICRU 83)

-VMAT technique
-10 days per fortnight

-GTV: gross primary as per CT/PET
-CTVp = primary + 3-4cm (wired)

-PET = CTV + 5mm

-Daily CBCT

58
Q

What are the indications for adjuvant RT to the primary site for melanoma?

A
  • Primary site indications:
    ○ High risk desmoplastic melanoma and positive margins where further resection is not feasible.
    § In general, at least 1 cm SM (T1), for thicker tumors 2 cm SM (≥ T3).
    If desmoplastic and SM-, H&N location, depth > 4 mm, Clark level V benefits from RT [Strom Cancer ‘14].
59
Q

What are the indications for adjuvant RT to the nodal basin for melanoma?

A
  • Nodal basin indications:
    ○ Parotid ≥ 1 LN, Neck/Axilla ≥ 2 LN, Inguinal ≥ 3 LN; Neck ≥ 3 cm, Axilla/inguinal ≥ 4 cm [TROG 02.01].
    ○ ENE, ≥ 4 LN, recurrent nodal dz, cervical LN [Ballo criteria].
    ○ Groin/pelvic nodes have a higher threshold for ENI due to morbidity of lymphedema.
    § BMI < 25 kg/m2: 1+ of following: ECE, >3-4 involved lymph nodes, recurrent dz, node >3-4 cm.
    § BMI > 25 kg/m2: ECE and 1+ of the following: >3-4 involved lymph nodes, node >3-4 cm.
    Positive SLN when completion LND is not planned.
60
Q

What are the options for radiotherapy fractionation in definitive, adjuvant, regional and palliative management of melanoma?

A
61
Q

What is the 5year OS for melanoma by stage?

A
62
Q

What is the incidence and aetiology for Merkle cell carcinoma?

A

Incidence (Australian statistics)
- Rare tumour (1 per 100,000 population)
- Incidence is increasing

Geographic incidence follows that of NMSC
- Highest = Australia & USA
Caucasian predominance
More common in males (2.5:1)

Aetiology

1) UV radiation exposure
	a. UV-B exposure is the biggest contributor in Australia
2) Merkel Cell Polyomavirus (MCV) exposure
	a. In low sun exposure areas, up to 80% of cases have this virus identified
3) Advancing age and caucasian ethnicity
4) Immunosuppression
	a. Transplants
	b. HIV
5) Other genetic mutations
	a. TP53 RB1
63
Q

What is the pathogenesis for Merkle cell carcinoma?

A

Pathogenesis

Traditionally, Merkel Cell Carcinoma was thought to arise from Merkel Cells
- Neurotactile cells of the dermis (mechanoreceptors)

This is controversial, with a differing view that they arise from a totipotent stem cell

Two main pathogenic pathways:
1) Merkel Cell Polyomavirus
a. Causally linked to MCC development (80% of all cases)
b. Good evidence that integration of virus into genome precedes clonal expansion
i. Large tumour (LT) antigen and small tumour (ST) antigen
2) Ultraviolet-B radiation
a. Particularly relevant for the MCV-negative group
i. This group is much higher in Australia than elsewhere
b. Typically occurs on sun-exposed skin (e.g. H+N)
Co-exists with other skin malignancies

64
Q

What is the histopathology for Merkle cell carcinoma?

A
  • Macroscopic
    ○ Rapidly growing, purple-red fleshy exophytic lesions
    ○ Variable degree of ulceration (often epidermis is preserved)
    • Microscopic
      ○ Expansile nodular or diffusely infiltrative tumour
      ○ Small blue round cell tumour with
      § High N:C ratio
      § Indistinct nucleoli
      § Scant cytoplasm
      § Salt and pepper chromatin
      ○ Majority are pure NE morphology
      ○ Frequent mitoses and apoptotic bodies
    • Immunohistochemistry
      ○ POS = synaptophysin, chromogranin, CD56, panCK, CAM5.2, AE1/AE3, CK20
      ○ NEG = CK7, TTF1(lung or other small cell cancer) CDX2, vimentin
    • Cytogenetics
      If MCV negative –> p53 and Rb1 mutations common
65
Q

What are the prognostic factors for Merkle cell carcinoma?

A

Patient Factors
- Age and performance status
- Immunosuppression

Tumour Factors
- TNM staging
○ Size
○ Depth of invasion
○ Nodal and distant metastases
- Head and neck location
- LVI
- TILs
- Merkel Cell Polyomavirus (absence of virus is negative prognosis)

Treatment Factors
-Positive Margins
Delayed RT

66
Q

What would be the expected presentation for a Merkle cell carcinoma?

A

Clinical features
* aggressive p[primary cutaneous neuroendocrine (small cell) carcinoma originating from the basal epidermis
* 50% occur in H+N region, then extremities
* Painless non-ulcerative dermal-based purplish lesion that may progress rapidly. MCC can spread through dermal lymphatics and cause satellite lesions.
* May present with node with n&obvious cutaneous primary (20%)
* Regional lymph nodes involved in 1/3 at presentation (predictor of poor outcome)
* Distant metastasis (<10% at presentation): liver, lung, bone, and brain.

Natural history
- High local recurrence rate, frequent nodal involvement and high risk of metastasis
Nodal status strongest predictor of distant spread and OS

67
Q

How would you work up a Merkle cell carcinoma?

A

History
- Lesion
○ Duration and rate of growth (typically rapid)
○ Pain/bleeding/ulceration (typically painless)
○ Location (most common on H+N)
- PMHx including contraindications to RT
○ immunosuppression
- Medications including contraindications to RT
- Social History
○ Sun exposure
○ Ability to attend for fractionated radiotherapy

Examination
- Lesion
○ Site and location
○ Depth and fixation
○ Colour (typically red-purple)
○ Ulceration (typically intradermal without overt surface erosion)
- Complete skin exam for synchronous lesions
- Palpate for lymphadenopathy

Investigations

Need histological confirmation
- Often excisional biopsy (especially if small)
- LN biopsy if clinically suspicious nodes

Staging
- At minimum, needs CT NCAP
- Ideally perform a FDG-PET (funded for rare cancer)

Sentinel LN Biopsy
- If clinically node negative, should be performed at time of biopsy/excision
Possibly not necessary if primary > 1-2cm, as adjuvant RT is indicated regardless

68
Q

What is the staging for Merkle cell carcinoma?

A
69
Q

What is the management for clinical lymph node negative and low risk Merkle cell carcinoma?

A

Low-risk (if primary is <1cm, non H+N site, and no immunosuppression)

* Surgical excision of the primary + SLNB
	○ Aim for 2cm surgical margin regardless of location due to high risk of dermal lymphatic spread 
		§ Do no re-resect if margins inadequate as XRT required w minimal delay. 
		§ If SLNB is microscopically positive, nodal RT without completion dissection is generally preferred.

* If clear wide margins,  and no risk factors (<1cm,  not H+N site, no immunosuppression, no LVI) - then no adjuvant therapy required, can be observe
	○ U. of Washington (2006-2019) [Goff '20, Bierma ARO '24]: Retro. Surgery→ Stage I Non H&N vs. H&N ± PORT.
	PORT is effective in decreasing LR for low risk H&N tumors, but may not be necessary for low risk non-H&N tumors
		§ 5y LR ± PORT of 10→ 0%, with 6 LRs observed in the surgery alone group. 
		§ 5y LR for H&N tumors ± PORT of 21→ 0%. 
		§ There were no LRs in non-HN MCC managed with surgery alone. 

	○ High risk regional relapse with local excision alone- 33% (5-10mm primary), 50% (>10mm primary). Veness et al.
	○ Retrospective study- risk of nodal met 4% vs 24% if >1cm size
	○ Another retrospective data showed false neg as high as 20% for SLN bx
	○ 30% of micromet in N0 pts- T1 20-30%, T2 >50%
70
Q

What is the management for clinical lymph node negative and high risk Merkle cell carcinoma?

A

Higher-risk (larger primary, H+N location, immunosuppressed)

* Surgical excision of the primary + SLNB
	○ Aim for 2cm surgical margin regardless of location
	○ Needs adjuvant radiotherapy- RT alone
		i. If no residual disease and SLNB negative
			1) 50Gy/25F to the primary and 1st echelon nodal region
		ii. If residual disease and SLNB negative
			1) Do not re-excise --> proceed directly to adjuvant RT
			2) Microscopic margin = 56Gy/28F to primary and 50Gy/25F to 1st echelon nodes
			3) Macroscopic margin = 60Gy/30F to primary and 50Gy/25F to 1st echelon nodes
		iii. If SLNB positive
			1) Consider role of nodal dissection
			2) Otherwise, RT to primary as appropriate and nodal irradiation
				a) Involved nodal level = 50-60Gy/25-30F
				b) Next echelon of nodes = 50Gy/25F
	○ NOTE: 
		§ SLNbx is unreliable, hence even when SLNbx is neg, we still need to treat the elective prophylactically. SLNbx is done to ensure the node is not pos, if it is then would need to dose escalate the node. 
		§ Currently there is emerging data looking into the role of Adj Nivolumab for completely resected Stage 1-2 and Stage 3-4 MCC- ADMEC-O
			□ Interim data: Adj Nivo-> absolute risk reduction of 9% at 1yr DFS and 10% 2yr DFS
			□ OS and events rate are inmature. 
	
* Alternative is definitive radiotherapy alone (if medically/technically unresectable)
	a. 60-66Gy/30-33F to the primary and 50Gy/25F to 1st echelon nodes

SIB technique
[VE Dose - 55Gy/20F to primary and 45Gy/20F to inguinal node prophylaxis]
[Rev 55/50Gy in 25F with Carboplatin
55Gy to SM+, scar, clips, tumour bed
50Gy to at risk area]

71
Q

What is the management for clinical lymph node positive Merkle cell carcinoma?

A

1) Surgical excision is favoured
a. Excision of the primary
b. Dissection of nodal region

2) Will need adjuvant radiotherapy
	a. Treat primary and nodal regions as appropriate (doses above)
		i. Adjuvant RT to nodal regions only indicated if multiple LN+ or ECE
		ii. Avoid nodal RT if a single LN+ is identified

3) Alternative is definitive RT (if medically/technically inoperable)
	a. 60-66Gy/30-33F to the primary and involved nodes 50Gy/25F to next echelon nodes
72
Q

What is the surgical technique with Merkle cell carcinoma?

A

DO NOT DELAY RADIOTHERAPY
- Must be simple and achieve primary closure
- Aim for rapid healing

Do not re-resect positive margins –> proceed to RT

Aim to commence Adj RT 4 week post op

73
Q

Is there a role for chemotherapy in Merkle cell carcinoma?

A

Adjuvant- controversial, but doesnt seem to have any benefit UNLESS male, T>3cm and pos margin.
- Rationale
§ ≥10mm primary
§ LN+ (62%)
§ residual dx post-sx
§ surgical recurrence
§ recurrence outside XRT field.
○ High metastatic potential of MCC
○ Responsive to CT
○ Improve OS
- Disadvantages
○ ↑XRT toxicity- esp. skin + CT toxicity- esp. neutropaenia
○ Short-lived responses 3-4mo, but clinically meaningful
Retrospective data showed incr OS if Pos margin, T >3cm, or male

74
Q

What would you do with recurrent or metastatic Merkle cell carcinoma?

A

RECURRENT Dx
- Main aim of treatment to prevent morbidity + mortality associated with recurrence.
- Median time to locoregional recurrence 8m. Most within 2yrs.
- Systemic recurrence- median survival 3-6m.
○ 20-30% develop LRR, poor prognosis but potentially curable. Restage fully if considering Rx with radical intent
- Can consider multimodal salvage Tx-> Salvage surgery, RT, CT (has shown best outcomes)

METASTATIC Dx
- Stage IV dx- median survival 9mo
- Clinical trial
- Chemo (CDDP or carbo ± etoposide, topotecan, cyclophosphamide, doxorubicin, vincristine)
○ ORR 50-80% but Short response time 3-4mo (not durable)
- Immunotherapy monotherapy (pembro [PD-1 inhib] RR 50%, avelumab [PD-L1 inhib] RR30%, with 75% response >1yr )
○ Seem to be the new standard - Based on small Studies- JAVELIN MERKEL 200 and KEYNOTE 17
○ larger scale data needed
Palliative RT- sx control

75
Q

What is the evidence for Adjuvant radiotherapy in Merkle cell carcinoma?

A

Adjuvant radiotherapy
- Retrospective NCDB study (Bhatia, 2016)
○ Stage I-II –> Improved OS (HR 0.71)
Stage III –> no improvement in OS

76
Q

What is the evidence for Concurrent chemotherapy in Merkle cell carcinoma?

A

Concurrent chemoradiotherapy
- RCT –> TROG 96:07
○ Only 53 patients
○ Historical comparison did not show any improvement in OS or LRC
- Retrospective NCDB study (Bhatia, 2016)
Neither adjuvant chemoRT or chemo alone improved OS

77
Q

What is the evidence for Definitive radiotherapy in Merkle cell carcinoma?

A

Definitive radiotherapy
- Small retrospective series only (Australian)
○ LC is 75-85%
High risk of distant relapse

78
Q

What would be your adjuvant radiotherapy technique for Merkle cell carcinoma?

A

Adjuvant setting

Pre-simulation

MDT discussion
Staging
Consider OPG + dental review if mandible is in-field

Simulation

Wire scar
Wire CTV
- 3-4cm margin from surgical scar
1cm bolus
If H+N, needs thermoplastic mask

Fusion

FDG-PET fusion

Dose Prescription

Primary
- Clear margins –> 50Gy/25F
- Microscopic margins –> 56Gy/28F
- Macroscopic margins –> 60Gy/30F

Nodes
- Elective –> 50Gy/25F
- ECE –> 56Gy/28F

VMAT technique
10 days per fortnight

Target Volumes

CTVp = Scar + 3-5cm margin (wired)
** 3cm for H&N MCC and 5cm for Non-H&N MCC
- minimum of 1.5cm deep down to fascia at depth
CTVn = 1st echelon nodal region
- If <20cm from primary, include primary in field
- * if excised node,:then excised node tumour+1cm

PTV = CTV + 5mm

Target Verification

Daily CBCT

OARs

As per H+N

Note that Merkel Cell is prone to in-transit cutaneous metastases

With respect to treatment of nodal volumes
- If separation is <20cm, aim to include primary and nodal volumes within the one field (i.e. treat in-transit skin)
If separation is greater, treat primary and nodes separately

79
Q

What would be your definitive radiotherapy technique for Merkle cell carcinoma?

A

Definitive setting

MDT discussion
Staging
Consider OPG + dental review if mandible is in-field

Wire CTV
- 3-4cm margin from primary lesion
1cm bolus
If H+N, needs thermoplastic mask

FDG-PET fusion

Primary
- Definitive = 60-66Gy/30-33F

Nodes
- Elective –> 50Gy/25F
- Definitive (cN+) = 60Gy/30F

VMAT technique
10 days per fortnight

CTVp = Primary + 3-5cm margin (wired)
** 3cm for H&N MCC and 5cm for Non-H&N MCC
- minimum of 1.5cm deep down to fascia at depth
defCTVn = LN + 7mm
elecCTVn = 1st echelon nodal region
- If <20cm from primary, include in field

PTV = CTV + 5mm

Daily CBCT

As per H+N

80
Q

What are the expected Local control outcomes for Merkle cell carcinoma after surgery alone and also after adjuvant RT?

A

<1cm , Ro= 5yr LR 0%
<1cm R1 30% to <5% with Adj RT
>1cm R0 50%

81
Q

What is the 5yr OS for Merkle cell carcinoma by stage?

A

OUTCOMES:
- Median Survival: Stage I/II – 40mo; Stage III – 13mo; Stage IV – 9mo
- Local Control with addition of RT
○ N0 50% -> 90%, 3yr OS 70-80%
○ N+ ve 50% -> 70-80%, 3yr OS 50%
○ RT alone in field control 75%, LRC 40%, 5yr OS 30-40%
- Possibly increase in DFS, OS- conflicting data
- Distant relapse in 50-60%
3yr OS 30% if DM

82
Q

What is the incidence and risk factors for Kaposi Sarcoma?

A

Rare vascular neoplasm caused by HHV8 in all cases
- 1-2 per 100,000 persons
Male predominance (3:1)
Geography and ethnicity dependent on subtype

Risk Factors
1) Immunosuppression
2) Previous malignancy (e.g. haematological)
3) Hygiene
a. Wearing shoes and bathing have both been associated
4) Smoking (reduces risk)
5) Lymphoedema
Small association is noted (?chronic vascular insufficiency –> inflammation)

83
Q

What are the four main subtypes of Kaposi sarcoma?

A

Four key subtypes
- Classical
○ Older men of Mediterranean, Eastern European or North African origin
○ Predominantly impacts lower limbs (lower legs, toes, soles)
○ Patches to nodules/plaques
○ Visceral invovlement is uncommon
○ Indolent course, rarely aggressive and disseminated

- Endemic (African)
	○ 2 types: lymphadenopathic (L) and cutaneous ©
	○ sub-Saharan Africa
	○ Bimodal: 
		§ children <10: LN/visceral invovlement 
		§ Men 20-50yrs: nodular, infiltrative skin lesions 
	○ Not related to HIV
	○ Predominantly impacts lower limbs
	○ More aggressive in children
	○ In adults: Initially indolent, but then aggressive course once nodal metastases established
	
- Iatrogenic
	○ Associated with transplantation (immunosuppression)
	○ Occurs when immunosuppressed pt contract HHV8 or receives an organ infected with HHV8
	○ Variable presentation
	○ Lower limbs ± disseminated, mucosa, visceral relatively common 
	○ Aggressive course
	○ Improves with removal of immunosuppression 
	
- AIDS-associated
	○ Associated with HIV1 (men or women), worse in africa 
	○ AIDS defining illness 
	○ Skin- H&N, trunk, genitals and LL; oral mucosa in 15-30%
	○ GIT , LN, lungs more common with poor HIV control 
	○ Most aggressive subtype
	○ May regress with effective HAART HIV treatment 
	○ Rapidly progressive from macules → papules, nodules, plaques 
	affect the genital, face, oral mucosa, LL
84
Q

What is the pathogenesis for Kaposi sarcoma?

A

All cases are caused by the HHV8 virus
- Detection of the virus assists with diagnosis

Key pathogenic steps
- Viral protein is transcribed and induces VEGF production –> endothelial growth
- Viral proteins disrupt proliferation
○ Synthesis of Cyclin D
Inhibition of p53

85
Q

What are the prognostic factors for Kaposi sarcoma?

A

Patient Factors
- Age and performance status
- Immunosuppression

Tumour Factors
- Clinical subtype
○ AIDS associated is most aggressive
○ Classical is most indolent
- Stage
○ Tumour/nodular stage often heralds nodal/visceral dissemination

Treatment Factors

86
Q

Describe the macroscopic and microscopic features of Kaposi sarcoma.

A

Note that the histopathology is independent of the clinical subtype
- They all look the same under the microscope

There are three stages of the disease
- Patch stage –> flat macules
- Plaque –> lesions thicken
- Nodular

Macroscopic
- Enlarging red/violaceous skin lesions
- Cut surface shows haemorrhagic appearance
Microscopic
- Patch stage
○ Dilated vascular channels
○ Dense lymphocytic infiltrate
- Plaque stage
○ More extensive with compressed and slit-like vascular channels
○ Infiltrative proliferation of spindled endothelial cells
- Nodule stage
○ Discrete nodules of uniform spindle cells
○ Intervening slit-spaces between spindle cells
- Mitoses are common, but pleomorphism is rare
Immunohistochemistry
- POS = HHV8, D2-40 (podoplanin), ERG, CD34 (endothelial progenitor)
- NEG = desmin, SMA, S100, melanA, CKs

Classic DDx is angiosarcoma (which will be HHV8 negative)

87
Q

How would you work up a patient with Kaposi sarcoma?

A

History
- Lesions
○ Duration and rate of growth
○ Pain/bleeding/ulceration
- Evidence of other lesions
○ Oral bleeding –> mucosal lesions
○ Abdominal pain –> visceral disease
○ Malaena –> GIT lesions
- PMHx including contraindications to RT
○ Immunosuppression including HIV/AIDS
- Medications including contraindications to RT
- Social History
○ Smoking status
○ Social supports and ability to attend RT

Examination
- Characterise visible lesions
○ Macule/plaque/nodule stage
○ Extent
- Examine entire skin as well as oral mucosa
- Lymphadenopathy

Investigations

Need histopathological confirmation

CT staging rarely indicated
- If symptoms indicated, image as appropriate
- If HIV-related, consider CT CAP due to aggressive disease course

Bloods
- FBC
○ Hb –> if bleeding
○ WCC + differential CD4+ count –> immunosuppression
- HIV serology (if aggressive disease course)
○ Viral load if positive

FOBT/Endoscopy only if evidence of GI bleeding

88
Q

What is the staging for Kaposi sarcoma?

A

No widespread staging system

Generally use a qualitative approach
- Type of lesion (macule stage, nodular stage, etc.)
- Location and extent

89
Q

Describe the management for Classical Kaposi sarcoma.

A

KS should be considered an “incurable” disease, in that lesions will recur given enough time
Management therefore is on the basis of symptomatology
- Cosmesis counts as symptomatic

1) Asymptomatic
	a. Observation alone
	
2) Symptomatic but limited extent
	a. Surgical excision
		i. Only consider if solitary lesion
		ii. Useful if no histopathological diagnosis
	b. Radiotherapy 
		i. Best if all disease is encompassable in field
		ii. Can use for palliation if single lesion is symptomatic
	c. Intralesional therapy (e.g. vincristine or bleomycin)
		i. This can be painful and lead to scarring
	d. Topical therapy (Retinoic acid, Imiquimod)
		i. Limited evidence on efficacy

3) Extensive disease
	a. Systemic therapy
		i. Pegylated liposomal doxorubicin --> Caelyx (71% significant response)
		ii. Paclitaxel
		§ Doxorubicin
		§ Vincristine+/- bleomycin
		§ gemcitabine vinoralbine
90
Q

Describe the management for HIV-AIDS related Kaposi sarcoma.

A

1) Refer to ID physician & introduction of HAART (thus improving immunosuppression)
a. Very commonly leads to improvement in disease

2) Systemic therapy (pegylated liposomal doxorubicin, Taxol or mTOR inhibitor )-> Doxo RR 50%, median duration 25m (time to response 4months)
	a. Indicated if:
		i. Widespread skin involvement
		ii. Disease is unresponsive to local therapy
		iii. Progression on HAART
		iv. Symptomatic visceral involvement

3) Local therapies (if symptomatic)
	a. As per classical Kaposi

* Immunotherapy-
	○ AIDS related KS→IFN-alpha shows response in 20-40%
 
* Experimental approaches-
	○ Molecular Targeted therapies are being investigated
	○ TKIs
	○ VEGFi/ antiangiogenic drugs eg pazopanib Checkpoint inhib immunotherapy
91
Q

Describe the management for Iatrogenic Kaposi sarcoma.

A

If possible, reversal of immunosuppression is best approach

92
Q

In general, what is the role of RT in management of Kaposi sarcoma?

A
  • Less role of XRT in advent of antiviral Rx for AIDS assoc KS
    • In Classic KS, local treatments can offer prolonged DFS as slow growing and indolent
    • Sensitive to RT, but can develop further lesions outside of RT field.
    • Use for Palliation –> pain, bleeding, oedema
    • Durable palliation of Symptoms + response >90%, CR in 70%, but recurrence in adjacent areas common
      Cosmetic result -> 50-60% remain purple
93
Q

Describe radiotherapy management of Kaposi sarcoma solitary lesions.

A
  • Solitary lesions:
    ○ Dose/F: 20Gy/10 acceptable in most scenarios, 30Gy/15 common, 8Gy/1 also good palliative dose. One study used 6MeV with 5mm bolus and 2-3cm margin. 30Gy/10# and 20Gy/5# also doses used for SKIN.
    ○ Range of margins used -> I would treat with 1.5-2cm margin, either 6MeV electrons with 1cm bolus or SXR (100kV)
    ○ 1 study showed higher response rate and duration with 40Gy vs 8Gy.
    § If single lesion consider 40Gy/20#
    If CKS and failed other therapies consider higher doses.
94
Q

Describe radiotherapy management of Kaposi sarcoma mucosal lesions.

A

○ AIDS assoc. KS has been reported to have more severe mucositis so are given lower doses, generally 15-20Gy/10# to avoid severe mucositis, also give antifungal treatment during RT
○ CKS can give higher doses of 30-40Gy.
○ [LVH- for mucosa KS, limited dose to 20Gy]
KV or Electrons for solitary superficial lesions, MV for lesions covering the majority of a limb (leave strip of skin so as not to ringbark the dermal lymphatics if possible.)

95
Q

Describe radiotherapy management of Kaposi sarcoma limb lesions.

A
  • Ext LL lesions:
    Techniques include AP/PA with bolus wrapped around leg in cast, or put leg in waterbath, commonly 20-30Gy, other technique for a limb is 6MeV electrons with 6 fields (60 degrees gantry rotations and in sup and prone positions) or extended SSD
96
Q

Describe radiotherapy management of Kaposi sarcoma when there are widespread lesions.

A

○ Consider TSE if disease above knee or hemibody skin electrons (disease below knee only)- treat to 15cm above lesion
○ TSE dose: 24Gy weekly 4Gy/F
1 study showed TSE of hemibody skin electrons with 100% RR median duration 4 yrs, more effective then “chasing” lesions as they occur

97
Q

What is the evidence for the management of Kaposi sarcoma with radiotherapy?

A

Very little randomised or comparative evidence
- Majority of data is retrospective series
- Majority of data is for the AIDS-related variant (data from the 1990s)

Summary of the retrospective data for RT

- Response
	○ CR rates of 80-90%
- Durability
	○ High rates of in-field control
	○ Considerable risk of distant relapse
- Dose
	○ Heterogeneity in outcome data
	○ Likely fractionated treatment is superior to single fraction EQD2 doses above 20Gy appear superior
98
Q

What would be your radiotherapy technique when definitively treating Kaposi sarcoma?

A

Higher-Dose –> Local Control

Patients

Limited disease extent in which the intent of treatment is to achieve durable control

Pre-simulation

Exclude HIV (serology)
Optomise HIV management
Rule out visceral involvement
Pathology review exclude angiosarcoma
MDT

Simulation

Wire field
- If VMAT, wire CTV
- If electrons, wire PTV
Consider 1cm bolus
Bite block
TPM

Dose prescription

Definitive
Electrons
- 30Gy/15F prescribed to 90% isodose
VMAT
- 30Gy/15F prescribed to PTV (ICRU 83)
MC:
* Consider lower doses 20Gy/10Fx

Palliative
-Electrons
- 20Gy/5F prescribed to 90% isodose
VMAT
- 20Gy/5F prescribed to PTV (ICRU 83)

9- 10 days per fortnight

Volumes

CTV
- 10mm
PTV
- VMAT = 5mm
- Electrons = 10mm

Target Verification

If VMAT, daily CBCT

OARs

Ensure a skin strip is preserved

99
Q

What would be your radiotherapy technique when definitively treating Kaposi sarcoma involving widespread limb surface?

A

Water Bath

Extensive disease involving multiple surfaces of a limb

  • Exclude HIV (serology)
  • Need significant RT and physicist multidisplinary involvement

Water bath
- Styrofoam at base to modulate height
- Thermoplastic immobilisation sleeve attached to bottom (foot to fit like a sandal)

Mark isocentre on the bath walls

Simulate water bath to plan RT (without patient)

MV photons (opposed laterals)
- 30Gy/15F prescribed to the midpoint

10 days per fortnight

N/A –> Treat entire limb

kV films daily

100
Q

What is the prognosis for Kaposi sarcoma?

A

Classical Kaposi Sarcoma

- Very rarely life threatening
- >90% chance of durable in-field control

AIDS-related Kaposi Sarcoma

- 5 year OS (with HAART)
	a. T0 disease = 92%  T1 disease = 83%
101
Q

A 63 year old male presents with extensive symptomatic Kaposi’s lesions on his right lower leg extending from just below the knee anteriorly, involving the calf posteriorly as well as both dorsal and plantar surfaces of his foot.

What probability of treatment response and potential toxicities would you discuss with the patient? (2 marks)

A

Response to RT high, upwards of 85-90%
Complete response 70-80%
High risk of recurrence/further lesions
Common to have residual pigemntation

Acute toxicities:
- Skin reaction, oedema, pain/tenderness
- Oedema ± compartment syndrome
Late:
- Dyspigmentation, telengiectasia, skin and soft tissue fibrosis
- Lymphoedema
- Joint dysfunction
- Secondary malignancy
- Risk of chronic non-healing ulcer (higher if pre-exisiting ulceration, poor vasculatuire, DM etc)

102
Q

What is the rationale and possible mechanism of action for the concurrent use of radiation therapy with immunotherapy?
In your answer discuss the impact of radiation timing, radiation techniques and dose fractionation. (6 marks)

A

Rationale and Mechanism of Action:
- RT+ IO potentially results in synergistic effects
- RT can enhance anti-tumour response and the efficacy of immunotherapy by:
○ Increasing Antigen Presentation: RT releases tumour antigens, improving immune recognition.
§ Also upregulates MHC and costimulatory molecules on tumour cells and antigen-presenting cells, enhancing tumour visibility and recognition by the immune system.
○ Modulating Tumour Microenvironment: RT alters the tumour environment,↑ expression of adhesion molecules and chemokines; promoting immune cell infiltration and activation.
○ Inducing Immunogenic Cell Death: RT causes cell death releasing damage-associated molecular patterns (DAMPs) that further stimulate immune responses
○ RT induced inflammation and cytokine release can enhance the efficacy of immune checkpoint inhibitors. Eg. can increase PD-L1 expression on tumour cells, making them more susceptible to PD-1/PD-L1 blockade.
○ RT induced tumour antigen release (locally) → systemic immune activation → regression of distant, untreated disease; enahnxced by immunotherapy

Impact of Radiation Timing, Techniques, and Dose Fractionation:
- Radiation Timing:
○ Sequential vs. Concurrent Administration:
§ Sequential administration of ICI after RT has shown more consistent benefits in clinical trials, as seen in the PACIFIC and CheckMate-577 trials.
§ Concurrent administration has shown mixed results, potentially due to differences in tumour burden and immune modulation.
○ Neoadjuvant RT:
§ Administering RT before ICI can increase antigen release and enhance immune priming, potentially improving ICI efficacy.
- Radiation Techniques:
○ Target Selection: Irradiation of the primary tumour and involved lymph nodes can reduce tumour burden and enhance local immune responses. However, irradiating tumour-draining lymph nodes may suppress systemic immune responses by reducing T-cell priming.
○ IMRT allows precise targeting of tumours while sparing normal tissues, which can reduce immune suppression caused by radiation-induced damage to healthy tissues
○ SBRT delivers high doses of radiation in fewer fractions, which can enhance local control and stimulate systemic immune responses.
- Dose Fractionation:
○ Hypofractionation: larger doses per fraction can enhance the immunogenic effects of radiation by inducing stronger tumour cell stress and more stronger immune responses.
○ Conventional fractionation maybe less immunogenic but can still modulate the tumour microenvironment and support immunotherapy.

103
Q

b. What is the pathophysiology behind the main side effects of immune checkpoint inhibitors?
Outline the common side effects. (2 marks)

A

ICI act by blocking proteins that inhibit immune responses, such as CTLA-4, PD-1, and PD-L1.
This boosts immune response to cancer, but at the cost of increased immune response to normal tissue
Primary mechanism is the enhanced activation of T-cells, which can effect not only cancer cells but also healthy cells, leading to inflammation and autoimmunity

  • Common Side Effects:
    • Dermatologic:
      ○ Rash
      ○ Pruritus
      ○ Vitiligo
    • Gastrointestinal:
      ○ Diarrhea
      ○ Colitis
    • Hepatic:
      ○ Hepatitis
      ○ Elevated liver enzymes
    • Endocrine:
      ○ Hypothyroidism or hyperthyroidism
      ○ thyroiditis
      ○ Adrenal insufficiency
      ○ Diabetes mellitus
    • Pulmonary:
      ○ Pneumonitis
    • Musculoskeletal:
      ○ Myalgia
      ○ Arthralgia
    • Renal:
      ○ Nephritis
    • Neurological:
      ○ Peripheral neuropathy
      ○ Meningitis
104
Q

Discuss the evidence that supports the routine use of immunotherapy in the management of melanoma with no BRAF driver mutation. (2 marks)

A

ICIs such as anti-PD-1 (pembrolizumab, nivolumab) and anti-CTLA-4 (ipilimumab) enhance T-cell mediated immune responses and widely used in treating melanoma, including cases without BRAF mutations;
Melanomas, regardless of BRAF status, express high levels of neoantigens, making them effective targets for immune checkpoint blockade.

Multiple randomised trials have demonstrated benefit of immunotherapy in all setting s(eg. Neoadjuvant, adjuvant) regardless of BRAF status with improvement in OS and PFS in adjuvant setting, and EFS benefit from NA ICI, but no OS benefit

Eg.
- CheckMate 067 Trial:
○ Compared nivolumab, ipilimumab, and combination in untreated advanced melanoma.
○ Significant improvement OS & PFS for nivolumab alone, and in combination with ipi, compared to ipi alone
○ Benefits were seen regardless of BRAF mutation status
- KEYNOTE-006 Trial:
○ Compared pembrolizumab with ipilimumab in advanced melanoma.
○ Pembrolizumab demonstrated superior OS and PFS.
○ Included patients with and without BRAF mutations, both benefiting from pembro
- Observational studies confirm durable responses and manageable safety profiles for ICIs in BRAF wild-type melanoma.

105
Q

An 80 year old patient has a basal cell carcinoma (BCC) excised from the midline left lower eyelid. The lesion is 5mm in size with a focally positive deep margin and clear peripheral margins of 3mm. There is no perineural or lymphovascular invasion.
a. (3 marks)
i. How would you further assess this patient?

A

· Take a detailed history including
o Lesion chronology and symptoms
o Co-morbidities, contraindicaitons to surgery or radiotherapy, genetic syndromes, immunosuppressed, medications
o Previous skin cancers. Previous RT
o Performance status
o Pre-existing visual issues with left eye?
o Patient preference for treatment
· Examination to determine location, surgical recovery and functional issues post operatively
o Palpate for lymphadenopathy
o Primary closure vs graft/flap
o Cranial nerve exam on V, VII
· Check pathology report for aggressive subtypes, thickness

106
Q

An 80 year old patient has a basal cell carcinoma (BCC) excised from the midline left lower eyelid. The lesion is 5mm in size with a focally positive deep margin and clear peripheral margins of 3mm. There is no perineural or lymphovascular invasion.

ii. Discuss the management options for this patient.

A

· Adjuvant treatment
o Preferred option given positive margin and moderate risk of recurrence. If this recurs then may grow deep to skin and affect intra-orbital contents, impact function of eyelid/eye/ cranial nerves
o RT
§ Given the sensitive location adjuvant radiotherapy would be preferred
§ Could use superficial/orthovoltage RT or VMAT
o Surgery
§ Likely to affect function of left lower eyelid, result in poor cosmetic outcome
§ May not be able to gain margin clearance
§ Would require experienced surgeon
o Moh’s micrographic surgery may be better alternative to ensure margin clearance

· Observation and treatment if recurrence
May be the preferred option in a poor prognosis patient

107
Q

An 80 year old patient has a basal cell carcinoma (BCC) excised from the midline left lower eyelid. The lesion is 5mm in size with a focally positive deep margin and clear peripheral margins of 3mm. There is no perineural or lymphovascular invasion.

Clinically the patient has a 12mm vertical scar in the midline lower eyelid. There is no obvious residual BCC. A decision is made to offer adjuvant radiation therapy. b. Describe an appropriate radiation therapy technique and dose fractionation schedule. (3 marks)

A

· Pre-SIM
o Ensure wound healing and commence RT 4-8 weeks post operatively
· SIM
o Supine
o Anaesthetic eye drops + Internal wax coated eye shield
o Review photos of previous lesion location
o Mark up 10mm around previous lesion location and use custom lead cutout to shield surrounding normal skin.
o Assess practical suitability of KV cone placement for location
o Take photos, notes on set up
· Prescription
o 50Gy/20# prescribed to skin surface, using 150kv photons to treat to 6mm depth (would depend on lesion DOI)
o Review depth dose/ filter tables for department
o Choose appropriate filter to ensure 90% isodose at treatment depth
o Treating 9 days per fortnight

108
Q

What are the potential complications of adjuvant radiation therapy for a skin lesion near the medial canthus? (2 marks)

A

· Acute
o Lethargy
o Radiation Dermatitis
o Alopecia
o Discomfort with internal eye shield
· Late
o Pigmentation changes
o Alopecia
o Dry eye
o Uveitis
o Cosmetic changes
o Functional changes to eyelid
o Weeping eye
o Graft failure (if graft present)
Secondary malignancy

109
Q

Following surgery for cutaneous Squamous Cell Carcinoma (SCC) in the Head and Neck area, what high-risk factors would you consider when recommending adjuvant radiation therapy. (2 marks)

A

Adjuvant RT to primary after surgical excision:
o Absolute indications:
· pT4
· margin+ or close
· PNI (symptomatic or named nerve)
· recurrent disease
o Relative indications: ≥2 of:
· pT2/3
· >5mm depth
· margin<5mm
· poorly diff
· extratumoural PNI of unnamed nerve
· extratumoural LVSI
· immunosuppression
Lesion location that may make further surgery technically complex or impact on patient function/cosmesis

110
Q

A well 50 year old man undergoes resection of a desmoplastic cutaneous melanoma medial to his right scapula. There is no disease beyond the primary site. Histology shows perineural invasion of multiple nerves up to 0.4mm diameter.

i. What other features would you like to review on the pathology report when deciding on further management? (0.5 marks)

A

· Lesion size
· Margin clearance
· Breslow’s thickness/ Clark level
· Ulceration
· Neurotropism
· LVI
· TILs
· SLNBx
Satellite lesions

111
Q

A well 50 year old man undergoes resection of a desmoplastic cutaneous melanoma medial to his right scapula. There is no disease beyond the primary site. Histology shows perineural invasion of multiple nerves up to 0.4mm diameter.

ii. If no other high risk features are present, what would be your recommended management and justify your answer? (2 marks)

A

· Patient should have SLNBx
· Given the desmoid histology and PNI – Adjuvant RT recommended to primary
o 48Gy/20# using VMAT given proximity to spinal cord
§ Treating to underlying facia.
§ Wire scar + use 1cm bolus
§ CTVp= Scar + 3-4cm. (Treatment margins would depend on surgical margin clearance.)
§ PTV= CTV + 0.5cm
· MIA and NCDM retrospective studies showed improved OS and LC with Adj RT for Desmoid histology.

One year later he presents with right axillary lymphadenopathy. Axillary dissection reveals complete resection of melanoma in the affected lymph nodes. b. What features in the pathology specimen could prompt consideration of adjuvant radiation therapy? (1.5 marks)
· ENE
· Large nodal burden
o >2 Axillary lymph nodes
o Axillary LN > 4cm
· Matted nodes
· Positive margin
· Mitosis
· TIL
Molecular markers: BRAF ect.

112
Q

A well 50 year old man undergoes resection of a desmoplastic cutaneous melanoma medial to his right scapula. There is no disease beyond the primary site. Histology shows perineural invasion of multiple nerves up to 0.4mm diameter.

Adjuvant radiation therapy is recommended. Describe a suitable radiation therapy technique and dose fractionation schedule. Include expected side effects in your answer. (3 marks)

A

· Pre-SIM
o Ensure wound healing (treat 4 weeks post op)
o PET scan
o MDT discussion
· SIM
o Wire scar
o 1cm bolus to skin
o Arms Akimbo
o Fuse PET
· Prescription
o 48Gy/20# to PTV, using VMAT technique, 10 days per fortnight
· Volumes
o CTVn is involved nodal region + surgically disturbed tissue
o PTV= CTVn + 5mm
· Target verification with daily CBCT
· OARs
o Ipsilateral lung V17Gy <35%
· Side effects:
o Acute
§ Lethargy
§ Radiation dermatitis
§ Pain/discomfort
§ Alopecia
o Late
§ Alopecia
§ Cosmetic changes
§ Lymphoedema
§ Radiation Pneumonitis
§ Secondary malignancy

113
Q

A well 50 year old man undergoes resection of a desmoplastic cutaneous melanoma medial to his right scapula. There is no disease beyond the primary site. Histology shows perineural invasion of multiple nerves up to 0.4mm diameter.

A year later he develops metastatic disease to the brain which is asymptomatic. Staging reveals no other disease. Performance status remains excellent.

d. i. If there were a single 1.5cm lesion in the motor cortex, what treatment would you recommend and why? (1 mark)

A

· Recommendation would be for SRS initially
o Given small size
o Eloquent area of brain that may result in moderate post-surgical morbidity
o Preservation of remaining brain tissue
o Lesion size may be too large for immunotherapy alone
Commence adjuvant Ipilimumab/ Nivolumab and then maintenance Nivolumab

114
Q

A well 50 year old man undergoes resection of a desmoplastic cutaneous melanoma medial to his right scapula. There is no disease beyond the primary site. Histology shows perineural invasion of multiple nerves up to 0.4mm diameter.

A year later he develops metastatic disease to the brain which is asymptomatic. Staging reveals no other disease. Performance status remains excellent.

If there were 15 small lesions scattered throughout the brain parenchyma, what therapeutic options exist and what are the advantages and disadvantages of each. Which is your preferred option? Justify your answer. (3 marks)

A

Consider Dexamethasone + PPI if pt is symptomatic. This may impact on effectiveness of immunotherapy
· Upfront immunotherapy (Ipilimumab/ Nivolumab)
o Adv: If small then good intracranial response, as per ABC trial. Good extracranial response as well.
o Dis: Takes time for response, so not a good option if patient is symptomatic. Immunotherapy less effective if pt on Dexamethasone for symptom management
· Dabrafenib + Trametinib - but short lived intracranial response
· SABR all mets
· WBRT
o Adv: Able to gain local control and facilitate weaning off Dexamethasone if symptomatic, so immunotherapy can commence
o Dis: Neurocognitive side effects. Does not deal with extracranial disease. No strong evidence for WBRT.
· Best supportive care
o Adv: no treatment side effects. Suitable if pt not appropriate for other treatments.
Dis: Poor outcomes

115
Q

A 70-year-old male undergoes excision of a 2.0cm skin lesion from the right forehead. Histopathology demonstrates a poorly differentiated squamous cell carcinoma (SCC) with extensive perineural invasion extending beyond the tumour edge but with clear resection margins.

b. He has a 4cm horizontal scar over the right lateral forehead 2cm above the eye with no signs of local recurrence. There is no altered sensation and no lymphadenopathy.
Histology shows a 2.0cm x 1cm x 7mm thick poorly differentiated SCC. Peripheral margins are well clear, the deep margin is 0.3mm. There is evidence of perineural spread involving several nerves up to 0.2mm in diameter at the deep margin.
There are no signs of further disease on examination or imaging.
A decision is made to proceed with adjuvant radiation therapy. Describe a suitable radiation therapy technique and dose fractionation schedule.
Your answer should include a detailed description and justification for your choice of target volumes and what potential complications you would discuss with this patient. (5 marks)

A

· Pre-SIM
o Ensure wound healing
· SIM
o Supine
o Wire scar
o 1cm Bolus to skin
o Wire up GTV based on clinical pre-operative photos
o Thermoplastic mask
· Prescription
o 60Gy/30# to high risk PTV. 54Gy/30# to low risk PTV, using a VMAT technique
o 10# per fortnight
· Volumes
o Pre-op GTV: as per mark up at SIM
§ Given the clear radial surgical margins, no need to treat entire surgical bed. Just the tumour bed. Identification of GTV helpful to minimise treatment volumes
o HR-CTV: GTV + 20mm margin, clipped to bone. Extend to Supra orbital foramen.
§ Lesion was 7mm thick with the deep margin having minimal clearance. Aim would to be to treat at least 10mm beyond deep margin, to cover potential microscopic disease. Given no bone invasion on path, no need to specifically treat bone.
§ Given extensive PNI at resection margin, treat skin supplied by supra-orbital nerve from tumour, back to superior orbital foramen. No anterior grade nerve cover, given no symptoms.
o LR-CTV: Frontal nerve (V1) path in orbit from Supra-orbital foramen to superior orbital fissure.
§ No radiological involvement of V1, so cover zone 1 (Supraorbital foramen to superior orbital fissure) at elective dose to cover microscopic involvement.
o PTV60: HR-CTV + 5mm
PTV54: LR-CTV + 5mm

116
Q

List factors which increase the risk of nodal involvement in cutaneous SCC of the head and neck region. (2 marks)

A

· Tumour size 2cm
· LVI
· DOI >4mm
· Recurrent disease
· Location: eg. ears/lip
· Histo: Poorly differentiated, desmoplastic
· PNI
Immunosuppression

117
Q

A fit, 30 year old woman with a past history of melanoma of the left calf presents with a palpable left groin mass. Biopsy confirms metastatic malignant melanoma. There is no further disease apparent on examination and staging investigations. She undergoes an inguinal nodal dissection.

a. What are the factors you would take into account when considering adjuvant radiation therapy? Justify your answer. (3 marks)

A

Stage 3 –node positive melanoma of calf N1-3, inguinal dissection.

Pt: age, comorbidity, fertility preservation, pregnancy, contraindications (immune ,RT, targeted), geography
Tumour
Primary: histology (desmoplastic), PNI, surgical margins, LVSI, DOI >4mm, ulceration, satellite lesions, in transit metastasis
Recurrence
Node:
ECE, Large nodal burden (>3 nodes (less for axilla/neck/parotid)
Node>4cm
margins
Treatment
Tumour spill at surgery
Neoadjuvant/adjuvant immune or BRAF inhibitors
Location and adjacent OAR
Previous RT

118
Q

A fit, 30 year old woman with a past history of melanoma of the left calf presents with a palpable left groin mass. Biopsy confirms metastatic malignant melanoma. There is no further disease apparent on examination and staging investigations. She undergoes an inguinal nodal dissection.

A decision is made to give her post-operative adjuvant radiation therapy. b. Describe a suitable radiation therapy technique and dose fractionation schedule. (3 marks)

A

Treat 4 weeks post op (if complete healing)
48Gy/20 10#/fortnight VMAT/IMRT to L inguinal region +- primary site
Sim: wire scar, consider 5mm superflab bolus, supine, frog leg CT L5-mid femur, fuse PET
Volume: CTVn –entire nodal basin (inguinal) and surgical scar
PTV = 1cm
Daily CBCT

119
Q

A fit, 30 year old woman with a past history of melanoma of the left calf presents with a palpable left groin mass. Biopsy confirms metastatic malignant melanoma. There is no further disease apparent on examination and staging investigations. She undergoes an inguinal nodal dissection.

This woman subsequently develops in transit subcutaneous metastases on her thigh. d. What are the potential management options for this situation? (2 marks)

A

Resectable:
Surgery +- adjuvant/palliative radiation therapy

Unresectable:

Systemic therapy –BRAF inhibitor, doublet immunotherapy

Limb Chemotherapy –infusion 30min melphalan and actinomycin D

120
Q

A 58 year old man presents after local excision of a 3cm Merkel cell carcinoma from the upper lateral right arm (over the deltoid). There is a positive radial margin of 0.2mm and there is prominent lymphovascular space invasion.

b. What management options are available for this patient? What would you recommend? Justify your answer. (4 marks)

A

adjuvant radiotherapy -60Gy to primary bed for positive margin 50Gy/25# for axilla

Consider adjuvant nivolumab

Consider concurrent chemotherapy (carboplatin +-etoposide) as per TROG/retrospective studies –large tumour and positive margin. For local control benefit

Observation –high risk of local and regional recurrence >40%

Not recommended:

Reresection of primary aiming for 2cm margin, followed by adjuvant RT required due to large size of primary. Nodal dissection only if clinical node pos disease. However very rapid aggressive tumour, delaying RT increases recurrence.

121
Q

A 58 year old man presents after local excision of a 3cm Merkel cell carcinoma from the upper lateral right arm (over the deltoid). There is a positive radial margin of 0.2mm and there is prominent lymphovascular space invasion.

a. What further information do you require to determine a management plan for this patient? (3 marks)

A

b. Pt –fitness/ability for radiothertapy, chemotherapy, access
c. immunosuppression
a. Healing from surgery and local symptoms
b. SCF/axilla exam
d. Tumour
a. Staging PET
b. Slnb
c. MCC virus detection
e. Treatment:
a. Radiation suitability
b. Time to radiation treatment
There is no other evidence of disease.

122
Q

A 58 year old man presents after local excision of a 3cm Merkel cell carcinoma from the upper lateral right arm (over the deltoid). There is a positive radial margin of 0.2mm and there is prominent lymphovascular space invasion.

Describe your radiation technique.

A

Presim: skin MDT, check wound healed.
SIM: wire to scar and 5cm margin. 1cm bolus for scar
Supine, R arm akimbo, head to left
CT C1to below diaphragm, IV contrast
Fuse PET

Dose: 60Gy/30# to tumour bed and 50Gy/25# to axilla, VMAT, 6MV photon to PTV, Daily CBCT
CTVp = scar/bed + 5cm circumferentially, deep to fascia (1.5cm)
CTVn = axilla LN
PTV = 5mm expansion

OAR:
Brachial plexus 55Gy
Lung V5gy <50%, V20gy < 30%, V30gy < 20%
Heart
Oesophagus

123
Q

A patient develops osteoradionecrosis of the mandible.
i What factors increase the risk of this complication? (2)

How would you manage this?

A

post dental tooth extraction / major dental work,
dose V60Gy<14%, V56<30%

124
Q

A wide local excision, sentinel node biopsy and axillary dissection are done for a cutaneous melanoma.

a) Discuss the prognostic factors that you would look for in:

i. the pathology report (3 marks)

A

For TNM staging, T and N stage can be determined on the pathological report. Clinical assessment for M as below

DOI- Clark score/Breslow thickness , which will correspond to T stage and guide the surgical margins aiming for
Surgical margins: As a guide would aim for > 2 cm for T2-4, <1 cm for T1. But this may depend on location and what is feasible with surgery
Histopathological subtype: Nodular, desmoplastic worse prognosis
Features at risk of nodal metastases: PNI and LVI, High mitoses, ulceration.
TILs positive prognostic biomarker
Molecular mutations: BRAF, NRAS, cKIT. BRAF mutation positive if suitable for targeted therapy, otherwise indicates poorer prognosis compared with wild-type BRAF. NRAS in general poorer prognosis in younger patients with higher recurrence rates and CNS involvement.
Number of nodes received and number positive
Size of positive LN, evidence of ENE
In transit mets or satellite mets

125
Q

Compare and contrast the biological behaviour and growth patterns of Merkel cell carcinoma and malignant melanoma. (3 marks)

A
126
Q

What is the management of Lentigo Maligna?

A

Lentigo Maligna = MIS with solar elastosis
- Margins are difficult to visualise
○ Add 10mm margin to disease visualised to eye
- Options for Rx
○ Surgery 90-95% cure rate
§ Large defect, GA
○ RT 56Gy/28# 80-85%
○ 5% imiquimod 70-75% cure rate
§ Requires daily treatment for 4 months
§ RADICAL ph 3 trial RPA (A Hong)
□ 56Gy/28F 5.5 weeks vs imiquimod 5% 5 applications per week for 12 weeks
□ 6 month 95% response rate both arms, slightly lower response rate longer term in imiquimod arm (but not significant)
○ Observation

127
Q

How would you manage lymph nodes in melanoma?

A

Management of LNs: MIA sentinel node risk calc
* SLNBx if: as per MSLT-I
○ No SLNB for T1a<0.8mm
○ Indications
§ >0.75mm (rounded up to 8mm) with 1 other risk factor (mitotic rate, ulceration, LVSI, Clark 4/5)
§ 1-4mm thick
○ If declines SLNB –US monitoring of 1st echelon nodes
* ⇒Positive SLNBx (pIIIa/b/c):
○ Completion LNDx MSLT-I- improved 10yr DFS 65-71% , DMFS, and CSS 42->63% ; or
§ Additional staging information
□ 15-20% of SN+ve pt had additional LN
□ However upstaging only 6%
○ Close ultrasound FU (if small deposit <1mm) is preferred with salvage LND MSLT-II: Immediate LN Dx small improvement in 3yr DFS 68 vs. 63% but no difference in OS significant morbidity (lymphoedema), DeCOG-SLT
§ Patients with stage IIIA melanoma and SLN tumour deposits ≥0.3 mm in maximum dimension are at higher risk of disease progression and may benefit from adjuvant systemic therapy.
§ Stage IIIA patients with SLN deposits <0.3 mm in maximum dimension demonstrate 5-year melanoma-specific survival similar to those with pathologic stage IB (T2aN0) melanoma, with consideration for less
intensive radiologic surveillance and follow-up (NCCN)
□ Consider if other higher-risk features (e.g. >4mm thick, ulceration)
□ IIIA: BRAF/MEK inhibitor
* BRAFV600E mutation: Dabrafenib/trametinib COMBI-AD trial: Improved 5yr RFS 53 vs. 36% and 5yr OS 86 vs. 77% cf. placebo

128
Q

How would you manage melanoma brain mets?

A
  • Brain metastases
    ○ Large and/or symptomatic - local control is priority since progression can lead to rapid functional deterioration, impaired QoL, or death
    * Surgical resection
    □ Posterior fossa - preferred for large >3cm due to risk of effacement of 4th ventricle/cerebral aqueduct
    □ rapid symptomatic improvement and resolution of peritumoral edema
    □ Prompt control of edema and discontinuation of glucocorticoids are particularly important for patients who will initiate immunotherapy postoperatively
    * or SRS
    * Surgical resection + postop RT
    * BSC +/- WBRT
    ○ Small (<1cm), minimally symptomatic or asymptomatic
    * Both targetted therapy and ipi/nivo have CNS activity
    * Targeted therapy produces rapid but short durations of intracranial response, whereas intracranial responses with immunotherapy are sometimes slower, but more durable
    * First line
    ▪ If BRAF positive:
    * Ipi/nivo (preferred);ABC phase 2 RCT: improved intracranial response rate ~46% vs. 20% cf nivolumab monotherapy. similar intracranial RR to extracranial RR. Or
    * Dabrafenib + trametinib; COMBI-MB phase 2 non-randomised. Intracranial response rate 58% but short lived, median PFS 5.6 months or
    ▪ If BRAF neg
    * Doublet as above
    ▪ Single agent immunotherapy (nivolumab, pembrolizumab or ipilimumab)
    ▪ SRS or surgical resection
    * Second line (at intracranial disease progression)
    ▪ SRS or surgical resection (preferred); or
    ▪ Dabrafenib + trametinib if BRAFV600E mutant and not previously used. Use in this setting should be limited to small and minimally symptomatic brain mets
    ▪ Immunotherapy if dab/tram used first line
129
Q

What is the evidence for adjuvant immunotherapy for Stage III Melanoma?

A

Stage 3 melanoma: Adjuvant immunotherapy multiple trials improvement in OS and PFS

Metastatic melanoma
* Ipilumab ph3 RCT: increased overall survival

* CheckMate 067 Trial:
	○ Compared nivolumab, ipilimumab, and combination in untreated advanced melanoma.
	○ Significant improvement OS & PFS for nivolumab alone, and in combination with ipi, compared to ipi alone
	○ Benefits were seen regardless of BRAF mutation status
* KEYNOTE-006 Trial:
	○ Compared pembrolizumab with ipilimumab in advanced melanoma.
	○ Pembrolizumab demonstrated superior OS and PFS.
	○ Included patients with and without BRAF mutations, both benefiting from pembro Observational studies confirm durable responses and manageable safety profiles for ICIs in BRAF wild-type melanoma.