Approximately 85% of patients aged under 45 years will survive for at least five years after chemo- and/or radiotherapy.1 Despite the gonadotoxicity of these treatments, the possibility for women to conceive a child after surviving cancer is a reality that can be envisaged thanks to advances in fertility preservation methods. Ovarian tissue cryopreservation (OTC) is one of the available options to preserve fertility before receiving treatment that could result in sterility.2 Currently, frozen/thawed ovarian tissue can only be re-used by autograft, and more than twenty live births have already been reported.43 However, there is legitimate concern about the risk of cancer re-seeding by transplanting the tissue in cases of cancer.5 In acute leukemia (AL), real-time quantitative polymerase chain reaction (RT-qPCR)6 or multicolor flow cytometry (MFC) based on leukemia-associated immunophenotype (LAIP)7 can be used to identify persisting leukemic cells at low level in ovarian tissue. Two studies have investigated minimal residual disease (MRD) in ovarian tissue in the setting of acute myeloid leukemia (AML) by RT-qPCR.98 MFC makes it possible to differentiate leukemic cells from normal progenitors and hematopoietic stem cells, based on a specific LAIP profile in 95% of AML patients in bone marrow (BM).10 MFC has been used before by our team to quantify MRD in the ovarian tissue of acute lymphoid leukemia (ALL) patients.11 The objective of the current study was to develop a technique for MRD detection using 8-color flow cytometry in ovarian tissue from AML patients.
Ovarian cortical fragments, obtained by oophorectomy or biopsies, were cryopreserved using slow cooling, stored in liquid nitrogen, thawed, and then underwent mechanical and enzymatic dissociation according to previously described protocols.11 Healthy reference ovarian tissue samples were obtained from women undergoing laparoscopic surgery for polycystic ovary syndrome (PCOS). AML blood or BM cells were used for validation of an experimental model of serial AML cell dilutions in ovarian cell suspensions. Cryopreserved ovarian tissue from 4 AML patients was used for MRD assessment. All 4 patients had received chemotherapy before harvesting of ovarian tissue. The use of human ovarian tissue for this study was approved by the Clinical Ethics Committee (9 June 2010) and by the Ethical Research Committee of Besançon University Hospital (5 March 2013). All patients gave written informed consent. MFC was performed using a FACSCanto II™ flow cytometer and FACSDiva™ software v.6.1.3 (BD Biosciences, San Jose, CA, USA). The freezing/thawing and dissociation steps did not modify cell surface marker expression.11 MFC gating strategy was based on the elimination of debris by an initial morphological gate using forward (FSC) and side (SSC) light scatter characteristics. Nucleated viable cells were then selected by their SYTO13/7AAD phenotype, giving the total events used for MRD quantification. Within these cells, we identified CD45 hematopoietic cells and CD3 T lymphocytes. The myeloid blasts were gated with 4 additional antibodies. Acquisition of at least 200,000 viable ovarian cells was performed as required in BM MRD assessment, except for one case where only 130,000 events were acquired (range 130,000–570,000). A significant abnormal cell population was defined as a homogeneous cluster of at least 20 LAIP events.7 To assess the specificity of the 8 antibody combinations used and the maximum sensitivity corresponding to the limit of detection that can be achieved for MRD, a quantitative study of ovarian cells from reference ovarian tissue was performed in the same conditions. The sensitivity of the MFC method was confirmed by 10-fold serial dilutions of AML cells in reference ovarian cell suspensions (10 to 10).11 Molecular quantification of NPM1 mutation A and CBFB-MYH11 were performed as previously described.1312 ABL1 was used as housekeeping gene.
Markers used in routine clinical practice for AML MRD detection by MFC in blood or BM (CD13 and CD33, CD14, HLA DR) were applied to AML cell detection in our experimental model of blood or BM leukemic cell dilutions (10 to 10) within reference ovarian cells. MRD quantification by MFC reached a plateau above the level of 10 (mean 4×10) (Figure 1A). This was explained by background noise due to the expression of CD13 and CD33, CD14 or HLA DR on ovarian cells, thus precluding achievement of sufficient sensitivity for MRD detection by MFC (Figure 1A). We therefore tested CD33 alone and other potentially useful AML markers (such as CD43 or CD361), and did not observe any background signal on ovarian cells with these new antibody combinations. We obtained robust sensitivity of 10 (Figure 1B), with a good correlation between experimental and theoretical MRD values (r=0.97, P<0.0001, n=37). Molecular MRD quantification was performed in dilution experiments using AML cells originating from patients carrying NPM1 mutation A. On analysis of 24 dilution points by both MFC and RT-qPCR, one sample was negative by both techniques and 19 were positive at or above 1×10 by both techniques with good correlation (r=0.95) (Figure 1C). Three dilution points were positive (>1×10) by RT-qPCR but negative by MFC (<1×10, between 5 and 10 LAIP events), and one sample was considered positive below 1×10 by RT-qPCR but negative by MFC. Thus, these results seem to indicate that there is a good correlation between MFC and RT-qPCR for positive MRD levels up to 10, and an increased sensitivity of RT-qPCR in cases of low positive MRD levels. Finally, for 4 AML patients, we investigated ovarian MRD by MFC (n=4) and RT-qPCR (CBFB-MYH11 quantification; n=1) (Table 1). Among these 4 patients, ovarian MRD results were positive by MFC in 2 cases. For Patient #1, 56 LAIP events presenting the same phenotype as the patient’s own AML cells at the time of diagnosis (Figure 2A) were detected among 3×10 viable ovarian cortical cells; MRD was quantified at 2×10 (Figure 2B). Moreover, AML cells were also found in the ovarian medulla of this patient at 5×10 (Figure 2C). For Patient #4, MRD was positive in the ovarian cortical tissue at a level of 3×10. We were not able to analyze more than 133,000 viable nucleated ovarian cells. However, we quantified 34 LAIP events, corresponding to positive MRD of 3×10. Patient #2 was MRD-negative by both MFC and CBFB-MYH11 RT-qPCR while Patient #3 was MRD-negative by MFC (Table 1).
This study demonstrates that MFC is a powerful tool for MRD detection in ovarian tissue from patients who undergo OTC before gonadotoxic treatment. Two studies published about ovarian MRD detection in AML patients98 suggest that immunohistochemistry is unable to identify a very low number of AML cells in ovarian tissue. Although RT-qPCR is known to have good sensitivity for MRD detection (10 to 10), this technique was only applied in one of 10 AML patients in these studies. Xenotransplanted ovarian tissue with positive MRD may have failed to induce AML development in mice because too few AML cells were present in the transplanted ovarian pieces. The xenograft model may not be accurate for ovarian AML MRD detection, but is currently the only available method to evaluate potential in vivo proliferation of leukemic cells. We describe here the use of MFC to quantify AML cells in ovarian tissue. Ovarian MRD detection by MFC can be applied to all AML patients using a single standardized myeloid MRD tube. The experimental model of serial dilutions of AML cells in an ovarian cell suspension made it possible to validate robust sensitivity of 10 (20 events with abnormal phenotype among 200,000 viable events). The determination of the appropriate antibody combination for MRD assessment is very important to achieve good specificity. In this study, switching two antibodies enabled us to obtain approximately one log increase in sensitivity, and we observed a good correlation between MFC and RT-qPCR MRD results in the dilution experiments. MRD detection by MFC was applied to cryopreserved ovarian tissue from 4 AML patients. MRD was positive in 2 AML patients at very low levels (10 to 10). The clinical significance of these positive MRD results must be interpreted with caution, since the ovarian MRD level that can induce a relapse after ovarian tissue transplantation is currently unknown. An MRD threshold mandatory for ovarian tissue transplantation should be specified in the future, and for this purpose ovarian tissue xenograft could be an interesting option. Two other options are currently under investigation to eliminate the risk of re-introducing disease during ovarian tissue autograft, namely in vitro maturation of oocytes14 and grafting of isolated ovarian follicles.15
In conclusion, we validate a technique to detect the presence of live AML cells in the ovarian cortex by MFC with high specificity and sensitivity. Improved MFC sensitivity could be achieved by performing the analysis on a larger number of cells. This MFC approach for ovarian MRD detection is currently the only available technique for ovarian MRD assessment when no molecular markers are available. MFC should promote the safe re-use of cortical ovarian fragments by autograft, without risk of re-introducing disease, in patients who have undergone gonadotoxic treatments.
References
- Howlader N, Noone A, Krapcho M, Neyman N, Aminou R, Altekruse S. SEER Cancer Statistics Review, 1975–2010. National Cancer Institute: Bethesda, MD; 2013. Google Scholar
- Donnez J, Dolmans MM. Fertility preservation in women. Nat Rev Endocrinol. 2013; 9(12):735-49. PubMedhttps://doi.org/10.1038/nrendo.2013.205Google Scholar
- Donnez J, Silber S, Andersen CY, Demeestere I, Piver P, Meirow D. Children born after autotransplantation of cryopreserved ovarian tissue. a review of 13 live births. Ann Med. 2011; 43(6):437-50. PubMedhttps://doi.org/10.3109/07853890.2010.546807Google Scholar
- Roux C, Amiot C, Agnani G, Aubard Y, Rohrlich PS, Piver P. Live birth after ovarian tissue autograft in a patient with sickle cell disease treated by allogeneic bone marrow transplantation. Fertil Steril. 2010; 93(7):2413e15-9. Google Scholar
- Bastings L, Beerendonk CC, Westphal JR, Massuger LF, Kaal SE, van Leeuwen FE. Autotransplantation of cryopreserved ovarian tissue in cancer survivors and the risk of reintroducing malignancy: a systematic review. Hum Reprod Update. 2013; 19(5):483-506. PubMedhttps://doi.org/10.1093/humupd/dmt020Google Scholar
- Cave H, van der Werff ten Bosch J, Suciu S, Guidal C, Waterkeyn C, Otten J. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia. European Organization for Research and Treatment of Cancer–Childhood Leukemia Cooperative Group. N Engl J Med. 1998; 339(9):591-8. PubMedhttps://doi.org/10.1056/NEJM199808273390904Google Scholar
- Campana D, Coustan-Smith E. Detection of minimal residual disease in acute leukemia by flow cytometry. Cytometry. 1999; 38(4):139-52. PubMedhttps://doi.org/10.1002/(SICI)1097-0320(19990815)38:4<139::AID-CYTO1>3.0.CO;2-HGoogle Scholar
- Greve T, Clasen-Linde E, Andersen MT, Andersen MK, Sorensen SD, Rosendahl M. Cryopreserved ovarian cortex from patients with leukemia in complete remission contains no apparent viable malignant cells. Blood. 2012; 120(22):4311-6. PubMedhttps://doi.org/10.1182/blood-2012-01-403022Google Scholar
- Rosendahl M, Andersen MT, Ralfkiaer E, Kjeldsen L, Andersen MK, Andersen CY. Evidence of residual disease in cryopreserved ovarian cortex from female patients with leukemia. Fertil Steril. 2010; 94(6):2186-90. PubMedhttps://doi.org/10.1016/j.fertnstert.2009.11.032Google Scholar
- Coustan-Smith E, Campana D. Should evaluation for minimal residual disease be routine in acute myeloid leukemia?. Curr Opin Hematol. 2013; 20(2):86-92. PubMedhttps://doi.org/10.1097/MOH.0b013e32835dd90aGoogle Scholar
- Amiot C, Angelot-Delettre F, Zver T, Alvergnas-Vieille M, Saas P, Garnache-Ottou F. Minimal residual disease detection of leukemic cells in ovarian cortex by eight-color flow cytometry. Hum Reprod. 2013; 28(8):2157-67. PubMedhttps://doi.org/10.1093/humrep/det126Google Scholar
- Gabert J, Beillard E, van der Velden VH, Bi W, Grimwade D, Pallisgaard N. Standardization and quality control studies of ‘real-time’ quantitative reverse transcriptase polymerase chain reaction of fusion gene transcripts for residual disease detection in leukemia - a Europe Against Cancer program. Leukemia. 2003; 17(12):2318-57. PubMedhttps://doi.org/10.1038/sj.leu.2403135Google Scholar
- Gorello P, Cazzaniga G, Alberti F, Dell’Oro MG, Gottardi E, Specchia G. Quantitative assessment of minimal residual disease in acute myeloid leukemia carrying nucleophosmin (NPM1) gene mutations. Leukemia. 2006; 20(6):1103-8. PubMedhttps://doi.org/10.1038/sj.leu.2404149Google Scholar
- Chian RC, Uzelac PS, Nargund G. In vitro maturation of human immature oocytes for fertility preservation. Fertil Steril. 2013; 99(5):1173-81. PubMedhttps://doi.org/10.1016/j.fertnstert.2013.01.141Google Scholar
- Luyckx V, Dolmans MM, Vanacker J, Legat C, Fortuno Moya C, Donnez J. A new step toward the artificial ovary: survival and proliferation of isolated murine follicles after autologous transplantation in a fibrin scaffold. Fertil Steril. 2014; 101(4):1149-56. PubMedhttps://doi.org/10.1016/j.fertnstert.2013.12.025Google Scholar