Cell-free DNA for the detection of emerging treatment failure in relapsed/ refractory multiple myeloma

Johannes M Waldschmidt, Andrew J Yee, Tushara Vijaykumar, Ricardo A Pinto, Julia Frede, Praveen Anand, Giada Bianchi, Guangwu Guo, Sayalee Potdar, Charles Seifer, Monica S Nair, Antonis Kokkalis, Jake A Kloeber, Samantha Shapiro, Lillian Budano, Mason Mann, Robb Friedman, Brea Lipe, Erica Campagnaro, Elizabeth K O'Donnell, Cheng-Zhong Zhang, Jacob P Laubach, Nikhil C Munshi, Paul G Richardson, Kenneth C Anderson, Noopur S Raje, Birgit Knoechel, Jens G Lohr, Johannes M Waldschmidt, Andrew J Yee, Tushara Vijaykumar, Ricardo A Pinto, Julia Frede, Praveen Anand, Giada Bianchi, Guangwu Guo, Sayalee Potdar, Charles Seifer, Monica S Nair, Antonis Kokkalis, Jake A Kloeber, Samantha Shapiro, Lillian Budano, Mason Mann, Robb Friedman, Brea Lipe, Erica Campagnaro, Elizabeth K O'Donnell, Cheng-Zhong Zhang, Jacob P Laubach, Nikhil C Munshi, Paul G Richardson, Kenneth C Anderson, Noopur S Raje, Birgit Knoechel, Jens G Lohr

Abstract

Interrogation of cell-free DNA (cfDNA) represents an emerging approach to non-invasively estimate disease burden in multiple myeloma (MM). Here, we examined low-pass whole genome sequencing (LPWGS) of cfDNA for its predictive value in relapsed/ refractory MM (RRMM). We observed that cfDNA positivity, defined as ≥10% tumor fraction by LPWGS, was associated with significantly shorter progression-free survival (PFS) in an exploratory test cohort of 16 patients who were actively treated on diverse regimens. We prospectively determined the predictive value of cfDNA in 86 samples from 45 RRMM patients treated with elotuzumab, pomalidomide, bortezomib, and dexamethasone in a phase II clinical trial (NCT02718833). PFS in patients with tumor-positive and -negative cfDNA after two cycles of treatment was 1.6 and 17.6 months, respectively (HR 7.6, P < 0.0001). Multivariate hazard modelling confirmed cfDNA as independent risk factor (HR 96.6, P = 6.92e-05). While correlating with serum-free light chains and bone marrow, cfDNA additionally discriminated patients with poor PFS among those with the same response by IMWG criteria. In summary, detectability of MM-derived cfDNA, as a measure of substantial tumor burden with therapy, independently predicts poor PFS and may provide refinement for standard-of-care response parameters to identify patients with poor response to treatment earlier than is currently feasible.

Conflict of interest statement

Competing interest statement:

J.M.W.: Advisory boards of Janssen and Sanofi. B.L.: Research funding from Amgen and Cellectar. Advisory boards of Bristol-Myers Squibb, Janssen and GlaxoSmithKline. C.-Z.Z.: Co-founder, advisor, and equity holder of Pillar BioSciences. P.G.R.: Research grants from Bristol-Myers Squibb, Oncopeptides, Celgene, Takeda, and Karyopharm. Advisory boards of Oncopeptides, Janssen, Sanofi, and Secura Bio. K.C.A.: Consultant to Bristol-Myers Squibb, Millennium, Janssen, Sanofi, Amgen, Gilead, Precision Biosciences. Scientific founder of Oncopep and C4 Therapeutics. N.S.R.: Consultant to Amgen, Bristol-Myers Squibb, Janssen, Sanofi, Takeda, AstraZeneca and C4 Therapeutics. Advisory boards of Caribou and Immuneel. Research funding from BluebirdBio. J.G.L.: Consultant for T2 Biosystems. Research funding from Bristol-Myers Squibb, Celgene. All other authors declare no conflicts of interest.

No disclosures related to this publication.

© 2021. The Author(s), under exclusive licence to Springer Nature Limited.

Figures

Figure 1.. Tumor fraction in cfDNA as…
Figure 1.. Tumor fraction in cfDNA as proxy for tumor burden and as prognostic marker.
A. Primary analysis of cfDNA in a test cohort of MM patients (n=16) undergoing a variety of therapeutic regimens. Tumor fraction in cfDNA was measured after a minimum of two months on the current regimen using ichorCNA. In a second validation cohort, patients with RRMM (n=45) received uniform treatment with elotuzumab, pomalidomide, bortezomib and dexamethasone (elo-PVD, clinicaltrials.gov identifier: NCT02718833). Tumor fraction in cfDNA was measured at baseline (n=44 available), and at cycle 3 day 1 (C3D1, n=42 available) of treatment. B. Kaplan-Meier curve for PFS (months) in MM patients with tumor fraction positivity (≥10%) or negativity (<10%) in cfDNA. C,D. Serial copy number profile, cfDNA tumor fraction and iFLC obtained at multiple time points in two patients, Pt01T (C, PFS 2.8 months) and Pt02T (D, PFS, 2.4 months). Abbreviations: RRMM= relapsed/refractory multiple myeloma, LPWGS= low-pass whole genome sequencing, elo-PVD= elotuzumab-pomalidomide-bortezomib-dexamethasone, C3D1= cycle 3 day 1 of elo-PVD treatment, TF= tumor fraction, EoT= end of treatment.
Figure 2.. Tumor fraction in cfDNA is…
Figure 2.. Tumor fraction in cfDNA is predictive of progression-free survival.
Kaplan-Meier survival for PFS (months) in MM patients with tumor fraction positive or negative results at screening (A, n=44) and C3D1 (B, n=42). Abbreviations: TF= tumor fraction.
Figure 3.. Comparing MM-derived cfDNA with serological…
Figure 3.. Comparing MM-derived cfDNA with serological parameters and bone marrow infiltration.
Correlation of paired samples between cfDNA tumor fraction and iFLC (A, n=79) or bone marrow infiltration (B, n=56). C. Copy number profile and matched bone marrow result for Pt01V (PFS 3.8 months) and Pt02V (PFS 1.2 months). Abbreviations: iFLC= involved serum-free light chain, BM= bone marrow, PFS= progression-free survival, TF= tumor fraction.
Figure 4.. Refining IMWG response criteria with…
Figure 4.. Refining IMWG response criteria with cfDNA as an orthogonal marker of response.
A. Kaplan-Meier survival for PFS (months) in 33 MM pts with SD or PR according to IMWG criteria separated by tumor fraction positive or negative cfDNA testing at C3D1. B. Detailed median PFS and hazard ratio values for patients with SD or PR according to respective cfDNA status at C3D1. C,D. Copy number profile, cfDNA tumor fraction, iFLC and serum M-protein over time in two patients, Pt03V (C, PFS 3.3 months) and Pt04V (D, PFS 4.3 months). Abbreviations: IMWG= International Myeloma Working Group, C3D1= cycle 3 day 1 (of elo-PVD treatment), PFS= progression-free survival, PR= partial response, SD= stable disease, TF= tumor fraction.
Figure 5.. Cox proportional hazard model for…
Figure 5.. Cox proportional hazard model for PFS and cfDNA tumor fraction after two cycles of elo-PVD.
Forest plots of hazard ratios for PFS according to cfDNA positivity (P=6.92e-05) at cycle 3 day 1 (C3D1) compared to achievement of VGPR (P=0.646) and PR (P=0.315) at the same time point (n=42), as well as age, having received >1 treatment, ISS stage III and high-risk cytogenetics. Cytogenetic information was not available for n=4 patients (*). Abbreviations: C3D1= cycle 3 day 1 (of elo-PVD treatment), TF= tumor fraction, VGPR= very good partial response, PR= partial response, SD= stable disease, ISS= International Staging System, HR= high-risk (del17p, t(4;14), t(14;16), t(14;20)).

References

    1. Kumar SK, Dispenzieri A, Lacy MQ, Gertz MA, Buadi FK, Pandey S et al. Continued improvement in survival in multiple myeloma: changes in early mortality and outcomes in older patients. Leukemia 2014; 28: 1122–1128.
    1. Keats JJ, Chesi M, Egan JB, Garbitt VM, Palmer SE, Braggio E et al. Clonal competition with alternating dominance in multiple myeloma. Blood 2012; 120: 1067–1076.
    1. Lohr JG, Stojanov P, Carter SL, Cruz-Gordillo P, Lawrence MS, Auclair D et al. Widespread genetic heterogeneity in multiple myeloma: implications for targeted therapy. Cancer Cell 2014; 25: 91–101.
    1. Bolli N, Avet-Loiseau H, Wedge DC, Van Loo P, Alexandrov LB, Martincorena I et al. Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat Commun 2014; 5: 2997.
    1. Bergsagel PL, Kuehl WM. Molecular pathogenesis and a consequent classification of multiple myeloma. J Clin Oncol; 23: 6333–6338.
    1. Chapman MA, Lawrence MS, Keats JJ, Cibulskis K, Sougnez C, Schinzel AC et al. Initial genome sequencing and analysis of multiple myeloma. Nature 2011; 471: 467–472.
    1. Morgan GJ, Walker BA, Davies FE. The genetic architecture of multiple myeloma. Nat Rev Cancer 2012; 12: 335–348.
    1. Kumar SK, Rajkumar SV, Dispenzieri A, Lacy MQ, Hayman SR, Buadi FK et al. Improved survival in multiple myeloma and the impact of novel therapies. Blood 2008; 111: 2516–2520.
    1. Kumar SK, Therneau TM, Gertz MA, Lacy MQ, Dispenzieri A, Rajkumar SV et al. Clinical course of patients with relapsed multiple myeloma. Mayo Clin Proc 2004; 79: 867–874.
    1. Landgren O, Rajkumar SV. New Developments in Diagnosis, Prognosis, and Assessment of Response in Multiple Myeloma. Clin Cancer Res 2016; 22: 5428–5433.
    1. Rasche L, Chavan SS, Stephens OW, Patel PH, Tytarenko R, Ashby C et al. Spatial genomic heterogeneity in multiple myeloma revealed by multi-region sequencing. Nat Commun 2017; 8: 268.
    1. Caers J, Paiva B, Zamagni E, Leleu X, Bladé J, Kristinsson SY et al. Diagnosis, treatment, and response assessment in solitary plasmacytoma: updated recommendations from a European Expert Panel. J Hematol Oncol 2018; 11: 10.
    1. Chawla SS, Kumar SK, Dispenzieri A, Greenberg AJ, Larson DR, Kyle RA et al. Clinical Course and Prognosis of Non-Secretory Multiple Myeloma. Eur J Haematol 2015. doi:10.1111/ejh.12534.
    1. Dupuis MM, Tuchman SA. Non-secretory multiple myeloma: from biology to clinical management. OncoTargets Ther 2016; 9: 7583–7590.
    1. Mills JR, Barnidge DR, Dispenzieri A, Murray DL. High sensitivity blood-based M-protein detection in sCR patients with multiple myeloma. Blood Cancer J 2017; 7: e590.
    1. Mithraprabhu S, Chen M, Savvidou I, Reale A, Spencer A. Liquid biopsy: an evolving paradigm for the biological characterisation of plasma cell disorders. Leukemia 2021. doi:10.1038/s41375-021-01339-6.
    1. Adalsteinsson VA, Ha G, Freeman SS, Choudhury AD, Stover DG, Parsons HA et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun 2017; 8. doi:10.1038/s41467-017-00965-y.
    1. Leary RJ, Kinde I, Diehl F, Schmidt K, Clouser C, Duncan C et al. Development of personalized tumor biomarkers using massively parallel sequencing. Sci Transl Med 2010; 2: 20ra14.
    1. Murtaza M, Dawson S-J, Tsui DWY, Gale D, Forshew T, Piskorz AM et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 2013; 497: 108–112.
    1. Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M et al. Circulating mutant DNA to assess tumor dynamics. Nat Med 2008; 14: 985–990.
    1. Scherer F, Kurtz DM, Newman AM, Stehr H, Craig AFM, Esfahani MS et al. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med 2016; 8: 364ra155.
    1. Corcoran RB, Chabner BA. Cell-free DNA Analysis in Cancer. N Engl J Med 2019; 380: 501–502.
    1. Manier S, Park J, Capelletti M, Bustoros M, Freeman SS, Ha G et al. Whole-exome sequencing of cell-free DNA and circulating tumor cells in multiple myeloma. Nat Commun 2018; 9: 1691.
    1. Guo G, Raje NS, Seifer C, Kloeber J, Isenhart R, Ha G et al. Genomic discovery and clonal tracking in multiple myeloma by cell-free DNA sequencing. Leukemia 2018; 32: 1838–1841.
    1. Gerber B, Manzoni M, Spina V, Bruscaggin A, Lionetti M, Fabris S et al.. Circulating tumor DNA as a liquid biopsy in plasma cell dyscrasias. Haematologica 2018; 103: e245–e248.
    1. Mithraprabhu S, Khong T, Ramachandran M, Chow A, Klarica D, Mai L et al. Circulating tumour DNA analysis demonstrates spatial mutational heterogeneity that coincides with disease relapse in myeloma. Leukemia 2017; 31: 1695–1705.
    1. Mithraprabhu S, Morley R, Khong T, Kalff A, Bergin K, Hocking J et al. Monitoring tumour burden and therapeutic response through analysis of circulating tumour DNA and extracellular RNA in multiple myeloma patients. Leukemia 2019; 33: 2022–2033.
    1. Kis O, Kaedbey R, Chow S, Danesh A, Dowar M, Li T et al. Circulating tumour DNA sequence analysis as an alternative to multiple myeloma bone marrow aspirates. Nat Commun 2017; 8: 15086.
    1. Rustad EH, Coward E, Skytøen ER, Misund K, Holien T, Standal T et al. Monitoring multiple myeloma by quantification of recurrent mutations in serum. Haematologica 2017; 102: 1266–1272.
    1. Li Q, Huang HJ, Ma J, Wang Y, Cao Z, Karlin-Neumann G et al. RAS/RAF mutations in tumor samples and cell-free DNA from plasma and bone marrow aspirates in multiple myeloma patients. J Cancer 2020; 11: 3543–3550.
    1. Oberle A, Brandt A, Voigtlaender M, Thiele B, Radloff J, Schulenkorf A et al. Monitoring multiple myeloma by next-generation sequencing of V(D)J rearrangements from circulating myeloma cells and cell-free myeloma DNA. Haematologica 2017; 102: 1105–1111.
    1. Biancon G, Gimondi S, Vendramin A, Carniti C, Corradini P. Noninvasive Molecular Monitoring in Multiple Myeloma Patients Using Cell-Free Tumor DNA: A Pilot Study. J Mol Diagn 2018; 20: 859–870.
    1. Mazzotti C, Buisson L, Maheo S, Perrot A, Chretien M-L, Leleu X et al. Myeloma MRD by deep sequencing from circulating tumor DNA does not correlate with results obtained in the bone marrow. Blood Adv 2018; 2: 2811–2813.
    1. Deshpande S, Tytarenko RG, Wang Y, Boyle EM, Ashby C, Schinke CD et al. Monitoring treatment response and disease progression in myeloma with circulating cell-free DNA. Eur J Haematol 2021; 106: 230–240.
    1. Mithraprabhu S, Sirdesai S, Chen M, Khong T, Spencer A. Circulating Tumour DNA Analysis for Tumour Genome Characterisation and Monitoring Disease Burden in Extramedullary Multiple Myeloma. Int J Mol Sci 2018; 19. doi:10.3390/ijms19071858.
    1. Manzoni M, Pompa A, Fabris S, Pelizzoni F, Ciceri G, Seia M et al. Limits and Applications of Genomic Analysis of Circulating Tumor DNA as a Liquid Biopsy in Asymptomatic Forms of Multiple Myeloma. HemaSphere 2020; 4: e402.
    1. Yee AJ, Laubach JP, Campagnaro EL, Lipe BC, Nadeem O, Friedman RS et al. A Phase II Study of Elotuzumab in Combination with Pomalidomide, Bortezomib, and Dexamethasone in Relapsed and Refractory Multiple Myeloma. Blood 2019; 134: 3169–3169.
    1. Ihorst G, Waldschmidt J, Schumacher M, Wäsch R, Engelhardt M. Analysis of survival by tumor response: have we learnt any better? Ann Hematol 2015; 94: 1615–1616.
    1. Chng WJ, Dispenzieri A, Chim C-S, Fonseca R, Goldschmidt H, Lentzsch S et al. IMWG consensus on risk stratification in multiple myeloma. Leukemia 2014; 28: 269–277.
    1. Avet-Loiseau H, Li C, Magrangeas F, Gouraud W, Charbonnel C, Harousseau J-L et al. Prognostic significance of copy-number alterations in multiple myeloma. J Clin Oncol 2009; 27: 4585–4590.
    1. Kortuem KM, Braggio E, Bruins L, Barrio S, Shi CS, Zhu YX et al. Panel sequencing for clinically oriented variant screening and copy number detection in 142 untreated multiple myeloma patients. Blood Cancer J 2016; 6: e397.
    1. Frede J, Anand P, Sotudeh N, Pinto RA, Nair MS, Stuart H et al. Dynamic transcriptional reprogramming leads to immunotherapeutic vulnerabilities in myeloma. Nat Cell Biol 2021. doi:10.1038/s41556-021-00766-y.
    1. Waldschmidt JM, Kloeber JA, Anand P, Frede J, Kokkalis A, Dimitrova V et al. Single-Cell Profiling Reveals Metabolic Reprogramming as a Resistance Mechanism in BRAF-Mutated Multiple Myeloma. Clin Cancer Res 2021. doi:10.1158/1078-0432.CCR-21-2040.
    1. Vrabel D, Sedlarikova L, Besse L, Rihova L, Bezdekova R, Almasi M et al. Dynamics of tumor-specific cfDNA in response to therapy in multiple myeloma patients. Eur J Haematol 2020; 104: 190–197.
    1. Engelhardt M, Ihorst G, Duque-Afonso J, Wedding U, Spät-Schwalbe E, Goede V et al. Structured assessment of frailty in multiple myeloma as a paradigm of individualized treatment algorithms in cancer patients at advanced age. Haematologica 2020; 105: 1183–1188.
    1. Alidousty C, Brandes D, Heydt C, Wagener S, Wittersheim M, Schäfer SC et al. Comparison of Blood Collection Tubes from Three Different Manufacturers for the Collection of Cell-Free DNA for Liquid Biopsy Mutation Testing. J Mol Diagn 2017; 19: 801–804.
    1. Mithraprabhu S, Spencer A. Analysis of Circulating Tumor DNA. Methods Mol Biol 2018; 1792: 129–145.
    1. Bianchi G, Kyle RA, Larson DR, Witzig TE, Kumar S, Dispenzieri A et al. High levels of peripheral blood circulating plasma cells as a specific risk factor for progression of smoldering multiple myeloma. Leukemia 2013; 27: 680–685.
    1. Lohr JG, Kim S, Gould J, Knoechel B, Drier Y, Cotton MJ et al. Genetic interrogation of circulating multiple myeloma cells at single-cell resolution. Sci Transl Med 2016; 8: 363ra147.
    1. Mishima Y, Paiva B, Shi J, Park J, Manier S, Takagi S et al. The Mutational Landscape of Circulating Tumor Cells in Multiple Myeloma. Cell Rep 2017; 19: 218–224.
    1. Kumar S, Rajkumar SV, Kyle RA, Lacy MQ, Dispenzieri A, Fonseca R et al. Prognostic value of circulating plasma cells in monoclonal gammopathy of undetermined significance. J Clin Oncol 2005; 23: 5668–5674.
    1. Gonsalves WI, Rajkumar SV, Dispenzieri A, Dingli D, Timm MM, Morice WG et al. Quantification of circulating clonal plasma cells via multiparametric flow cytometry identifies patients with smoldering multiple myeloma at high risk of progression. Leukemia 2017; 31: 130–135.
    1. Gonsalves WI, Jevremovic D, Nandakumar B, Dispenzieri A, Buadi FK, Dingli D et al. Enhancing the R-ISS classification of newly diagnosed multiple myeloma by quantifying circulating clonal plasma cells. Am J Hematol 2020; 95: 310–315.
    1. Paiva B, Paino T, Sayagues J-M, Garayoa M, San-Segundo L, Martín M et al. Detailed characterization of multiple myeloma circulating tumor cells shows unique phenotypic, cytogenetic, functional, and circadian distribution profile. Blood 2013; 122: 3591–3598.
    1. Sanoja-Flores L, Flores-Montero J, Puig N, Contreras-Sanfeliciano T, Pontes R, Corral-Mateos A et al. Blood monitoring of circulating tumor plasma cells by next generation flow in multiple myeloma after therapy. Blood 2019; 134: 2218–2222.
    1. Garcés J-J, Bretones G, Burgos L, Valdes-Mas R, Puig N, Cedena M-T et al. Circulating tumor cells for comprehensive and multiregional non-invasive genetic characterization of multiple myeloma. Leukemia 2020; 34: 3007–3018.
    1. Kubiczkova L, Kryukov F, Slaby O, Dementyeva E, Jarkovsky J, Nekvindova J et al. Circulating serum microRNAs as novel diagnostic and prognostic biomarkers for multiple myeloma and monoclonal gammopathy of undetermined significance. Haematologica 2014; 99: 511–518.
    1. Wang W, Corrigan-Cummins M, Barber EA, Saleh LM, Zingone A, Ghafoor A et al. Aberrant Levels of miRNAs in Bone Marrow Microenvironment and Peripheral Blood of Myeloma Patients and Disease Progression. J Mol Diagn 2015; 17: 669–678.
    1. Navarro A, Díaz T, Tovar N, Pedrosa F, Tejero R, Cibeira MT et al. A serum microRNA signature associated with complete remission and progression after autologous stem-cell transplantation in patients with multiple myeloma. Oncotarget 2015; 6: 1874–1883.
    1. Lionetti M, Musto P, Di Martino MT, Fabris S, Agnelli L, Todoerti K et al. Biological and clinical relevance of miRNA expression signatures in primary plasma cell leukemia. Clin Cancer Res 2013; 19: 3130–3142.

Source: PubMed

3
Suscribir