Current applications of multiparameter flow cytometry in plasma cell disorders

T Jelinek, R Bezdekova, M Zatopkova, L Burgos, M Simicek, T Sevcikova, B Paiva, R Hajek, T Jelinek, R Bezdekova, M Zatopkova, L Burgos, M Simicek, T Sevcikova, B Paiva, R Hajek

Abstract

Multiparameter flow cytometry (MFC) has become standard in the management of patients with plasma cell (PC) dyscrasias, and could be considered mandatory in specific areas of routine clinical practice. It plays a significant role during the differential diagnostic work-up because of its fast and conclusive readout of PC clonality, and simultaneously provides prognostic information in most monoclonal gammopathies. Recent advances in the treatment and outcomes of multiple myeloma led to the implementation of new response criteria, including minimal residual disease (MRD) status as one of the most relevant clinical endpoints with the potential to act as surrogate for survival. Recent technical progress led to the development of next-generation flow (NGF) cytometry that represents a validated, highly sensitive, cost-effective and widely available technique for standardized MRD evaluation, which also could be used for the detection of circulating tumor cells. Here we review current applications of MFC and NGF in most PC disorders including the less frequent solitary plasmocytoma, light-chain amyloidosis or Waldenström macroglobulinemia.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Time axis highlighting the most important discoveries concerning multiparameter flow cytometry and its use in plasma cell dyscrasias.
Figure 2
Figure 2
Example of MRD analysis in MM using next generation flow approach and Infinicyt software (Cytognos). (a) Bone marrow PC compartment represents 0.04% of total nucleated cells including 98.5% of normal PCs (blue) and 1.5% of aberrant PCs (red). These aberrant plasma cells represent 0.0004% of total nucleated cells translating in MRD positive result reaching the sensitivity of 10−6. The typical aberrant phenotype: CD45−/CD38dim/CD19−/CD56+/CD27−/CD81−/CD117+/cyKappa+. (b) NGF is optimal tool also for follow-up of patients with non-secretory multiple myeloma. Bone marrow PC compartment represents 0.16% of total nucleated cells including 50% of normal PCs (blue) and 50% of aberrant PCs (red). Aberrant PCs in this case have rare immunophenotype with CD38- and lack of cytoplasmic staining of kappa or lambda light chains: CD45+/CD38−/CD19−/CD56+/CD27−/CD81+/CD117−/cyKappa−/cyLambda−.
Figure 3
Figure 3
Next-generation flow approach used for identification of CTCs in MGUS, smoldering MM and active MM patients.

References

    1. Castillo JJ. Plasma cell disorders. Prim Care 2016; 43: 677–691.
    1. Sant M, Allemani C, Tereanu C, Angelis RD, Capocaccia R, Visser O et al. Incidence of hematologic malignancies in Europe by morphologic subtype: results of the HAEMACARE project. Blood 2010; 116: 3724–3734.
    1. Rajkumar SV, Dimopoulos MA, Palumbo A, Blade J, Merlini G, Mateos M-V et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol 2014; 15: e538–e548.
    1. Kyle RA, Therneau TM, Rajkumar SV, Larson DR, Plevak MF, Offord JR et al. Prevalence of monoclonal gammopathy of undetermined significance. N Engl J Med 2006; 354: 1362–1369.
    1. Bladé J. Clinical practice. Monoclonal gammopathy of undetermined significance. N Engl J Med 2006; 355: 2765–2770.
    1. Kyle RA, Rajkumar SV. Epidemiology of the plasma-cell disorders. Best Pract Res Clin Haematol 2007; 20: 637–664.
    1. Kyle RA, Therneau TM, Rajkumar SV, Offord JR, Larson DR, Plevak MF et al. A long-term study of prognosis in monoclonal gammopathy of undetermined significance. N Engl J Med 2002; 346: 564–569.
    1. Kyle RA, Remstein ED, Therneau TM, Dispenzieri A, Kurtin PJ, Hodnefield JM et al. Clinical course and prognosis of smoldering (asymptomatic) multiple myeloma. N Engl J Med 2007; 356: 2582–2590.
    1. Kaleem Z, Crawford E, Pathan MH, Jasper L, Covinsky MA, Johnson LR et al. Flow cytometric analysis of acute leukemias. Diagnostic utility and critical analysis of data. Arch Pathol Lab Med 2003; 127: 42–48.
    1. Braylan RC. Impact of flow cytometry on the diagnosis and characterization of lymphomas, chronic lymphoproliferative disorders and plasma cell neoplasias. Cytom Part J Int Soc Anal Cytol 2004; 58: 57–61.
    1. Kwok M, Rawstron AC, Varghese A, Evans PAS, O’Connor SJM, Doughty C et al. Minimal residual disease is an independent predictor for 10-year survival in CLL. Blood 2016; 128: 2770–2773.
    1. Theunissen P, Mejstrikova E, Sedek L, van der Sluijs-Gelling AJ, Gaipa G, Bartels M et al. Standardized flow cytometry for highly sensitive MRD measurements in B-cell acute lymphoblastic leukemia. Blood 2016; 129: 347–357.
    1. Grimwade D, Freeman SD. Defining minimal residual disease in acute myeloid leukemia: which platforms are ready for ‘prime time’? Blood 2014; 124: 3345–3355.
    1. Caraux A, Klein B, Paiva B, Bret C, Schmitz A, Fuhler GM et al. Circulating human B and plasma cells. Age-associated changes in counts and detailed characterization of circulating normal CD138- and CD138+ plasma cells. Haematologica 2010; 95: 1016–1020.
    1. Jelinek T, Hajek R. Monoclonal antibodies - A new era in the treatment of multiple myeloma. Blood Rev 2016; 30: 101–110.
    1. Arroz M, Came N, Lin P, Chen W, Yuan C, Lagoo A et al. Consensus guidelines on plasma cell myeloma minimal residual disease analysis and reporting. Cytometry B Clin Cytom 2016; 90: 31–39.
    1. Flores-Montero J, de Tute R, Paiva B, Perez JJ, Böttcher S, Wind H et al. Immunophenotype of normal vs. myeloma plasma cells: toward antibody panel specifications for MRD detection in multiple myeloma. Cytometry B Clin Cytom 2016; 90: 61–72.
    1. Paiva B, Puig N, Cedena MT, de Jong BG, Ruiz Y, Rapado I et al. Differentiation stage of myeloma plasma cells: biological and clinical significance. Leukemia 2017; 31: 382–392.
    1. Ocqueteau M, Orfao A, Almeida J, Bladé J, González M, García-Sanz R et al. Immunophenotypic characterization of plasma cells from monoclonal gammopathy of undetermined significance patients. Implications for the differential diagnosis between MGUS and multiple myeloma. Am J Pathol 1998; 152: 1655–1665.
    1. Paiva B, Merino J, San Miguel JF. Utility of flow cytometry studies in the management of patients with multiple myeloma. Curr Opin Oncol 2016; 28: 511–517.
    1. Olteanu H, Harrington AM, Hari P, Kroft SH. CD200 expression in plasma cell myeloma. Br J Haematol 2011; 153: 408–411.
    1. Olteanu H. Role of flow cytometry in the diagnosis and prognosis of plasma cell myeloma. Surg Pathol Clin 2016; 9: 101–116.
    1. Flores-Montero J, Sanoja-Flores L, Paiva B, Puig N, García-Sánchez O, Böttcher S et al. Next Generation Flow for highly sensitive and standardized detection of minimal residual disease in multiple myeloma. Leukemia 2017; 31: 2094–2103.
    1. Nadav L, Katz B-Z, Baron S, Yossipov L, Polliack A, Deutsch V et al. Diverse niches within multiple myeloma bone marrow aspirates affect plasma cell enumeration. Br J Haematol 2006; 133: 530–532.
    1. Rawstron AC, Orfao A, Beksac M, Bezdickova L, Brooimans RA, Bumbea H et al. Report of the European Myeloma Network on multiparametric flow cytometry in multiple myeloma and related disorders. Haematologica 2008; 93: 431–438.
    1. Seegmiller AC, Xu Y, McKenna RW, Karandikar NJ. Immunophenotypic differentiation between neoplastic plasma cells in mature B-cell lymphoma vs plasma cell myeloma. Am J Clin Pathol 2007; 127: 176–181.
    1. Olteanu H, Wang H-Y, Chen W, McKenna RW, Karandikar NJ. Immunophenotypic studies of monoclonal gammopathy of undetermined significance. BMC Clin Pathol 2008; 8: 13.
    1. Paiva B, Almeida J, Pérez-Andrés M, Mateo G, López A, Rasillo A et al. Utility of flow cytometry immunophenotyping in multiple myeloma and other clonal plasma cell-related disorders. Cytometry B Clin Cytom 2010; 78: 239–252.
    1. Pérez-Persona E, Vidriales M-B, Mateo G, García-Sanz R, Mateos M-V, de Coca AG et al. New criteria to identify risk of progression in monoclonal gammopathy of uncertain significance and smoldering multiple myeloma based on multiparameter flow cytometry analysis of bone marrow plasma cells. Blood 2007; 110: 2586–2592.
    1. Paiva B, Vídriales M-B, Rosiñol L, Martínez-López J, Mateos M-V, Ocio EM et al. A multiparameter flow cytometry immunophenotypic algorithm for the identification of newly diagnosed symptomatic myeloma with an MGUS-like signature and long-term disease control. Leukemia 2013; 27: 2056–2061.
    1. Greipp PR, Miguel JS, Durie BGM, Crowley JJ, Barlogie B, Bladé J et al. International Staging System for Multiple Myeloma. J Clin Oncol 2005; 23: 3412–3420.
    1. Palumbo A, Avet-Loiseau H, Oliva S, Lokhorst HM, Goldschmidt H, Rosinol L et al. Revised international staging system for multiple myeloma: A Report From International Myeloma Working Group. J Clin Oncol Off J Am Soc Clin Oncol 2015; 33: 2863–2869.
    1. Avet-Loiseau H, Durie BGM, Cavo M, Attal M, Gutierrez N, Haessler J et al. Combining fluorescent in situ hybridization data with ISS staging improves risk assessment in myeloma: an International Myeloma Working Group collaborative project. Leukemia 2013; 27: 711–717.
    1. Pérez-Persona E, Mateo G, García-Sanz R, Mateos M-V, de Las Heras N, de Coca AG et al. Risk of progression in smouldering myeloma and monoclonal gammopathies of unknown significance: comparative analysis of the evolution of monoclonal component and multiparameter flow cytometry of bone marrow plasma cells. Br J Haematol 2010; 148: 110–114.
    1. Paiva B, Johnson SK, Mateos M-V, Alapat DV, Puig N, Hernandez M-T et al. Automated multiparameter flow cytometry (MFC) immunophenotyping for reproducible identification of high risk smoldering multiple myeloma (SMM). Blood 2016; 128: 373–373.
    1. Paiva B, Vidriales M-B, Mateo G, Pérez JJ, Montalbán MA, Sureda A et al. The persistence of immunophenotypically normal residual bone marrow plasma cells at diagnosis identifies a good prognostic subgroup of symptomatic multiple myeloma patients. Blood 2009; 114: 4369–4372.
    1. Lahuerta J-J, Paiva B, Vidriales M-B, Cordón L, Cedena M-T, Puig N et al. Depth of response in multiple myeloma: a pooled analysis of three PETHEMA/GEM Clinical Trials. J Clin Oncol 2017; 35: 2900–2910.
    1. Edling CE, Hallberg B. c-Kit—a hematopoietic cell essential receptor tyrosine kinase. Int J Biochem Cell Biol 2007; 39: 1995–1998.
    1. Mateo G, Montalbán MA, Vidriales M-B, Lahuerta JJ, Mateos MV, Gutiérrez N et al. Prognostic value of immunophenotyping in multiple myeloma: a study by the PETHEMA/GEM cooperative study groups on patients uniformly treated with high-dose therapy. J Clin Oncol Off J Am Soc Clin Oncol 2008; 26: 2737–2744.
    1. Schmidt-Hieber M, Pérez-Andrés M, Paiva B, Flores-Montero J, Perez JJ, Gutierrez NC et al. CD117 expression in gammopathies is associated with an altered maturation of the myeloid and lymphoid hematopoietic cell compartments and favorable disease features. Haematologica 2011; 96: 328–332.
    1. Murray ME, Gavile CM, Nair JR, Koorella C, Carlson LM, Buac D et al. CD28-mediated pro-survival signaling induces chemotherapeutic resistance in multiple myeloma. Blood 2014; 123: 3770–3779.
    1. Paiva B, Gutiérrez N-C, Chen X, Vídriales M-B, Montalbán M-Á, Rosiñol L et al. Clinical significance of CD81 expression by clonal plasma cells in high-risk smoldering and symptomatic multiple myeloma patients. Leukemia 2012; 26: 1862–1869.
    1. Paiva B, Puig N, Arana P, Cedena T, Cordon L, Vidriales MB et al. Prognostic value of antigen expression in multiple myeloma (MM): a large GEM/Pethema Study based in four consecutive clinical trials. Blood 2015; 126: 19–19.
    1. San Miguel JF, García-Sanz R, González M, Orfão A. DNA cell content studies in multiple myeloma. Leuk Lymphoma 1996; 23: 33–41.
    1. García-Sanz R, Orfão A, González M, Moro MJ, Hernández JM, Ortega F et al. Prognostic implications of DNA aneuploidy in 156 untreated multiple myeloma patients. Castelano-Leonés (Spain) Cooperative Group for the Study of Monoclonal Gammopathies. Br J Haematol 1995; 90: 106–112.
    1. Paiva B, Vídriales M-B, Montalbán M-Á, Pérez JJ, Gutiérrez NC, Rosiñol L et al. Multiparameter flow cytometry evaluation of plasma cell DNA content and proliferation in 595 transplant-eligible patients with myeloma included in the Spanish GEM2000 and GEM2005<65y trials. Am J Pathol 2012; 181: 1870–1878.
    1. Bladé J, Samson D, Reece D, Apperley J, Björkstrand B, Gahrton G et al. Criteria for evaluating disease response and progression in patients with multiple myeloma treated by high-dose therapy and haemopoietic stem cell transplantation. Myeloma Subcommittee of the EBMT. European Group for Blood and Marrow Transplant. Br J Haematol 1998; 102: 1115–1123.
    1. Durie BGM, Harousseau J-L, Miguel JS, Bladé J, Barlogie B, Anderson K et al. International uniform response criteria for multiple myeloma. Leukemia 2006; 20: 1467–1473.
    1. Rajkumar SV, Harousseau J-L, Durie B, Anderson KC, Dimopoulos M, Kyle R et al. Consensus recommendations for the uniform reporting of clinical trials: report of the International Myeloma Workshop Consensus Panel 1. Blood 2011; 117: 4691–4695.
    1. van de Velde HJK, Liu X, Chen G, Cakana A, Deraedt W, Bayssas M. Complete response correlates with long-term survival and progression-free survival in high-dose therapy in multiple myeloma. Haematologica 2007; 92: 1399–1406.
    1. Gay F, Larocca A, Wijermans P, Cavallo F, Rossi D, Schaafsma R et al. Complete response correlates with long-term progression-free and overall survival in elderly myeloma treated with novel agents: analysis of 1175 patients. Blood 2011; 117: 3025–3031.
    1. Kumar S, Paiva B, Anderson KC, Durie B, Landgren O, Moreau P et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol 2016; 17: e328–e346.
    1. Barlogie B, Mitchell A, van Rhee F, Epstein J, Morgan GJ, Crowley J. Curing myeloma at last: defining criteria and providing the evidence. Blood 2014; 124: 3043–3051.
    1. San Miguel JF, Almeida J, Mateo G, Bladé J, López-Berges C, Caballero D et al. Immunophenotypic evaluation of the plasma cell compartment in multiple myeloma: a tool for comparing the efficacy of different treatment strategies and predicting outcome. Blood 2002; 99: 1853–1856.
    1. Rawstron AC, Davies FE, DasGupta R, Ashcroft AJ, Patmore R, Drayson MT et al. Flow cytometric disease monitoring in multiple myeloma: the relationship between normal and neoplastic plasma cells predicts outcome after transplantation. Blood 2002; 100: 3095–3100.
    1. Paiva B, Vidriales M-B, Cerveró J, Mateo G, Pérez JJ, Montalbán MA et al. Multiparameter flow cytometric remission is the most relevant prognostic factor for multiple myeloma patients who undergo autologous stem cell transplantation. Blood 2008; 112: 4017–4023.
    1. Rawstron AC, Child JA, de Tute RM, Davies FE, Gregory WM, Bell SE et al. Minimal residual disease assessed by multiparameter flow cytometry in multiple myeloma: impact on outcome in the Medical Research Council Myeloma IX Study. J Clin Oncol Off J Am Soc Clin Oncol 2013; 31: 2540–2547.
    1. Paiva B, Gutiérrez NC, Rosiñol L, Vídriales M-B, Montalbán M-Á, Martínez-López J et al. High-risk cytogenetics and persistent minimal residual disease by multiparameter flow cytometry predict unsustained complete response after autologous stem cell transplantation in multiple myeloma. Blood 2012; 119: 687–691.
    1. Paiva B, Cedena M-T, Puig N, Arana P, Vidriales M-B, Cordon L et al. Minimal residual disease monitoring and immune profiling in multiple myeloma in elderly patients. Blood 2016; 127: 3165–3174.
    1. Paiva B, García-Sanz R, San Miguel JF. Multiple Myeloma Minimal Residual Disease. Cancer Treat Res 2016; 169: 103–122.
    1. Stetler-Stevenson M, Paiva B, Stoolman L, Lin P, Jorgensen JL, Orfao A et al. Consensus guidelines for myeloma minimal residual disease sample staining and data acquisition. Cytometry B Clin Cytom 2016; 90: 26–30.
    1. Martinez-Lopez J, Sanchez-Vega B, Barrio S, Cuenca I, Ruiz-Heredia Y, Alonso R et al. Analytical and clinical validation of a novel in-house deep-sequencing method for minimal residual disease monitoring in a phase II trial for multiple myeloma. Leukemia 2017; 31: 1446–1449.
    1. Martinez-Lopez J, Lahuerta JJ, Pepin F, González M, Barrio S, Ayala R et al. Prognostic value of deep sequencing method for minimal residual disease detection in multiple myeloma. Blood 2014; 123: 3073–3079.
    1. Puig N, Sarasquete ME, Balanzategui A, Martínez J, Paiva B, García H et al. Critical evaluation of ASO RQ-PCR for minimal residual disease evaluation in multiple myelomaA comparative analysis with flow cytometry. Leukemia 2014; 28: 391–397.
    1. Jelinek T, Kryukova E, Kufova Z, Kryukov F, Hajek R. Proteasome inhibitors in AL amyloidosis: focus on mechanism of action and clinical activity. Hematol Oncol. 2016 Sep 20; doi:10.1002/hon.2351.
    1. Nowakowski GS, Witzig TE, Dingli D, Tracz MJ, Gertz MA, Lacy MQ et al. Circulating plasma cells detected by flow cytometry as a predictor of survival in 302 patients with newly diagnosed multiple myeloma. Blood 2005; 106: 2276–2279.
    1. Gonsalves WI, Rajkumar SV, Gupta V, Morice WG, Timm MM, Singh PP et al. Quantification of clonal circulating plasma cells in newly diagnosed multiple myeloma: implications for redefining high-risk myeloma. Leukemia 2014; 28: 2060–2065.
    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. Granell M, Calvo X, Garcia-Guiñón A, Escoda L, Abella E, Martínez CM et al. Prognostic impact of circulating plasma cells in patients with multiple myeloma: implications for plasma cell leukaemia definition. Haematologica 2017; 102: 1099–1104, haematol.2016.158303.
    1. Vagnoni D, Travaglini F, Pezzoni V, Ruggieri M, Bigazzi C, Dalsass A et al. Circulating plasma cells in newly diagnosed symptomatic multiple myeloma as a possible prognostic marker for patients with standard-risk cytogenetics. Br J Haematol 2015; 170: 523–531.
    1. San Miguel JF, Gutiérrez NC, Mateo G, Orfao A. Conventional diagnostics in multiple myeloma. Eur J Cancer Oxf Engl 1990 2006; 42: 1510–1519.
    1. Burgos L, Alignani D, Garces J-J, Ortiz L, Jelinek T, Segura V et al. Non-invasive genetic profiling is highly applicable in multiple myeloma (MM) through characterization of circulating tumor cells (CTCs). Blood 2016; 128: 801–801.
    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. Dingli D, Nowakowski GS, Dispenzieri A, Lacy MQ, Hayman SR, Rajkumar SV et al. Flow cytometric detection of circulating myeloma cells before transplantation in patients with multiple myeloma: a simple risk stratification system. Blood 2006; 107: 3384–3388.
    1. Pardanani A, Witzig TE, Schroeder G, McElroy EA, Fonseca R, Dispenzieri A et al. Circulating peripheral blood plasma cells as a prognostic indicator in patients with primary systemic amyloidosis. Blood 2003; 101: 827–830.
    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. 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. Peceliunas V, Janiulioniene A, Matuzeviciene R, Zvirblis T, Griskevicius L. Circulating plasma cells predict the outcome of relapsed or refractory multiple myeloma. Leuk Lymphoma 2012; 53: 641–647.
    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, Witzig TE, Greipp PR, Rajkumar SV. Bone marrow angiogenesis and circulating plasma cells in multiple myeloma. Br J Haematol 2003; 122: 272–274.
    1. An G, Qin X, Acharya C, Xu Y, Deng S, Shi L et al. Multiple myeloma patients with low proportion of circulating plasma cells had similar survival with primary plasma cell leukemia patients. Ann Hematol 2015; 94: 257–264.
    1. Pellat-Deceunynck C, Barillé S, Jego G, Puthier D, Robillard N, Pineau D et al. The absence of CD56 (NCAM) on malignant plasma cells is a hallmark of plasma cell leukemia and of a special subset of multiple myeloma. Leukemia 1998; 12: 1977–1982.
    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. Bretones G, Paiva B, Valdes-Mas R, Alignani D, Garcia M, Burgos L et al. Genomic profiles of bone marrow (BM) clonal plasma cells (PCs) vs circulating tumor cells (CTCs) and Extramedullary (EM) plasmacytomas in multiple myeloma (MM). Blood 2016; 128: 4442–4442.
    1. Hill QA, Rawstron AC, de Tute RM, Owen RG. Outcome prediction in plasmacytoma of bone: a risk model utilizing bone marrow flow cytometry and light-chain analysis. Blood 2014; 124: 1296–1299.
    1. Dimopoulos MA, Terpos E. Solitary bone plasmacytomas need to flow. Blood 2014; 124: 1209–1210.
    1. Katodritou E, Terpos E, Symeonidis AS, Pouli A, Kelaidi C, Kyrtsonis M-C et al. Clinical features, outcome, and prognostic factors for survival and evolution to multiple myeloma of solitary plasmacytomas: a report of the Greek myeloma study group in 97 patients. Am J Hematol 2014; 89: 803–808.
    1. Jelinek T, Kryukova E, Kufova Z, Kryukov F, Hajek R. Proteasome inhibitors in AL amyloidosis: focus on mechanism of action and clinical activity. Hematol Oncol 2016, e-pub ahead of print 20 September 2016 doi:10.1002/hon.2351.
    1. Jelinek T, Kufova Z, Hajek R. Immunomodulatory drugs in AL amyloidosis. Crit Rev Oncol Hematol 2016; 99: 249–260.
    1. Paiva B, Martinez-Lopez J, Corchete LA, Sanchez-Vega B, Rapado I, Puig N et al. Phenotypic, transcriptomic, and genomic features of clonal plasma cells in light-chain amyloidosis. Blood 2016; 127: 3035–3039.
    1. Lisenko K, Schönland SO, Jauch A, Andrulis M, Röcken C, Ho AD et al. Flow cytometry-based characterization of underlying clonal B and plasma cells in patients with light chain amyloidosis. Cancer Med 2016; 5: 1464–1472.
    1. Paiva B, Vídriales M-B, Pérez JJ, López-Berges M-C, García-Sanz R, Ocio EM et al. The clinical utility and prognostic value of multiparameter flow cytometry immunophenotyping in light-chain amyloidosis. Blood 2011; 117: 3613–3616.
    1. Muchtar E, Jevremovic D, Dispenzieri A, Dingli D, Buadi FK, Lacy MQ et al. The prognostic value of multiparametric flow cytometry in AL amyloidosis at diagnosis and at the end of first-line treatment. Blood 2017; 129: 82–87.
    1. Gertz MA. Waldenström macroglobulinemia: 2017 update on diagnosis, risk stratification, and management. Am J Hematol 2017; 92: 209–217.
    1. Waldenstrom J. Macroglobulinaemia. Acta Haematol 1958; 20: 33–39.
    1. Owen RG, Treon SP, Al-Katib A, Fonseca R, Greipp PR, McMaster ML et al. Clinicopathological definition of Waldenstrom’s macroglobulinemia: consensus panel recommendations from the Second International Workshop on Waldenstrom’s Macroglobulinemia. Semin Oncol 2003; 30: 110–115.
    1. Castillo JJ, Garcia-Sanz R, Hatjiharissi E, Kyle RA, Leleu X, McMaster M et al. Recommendations for the diagnosis and initial evaluation of patients with Waldenström Macroglobulinaemia: A Task Force from the 8th International Workshop on Waldenström Macroglobulinaemia. Br J Haematol 2016; 175: 77–86.
    1. Paiva B, Corchete LA, Vidriales M-B, García-Sanz R, Perez JJ, Aires-Mejia I et al. The cellular origin and malignant transformation of Waldenström macroglobulinemia. Blood 2015; 125: 2370–2380.
    1. Treon SP, Xu L, Yang G, Zhou Y, Liu X, Cao Y et al. MYD88 L265P somatic mutation in Waldenström’s macroglobulinemia. N Engl J Med 2012; 367: 826–833.
    1. Landgren O, Staudt L. MYD88 L265P somatic mutation in IgM MGUS. N Engl J Med 2012; 367: 2255–2256-2257.
    1. García-Sanz R, Jiménez C, Puig N, Paiva B, Gutiérrez NC, Rodríguez-Otero P et al. Origin of Waldenstrom’s macroglobulinaemia. Best Pract Res Clin Haematol 2016; 29: 136–147.
    1. San Miguel JF, Vidriales MB, Ocio E, Mateo G, Sánchez-Guijo F, Sánchez ML et al. Immunophenotypic analysis of Waldenstrom’s macroglobulinemia. Semin Oncol 2003; 30: 187–195.
    1. Paiva B, Montes MC, García-Sanz R, Ocio EM, Alonso J, de Las Heras N et al. Multiparameter flow cytometry for the identification of the Waldenström’s clone in IgM-MGUS and Waldenström’s Macroglobulinemia: new criteria for differential diagnosis and risk stratification. Leukemia 2014; 28: 166–173.
    1. Paiva B, Chandia M, Vidriales M-B, Colado E, Caballero-Velázquez T, Escalante F et al. Multiparameter flow cytometry for staging of solitary bone plasmacytoma: new criteria for risk of progression to myeloma. Blood 2014; 124: 1300–1303.
    1. Avet-Loiseau H, Garand R, Lodé L, Robillard N, Bataille R. 14q32 Translocations discriminate IgM multiple myeloma from Waldenstrom’s macroglobulinemia. Semin Oncol 2003; 30: 153–155.
    1. Willenbacher W, Willenbacher E, Brunner A, Manzl C. Improved accuracy of discrimination between IgM multiple myeloma and Waldenström macroglobulinaemia by testing for MYD88 L265P mutations. Br J Haematol 2013; 161: 902–904.
    1. García-Sanz R, Ocio E, Caballero A, Magalhães RJP, Alonso J, López-Anglada L et al. Post-treatment bone marrow residual disease>5% by flow cytometry is highly predictive of short progression-free and overall survival in patients with Waldenström’s macroglobulinemia. Clin Lymphoma Myeloma Leuk 2011; 11: 168–171.
    1. Jelinek T, Hajek R. PD-1/PD-L1 inhibitors in multiple myeloma: The present and the future. OncoImmunology 2016; 5: e1254856.
    1. Seckinger A, Delgado JA, Moser S, Moreno L, Neuber B, Grab A et al. Target expression, generation, preclinical activity, and pharmacokinetics of the BCMA-T cell bispecific antibody EM801 for multiple myeloma treatment. Cancer Cell 2017; 31: 396–410.
    1. Robillard N, Jego G, Pellat-Deceunynck C, Pineau D, Puthier D, Mellerin MP et al. CD28, a marker associated with tumoral expansion in multiple myeloma. Clin Cancer Res Off J Am Assoc Cancer Res 1998; 4: 1521–1526.
    1. Robillard N, Wuillème S, Lodé L, Magrangeas F, Minvielle S, Avet-Loiseau H. CD33 is expressed on plasma cells of a significant number of myeloma patients, and may represent a therapeutic target. Leukemia 2005; 19: 2021–2022.
    1. Lee KH, Seo HS, Sohn JY, Lee E, Lee H, Eom H-S et al. Abstract 3123: Aberrant expression of CD33 is associated with poor prognosis in patients with multiple myeloma and tumor progression. Cancer Res 2016; 76(14 Supplement): 3123–3123.
    1. Gonsalves WI, Timm MM, Rajkumar SV, Morice WG, Dispenzieri A, Buadi FK et al. The prognostic significance of CD45 expression by clonal bone marrow plasma cells in patients with newly diagnosed multiple myeloma. Leuk Res 2016; 44: 32–39.
    1. Pojero F, Flores-Montero J, Sanoja L, Pérez JJ, Puig N, Paiva B et al. Utility of CD54, CD229, and CD319 for the identification of plasma cells in patients with clonal plasma cell diseases. Cytometry B Clin Cytom 2016; 90: 91–100.
    1. Iqbal MS, Otsuyama K-I, Shamsasenjan K, Asaoku H, Mahmoud MS, Gondo T et al. Constitutively lower expressions of CD54 on primary myeloma cells and their different localizations in bone marrow. Eur J Haematol 2009; 83: 302–312.
    1. Van Camp B, Durie BG, Spier C, De Waele M, Van Riet I, Vela E et al. Plasma cells in multiple myeloma express a natural killer cell-associated antigen: CD56 (NKH-1; Leu-19). Blood 1990; 76: 377–382.
    1. Van Riet I, De Waele M, Remels L, Lacor P, Schots R, Van Camp B. Expression of cytoadhesion molecules (CD56, CD54, CD18 and CD29) by myeloma plasma cells. Br J Haematol 1991; 79: 421–427.
    1. Rawstron A, Barrans S, Blythe D, Davies F, English A, Pratt G et al. Distribution of myeloma plasma cells in peripheral blood and bone marrow correlates with CD56 expression. Br J Haematol 1999; 104: 138–143.
    1. Conticello C, Giuffrida R, Parrinello N, Buccheri S, Adamo L, Sciuto MR et al. CD200 expression in patients with Multiple Myeloma: another piece of the puzzle. Leuk Res 2013; 37: 1616–1621.
    1. Tai Y-T, Dillon M, Song W, Leiba M, Li X-F, Burger P et al. Anti-CS1 humanized monoclonal antibody HuLuc63 inhibits myeloma cell adhesion and induces antibody-dependent cellular cytotoxicity in the bone marrow milieu. Blood 2008; 112: 1329–1337.
    1. Muccio VE, Saraci E, Gilestro M, Gattei V, Zucchetto A, Astolfi M et al. Multiple myeloma: New surface antigens for the characterization of plasma cells in the era of novel agents. Cytometry B Clin Cytom 2016; 90: 81–90.
    1. Roussel M, Lauwers-Cances V, Robillard N, Hulin C, Leleu X, Benboubker L et al. Front-line transplantation program with lenalidomide, bortezomib, and dexamethasone combination as induction and consolidation followed by lenalidomide maintenance in patients with multiple myeloma: a phase II study by the Intergroupe Francophone du Myélome. J Clin Oncol 2014; 32: 2712–2717.
    1. Paiva B, Chandia M, Puig N, Vidriales M-B, Perez JJ, Lopez-Corral L et al. The prognostic value of multiparameter flow cytometry minimal residual disease assessment in relapsed multiple myeloma. Haematologica 2015; 100: e53–e55.
    1. Fukumoto K, Fujisawa M, Suehara Y, Narita K-T, Usui Y, Takeuchi M et al. Prognostic impact of immunophenotypic complete response in patients with multiple myeloma achieving better than complete response. Leuk Lymphoma 2016; 57: 1786–1792.
    1. Attal M, Lauwers-Cances V, Hulin C, Leleu X, Caillot D, Escoffre M et al. Lenalidomide, bortezomib, and dexamethasone with transplantation for myeloma. N Engl J Med 2017; 376: 1311–1320.

Source: PubMed

3
Abonnieren