MM: interpreting cytogenetic results for clinical practice · Clinical impact –IgH translocations...

Post on 09-Jul-2020

1 views 0 download

Transcript of MM: interpreting cytogenetic results for clinical practice · Clinical impact –IgH translocations...

MM: interpreting cytogenetic results for clinical practice

Dr Sally MooreConsultant Haematologist

Royal United Hospital, BathChurchill Hospital, Oxford

This medical education meeting was organised and funded by Janssen-Cilag Ltd. EM-12168 | May 2019

▼ This medicinal product is subject to additional monitoring. This will allow quick identification of new safety information. Healthcare professionals are asked to report any suspected adverse events.

Click below for prescribing information and adverse events reporting:For daratumumab▼click here

For bortezomib click here

This meeting was organised and funded by Janssen

The slide content has been reviewed by Janssen to ensure compliance with the ABPI Code of Practice

for the Pharmaceutical Industry

The faculty may express personal opinions that are not necessarily shared by Janssen

Adverse events should be reported.▼Reporting forms and information can be found at

www.mhra.gov.uk/yellowcard or search for MHRA Yellow Card in the Google Play or Apple App Store.

Adverse events should also be reported to Janssen-Cilag Limited on 01494 567447 or at

dsafety@its.jnj.com

Any adverse events presented in this slide deck (related to Janssen products) have been reported to

drug safety

Prescribing Information is available at this meeting

Janssen-Cilag Ltd, 50–100 Holmers Farm Way, High Wycombe, Buckinghamshire HP12 4EG

Congress sponsorship: Amgen, Celgene, Takeda

Advisory: Amgen, Celgene, Takeda

Educational activities: Amgen, Janssen, Roche

Disclosures – Dr Sally Moore

Learning objectives

• To understand what CAs are and how they may affect prognosis

• To explore treatment decisions regarding CAs through the use of patient case studies

• To explore factors that might affect future considerations regarding CAs in multiple myeloma

CA: cytogenetic abnormalities

Disease profiling

Landgren O, Morgan GJ. Clin Cancer Res. 2014; 20:804–13.FISH: fluorescence in situ hybridisation;

MGUS: monoclonal gammopathy of undetermined significance; miRNA: micro ribonucleic acid.

Frequency of CAs

Sonneveld P, et al. Blood. 2016; 127:2955–62.

Primary and secondary genetics events that can be identified by FISH

Ig: immunoglobulin; NA: not applicable.

Clinical impact – IgH translocations

• t(4;14) = ↓ PFS/OS1

Bortezomib improves survival vs vincristine2

↑ OS with HDT and tandem ASCT3

• t(14;16) and t(14;20) = predictors of poor outcome4

• t(11;14) associated with CD20 expression and LPL morphology. Reported as favourable in some studies, no impact in others5,6

• In general, t(6;14), t(11;14), gain(5q) and hyperdiploidy do not confer poor prognosis

1. Gertz MA, et al. Blood. 2005; 106:2837–40; 2. Sonneveld P, et al. J Clin Oncol. 2012; 30:2946–55; 3. Moreau P, et al. Leukemia. 2007; 21:2020–4; 4. Boyd KD, et al. Leukemia. 2012; 26:349–55; 5. Fonseca R, et al. Blood. 2002; 99:3735–41; 6. Avet-Loiseau H, et al. Blood. 2007; 109:3489–95.

ASCT: autologous stem cell transplantation;HDT: high-dose therapy; LPL: lymphoplasmacytic lymphoma;

OS: overall survival; PFS: progression-free survival.

Clinical impact – genomic imbalance

General• Due to selection of subclonal disease, the prognostic impact of CA may vary from diagnosis to refractory disease1

Diploidy• Hyperdiploidy (50% of NDMM) = ↑ PFS/OS2. Co-existing hyperdiploidy may not abrogate poor prognosis of adverse CA3

• Hypoploidy is regarded as a poor prognostic CA4

• t(4;14) PFS and OS can be negatively affected by hypodiploidy4

Deletions• Adverse impact of del(13q) is associated with del(17p) and t(4;14). Del(13q) alone does not confer poor survival1,5

• Del(17p) or del(17) has a negative impact on PFS/OS. DelTP53 induces clonal immortalisation and survival of tumour cells6

• What minimum % del(17p) cells = adverse prognosis? Minimums of 20% and 60% have been suggested4,5

• In solitary plasmacytoma or extramedullary disease, del(17p) may occur more frequently7

Gains• >3 copies of 1q = worse outcome (dosage effect)5

• Gain(1q) often associated with del(1p32), which confers poor prognosis8

NDMM: newly diagnosed multiple myeloma.

1. Walker BA, et al. J Clin Oncol. 2015; 33:3911–20; 2. Neben, et al. Haematologica. 2010; 95:1150–7; 3. Pawlyn C, et al. Blood. 2015; 125:831–40; 4. Hebraud, et al. Blood. 2015; 125:2095–100; 5. Boyd KD, et al. Leukemia. 2012; 26:349–55; 6. Teoh PJ, et al. Leukemia. 2014; 28:2066–74; 7. Billecke, et al. Br J Haematol. 2013; 161:87–94; 8. Hebraud B, et al. Leukemia. 2014; 28:675–9.

Clinical impact – multiple abnormalities

• Adverse IgH translocation: 62% have gain(1q) compared with 32.4% in controls1

• Del(17p) frequency is similar in patients without adverse IgH translocations1

• Triple combination of an adverse IgH translocation, gain(1q) and del(17p) had OS of 9.1 months, demonstrating impact of multiple adverse CAs on OS1

• In 110 patients with either t(4;14) or del(17p), 25 had both abnormalities2

• t(4;14): ↓ PFS seen in del(1p32), del(22q), and/or structural changes

↓ OS seen in del(13q14), del(1p32) and higher number of CAs2

• del(17p): ↓ PFS seen in del(6q)

↓ OS seen in del(1p32)2

1. Boyd KD, et al. Leukemia. 2012; 26:349–55; 2. Hebraud B, et al. Blood. 2015; 125:2095–100.

Clinical impact – good and adverse

• Hyperdiploidy: gain5(q31), trisomies 3 and 5 confer a favourable prognosis1,2

• In the Myeloma IX study, 58% had hyperdiploidy. Of these, 61% had ≥1 adverse lesion [t(4;14), t(14;16), t(14;20), gain1q or del(17p)]3

• PFS and OS were shorter in those with hyperdiploidy plus adverse lesion vshyperdiploidy alone (PFS: 23 vs 15.4 months; OS: 60.9 vs 35.7 months)3

• Alternatively, presence of hyperdiploidy did not change the outcome in patients with an adverse lesion3

• Trisomies in patients with t(4;14), t(14;16), t(14;20) or delTP53 reduced their adverse impact4

1. Hebraud B, et al. Blood. 2015; 125:2095–100; 2. Avet-Loiseau H, et al. J Clin Oncol. 2009; 27:4585–90;3. Morgan, et al. Blood. 2012; 119:7–15; 4. Kumar S, et al. Blood. 2012; 119:2100–5.

FISH – what is high risk?

• IMWG 2009 – at least one of: del(17p), t(4;14), t(14;16)1

• IMWG 20162 –

• Mayo Clinic – as above including hypodiploidy and t(14;20)3

• MRC IX – adverse lesions include t(4;14), t(14;16), gain(1q), del(13q) and del(17p)

• FR: absence of genetic lesion

• IR: one adverse CA

• HR: >1 adverse CA

• UHR: ≥3 adverse CAs4

1. Fonseca, et al. Leukemia. 2009; 23:2210–21; 2. Sonneveld P, et al. Blood. 2016; 127:2955–62;3. Stewart, et al. Leukemia. 2007; 21:529–34; 4. Boyd KD, et al. Leukemia. 2012; 26:349–55.

FR: favourable risk; GEP: gene expression profiling; HR: high-risk;IMWG: International Myeloma Working Group; IR: intermediate risk;

MRC: Medical Research Council; UHR: ultra high risk.

FISH cut-off

• IFM1

Percentage of plasma cells with adverse FISH correlates with outcome:

Del(13q) – cut-off 74% OS rate at 41 months 59% vs 80%

Del(17p) – cut-off 60% OS rate at 22.4 months 50% vs 75%

• IMWG R-ISS:2

FISH analyses were performed in a few European laboratories. Despite interlaboratory variability, the analyses of p53 deletion, t(4;14), and t(14;16) were commonly included in each multiple myeloma panel and tested using commercial probes. Of note, however, the cut-off levels were not identical, ranging from 8% to 20% for numerical aberrations and from 10% to 15% for immunoglobulin H translocations

Patients were considered positive for a given CA when it was present in a percentage higher than the cut-off threshold, defined by each local laboratory2

• Eurozone: Spanish 20%, French 60%, UK – number of abnormalities

1. Avet-Loiseau H, et al. Blood. 2007; 109:3489–95; 2. Palumbo A, et al. J Clin Oncol. 2015; 33:2863–9.IFM: Intergroupe Francophone du Myélome; R-ISS: Revised International Staging System.

Double-hit myeloma

Walker BA, et al. Leukemia. 2019;33:159–70. ISS: International Staging System.

• N=1,273, whole genome and exome data

• HR = ISS III with either bi-allelic TP53 inactivation or ≥4 copies of 1q (6.1% of patients)

100

80

60

40

20

0

0 12 24 36 48 60

OS

(%)

100

80

60

40

20

0

0 12 24 36 48 60

Months

PFS

(%

)

Log-rank p-value <0.0001

MonthsDeaths / N 18-month estimate

IMWG: low/standard risk, RP-Risk: Low Risk 21 / 357 96% (94, 98)

IMWG: low/standard risk, RP-Risk: Intermediate Risk 59 / 296 85% (81, 90)

IMWG: low/standard risk, RP-Risk: Double Hit 10 / 24 73% (53, 93)

IMWG: high risk, RP-Risk: Low Risk 6 /30 88% (74, 100)

IMWG: high risk, RP-Risk: Intermediate Risk 6 / 53 94% (86, 100)

IMWG: high risk, RP-Risk: Double Hit 16 / 24 37% (16, 58)

Log-rank p-value <0.0001

Events / N 18-month estimate

IMWG: low/standard risk, RP-Risk: Low Risk 65 / 357 87% (84, 91)

IMWG: low/standard risk, RP-Risk: Intermediate Risk 126 / 296 67% (62, 73)

IMWG: low/standard risk, RP-Risk: Double Hit 15 / 24 44% (23, 65)

IMWG: high risk, RP-Risk: Low Risk 10 /30 69% (51, 88)

IMWG: high risk, RP-Risk: Intermediate Risk 21 / 53 74% (61, 87)

IMWG: high risk, RP-Risk: Double Hit 18 / 24 35% (15, 54)

IMWG Guidelines 2016• Consensus updates: abnormalities such as t(4;14), del(17/17p), t(14;16), t(14;20),

non-hyperdiploidy and gain(1q) confer poor prognosis. The prognosis of patients showing

these abnormalities may vary with the choice of therapy

• Treatment strategies have shown promise for HR cytogenetic diseases, such as PI +

lenalidomide combination, pomalidomide, double ASCT + PI, or IT with lenalidomide and

pomalidomide

• Bortezomib and carfilzomib appear to improve CR, PFS and OS for t(4;14) and del(17p)

• Lenalidomide may be associated with improved PFS in t(4;14) and del(17p)

• Patients with multiple adverse CAs do not benefit from these agents

• FISH data are implemented in the R-ISS for risk stratification

Sonneveld P, et al. Blood. 2016; 127:2955–62. CR: complete response; IT: immunotherapy; PI: proteasome inhibitor.

R-ISS 2005–2012

n=3,060 of 4,4451

HR: del(17p) and/or t(4;14)1

Data are generated from 3,060 patients from 11 international trials conducted from 2005 to 2012 that were pooled and analysed by the IMWG.1

1. Palumbo A, et al. J Clin Oncol. 2015; 33:2863–9; 2. Dispenzieri A. Hematology Am Soc Hematol Educ Program. 2016; 2016:485–94.

Figure adapted from Dispenzieri 2016.2

LDH: lactate dehydrogenase; Nml: normal; SR: standard risk.

R-ISS

Palumbo A, et al. J Clin Oncol. 2015; 33:2863–9. CI: confidence interval; F: female; HR: hazard ratio; M: male; NR: not reached.

• 69-year-old lady. PMH of well-controlled asthma; family history of breast cancer

• November 2016: numb lip – mandibular plasmacytoma

• PET-CT: extremely extensive disease. Cauda equina at L2–4

• IgG lambda paraprotein 33 g/l, Hb 76 g/l

• BMT November 2016: 100% plasma cells

• FISH: gain(1q), t(14;16)

• R-ISS III, ISS IIIBMT: bone marrow transplant; CT: computed tomography; Hb: haemoglobin;

PET: positron emission tomography; PMH: previous medical history.

Case study 1

1. CTD

2. Bortezomib-based regimen

3. Myeloma XI

4. CARDAMON trial (KCD)

5. Other

CTD: cyclophosphamide, thalidomide, dexamethasone; KCD: carfilzomib, cyclophosphamide, dexamethasone; Tx: treatment.

Which initial induction Tx?

• CTD vs CVAD (up to six cycles) and ASCT

• FISH – 50%

• n=293 adverse CA [t(4;14), t(14;16), t(14;20) gain(1q), del(1p32) del(17p)], n=333 favourable

Morgan GJ, et al. Haematologica. 2012;97:442–50.

PFS = 20 months with adverse interphase FISH vs 34 months with favourable interphase FISH

Transplant-eligible – CTD Myeloma IX

CVAD: cyclophosphamide, vincristine, doxorubicin, dexamethasone.

Transplant-eligible – bortezomib induction

Meta-analysis: n=1,572; four studies

All-comer CR rates 33–55% for bortezomib-based induction vs 20–35% for non-bortezomib-based induction

Sonneveld P, et al. J Clin Oncol. 2013; 31:3279–87.

PFS (intent-to-treat)

Time (years)

PFS OS (%)

All t(4;14) del(17p) All t(4;14) del(17p)

VD1,2

3/4 36/12 28/12 14/12* 81 63 49

VAD1,2 30/12 16/12 NR 77 32 50

VTD3

368% 31% NR 86 NR NR

TD3 56% 63% NR 84 NR NR

*Event-free survival.1. Harousseau JL, et al. J Clin Oncol. 2010; 28:4621–9; 2. Avet-Loiseau H, et al. J Clin Oncol. 2010; 28:4630–4;3. Cavo M, et al. Lancet. 2010; 376:2075–85.

Transplant-eligible – bortezomib induction

NR: not reported; TD: thalidomide, dexamethasone; VAD: vincristine, doxorubicin, dexamethasone;

VD: bortezomib, dexamethasone; VTD: bortezomib, thalidomide, dexamethasone.

Jackson G, et al. Presentation at ASH 2018. Abstract 302 and oral presentation.

Myeloma XI

Bortezomib-based

regimen

No bortezomib-

based regimen

Maximum response

PDSD

MRPR

CRVGPR

R 1:1

R 1:1

R 1:1

• Primary endpoints: PFS and OS for each randomisation

• Median follow-up of 34.5 monthsCRD: cyclophosphamide, lenalidomide, dexamethasone; K: carfilzomib;

MR: minimal response; PD: progressive disease; PR: partial response; R: randomised; SD: stable disease; TE: transplant-eligible; VGPR: very good partial response.

Myeloma XI

Jackson G, et al. Presentation at ASH 2018. Abstract 302 and oral presentation. BM: bone marrow.

Myeloma XI

Jackson G, et al. Presentation at ASH 2018. Abstract 302 and oral presentation.

• SR: absence of any high-risk lesions

• HR: presence of any one of t(4;14), t(14;16), t(14;20), del(17p) or gain(1q)

• UHR: presence of more than one lesion

HR: hazard ratio.

CARDAMON trial• CARDAMON: KCD-ASCT-Kd vs KCD-Kd

• FORTE: n=474

Gay F, et al. Presentation at ASH 2018. Abstract 121 and oral presentation;ClinicalTrials.gov. NCT02315716. Available at: https://clinicaltrials.gov/ct2/show/NCT02315716 [accessed January 2019].

KRd: carfilzomib, lenalidomide, dexamethasone; MRD: minimal residual disease; neg: negative;

sCR: stringent complete response.

Transplant-eligible – ASCT

• RV-MM-EMN-441 (n=127)

• Rd induction – RCD vs ASCT (some tandem) – lenalidomide vs lenalidomide/prednisone

• HR FISH: three-year PFS 43% (ASCT) vs 17% (RCD); three-year OS 78% vs 67%

Dispenzieri A. Hematol Am Soc Hematol Educ Program. 2016; 2016:485–94; Greipp PR, et al. J Clin Oncol. 2005; 23:3412–21; Avet-Loiseau H, et al. Leukemia. 2013; 27:711–7; Palumbo A, et al. J Clin Oncol. 2015; 33:2863–9; Gay F, et al. Lancet Oncol. 2015; 16:1617–29.

RCD: lenalidomide, cyclophosphamide, dexamethasone; Rd: lenalidomide, dexamethasone; R/P: lenalidomide/prednisone.

Tandem ASCT• Pooled analysis of four trials, n=606, 23% of patients with t(4;14)/del(17p)1

• All received bortezomib induction. Single ASCT n=254, tandem=3521

• <CR post-bortezomib induction resulted in longer PFS and higher OS rate with tandem ASCT vs single ASCT1

• PFS: 42 months vs 21 months; OS: 67 months vs 31.5 months1

1. Cavo M, et al. Blood. 2013; 122: Abstract 767; 2. Cavo M, et al. Blood. 2012; 120:9–19.

• GIMEMA MM-BO20052

• VTD or TD induction, tandem ASCT, consolidation. Patients with t(4;14) on bortezomib-based induction had almost identical PFS as SR patients

PFS for patients with HR cytogenetics and who failed CR after bortezomib-based induction regimens

0.75

0.50

0.25

0.00

Months240 12 36 48

HR=0.41, p=0.006

Single ASCT

Double ASCT

1.00

HR=0.41, p=0.006

Single ASCT

Double ASCT

Months240 12 36 48

0.75

0.50

0.25

0.00

OS for patients with HR cytogenetics and who failed CR after bortezomib-based induction regimens1.00

EMN02/HO95

• Three-year PFS:

• Single ASCT 64%, double ASCT 73%; HR 0.7; p=0.04

• ↑ PFS with double ASCT in all groups including:

• High-risk cytogenetic profile [cyto-3; t(4;14), t(14;16), or del(17p)]:

Hazard ratio 0.42 (95% CI 0.21–0.84; p=0.014)

• Highest-risk cytogenetic profile [(cyto-5; amp(1q), del(1p) and t(4;14),

t(14;16) or del(17p)]:

Hazard ratio 0.65 (95% CI 0.42–1.01; p=0.059)

Cavo M, et al. Presentation at ASH 2017. Abstract 401.

What would you do?

Treatment

DXT: radiotherapy; VTD: bortezomib, thalidomide, dexamethasone.

Par

apro

tein

g/l

50

40

30

20

10

0

01/09/2016 30/11/2016 01/03/2017 01/06/2017 31/08/2017 29/11/2017

Date

IMWG recommendations 2016

• Thalidomide does not abrogate the effect of t(4;14), t(14;16), t(14;20), del(17), del(17p) and gain(1q) in transplant-eligible patients

• Bortezomib partly overcomes the effect of t(4;14) and possibly del(17p) on CR, PFS and OS. There is no effect in t(4;14) and del(17p) in transplant-eligible patients

• Lenalidomide partly improves the adverse effect of t(4;14) and del(17p) on PFS, but not OS, in transplant-eligible patients

• Combining a PI with lenalidomide/dexamethasone greatly reduces the adverse effect of t(4;14) and del(17p) on PFS in NDMM. Carfilzomib/lenalidomide seems effective in patients with HR cytogenetics

• ASCT is standard therapy for transplant-eligible patients with NDMM. It contributes to improved outcomes across prognostic groups

• Double HDT/ASCT combined with a PI may improve PFS in t(4;14) or del(17p), or both. HDT plus double ASCT is recommended for patients with HR cytogenetics

• Allogeneic SCT or tandem auto-allo-SCT may improve PFS in t(4;14) or del(17p)

Sonneveld P, et al. Blood. 2016; 127:2955–62.

Maintenance?

1. None

2. Thalidomide

3. Lenalidomide

4. Carfilzomib

5. Ixazomib

6. Other

Maintenance post-ASCT – thalidomide

MRC-IX

Morgan, et al. Blood. 2012; 119:7–15.

Pat

ien

ts (

%)

Pat

ien

ts (

%)

PFS (months) OS (months)

CTDa: cyclophosphamide, thalidomide, dexamethasone attenuated; ITT: intent-to-treat; MP: melphalan, prednisone.

Maintenance post-ASCT – lenalidomide

• Meta-analysis, lenalidomide maintenance vs placebo/observation post-ASCT, n=1,2081

• Improved PFS if HR for lenalidomide maintenance vs observation, but no improvement in OS (most patients did not have FISH data)

• Myeloma XI2

• FISH data available: 22.8% lenalidomide maintenance arm, 30.1% observation arm

1. McCarthy PL, et al. J Clin Oncol. 2017; 35:3279–89; 2. Jackson G, et al. Blood. 2016; 128: Abstract 1143.

Maintenance post-ASCT – ixazomib

• TOURMALINE MM3

• Ixazomib maintenance for 26 cycles vs placebo

• Discontinuation rates similar in both arms

• PFS 26.5 months for ixazomib vs 21.3 months for placebo;benefit seen in HR patients and those ISS III

• Maintenance in patients not eligible for transplant pending

Dimopoulos MA, et al. Presentation at ASH 2018. Abstract 301.

Continuous Tx post-ASCT (+/- consolidation) KRD

• KRD x 4 – ASCT – KRD x 4 – KRD reduced dose to C18

• sCR: 20% post-ASCT, 69% post-4 x consolidation, 82% post-KRD maintenance

• MRD-negative: 66% at C8, 71% C18

• Two-year PFS: 97%

• Two-year OS: 99%

• No difference in outcomes with HR and SR

Zimmerman T, et al. Blood. 2016; 128:675. C: cycle.

Allogeneic SCT

• Limited data

• n=73, auto-allo tandem SCT1

• 21.9% had t(4;14)/del(17p); five-year PFS 29% equivalent to non-HR group

• n=143 (1999–2008) retrospective, multicentre analysis2

• HR vs normal: three-year OS 45% (HR) vs 39% (normal)

1. Kröger N, et al. Biol Blood Marrow Transplant. 2013; 19:398–404; 2. Roos-Weil D, et al. Haematologica. 2011; 96:1504–11.

VTD and DXT

HarvestASCT

CD

IRd

Daratumumab

PCd

CD: cyclophosphamide, dexamethasone;IRd: ixazomib, lenalidomide, dexamethasone;

PCd: pomalidomide, cyclophosphamide, dexamethasone.

Progress update

Par

apro

tein

g/l

50

40

30

20

10

001/09/2016

Date

30/11/2016 01/03/2017 01/06/2017 31/08/2017 29/11/2017 28/02/2018 31/05/2018 30/08/2018

Case study 2

• 70-year-old lady. PMH breast cancer (lumpectomy, DTX, tamoxifen), complex nerve pain, BCC nose and arm, recurrent UTIs

• March 2014: incidental finding, IgG kappa 14 g/l

• SS: NAD, MRI spine: degenerative changes only

• Hb 120, normal U&Es

• ISS I

• BM: 17% plasma cells on aspirate

• FISH: gain(1q), gain(4p) and gain(11q)

BCC: basal cell carcinoma; MRI: magnetic resonance imaging; NAD: no abnormality detected; SS: skeletal survey;

U&E: urea and electrolytes; UTI: urinary tract infection.

Which initial induction Tx?

1. CTD

2. Bortezomib-based regimen

3. Myeloma XI

4. Other

Transplant-ineligible – bortezomib

VISTA1

• VMP vs MP – normalised outcomes with HR

1. Mateos MV, et al. J Clin Oncol. 2010; 28:2259–66. MP: melphalan, prednisone; VTP: bortezomib, thalidomide, prednisone.

Figure adapted from Mateos, et al. 20101

OS in patients treated with VMP

Transplant-ineligible – bortezomib

n=902, HR as per IMWG (27% HR)

VMP plus maintenance vs VMP (GIMEMA-MM-03-05) and MPR vs CPR (EMN01)

Larocca A, et al. Blood. 2017; 130:744.BORT: bortezomib; CPR: cyclophosphamide, prednisone, lenalidomide; HiR: high-risk;

LEN: lenalidomide; MPR: melphalan, prednisone, lenalidomide; PS: performance status; StR: standard-risk.

PFS

Months Months

OS

OverallSex

FemaleMale

Age≤75 years>75 years

FISHStRHiRMissing

ISSIIIIII

Karnofsky PS90–10070–8950–69

LDH≤450>450Missing

Hazard ratio (95% CI) Interaction-p

0.76 (0.66–0.88)

0.78 (0.64–0.95)0.74 (0.61–0.90)

0.74 (0.63–0.88)0.81 (0.62–1.06)

0.87 (0.72–1.05)0.54 (0.41–0.72)0.78 (0.59–1.04)

0.85 (0.65–1.12)0.74 (0.60–0.91)0.71 (0.54–0.94)

0.69 (0.56–0.84)0.83 (0.67–1.02)0.87 (0.56–1.34)

0.78 (0.67–0.92)0.68 (0.44–1.05)0.69 (0.48–0.98)

0.66

0.55

0.01

0.62

0.34

0.71

0.41 1 1.34

Transplant-ineligible – lenalidomide

• Lenalidomide/dexamethasone – does not normalise HR FISH1,2

• FIRST3

• MPT vs lenalidomide/dexamethasone

• Lenalidomide/dexamethasone confers no advantage with HR FISH

• Myeloma XI4

1. Kapoor, et al. Blood. 2009; 114:518–21; 2. Jacobus, et al. Br J Haematol. 2011; 153:340–8; 3. Avet-Loiseau. Blood. 2015; 126: Abstract 730; 4. Jackson G, et al. Blood. 2018; 128: Abstract 1143. MPT: melphalan, prednisone, thalidomide.

Progress update

Complicated by pseudo-obstruction, urosepsis, D and V, AKI

Hb 78, WCC 6.1, neuts 4.3, plts 243BM 30% plasma cells

What would you do?

Par

apro

tein

g/l

AKI: acute kidney injury; D and V: diarrhoea and vomiting; neuts: neutrophils; plts: platelets; WCC: white cell count.

01/06/2015 30/11/2015 01/06/2016

0

40

30

20

10

Progress update

Cyclophosphamide omitted for last two cycles Vomiting, pneumococcal sepsis

Par

apro

tein

g/l

What would you do?

01/06/2015 30/11/2015 01/06/20160

30/11/2016 02/06/2017 01/12/2017

40

30

20

10

VMP x8

CDTa

IRD (PAS)

Par

apro

tein

g/l

01/06/2015 30/11/2015 01/06/2016

0

30/11/2016 02/06/2017 01/12/2017

40

30

20

10

03/06/2018 02/12/2018 04/06/2018

PAS: patient access scheme.

Progress update

Case study 3

• 58-year-old nurse consultant. PMH Addison’s disease, idiopathic urticaria, lichen sclerosis, osteopenia

• June 2015: lytic lesion R femoral diaphysis ‘thigh pain’

• SS:

• Hb 126, Cr 103, Ca 2.54

• Paraprotein IgA lambda 19 g/l

• ISS I

• BM 60% plasma cells

• FISH t(4;14), gain(1q)

CA: calcium; Cr: creatinine.

Initial treatment

IgA levels

What would you do?

25

20

15

10

5

0

01/06/2015 30/11/2015

Which re-induction Tx?

1. Induction with CTD and second ASCT

2. ACCORD trial with plan for second ASCT

3. PI-based regimen

4. Other

Myeloma X

Cook G, et al. Lancet Oncol. 2014; 15:874–85.

Myeloma X

Included: • Progression ≥12/12

Exclusion criteria:• Tx for relapsed disease

• ECOG PS 3–4

• Grade 2 peripheral neuropathy

• Comorbidity precluding ASCT

1. Cook G, et al. Lancet Oncol. 2014; 15:874–85. ECOG: Eastern Cooperative Oncology Group; TTP: time to progression.

Table adapted from Cook, et al. 20141

Myeloma X

Patients missing from the table received no treatment; *Includes both sCR and CR.†Early death after induction was defined as death between registration and up to and including 21 days after the final cycle commenced; early death after randomised treated was defined as death between randomisation and up to and including 100 days after randomisation.Cook G, et al. Lancet Oncol. 2014; 15:874–85.

Best response in all registered patients after induction and in all randomly assigned patients after randomised treatment

Response after induction (n=297)

Myeloma X

OS 67 vs 52/12

Cook G, et al. Lancet Oncol. 2014; 15:874–85.

Subgroup analysis of TTPOS in the intent-to-treat population

Myeloma XII – ACCoRd

• RRMM post-ASCT relapse: • ITD-ASCT vs ITD-ASCT(augmented)

• Ixazomib vs observation post-ASCT maintenance

• 68.2% SR, 24.2% HR, 6.1% UHR at relapse

• ORR did not differ by genetic risk (SR/HR: ORR 57.8% vs 50.0%, p=0.55)

• Previous PI exposure important (exposed vs naïve: ORR 57.8% vs 75.9%, p=0.03)

• PD more commonly seen in PI-exposed (18.6%) and HR (18.8%) vs overall population (11.9%)

Cook G, et al. Presentation at ASH 2018. Abstract 255.ITD: ixazomib, thalidomide, dexamethasone;

ORR: overall response rate; RRMM: relapsed/refractory multiple myeloma.

ReLApsE – ASH 2018• n=282

• Arm A: Rd continuous, Arm B: Rd x3 – ASCT #2 – R maintenance

• Arm B: 29.5% did not receive planned ASCT

• HR FISH: A: 31.6%; B: 42.9%

• ORR at LM analysis (post C5): A: 69.6%; B: 82.3%

• Multivariate LM analysis: superior PFS (hazard ratio 0.6, p=0.01) and OS (hazard ratio 0.39, p=0.006)

• LR FISH and R-ISS I demonstrated superior OS with B over A. Benefit not seen with HR FISH or R-ISS II/III

Arm A Arm B

ORR 74.6% 77.9%

≥VGPR 47.1% 49.3%

PFS 18.8 months 20.7 months

OS NR 62.7 months

LM analysis Arm A Arm B p-value

PFS 20.1 months 23.3 months 0.09

OS NR 57 months 0.046

Goldschmidt H, et al. Presentation at ASH 2018. Abstract 253 and 254. LM: landmark; LR: low risk; R: lenalidomide.

Progress update

VTD

ASCT

ITD (Myeloma XII) DT-PACE

Rd

40

30

20

10

001/06/2015 30/11/2015 01/06/2016 30/11/2016 02/06/2017 01/12/2017 03/06/2018 02/12/2018 04/06/2019

25

20

10

001/06/2015 30/11/2015 01/06/2016 30/11/2016 02/06/2017 01/12/2017 03/06/2018 02/12/2018 04/06/2019

15

5

What would you do?

Progress update

VTD

ASCT

ITD (Myeloma XII) DT-PACE

RD

40

30

20

10

001/06/2015 30/11/2015 01/06/2016 30/11/2016 02/06/2017 01/12/2017 03/06/2018 02/12/2018 04/06/2019

25

20

10

001/06/2015 30/11/2015 01/06/2016 30/11/2016 02/06/2017 01/12/2017 03/06/2018 02/12/2018 04/06/2019

15

5

DT-PACE: dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide, etoposide.

Which subsequent Tx?

1. RD

2. IRD

3. Daratumumab

4. Pomalidomide-based triplet

5. Pomalidomide (+/- clarithromycin)

6. Pomalidomide (+/- cyclophosphamide)

7. Bortezomib-based regimen

Principles of treating HR disease

• Accurately identifying patients

• Rapid disease control

• Scheduling

• Tailoring treatment to disease biology

• Avoid alkylating agents

• Aim to achieve outcomes similar or ‘less than expected’ difference to SR disease

Dr Sally Moore, personal communication.

CA considerations

• Most labs only test FISH according to R-ISS

• Sample quality is essential

• Much unknown re. confounding factors

• Role of repeat FISH at relapse – clonal evolution

• Limited by Tx algorithm/NICE appraisals

• Role of clinical trials

• Balance of toxicity with response

• Elderly add in third drug to doublet if issues with tolerability?

Dr Sally Moore, personal communication.

2018 updated guidelines on diagnosis, risk stratification and management

Rajkumar SV, et al. Am J Hematol. 2018; 93:1091–110.

Non-transplant-eligible

mSMART risk stratification

Bortezomib-based regimen

Bortezomib-based regimen x 3–4 cycles

PI-based maintenance

Bortezomib-based regimen

Risk groupPercentage of newly diagnosed patients with the abnormality

SR 75%

Trisomies

t(11;14)

t(6;14)

IR 10%

t(4;14)

Gain(1q)

HR 15%

t(14:16)

t(14;20)

del(17p)

Transplant-eligible

PI-based maintenance

PI-based maintenance

Thank yousallymoore5@nhs.net