A krónikus limfoid leukémia (CLL) a leggyakoribb felnőttkori leukémia a fejlett országokban. A CLL egy indolens lefolyású, gyógyíthatatlan betegség, mely a kezelést követő relapszusokkal járhat, emiatt a betegek folyamatos követése szükséges. Az elmúlt évek bővülő terápiás lehetőségei (pl.: alemtuzumab, venetoclax) lehetővé tették a daganatos sejtek mennyiségének tartós és drasztikus csökkenését, ezért szükségessé vált a visszatérő betegség minél érzékenyebb detektálása. Erre nyújt lehetőséget a minimális, újabban mérhető reziduális betegség (minimal residual disease: MRD) meghatározása áramlási citometriai és molekuláris genetikai vizsgálati módszerekkel. Az MRD-szint és a túlélés összefüggését vizsgáló klinikai tanulmányok egyértelművé tették, hogy az alacsonyabb MRD-szint hosszabb túléléssel jár együtt. Bár az MRD meghatározása jelenleg nem a klinikai rutin része, amennyiben a jelenleg még futó klinikai vizsgálatok eredményei indokolttá teszik meghatározását, áramlási citometriai módszeren alapuló vizsgálata a rutin diagnosztika része lehet majd.
Vuillier F, et al. Evaluation of residual disease in B-cell chronic lymphocytic leukemia patients in clinical and bone-marrow remission using CD5-CD19 markers and PCR study of gene rearrangements. Leuk Lymphoma 1992; 7(3): 195–204.
Rawstron AC, et al. Quantitation of minimal disease levels in chronic lymphocytic leukemia using a sensitive flow cytometric assay improves the prediction of outcome and can be used to optimize therapy. Blood 2001; 98(1): 29–35.
Hillmen P, et al. Alemtuzumab compared with chlorambucil as first-line therapy for chronic lymphocytic leukemia. J Clin Oncol. 2007; 25(35): 5616–5623.
Böttcher S, et al. Minimal residual disease quantification is an independent predictor of progression-free and overall survival in chronic lymphocytic leukemia: a multivariate analysis from the randomized GCLLSG CLL8 trial. J Clin Oncol. 2012; 30(9): 980–988.
Stilgenbauer S, et al. Venetoclax for patients with chronic lymphocytic leukemia with 17p deletion: Results from the full population of a phase II pivotal trial. J Clin Oncol. 2018; 36(19): 1973–1980.
Hallek M, et al. iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL. Blood 2018; 131(25): 2745–2760.
Kwok M, et al. Minimal residual disease is an independent predictor for 10-year survival in CLL. Blood 2016; 128(24): 2770–2773.
Eichhorst B, et al. First-line chemoimmunotherapy with bendamustine and rituximab versus fludarabine, cyclophosphamide, and rituximab in patients with advanced chronic lymphocytic leukaemia (CLL10): an international, open-label, randomised, phase 3, non-inferiority trial. Lancet Oncol. 2016; 17(7): 928–942.
Geisler CH, et al. Frontline low-dose alemtuzumab with fludarabine and cyclophosphamide prolongs progression-free survival in high-risk CLL. Blood 2014; 123(21): 3255–3262.
Roberts AW, et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016; 374(4): 311–322.
Seymour JF, et al. Venetoclax-rituximab in relapsed or refractory chronic lymphocytic leukemia. N Engl J Med. 2018; 378(12): 1107–1120.
Hallek M, et al. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute – Working Group 1996 guidelines. Blood 2008; 111(12): 5446–5456.
Kovacs G, et al. Minimal residual disease assessment improves prediction of outcome in patients with chronic lymphocytic leukemia (CLL) who achieve partial response: Comprehensive analysis of two phase III studies of the German CLL Study Group. J Clin Oncol. 2016; 34(31): 3758–3765.
Ghia P, Rawstron A. Minimal residual disease analysis in chronic lymphocytic leukemia: A way for achieving more personalized treatments. Leukemia 2018; 32(6): 1307–1316.
European Medicines Agency. Guideline on the use of minimal residue disease as an endpoint in chronic lymphocytic leukaemia studies. 2014. https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-use-minimal-residue-disease-endpoint-chronic-lymphocytic-leukaemia-studies_en.pdf
Thompson PA. MRD negativity as a surrogate for PFS in CLL? Blood 2018; 131(9): 943–944.
Dimier N, et al. A model for predicting effect of treatment on progression-free survival using MRD as a surrogate end point in CLL. Blood 2018; 131(9): 955–962.
Fürstenau M, et al. Minimal residual disease assessment in CLL: Ready for use in clinical routine? Hemasphere 2019; 3(5): e287.
Burger JA, et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med. 2015; 373(25): 2425–2437.
Shanafelt TD, et al. ibrutinib-rituximab or chemoimmunotherapy for chronic lymphocytic leukemia. N Engl J Med. 2019; 381(5): 432–443.
Fraser G, et al. Updated results from the phase 3 HELIOS study of ibrutinib, bendamustine, and rituximab in relapsed chronic lymphocytic leukemia/small lymphocytic lymphoma. Leukemia 2019; 33(4): 969–980.
Ahn IE, et al. Depth and durability of response to ibrutinib in CLL: 5-year follow-up of a phase 2 study. Blood 2018; 131(21): 2357–2366.
Hillmen P, et al. Ibrutinib plus venetoclax in relapsed/refractory chronic lymphocytic leukemia: The CLARITY study. J Clin Oncol. 2019; JCO1900894.
Thompson PA, et al. Fludarabine, cyclophosphamide, and rituximab treatment achieves long-term disease-free survival in IGHV-mutated chronic lymphocytic leukemia. Blood 2016; 127(3): 303–309.
Thompson PA, Wierda WG. Eliminating minimal residual disease as a therapeutic end point: working toward cure for patients with CLL. Blood 2016; 127(3): 279–286.
Rawstron AC, et al. A complementary role of multiparameter flow cytometry and high-throughput sequencing for minimal residual disease detection in chronic lymphocytic leukemia: a European Research Initiative on CLL study. Leukemia 2016; 30(4): 929–936.
Rawstron AC, et al. International standardized approach for flow cytometric residual disease monitoring in chronic lymphocytic leukaemia. Leukemia 2007; 21(5): 956–964.
Logan AC, et al. Minimal residual disease quantification using consensus primers and high-throughput IGH sequencing predicts post-transplant relapse in chronic lymphocytic leukemia. Leukemia 2013; 27(8): 1659–1665.
Farina L, et al. Qualitative and quantitative polymerase chain reaction monitoring of minimal residual disease in relapsed chronic lymphocytic leukemia: early assessment can predict long-term outcome after reduced intensity allogeneic transplantation. Haematologica 2009; 94(5): 654–662.
Böttcher S, et al. Comparative analysis of minimal residual disease detection using four-color flow cytometry, consensus IgH-PCR, and quantitative IgH PCR in CLL after allogeneic and autologous stem cell transplantation. Leukemia 2004; 18(10): 1637–1645.
Rawstron AC, et al. Improving efficiency and sensitivity: European Research Initiative in CLL (ERIC) update on the international harmonised approach for flow cytometric residual disease monitoring in CLL. Leukemia 2013; 27(1): 142–149.
Sartor MM, Gottlieb DJ. A single tube 10-color flow cytometry assay optimizes detection of minimal residual disease in chronic lymphocytic leukemia. Cytometry B Clin Cytom. 2013; 84(2): 96–103.
Dowling AK, et al. Optimization and validation of an 8-color single-tube assay for the sensitive detection of minimal residual disease in B-cell chronic lymphocytic leukemia detected via flow cytometry. Lab Med. 2016; 47(2): 103–111.
Stehlíková O, et al. Detecting minimal residual disease in patients with chronic lymphocytic leukemia using 8-color flow cytometry protocol in routine hematological practice. Int J Lab Hematol. 2014; 36(2): 165–171.
Abrisqueta P, et al. Rituximab maintenance after first-line therapy with rituximab, fludarabine, cyclophosphamide, and mitoxantrone (R-FCM) for chronic lymphocytic leukemia. Blood 2013; 122(24): 3951–3959.
Fischer K, et al. Bendamustine in combination with rituximab for previously untreated patients with chronic lymphocytic leukemia: a multicenter phase II trial of the German Chronic Lymphocytic Leukemia Study Group. J Clin Oncol. 2012; 30(26): 3209–3216.
Li J, et al. Population pharmacokinetics of rituximab in patients with chronic lymphocytic leukemia. J Clin Pharmacol. 2012; 52(12): 1918–1926.