28 november 2022: Zie ook dit artikel: https://kanker-actueel.nl/isatuximab-naast-pomalidomide-dexamethason-geeft-betere-overall-overleving-mediaan-69-maanden-dan-alleen-pomalidomide-dexamethason-bij-patienten-met-multiple-myeoloma-kahler-botkanker-die-zwaar-voorbehandeld-waren.html

28 november 2022: Bron: The Lancet Published:November, 2022

Toevoeging van isatuximab , een anti-CD38 monoklonaal antilichaam, aan de medicijnencombinatie lenalidomide, bortezomib en dexamethason voooraf gegeven bij patiënten met de ziekte van Kahler - multiple myeloma (botkanker)  die in aanmerking komen voor een stamceltransplantatie geeft betere MRD-negativiteit (= meetbare restziekte) op moment van de stamceltransplantatie. Aan het einde van de voorbereidende chemotherapie was de mate van meetbare ziekte ( MRD-negativiteit, zoals gemeten met flowcytometrie met een gevoeligheidsgrenswaarde van 10-5, 50% voor patiënten in de isatuximab-VRd-groep en 36% voor patiënten in de VRd-groep.
Subgroepanalyses suggereerden een vergelijkbare effectgrootte in subgroepen, inclusief die met cytogenetische afwijkingen met een hoog risico. Isatuximab verhoogde het aantal bijwerkingen van graad 3/4 niet, hoewel ze in beide groepen vaak voorkwamen. Wel waren de percentages van graad 3/4 neutropenie hoger bij patiënten in de isatuximab-VRd-groep. Stamcelmobilisatie was succesvol bij bijna alle patiënten, hoewel patiënten die isatuximab-VRd kregen meer plerixafor nodig hadden om te helpen bij het verzamelen van stamcellen, en het mediane aantal verzamelde CD34+-stamcellen was lager voor patiënten in de isatuximab-VRd-groep.

Dat blijkt uit de resultaten van een fase III studie bij totaal 660 patiënten die waren opgenomen in de ITT-analyse (331 in de isatuximab-groep en 329 in de controlegroep).

Vertaling van de resultaten uit het abstract:

  • 654 (99%) patiënten waren blank, twee waren Afrikaans, één was Arabisch en drie waren Aziatisch.
  • 250 (38%) waren vrouwen en 410 (62%) waren mannen.
  • De mediane leeftijd was 59 jaar (IQR 54-64).
  • MRD-negativiteit - meetbare restziekte na inductietherapie werd bereikt bij 166 (50%) patiënten in de isatuximab-groep versus 117 (36%) in de controlegroep (OR 1,82 [95% BI 1,33–2,48]; p=0· 00017).
  • De mediane follow-uptijd vanaf het begin tot het einde van de inductietherapie was 125 dagen (IQR 125–131) versus 125 dagen (125–132).
  • Ten minste één bijwerking van graad 3 of 4 deed zich voor bij 208 (63%) van de 330 patiënten versus 199 (61%) van de 328 patiënten.
  • Neutropenie van graad 3 of 4 trad op bij 77 (23%) versus 23 (7%) patiënten en infecties van graad 3 of 4 kwamen voor bij 40 (12%) versus 32 (10%) patiënten.
  • Van de 12 sterfgevallen tijdens inductietherapie werd één sterfgeval als gevolg van septische shock in de isatuximab-groep en vier sterfgevallen (één cardiale decompensatie, één lever- en nierfalen, één hartstilstand en één door geneesmiddelen geïnduceerde enteritis) in de controlegroep beschouwd als behandeling. verwant.

Hier het abstract van de studie gepubliceerd in The Lancet:

ARTICLES| VOLUME 9, ISSUE 11E810-E821, NOVEMBER 01, 2022

Addition of isatuximab to lenalidomide, bortezomib, and dexamethasone as induction therapy for newly diagnosed, transplantation-eligible patients with multiple myeloma (GMMG-HD7): part 1 of an open-label, multicentre, randomised, active-controlled, phase 3 trial

Published:November, 2022 DOI:https://doi.org/10.1016/S2352-3026(22)00263-0

Summary

Background

Anti-CD38 monoclonal antibodies have consistently shown increased efficacy when added to standard of care for patients with multiple myeloma. We aimed to assess the efficacy of isatuximab in addition to lenalidomide, bortezomib, and dexamethasone in patients with newly diagnosed transplantation-eligible multiple myeloma.

Methods

This open-label, multicentre, randomised, active-controlled, phase 3 trial was done at 67 academic and oncology practice centres in Germany. This study is ongoing and divided into two parts; herein, we report results from part 1. Eligible patients were aged 18–70 years; had a confirmed diagnosis of untreated multiple myeloma requiring systemic treatment and a WHO performance status of 0–2; and were eligible for induction therapy, high-dose melphalan and autologous haematopoietic stem-cell transplantation, and maintenance treatment. Patients were randomly assigned (1:1) to receive three 42-day cycles of induction therapy either with isatuximab plus lenalidomide, bortezomib, and dexamethasone (isatuximab group) or lenalidomide, bortezomib, and dexamethasone alone (control group) using a web-based system and permuted blocks. Patients in both groups received lenalidomide (25 mg orally on days 1–14 and 22–35), bortezomib (1·3 mg/m2 subcutaneously on days 1, 4, 8, 11, 22, 25, 29, and 32), and dexamethasone (20 mg orally on days 1–2, 4–5, 8–9, 11–12, 15, 22–23, 25–26, 29–30, and 32–33). Isatuximab was given as 10 mg/kg intravenously on days 1, 8, 15, 22, and 29 of cycle 1 and on days 1, 15, and 29 of cycles 2 and 3. The primary endpoint was minimal residual disease (MRD) negativity assessed by flow cytometry, in the intention-to-treat (ITT) population. This study is registered with ClinicalTrials.gov, NCT03617731.

Findings

Between Oct 23, 2018, and Sep 22, 2020, 660 patients were included in the ITT analysis (331 in the isatuximab group and 329 in the control group). 654 (99%) patients were White, two were African, one was Arabic, and three were Asian. 250 (38%) were women and 410 (62%) were men. The median age was 59 years (IQR 54–64). MRD negativity after induction therapy was reached in 166 (50%) patients in the isatuximab group versus 117 (36%) in the control group (OR 1·82 [95% CI 1·33–2·48]; p=0·00017). Median follow-up time from start to end of induction therapy was 125 days (IQR 125–131) versus 125 days (125–132). At least one grade 3 or 4 adverse event occurred in 208 (63%) of 330 patients versus 199 (61%) of 328 patients. Neutropenia of grade 3 or 4 occurred in 77 (23%) versus 23 (7%) patients and infections of grade 3 or 4 occurred in 40 (12%) versus 32 (10%) patients. Among 12 deaths during induction therapy, one death due to septic shock in the isatuximab group and four deaths (one cardiac decompensation, one hepatic and renal failure, one cardiac arrest, and one drug-induced enteritis) in the control group were considered treatment-related.

Interpretation

Addition of isatuximab to lenalidomide, bortezomib, and dexamethasone for induction therapy improved rates of MRD negativity with no new safety signals in patients with newly diagnosed transplantation-eligible multiple myeloma.

Funding

Sanofi and Bristol Myers Squibb (Celgene).

References

  1. 1.
    • Dimopoulos MA 
    • Moreau P 
    • Terpos E 
    • et al.
    Multiple myeloma: EHA-ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up.
    Ann Oncol. 2021; 32309-322
  2. 2.
    • Attal M 
    • Lauwers-Cances V 
    • Hulin C 
    • et al.
    Lenalidomide, bortezomib, and dexamethasone with transplantation for myeloma.
    N Engl J Med. 2017; 3761311-1320
  3. 3.
    • Durie BGM 
    • Hoering A 
    • Abidi MH 
    • et al.
    Bortezomib with lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with newly diagnosed myeloma without intent for immediate autologous stem-cell transplant (SWOG S0777): a randomised, open-label, phase 3 trial.
    Lancet. 2017; 389519-527
  4. 4.
    • Joseph NS 
    • Kaufman JL 
    • Dhodapkar MV 
    • et al.
    Long-term follow-up results of lenalidomide, bortezomib, and dexamethasone induction therapy and risk-adapted maintenance approach in newly diagnosed multiple myeloma.
    J Clin Oncol. 2020; 381928-1937
  5. 5.
    • Rosiñol L 
    • Oriol A 
    • Rios R 
    • et al.
    Bortezomib, lenalidomide, and dexamethasone as induction therapy prior to autologous transplant in multiple myeloma.
    Blood. 2019; 1341337-1345
  6. 6.
    • Attal M 
    • Richardson PG 
    • Rajkumar SV 
    • et al.
    Isatuximab plus pomalidomide and low-dose dexamethasone versus pomalidomide and low-dose dexamethasone in patients with relapsed and refractory multiple myeloma (ICARIA-MM): a randomised, multicentre, open-label, phase 3 study.
    Lancet. 2019; 3942096-2107
  7. 7.
    • Moreau P 
    • Dimopoulos MA 
    • Mikhael J 
    • et al.
    Isatuximab, carfilzomib, and dexamethasone in relapsed multiple myeloma (IKEMA): a multicentre, open-label, randomised phase 3 trial.
    Lancet. 2021; 3972361-2371
  8. 8.
    • Deckert J 
    • Wetzel MC 
    • Bartle LM 
    • et al.
    SAR650984, a novel humanized CD38-targeting antibody, demonstrates potent antitumor activity in models of multiple myeloma and other CD38+ hematologic malignancies.
    Clin Cancer Res. 2014; 204574-4583
  9. 9.
    • Jiang H 
    • Acharya C 
    • An G 
    • et al.
    SAR650984 directly induces multiple myeloma cell death via lysosomal-associated and apoptotic pathways, which is further enhanced by pomalidomide.
    Leukemia. 2016; 30399-408
  10. 10.
    • Moreno L 
    • Perez C 
    • Zabaleta A 
    • et al.
    The mechanism of action of the anti-CD38 monoclonal antibody isatuximab in multiple myeloma.
    Clin Cancer Res. 2019; 253176-3187
  11. 11.
    • Moreau P 
    • Attal M 
    • Hulin C 
    • et al.
    Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study.
    Lancet. 2019; 39429-38
  12. 12.
    • Voorhees PM 
    • Kaufman JL 
    • Laubach J 
    • et al.
    Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial.
    Blood. 2020; 136936-945
  13. 13.
    • Facon T 
    • Kumar S 
    • Plesner T 
    • et al.
    Daratumumab plus lenalidomide and dexamethasone for untreated myeloma.
    N Engl J Med. 2019; 3802104-2115
  14. 14.
    • Mateos MV 
    • Dimopoulos MA 
    • Cavo M 
    • et al.
    Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma.
    N Engl J Med. 2018; 378518-528
  15. 15.
    • Lahuerta JJ 
    • Paiva B 
    • Vidriales MB 
    • et al.
    Depth of response in multiple myeloma: a pooled analysis of three PETHEMA/GEM clinical trials.
    J Clin Oncol. 2017; 352900-2910
  16. 16.
    • Munshi NC 
    • Avet-Loiseau H 
    • Rawstron AC 
    • et al.
    Association of minimal residual disease with superior survival outcomes in patients with multiple myeloma: a meta-analysis.
    JAMA Oncol. 2017; 328-35
  17. 17.
    • Paiva B 
    • Puig N 
    • Cedena MT 
    • et al.
    Measurable residual disease by next-generation flow cytometry in multiple myeloma.
    J Clin Oncol. 2020; 38784-792
  18. 18.
    • Perrot A 
    • Lauwers-Cances V 
    • Corre J 
    • et al.
    Minimal residual disease negativity using deep sequencing is a major prognostic factor in multiple myeloma.
    Blood. 2018; 1322456-2464
  19. 19.
    • Kumar S 
    • Paiva B 
    • Anderson KC 
    • et al.
    International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma.
    Lancet Oncol. 2016; 17e328-e346
  20. 20.
    • Rajkumar SV 
    • Dimopoulos MA 
    • Palumbo A 
    • et al.
    International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma.
    Lancet Oncol. 2014; 15e538-e548
  21. 21.
    • Palumbo A 
    • Avet-Loiseau H 
    • Oliva S 
    • et al.
    Revised international staging system for multiple myeloma: a report from International Myeloma Working Group.
    J Clin Oncol. 2015; 332863-2869
  22. 22.
    • Flores-Montero J 
    • Sanoja-Flores L 
    • Paiva B 
    • et al.
    Next generation flow for highly sensitive and standardized detection of minimal residual disease in multiple myeloma.
    Leukemia. 2017; 312094-2103
  23. 23.
    • Neben K 
    • Lokhorst HM 
    • Jauch A 
    • et al.
    Administration of bortezomib before and after autologous stem cell transplantation improves outcome in multiple myeloma patients with deletion 17p.
    Blood. 2012; 119940-948
  24. 24.
    • Sonneveld P 
    • Avet-Loiseau H 
    • Lonial S 
    • et al.
    Treatment of multiple myeloma with high-risk cytogenetics: a consensus of the International Myeloma Working Group.
    Blood. 2016; 1272955-2962
  25. 25.
    • Paiva B 
    • San-Miguel JF 
    • Avet-Loiseau H
    MRD in multiple myeloma: does CR really matter?.
    Blood. 2022; (published online May 13.)
  26. 26.
    • Avet Loiseau H 
    • Sonneveld P 
    • Moreau P 
    • et al.
    Daratumumab (DARA) with bortezomib, thalidomide, and dexamethasone (VTd) in transplant-eligible patients (pts) with newly diagnosed multiple myeloma (NDMM): analysis of minimal residual disease (MRD) negativity in Cassiopeia part 1 and part 2.
    Blood. 2021; 138 (abstr).82
  27. 27.

    Moreau P. Treatment of newly diagnosed myeloma transplant eligible: what is the optimal induction? 18th International Myeloma Workshop; Sept 9, 2021.

  28. 28.
    • Laubach JP 
    • Kaufman JL 
    • Sborov DW 
    • et al.
    Daratumumab (DARA) plus lenalidomide, bortezomib, and dexamethasone (RVd) in patients (pts) with transplant-eligible newly diagnosed multiple myeloma (NDMM): updated analysis of Griffin after 24 months of maintenance.
    Blood. 2021; 138 (abstr).79
  29. 29.
    • Sonneveld P 
    • Dimopoulos MA 
    • Beksac M 
    • et al.
    Consolidation and maintenance in newly diagnosed multiple myeloma.
    J Clin Oncol. 2021; 393613-3622
  30. 30.
    • Cavo M 
    • Gay F 
    • Beksac M 
    • et al.
    Autologous haematopoietic stem-cell transplantation versus bortezomib-melphalan-prednisone, with or without bortezomib-lenalidomide-dexamethasone consolidation therapy, and lenalidomide maintenance for newly diagnosed multiple myeloma (EMN02/HO95): a multicentre, randomised, open-label, phase 3 study.
    Lancet Haematol. 2020; 7e456-e468
  31. 31.
    • Costa LJ 
    • Chhabra S 
    • Medvedova E 
    • et al.
    Daratumumab, carfilzomib, lenalidomide, and dexamethasone with minimal residual disease response-adapted therapy in newly diagnosed multiple myeloma.
    J Clin Oncol. 2022; 402901-2912

Linked Articles


Plaats een reactie ...

Reageer op "Isatuximab aanvullend op lenalidomide, bortezomib en dexamethason vooraf aan stamceltransplantatie geeft betere meetbare restziekte - MRD (50 vs 36 procent) bij patiënten met nieuw gediagnosticeerde multiple myeloma die stamceltransplantatie ondergaan"


Gerelateerde artikelen