Haematology Flashcards

1
Q

What is Tumour Lysis syndrome?

A

Increase uric acid, calcium, K, PO4, serum Cr (more common with aggressive tumours)

eg, diffuse large B cell lymphoma [DLBCL],
Burkitt lymphoma,
high-grade B cell lymphoma with MYC and BCL2 and/or BCL6 gene rearrangements

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

What is the Epidemiology and risk factors for Hodgkin’s Lymphoma?

A

Incidence (Australian statistics)
- 800 cases annually
- Represents 10% of all lymphomas
- Uncommon malignancy (0.5% of all cancers)

Gender distribution varies with subtype
Median age = 30 years, bimodal at 70
- Most common cancer adolescents/young adults (not young children)
- Good prognosis 98%
- Rarely familial/geographic clustering

6% of childhood cancers
M:F ratio 2.5:1 for children <5yr; 1:1 for children 15-19yr
EBV found in 60% of childhood cases

Aetiology

1) EBV-related
	a. Most relevant for mixed cellularity and lymphocyte-depleted subtypes
2) Smoking
3) Socioeconomic status
	a. High SE = nodular sclerosing
	b. Low SE = mixed cellularity
4) Immunosuppression
	a. Generally results in EBV-related 
5) Familial
	a. Related to HLA variability (multi-gene)
	b. No single high-risk allele
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is the pathogenesis for Hodgkin’s Lymphoma?

A

EBV-mediated

Often EBV-mediated
	○ Latent infection
	○ Leads to expression of LMP1 protein
		§ Assists in evasion of apoptosis
		§ Activates NF-kB pathway
		§ Upregulates PD-L1 expression (Contributes to evasion of T cell immunity)
EBV infection alone is not sufficient for Hodgkin's Lymphoma
	○ Very common infection, but malignancy is rare
	○ Abnormal HLA variations likely increase susceptibility
	○ Immunodeficiency eg HIV more EBV related lymphoma

Somatic Hypermutation

Clonal Immunoglobin heavy-chain (IgH) re-arrangements are characteristic in Reed-Sternberg cells
	○ This leads to somatic hypermutation
Activation of NK-kB transcription factor is a common event
	○ Further mutational events are broad and poorly understood Leads to a cell with varied and broad mutational profiles
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are the subtypes of Hodgkin’s Lymphoma?

A

1) Classical Hodgkin’s
a. Nodular Sclerosing (70%)
b. Mixed Cellularity (20%)
c. Lymphocyte Rich (5%)
d. Lymphocyte Depleted (<1%)
2) Nodular Lymphocyte Predominant Hodgkin’s (5%)

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

Describe the pathology for classic Hodgkin’s Lymphoma

A

Macroscopic
○ Rubbery nodes with abnormal morphology
§ Effacement of the nodal architecture
§ Variable fibrosis (prominent in nodular sclerosing)

Microscopic
	○ General
		§ Effacement of normal nodal architecture
		§ Infiltration with heterogenous lymphocytic infiltrate
	○ Reed-Sternberg cells are diagnostic, with mixed inflammatory background
		§ Large abnormal lymphocytes with multi-lobulated nuclei Prominent nucleolus and abundant cytoplasm Further aspects vary by subtype: NB all treated the same mmunohistochemistry
	○ POS = CD15 & CD30 pos (RS cells) & PAX5
	○ NEG = CD19/20 (B-cell), CD3 (T-cell), CD45 (leukocyte common antigen)
	○ EMA= epithelial membrane antigen
No clonal cells on flow cytometry (RS cells too infrequent)

PDL1
	○ overexpression of PD1 ligands including PD-L1 and PD-L2 common on RS cells 
	○ copy no. variation and genetic alterations at chromoseome 9p24.1 and increased JAK2 signalling account for majority of cases with overexpression of PDL1/2
	○ PDL1/2 alterations are a defining feature of HL and result in very high expression of PDL1/2 on the cell surface
		§ protects from T cell mediated killing 
	○ PDL1 expression and MHC class II positivity on RS cells are predictors of favourable outcome after PD1 blockade
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Describe the pathology for Nodular lymphocyte predominant Hodgkin’s Lymphoma

A

Similar behaviour to Follicular lymphoma

Macroscopic
	○ Rubbery nodes with abnormal morphology
	○ Effacement of the nodal architecture

Microscopic
	○ RS cells are infrequently seen
	○ Lymphocytic and histiocytic (L&H) cells are much more plentiful
		§ Multilobed nuclei and thin nuclear membrane ("popcorn cells")
	○ Heterogenous inflammatory infiltrate remains

Immunohistochemistry
	○ POS = CD20 (B cell), CD45 (leucocyte common antigen), PAX5 
	○ NEG = CD15, CD30 (RS cells), CD3 (T cell marker)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What is the relationship between HIV and Hodgkin’s Lymphoma?

A

HL is a non AIDS defining cancer
Relationship between HIV and EBV
- Immunological factors:
· HIV+ pt are immunosuppressed with decrease CD4 T cells-> decreased cellular surveillance mechanism and increases risk of malignancies, including HL
· HIV infection leads to Chronic B cell activation and cytokine dysregulation which promote lymphoproliferative disorders
- Co-infection
· Coinfection of HIV and EBV lead to higher prevalence of EBV in the host (x8), due to impaired immune response -> result in EBV persistence and oncogenic process.
· HIV pt are also more susceptible to other oncogenic virus eg HHV-8
- Clinical feat of HL in HIV pts
· HL in HIV+ pt are more aggressive and more advanced at dx
· Higher incidence of atypical and extranodal presentation
· Subtype mixed cellularity and lymphocyte depleted are more common
- Impact on HIV progression and treatment
· Risk of HL increases with more advanced HIV disease, but HL can still occur in pts on preserved immune system while on ART
· HIV pt have lower tolerance to chemo regimen due to immunosuppression and potential organ compromise, increase risk of opportunistic infection and complications
· ART has changed the outcomes of HL in HIV+ pts
· ART has extended life expectancy in HIV+ pt hence more likely to develop cancer prognosis and outcome
- Prognosis and surveillance
· If HIV is well controlled, outcome and RR of HL is similar to normal population
· Longterm surveillance- continuous monitoring of both HIV related complication and secondary malignancies is crucial

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

What are the prognostic factors for Hodgkin’s Lymphoma?

A

Patient Factors
- Age
- Performance Status
- B-symptoms

Tumour Factors
- Lugano Stage
○ Including number of nodal sites
- Size/bulk of tumour
- Blood markers
○ ESR
○ Hb (<105)
○ WCC (>15)
○ Lymphopaenia (<0.6)
○ Albumin (<40)
- Presence of EBV
○ Inferior prognosis
- Extranodal disease

Treatment Factors
- Incorporation of radiotherapy into management
- High-volume treatment centre

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

Describe the presentation and history/ examination for Hodgkin’s Lymphoma

A

Presentation
- Usually presents with painless, superficial enlarged LN , often contiguous LN chains in a predictable manner
- Later in disease course&raquo_space; haematogenous spread occurs with vascular invasion
- Majority supradiaphragmatic disease, isolated infra-diaphragmatic disease in 3-7%
- Approx 60-70% present with cervical and/or SCF LN
- 30% axillary
- Majority 50-60% mediastinal
- Involvement of abdominal organs uncommon
- Only 10-15% had extranodal disease
○ Bone, BM, lung, liver
○ CNS rare
- 25% have B symptoms (fevers, night sweats, weight loss >10%)before LN; carriers worse prognosis in both early and advanced disease
- Alcohol induced painful lymphadenopathy (Mechanism ?prostaglandins ?vasodilation) unclear prognostic significance

Consultation

- History
	○ HPI
		§ Palpable lymphadenopathy (non-tender)
			□ Duration and tempo of disease
		§ Mediastinal mass
			□ Incidental on CXR
			□ Painful
			□ Superior vena cava syndrome
		§ Constitutional symptoms 25%
			□ Fever
			□ Night sweats (drenching)
			□ Weight loss (10% within 6mo)
		§ Cutaneous itch (pruritis) (not a B symptom)
		§ Alcohol induced pain occurring minutes after intake and localising to regions of LN 
		§ May have multiple GP visits but delayed diagnosis
	○ PMHx:
		§ Previous radiotherapy or chemotherapy
		§ Immunosuppression (including HIV)
	○ Medications
		§ Immunosuppression
	○ Social:
		§ Smoking
	○ Family History
		§ Lymphomas

- Examination
	○ General inspection
	○ Lymphatic examination
		§ Cervical
		§ Axillary
		§ Epitrochlear
		§ Inguinal
		§ Popliteal
	○ Abdominal palpation
		§ Hepatomegaly
		§ Splenomegaly
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What would be you’re workup for Hodgkin’s Lymphoma?

A

Work-Up

- Bloods
	○ FBC + ESR (prognostic)
	○ EUC, CMP and LFT
	○ LDH
	○ HepB/HepC/HIV serology (pre-chemotherapy)
	○ Vitamin D (people with normal vitamin D do better)
	○ pregnancy

- Histopathology
	○ Excisional biopsy preferred
		§ Large amounts of tissue may be required to identify the scant RS cells
	○ Core biopsy is a reasonable alternative
		§ FNAB is insufficient

- FDG-PET (staging) -uptake by all the reactive cells

- Cardiac TTE
	○ LVEF prior to anthracycline chemotherapy
- Lung function tests
	○ Prior to bleomycin
	○ Prior to radiotherapy
- Fertility management

- Routine bone marrow biopsy is NOT routine anymore, PET is sufficient for assessment of bone involvement
	○ FDG-PET has sufficient sensitivity
	○ A study of 454 HL pt: Stage I/II disease on PET, none had a positive BMT
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Describe the Cotsworld staging for Lymphoma.

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

Describe the Lugano Classification for Lymphoma. Also the factors impacting unfavourable Hodgkin’s Disease.

A

Suffixes
- A/B = presence or absence of constitutional symptoms
- E = involvement of extralymphatic organs (for stage I or II only)
- X = bulky disease (old system, not commonly used)

HD Favourable vs unfavourable:
3-BEE
3+ lymph nodes
Bulky mediastinum >1 third/10cm
ESR >50, or B symptoms ESR>30
Extranodal disease

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

Describe the unfavourable favourable risk grouping for Hodgkins lymphoma

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Describe the Deauville scoring for Lymphoma.

A

PET-response to chemotherapy

Deauville X = new areas of uptake unlikely related to lymphoma

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

Describe the general approach to management of Lymphoma patients

A

Many patients are young, and so treatment should be considered in this context
Risk base response adapted treatment.
Survival excellent, aim to decrease long term morbidity.
Most cases are chemosensitive, and OS is 86%

1) Discussion and consideration of late toxicities
2) Fertility
	a. Referral/Consultation for fertility preservation
	b. bHCG for all female patients of appropriate age
3) Smoking cessation
4) Psychosocial input
	a. Preferably within the context of a AYA service

Long-term follow-up (late effects clinic) is required
- Primary cancer surveillance
- Secondary cancer surveillance
- Late effects surveillance

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

Describe the management of early stage (Stage I + II) Hodgkin’s Lymphoma.

A

ABVD = adriamycin (doxorubicin), bleomycin, Vinblastine, Dacarbazine. 1 cycle = 1 month, chemo given fortnightly (D1 and D15)
SE: Adriamycin cardiac (sharp increase risk doses >400 mg/m2), Bleomycin pulmonary/allergic reaction, Vinblastine haematologic, dacarbazine nausea and vomiting, alopecia

BEACOPP =bleomycin, etoposide, (A) doxorubicin, cyclophosphamide, (O) vincristine, procarbazine, prednisolone, 21 day cycle, chemo day 1, 8
(Not for >65yo -high mortality)
○ More etop and procarbazine which are carcinogenic
○ Less bleomycin and adriamycin then ABVD

Similar response rates with all chemo, however BEACOPP slightly better cure rates; at cost of significant toxicity including development of secondary acute myeloid leukemia/MDS and sterility
-A Deauville 5 score post initial chemo would warrant a biopsy to inform subsequent therapy. If a biopsy is not feasible, treatment should be escalated (NCCN).

NOTE:
- Chemotherapy alone protocols result in inferior PFS (H10-F and H10-U trials)
○ Radiotherapy should in general not be omitted
○ Exception is –> unfavourable risk disease, with 2x2 chemotherapy, with CMR on PET4 (HD17)

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

Describe the management of Advanced stage (Stage III + IV) Hodgkin’s Lymphoma.

A

Consider including stage IIX (bulky disease) in the advanced stage treatment protocols
- Excluded from many early-stage trials

Chemotherapy

- x2 ABVD -> restage with PET
	○ If CMR -> x4 AVD (RATHL)
	○ If D4+ -> x3 BEACOPPesc -> Restage with PET
		§ CMR -> x1 BEACOPPesc +/- ISRT 
		§ D4+ -> biopsy -> treat as refractory if positive 
- x6 Bv-AVD (from ECHELON-1) -> restage with PET
	○ D4+ -> biopsy -> treat as refractory if positive 
	
- esc-BEACOPP in patients <60, those with neuropathy.
	○ 4 cycles then repeat PET
	
- Brentuximab only on PBS for relapsed/refractory disease

Radiotherapy

GHD0607 and HD0801 showed no statistically significant benefit of RT for bulky disease
	○ If PET2(-), PET6(-) 

- After completion of chemotherapy, perform a PET6
	○ If CMR, deliver 30Gy ISRT
	○ If D4+, consider Bx 
		§ If positive additional therapy as per relapsed/refractory disease
		§ Alternatively: deliver 36Gy ISRT
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Describe the management of Nodular lymphocyte predominant Hodgkin’s lymphoma.

A

All treatment decisions need to be made in the context of natural history:
- Indolent progression
- Excellent prognosis (even with advanced disease)
- Typically asymptomatic at most stages

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

What would be your approach to managing relapse/ refractory Hodgkin’s Lymphoma?

A
  • Goal is to achieve cure while limiting toxicity and complications from Tx.
    • Generally salvage/targeted chemo - > PET-> If CR then ASCT if eligible.
      ○ Can use sequential chemo, RT for any PET positive site before ASCT, targeted chemo, or immuno to help achieve CR before ASCT
      ○ Cure 50% with ASCT
      ○ ASCT long term toxicity: AML, MDS, infertility
      If ineligible for ASCT, then consider single agent/comb chemo, targeted chemo, immuno, and/or RT
    • Decision for RT must balance between LC vs longterm toxicity
      ○ Factors to consider:
      § RT for CR prior to HCT or consolidation in pts with CR who had bulky disease at relapse
      □ Consolidation RT can be given prior to ASCT in p who had PR
      □ Consolidation RT can be given after ASCT in pt who had bulky disease at relapse
      ○ Although no RCT data, several studies showed RT can control limited residual disease and contribute to improved prognosis
      ○ late first relapse- salvage chemo followed by RT without ASCT is an option for pt with late relapse ( several years)-> long term OS 50-80%.
      ○ Palliation RT with/without chemo for symptom relief
    • Chemo 6-8x then PET/CT
      ○ Localized area of metabolically active dx→ RT
      ○ Widespread metabolically active dx→ stem cell tx (if older + not suitable for tx- then give RT)
    • Prognosis depends on
      ○ Relapse free interval (most relapse will present within 2-3yrs)
      ○ Initial therapy (CT or not)
      ○ response to salvage chemo- If CR on PET-> 93% 2yrs PFS
    • Note:
      ○ For patients who relapse or become refractory, secondary therapies are often successful in providing another remission and may even cure the disease.
      ○ Multiple treatment options available but depend on several factors:
      § timing of the relapse (better if >12mo),
      § ECOG,
      § anaemia,
      § scope of disease,
      § suitability for further chemo/RT,
      § response to salvage chemo
      ○ Salvage RT dose up to 40Gy, yield higher RR and LC. Higher dose = more durable response if tolerated.
      ○ RT also play cytoreduction and consolidation in peritransplantation period in some programs to decrease rate of ASCT failures. 30-36Gy is recommended within 6 weeks post stem cell infusion.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Describe manage of Hodgkin’s Lymphoma in pregnancy

A

Management
- First trimester
○ Greatest fetal risk in this trimester due to implantation/embryogenesis and major manifestation of drug toxicity are spontaneous abortion or major morphological abnormalities.
○ Defer until second/third trimester if possible
§ Asymptomatic, non-bulky, sub-diaphragmatic
§ Need treatment if progressive disease/symptomatics/threatened well being (e.g. spinal cord compression, SVCO)
○ If need to treat:
§ ABVD*
§ Can give vinblastine alone then ABVD
- Second / third trimester
○ Defer to third trimester / post partum if possible- this allow adequate staging and selection of appropriate therapy
§ Don’t defer if: Bulky, stage III-IV, symptomatic
○ If treating:
§ ABVD
§ RT - only if must be given for local control of troubling symptoms above the diaphragm
□ Shield abdomen
□ Whole fetus dose <0.1Gy
- Post-partum
○ Treat as per normal
- Adverse fetal effects
○ Intrauterine growth restriction
○ Decreased IQ
○ Malformations
○ Low birth weight
○ Premature
○ Stillborn
○ Gonadal//ovarian damage
○ Carcinogenesis
- Indications for termination
○ Relapse -> needs autologous stem cell transplant
○ Prognosis - unable to care for baby
Need to discuss: Effects of treatment on future fertility

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

What is the evidence for non-risk adaptive management of favourable risk, early stage Hodgkin’s Lymphoma?

A

Note: Be aware “H” trials are for EORTC risk classifications, while “HD” trials are for GHSG risk classifications.

HD7 trial (Engert, 2007)
- 650 patients with favourable risk disease randomised to
○ 30Gy IFRT with 10Gy boost
○ 2x ABVD with same RT
- Outcomes
○ No difference in OS
○ RT alone resulted in inferior FFTF (67% vs 88%)
○ No difference in second malignancy risk

HD10 trial (Engert, 2010)
- Pre-PET era (1998-2003)
- 1370 patients with favourable risk disease randomised in 2x2 fashion to
○ 4 cycles vs 2 cycles of ABVD
○ 30Gy vs 20Gy IFRT

  • Outcomes
    ○ No difference in OS or FFTF for any comparison
    § I.e. 4x ABVD + 30Gy was not superior to 2x ABVD + 20Gy
    4x ABVD and 30Gy both independently increased toxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is the evidence for management of unfavourable risk, early stage Hodgkin’s Lymphoma?

A

HD11 trial (Eich, 2010) = current standard of care
(4x ABVD+30Gy)
- 1570 patients with unfavourable risk disease randomised in 2x2 fashion to
○ 4 cycles BEACOPP vs 4 cycles of ABVD
○ 30Gy vs 20Gy IFRT
- Outcomes
○ The only inferior arm was 4x ABVD + 20Gy IFRT
§ Inferior to 4x ABVD + 30Gy
○ 20Gy is acceptable if combined with 4x BEACOPP

HD14 (von Tresckow, 2012)
- 1655 patients with unfavourable risk disease randomised to
○ 4x ABVD + 30Gy IFRT
○ 2x ABVD + 2x BEACOPP + 30Gy IFRT
- Outcomes
○ BEACOPP improves 5 year FFTF rates (87.7% vs 94.8%)
○ No difference in OS
○ Marked increase in G3+ toxicity with BEACOPP (87.1% vs 50.7%)

H10-U trial (Andre, 2017)
- 1137 patients with favourable risk disease received 2x ABVD and then a PET, before being randomised to
○ Non-adaptive (1x ABVD + 30Gy INRT)
○ Adaptive approach
§ If CMR (D1-2), then 2x ABVD
§ If PR, 2x BEACOPPesc + INRT
- Outcomes
○ Following negative PET2, omission of RT could not be deemed non-inferior (5 year PFS –> HR 1.45)
○ Following positive PET2, BEACOPPesc improved 5 year PFS compared with standard non-adaptive arm (HR 0.42)
○ 10year update (2024): PFS no stat difference (2-4%)

HD17 (Borchmann, 2021)
- 1100 patients with unfavourable risk disease were given 2x ABVD + 2x BEACOPP, before a repeat PET
○ Non-adaptive (30Gy IFRT)
○ Adaptive
§ If CMR (D1-2), nil firther
§ If PR, 30Gy IFRT
- Outcomes
○ Following negative PET4, omission of RT was deemed non-inferior (5 year PFS difference of 2.2%)
○ Non-inferiority was sustained in bulky disease subgroup

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

What is the evidence for PET risk adaptive management of favourable risk, early stage Hodgkin’s Lymphoma?

A

Randomised PET Directed Therapy Trials for Early Stage Favourable HL (UK rapid, H10, HD16)
-PET does not mean omission of RT (ISRT cannot be omitted from SOC if interim PET D 1-2) without change in PFS and OS, ommission of RT as part of shared care with patient factors
-Allows de-escalation of chemo or escalation of chemo

H10 trial (Andre, 2017)
- 1137 patients with favourable/ unfavourable risk disease received 2x ABVD and then a PET, before being randomised to
○ Non-adaptive (1x ABVD (x2 ABVD for unfav) + 30Gy INRT) = ABVDx3 + INRT 30Gy
○ Adaptive approach
§ If CMR (D1-2), then 2x ABVD (fav) or 4x ABVD (Unfav)
§ If PR, 2x BEACOPPesc + INRT
- Outcomes
○ Following negative PET2, omission of RT could not be deemed non-inferior (5 year PFS –> HR 15.8)
○ Update 10 year (2024) PFS difference:
§ ABVD + RT = 99%, ABVDx4 = 85% p0.001
§ Both arms 4% secondary malignancy
○ Following positive PET2, BEACOPPesc improved 5 year PFS compared with standard non-adaptive arm (HR 0.42)

HD16 trial (Fuchs, 2019)
- 1150 patients with favourable risk disease received 2x ABVD and then a PET, before being randomised to
○ Non-adaptive (20Gy IFRT)
○ Adaptive approach
§ If CMR (D1-2), nil further
§ If PR, proceed to INRT
- Outcomes
○ Following negative PET2, omission of RT could not be deemed non-inferior (5 year PFS –> HR 1.78)
○ 2024 update: Per protocol 5year PFS better with RT 93% vs 86% p 0.04 =7%
○ No detriment to survival

UK RAPID
- Looked to demonstrate non-inferiority of x3 cycles of ABVD and if PET negative observation
○ Was not able to show non-inferiority, but the prognosis of HL with or without RT is still very good *
- 602 pts RCT ABVDx3 and PET negative observation vs 30Gy IFRT
- RT gives 3-6% PFS advantage*

CALGB 50604 –not randomised. ABVDx2 PET neg ABVD x2 = ABVDx4. PFS = 91% = acceptable
Acceptable to quote in patients you wish to avoid RT in

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

Describe the evidence for management of advanced stage Hodgkin’s Lymphoma.

A

EORTC trial (Aleman, 2007)
- 739 patients with advanced disease were given 6x MOPP chemotherapy and stratified and randomised to
○ If CR, randomised to
§ Observation
§ IFRT
○ If PR, given 30Gy IFRT
- Outcomes
○ 8 year PFS and OS for PR patients was 76% and 84% respectively
§ Not significantly different to either CR arm
§ Suggestive of good benefit to RT for PR
○ No benefit to additional RT in the CR arm
○ Note that PR patients had bulky disease more often than CR disease

Retrospective review (Phan, 2011)
- 118 patients with stage III disease were reviewed
- Outcomes
○ Presence of bulky disease was associated with worse DFS
○ Mediastinal RT was associated with improved DFS and OS
- Extrapolate that RT for patients with bulky disease improves outcomes

RATHL trial
- IIA unfavourable or advanced disease (III, IV), Interim PET neg
- ABVD x2
○ If PET positive (centre choice)
§ Esc BEACOPP x3
§ BEACOPP-14 x4
○ If PET negative (randomised)
§ ABVD or AVD x4
- No difference if OS or PFS in PET negative group. 7 years PFS 80%
- No difference between esc-BEACOPP or BEACOPP in PET positive group
- ABVD x2 followed by 4 cyles AVD accepatble

GHSG Escalated BEACOPP for advanced stage
- Randomised baseline BEACOPP (bleomycin, etoposide, adriamycin, cylophosphamide, vincristine, procarbazine and prednisone) + escalated BEACOPP
- Improvement in FFTF and OS in esc BEACOPP
- There have been other RCT comparing escBEACOPP with ABVD, all have favoured escBEACOPP PFS

ECHELON 1- Brentuximab (Swap bleomycin for brentuximab) 2018
- Phase 3, Stage III-IV Hodgkin lymphoma
- Brentuximab vedotin, doxorubicin, vinblastin and dacarbazine (A+AVD) vs ABVD
- Radiation was allowed for PRs PET+ and >2.5cm
- 6yr OS 94% vs 89%
- Improved OS with A+AVD

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Describe your Involved site radiotherapy technique for Hodgkin’s Lymphoma

A

Involved Site Radiotherapy
Patients
1) Favourable Risk - Stage I-II (20Gy)
2) Unfavourable Risk - Stage I-II (30Gy)
3) Advanced Disease with Initial Bulk (30Gy)

Pre-simulation
MDT discussion
Fertility consideration
- bHCG in all females
- Fertility preservation
Smoking cessation

Aim commencement 4-6 weeks post-chemo

Simulation
Supine with arms above head -decide if up or down or abducted
Bra off
Vacbag

DIBH if superior mediastinum

Generous CT (2mm with contrast)

Fusion
Pre-chemotherapy PET

Dose prescription Favourable risk
- PET neg 20Gy/10F
Unfavourable risk
- 30Gy/15F -(don’t need any higher)

Advanced disease (with bulk)
        - 30Gy/15F
        - Boost to 36Gy/18F if residual disease

Children 19.8-25.5Gy see ILROG guideline

VMAT technique or butterfly IMRT (women)
10 days per fortnight

Volumes
GTV (residual)
- Residual disease on PET

CTV
        - Use pre-chemo volume to establish superior/inferior margins  (note  PET free breathing)
                ○ Consider extra 1cm sup/inf if significant imaging uncertainty
        - Use current CT to delineate radial margins (tumour shrinkage)
                ○ Infiltrated chest wall/muscle
CTVboost
        - GTV + 1cm
ITV
        - Consider if mediastinal disease
PTV
        - CTV + 7mm

Target Verification
Daily CBCT

OARs
Bilateral lung
- V20 < 30%
- V5 < 50%

Heart
- Mean < 5Gy (critical 15Gy) (similar to 1 cycle ABVD)

Breast
- Mean < 4Gy (critical 15Gy)

Spinal Cord
- Dmax < prescribed dose
Thyroid V30<60%

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

Describe the difference between EFRT, IFRT, ISRT, INRT; in Lymphoma Treatment

A
  • movement to decrease field of RT to ↓acute and long term tox, whilst maintaining survival rates
    • EFRT: extended field, includes clinically involved and adjacent clinically uninvolved sites (eg. mantle, inverted Y)
    • IFRT: limited to involved regions eg. mediastinal + low bilateral SCF (entire mediastinum)
    • Large fields, now reserved for salavge patients of patients who fail chemotherapy and are not suitable for further lines of more intense chemo
    • Defined as the anatomical boundaries of a whole lymphatic region
    • ISRT: only pre- and post-chemo nodal volumes + limited margin of healthy tissue to accommodate uncertainties in determining pre-chemo tumour volume
      ○ originally involved SITE,
      ○ ISRT preferred by ILROG
      ○ volume accounts for tumour shrinkage,αnatomy changes post chemo, spares adjacent uninvolved organs
    • ‘Generous’ ISRT for early/indolent stage lymphomas as no chemo
    • For extra-nodal disease: CTV generally consists of the entire organ (Stomach, thyroid). Partial organ ISRT is well defined CTV volume e.g. orbit, breast. Uninvolved LN not targeted○ consider an ITV
      ○ PTV expansion appropriate for site/immobilisation/image guidance
    • INRT: includes pre-and post chemo nodal volumes + ver limited margin of healthy tissue (5-10mm)
      ○ requires optimal pre-chemo PET/CT acquired with pt in treatment position, fused with post chemo imaging
      considered form of ISRT with more strict requirements for pre chemo imaging
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is the prognosis of Hodgkin’s Lymphoma based on risk group/ stage?
How would you follow up a patient with Hodgkin’s Lymphoma?

A

Follow-Up

* Clinical review every three months for the first two years (shared care)
	○ Clinical examination
	○ FBC, ESR at each visit
* Clinical review every six months for years 3-5
	○ Clinical examination
	○ FBC, ESR at each visit
* Annual surveillance thereafter for late effects
	○ Long-term follow-up required especially for NLPHL

* Single FDG-PET at 12 weeks post RT
* Consider routine CT every 6-12 months for the first two years
	○ Minimise, particularly in the young

* Consider annual TFTs if neck irradiated
	○ Up to 60% hypothyroid. 1/3000 risk of cancer
* Consider TTE and carotid US
	○ Stress echo at 10 years
	○ Coronary artery exposure
* Consider early breast screening
	○ 10 years after RT
	○ Age 40 years

Survivorship
cardiac, -IHD, heart failure, valve dysfunction
secondary cancer (9% @ 30 years)
Breast cancer -high dose 30Gy = 25% cancer
No risk if age >35
48.5% @ 40 years (20% general population
More chronic health conditions (40%)
Fatigue -exercise phyiologist

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

Discuss the expected toxicity after lymphoma treatment.

A

Toxicity
- In 1st 10yrs after treatment- most likely to die from HD; after that more likely to die from Rx related toxicity (in particular cancer + cardiac dx).
- Second malignancy
○ ~15% at 15yrs (higher than other cancer treated in young pts- ? genomic instability in pts with HD). Relative risk related to age at treatment. Survival after diagnosis of cancer is poor.
§ ALL. 3% risk at 15yrs. Most pts die within a year of diagnosis.
§ NHL. 1.5% risk at 15yrs (latency ~7yrs)
§ Solid tumours (75% of all 2nd malignancies), 12% risk at 15 yrs. Main solid tumours: lung, breast due to XRT. Other sites: thyroid, GI, bone + soft tissue, melanoma.
- Cardiac dx
○ IF MHD <4-4.5Gy, then risk is insignificant. However each increase in MHD >5Gy, yields the same excess risk of cardiac event as an increase in Anthracycline dose of 50mg/m2 (eg. 1x cycle of ABVD or CHOP)
Latent toxicities of Cardiac dx: arrhythmias, MI, pericarditis, myocarditis, pericardial effusion, pericardial fibrosis, cardiac death.
○ Stanford data (1998) 3.9% survivors died from cardiac dx at 10yrs. RR death 3.1, but only in pts getting > 30Gy to mediastinum.
○ Related to XRT field size, risk lingers even 30+ yrs after Rx.
- Thyroid failure
○ Due to XRT lower neck
○ ~50% pts
- Infertility
○ Esp. procarbazine, alkylating agents

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

What is the epidemiology and risk factors for DLBCL?

A

Incidence
- Around 2000 people diagnosed annually
- Most common form of non-Hodgkin lymphoma (30% of all lymphomas)

Median age is 60 years
Slight male predominance (1.5:1)

Clinically aggressive, rapid but curable
Often arises in LNs but can be present anywhere
70% of cases are nodal
30% are extranodal
- GIT most common extranodal
- Testes, eyes, Mediastinum, CNS

Aetiology

1) Previous low-grade lymphoma
	a. Transformation (e.g. follicular, CLL)
2) Immunosuppression
	a. Iatrogenic (organ transplant)
	b. HIV-associated
3) Environmental exposures
	a. Hair dyes
	b. Pesticides

Limited familial link

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

Describe the pathogenesis for DLBCL

A

Pathogenesis

* Cell of Origin 'GCB vs ABC'
	○ DLBCL can arise from two B-cell ancestor cells
		§ Germinal-centre type (CD10, BCL6) --> favourable
		§ Activated B-cell type (MUM1) ~50%
		§ "Type 3" 

* BCL-6
	○ One of the key mutations that occurs is the overexpression of BCL6 gene (near 100% of cases)
		§ Usually functions as an anti-apoptotic gene
		§ Over-expression leads to down-regulation of pro-apoptotic genes such as p53
	○ Multiple rearrangements exist to manifest this change
		§ t(3:14) --> BCL6 and IGH is present in 30%
		
* BCL2:
	○ Inhibit apoptosis: primary function of the BCL2 gene is to inhibit apoptosis, the programmed cell death mechanism in cells. By doing so, it helps regulate cell survival and maintain a balance between cell death and cell growth.
	Cell Survival: Proteins encoded by the BCL2 gene are integral to the survival of cells, as they prevent the activation of caspases, which are enzymes that play a key role in initiating apoptosis.

* TP53
	○ Separate to the BCL6-mediated down-regulation, p53 mutations are common in DLBCL
		§ Loss of function mutations
		§ Deletions
	○ Again, this impairs apoptosis and cell-cycle arrest machinery

Double-hit and triple-hit

* MYC rearrangements occur in 10% of patients
	○ Associated with a very poor prognosis
	○ Rearrangements often involve the IGH (chomosome 14) or BCL6 (chromosome 3) genes

* When combined with BCL2 or BCL6 rearrangements, prognosis is particularly poor

* Terminology:
	○ Double-expressor = based on IHC (protein over-expression)
	○ Double-hit = based on ISH/genotyping (genetic rearrangement)
* Both forms are associated with poor prognosis

Low grade lymphomas can develop into high grade through transformation
Triggered by accumulation of additional transforming mutations

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

Describe the pathology of primary B-Cell Mediastinal Lymphoma.

A

Aggressive rapidly progressive, anterior mediastinal mass causing obstruction.
Arises from thymic B cell mass in 20-40yo, female

Micro: mimics DLBCL or hodgekins –sheets of large lymphocytes, clear cytoplasm dense collagen deposition (compartmental fibrosis)

IHC
Pos: CD19 , 20, 79a, CD45, BCL6 (same as DLBCL)
CD30 (same as hodgekins)
Negative:
Immunoglobulin (DLBCL), CD15 (Hodgekins), CD 3,4,5,8, EBV

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

Describe the pathology of DLBCL

A

Macroscopic
- Large matted lymphadenopathy
- Effacement of normal nodal architecture

Microscopic
- Diffuse growth pattern with effacement of normal nodal architecture
- Sheets or large cells: transformed mature proliferating B cells
○ Nuclei enlarged 5x normal lymphocytes
○ Resemble centroblasts (pale abundant cytoplasm with non-cleaved nuclei)
○ Large, irregular and multi-lobulated nuclei
- Mod- High proliferation index (Ki67 often > 70%)

Immunohistochemistry
- POS = CD19, CD20, CD79a (indicating B cell lymphoma, Non hodgkin type), surface Ig (often IgM) (stain positive for B cell markerts), CD45
○ GCB-type = CD10 (=GCB type), BCL6
○ ABC-type = MUM1 (50%)
○ Poor prognostic = FISH-MYC, BCL2 t(14:18)
CD5
§ p53
- NEG = CD3, CK

Molecular
- 30% have t(3:14) –> BCL6 and IGH
- 30% have t(14:18) –> BCL2 and IGH (as in follicular lymphoma)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Describe the prognostic factors for DLBCL

A

Patient Factors
- Age
- Performance status
- Constitutional symptoms (B symptoms)
- Serum markers
○ LDH above ULN

Tumour Factors
- Histological grade (i.e. aggressive lymphoma)
- Lugano Stage
○ Number of nodal sites
○ Number of extranodal sites (>1)
- Histological subtype
○ GCB vs ABC (ABC worse)
- Gene expression
○ Double/triple hit
- p53
- “Bulk of disease” (usually defined as > 7-10cm)

Treatment Factors
- Response to first-line therapy (PET evaluation)

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

Describe the presentation and history/examination for a patient with DLBCL.

A

Presentation

Clinically aggressive
Often arises in LNs but can be present anywhere
70% of cases are nodal
30% are extranodal
- GIT most common extranodal
- Testes, eyes, Mediastinum, CNS, thyroid, bone, kidney, liver, breast and skin

20% stage I disease
40% disease limited to one side of diaphrgam (stage II)
20% above and below
40% widespread with extranodal (Lung, liver, kidney, BM; in contrast of PRIMARY extranodal)

30% of pt reprot B symptoms

Consultation

- History
	○ HPI
		§ Palpable lymphadenopathy (non-tender)
			□ Duration and tempo of disease (typically rapidly growing)
		§ Emergency presentation is common
			□ SVCO
			□ Spinal Cord Compression
			□ Tumour lysis syndrome
			□ Airway obstruction
			□ Renal obstruction
		§ Constitutional symptoms
			□ Fever
			□ Night sweats (drenching)
			□ Weight loss (10% within 6mo)
		§ Cutaneous itch
	○ PMHx:
		§ Previous radiotherapy or chemotherapy
		§ Previous low-grade lymphoma 
		§ Immunosuppression (including HIV)
	○ Medications
		§ Immunosuppression
	○ Social:
		§ Smoking
	○ Family History
		§ Lymphomas

- Examination
	○ General inspection
	○ Lymphatic examination
		§ Cervical
		§ Axillary
		§ Epitrochlear
		§ Inguinal
		§ Popliteal
	○ Abdominal palpation
		§ Hepatomegaly
		§ Splenomegaly
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Describe the investigations you would organise for a patient with DLBCL

A

Work-Up

- Bloods
	○ FBC
	○ EUC, CMP and LFT
	○ LDH
	○ HepB/HepC/HIV serology (pre-chemotherapy)

- Histopathology
	○ Excisional biopsy preferred
		§ Large amounts of tissue may be required 
	○ Core biopsy is a reasonable alternative
		§ FNAB is insufficient

Staging

- FDG-PET (staging)

- Consider lumbar puncture if at high-risk of CNS involvement
	○ Subtypes (double-hit DLBCL, Burkitt's, ATCL)
	○ HIV-associated
	○ Location (epidural/eye, bone marrow, kidney/adrenal, testicular, paranasal sinus)

- Routine bone marrow biopsy is NOT required (only to confirm limited stage)
	○ FDG-PET has sufficient sensitivity
	○ Additionally, BM can be omitted for stage III DLBCL because does not change management

- Lumbar puncture/MRI if high risk disease
- Opthalmic assessment if CNS
- Testicular Ex + US
- GI endoscopy and biopsy if seen on PET

Pre-treatment

- Cardiac TTE
	○ LVEF prior to anthracycline chemotherapy
- Lung function tests
	○ Prior to bleomycin
	○ Prior to radiotherapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What would be your considerations prior to treatment of a patient with DLBCL

A

General Approach

Most patients should be treated with curative intent
- Even stage IV disease is curable

Considerations prior to treatment
- Performance status and fitness
- Organ function
○ TTE (anthracyclines)
○ PFTs (RT +/- bleomycin)
- Risk of CSF involvement
○ May need LP
○ Consider CNS prophylaxis (HD-MTX or IT-MTX)
- Consider fertility

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

Describe the management of early stage, non-bulky, IPI low DLBCL

A

In general:
Favour chemotherapy alone if non-bulky

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

Describe the management of early stage, non-bulky, IPI high DLBCL

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

Describe the management of early stage, bulky DLBCL

A

Key Points

- If initially bulky disease
	○ Should have combined modality therapy (RT improves OS and PFS)
	○ Give 36Gy ISRT
	
- Should perform a post-chemotherapy FDG-PET before proceeding
- If FDG-avid residual disease after chemotherapy Give 40Gy ISRT
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Describe the toxicity of CHOP chemotherapy in DLBCL

A

CHOP: cyclophosphamide, doxorubicin, vincristine, prednisolone, 6 doses every 21 days
Toxicity:
* Cyclophosphamide: alopecia, myelosuppression, N&V, infertility, 2nd leukaemia
* Doxorubicin: cardiac, lifetime dose, myelosupp, 2nd leuk
* Vincristine: PN, not much myelosupp
* Pred: high dose for 5 days, cytotoxic to WBC
Note: Pt who are exposed to HBV are at risk of reactivation during treatment with RCHOP-> NEED antiviral prophylaxis or periodic HBV DNA monitoring and antiviral treatment in the case of reactivation

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

Describe the management of Advanced stage DLBCL

A
  • Induction chemotherapy with 6x R-CHOP
    ○ No rationale for maintenance rituximab
    • Use PETCT to assess response to chemo and guide need for RT
    • Consideration should be given to consolidation radiotherapy -> to improve PFS, OS and LC benefit
      ○ Indications for consolidation RT
      § Initially bulky disease >7.5cm
      § Skeletal metastases
      § Extranodal sites of disease
      § Metabolic partial response
      ○ Technique
      § 36Gy ISRT if complete metabolic response
      * consider dose reducing to 30Gy/15F if large volume with CMR
      40Gy ISRT if residual avidity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Describe CNS Prophylaxis in management of NHL

A
  • CNS relapse of NHL is usually fatal despite Tx.
    • CNS IPI used by haematologist as score to stratify risk
      Prophylaxis desirable in high risk cases for CNS relapse
    • Use in
      ○ CSF positive
      ○ Adrenal, renal and Testicular, (NB prophylactic treatment also of contralat testicle)
      ○ Paranasal sinus
      ○ BM involvement
    • Retro analysis:>1 extranodal site and ↑LDH independent predictors of CNS relapse.
      Usually given as intrathecal methotrexate (+/- cytarabine) or IV high dose MTX.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Discuss management of Relapsed DLBCL

A

Relapse
* More than 30% of DLBCL relapse, 1/100000 annually
* Refractory/Relapse disease 3yr PFS 37% and 3yr OS 49%
* Important prognostic factors: initial prognostic factors, nature of previous treatment and time interval
* Dx- core biopsy should be repeated before proceeding with 2nd line therapy
* Re-staging
* Treatment:
○ IF good PS, salvage Rituximab and chemo followed by HDC and ASCT (only 50% of pts proceed to ASCT)
* R-DHAP, R-ICE, R-GDP (less toxic), BEAM
* R-GEMOX if cannot tolerated high dose therapy. Pixantrone is a new anthracycline with less cardiac toxicity
○ IFRT or iceberg RT may be used in limited stage disease
○ No role for maintenance rituximab
○ Allogeneic transplant (may be considered in pt with refractory disease, early relapse or relapse after ASCT)
* Site of further relapse after ASCT is usually the original site of relapse or refractory disease. PARMA trial-> consolidation RT can reduce this despite CR to salvage chemotherapy
* Indication for RT in relapse/refractory disease–> reduce LRR and improve RFS
○ Localised relapse/refractory disease that can be encompassed by RT with acceptable toxicity
○ IF disseminated disease, RT to selected area
* Locoregional site with Incomplete response to salvage chemo
* Dominant skeletal relapse
* Site where LC is critical eg. threatened spinal cord, nerve root compression, SVCO, aerodigestive or urological tract obstruction
* Peri-transplant RT for potential cytoreduction residual active disease

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

Describe the management of Testicular DLBCL and Primary mediastinal DLBCL

A

Testicular
- Orchidectomy
- 6x R-CHOP chemotherapy
- CNS prophylaxis (IT or HD-MTX)
- 30Gy/15F to contralateral testis
○ 15% risk of relapse

Primary Mediastinal DLBCL –generally bulky at dx
- Limited stage (encompassable with RT field)
○ 6x RCHOP + 36Gy ISRT (same as DLBCL)
* If giving R-CHOP need RT, if not need more aggressive chemo DA-REPOCH
○ 6-8x R-EPOCH alone (if PET complete response no RT)
§ If PET positive –biopsy –consider salvage
§ RT not effective if large residual
- Advanced stage (unencompassable)
○ 6-8x R-CHOP (+ 36Gy ISRT if disease limited to chest)
○ 6-8x R-EPOCH alone

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

Describe the evidence for management of Non-bulky DLBCL

A

Summary:
- RT is associated with improvement in OS when there is initial bulk
- This benefit is uncertain in non-bulky disease
○ Chemotherapy alone could be considered

SEER analysis (Ballonoff, 2008)
	○ 13420 patients with early-stage DLBCL were analysed
		§ 41% had received RT
	○ Outcomes
		§ Use of RT was associated with a significant CSS and OS benefit
			□ This persists to 15 years of follow-up
			□ This persists on multivariate analysis

LYSA 02-03 randomised trial (Lamy, 2018)                                    Radiation deescalation is ok for nonbulky
	○ 334 patients with stage I-II DLBCL without bulk and IPI=0, completed R-CHOP-14 chemo and proceeded to PET
		§ If CMR achieved, randomised to 40Gy RT consolidation vs observation
	○ Outcomes
		§ Addition of RT did not improve EFS or OS at 5 years
	
	○ RICOVER-60 (Pfreundschuh, 2008)
		§ 1222 elderly (>60yo) patients with DLBCL (any stage or IPI) were enrolled on RICOVER-60 and randomised to
			□ 6 vs 8 cycles of CHOP
			□ With or without 8x rituximab
			□ 36Gy ISRT allowed if bulky disease
		§ Outcomes
			□ 8 cycles of chemotherapy was not superior to 6 cycles for OS or PFS
			□ Addition of rituximab to 6x CHOP improved OS (HR 0.63) and PFS (HR 0.50)
			□ No major differences between 6x R-CHOP vs 8x R-CHOP
		§ Thus, 6x R-CHOP is the preferred approach

Choice of Chemotherapy

FLYER trial (Poeschel, 2019)
	○ 592 patients with stage I-II non-bulky DLBCL (favourable IPI score) were randomised to
		§ 4 vs 6 cycles of R-CHOP
		§ Non-inferiority design
	○ Outcomes
		§ 4 cycles was non-inferior to 6 cycles
			□ 3 year PFS 96% (4x) vs 94% (6x) No difference in 3-year OS (99% vs 98%)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Describe the evidence for management of Bulky DLBCL

A

○ RICOVER-noRTh (Held, 2014) Radiation de-escalation is not ok for bulky
§ Patients on the preferred arm (6x R-CHOP with 36Gy RT for bulky disease) were compared with a prospective cohort where RT was not allowed
□ Neither arm had RT for nonbulky disease (RT only if bulky disease)
§ Outcomes
□ On multivariate analysis, RT for bulky disease was associated with improved 3yr PFS and OS
○ 3yr OS 65 -> 90% (HR 4.3; p=0.002)
○ 3yr PFS 62 -> 88% (HR 4.4; p=0.001)
□ RT abrogates the prognostic impact of disease bulk (should be standard of care)

UNFOLDER (Pfreundschuh, 2018)
	○ Patients with stage I-II favourable IPI DLBCL were randomised to
		§ 6x R-CHOP-14 vs 6x R-CHOP-21
		§ Consolidation RT for bulk vs observation
	○ Trial closed early due to significant benefit in the RT arm
	○ Outcomes
		§ RT was associated with improved 3-year EFS (84% vs 68%)
			□ No difference in 3yr OS
		§ No difference in chemotherapy arms (EFS, PFS, OS)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Describe the evidence for RT Dose in NHL

A

UK-based randomised phase III trial (Lowry, 2011)
○ RT dose recommended for indolent NHL is 24Gy and 30Gy in aggressive NHL.
○ 161 patients with indolent NHL and 640 patients with aggressive NHL
§ Indolent = 24Gy/12F vs 40-45Gy
§ Aggressive = 30Gy/15F vs 40-45Gy
○ Outcomes
§ No difference in ORR in either group
□ Indolent (93% vs 92%)
□ Aggressive (91% vs 91%)
§ No difference in in-field progression
□ Indolent (HR 1.09)
□ Aggressive (HR 0.98)
§ No difference in PFS or OS

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

Describe the evidence for management of advanced stage DLBCL

A

MDACC retrospective study (Phan, 2010)
- Of 279 patients with advanced stage DLBCL, 39 had bulky disease
○ 23 of these received RT
- Outcomes
○ RT was associated with improved OS and PFS

RICOVER-60 - any stage
§ Outcomes
□ 8 cycles of chemotherapy was not superior to 6 cycles for OS or PFS
□ Addition of rituximab to 6x CHOP improved OS (HR 0.63) and PFS (HR 0.50)
□ No major differences between 6x R-CHOP vs 8x R-CHOP

RICOVER-noRTh trials - any stage
□ On multivariate analysis, RT for bulky disease was associated with improved PFS and OS
□ RT abrogates the prognostic impact of disease bulk (should be standard of care)

Freeman 2021- Risk-adapted era supporting using PETCT to select people who need Consolidative RT
- 723 pt, Retrospective, PETCT guided approach. 6-8x RCHOP -> Omit RT if PETCT neg vs ISRT if PETCT pos.
- PETCT Neg => No RT 3yr TTP 83%, 3yr OS 87%
- PETCT Pos => RT 3yr TTP 76% 3yr OS 80%
○ Note: Even PETCT Neg Bulky >10cm had similar outcome to those without bulk suggesting that EOT PETCT can reliably guide selective administration of consolidative RT

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

Describe the evidence for management of relapsed DLBCL

A

CAR-T cells
ZUMA-1 trial phase II single arm multi centre trial
-Axi-cell autologus anti-CD19 CAR-T cell therapy
-Demonstrated durable response in patients with refraction DLBCL, 5 year follow up 30% of patients achieved durable response

- Polatuzumab vedotin (pola), an anti-CD79b antibody drug conjugate (ADC) linked with monomethyl auristatin E (MMAE), in combination with rituximab, cyclophosphamide, doxorubicin, and prednisone (Pola-R-CHP), essentially replacing vincristine in the CHOP backbone
- Pola-R-CHP superior to R-CHOP in adult patients with newly diagnosed DLBCL with IPI >2 (PFS) 
- Toxicity ad no OS benefit 
- ?Emerging new front-line management instead of R-CHOP for DLBCL, trials emerging
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What are the indication for radiotherapy in DLBCL?

A

Indications for Radiotherapy
* first-line management:
○ chemo minimisation strategy in LS-DLBCL
§ consider R-CHOPx4c →ISRT (SWOG 8736)
§ of value to elderly pt, where longer courses of chemo associated with ↑ risk of myelosuppression, infections, treatment related mortality (& late effects less relevant)
○ consolidation therapy in sites with higher risk of local relapse
§ initial bula ≥ 7.5cm (RICOVER-60-no-RT)
§ non-bulky initial SUV ≥15 (5yr LRFS 100→69%)
§ residual mass. >2cm on CT despite PET EOT neg
§ no RCT data to quantify benefit of consolidation RT (and unlikely, as pt in non-RT arms of UNFOLDER closed early due to higher event rates in pt not assigned to RT
○ conversion of incomplete metabolic responses after initial immunochemotherapy to complete response
§ nuclear how to manage pt who are PET EOT negative
§ no prospective evidence to omit RT in pt with initial bulky and/or extra lymphatic disease

Aggressive NHL dose considerations
* Upfront DLBCL after chemo: 30-40 Gy. Give 30 Gy if DS 1-3. Boost to higher doses if DS4.
* Upfront double-hit DLBCL: Correct dose is unknown, but may lean toward the higher end of the dose spectrum when consolidating.
* Upfront PMBCL:
○ Avoid RT if given DA-R-EPOCH if possible.
○ After R-CHOP x6, given 30-40 Gy depending on PET response.
* Relapsed/refractory DLBCL:
○ Localized disease, Bulky > 5 cm [PARMA], incomplete response to salvage chemo or ASCT, skeletal involvement, critical sites where LC is important.
○ Cytoreduction prior to ASCT: 40-50 Gy or higher if refractory.
○ DS1-3 with salvage chemo and ASCT: 30-36 Gy. Residual avidity: 40-50 Gy.
○ Transplant ineligible, curative intent: 45-55 Gy.
○ Palliative intent with limited life expectancy: hypofractionated schedule of 8-30 Gy.

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

Discuss the management of breast lymphoma

A
  • Epidemiology: Rare (1% of extranodal lymphomas), Assoc w Breast feeding and pregnancy
    • Histology:
      • 50% DLBCL
      • Follicular lymphoma
      • Marginal zone lymphoma
    • Biologic behaviour: 10-30% of breast DLBCL have CNS involvement
    • Treatment:
      • Aggressive
        w R-CHOPx6 + RT 30Gy/15#
        w Consider CNS prophylaxis
      • Indolent
        w IFRT 24Gy/12#
      • Mastectomy not recommended
    • RT technique
      • CTV = whole breast (uninvolved LNs need not be included)
      • Dose: 30Gy/15F
      • Technique as per breast carcinoma
    • Outcomes
      • Good LC with RT
      • 5yr OS 50%
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

Describe your radiotherapy technique for DLBCL

A

Involved Site Radiotherapy
Patients
1. Stage I-II disease (chemo-de-escalation)
2. Stage I-II disease (bulky)
3. Stage III-IV disease (bulky)

Pre-simulation
MDT discussion
Fertility consideration
- bHCG in all females
- Fertility preservation
Smoking cessation

Simulation
Supine with arms above head
Vacbag
DIBH if superior mediastinum
Generous CT (2mm with contrast)

Fusion
Pre-chemotherapy PET

Dose prescription
Single dose-level
- 30Gy/15F (if chemo de-escalation)
- 36Gy/18F (if bulky)
- 40Gy/20F (if FDG-avid residual disease)
VMAT technique
10 days per fortnight

Volumes
GTVpre
- Gross disease on pre-chemo CT and PET
GTVres
- Gross residual disease on post-chemo PET
CTV
- Use GTV volume to establish superior/inferior margins
○ Consider extra 1-2cm superior/inferior to account for imaging uncertainty
- Use current CT to delineate radial margins
ITV
- Consider if mediastinal disease
PTV
- CTV + 7mm

Target Verification
Daily CBCT

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

Discuss the work up and management of ocular lymphoma

A
  • Assoc with poor prognosis
    • 2 patterns of intra-ocular lymphoma
      ○ Commonly assoc with CNS involvement:
      □ Optic n, retina, vitreous; usually high grade
      □ 20% PCNSL have ocular involvement at diagnosis
      □ 80% of ocular lymphomas will develop CNS lymphoma (latency ~1y)
      □ Present with blurred vision or floaters (identified by slit-lamp examination)
      □ Most pts eventually develop bilat dx
      □ Dx: lymphoma cells in vitreous sample
      ○ Lymphoma of the uveal tract:
      § Commonly assoc with visceral involvement; choroids, ciliary body, iris
      § Bilat dx common, although Sx usually affect 1 eye more
      § Usually DLBCL
      § DDx: inflammatory syndrome: uveitis, conjunctivitis, glaucoma
    • Biologic behaviour
      • Brain relapse was universal in older series, however recent data suggest many patients do not relapse in the brain after eye-directed therapy alone and if they do, salvage options remain effective
      • Majority present with pathologic or clinical suspicion of bilateral ocular involvement
        w Although most series have treated both eyes, if there is no suspicious of disease in the contralateral eye, it is reasonable to treat only the involved eye
    • Workup:
      • Full ophthalmologic review
      • Brain/orbital MRI
      • Ocular coherence tomography and US- to assess extent and nature of lesion
      • Biopsy- vitrectomy bx or cytology/ flow cytometry of the vitrous fluid or tissue bx
      • Lumbar puncture and CSF evaluation for cytology and raised prot levels.
    • Optimal Rx approach not well-defined
    • Management
      • The globe is not well treated by systemic therapy
      • Options include
        w Definitive RT alone
        w Intra-ocular methotrexate
        w Rituximab- either sytemically or intravitreally injected
        w Steroids to reduce inflammation and oedema
    • RT technique
      • CTV: Globe of the eye(s), optic nerve(s) to the level of the chiasm
      • PTV: 5mm expansion
      • Technique:
        w If both eyes are involved (or there is suspicion of bilateral involvement) an opposed lateral beam arrangement should include both globes and the optic nerves to the chiasm. The isocentre should be set at the posterior border to reduce divergence in case of need for subsequent WBRT.
        w If only one eye is involved, the field is limited to the involved eye using conformal RT or IMRT, or wedge pair
      • Dose: 36Gy/20F
    • Outcome:
      • Rx results poor: high local failure, RT provides temporary palliation, MS 6-18m
    • For relapse: intra-vitreal MTX
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Describe the epidemiology, biological behaviour and management of primary testicular lymphoma

A
  • Special considerations: High risk CNS site
    • General management: R-CHOP, IT MTX + RT to contralateral testes
    • Epidemiology
      • I:
        w 1-7% of all testicular cancer
        w Most common testicular tumour in >60 yo
      • A: Mostly elderly men >60 years
      • S: Men
    • Pathology
      • 80-90% of all primary testicular lymphomas are DLBCL, the rest are either plasmacytic, Burkitt’s, mantle cell lymphoma, LG follicular lymphoma or T-cell lymphoma.
      • Has altered surface adhesion molecules- hence the propensity to disseminate to extranodal sites
      • Majority ABC pattern.
      • Common mutation MYD88
    • Biologic behaviour
      • Majority present with st I/II disease, IPI 0-2, no B sx
      • Tendency to disseminate to contralateral testis and CNS (meningeal or parenchymal) → 40-50%
      • Poor prognosis (even if stage I) cf. DLBCL at other sites
      • Usually unilateral, but 10% can have bilateral involvement
      • Aggressive behaviours- spread to multiple extranodal sites: lung, pleura, skin, soft tissue and waldeyer’s ring
    • Prognosis
      • median PFS 4 years
      • 5 yr OS ~50% (all stages)
      • Median OS 5 years
      • Failures usually within 1-3 years of initial Rx, survival after failure is short
      • Extranodal recurrence in ~70% (+/- nodal disease)
      • Most common sites of failure
        w Contralateral testis (30%)
        w CNS (30%)
        w Other less common sites: bone, skin, BM, lung, soft tissue, adrenals, liver, GIT + spleen
    • Refractory/Relapsed- no std therapy defined.-
      > HDCT + ASCT. clinical trials
    • RT technique:
      • Sim: be present during sim to confirm position and setup, Frog-leg, penis lifted and taped to abdominal wall, scrotum supported and immobilised with bolus under and around the scrotum
      • Dose: 30Gy/15F
      • Target volume
        w Ipsilateral (if not removed by surgery) and contralateral testis
        Technique: Anterior electron field with energy calculated according to thickness of scrotum/testis. Bolus may be required
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

Discuss the work up and management of Osteolymphoma

A
  • Epidemiology: Rare; median age 60yrs
    • Prognosis: 5y OS 50%, 80% LC.
    • Histology: Majority DLBCL (90%)
    • Location: Any bone, usually diaphysis (central portion), often multifocal dx.
    • Investigations
      • Bloods, bx, imaging incl MRI, PET, BMAT
      • Large proportion pts have dx elsewhere, therefore Ix for other sites of involvement
    • Natural History:
      • Majority present with stage IE disease
      • ~10% have polyostotic presentation (multiple bone)
        w Most common bones involved: femurs, pelvic bones, tibia and fibula
      • Often recur at multiple sites, exclusively bone (i.e. different disease to systemic lymphoma)
    • Good prognostic factors: age <60, Nodes, LDH, ECOG 0, mono-ostotic dx
    • RT technique:
      • Volume
        w CTV: Prechemotherapy GTV (preferably on MRI) with margins to accommodate uncertainties in subclinical tumour extension and quality of imaging and fusion
        w PTV = CTV + 0.5-1cm depending on site and immobilisation
      • Dose:
        w After chemo, complete regression of PET activity may not be clear at time of RT
        w 30-40Gy depending on certainty that a CR has been achieved with chemotherapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

In general what is the management of extra-nodal DLBCL

A

30-40% of DLBCL present as extranodal DLBCL with no nodal involvement

[ILROG]
Volume:
* Whole organ/ Compartment irradiation is the most widely reported approach for many sites (eg, stomach, thyroid, breast) and is mandatory for sites in which disease is usually multifocal (eg, primary CNS lymphoma).
* Partial organ irradiation is appropriate for unifocal DLBCL involving long bones when disease extent is well defined on baseline PET and MRI
* For DLBCL of lung and skin, RT is confined to the tissue volume involved before systemic therapy.
Dose: 30-36Gy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What is the prognosis for DLBCL?

A

Components of IPI score
- Age > 60 years
- ECOG 2+
- Lugano Stage III - IV
- 2+ extranodal sites
- Elevated LDH

CNS Internal Prognostic Index

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

Describe the epidemiology for follicular lymphoma.

A

Incidence (Australian statistics)
- 1500 cases annually
- Second most common variant of non-Hodgkin Lymphoma (25%)

Slight female predominance (1:1.5)
Median age 60-65 years

Aetiology

No strong aetiological causes known

Family history
- First degree relatives

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

Describe your follow up for a patient with DLBCL.

A

Follow-Up

* Clinical review every three months for the first two years (shared care)
	○ Clinical examination
	○ FBC, LDH at each visit
* Clinical review every six months for years 3-5
	○ Clinical examination
	○ FBC, LDH at each visit
* Annual surveillance thereafter for late effects

* Single FDG-PET at 12 weeks post RT
* Consider routine CT every 6-12 months for the first two years
	○ Minimise, particularly in the young

* Consider annual TFTs if neck irradiated
* Consider TTE and carotid US
* Consider early breast screening
	○ 10 years after RT
	○ Age 40 years
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Describe the pathology for follicular lymphoma.

A

Macroscopic
- Variable degree of effacement of normal nodal structure
- Retention of follicular/nodular pattern with some disordering
○ Higher-grade disease may have a more diffuse pattern

Microscopic
- Follicular proliferation and expansion within the LN: uniformly appearing, numerous, closely packed follicles
- Two cell types are present = small and large cells proliferate
○ Centrocytes (small cells with scant cytoplasm and cleaved nuclear contours)
▪ (Most cells)
○ Centroblasts (larger cells with more cytoplasm and open nuclear chromatin)
○ Instead of combination of B cells (CD20+) and some T cells (CD3+), follicles are composed of mostly B cell positive for BCL-2
- Two types of pattern
○ Nodular (lower-grade)
Diffuse (higher-grade) –> loss of normal follicular dendritic cell meshwork

Immunohistochemistry
- POS = CD19/CD20/CD79a (B-cell markers), CD10/BCL6 (germinal centre), BCL2
- NEG = CD3/CD4/CD8 (T-cell markers)

Molecular
- t(14:18) –> IGH/BCL2 translocation
BCL6 rearrangement can be seen in grade 3a/3b disease

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

Describe the pathogenesis of follicular lymphoma

A

Originates from the follicular cells in the germinal centre of the LN
Varying proportions of centrocytes and centroblasts with at least partial follicular growth pattern.

Pathogenesis
Strongly associated with BCL2 translocations (BCL2 = anti-apoptotic protein)
Characteristic translocation is the t(14:18)
- IGH and BCL2
BCL2 is an anti-apoptotic gene –> upregulation
- Very few apoptotic bodies are seen

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

List the WHO 5th edition subtypes of follicular lymphoma

A

NEW WHO 5th edition 2023:
- Classic follicular lymphoma
○ Gr 1,2,3a –grading by counting centroblasts is unreliable and treated the same
- Follicular large B (was grade 3B) -treat like DLBCL
- Predominatnly diffuse FL
- FL with unusual cytological feature (large centrocytes, high KI67)
○ Subavriants; blastoid, large centrocyte
○ Favourable prognosis
○ Diffuse growth, CD23, 10 pos.
- Other
- In situ follicular B-cell neoplasm
Paediatric-type follicular lymphoma
Duodenal-type follicular lymphoma
Primary cutaneous follicle center lymphoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Discuss the natural history of follicular lymphoma.

A

Early follicular lymphoma is incurable but has an indolent long waxing and waning natural history with good prognosis, 10Yr PFS 80-90%
In 10-20% of pt, it can spontanenously regress in some patient without needing treatment
- Upfront RT can improve PFS and OS compared to observation.
- It also decreases the risk of malignant transformation
Very radiosensitive and chemosensitive, after RT, LC >90% with <5% infield recurrence. Most of the recurrence are out of field (75%) and 25% are DM+.

In advanced stages, it natural disease is also variable with 50% have DM+ at dx. There is 2-5%/yr risk of transformation to DLBCL. 30% of pt transform into DLBCL. Transformed DLBCL from follicular lymphoma are more aggressive than primary DLBCL.

Upfront treatment is depending on the modified GELF criteria.

Richters transformation: usually DLBCL, but also Burkitt’s
Path feat: Loss of follicular architecture and accumulation of large tumour cells resembling aggressive NHL cells with increasing centroblast and high ki67

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What are the prognostic factors for Follicular lymphoma?

A

Patient Factors
- Age (>60 years)
- Performance status
- Blood results
○ Hb level (<120g/L)
○ Serum LDH level (above ULN)
- Symptomatology

Tumour Factors
- Lugano staging
○ Bone marrow involvement
- Number of nodal regions involved (>4)
- Serum beta-2 microglobulin
- High Ki67

Treatment Factors
- Contraindications/ access to treatment

FLIPI was developed for overall survival and established in the pre-rituximab era. In contrast, FLIPI-2 was designed for progression-free survival, and, in its development cohort, more than one-half of patients had been treated with rituximab-containing regimens.

“BBASH 6”
B2 Microglobulin high
BM involved
Age >60
Size >6cm
Hb <120

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Discuss the presentation and history for a patient with follicular lymphoma.

A

Presentation
- Subacute, or chronic asymptomatic peripheral lymphadenopathy
- Indolent: may persist, or wax and wane over years
- Abdominal, pelvic or retroperitoneal LN can be bulky without symptoms
- Nodal masses not locally invasive and destructive
- LN invovlement not in an orderly manner
- Early haematogenous dissemenation is common
- B symptoms in less than 20% (if present should consider transformation)

FL is mainly a nodal disease, but involvement of the spleen and bone marrow is frequently encountered.

Consultation

- History
	○ HPI
		§ Palpable lymphadenopathy (non-tender)
			□ Duration and tempo of disease
			□ Commonly waxes and wanes
		§ Splenomegaly or hepatomegaly
		§ Constitutional symptoms
			□ Fever
			□ Night sweats (drenching)
			□ Weight loss (10% within 6mo)
	○ PMHx:
		§ Previous radiotherapy or chemotherapy
		§ Immunosuppression (including HIV)
	○ Medications
		§ Immunosuppression
	○ Social:
		§ Smoking
	○ Family History
		§ Lymphomas

- Examination
	○ General inspection
	○ Lymphatic examination
		§ Cervical
		§ Axillary
		§ Epitrochlear
		§ Inguinal
		§ Popliteal
	○ Abdominal palpation
		§ Hepatomegaly
		§ Splenomegaly
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

How would you work up a patient with follicular lymphoma?

A

Work-Up

- Bloods
	○ FBC + ESR (prognostic)
	○ EUC, CMP and LFT
	○ LDH
	○ HepB/HepC/HIV serology (pre-chemotherapy)
	○ B2 microglobulin

- Histopathology
	○ Excisional biopsy referred
		§ Large amounts of tissue may be required 
	○ Core biopsy is a reasonable alternative
		§ FNAB is insufficient

- FDG-PET (staging)

- Routine bone marrow biopsy IS required to confirm limited stage (if PET negative)
	○ 85% have bone marrow involvement at diagnosis
	○ FDG-PET alone has insufficient sensitivity

- Only if considering chemotherapy:
	○ Cardiac TTE
		§ LVEF prior to anthracycline chemotherapy
	○ Lung function tests
		§ Prior to bleomycin
		§ Prior to radiotherapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

Describe the management of Stage I and Stage II Follicular lymphoma.

A

Stage I

- Therapeutic options include:
	○ Curative radiotherapy (Generous ISRT as no chemo planned)
	○ Observation alone
		▪ Watchful waiting for G1-2, stage I/II FL may be appropriate in some patients, as ~50% of patients will avoid treatment at 15y. However, the rate of transformation to DLBCL can be as much as 20% (decreased to < 5% with RT)
			* Guidelines do not recommend this approach

- Low-dose radiotherapy is associated with high-rates of durable control
	○ 24Gy/12F (or 30Gy/15F)
		§ Modified ISRT (or IFRT)
		§ Generous volumes to incorporate some surrounding lymphoid tissue (not getting chemo for microscopic disease)
	○ 36Gy/20# for bulky sites >5cm
	○ Outcomes
		§ Local control 90%, but up to 40% relapse out of field, (25% in PET era)
		§ 10 year relapse-free survival of 50%
		§ Median OS >15 years

Stage II

- Therapeutic options can be as per Stage I or stages III-IV
	○ Observation
	○ Definitive Radiotherapy
	○ Systemic therapy

- Only consider RT if disease can be safely and easily encompassed by one radiotherapy field
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

In general what factors should be taken into account when managing follicular lymphoma?

A

Any treatment decisions should consider the overall context
- Age and performance status
- Current burden of disease
- Current symptomatology
- Kinetics of disease

True cure is rarely feasible (even in stage I disease)
- However, long-term durable control is often achieved

All grade 3b patients should be managed as per DLBCL (high-grade)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

Discuss the management for Stage III/IV follicular lymphoma.

A

Stage III and IV

- Cure cannot be achieved, regardless of treatment approach

- Upfront therapy does not result in:
	○ Improved OS
	○ Reduction in risk of transformation to high-grade disease
	○ RCT indolent FL 
		§ local control 24Gy/12F  =90-98% 
				 vs 4Gy/2F  = 80-96%
		Also included marginal zone lymphoma 50-70% complete response
	○ Therefore, there is no rationale for routine upfront therapy

- Most patients should be observed in the first instance
	○ 10-20% of cases have spontaneous regression
- Indications for upfront chemotherapy(GELF criteria)
		§ L337 BBEAMSS
	○ LDH Elevated or beta-2-microglobulin
	○ 3 areas >3cm (extensive nodal disease)
	○ Bulky >7cm
	○ 
	○ B symptoms
	○ Elevated lymphocytes>5
	○ Asctes/effusion
	○ Marrow failure or neut <1, platelets<100
	○ Symptoms -compression
	○ Splenic symptoms

- Active treatment options:
	○ Chemotherapy -  improve RR and PFS, but no OS benefit
		§ Single agent Rituximab
		§ Rituximab + bendamustine
		§ R-CHOP (high-grade disease)
	○ Chemotherapy + Rituximab - improve RR, PFS and OS cf chemo alone
	○ Myeloablatic Tx and ASCT -  prolong PFS after Chemo, but no OS benefit
	○ Radioimmunotherapy (Zevalin Y-90 attached to CD20 antibody)- prolong PFS after chemo, no OS benefit
- Rituximab maintenance every 2 months for 2 years is recommended after immunochemotherapy- improve mPFS 10 vs 4yr
- Palliative RT: 70-90% (4-24gy) local control @ 5 years
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

Discuss the management of relapsed follicular lymphoma.

A

○ If relapse/ progression> strongly recommended to repeat bx to exclude transformation to an aggressive lymphoma (Richters transformation)

○ Adverse prognostic factors: B sx, bulky dx, >2 relapses, LDH >400, Hb <120
○ Median OS:
* 0 factor- >6yrs
* 1 factor- 2yrs
* 2+ factors- 8.5months
○ Treatment options
* No adverse factors: watch+ wait, CT, Rituximab
* 1+ factor depends on age
election of salvage treatment is dependent on previous efficacy and duration of response- prefer non cross-resistant regimen eg. Bendamustine after CHOP, or vice versa
* IF low disease burden-> rituximab alone can be considered
* IF short remission after Chemo-> then lenalidomide+ Rituximab
* Radioimmunotherapy could be considered in elderlies who are not fit for chemo
* consider HDCT + ASCT if early relapse; consider allo SCT if late relapse
* IF rituximab duration <6mo-> then give obinutuzumab-bendamustine

	○ Proportion + duration of response ↓with each episode of treatment.
	○ Radioimmunotherapy (Zevalin/Bexxar) recently showing promise. CR 60-80% in relapsed/refractory dx.
	○ Palliative RT- for symptomatic sites
		* RR 80-90% with low dose 4Gy/2F. 
		* Well tolerated, can give high dose XRT or subsequent CT if needed. 
		* NB: RCT (MRC) shows inferior RR and LC with 4Gy than with 24Gy so if durable response required, give higher dose.
 
○ Maintenance can prolong PFS and OS in relapsed disease. Rituximab every 3 mo for 2 years.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

Describe transformed follicular lymphoma and its management.

A
  • Up to 70% transform to higher grade lymphoma during course of dx.
    • Histological transformation of FL occurs approx. 1-2% per year, and is assoc w/ rapid progression of lymphadenopathy, infiltration of extranodal sites, development of systemic symptoms, elevated serum lactate dehydrogenase, hypercalcemia, and often a poor prognosis.
      ○ In most cases, the pathology is consistent with DLBCL. Less commonly, FL evolves into Burkitt lymphoma, high grade B cell lymphoma with MYC and BCL-2 and/or BCL-6 rearrangements (“double-hit” lymphoma), or into precursor lymphoid neoplasms (blastic/blastoid transformation). HT includes FL that evolves to grade 3b FL
      • Clinical feat:
        ○ Rapid progression of lymphadenopathy
        ○ Invasion of new unusual extranodal site- esp. CNS, pleura, bone, liver mass.
        ○ B-Symptoms out of proportion to dx extent:
        ○ ↑ LDH (esp. sudden. marked), ↑ Ca. fever, night sweat, WT loss
        ○ Early treatment failure ( 2 years)
        ○ PET >SUV10
        ○ Advanced Stage
      • Path feat: Loss of follicular architecture and accumulation of large tumour cells resembling aggressive NHL cells with increasing centroblast and high ki67
      • Poor prognosis post-transformation: mOS 1-2y
        ○ possibly improved in cohort of patients who had never been exposed to systemic therapy prior to transformation
      • Treatment: no data from randomised trials to support optimal treatment for these patients as they are often excluded from clinical trials
        ○ Consider Rituximab monotherapy or combination with antracycline-based chemo eg. R-CHOP
        * Retrospective reviews show an improved OS with anthrax-based chemo + Rituximab
        ○ Little impact of anthracycline based CT; possible role of autologous SCT
        ○ High grade follicular lymphoma: (G3)
        * Stage 1-2:
        ▪ Manage as per DLBCL
        * 3-6x RCHOP + ISRT 30Gy/15# for CR
        * Stage 3-4:
        ▪ Palliation
        ▪ RT: as per boom-boom 4Gy/2#
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

Discuss paediatric follicular lymphoma and its management

A
  • FL variant originally described in children, but rarely occurs in adults.
    • Characterised by typically
      ○ localised disease
      ○ absence of BCL2 aberrations
      ○ lack of t(14;18)
      ○ grade III histolog, and
      ○ A high proliferation rate.
      It shows a much more indolent disease course and can be managed with local therapy only, despite displaying histologically more aggressive features.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What is the evidence for managing early stage follicular lymphoma.

A

Summary:
- Radiotherapy is associated with OS benefit in early-stage FL
Addition of chemotherapy does not improve OS in this group
There is a PFS advantage however

TROG 99.03 trial (MacManus, 2018)
○ 150 patients with stage I-II low-grade follicular lymphoma were randomised to
§ 30Gy IFRT alone
§ IFRT + 6x CVP chemotherapy (rituximab added during trial)
○ Outcomes
§ 10 year PFS was superior in the chemo arm (59% vs 41%)
§ No difference in 10 year OS (95% vs 87%; p=0.40)
§ Updated data presented in ESTRO 2023 (MFU 11.3yrs)- systemic therapy esp with Rituximab increased PFS and sig reduced risk of death/transformation to aggressive lymphoma (most benefit in PET staged pt). No OS benefit

NCDB analysis (Vargo, 2015)
	○ Retrospective analysis of 35961 patients with stage I-II and grade 1-2 follicular lymphoma
		§ 63% were stage I
	○ Outcomes
		§ RT use declined from 37% to 24% between 1999 and 2012
		§ RT was associated with improved OS (10 year OS 68% vs 54%)
			□ This association remained independently valid on multivariate analysis
70
Q

Discuss the evidence for managing advanced follicular lymphoma.

A

Summary:
- There is no OS advantage to upfront therapy in asymptomatic advanced-stage FL
○ PFS and FFTF benefits do exist however
- There is some variability in the literature regarding risk of transformation
○ Most likely, no advantage with upfront therapy

UK-based randomised phase III trial (Ardeshna, 2014)
	○ 379 patients with asymptomatic advanced stage follicular lymphoma were randomised to
		§ Surveillance
		§ 4 weeks of rituximab (induction)
		§ Induction rituximab + maintenance for 2 years
	○ Outcomes
		§ Both rituximab arms were associated with longer PFS and time to second-line therapy
		§ Rituximab was associated with improved psychological measures
			□ No difference in overall QoL
		§ No difference in OS
		§ No difference in rate of transformation

French retrospective database analysis (Solal-Celigny, 2012)
	○ 120 patients from a larger 1093 patient cohort were incorporated
		§ Patients registered on the database without any upfront therapy (80% had disseminated disease)
		§ Matched to a cohort of 242 patients who received upfront rituximab
	○ Outcomes
		§ After 5 years of follow-up, 50% of the cohort had commenced therapy
		§ Non-significant trend to improved FFTF in a group with upfront rituximab (HR 0.63; p=0.103)
		§ No difference in rates of transformation
		§ No difference in OS at 5 years (88% vs 87%)

Mayo Clinic retrospective analysis (Link, 2013)
	○ 631 patients with follicular lymphoma were included
	○ Outcomes
		§ Overall transformation rate of 10.7% over 5 years (2% per year)
		§ Transformation was highest in those observed compared with rituximab monotherapy (14.4% vs 3.2%; p=0.021)
70
Q

Discuss the evidence for the radiotherapy dose used in follicular lymphoma.

A

Summary:
- In the definitive setting, 24Gy/12F is optimal in indolent NHL. Use 30Gy/15F for Aggressive NHL
○ No benefit to escalation to 40Gy
○ 4Gy cannot be declared non-inferior
- 4Gy/2F is a reasonable palliation dose (“boom-boom”)

UK-based randomised phase III trial (Lowry, 2011)
	○ 161 patients with indolent NHL and 640 patients with aggressive NHL
		§ Indolent = 24Gy/12F vs 40-45Gy
		§ Aggressive = 30Gy/15F vs 40-45Gy
	○ Outcomes
		§ No difference in ORR in either group
			□ Indolent (93% vs 92%)
			□ Aggressive (91% vs 91%)
		§ No difference in in-field progression
			□ Indolent (HR 1.09)
			□ Aggressive (HR 0.98)
		§ No difference in PFS or OS

	Russo (IJROBP 2013): 187 sites / 127 pts. Retrospective, single institution " Boom Boom" 4Gy/2F
		○ Highly effective for palliation of advanced stage, relapsing or even most chemo-refractory patients with indolent NHL
		○ Overall response - 82% (Complete response 57%, partial response 25%)
		○ CLL histology associated with a lower response rate
		○ Tumour size, site, age at diagnosis and prior systemic therapy was not assoc with response
		○ Median time to recurrence was 13.6 months

FORT RCT (Hoskin, 2021)
	○ 614 targets in 548 patients with follicular lymphoma were randomised to
		§ 24Gy/12F vs 4Gy/2F
		§ Non-inferiority design
	○ Outcomes
		§ 4Gy inferior
			□ Time to progression was shorter in the 4Gy group (HR 3.42)
			□ 4Gy associated with worse 5 year PFS (89.9% vs 70.4%)
		§ 4Gy/2F achieved 50% CR and a further 31% PR rate
			□ Good for palliation, or if patient getting chemo
71
Q

Describe the radiotherapy technique for early stage follicular lymphoma.

A

Involved Site Radiotherapy
Patients
1. Stage I disease
2. Stage II disease (consider)

Pre-simulation
MDT discussion
Fertility consideration
- bHCG in all females
- Fertility preservation
Smoking cessation

Simulation
Supine with arms above head
Vacbag
DIBH if superior mediastinum
Generous CT (2mm with contrast)

Fusion
Pre-chemotherapy PET

Dose prescription
Single dose-level
- 24Gy/12F prescribed to PTV
VMAT technique
10 days per fortnight

Volumes
GTV
- Gross disease on CT and PET
CTV
- Use GTV volume to establish superior/inferior margins
○ Consider extra 2-3cm superior/inferior
- Extend radially to incorporate entire nodal compartment
- Incorporate any suspicious adjacent nodes
ITV
- Consider if mediastinal disease
PTV
- CTV + 7mm

Target Verification
Daily CBCT

72
Q

Describe the prognosis and follow up for follicular lymphoma.

A

Prognosis

Stage I disease
- 10 year relapse-free survival > 50%
- 10 year OS > 80%

Jb/Lm cure rate 70-80%
LC - 80-90%
Peter Hoskins studies

Prognosis for Stage III and IV disease
- As per FLIPI score

Risk of transformation to DLBCL
- Approximately 30% overall
- Estimated at 2% per year

Follow-Up

* Clinical review every three months for the first two years (shared care)
	○ Clinical examination
	○ FBC, LDH at each visit
* Clinical review every six months for years 3-5
	○ Clinical examination
	○ FBC, LDH at each visit
* Annual surveillance thereafter for late effects
	○ Long-term follow-up required

* Single FDG-PET at 12 weeks post RT
* Consider routine CT every 6-12 months for the first two years
	○ Minimise, particularly in the young

* Consider annual TFTs if neck irradiated
* Consider TTE and carotid US
* Consider early breast screening
	○ 10 years after RT
	○ Age 40 years
73
Q

Discuss the epidemiology of MALT

A

MALT (Mucosa-associated lymphoid tissue) (part of Marginal Zone):
* Clinical features of MALT lymphoma
○ Low-grade NHL, median age 60yrs, relatively uncommon
○ 15-20% of lymphomas arise at extra-nodal sites, most commonly GIT
§ Common: stomach, orbit, salivary glands, lung + skin.
§ Less common: GI mucosa, thyroid, breast, bladder, liver.
○ Assocn of gastric dx with other sites, so non-gastric site pt should have endoscopy.
○ 70-90% have Stage I-II dx
○ High CR rate + long survival, 80% at 10yrs.
○ Tendency to remain localized to tissue of origin for a long time but frequently recurs locally and potential for systemic spread and transformation to aggressive B cell lymphoma. 5-10% transformation risk to DLBCL. Gastric subtype tends to be more localized
○ Uncommon to have B sx or LDH rise.
○ Peripheral blood is usually not involved
○ Can spread to nodes or marrow in about ~30% cases
○ Present with non-specific sx related to site
§ Gastric- epigastric pain, dyspepsia, N+V, anorexia, weight loss.
§ Ocular: growing mass, eye redness, excessive eye watering
§ Salivary gland: slow growing mass, minority have bilateral involvement
§ Skin: red papules, plaques, nodules on trunk or upper extremities
§ Intestine: diarrhea, cramping pain, malabsorption
○ Increasing evidence suggests caused by chronic antigen stimulation due to infection or Autoimmune dx
§ H.pylori - stomach
§ Chlamydia psittaci - orbital
§ Campylobacter – small bowel
§ Sjogren’s Syndrome - salivary glands
§ Hashimoto’s thyroiditis - thyroid
§ Borrelia spirochete of Lyme dx – skin
§ Hepatitis C viral infection
○ Associated with autoimmune diseases:
w Sjogrens, SLE, Hashimotos

73
Q

Discuss the pathology for marginal zone lymphoma and its three subtypes.

A
  • Normal mucosal tissue contains non-continuously spread collections of lymphocytes called mucosal-associated lymhoid tissue
    • Lymphoma arising from the marginal zone of such collections = MALT lymphoma
    • Generally slow growing, indolent
    • Named because of involvement of marginal zone around lymphoid follicles.
    • Composed of morphologically heterogenous small B-cells including marginal zone (centrocyte-like) cells, cells resembling monocytoid cells, small lymphocytes and scattered immunoblasts and centroblast-like cells
    • The infiltrate is in the marginal zone of the reactive B cell follicles and extends into the interfollicular region
    • There are 3 subtypes of MZL:
      1) Extranodal MZL (MALT)
      Also called low grade B cell lymphoma of mucosa associated lymphoid tissue or MALT lymphoma
      2) Nodal marginal zone B cell lymphoma
      Primary nodal lymphoma with LN infiltration by monocytoid B cells, but without extranodal involvement.
      Rare; Good prognosis
      3) Splenic MZL
      Marked massive splenomegaly with peripheral blood + BM involvement. Usually no adenopathy.
      Splenectomy may result in prolonged remission.
    • Symptoms depending on sites of involvement
      B symptoms rare - consider transformation
74
Q

What is the pathogenesis for MALT?

A
  • Develops in the post-germinal centre memory B cells with capacity to differentiate into marginal zone cells and plasma cells
    • Characteristic lymphoepithelial lesions - infiltration of neoplastic small lymphocytes in epithelial tissue of organ.
      ○ Dense, monotonous population of centrocyte-like cells, often with residual germinal centres and lymphoepithelial lesions
      Chronically inflamed tissue – lies on continuum between reactive lymphoid hyperplasia and full-blown lymphoma. Initially, B-cell clone emerges which is still dependent on antigen-stimulated T-helper cells for growth and survival. Thus, withdrawal of responsible antigen causes tumour involution (eg antibiotics against H pylori in MALToma). Then tumour acquires additional mutations eg (11;18), (14;18) or (1;14) translocations -> upregulated BCL10 and MALT1 ->activation of NFkB and further transformation to antigen-independent growth
75
Q

Describe the pathology of MALT

A
  • Micro:
    ○ Small size lymphocytes
    ○ Architectural effacement by atypical centrocyte-like cells (small cleaved follicular cells with abundant cytoplasm), that infiltrate around reactive B cell follicles in a marginal zone distribution with spread into the interfollicular area
    ○ May invade epithelial structures to form lymphoepithelial lesions; cells are at various stages of B cell differentiation including monocytoid B cells, small lymphocytes, plasma cells, centroblasts and immunoblasts in small numbers
    ○ May have follicular colonization of neoplastic cells resembling follicular lymphoma
    • IHC:
      ○ Positive:
      w CD19, CD20, CD22, CD79a, surface Ig, cytoplasmic Ig in plasma cell component, BCL10
      w Variable CD11c, variable BCL2, variable monoclonal light chain expression
      w Note: plasmacytoid cells may be negative for B cell markers but are strongly immunoreactive for kappa or lambda
      ○ Negative:
      w IgD, CD5, CD10, CD23; usually CD43 and BCL1
    • Molecular/ cytogenetics:
      ○ t(11;18)(q21;q21): API2 and MALT1 (50%): associated with aggressive disease and poor response to antibiotics
      ○ t(14;18)(q32;q21): IgH and MALT1: described in nongastric extranodal MALT lymphomas; indistinguishable from IgH / BCL2 by cytogenetics; associated with other karyotypic abnormalities including various trisomies
      ○ t(1;14)(p22;q32): BCL10 and IgH (% unknown): inactivates the “pro-apoptotic” BCL10 protein; associated with advanced MALT
      ○ t(1;2)(p22;p12): BCL10 and Ig Kappa
      ○ Trisomy 18 is also common; trisomy 3 is present, whole or partial in 26 - 55%
      ○ Multiple lesions often have 1 - 3 neoplastic clones
      ○ Low grade lesions often are BCL6 negative, but have BCL6 mutations by PCR
76
Q

Discuss Non-gastric malt, its treatment and the evidence for this.

A

Non-gastric MALT
* Commonly involves conjunctiva (most common non-gastric MALT), salivary glands, lung, thyroid
* Assoc with autoimmune dx + chronic inflammatory response.
* May have involvement of paired organs (e.g. both parotids)
* Work-up similar to other NHL

Treatment:
* ISRT is an effective initial treatment (NCCN) for early stage or symptomatic extensive stage
○ RT > 95% LC → high cure rates in retrospective series.
* Ritux single agent is an option (NCCN) - ORR of 80%
* Surgical excision: lung, colon, small intestine, breast for early stage disease
* Extensive stage: similar to other low grade lymphomas, treat only with an indication
* Up to 40% relapse → distant or contralat site (e.g. orbit, salivary gland); tendency for relapse at other MALT sites.

Evidence:
* Goda (cancer 2010). Retrospective series, 167 pts with localized MALT treatment with RT +/- chemo (non gastric MALT = 142 pts)
○ RT alone: 10 year RFS 74% and OS 89%
○ Mean dose to non-orbital sites 30Gy, orbits 25Gy
Thyroid and gastric MALT significantly lower risk of distant relapse
Other retrospective series that show good response with local RT

77
Q

Discuss Salivary/ other MALTs and their management

A

Salivary MALT
* Classically association with Sjogrens S, frequently bilateral; median age 50yrs.
* Presentation - painless parotid mass
* Ax: Superficial parotidectomy (FNA insufficient); dental rv + r/o teeth if required
* RT technique
○ Dental rv; rv imaging/path; consent
○ Supine, neck extended, thermoplastic mask immobilization
○ Target volumes: whole parotid (inc. deep lobe)
§ IF Node -ve-> Ipsilat L2
§ IF Node +ve-> Ipsilat L2-4
○ Dose: 24Gy/12F to ICRU ref pt, 3D CRT/IMRT, 6MV photons. Can use a wedge pair technique.
○ OAR- contralat parotid mean <26Gy, lens <8Gy
○ Toxicity
§ Acute- lethargy, erythema, mucositis, odynophagia
Late- xerostomia, dental complications

MALT of other Sites
* Stage IE → RT alone 24Gy. For Waldeyer’s ring, treat adjacent LN
* Low grade thyroid MALT is treated w higher doses – 40 Gy.
* Skin- resection for small lesion. Larger lesions - XRT 85-100% LC. Frequent relapse with new skin lesions, treated with XRT if Symptoms.
Consider surgery in some sites as primary modality, would not require IFRT.

78
Q

Describe the pathology for gastric MALT.

A
  • Dense monotonous lymphoid infiltrate in lamina propria
    • Adjacent mucosa has epithelial erosion, intestinal metaplasia, H pylori, lymphoid follicles, atrophy, atypical regenerative changes, dysplasia
    • Signet ring epithelial cells are present in 1/3 of superficial lamina propria associated with lymphoid areas, may represent lymphoepithelial lesions
    • IHC:
      w Positive: B cells markers (CD19, 20, CD79a) CD43. Monoclonal light chain restriction
      w Negative: CD5 (mantle), BCL6, CD10 (follicular), cyclin D1
    • Molecular/cytogenetics:
      w Gene rearrangement positive, but PCR may be falsely positive as gastritis contains monoclonal B cells
      w T(11;18)(q21;q21) in 6-26% - shown to be assoc with disseminated disease and resistance to abx H Pylori treatment
      w T(14;18)(q32:q21) in 1-5%
      w Trimsoy 3 (60%)
    • Exclude other B cell lymphoma:
      • SLL/XLL: CD5 +ve
      • Mantle cell: CD5 +ve
      • FL: CD10+, CD43 –
      • MYD88: positive in WM/LPL but rarely (still possible) positive in MZL
79
Q

How would your work up and stage a gastric MALT?

A

Evaluation
* Bloods- FBC, UECs, LDH, serum protein electrophoresis
* Exam:
w Look for other sites of MALT lymphoma ie eyes, ears, nose, throat, LN, spleen, skin
* H.pylori breath test
* CT CAP, Ba studies (gastric)
* Bone marrow bx (10% bm infiltration)
* Endoscopy and bx
w Important: multiple sites including duodenum to exclude other involved areas
* Tumour biopsy should be tested for H pylori
w FISH or PCR testing for t(11;18) to identify tumors unlikely to respond to H Pylori treatment
* Infectious agent presence e.g. H.pylori serology.
* Endoscopic USS stomach (look for depth invasion + LN presence).
Staging: (for gastric lymphoma)
* NOT as per Ann Arbor (does not incorporate depth of tumour invasion which is known to affect prognosis)
* Lack of no unifying staging system makes interpreting trials difficult
* Most widely accepted: Lugano staging system

80
Q

Describe the management of H Pylori Positive Stage I-II gastric MALT.

A

Stage 1-2:
H Pylori positive:
* HP eradication therapy with triple tx (PPI + clarithromycin + amoxicillin or metronidazole for those allergic to penicillin)
w No randomized clinical trials comparing diff treatment options in this population
w Prospective single-arm studies and retrospective reviews look at H Pylori tx vs RT vs surgery - H pylori treatment is preferred treatment given low side effects and treatment efficacy (up-to-date) [ 50-80% CR, median time to clear 15mo, 7yr LF 22%]
* Needs 3 monthly endoscopy + biopsy post treatment to ensure disease response, recurrence or development of adenocarcinoma, then 6 monthly for two years, annual
w Treatment failures: localized RT with curative intent
w Resection: usually not indicated unless perforation, bleeding or obstruction
* RT indicated if:
w t(11;18)
w Penetration deeper than submucosa or LN+ (stage II)
w High-grade dx
w Progression/persistence despite H. pylori therapy
w H.pylori negative (can still trial triple therapy, but unlikely to respond)
* XRT- CR near 100%, high cure rate, low toxicity. Dose 24Gy/12F (Lowry study)
* CT is another option in abx resistant lymphoma - e.g. chlorambucil, Rituximab
* Surgery indicated if risk of perforation. If complete gastrectomy then RT not required, but RT should be given if partial gastrectomy.
* Salvage options after RT:
w CHOP CT- high RR but relapse common. Use if CI to XRT or advanced dx
w Rituximab
w Surgery - but morbidity
H Pylori negative:
* RT is indicated
* Ritux is an option for those where RT is contraindicated
* Endoscopy + biopsy after 3-6 months
Evidence:
* Several single arm, trials and large retrospective series look at H Pylori treatment as treatment for gastric MALT. Comparison is limited due to lack of consensus of response evaluation and definition of CR. Summary of trials:
w Almost all patients able to clear HP – 20% needed a second course of therapy
w Histologic CR in 50-80%
w Median time to clear was 15months
w Median FU 7 years, 16/74 relapsed (22%)
* Aviles (Med Oncol 2005): 241 pts, randomized trial, early stage low grade gastric MALT, randomized to surgery vs RT vs chemotherapy
w No diff in OS
w Mild acute and late toxicities
* Schmelz (J gastro 2019): prospective multicenter trial, 102 patis stage 1E or 21E, refractory or HP negative pts randomized to 25.2Gy vs 36Gy RT
w 76% showed a complete remission at 3 months.
w All pts achieved stable CR regardless of dose
w Presence of t(11;18) and monoclonality of tumour cells were predictors for persistent lymphoma post eradication therapy
w Overall: most HP+, low grade gastric MALT will respond to eradication therapy. In those who dose, dose reduced RT with 25Gy is reasonable

81
Q

Describe the management of H Pylori negative Stage I-II gastric MALT.

A

H Pylori negative:
* RT is indicated
* Ritux is an option for those where RT is contraindicated
* Endoscopy + biopsy after 3-6 months
Evidence:
* Several single arm, trials and large retrospective series look at H Pylori treatment as treatment for gastric MALT. Comparison is limited due to lack of consensus of response evaluation and definition of CR. Summary of trials:
w Almost all patients able to clear HP – 20% needed a second course of therapy
w Histologic CR in 50-80%
w Median time to clear was 15months
w Median FU 7 years, 16/74 relapsed (22%)
* Aviles (Med Oncol 2005): 241 pts, randomized trial, early stage low grade gastric MALT, randomized to surgery vs RT vs chemotherapy
w No diff in OS
w Mild acute and late toxicities
* Schmelz (J gastro 2019): prospective multicenter trial, 102 patis stage 1E or 21E, refractory or HP negative pts randomized to 25.2Gy vs 36Gy RT
w 76% showed a complete remission at 3 months.
w All pts achieved stable CR regardless of dose
w Presence of t(11;18) and monoclonality of tumour cells were predictors for persistent lymphoma post eradication therapy
w Overall: most HP+, low grade gastric MALT will respond to eradication therapy. In those who dose, dose reduced RT with 25Gy is reasonable

82
Q

Describe the management of Stage IIE to IV Gastric Malt

A
  • Distant nodal or advanced stage disease (uncommon), treatment similar to advanced stage FL
    • Asymptomatic patients should be observed
    • Indications for treatment:
      w End organ dysfunction
      w Presence of symptoms (GI bleeding, early satiety)
      w Bulky disease at presentation
      w Steady progression of disease
      w Patient preference
    • Options of treatment:
      w Palliative RT
      w Chemoimmunotherapy (as per NCCN)
      * First line: bendamustine + ritux, RCHOP or RCVP, lenalidomide + ritux
      Second line: lenalidomide + ritux, bendamustine + obinutuzumab, iributinib, P13K inhibitors
83
Q

Describe your radiotherapy technique for gastric MALT.

A

XRT technique
* Most series report close to 100% response rate to XRT, with no failures.
* Pre-simulation
w Hx+PE, rv imaging and endoscopic findings, path
w Consent
w Sim + treat on empty stomach (early am), fast for 4hrs, Breath hold (reduces heart dose)
w Check renal function +/- DTPA scan
* Simulation/Planning
w Supine, vacbag, arms up (4D-CT)
w 4DCT scan with oral + iv contrast
w Prescribe Zofran pre-XRT daily.
* Target volumes:
w GTV= visible dx
w CTV= whole stomach + abnormal perigastric nodes
w ITV= changes in stomach as per 4DCT OR CTV+1cm
w PTV= CTV + 1cm
* Dose prescription: 24Gy/12F 3D-CRT/IMRT (usually AP/PA) to ICRU ref pt, 6/10MV P.
* Daily CBCT ensure stomach coverage
* OAR:
w Kidney <18-20Gy to 2/3 of kidney
w Liver mean <30Gy
w Spleen mean < 10Gy
w Heart and Lung
* Freq SEs: Anorexia, N+V

84
Q

Compare and contrast the types of orbital lymphoma

A
85
Q

Describe epidemiology of orbital MALT

A
  • Affects elderly pts. Most common non-gastric site, 10% of orbital tumours.
    • Common orbital tumour, 50% of all orbital malignancies
    • B cell NHL, mostly arise from MALT (orbital adenxal MALT lymphoma)
    • Non-orbital adnexal MALT lymphomas vary in histology, DLBCL common
    • Among the common OAL tumor types, EMZL, FL, and LPL are considered low grade or indolent, whereas DLBCL and MCL are considered high grade. Mortality for EMZL ranges from 0-20%, FL 20-37%, MCL 38-100% and LPL 14-100%
    • 2% of all lymphomas, 5-15% of extranodal lymphomas
    • More common in females.
    • Arises in
      w Superficial tissues: salmon-flesh appearance in conjunctiva, eye-lids.
      w Deeper tissues: lacrimal gland, retrobulbar areas → proptosis, diplopia.
    • MALT often in conjunctiva; retrobulbar lesions can have more aggressive histology
    • Chlamydia psittaci – orbital ~25%
    • Bilateral dx common → synchronous or metachronous ~10-15%
    • T(11;18)(q21;q21) is seen in 15-40% orbital MALT
    • Behaviour: indolent, uncommon to metastasise.
    • Intraocular less indolent
      w 15% bilateral
86
Q

Describe the presentation and work up for orbital MALT

A
  • Variable clinical presentation
    • Involves conjunctiva, ocular adnexa, lacrimal gland, eyelid, retrobulbar soft tissues.
    • Usually painless, 25% conjunctival invovlement is present
    • Without conjunctival (75%) - usually presents in the superior lateral quadrant, close proximity to lacrimal gland
      § Palpable mass
      § Exopthalmos
      § Ptosis
      § Diplopia, abnormal occular movement s
    • Direct infiltration of globe is rare
    • Vision is usually preserved
    • By definition 1E disease
    • Need Ophthalmologist for conjuctival disease

Workup: Bx, CT/MRI local extent, CT CAP, PET, bloods
Look for salmon-colored conjunctival mass. Fundoscopy, slit lamp.

87
Q

What are the prognostic features for orbital MALT

A

Prognositic Factors:
high-grade disease, stage IVE, LDH, lacrimal gland involvement (40% nodes).

88
Q

What is the differential diagnosis for orbital MALT?

A

DDx:
* Malignant
§ Skin: SCC/BCC/Melanoma
§ Orbital RMS (predominantly in kids)
§ Uveal melanoma
§ Lacrimal gland tumour
§ Mets: melanoma, breasty
* Benign
§ Inflammatory pseudotumour
§ IG4 orbital disease
§ Thyroid eye disease
§ Infection, abscess
§ Cavernous haemangioma

89
Q

What is the treatment for Orbital MALT?

A
  • Asymptomatic + Chlamydia positive:
    w Consider treatment with abx i.e doxy

Radiotherapy:
* Dose:
w Stage 1: 24-30Gy
* Lowry paper: 24Gy/12#
w Disseminated: 4Gy/2# “boom boom”
w [ILROG guidelines] from 2020 suggest 4/2 [is acceptable] for orbital low grade lymphomas (e.g. MZL, MALT, etc).
* Boom boom typically has a CR of 50% and a 2y local failure of 25% for low-grade lymphoma
w Doses of >36Gy assoc with significant toxicity: ischemic retinopathy, corneal ulceration, optic atrophy, neovascular glaucoma, risk of vision loss
Ongoing small trial of 100 4Gy/2# = 90% complete response. If fail give remaining 20Gy

90
Q

What would be the radiotherapy technique for orbital MALT?

A
  • RT technique:
    w Anterior orbital lesions → direct KV (120kV) or E (4-6 MeV) for adequate surface dose. Eyes closed, may require additional bolus.
    w If disease is confined to conjunctiva/eyelid, can consider sparing rest of orbit
    w Higher rates of local failure with partial orbital RT
    w Unilateral Retro-orbital lesion
    * Supine, immobilize with mask, CT scan
    * Target volume:
    ® GTV: gross disease
    ® CTV: whole orbit
    ® PTV: CTV + 5mm
    * Dose prescription: 24Gy/12F to ICRU ref pt, using 6MV photons, 3D CRT/IMRT
    * Wedge pair photon technique (ant + lat-obliques).
    w Lat field has ant edge at lat bony canthus, half beam blocked + angled 5-10deg post, to avoid divergence thro contralat lens. Minimise dose to other eye
    w If proptosis of eye, wedge pair with sup-inf obliques for better dose distn
    * Eyes open- spare conjunctiva
    * TLDs for dose verification
    w Bilateral Retro-orbital lesion
    * Opposed laterals, 6MV photons.
    * Front of beams should be at the outer canthus to avoid irradiating front of eye, angle each posteriorly (5-10deg) to avoid the contralat eye.
91
Q

What is the expected prognosis and toxicity for orbital MALT RT?

A
  • Toxicity
    w Acute - erythema, conjunctivitis, epilation, epiphora (excess tear production)
    w Late - cataracts, dry eye (keratitis sicca) which may lead to corneal ulcer
    w Cataracts occur in ~33% of patients after 2.5-6.5 Gy with 8y latent period.
    w Cataracts occur in ~66% of patients after 6.5-11.5 Gy with 4y latent period.
    § Lens opacification at doses >13-16 Gy.
    § 2 Gy/single fraction firm cutoff. Fractionation: < 4 Gy no cataracts, > 10 Gy 100% cataracts.
    § Cataracts are the classical deterministic (threshold) effect.
    § Stochastic effects have severity independent of dose (e.g. development of SMNs).
    w Loss of eyelashes occurs at 20 Gy and conjunctivitis can be seen at 30 Gy.
    w Retinal damage does not occur with < 24 Gy, but can occur when >34 Gy given.
    w Xerophthalmia TD 5/5 of 35 Gy.
    • LC rates of 85-100%
    • Distant failures vary from 20-50% and usually involve other MALT tissues with indolent behavior and prolonged survival
      w Primary site correlates with risk for systemic involvement:
      * Eyelid: highest risk
      * Conjunctival: lowest risk
92
Q

What is the evidence for management of orbital MALT?

A

Evidence:
* Multiple papers (old and new) showing effectiveness of RT for orbital lymphoma - high rates of LC with minimal toxicity
* Tran (Leuk Lymphoma 2013): retrospective study, Peter Mac, 24 pts and 27 orbits, RT to orbit with 24-25Gy
w All pts had complete response
w 1 local relapse, 1 contralateral, 1 distant
w OS 100% at 2 years and 5 years
Late toxicity: cataracts but nil else

Boom boom for orbital lymphoma may now be acceptable (n=20, 27) [Fasola IJROBP ‘13]

93
Q

Describe the epidemiology, pathology and management of Nodal marginal zone lymphoma.

A
  • 1-2% of all NHL
    * Age >60, F>M
    * >95% have peripheral lymphadenopathy, extranodal sites are uncommon
    * Involvement of bone marrow and peripheral blood in 30-40% cases
    * Usually advanced stage disease but tumors are non-bulky
    * Rarely B symptoms present ~15%
    * Rare, Good prognosis, indolent course; but long-term survival outcomes are less favorable compared to other MALT lymphomas
    * Pathology:
    w Partial/total effacement of architecture, interfollicular infiltrate of monocytoid, centrocyte-like B cells that are 2-3x larger than small lymphocytes
    w Tumour cells surround and infiltrate follicles, moderately abundant pale cytoplasms, round/irregular nuclei with clumped chromatin
    w Similar morphology to MALT lymphoma or splenic marginal zone
    w 30% have neoplastic plasma cells
    w Presence of follicular dendritic meshwork remnant suggestive of colonised follicles favors diagnosis of marginal zone lymphoma
    w IHC:
    * Positive: CD19, CD79a, BCL2, CD22, IgM, IgD
    * Negative: CD5, CD10, CD23, CD25, BCL10, CyclinD1
    w Molecular/cytogenetics:
    * Clonal rearrangements of IgH and light chains
    * Trisomy 18 and 3
    * Treatment options:
    w Treatment not required immediately. Unless symptomatic, most people are “watch and wait”
    w For early disease, use ISRT
    w Chemotherapy(R-Bendamustine), Rituximab, anti-HCV tx
    w SC T for relapse NMZL (no prospective studies)
    w Observe in stage 3-4 with treatment initiation for symptomatic reasons (based on GELF criteria)

Individualize Rx

94
Q

Describe the epidemiology, pathology and management of Splenic marginal zone lymphoma

A
  • Massive splenomegaly w peripheral blood + BM involvement. Usually no ↑LN.
    • Characterized by splenomegaly which is symptomatic when massive or can be assoc with cytopenia
    • Peripheral LN often not involved while splenic hilar nodes often are involved
    • Thoracic or abdominal nodes in about 1/3 pts
    • Bone marrow involvement in 85% and peripheral blood in 30-50%
    • Thought to be uncommon variant of CLL - distinguished from CLL with the absence of CD5 and strong CD20 expression
    • Indolent course
    • Pathology:
      w Clinical, immunophenotypic and genetic features of this are diff from other marginal zone lymphomas - unrelated to MALT or nodal marginal zone
      w Micro:
      * Spleen: micronodular lymphoid infiltrate in white pulp follicles
      w Infiltration by two cell types: small lymphocytes and marginal zone cells - leads to mixed mantle zone and marginal zone involvement pattern
      w Diffuse infiltration in the red pulp
      w Cells are centrocyte-like, monocytoid or lymphoplasmacytic
      * Bone marrow:
      w Combination of nodular, interstitial and intrasinusoidal - nodular pattern is assoc with disease progression
      * Peripheral smear:
      w Cells with scant cytoplasm and cleaved nucleus, confirm neoplastic with flow cytometry
      w IHC:
      * Positive: CD19, CD20, CD22, CD79a, BCL2
      * Negative: CD5, CD3, BCL1, cyclin D1, CD10
      w Molecular:
      * Lacks recurrence chromosomal translocations
      * Clonal rearrangements of IgH and IgL are common
      * Deletions in 7q (up to 40%) + NOTCH2 mutation are specific for splenic marginal zone
    • Treatment:
      w Observe in those who are asymptomatic with no splenomegaly or progressive cytopenias
      w Symptomatic: ritux (high response rates with durable remissions)
      w Splenectomy if disease is not responsive to ritux
      w Consider RT where/if symptomatic
      w Splenectomy may result in prolonged remission.
      Responds less well to CT cf CLL.
95
Q

Describe the epidemiology and pathology of Mantle cell lymphoma.

A

6% of all lymphomas
Worse prognosis
MS of 5 years
In some older pt has slow progressive course
Male predominance
Usually presents with widespread disease, BM invovlement and GIT (stage IV diseasE)
Bone marrow invovlement common
Leukemic phase seen in 75%
GIT frequently involved
Can be associated with formation of multiple polyps (lymphomatous polypoisis)
Other common sites: spleen and walders ring

Histo:
- Arises in the mantle zone of the lymph node which is a post germinal centre B cell
- Micro
○ Small lymphoid cells, arising from mantle zone directly ins ome cases
- IHC
Charactersitic pos for cyclin D1 (translocation of gene encoding for cyclinD1 on chromosome 11, to one for Ig on 14 t(11;14)

96
Q

Describe the management of Mantle cell lymphoma.

A
  • Aggressive, but very radiosensitive
    • Patients rarely present with early stage disease
    • Involved site radiotherapy or radiotherapy for palliation of symptomatic lesions

General Management

Early Stage

* Stage I or Stage II Non Bulky Disease
	○ Chemo* or ISRT (24-36 Gy)
* If Bulky disease
	○ Chemo*

Advanced Stage

* Stage II bulky, Stage III, Stage IV 
* Determine is intent is for auto transplant vs no transplant upfront
	○ Induction chemo**
		§ Clinical response: Maintenance BTKI (e.g. ibruninib) + rituximab
		§ Partial response: BTKI or chemoimmunotherapy 
		§ No response: Second line BTKI or clinical trial or CART cell 

*Chemo 'Less aggressive' e.g. Bendamustine + rituximab, RCHOP, lenolidamide + rituximab

** Induction chemo ‘aggressive’ e.g. Maxi-CHOP (dose intensification immunochemotherapy with R CHOP and alternating rituximab w high dose cytarabine)

Palliation
* Studies have demonstrated effectiveness of ultra-low dose RT (4Gy) for Mantle Cell Lymphoma  19 patients with relapsed refractory mantle cell: 81% CR rate with 4Gy/2#
97
Q

Describe the epidemiology of Primary CNS lymphoma.

A

Incidence
- <1 per 100000 population
- Accounts for 5% of all primary brain tumours
- Rising incidence due to HIV and transplant-related immunosuppression

Median age = 55 years
- Younger in immunocompromised patients
Male predilection in immunosuppressed group only (9:1)

Aetiology

1) Immunosuppression
	a. HIV and transplant-related are most common
	b. EBV infection is thought to be causal
98
Q

Describe the pathology of primary CNS Lymphoma.

A

Pathology

- Most common histopathologies
	○ DLBCL
	○ Burkitt's lymphoma
	○ T-cell lymphoma (uncommon)
	○ Low-grade lymphomas (uncommon)

Primary CNS Lymphoma
- Macroscopic
○ Variable demarcation from surrounding parenchyma
○ Heterogenous appearances (yellow to grey to brown)
○ Typically firm and homogenous
- Microscopic
○ Typical angiocentric growth pattern of neoplastic lymphocytes
○ Diffuse parenchymal invasion by small clusters of cells
○ Reactive astrocytic and microglial activation
- Immunohistochemistry
○ POS = CD19, CD20, CD79a, BCL6, BCL2, MYC, EBV
○ NEG = CD38 (plasma cell), CD30, ALK1, HHV8
- Molecular
○ EBV EBER-ISH is often positive

98
Q

What are the prognostic features for primary CNS Lymphoma?

A

atient Factors
- Age and performance status (most important)

Tumour Factors
- Histopathology
- Deep brain location
- Serum LDH
- CSF protein

Treatment Factors
- Incomplete response to induction chemotherapy

IELSG prognostic score

- 5 factors
	○ Age > 60yo
	○ Performance status (ECOG 3+)
	○ Elevated LDH (above normal)
	○ Elevated CSF protein (above normal)
	○ Deep brain involvement
	
- Prognostic groups (2yr OS)
	○ 0-1 factor = 80%
	○ 2-3 factors = 48%
	○ 4-5 factors = 15%
99
Q

Describe the definitive management of primary CNS Lymphoma.

A

Immediate

- Dexamethasone for symptomatic benefit
	○ Hold off until diagnosis established

Definitive management

- Definitive chemotherapy 
	○ MATRIX protocol (4 cycles)
		§ High-dose methotrexate (3.5g/m2)
		§ Ara-C (cytarabine)
		§ Thiotepa
		§ Rituximab
		§ Leucovorin (24 hours after MTX)

- Response assessment
	○ Repeat MR after cycle 4
		§ If complete response, proceed to consolidation chemotherapy
		§ If incomplete response, should consider WBRT vs BSC

- Consolidation options (if complete response)
	○ HD chemotherapy with ASCT salvage (preferred due to cognitive effects of WBRT)
		§ Carmustine
		§ Thiotepa
	○ Chemo for elderly patients (high dose methotrexate) –ph2 trials showing similar overall survival
	○ 
	
	○ WBRT (ideally if <65 years old and only partial response)
		§ 36Gy/20F via helmet field (Complete Response)
			□ 9Gy/5# boost to residual disease (Partial Response)
		§ Consider extending to CSI at 36Gy/20F if CSF positive
		§ Avoid hypofractionation to brain after methotrexate Complete response to chemo: WBRT 23.4Gy/13# -minimal neurocognitive toxicity

Ocular management:
Options: intraoperative chemo
Orbital RT 23-36Gy (included in lymphoma WBRT PR or CR) = 90% ocular control

100
Q

What is the history and work up of primary CNS lymphoma?

A

Consultation

- History
	○ Presenting symptoms
		§ Raised ICP (Headache, nausea, confusion)
		§ Focal neurological deficits (including seizures)
		§ Time course of symptoms
		§ Constitutional symptoms
	○ PMHx:
		§ Immunosuppression
		§ HIV
	○ Medications
		§ Immunosuppression
	○ Social
		§ Performance status

- Examination
	○ Cognitive assessment (incl performance status)
	○ Full neurological examination
	○ Ophthalmological examination
		§ Papilloedema
		§ Exclude ocular primary
		§ Vitreoretinal involvment
	○ Testicular examination (exclude testicular primary)

Work-Up

- Ophthalmology review
- MR Brain
	○ Typically periventricular location of masses with FLAIR
	○ Lesions have intense enhancement
		§ Immunocompetent = homogenous appearance
		§ HIV = heterogenous irregular enhancement
	○ May be solitary or multifocal

- MR Whole Spine
	○ Should be performed to complete staging

- FDG-PET
	○ Exclude extracranial disease
- Testicular Ex/US

- Bloods
	○ FBC, EUC, CMP, LFT (chemo)
	○ LDH
	○ HIV screening (if not already known)

- Histopathology
	○ If CNS lymphoma suspected, should have stereotactic biopsy only (not resection)

Lumbar Puncture (unless contraindicated)
CSF cytology + protein

101
Q

Discuss the management of relapse refractory primary CNS lymphoma and also palliative management.

A

Relapse/Refractory PCNSL
* Poor prognosis with mOS 3.5mo
○ Small non-randomised trials predominantly
○ Options of therapy:
* Trials
* Retreatment with HD-MTX based chemotherapy schedules followed by thiotepa based HDT ASCT - median PFS 24-41mo (Soussain et al 2008 & 2012m Kasendra et al 2017)
* Ifosfamide based salvage (R-ICE) followed by ASCT (Choquet et al, 2015)
▪ ORR 70%, 23/41pt relapsed with median PFS 133 days
* RTx
▪ WBRT 36-45Gy depending on performance status and life expectancy
▪ CR 37-58% with median survival 10-16mo in RT naïve pts who are refractory to or relapsed after HD-MTX (Hottinger et al 2007, Khimani et al 2011)
▪ IF >60 yo with limited volume relapse, consider rdWBRT 24Gy/16F with sequential boost 21Gy/14F to gross residual disease (Total 45Gy). with orbital dose of 36 Gy/24F if evidence of ocular involvement

Palliative Management

- WBRT
	○ Dose depends on performance status
		§ Good = 36Gy/20F
		§ Poor = 20Gy/5F

- Dexamethasone

- BSC
102
Q

What is the evidence for radiotherapy in management of primary CNS lymphoma?

A

Addition of Radiotherapy to HD-MTX chemotherapy

- Consider in younger patients with a partial response
	○ No OS advantage
	○ Borderline PFS advantage only

	G-PCNSL-SG-1 trial (Thiel, 2010)
		§ 551 patients with newly diagnosed PCNSL were randomised to
			□ HD-MTX-based chemotherapy with or without whole brain radiotherapy
			□ 45Gy/30F
		§ Non-inferiority design (OS is primary endpoint)
		§ Outcomes
			□ No difference in median OS (HR 1.06; p=0.71)
			□ Improved PFS (18.3mo vs 11.9mo)
			□ Significant increase in neurotoxicity with WBRT

Consolidation WBRT vs high-dose chemotherapy with ASCT salvage

- WBRT is a reasonable option with comparable results
- Consideration must be given to neurotoxicity

	○ IELSG-32 trial (Ferreri, 2017)
		§ 227 patients were randomised in a 3x2 factorial design
			□ MTX/cytarabine vs MTX/cytarabine/rituximab vs MTX/cytarabine/rituximab/thiotepa
			□ Consolidation WBRT vs HD chemotherapy with ASCT salvage
		§ Outcomes (second randomisation)
			□ No difference in OS (HR 1.03)
			□ No significant PFS difference between arms (HR 1.50; p=0.17)
				® Trend towards WBRT Increased haematological toxicity in HD chemo arm
103
Q

Describe the radiation technique for primary CNS lymphoma.

A

Consolidation WBRT
Patients
1) CR following induction chemotherapy
2) Palliation

Pre-simulation
MDT discussion
Consider baseline neurocognitive assessment
Fertility discussion (more relevant for chemotherapy)

Simulation
Supine with arms by sides
- Handgrips
Immobilisation mask
Generous CT (2mm)
- Vertex to mid neck

Fusion
MR brain fusion
- T1 GAD + T2-FLAIR

Dose prescription
Two Dose Levels
- 36Gy/20F to the whole brain
- 9Gy/5F boost (if partial response)

3D-CRT technique
        - Helmet field
10 days per fortnight

Volumes
* WBRT
○ Helmet field, incorporating:
○ Cribriform plate
○ Posterior orbit
○ Including C1 & C2 (caudal edge of C2)
* Boost volume (if partial response)
○ GTV
○ Residual disease
○ CTV
○ GTV + 10mm
○ PTV
○ CTV + 5mm

Target Verification
Daily CBCT

OARs
Lens - Dmax < 7Gy

104
Q

What is the expected survival for primary CNS lymphoma?

A

Prognosis

With treatment:

Median OS 35 months
2-Year OS 69%
5-Year OS 20%

Without any treatment
- Median OS = 3-4 months

WBRT: 80% severe neurological toxicity after 1 year (progressive leukoencephalopathy = dementia, ataxia, urinary incontinence), especially age >60

105
Q

What is the classification for primary skin lymphoma?

A

Diverse group of diseases, 5-10% of NHL
Three main catergories with predominant invovlement
- Leukemic
- Nodal
- Extranodal

Chonic, slow progression with long term remission

Classification
* Cutaneous lymphomas may either by ‘primary’ (arisiing initially in skin) or may be a site of extranodal involvement of predominantly nodal lymphoma
* PCL are either T cell (75%) or B cell (25%) in origin
* Heterogenous group of lymphomas from very indolent to aggressive
* The WHO-EORTC classification of primary cutaneous lymphomas is

  1. T cell lymphomas - 75% of PCL
    § Mycosis fungoides (indolent) and Sezary’s syndrome (aggressive)
    § Primary cutaneous CD30+ lymphoproliferative disorders (indolent)
    § Primary cutaneous anaplastic large cell lymphoma (PC-ALCL)
    § Lymphomatoid papulosis (LyP)
    § LyP and PC-ALCL are pathologically distinct but the distinction carries not therapeutic or prognostic differences
    § Rare primary cutaneous T cell lymphomas (cumulatively <10% of PCTCL)
    § Subcutaneous panniculitis-like T cell lymphoma
    § α/β T cell phenotype (indolent)
    § γ/δ T cell phenotype (aggressive)
    § Extranodal NK/T cell lymphoma, nasal type skin is second most common site of involvement after nasal cavity/nasopharynx
  2. B cell lymphomas - 25% of PCL
    § Primary cutaneous marginal zone cell lymphoma (indolent)
    § Primary cutaneous follicular centre cell lymphoma (indolent)
    § Primary cutaneous DLBCL, leg type (aggressive)
106
Q

What is the staging for primary cutaneous lymphoma?

A

Staging (ISCL) “
Note: This staging system applies to primary cutaneous lymphoma except for mycosis fungoides which has a separate staging system

T classification
* T1: Solitary skin involvement (single lesion)
○ T1a: <5cm diameter
○ T1b: >5cm diameter
* T2: Regional skin involvement: multiple lesions limited to 1 body region or 2 contiguous body regions
○ T2a: all disease encompassable in a <15cm diameter circular area
○ T2b: all disease encompassable in a 15-30cm diameter circular area
○ T2c: all disease encompassable in a >30cm diameter area
* T3: Generalised skin involvement
○ T3a: Multiple lesions involving 2 non-contiguous body regions
○ T3b: multiple lesions involving ≥3 body regions

N classification
* N0 : no clinical LN involvements
* N1: Involvement of 1 peripheral LN region that drains an area of current or prior skin involvement
* N2: Involvement of 2 or more peripheral LN regions or involvemet of any LN region that does not drain an area of current or prior skin involvement
* N3: Involvement central LNs

M classification
* M0: No extracutaneous non-LN involvement
* M1: Extracutaneous non-LN disease present

Note:
* Except early stage MF, all pts staged to exclude extracutaneous dx with FBC, UECs, LDH, imaging +/- BM Bx
* Separate staging systems for MF and other non-MF PCL

107
Q

Describe the management of Cutaneous follicular, marginal zone and anaplastic large cell lymphomas

A
  • Behaviour: Indolent
    • Intent: Curative
    • Prognosis:
    • T1 (solitary) - T1a (<5m diameter), T1b (≥5cm diameter)
      ○ Involved lesion RT
      ○ Margins beyond area of clinically evident erythema / induration vary depending on lesion size and body site
      § 1.0-1.5cm
      ○ Thickness of the lesions must be determined to ensure adequate coverage at depth
      ○ Technique:
      § 6-9 MeV electrons, bolus to provide Dmax at surface
      § Low energy x-rays 100 kV photons
      ○ Dose:
      § Definitive: 24 to 30Gy
      § Palliative: 4Gy/2# achieves 72% CR rate, with 30% requiring re-treatment within a median of 6 months
108
Q

Describe the management of Cutaneous DLBC, leg-type.

A

DLBCL, leg-type
* Clinical presentation:
○ Rapidly growing red to bluish tumours, often located on lower extremity
○ The term ““leg type”” reflects the predominant but not exclusive anatomic location of these tumours
* Epidemiology
○ A: Often elderly patients
* Biologic behaviour
○ Aggressive lymphoma
○ Often relapse in extra-cutaneous sites
* Management
○ Solitary or localised disease:
§ R-CHOP → local RT
§ Unable to tolerate chemo:
□ RT alone or combined with rituximab
* RT technique:
○ CTV: Pre-chemotherapy GTV + 1-2cm margin
○ Technique; 6-9MeV electrons with bolus to avoid skin sparing
§ For certain body surfaces, higher-energy photon fields adn opposed field treatment (with bolus) may be required to provide adequate coverage
○ Dose:
§ 36-40Gy
§ 40Gy recommended if no systemic therapy given

109
Q

Describe the management of primary cutaneous T-cell lymphoma

A
  • Subcutanoeus panniculitis-like T cell lymphoma
    ○ Patients with α/β T-cell phenotype and solitary skin lesions should be treated with RT, usually with electrons
    ○ Little information on dose is available, but doses ≥40 Gy have been used
    • Primary cutaneous γ-δ T-cell lymphoma
      ○ Patients often present with generalised skin lesions
      ○ Respond poorly to systemic treatment
      ○ Local control may be achieved with RT to doses of 24 to 30 Gy, but patients tend to relapse
    • Primary cutaneous NK/T-cell lymphoma, nasal type
      ○ In patients with localised skin disease, RT is the first choice of treatment
      ○ Recommended doses are as for NK/T cell lymphoma in the upper aerodigestive tract:
      § 50Gy to the initial lesion and a boost of 5-10 Gy to residual disease
110
Q

Describe the pathology for mycosis fungoides and Sezary syndrome

A

Pathology
* Evolves from localised patches -> plaques -> tumours.
* Infiltration of epi +upper dermis by characteristic neoplastic T cells with cerebriform nuclei= Sezary cells.
* Intraepidermal aggregates of Sezary cells/dendritic cells (Pautrier microabscesses) are pathognomonic.
* Malignant T cells accumulate along basal layer of epidermis→ string of pearls
* Tumour stage- lose epidermotropism

110
Q

Describe the clinical features of mycosis fungoides and sezary syndrome

A
  • Indolent NHL caused by skin-homing CD4+ T cell that cause patches, plaques and tumours.
    • Unique clinical features + histopathology.
    • Characterised by patches + plaques where malignant T lymphos have invaded skin
    • Peak 60yrs; M:F à 2:1; Cause unclear. DDx atopic dermatitis, tinea corporis, psoriasis. Sun exposure appears protective (?immunomodulatory effect dendritic cells)
    • Clinical Features
      • Sx may be present for yrs with waxing/waning cutaneous eruptions.
      • Typically present with erythematous scaly patches or plaques.
      • Initial lesions “patches” resemble eczema or psoriasis→ may delay diagnosis.
      • Lesions typically pruritic, in a bathing trunk distribution (non exposed areas).
      • Evolves from localised patches àplaques àtumours.
      • Another phase of skin dx is generalised erythroderma→ intensely symptomatic.
      • Sezary syndrome= erythrodermic leukaemic variant of MF with 3 components:
        i) T4 disease with Generalised erythroderma (>80% body area)
        ii) ↑LN
        iii) B2: >1000/ml circulating Sezary cells in peripheral blood (atypical T-cells with cerebriform nuclei)
111
Q

Describe the work up for mycosis fungoides and Sezary syndrome

A

Work Up
* Staging system for MF is TNMB (B = blood).
* To stage: examn of skin inc. scalp, palms, soles + perineum.
* Calculate BSA involved using the mSWAT system
* Skin Bx
* Abdo exam to exclude hepatosplenomegaly
* FBC with Sezary cell analysis
* LN biopsy if ↑LN present.
* CT scan in advanced dx (assess visceral + nodal dx).
PET in T2+ dx

112
Q

Describe the staging for mycosis fungoides/ sezary syndrome

A

Presentation: T2- 40%, T1-25%, T3-25%, T4-10%.
* ‘Premycotic’ phase
* Non-specific, slightly scaling skin lesions that wax and wane over a period of years
* May respond to topical corticosteroids
* Biopsies non-diagnostic
* ‘Patch’ phase (T1<10% body surface/T2>10% body surface)
* Histologic findings
* Epidermal lymphocytes with medium to large, extremely convoluted nuclei
* Haloed epidermal lymphocytes
* Pautrier’s microabscesses

Immunophenotype: CD2, 3, 4, 5 +ve, CD45RO+ve, CLA +ve; CD8-ve CD30-ve
* PCR for Tcell receptor rearrangement (ie clonal)
* ‘Plaque’ phase (T1/T2)
* Density of neoplastic cells within dermis increases
* Degree of epidermotropism more exaggerated
* ‘Tumour’ phase (T3)
* Ulcerating or fungating tumours
* Very dense dermal infiltrate involving full breadth of dermis, often extending into subcutaneous fat
* Infection of often cause of death
* Erythroderma (T4)
* Generalised erythroderma ie >80% body involved with patches and plaques, intense pruritus, hyperkeratosis of palms and soles
* Metastases to non-cutaneous sites
* Lymph nodes
* Lung
* Liver

113
Q

What is the diagnostic criteria for mycosis fungoides?

A

Diagnostic Criteria- need 4pts to dx MF
* Diagnosis of MF can be made using the point-based algorithm by EORTC which is based on clinical, histopathologic, molecular, and immunopathologic criteria:
* Clinical criteria: persistent and/or progressive patches and plaques plus (two points given if two of the following are present, one point is given if one of the following is present): lesions in a non-sun-exposed location, size/shape variation of lesions, poikiloderma
* Histopathologic criteria: Superficial lymphoid infiltrate present plus (two points are given if both of the following are present, one point is given if only one of the following is present): epidermotropism (lymphocytes in epidermis) without spongiosis (epidermal oedema common in benign skin conditions), lymphoid atypia.
* Molecular biological criteria: If clonal TCR gene rearrangement is present, give one point.
* Immunopathologic criteria: Give one point if any of the following is present: <50% of the T cells express CD2, CD3, or CD5; <10% of the T cells express CD7; there is discordance of the epidermal and dermal cells with regard to expression of CD2, CD3, CD5, or CD7.

114
Q

Describe the management for mycosis fungoides/ sezary syndrome

A

Topical Chemotherapy
Topical Nitrogen Mustard (mechlorethamine)
* Effectively for T1/T2 dx. All skin daily, then only on clinically affected areas→ maintenance tx for 2m. High relapse, but respond to further Rx.
Phototherapy
* UVB or PUVA (psoralen + UVA photoCT).
* Psoralen orally 2 hrs pre UVA; intercalates with DNA upon exposure to UVA.
* Given 3x/wk until CR, + maintenance up to 1 year.

Radiotherapy
* MF is exquisitely radiosensitive. E or KV RT
* Useful for individual plaques: dose 15Gy/5F, with high likelihood local control.
* TSE considered for thick plaque lesions or pts who failed topical CT or photoTx.
* CR 80% of T1/2. 50% relapse at 1yr, but durable response achieved with more TSE.
* TSE followed by topical nitrogen mustard.
* ↑ CR with TSE in T2 dx vs. topical nitrogen or PUVA. But no ↑ OS.

RT Technique (good coverage Gunderson)
i) Localised fields:
* Palliation of areas of bulky dx or symptomatic skin dx
* Target volume 4-5mm depth for patches/plaques, or to depth of tumours.
* Clinical mark-up. Add 2-3 cm laterally to encompass microscopic dx.
* Choose beam E for adequate dose at depth. KV <1.5cm; E >1.5cm.
* Control rate >95% with 30Gy.
* Dose:
* 15Gy/5F prescribed to 90% isodose with E;
* IF poor PS pts, single # 4 or 8Gy.

ii) TSE RT:
* Indications: thickened plaques, failed topical CT. Delivered with curative intent.
* Target volume = epidermis + dermis

* Dose:
	* 36Gy/18F to skin surface, 2Gy/F, 2 days per week. Hence, takes 9 weeks
		OR
	* 36G/18F to skin surface, 2Gy/F, 5d/wk with 2 wk break in middle. 
	* ICRU 23
	* 6MeV
115
Q

Describe the technique and side effects of total skin electron bream treatment

A
  • Technique:
    Stanford technique:
    * Pt undressed with light underwear stands on rotating table in a wooden frame at 3.5m from linac. Patient rotates through 6 positions involving 6 feet positions and twisting motions. (3 positions/fraction, 1 fraction over 2 days)
    * Scattering filter (clear perspex) at 20cm from the patient for dose uniformity + minimise photon contamination (have guidelines for max photon dose)
    * Position – alternate arms up/down + leg front/back; start at diff angle to gantry each day (smooth out dose distn)
    § Day 1: AP, RAO, LAO
    § Day 2: PA, RPO, LPO
    * TLDs to check doses received, esp. sites where may get underdosing (skin flexures) + need boost (50 TLDs/day)
    * 2 fields for treatment with beam angled down @ 72 and up @ 108 degrees (dep on height of pt)
    § Gap at centre calculated so get 90% dose in centre but no sig overlap
    § Beam divergence matched for sup/inf beams
    * High dose rate used to reduce treatment time.
    * Coverage to about 1cm (90% isodose)
    * Dose uniformity +/- 10%.
    * 6MeV used. (beam degraded to equivalent of 4.2Mev). 80% isodose > 4mm so epidermis + dermis in high dose region. 20% isodose line should be <20mm from skin to minimise dose to deeper structures.
    * 80% isodose should receive minimum total dose 26Gy
    * Electron boost to scalp, perineum, soles of feet, axillae, under breasts + to bulky tumours that are not receiving adequate dose at depth. Max 20Gy/5F.
    * Shields:
    § eyes- external shields (can boost eyelids if required with internal lead shields after)- lead lined googles
    § nail beds (fingers + toes).- lead nail bed gloves
    § scrotum/penis - if concerned about fertility - lead line cricket box

SEs of TSE:
* Acute- erythema + dry desquamation 10Gy. Oedema + blistering 25Gy.
* Subacute- alopecia (at 25Gy), temporary loss of finger + toenails
* Late- chronically dry skin + inability to sweat properly for 6-12 months. Telangiectasia + uneven pigmentation. ↑ risk of cutaneous malignancy.

116
Q

What is the expected outcomes for mycosis fungoides/ Sezary syndrome?

A

Prognosis
* T1- excellent long-term MS, as per N popln.
* Only 9% → advanced stage dx.
* RT curative in pts with early localised dx
* T2- MS 11 yrs
* T3 + T4 without extradermal dx- MS 3-5 yrs.
* Poor PFs: age > 65yrs, extracutaneous dx, large cell transformation, ↑LDH.
* Patients with extracutaneous dx at presentation have MS 1 yr.

Outcome:
* 5yr LC~ 80%
* 5yr OS >90%

117
Q

Describe the radiation technique when treating the spleen for haematological conditions.

A
  • No firm RT schedules, but low dose (<=1Gy) intermittent (2-3 fx/week), to total <=10 Gy recommended. One report suggests conventional 2 Gy/fx may be equally well tolerated. Consider dosing as follows:
    w Lymphoproliferative: 4-10 Gy total in 1 Gy/fx
    w Myelofibrosis: 1-9 Gy total, with 0.25-0.5 Gy/fx
    w Hypersplenism: RT typically not useful
    • Patients should be continuously monitored, and fields adjusted with shrinking spleen
    • RT should be stopped when therapeutic goal is achieved, rather than when prescription is reached
    • Retreatment as necessary, with consideration of renal dose
    • Complications are primarily myelosuppression, and need FBE and exam to be monitored daily
      w Toxicity tolerable in lymphoid disorders, higher toxicity in myeloproliferative disorders
      w Decline in hemoglobin - can lead to MI
      Thrombocytopenia - can result in significant bleeding
118
Q

Discuss the indications for splenic radiotherapy.

A
  • Symptomatic LUQ pain, mechanical discomfort related to large spleen, early satiety due to stomach compression
    • RT used in patients with indications for splenectomy, who are high risk surgical candidates or who decline surgery
    • Mechanism of splenomegaly important for RT
    • Proliferative cell infiltration (lymphoproliferative): chronic lymphocytic leukaemia (CLL), prolymphocytic leukaemia (PLL), acute myelogenous leukemia (AML), hairy cell leukaemia (HCL), splenic marginal zone lymphoma (SLVL)
    • Bone marrow myelofibrosis: resulting in extramedullary hematopoiesis: primary myeloid metaplasia; or secondary to CML, polycythemia vera, or essential thrombocythemia
    • Immune thrombocytopnia (ITP)
    • Hypersplenism: typically seen as cytopenia caused by splenomegaly (e.g. portal hypertension)
119
Q

Describe burkitts lymphoma

A
  • Doubling time 24hrs, highly aggressive, usu children or HIV+
    • Characteristic translocation and deregulation of MYC gene on Chr 8
    • Sporadic type present with abdo mass+ ascites
      ○ Patients with limited stage dx have excellent prognosis.
      NO ROLE for RT, even in localised dx, limited stage disease.
      ○ Treat: Aggressive chemotherapy regimen
      § CODOX-M (cyclophosphamide, vincristine, doxorubicin, high-dose methotrexate) with
      § IVAC (ifosfamide, cytarabine, etoposide, and intrathecal methotrexate)
      ○ High rate of CNS relapse, hence requires CNS prophylaxis.
      ○ Disseminated disease, responsive to chemotherapy
      BMT at 1st relapse.○ Survival decreases with age (SEER data)
      § 5yr OS
      □ <19yo 85%
      □ 20-39yo: 60%
      □ 40-59yo: 50%
      □ >60yo: 33%
120
Q

Describe peripheral T-Cell lymphoma and its management.

A
  • Lymphoma of mature (post-thymic) T-cells →express either CD4 or CD8.
    • Treatment
      w Trial
      w Induction chemo: CHOP (>60) or CHOEP (+Etoposide) (<60), if <60yo x6-8
      * Add brentuximab if CD30+
      w Consolidation:
      * ISRT for early disease/bulky, CR to chemo
      * Autologous SCT in chemosensitive patients indicated if high risk features exist even at first CR
      w HR features = high IPI score, anaplastic large cell and ALK negative
      * ISRT + SCT localised but high IPI; localised with PR to chemo)
121
Q

Describe Extranodal NK/T-cell lymphoma Nasal type and its management

A
  • Localized, aggressive and destructive disease that rarely involves LN. Extensive necrosis and angioinvasion.
    • Presentation:
      w Mostly presents in nose and paranasal sinuses (other extranodal sites- palate, trachea, skin, GIT)
      w Risk of CNS involvement (in advanced stage disease) and local recurrence
      w Highly aggressive, poor response, short OS
    • Prognosis: Poor 5yr OS 40-60%
      w High Px w/ localised dx
      w Low Px w/ Extranasal sites
    • Treatment (no RCTs, retrospective and small prospective series only):
      w SMILE (Dexamethasone, methotrexate, ifosfamide, L-asparaginase, and etoposide regime) preferred to CHOP-> RTx IFRT (50Gy/25Fr)-> Autologous SCT (if young) (SMILE now being used in upfront setting due to promising response to L-asparaginase)
      w SCT : no consensus. consider if CR pts, young pts
      w CNS prophylaxis if 3/4 of the NKPI (NK/T Cell lymphoma Prognostic index):- this determine 5yr OS and risk of CNS involvement without prophylaxis:
      * Presence of B symptoms (fever/chills, drenching night sweats, weight loss)
      * Stage III or IV disease
      * Elevated LDH
      * Regional lymph node involvement
121
Q

Describe the epidemiology and risk factors for multiple myeloma.

A

Incidence (Australian statistics)
- 1800 cases annually
- 0.85% lifetime risk
Australia has the highest rate in the world

Male predominance (1.5:1)
Median age = 70 years
More common african, pacific island, less asian

Aetiology

1) Advancing age
	a. 3% incidence of MGUS in older population
2) Immunosuppression
3) Environmental (chemical)
	a. Benzene
	b. Pesticides
	c. Agent Orange
	d. Farming, wood working, leather working, petrol workers
	
4) Previous radiotherapy
5) Familial link
	a. First degree relative --> RR 3.7
	b. ?need 2nd hit
6) 20% from MGUS
122
Q

What is multiple myeloma?

A
  • Neoplastic proliferation of monoclonal plasma cells producing a monoclonal immunoglobulin or light chains.
    • Plasma cells = mature B cells that produce antibodies
    • Antibody = immunoglobulin
    • Multiple myeloma has different types and subtypes. These types are based on the immunoglobulin (protein) produced by the myeloma cell
    • Each antibody has a light chains (kappa or lambda) and heavy chains (A,E,G,D,M)
      ○ Each antibody has a different number of light and heavy chain pairs -> characteristic normal distribution of antibodies by molecular weight
      ○ Malignant clone causes an overproduction of an antibody- monoclonal Ig which cannot fight infection
    • Most common type of myeloma is IgG Kappa myeloma- produced Ig made from 2 heavy chain IgG bound to 2 kappa light chain
    • 15% of people have light chain myeloma- only produced light chain and no heavy chain- aka Bence Jones proteins
    • 1-2% of pt have “non-secretory myeloma”- produces very little or no monoclonal proteins of any type
    • IgM myeloma is very rare, IgM production occurs in a Walderstroms macroglobulinaemia, which is more like a lymphoma than myeloma.
    • Paraprotein = an abnormal protein, monoclonal immunoglobulin or light chain, present in blood or urine produced by clonal proliferation of plasma cells
  • Clinicopathological entity
    • The clones proliferate in BM→ lytic bone destruction, osteopaenia, path fracture and ↑ Ca. BM suppression.
    • The secreted paraproteins can cause nephropathy and hyperviscosity syndromes.
123
Q

Compare MGUS and Smouldering Myeloma.

A
124
Q

Compare Solitary plasmacytoma and multiple myeloma

A
125
Q

Describe the types of plasmacytoma and the pathogenesis.

A

Plasmacytoma Types

- Osseous
	○ 50% risk of transformation to multiple myeloma
	○ Low risk of local recurrence after radiotherapy (10%)
	
- Extraosseous
	○ Sites
		§ COMMON = pharynx, paranasal sinuses, lungs
		§ UNCOMMON = GT, connective soft tissue, liver, nodes
	○ Prognosis is much better than osseous type
		§ Low risk of recurrence with RT (<10%)
		§ Low risk of transformation to multiple myeloma (<15%)

Pathogenesis

Heterogenous genetic mutational profiles are typical

IGH gene (chromosome 14) is frequently involved
- Cyclin D1 (chromosome 11)
- Cyclin D3 (chromosome 6)

Poor prognosis has been associated with
- p53 deletion
- MYC amplification

Bone destruction

Factors secreted by myeloma cells augment bone resorption pathways (favouring destruction)
- Osteoclast activation = CCL3 & RANKL
- Osteoblast inhibition = modulators of the Wnt pathway (e.g. DKK1)

126
Q

Describe the pathology for multiple myeloma

A

Macroscopic
- Radiographic punched out lesions within bone
- Soft, gelatinous and red tumour masses (“fish flesh”)

Microscopic
- Sheets of malignant plasma cells or plasmablasts, show classic plasma cell features; abundant basophilic cytoplasm, perinuclear hof, round eccentric nuclei, clockface chromatin, Mott cells, Russell bodies, dutcher bodies, flame cells
- Tumours are comprised with populations of monoclonal plasma cells with varying maturity
○ Mature plasma cells
○ Plasmablast = vesicular chromatin and prominent single nucleolus
○ Bizarre multinucleated cells
- May have globular cytoplasmic inclusions (Russell or Dutcher bodies)

Immunohistochemistry
- POS = CD138/CD79a (plasma cells), CD56
○ Light chain restriction (stain for EITHER kappa or lambda LCs - but not both)
- NEG = CD19/20 (normal B-cells), CD 45, PAX,
○ CD 27, 81 (normal plasma cells)

Molecular/cytogenetics:
* Abnormal cytogenetics in only 1/3 cases
* Some cytogenetics associated with worse outcome - del (13q), t(4;14), t(14;16), t (14, 20), del (13p), del (17p)
* IgH rearrangements in 75% pts

127
Q

What are the prognostic factors for multiple myeloma?

A

patent Factors
- Age
- Performance status
- Serum studies
○ Albumin < 30g/L
○ Creatinine > 200
○ Hypercalcaemia
○ Anaemia

Tumour Factors
- Burden of disease
○ MM vs solitary plasmacytoma
○ Bone marrow (>50% is adverse)
- Maturity of cells (plasmablastic cells impart worse prognosis)
- SFLC ratio
- Beta-2 microglobulin
- LDH
- Cytogenetics
○ Worse: hypoploidy, t(4;14), MAF translocations t(14;16), t(14;20), del(17p), del(13)
○ Better: hyperdiploid, t(11;14)

Treatment Factors
- Time to relapse following therapy

128
Q

What are the risk factors for progression to multiple myeloma from MGUS and for smouldering myeloma?

A

Risk of Progression (MGUS –> MM)

1) Elevated monoclonal globulin (>1.5g/dL)
2) Abnormal serum light-chain ratio
3) Non IgG gammopathy (e.g. IgA)

Risk of Progression (Smouldering Myeloma –> MM)

1) Elevated monoclonal globulin (>2g/dL)
2) Marrow plasma cells > 20%
3) SFLC ratio > 20
129
Q

Describe the history and examination for multiple myeloma.

A
  • History
    ○ HPI
    § Classical symptoms
    □ Bone pain
    □ Hypercalcaemia (neurological, renal stones, bone pain)
    □ Renal failure (incl pruritis)
    □ Anaemia (fatigue, dyspnoea)
    § Neurological deficits (spinal cord compression)
    § Constitutional symptoms
    § Recurrent infection (impaired immunity)
    ○ PMHx:
    § Immunosuppression
    § Previous radiotherapy or chemotherapy
    ○ Medications:
    § Immunosuppressants
    § Nephrotoxics
    ○ Family History
    ○ Social
    § Performance status
    § Smoking
    • Can also have myelosuppression, immunosupression
    • Examination
      ○ General inspection
      ○ Bony palpation
      § Spine
      § Calvarium
      § Ribs
      § Shoulder girdle
      § Pelvic girdle (incl hips)
      ○ Nodal examination
      ○ Skin -plasmacytomas -purple nodules -very poor prognosis
130
Q

Describe the work up for multiple myeloma

A
  • Laboratory
    ○ Serum
    § FBC
    § EUC, CMP, LFTs
    § LDH - for prognostic/staging factor
    § Albumin - prognostic/ staging factor
    § Beta-2-microglobulin - prognostic/ staging factor
    § Serum free light chains (SFLC) - to determine kappa or lambda light chain
    § Electrophoresis (EPG/iEPG) - to determine if monoclonal band present (IgA, IgM, IgG)○ Urine
    § Bence Jones protein (ideally 24 hour collection)
    § Urine electrophoresis
    ○ Peripheral blood film- Rouleaux formation- stack of RBC, related to the quantity and type of M protein
    • Histopathology
      ○ Biopsy of any plasmacytoma
      ○ Bone marrow biopsy
      § Proportion of involvement. Need > 10% of BM involvement for MM
      § Cytogenetics and Myeloma FISH
    • Imaging
      ○ Whole-body CT Skeletal Survey
      ○ Consider FDG-PET (1 funded under rare cancer medicare)
      § Mandatory as per ILROG
      § NOT for WBBS (no osteoblastic activity)
      § PET better for looking at estramedullary sites
      ○ Consider MR spine as appropriate
      § 5mm lesion
131
Q

What is the staging for multiple myeloma?

A
132
Q

Describe the management of MGUS and smouldering myeloma.

A

MGUS

- No specific management is indicated
- Treatment does not reduce risk of transformation

- Annual surveillance should be performed to watch for progression
	○ Clinical history and examination
	○ Laboratory studies
		§ FBC, EUC, CMP
		§ Serum EPG/iEPG
		§ Serum free light chains

Smouldering Myeloma
management of smouldering myeloma primarily involves regular monitoring and observation to detect any progression to active myeloma. In recent years, there has been growing interest in early intervention for patients with high-risk smouldering myeloma, though this is typically done within the context of clinical trials. The decision to start treatment is based on a comprehensive assessment of the risk of progression, the patient’s overall health, and potential benefits versus risks of therapy.

132
Q

Describe the management of multiple myeloma.

A
  • Cure is not possible although treatment always is.- Wide range of treatment available
    ○ course is variable and diff people have diff risk of disease progression
    ○ Aim= achieve control of disease = complete remission or MRD (minimal residual disease)
    • Supportive care is important
    • Factors affecting Mx:
      ○ Age and general health - elderly might not be transplant eligible
      ○ Pt’s preference
      ○ Eligibility for ASCT - improve ORR, PR, CR, EFS and OS (by 9-18mo)
      § For pts <75
      § Mortality <0.5%
      § Recover blood cells in 12 days, QOL baseline in 3 months.
      ○ MM disease burden and aggressiveness
      ○ Risk profile
      ○ Access to new agents/ cost/ clinical trials
    • Address organ damage and preserve function:
      ○ renal - protection and hydration
      ○ bone- zoledronic acid
      ○ immune - antimicrobial prophylaxis and IVIg
    • Urgent complications of myeloma should be managed first
      ○ Renal failure - renal review, dialysis
      ○ Hypercalcaemia - biphosphonate, IVF
      ○ Spinal cord compression- Surg+ RT, Dex, analgesia
      ○ Hyperviscosity syndrome- anticoag
    Treatment strategy:
    ○ Manage MM complications
    ○ Induction (lite chemo if no transplant)
    ○ Consolidation (autologous stem cell transplant)
    ○ maintenance
    - Manage systemic disease in first instance
    ○ First-line = CyBorD induction
    § Consider autologous SCT for consolidation (if eligible)
    § Alternative is lenalidomide maintenance
    ○ Second-line = as per PBS/patient factors
    § carfilzomib/lenalidomide/dexamethasone
    § Alternative is daratumumab/lenalidomide/dexamethasone
    • Radiotherapy has solely a palliative role
      ○ Quite radiosensitive
      ○ Higher doses are associated with improved durability of control
    • ILROG recommendation:
      ○ Bony sites for pain control: either 8Gy/1F, 20Gy/5F or 30Gy/10F.
      § Single 8/1 preferred for poor prognosis patients
      ○ Large volumes or re-treatment: consider 30Gy/10-15F
      ○ Cord compression or bulky mass where durable control needed: 30Gy/10-15F
      ○ No need to add additional margin GTV-CTV as not critical to cover subclinical disease in the context of wider systemic disease. Whole bone coverage not required.

Biologicals/monoclonals, used in combination
Often CD38, CD138 eg Daratumumab
Immunotherapies -
Targeted therapies -plasma cells, osteoclasts, IL-6 pathways -RAF, PI3k
CAR-T or Bispecific T cell engager

133
Q

Discuss the management of solitary plasmacytoma.

A

Solitary Plasmacytoma

- Immediate management should be considered as appropriate
	○ Bone stabilisation
	○ Reverse airway obstruction

- Dose: No high-level evdience, snall retrospective series.  ILROG > 5cm 40-50 Gy, <5cm 35-40Gy/ SEPs 40-50Gy (1.8-2 Gy per fraction)
- Definitive management consists of radiotherapy alone
	○ 45Gy/25F
		§ Small lesion <5cm consider 40Gy
	○ GTV + 2cm
- If tumour resection has occurred, RT to the tumour bed is still indicated

- No rationale for adjuvant systemic therapy
	○ Surveillance alone
134
Q

Describe your radiotherapy technique for plasmacytoma

A

Definitive RT
Patients
1. Solitary Osseous Plasmacytoma
2. Solitary Extra-Medullary Plasmacytoma

Pre-simulation
Full myeloma screen
MDT discussion
- Exclude diagnosis of multiple myeloma
Consider need for urgent management
- E.g. surgical decompression or fixation
Fertility consideration
- bHCG in all females
- Fertility preservation
Smoking cessation
Bortezomib and abdo GIT toxicity

Simulation
Supine with arms above head
Vacbag
Generous CT (2mm without contrast)
MRI for H+N, epidural extension,

Fusion
Pre-surgical imaging (PET or MR)

Dose prescription
Single dose level
- 45Gy/25F prescribed to PTV
VMAT technique
10 days per fortnight

Volumes
GTV
- Plasmacytoma as seen on imaging
CTV
- GTV + 2cm
CTV (post-op)
- Pre-op GTV + 2cm
- Ensure surgical bed is incorporated
PTV
- CTV + 7mm

Target Verification
Daily CBCT

OARs
Spinal Cord
- Dmax < 45Gy

Kidneys (bilateral)
        - Mean < 15Gy
H+N constraints
Bone marrow in adults
Pelvis 25%, T spine 20% L spine 20% Ribs/calv 9%
Everything else ~3-5% each bone
134
Q

What is the evidence for the use of radiotherapy in solidary plasmacytoma?

A

NCDB retrospective analysis (Goyal, 2018)
- 5056 patients are included for retrospective review
○ 70% are osseous plasmacytoma
- Outcomes
○ Most common extra-medullary sites were upper aerodigestive tract (45%) and soft tissue (19%)
§ Soft tissue had worse OS
○ Both advancing age and bone primary were associated with worse OS
○ Combination of surgery and adjuvant RT had the best OS
○ RT at a dose > 40Gy was associated with improved OS

Retrospective registry review (Ozsahin, 2005)
- 258 patients with plasmacytoma included
○ 80% are osseous plasmacytoma
- Outcomes
○ Median RT dose 40Gy
§ No dose response gradient seen above 30Gy
○ Local control was superior when size < 4cm
Persistent paraprotein elevation at 12mo are at increased risk to progression to MM

134
Q

What is the evidence for the use of radiotherapy in multiple myeloma?

A

Retrospective review (Rades, 2016)
- 237 patients with multiple myeloma causing spinal cord compression were included
○ Patients who required neurosurgery were excluded
- Outcomes
○ Overall RR was 97% (with53% being improvement)
○ Ambulation maintained in 88% of patients
§ Of patients who were non-ambulatory at presentation, 64% regained ambulation
○ No improvement in local control based on length of RT course
§ Short course = 8Gy/F or 20Gy/5F
§ Long course = 30Gy/10F, 37.5Gy/15F or 40Gy/20F
- Summary: RT alone provides excellent neurological and local control results in myeloma (radio-sensitive disease)

Palliative dose: all restrospective series 30Gy for spinal cord compression, otherwise 20Gy/5 or 8 in 1 ok

135
Q

What is the expected prognosis and follow up for plasmacytoma and myeloma?

A

Complete remission of 40-60%

Follow-Up

- Clinical review every three months for the first two years
	○ Clinical history and examination
	○ FBC, EUC, CMP, LDH, serum FLC, EPG/iEPG
- Clinical review every six months for the indefinite future
	○ Clinical history and examination
	○ FBC, EUC, CMP, serum FLC, EPG/iEPG

- Consider urine Bence Jones as indicated
- Imaging as indicated only Routine annual imaging for solitary plasmacytoma & smouldering myeloma
136
Q

What is the epidemiology and risk factors of ALL

A

Incidence of ALL (Australian statistics)
* 200 cases annually
* 4.2 cases per 100000 children
* Accounts for 75% of cases of childhood leukaemia
Leukaemia is the most common form of childhood cancer (30%), ALL (70%)&raquo_space;> AML
* Majority of paeds leukaemia are B-cell origin (85%)
Peak incidence is 2-5 years of age
Male predominance (1.5:1)

Among leukaemia, 80% are ALL and CML/AML account for 20%

Aetiology
1. Genetics
a. Trisomy 21 (Down’s)- increased risk of ALL, and B-ALL have increased relapse and toxicities
b. Ataxia Telangiectasia (ATM)
c. NF1
2. Previous radiation exposure
3. Previous chemotherapy
4. Environmental exposure
a. Benzene
5. infective viral- HTVL-1 assoc with T cell ALL, decreased immunity

137
Q

What is the epidemiology and risk factors for AML?

A

AML

Epidemiology
* rare, just over 1% of adult cancer
* 2nd most common leukaemia in adult after CML
* median age 65yo
* incidence increases with age
* Men predominance
* High incidence in caucasian

Aetiology
* Preceeding by previous clonal haematopoesis conditions eg. myelodysplastic syndrome, myeloproliferative disorders, paroxysmal noctural haemoglobinuria, aplastic anaemia
* Environmental exposure
○ industrial chemicals
○ tobacco smoking
○ chemotherapy
○ RT
○ retrovirus
* Genetic :
○ Trisomy 21, Fancomi anaemia, Blooms syndrome

138
Q

What is the history and work up for Leukaemia?

A
  • History
    ○ HPI
    * Constitutional symptoms- fever, lethargy
    * Bleeding, Bruising and petechiae
    * Pallor and fatigue
    * Lymphadenopathy
    * Bone pain
    * mass effect- bone pain, liver/renal/ testes
    ○ PMHx
    * Genetic syndromes
    * Previous RT or chemotherapy
      * Examination
          ○ Nodal examination
          ○ Abdominal examination
              * Hepatomegaly
              * Splenomegaly
          ○ Skin examination
              * Bruising, petechiae
              * Include oral mucosa and conjunctivae
          ○ Full neurologic examination - to exclude CNS involvement
          ○ Testicular examination - exclude testis involvement

Work-Up
* FBC with blood film- exclude thrombocytopaenia
* EUC, CMP, LFT, LDH, uric acid - assess kidney, liver involvement, and cell turnover
* Coags
* Viral serology (CMV, EBV, HIV, HepB, HepC)

* CT Chest, abdo, pelvis
* MR Brain
	 
		* Bone Marrow Biopsy
		* Lumbar Puncture (if Ptl >20)
 
* Prior to chemotherapy
	○ Urinalysis and blood cultures
	○ TTE Lung function tests
139
Q

Describe the pathology for ALL

A
  • Proliferation of malignant T or B blast cells-> failure to replicate and mature, crowding out the normal cells
    • Dx: 25% of BM blasts
    • Micro: hypercellular BM with small cells with little cytoplasms, might mitotic rate, interspersed with macrophages “starry sky”
    • Flow: TDT (only on blast cells)
    • B- CD19/10 (not CD20 as too immature)
    • T- CD1,2,57
    Cytogenetics
    * t(12:21)= good prognosis
    ○ Hyperploidy= good prognosis
    ○ t(9:22)= philadelphia fusion, BCR-ABL gene = bad prognosis- increased recurrence and CNS disease
    * result in constant activation of the TKI
140
Q

What are the prognostic factors for leukaemia?

A

Poor prognostic groups
* T-cell –> 5 year EFS < 80%
* Ph+ (t[9:22]) –> 5 year EFS 70%

Prognostic factors:
* Patient:
○ age <1yo or >10yo
○ male
* Tumour:
○ WCC high >50
○ T-cell ALL
○ CD10 neg
○ CNS involvement
○ Testis involvement
○ Hypodiploidy, Ph+
* Treatment:
○ slow response to MRD
○ measurable residual disease post Chemo

141
Q

What is the risk stratification and staging for leukaemia?

A

High risk ALL:
* all adults
* Older children > 6-7yo
* T-cell phenotype
CNS2-3 disease

142
Q

What is the management for ALL?

A

In general, treatment comprises 4 phases:
1. Induction chemotherapy- aim remission and normal haematopoesis
a. pred, vincristine, L-asparginase +/- IT MTx if CNS disease
2. CNS prophylaxis -treat if high risk of CNS - Dz at Dx, T-cell, Ph+, High WCC
Options:
3. IT-MTX + Cytarabine (preferred)- has CNS penetrance
b. Cranial irradiation only used in high risk patients
i. CNS3 disease (proven CNS dissemination at diagnosis) –> biggest indication
ii. High WCC (>100)
iii. T-cell phenotype
iv. Adverse cytogenetics (t[4:11] or Ph+)
v. Lymphomatous presentation
3. Consolidation -avoid early relapse
a. mercaptopurine, vincristine, asparginase
b. May include allogenic SCT (Might need TBI for conditioning)
c. If testicular residual involvement, consider irradiation (santuary site)
i. 24Gy/12F bilateral testes
4. Maintenance- maintain remission, suppress any drug resistant clones- oral mercapto, oral MTX, 3/12 IT MTX

143
Q

What is the role of radiotherapy in ALL?

A
  • CNS prophylaxis
    • CNS therapy for active disease
    • TBI for conditioning prior to ASCT
    • Testis RT in relapse
    • Splenic RT for Myeloinfiltration
    • Palliation
      ? Role of CSI 14.4Gy/8F in active CNS disease but concern on BM suppression for future chemo and consideration of possible future TBI
144
Q

Describe non-myeloablative haematopoietic stem cell transplant conditioning.

A
  • no eradication of the host BM cell to achieve graft versus tumour effect to eradicate any residual AML cells
    ○ In non myeloablative/RIC regimen, TBI can also be used in combination with chemotherapy agent to suppress the host’s immune cells with the goals of increasing successful stem cell transplant and allow graft vs tumour cell effect. This is used in previously treated patient with stem cell transplant or frail patient who might not tolerate myeloablative regimen
    • allows transplantation in high-risk or previously heavily treated pts with chemo to decr regimen toxicity and mortality
    • it is also used in circumstances where cancer cells cannot be eradicated entirely by HDCT.
    • Options:
      ○ Flu/TBI- fludarabine over 3 days with 2Gy TBI on the day of the graft
      ○ TLI/ATG (total lymphoid irradiation 8-12Gy over 11 days with anti-thymocyte globulin administered over 5 days)
      * lower risk of non-relapse mortality but higher risk of relapse
      * similar EFS and OS
      ○ Flu/Mel, Flu/Bu2, Flu/Cy, Flu/Bu/TT.

NMA vs Myeloablative
* MA - higher RFS BUT higher treatment related mortality. Similar OS [as per multicentre Ph3 trial]

144
Q

Describe Haematopoietic Stem Cell Transplant (HSCT)

A
  • IV administration of haematopoetic stem cells to eradicate malignant cells and re-establish BM function
    ○ Autologous: stem cells from pt, only irradiates
    * ADV: decr mortality, nil donor issues, nil GVHD, combat chemo resistance with HDCT, incr OS
    * DISADV: no GVT effect, expensive, still toxic+morbid “sepsis risk”, risk of infusing malignant cells
    ○ Allogeneic: stem cells from a HLA matched donor, need immunosuppression
    * Need HLA matching
    * increase mortality dt GVHD but
    * increase cure dt Graft vs Tumour effect
    ○ Mini-Allogeneic (NMA): recipient BM not eradicated but donor WCC causes GvT effect.
    * avoid myeloablation toxicity
    * Graft vs tumour effect
    • Stem cells are harvested from the BM in the iliac crest or peripherally with GCSF
      ○ BMT - harvested from post iliac crests by repeated aspirations under GA
      ○ PSCT - mobilisation of stem cells by chemotherapy and G-CSF into the peripheral circulation and harvested by leukopheresis
145
Q

Describe myeloablative haematopoietic stem cell transplant conditioning.

A
  • Use of chemo only (Bu/Cy - Busulfan+ cyclophosphamide) or chemo+RT (Cy/TBI 12Gy/6F BD over 3 days) to potentiate and maximise tumour eradication and immunosupression before stem cell transplant
    • Minimise risk of graft failure but also increases treatment related toxicity and mortality
    • MA comparing Bu/Cy vs Cy/TBI
      ○ Bu/Cy
      * Higher hepatic sinusoidal obstructive syndrome (13 vs 6%) AND higher relapse risk
      * No diff in treatment related mortality, risk of GvHD, DFS or OS
    • TBI - 12 or 16Gy?
      ○ RCT showed that relapse risk and graft failure is lower with 16Gy BUT higher transplant related morbidity and mortality-> hence OS unchanged
146
Q

What are the pros and cons of TBI vs chemotherapy conditioning?

A

Pros of TBI
* Data suggests OS advantage in ALL
* Independent of blood supply (no sanctuary sites). Allows treatment of sanctuary sites
* Independent of drug bioavailability, clearance, etc.
* No cross resistance with other agents (chemo resistant clones may be sensitive to RT)

Cons of TBI
* Acute toxicities
○ GI (nausea, vomiting, diarrhoea), H&N (xerostomia, parotitis, oral mucositis), general (fatigue, headache, fever), skin (erythema, alopecia)
* Marked late toxicity profile
○ CNS (slightly lower IQ), H&N (cataracts, xerostomia, dental decay), endocrine (GH, TSH, gonadal hormones - delayed puberty, infertility, early menopause), growth and development (impact on bone development), lung (radiation pneumonitis), liver (veno-occlusive disease 10-15%), kidney (nephropathy), second malignancy (predominantly haematological)
* Logistical challenges
Might not be suitable for elderly with morbidities

147
Q

What are the indications for TBI?

A
  • No role in autologous HCT
    • Indicated in:
      ○ Myeloblative transplants
      * Leukaemias
      ▪ AML
      * First remission with intermediate to poor risk molecular and cytogenetic prognostic features
      * Refractory disease or non-responsive to induction chemotherapy
      * Remission after treatment for relapse
      * Refractory disease after relapse
      ▪ ALL
      * First remission with poor risk features or Philadelphia chromosome positive disease
      * Refractory disease or non-responsive to induction chemotherapy
      * Remission after treatment for relapse
      * Disease that remains refractory after relapse
      ▪ CLL uncommonly used
      * Consider in relapsed/refractory CLL in young fit patient (minority - median age 70yo)
      ▪ CML uncommonly used
      * Consider in accelerated / blast phase (first line therapy in chronic phase is imatinib)
      * Lymphomas
      ▪ Relapsed aggressive NHL (PARMA trial)
      ▪ Relapsed/refractory Hodgkin lymphoma
      * Solid malignancies
      ▪ Neuroblastoma
      ▪ Metastatic Ewing sarcoma
      ▪ Testicular tumours
148
Q

Describe testicular radiotherapy and technique

A

Rationale:
* testes are sanctuary sites
* need intensive high dose MTX to penetrate blood testis barrier
* relapse after chemo <5%,
* Testicular RT is ONLY given in relapse to avoid sterility
Need fertility discussion as both testes needs to be irradiated!!!
Technique:
* S- supine, frog leg, vacbag, mark up clinically, tape penis up. Ensure cremasteric reflex doesnt pull testes up into the inguinal canal
* V- bilateral testis in scrotum
* D- 24Gy/12F
* T:
○ 10-12MeV/ 250KV, dose to 90% isodose (If scrotal/testes thickness <2cm; children)
○ 6MV photon beam with bolus, angle the photon beam anterior inferior oblique (if scrotal/tests thickness >2cm; adults)
Toxicity: hypogonadism, sterility, hypoplasia, 2nd malignancy

149
Q

What is the evidence for TBI in HSCT myeloablation?

A

FORUM trial (Peters, 2021)
* 417 patients with ALL who were in remission following induction chemotherapy
○ <18yo
○ High-risk ALL
* For consolidation allogenic stem-cell transplant, patients were randomised to
○ Chemotherapy alone conditioning (fludarabine, thiotepa and busulfan)
○ TBI containing conditioning (12Gy/6F BD TBI + etoposide)
* Non-inferiority design
* Trial ceased early due to futility
* Outcomes
TBI associated with significantly higher 2 year OS (91% vs 75%; p<0.0001)

150
Q

What is your radiotherapy technique for TBI?

A

Presim:
- Haem MDT discussion, Ensure complete remission with induction chemo
- Consent
- Baseline blood (daily blood test during TBI)
- Assess baseline renal, pulmonary and hepatic function—Acquire baseline function for each.
- Baseline ophthalmologic examination.
- Testicular examination +/- USS to rule out testicular involvement with leukaemia.
- Counsel on family planning and sperm banking if needed
- Counsel on sex abstinence or protected sex with contraception after RT
- ensure no previous RT delivered
- Smoking cessation
- Coordinate Start date of TBI with stem cell transplant, chemo dates, and Timing of admission
- Ensure no active infection
- Premed with ondansetron and Dex 4mg daily before each RT

SIM:
- Present during mock up sim to oversee and check set up with physicist and RT
- Set up: Arm on chest, supine with full body vacbag. knee and hip flexion and knee block blocks if height > 165cm
- Perspex blocks on either side of the head and rice bags for neck compensation
- Perspex shielding for spoiler at 15cm from the pt bed (in between the gantry and bed)
- Measurement taken for extended FSD (gantry at 90 degree with rotated collimator, 40x40cm, pt planned at extended FSD ~4m on either side of the room)
- Use light field to check pt is completely covered by the treatment beam
- CT sim whole body

Dose and technique
- Dose: 12Gy/6F BD , 2Gy/F, 6 hour apart, over 3 days until the day of stem cell transplant, Prescribed to MPD, isocentre 2cm from umbilicus
- Opposed lateral, 6MV photons, extended SSD 365cm
- turn pt at 180 degree (rather than gantry) for each field.
- Dose rate 5-7 cgy/min for ALARA and reduce need for shielding of lung, heart and kidneys

Volume:
- Whole body – based on measurement from FSD and divergence rather than CT-volume based

OAR:
- Lung mean dose < 10Gy
- Lens Dmax <8Gy
- Perspex block decrease dose to brain and head

Target verification:
- Ion chamber placed in between thighs for in vivo dosimetry
- multiple TLDs in the body- bilateral head, neck, chest , hip and groin

151
Q

What are the challenges with TBI?

A

Organisational challenges:
- Treatment typically delivered BiD with long treatment time, hence, timing and scheduling of machine is critical and need for a back-up plan in case of failure.
- Timing and scheduling of TBI with chemotherapy and transplant—need well coordination and communication with haematology team to ensure timely and smooth delivery of treatment.

Clinical challenges:
- Long treatment time may be a challenge for a patient who is unwell to maintain the same position—ensuring that patient is in comfortable position to minimise movement.

Technical/ physical challenges:
- Field size: maximum field size is 40cm for conventional Linac, hence Extended SSD of 4cm from isocentre is needed to ensure whole body coverage
- Technique: with conventional linac, typically limited to opposed lateral fields in supine position. Alternatively, standing or lying position with AP/PA fields and need lung shielding
- Dose rate: to decrease risk of pneumonitits, need reduced dose rate to <7cGy/min when using extended SSD
- Dose homogeneity: Due to changes in separation/thickness of patient- use of tissue compensators to the thinner parts of the body e.g. legs and ankles to improve dose homogeneity, using higher beam energy to decrease inhomogeneity between superficial and deep tissue (but careful consideration of skin sparing and potential underdosing to superficial structures e.g. skull)
- Tissue shielding/ compensation: aim to deliver TBI to prescribed dose +/- 10% to account for dose inhomogeneity. This can be minimised by using tissue compensator esp in head, neck, legs, ankles. Additional compensator/block can be used for lung, kidney, liver in pt with organ dysfunction
- Commissioning: beam calibration, PPD and Tissue-phantom ratios and off-axis ratios needs to be measured in the same condition as in TBI setup. Need to account for large RT fields and scattering from the treatment room, floor and walls, size of pt and calibration phantom. To ensure adequate dose delivered, use invivo measurement with TLD (to verify entrance and exit dose) and ion chamber

152
Q

Describe prophylactic and therapeutic cranial irradiation for haematological patients.

A

Rationale:
* PCI for high risk pts due to high risk CNS disease (T cell, Ph+, High WCC, poor responders)
* treat CNS disease at dx
Technique:
* S- supine, mask, TLDs on eyes- lens dose
* V-
○ entire brain, cranial meninges to C2, include temporal lobe, cribriform plate, post 1/2 of the globe
* D- 12Gy/8F for PCI, 24Gy/12F if CNS disease
[Bible 12Gy/8F PCI and 18Gy/10F therapeutic]
* T:
○ opposed lateral beams, prescribed to MPD, 6MV photons
○ avoid lens by moving gantry angle by 5 degrees, isocentre at the lateral canthus + half beam block
Toxicity:
* Acute: headache, nausea, vomiting, alopecia, lethargy, somnolence syndrome, pharyngitis, otitis
* Latent: neurocognitive decline, cataract, hypopituitarism, 2nd malignancy

[FRANZCR]
Cranial RT Late toxicity (for 24Gy/12F)
* Somnolence syndrome
○ 1mo post RT, 40-50% pts gets lethargic, irritable, anorexia +/- fevrs
○ usually improves in 2-3 weeks
○ steroids during cranial RT may help
* Cataract
* Pituitary Dysfunction
○ usually affect only GH production at doses used, and more likely in children <5yo
* NeuroCognitive impairment
* Leukoencephalocapthy- demyelination conditions occurs months post RT + IT/IV MTX, seen with 24Gy but risk decreases if <20Gy PCI RT
* Secondary cancers
○ not that common with cranial RT alone, more common when PCI RT combined with TBI
○ HGG/ meningioma and thyroid cancers

153
Q

A fit 66-year-old male with nausea and reflux undergoes gastroscopy showing diffuse thickening of the gastric mucosal folds in the body of the stomach. Multiple endoscopic biopsies confirm MALT/extra-nodal marginal zone lymphoma. Outline your initial assessment. (2 marks)

A

Acute Management:
Gastric MALTs can present with GI bleeding/maleana. Acute assessment of this based on endoscopy findings and patient symptoms, may require supportive management with transfusions if evidence of HB drop.

If H pylori positive- commence PPI, Clarithryomycin, amoxicillin and metronidazole

History:
Symptom review: Dyspepsia, GI bleeding, abdominal pain or discomfort, ensure no features of gastric outlet obstruction
History of H.pylori infection, history of malignancy, family history of haematological malignancies or cancer. Previous RT.
Assess co morbidities and fitness for treatment

Examination:
Assess performance status
Examine for other sites of MALT: Eyes, nose, ears, thyroid, spleen, skin
Full lymph node examination

Investigations:
Bloods: FBC, EUC, (+/- Group and Hold/coags), LDH, SPEP, hep B/c
H.pylori Testing (Breath test and on biopsy)
CT CAP or PET if available
+/- Bone marrow biopsy
Review endoscopy for tumour location, size, adverse features (bleeding, obstruction)

154
Q

In general, outline the management options for gastric MALT/extra-nodal marginal zone lymphoma and the indications for each option. (3 marks)

A

EARLY STAGE I-11( T1-3N0-1)
· H pylori eradication
o 70-95% lymphoma regression after H pylori eradication
o T11:18 predicts lack of tumour response to antibiotics
o If T11:18 negative and early Stage I-II disease, observe
· Stage I or II disease H pylori positive but T11:18 positive or H.pylori negative
o Involved site RT
o Rituximab (If Involved Site RT is contraindicated)
ADVANCED STAGE IIe-IV
· Evaluate if candidate for clinical trial (incurable with conventional therapy)
· If symptomatic (GI bleeding, bulky disease, obstructive symptoms)
o Consider palliative ISRT or RCHOP
· If asymptomatic
o Recommend best supportive care/treat on symptoms

155
Q

A fit 66-year-old male with nausea and reflux undergoes gastroscopy showing diffuse thickening of the gastric mucosal folds in the body of the stomach. Multiple endoscopic biopsies confirm MALT/extra-nodal marginal zone lymphoma. Outline your initial assessment. (2 marks)

The patient has localised gastric MALT lymphoma. Discuss a suitable radiation therapy technique and dose fractionation schedule. (3 marks)

A

PRE SIM: Acute management and stabilisation of patient, H.pylori eradication therapy, consent, pre-RT medication prescribing ondansetron 4mg pre RT
SIM: Supine in vac bag with knee blocks and ankle stocks. Arms above head. Fasted 4 hours. Attempt DIBH or 4DCT for motion management. RT tattoos
Fusion: Available scans e.g. PET
Volumes: CTV entire stomach + and involved lymph nodes. ITV using 4D CT. PTV ITV + 1cm.
Dose: 24 Gy in 12 fractions (2 Gy per fractions) prescribed as per ICRU 83. 9 fractions per fortnight. VMAT 6 MV photons.
Verification: Daily CBCT to evaluate stomach filling and target coverage
OARS: Heart Mean <8 Gy, Lungs V20<20, V5<12, Spleen <10Gy

156
Q

(2 marks) i. In general, discuss the outcomes associated with the different treatment options for localised Gastric MALT lymphoma. Include in your answer a discussion of response rates and relapse rates.
ii. Discuss the potential toxicity associated with radiation treatment in this setting and an appropriate follow up schedule. (2 marks)

A

· Early stage disease, with H Pylori eradication: 70-95% response rate
o 5 year overall survival 90%, but 5 year DFS lower ~ 75%.
o Recommend observation, as still reasonable risk of disease relapse requiring salvage treatment
· Advanced disease, considered incurable

Radiotherapy associated toxicity
Acute: Fatigue, nausea and vomiting, dyspepsia, oesophagitis.
Late: Pneumonitis, cardiac disease, oesophageal stricture, secondary malignancy
Following treatment of early disease, endoscopic assessment in 6 months. If still disease at re-evaluation, can consider ongoing observation as clinical response can take up to 18 months be be achieved.

Clinical follow up every 3 months with bloods prior for 1st 2 years, then 6 monthly up to 5 years.
As per NCNN ‘Optimal interval for follow-up endoscopy and imaging is not known. Follow-up endoscopy and imaging using the modalities performed during workup is driven by symptoms. Relapse rates are higher after treatment with rituximab and may warrant serial endoscopy.’

157
Q

A fit 48 year old woman presents with a painless 4cm mass in the left lower axilla. An excision biopsy is performed. Pathological review demonstrates a lymph node replaced by follicular lymphoma (Grade 1).

a. What further information do you require to decide on a management plan for this patient? Justify your answer. (2 marks)

A

Patient factors - to determine fitness for treatment and need of acute management if symptomatic
o Age, performance status
o Symptomatology/ B symptoms
o Hep B/pregnancy test
· Tumour factors – to determine the appropriate therapeutic options
o Burden of disease/ Stage
§ GELF criteria
§ L337 BBEAMSS
□ LDH Elevated or beta-2-microglobulin
□ 3 areas >3cm (extensive nodal disease)
□ Bulky >7cm
□ B symptoms
□ Elevated lymphocytes>5
□ Asctes/effusion
□ Marrow failure or neut <1, platelets<100
□ Symptoms -compression
□ Splenic symptoms
o Kinetics of disease
· Treatment factors – to determine if the general recommended therapeutic options are available to this patient
o Suitability for chemotherapy or radiotherapy
§ Including contra-indications
§ Previous treatment
o Access to treatment

158
Q

A fit 48 year old woman presents with a painless 4cm mass in the left lower axilla. An excision biopsy is performed. Pathological review demonstrates a lymph node replaced by follicular lymphoma (Grade 1).

b. PET CT shows avidity at the site of the excised node, with a smaller avid mass situated superiorly at the left axillary apex (Stage 1 Follicular lymphoma). (6 marks) -. Outline the treatment options available for this patient. =: If radiation therapy were to be used in her management, describe a suitable radiation therapy technique and dose fractionation schedule.

A

Options for stage 1 follicular lymphoma
· Involved site radiotherapy (Preferred)
· Involved site radiotherapy + Anti-CD20 monoclonal antibody (Rituximab) +/- RVP Chemo
o Addition of systemic therapy has not been shown to improve OS, but can improve FFS
· Anti-CD20 monoclonal antibody (Rituximab) +/- RVP Chemo alone
· Observation (not recommended)
Radiotherapy technique
· Pre-sim
o MDT Discussion
o Fertility discussion
o Smoking cessation
· Simulation
o Supine in vacbag with arms above head
o CT from C3 to below diaphragm, 2mm slices with contrast
o Fuse Pre-chemotherapy PET
· Dose prescription
o 24Gy/12# prescribed as per ICRU83
o VMAT technique, 9-10 days per fortnight
· Volumes
o GTV: Gross disease seen on CT/PET
o CTV: GTV + 2-3cm sup/inf to include entire nodal compartment
§ Include pre-chemotherapy nodal region (if received)
o PTV: CTV+7mm
· Daily CBCT for target verification
· OAR
o Bilateral lung: V20<30
o Heart Mean <5Gy
o Spinal cord Dmax <prescribed
o Brachial plexus Dmax <prescribed

159
Q

A fit 48 year old woman presents with a painless 4cm mass in the left lower axilla. An excision biopsy is performed. Pathological review demonstrates a lymph node replaced by follicular lymphoma (Grade 1). She receives radiotherapy.

Six years after obtaining a complete response to radiation therapy, the patient represents with a 3 cm painless mass in the post triangle of the right neck. A CT scan shows small volume para-aortic lymphadenopathy. The neck and para-aortic adenopathy are avid on PET CT. C. How would you manage this patient now? (2 marks)

A

· Assess pt
o Performance status
o Organ function – bloods
o Symptoms due to bulky disease
o Constitutional symptoms
o Cytopaenias
o Disease kinetics
· Repeat biopsy to exclude transformation to aggressive lymphoma

- In Stage III disease, Upfront therapy does not result in:
	○ Improved OS
	○ Or Reduction in risk of transformation to high-grade disease

· Would advocate for observation, unless symptomatic

160
Q

A fit 21 year old female is found to have a mediastinal mass. CT guided biopsy confirms Nodular Sclerosing Hodgkin’s Lymphoma. CT and PET staging shows the disease is confined to the mediastinum and left supraclavicular fossa.

a. What factors do you need to consider when deciding on this patient’s management? (3 marks)

A

Patient: fertility preservation, smoking cessation, psychosocial/AYA service
Suitability for chemo, radiation
Lung, heart function, radiosensitising syndrome
HIV
Family history/genetics
Tumour
Prognostic factors re favourable vs unfavourable:
ENE, ESR (depending on B symptoms), >3 nodal regions, bulky disease
Treatment
PET response to chemo
Late toxicities of treatment
Patient preferences
Access to treatment
Previous RT

161
Q

A 28-year-old man presents with an asymptomatic 3 cm mass in his left groin. The rest of the examination is normal. Biopsy confirms this to be a mixed cellular Hodgkins Lymphoma.

a. What further information do you require in order to decide on a management plan for this patient and justify your answer? (3 marks)

A

Management plan based on staging and prognostic risk factors. Staging grouped into Early Stage (I and II) and Advanced Stage (III and IV). If early stage prognostic factors into favourable and unfavourable (Bulky disease, Mediastinal disease ratio >0.3, ESR, Extra nodal disease, 3+ nodal sites, B symptoms).

History:
-B symptoms (Fevers, night sweats, weight loss)
-Alcohol induced lymphadenopathy/prutitis
-Previous EBV infection
-Past medical history: previous radiotherapy, chemotherapy or cancer.
-Family history of blood cancers or malignancy
-Fertility preservation,
Examination:
Lymph node examination and examination for extra nodal disease, the stem reports is normal

Investigations:
Baseline ECHO, PFTs
Bloods: HIV, Hep B/C, ESR, Albumin, Lymphocytes, LDH
Biopsy- ensure atleast a core or excisional for diagnosis: Mixed cellular type, EBV status
Staging PET scan

162
Q

A 28-year-old man presents with an asymptomatic 3 cm mass in his left groin. The rest of the examination is normal. Biopsy confirms this to be a mixed cellular Hodgkins Lymphoma.

Staging CT scan does not show any further disease. The rest of the investigations are normal. A decision is made to treat with sequential chemotherapy and radiation therapy.
c. Describe a suitable radiation treatment technique and dose prescription for this patient. (3 marks)

A

Involved site radiotherapy 20 Gy in 10 fractions to the LEFT groin
Pre Sim: Fertility discussion/management (check this was done prior to chemo), MDT discussion, identify if any issues with tolerating chemotherapy, consent
Sim: Supine, vac bag and hands on chest. Frog leg. Palpate nodal disease at SIM and wire node. 5 mm bolus. CT 2 mm slices with IV contrast
Fusion: Pre-chemo Pet
Prescription: 20 Gy in 10 fractions 2 Gy per fraction, 6 MV photons VMAT technique
Volumes: Contour pre-chemo GTV on PET, if residual disease contour post-chemo GTV. CTV defined by superior/inferior extent of pre-chemo GTV. Clip to bone, air, other structures. PTV= CTV + 1cm
OARs: Femoral heads, bladder, prostate, testes, rectum ALARA,

163
Q

A 28-year-old man presents with an asymptomatic 3 cm mass in his left groin. The rest of the examination is normal. Biopsy confirms this to be a mixed cellular Hodgkins Lymphoma.

Staging CT scan does not show any further disease. The rest of the investigations are normal. A decision is made to treat with sequential chemotherapy and radiation therapy.
How would you counsel this patient with regards to the risk of recurrence and long-term side effects? (3 marks)

A

Overall good prognosis and survival rates
Early stage disease, survival >90% . Risk of relapse/progression ~20% 5-10 years, in the case of relapse/progression further salvage treatments are effective. Hence Hodgkins Lymphoma is considered to have an excellent prognosis and survival outcomes.

Monitoring for relapse clinically
-3 month post-treatment PET
-clinical examination, bloods and review ever 3 months for 2 years, then 6 monthly until 5 years
-Annual surveillance for late effects
Long-term side effects to discuss related to groin
-Secondary malignancy risk (Increased risk with chemo and RT, estimate to by 15% @15 years), infertility, femoral head #

164
Q

A 40-year-old man with acute myeloid leukaemia is referred for total body irradiation (TBI) prior to a bone marrow transplant for relapsed acute myeloid leukaemia.

a. What would you explain to the patient about the rationale for TBI? (2 marks)

A

Marrow transplant needs conditioning, rationale is to eliminate disease, and deplete the bone marrow to allow engraftment.

RT penetrates sanctuary sites and areas with poor blood supply (CNS, testes, orbits), as well has no cross resistance with chemotherapy.

165
Q

A 40-year-old man with acute myeloid leukaemia is referred for total body irradiation (TBI) prior to a bone marrow transplant for relapsed acute myeloid leukaemia.

b. Describe a suitable radiation therapy technique and dose fractionation schedule for TB1. In your answer, include the technical difficulties that arise in TBI and how these are overcome. (5 marks)

A

6MV photon extended SSD, opposed lateral at low dose rate
12Gy in 6 fraction treated BD (6 hours apart), prescribed to midpoint
After conditioning chemo and prior to transplant.

Technical difficulties:
-encompass entire body within a field (use extended field to increase field size and decrease beam attenuation due to inverse square law
-achieve dose uniformity within 10% -body has varying widths
Long delivery time and strict scheduling re chemo/transplant.

Technique
Sim/mock up –with physics
SSD at 4m, patient on couch with knees bent, arms by side (to shield lungs). CT sim 2mm slices entire body

Colimator 45degrees to maximise horizontal field length, light field projected.

Perspex beam spoiler on side of couch –reduce skin sparing effect

Planning:
Tissue compensators –head (perspex), neck
+-lungs +- kidney

Treatment:
Premedication with dexamethasone/ondansetron
Target verification: patient within light field
Dose verification with multiple TLD (bilateral head, neck, chest, hip, groin) and midline ion chamber (between thighs).
Each beam split into 2 phases, first phase 30s to double check dosimetry invivo.

Turn patient/bed 180degrees for second field.

166
Q

What are the potential acute and late toxicities of TBI? (3 marks)

A

RT toxicities combine with chemotherapy toxicities, can be difficult to separate.
Acute: pancytopenia, GIT –nausea, vomiting, diarrhoea, sialdenitis, venoocclusive disease (liver)
Subacute: pneumonitis –dose limiting toxicity

Late:
Cataracts
Gonadal failure
Hypothyroidism
Osteoporosis
Cardiac failure
Secondary malignancy

167
Q

When treating patients with Hodgkin’s disease in general, how would you reduce the treatment and patient related risk factors?

A

Treatment factors:
- Before/during treatment: minimise dose to heart as much as possible
○ MHD ALARA (noting every 1Gy increases risk of cardiac event)
§ Aim for mean <4-5Gy
§ Contour Substructures: LV Mean <8Gy, Valves Dmax <25, Vessels mean <7Gy, minimse Dmax to major vessels
○ Consideration of alternative treatments (including chemo alone vs. RT if heart DVHs are unacceptable eg. MHD >15Gy)
○ Consideration to alternative systemic therapies if baseline risk of cardiac toxicity high
Ie. Avoid anthracycline chemo
○ DIBH/ positioning techniuqes
- After treatment: Regular surveillance and optimisation of cardiac function if impairment detected

Patient factors:
- Optimise cardiovascular risk factors
○ Encourage physical activity
○ Stop smoking
- Surveillance and early intervention: Echo/Coronary artery calcium core 10 years

Referral to late effects clinic

168
Q

With patients undergoing Hodgkin Lymphoma treatment what education would you give the patient on risk reduction strategies?

A

Cardiac:
- Optomise cardiac risk factors - diet, physical acrtivity, smoking
- Treatment of HTN, Hyperchol, DM etc
- Maintain normal weight
- Regular cardic review 5-10 years ECHO
Pulmonary:
- Stop smoking
Thyroid
- Annual TFTs, thyroid exam
Secondary malignancy:
- Consider low dose CT in high risk pts for lung ca
- Stop smoking
- Regular breast screening 8-10 years after RT or 40 which every is earlier
- MMG/USS/MRI (increases sensitivity) annually (can alternate 6 monthly with modalities)
- Regular self examination/ breast awareness, and physician examinations

169
Q

Describe the pathogenesis of lytic metastases in multiple myeloma. (2 marks)

A

Uncoupling of bone-remodelling process. Myeloma cells interacting with the bone microenvironment leads to activation of osteoclasts and suppression of osteoblasts, resulting in bone loss. The Myeloma cells achieve this through several intercellular and intracellular signalling cascades such as; RANK/RANKL/OPG, NOTCH and Wnt.
Osteoclasts promote plasma cell survival via interleukin 3, 6, TNF alpha