Abstract
Wilms tumor gene 1 (WT1) is an important gene which is involved in growth and development of many organs. It is identified as a tumor suppressor gene in nephroblastoma. However, its role as a tumor oncogene has been highlighted by many studies in haematological as well as non haematological malignant neoplasm. The expression of WT1 on leukemic blast cells sensitised us to explore its impact on neoplastic phenomenon. WT1 is has been found both mutated as well as over expressed in different subsets of acute myeloid leukemia (AML). WT1 is a gene has been used as a biomarker for diagnosis, monitoring of minimal residual disease (MRD) and detection of relapse for molecular remission in AML. It also has potential of being a predictive molecular predictive biomarker for the treatment of leukemic cases after allogeneic transplantation. The WT1 specific expression on blast cells and its interaction with cytotoxic T cell has also been explored for its potential usage WT1 based immunotherapy. Here, we are reviewing molecular updates of WT1 gene and discuss its potential clinical applications as a predictive molecular biomarker for diagnosis, as MRD detection and as immunotherapy in AML.
Keywords: WT1 gene, WT1 mutation, WT1 expression, AML
Introduction
Acute myeloid leukemia (AML) has been a heterogeneous group of disease with various combinations of clinical symptoms, aggressiveness and complications [1]. AML is the second most prevalent type of leukemia diagnosed in adults and children [2]. However, incidence of AML is directly proportional to the increasing age [3]. Several chromosomal rearrangements and mutations have been detected in AML and linked with the diagnosis, pathogenesis, and prognosis of AML [4,5]. The overall survival is also dependent on genomic profile of mutational burden [6]. In addition to mutations, significant modification at post transcriptional and posttranslational level may produce malignant cell changes [7]. WT1 gene is a tumor suppressor gene has been primarily linked with nephroblastoma and now being considered as a tumor oncogene, which plays a critical substantial role in neoplastic process related to hematopoietic malignancies [8].
WT1 gene has dual functioning of acting as tumor suppressor as well oncogenic gene and controls transcription, translation, RNA metabolism at cellular level [9,10]. WT1 mediated pathway of transcriptional regulation plays an impactful role in normal and malignant hematopoiesis [11]. Overexpression of WT1 specific iso-forms in myeloblast cells of AML with minimal maturation results in the induction of apoptosis and G1 arrest as well [12]. WT1 is a potent transcriptional regulatory molecule that plays a crucial role in the regulation of apoptosis, cell survival, promotes cell proliferation, cell growth, metastasis, differentiation, and normal cellular development [13,14].
Structure of WT1
The WT1 gene is located on chromosome 11p13 and is 50 kb in length. It consists of 3.2 kb of mRNA produced from total 10 exons. It regulates the expression and encodes a zinc finger transcription factor, which controls cellular growth and metabolism, including growth factors, extracellular matrix components through binding to GC-rich homologous sequence, and regulates transcription activator or represses expression of specific target genes (ECM) [15,16]. Mutations of the WT1 gene lead to disorders such as Wilms’ tumors or Denys-Drash syndrome [17,18]. It is implicated in the development of organ systems such as the kidney, retina, spleen, and heart through various signalling pathways [19,20]. It also promotes epithelial-to-mesenchymal transition (EMT) [21]. WT1 mRNA shows two important splicing regions [22,23]. These include splicing of exon 5, which encodes 17 amino acids (AA) and another segment of nine nucleotides which code for 3 amino acids such as lysine, threonine, and serine (KTS) on exon 9 [24]. Alternative splice 2 inserts three amino acids-lysine [K], threonine [T], and serine [S] between exons 9 and 10, alter the conserved spacing between zinc fingers 3 and 4, leads to the significant reduction of the DNA binding ability but enhances RNA binding [23,25]. The alternative splicing of these two positions gives rise to four different protein isoforms: WT1 A (17AA-/KTS-), WT1 B (17AA-/KTS+), WT1 C (17AA+/KTS-), and WT1 D (17AA+/KTS+). KTS-positive isoforms constitute 80% of cellular WT1 [16,26,27]. The full-length product encoded by WT1 is a 57 kD protein.
Function of WT1
The N-terminal terminal transactivation domain of WT1 is composed of proline, glutamic acid, serine, and glycine-rich sequences [28,29]. This N-terminal domain is relevant for transcriptional regulatory function of WT1, such as transcriptional repression [30]. The C-terminal of WT1 is composed of four zinc-fingers moieties, each of which has two cysteine and two histidine [31]. This zinc finger permits binding of target DNA sequences, regulating gene transcription such as RNA and protein interactions. Thus truncated WT1 might exhibit oncogenic properties [32].
WT1 in normal haematopoiesis
Differential expression of WT1 isoforms may support isoform-specific differentiation in hematopoiesis and leukemogenesis [33]. The KTS insert significantly increases the flexibility of protein [34] by limiting related binding sites at the major groove in DNA. KTS2 isoforms may repress or activate transcription in normal hematopoietic cells [35]. KTS2 isoforms is arrests cells in G1 phase and induces myelomonocytic differentiation of CD34 positive hematopoietic progenitor cells [36]. The KTS forms also co-localize preferentially with already available ubiquitous transcription factors. In contrast, most of KTS(+) isoforms are found in a speckled pattern and co-localize with small nuclear ribonucleoprotein particles (snRNPs), suggesting a role in site specific splicing [37]. WT1 KTS(+) does not influences p21 expression however it promotes EMT, specially within solid neoplastic diseases [38]. However, WT1 KTS(-) increases p21 expression and cell proliferation with diminishing reproductive potency and G2 arrest [39]. The WT1 KTS(-) isoform more strongly enhances CD95L mediated cell death in T-cell acute lymphoblastic leukemia (T-ALL) [40]. The major WT1 subtypes have inhibitory functions, e.g., WT1 KTS [+], WT1 KTS(-) can inhibit the expression of the apoptotic genes such as p53, Bak, Bax or caspase-9, also induces expression of transcription factor BCL2; thus promoting an anti-apoptotic effect. One study on targeted transgenic murine model reported that WT1 was not found in long term-hematopoietic stem cells. Deletion of WT1 among young and adult mice resulted in death of animals in time frame of approximately 10 days with following causes such as glomerulosclerosis, atrophy of pancreas, and diminished extramedullary hematopoiesis [41].
WT1 in AML
The impact of WT1 gene mutation on AML was first identified in 1994 by King-Underwood and his colleagues on its possible role of drug resistance [42]. Recurrent somatic mutations in WT1 appear to occur in approximately 6-15% of newly diagnosed cases of AML [41]. These include deletions, insertions, and base substitutions mutations, primarily targeting exon 7 and 9 [43-48]. However, the vast majority of mutations resulting in loss-of-function, and expression of truncated proteins perform in a dominant-negative manner, which may contribute to a myeloid differentiation block present in AML blasts [46].
WT1 mutations are usually denoted by loss-of-normal function. Mutational analysis of a large cohort of AML cases demonstrated that mutations of WT1 are mutually exclusive with ten-eleven translocation methylcytosine dioxygenase 2 (TET2), isocitrate dehydrogenase 1 (IDH1), isocitrate dehydrogenase 2 (IDH2), or CCAAT enhancer-binding protein alpha [CEBPA] mutations [47,48]. Rampal et al. revealed that TET2 catalyzes the conversion of 5-methylcytosine (5-mC) to 5-hydroxymethylcytosine (5-hmC) [49]. TET2 loss-of-function mutations and IDH1/2 mutations result in inhibition of the DNA demethylation pathways with an accumulation of 5-mC and a decrease of 5-hmC. WT1 mutations attenuate the TET2 function; a reduction in WT1 would potentially reduce TET2 activity in AML. Several studies confirmed that similar epigenetic alterations characterize WT1-mutant AMLs, as found in TET2 and IDH1/2 mutant AML. WT1-mutant AMLs presented a global reduction in 5-hydroxymethylcytosine (5-hmC) levels and interruption of gene-regulatory interfaces required for normal hematopoiesis. WT1 with TET2 as a cofactor, transcriptionally regulates maternally expressed gene 3 (MEG3) expression. MEG3 induces GO/G1-phase & apoptosis and decreases cell proliferation by regulating p53 expression. In WT1 or TET2 mutated AML cell lines, the lncRNA of maternally expressed gene 3 (MEG3) is significantly downregulated [50].
Subsequently, numerous studies emphasized possible impact of WT1 gene expression irrespective of their mutational status as an independent factor. Various researches have established relationship between WT1 gene expression and AML. There has been a most recent study by Liu et al. [51] from china where WT expression was studied on 195 odd cases of AML and established as marker of MRD. Rautenberg et al. [52] studied WT gene expression on post allogeneic transplant cases of AML & MDS to utilize it as marker for prediction of relapse. Nomedede et al. [53] proved it as marker of prognostic marker among 585 cases of AML in European region. A study from china [54] proved WT1 gene expression as an independent prognostic marker in cases of acute leukemia. There have been two independent studies [55,56] from Italy where WT1 gene expression has been evaluated as marker for risk stratification and long term progression respectively.
Therapeutic implications of WT1 in AML
There has been a trial to attempt to use peptide based vaccines against WT1 especially in cases where given overexpression has been documented. Numerous studies on WT immunotherapy have been carried till date using a variety of different vaccination strategies, for example (HLA-restricted versus non HLA-restricted peptides). Clinically meaningful responses have been reported in several trials in both AML and MDS cases, with associated increases in WT1-specific T-cell frequencies (Table 1) [57-66]. The expression of WT1 on normal tissues and its role in normal haematopoiesis has been an issue of probable possibility of autoimmune phenomenon. However, there have been no reports till date [49]. This approach has, thus demonstrated clinical efficacy but still requires further large-scale evaluation. Another alternative approach has been development of monoclonal antibodies that may recognizes a peptide fragment of WT1 complexed with HLA-A0201. This antibody demonstrated efficacy in a NOD/SCID mouse xenotransplanted with human leukemias [49].
Table 1.
Year | Research Group | Trial no clinicaltrials. gov/show/ | Inclusion criteria | Clinical Outcome | Ref no |
---|---|---|---|---|---|
2018 | Maslak et al. | NCT01266083 | AML in CR | Stimulated specific immune response | [57] |
2018 | Nakata et al. | UMIN000015870 | AML in CR | Molecular CR maintained for 3.5 years | [58] |
2018 | Liu et al. | NCT01842139 | AML in CR | WT1 vaccine with Montanide induced CD8 response | [59] |
2017 | Anguille et al. | NCT00965224 | AML in CR | Delay in relapse was seen in vaccine group | [60] |
2017 | Kobayashi et al. | NCT2001440920 | AML in CR | Reduction in WT mRNA transcripts | [61] |
2015 | Brayer et al. | NCT00665002 | AML in CR | Vaccinations well tolerated. Relapse-free survival >1 year | [62] |
2011 | Rezvani et al. | NCT00488592 | AML in CR & MDS | CD8 response detected in all evaluable patients | [63] |
2010 | Maslak et al. | NCT00398138 | AML in CR | Promising disease-free and overall survival | [64] |
2008 | Rezvani et al. | NCT00433745 | AML in CR & MDS | Reduced in WT1 mRNA expression | [65] |
2004 | Mailander et al. | NCT00153582 | Recurrent AML | Vaccine induced CR in recurrent AML absence of toxicity | [66] |
CR, complete remission; AML, acute myeloid leukemia; MDS, Myelodysplastic Syndrome.
Prognostic implications of WT1 in AML
The undetectable leukemic stem cells (LSC) on morphology are strong independent factor for future relapse [67]. The monitoring of MRD is of utmost importance to prevent future relapse and improve overall survival. Detectable levels of WT1 expression during follow-up in AML cases are a potential marker for assessment of residual blast populations or even to predict future relapse of AML [68]. However, there are indicators for the involvement of WT1 in malignant events of AML blasts, such as the interactions of WT1 with the proto-oncogene bcl-2 and tumor suppressor gene p53 [69]. Despite advancement in development of new treatments protocols, many cases are refractory to ongoing therapeutic strategies. They have a higher relapse rate, with overall long-term survival of patients signifying below 40% and more than 60% among cases over 65 years of age succumbing to disease within one year of diagnosis [70]. It is assumed that relapses originate from undetectable populations of LSC, which are characterized by a pronounced self-renewal capacity that evade traditional chemotherapy [71]. In vitro killing of tumor cells by WT1-specific CD81 cytotoxic T lymphocytes facilitated the development of a WT1 based vaccine. WT1-specific immunotherapy might be useful to optimize multimodal therapy of haematological malignancies [72,73]. There are various studies which have correlated the expression of WT1 and data of AML cases. Lovvik et al. observed that in AML cases at primary diagnosis, 66% had more than 20-fold WT1 overexpression in peripheral blood (PB) or bone marrow (BM) (PB 74%; BM 45%) [74]. In another study, Ho et al. revealed that in a group of paediatric AML, a significant difference in event-free survival and leukemia-free survival for cases with high versus low WT1 expression was found [75]. Nomdedeu et al. reported that de novo AML patients with high-level WT1 expression with 3-year overall survival (OS) was only 19%, whereas patients with low-level WT1 expression 3-year overall survival (OS) was about 64% [54]. Brieger et al. showed in his study of 52 AML cases WT1 gene was overexpressed in 41/52 (79%) patients at the time of first diagnosis. The majority of the 14 patients lost WT1 expressions that were studied in CR, where as in 4 cases reappearance of WT1 expression estimated before relapse [76]. In one of the study with background knowledge of WT1 and Bcl2 controlling apoptosis, WT1 expression and proto-oncogene Bcl-2 was estimated simultaneously as prognostic markers of AML treatment outcome. The study showed that increased WT1 and Bcl-2 expression was associated with reduced rate of continuing complete remission and increased deaths among AML cases with age less than 60 years [77].
Application of WT1 for detection of minimal residual disease
WT1 is a potential marker for detection of MRD in AML. The identification of MRD has led to substantial improvements in early recognition of recurrence of AML [78]. Many studies have determined that the MRD assessment provided evidence to stratify high-risk AML patients better and give significant insight into the effectiveness of treatment. Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) serves as an effective treatment strategy for high-risk patients with AML in CR [79]. WT1 expression affects prognosis of Allo-HSCT in AML [80]. Furthermore, increased WT1 expression was associated more with higher rate of relapse compared, with cases under remission were constantly associated with low levels [81]. There are studies which have showed association between expression of WT1 gene and chances of relapse after allogeneic stem cell transplant (Allo-HSCT). Ogawa et al. found that higher WT1 levels after Allo-SCT were associated with increased chances of relapse. There was constant doubling time in WT exponential expression in sust of cases having relapse. The significance of WT1 log reduction after induction chemotherapy to be an independent predictor of relapse [82]. Weisser et al. observed that higher than 2 log decline in WT1 transcript levels from the beginning of chemotherapy was correlated with a significantly improved Overall Survival (OS) and event-free survival (EFS). After induction chemotherapy, decreased WT1 gene expression in AML patients conferred a more favourable prognosis and correlated with higher Overall Response Rate (ORR), and 2-yr overall survival rates and disease-free survival (DFS) rate were highly significant (P-value <0.05) [83]. In an another study, Cilloni et al. demonstrated that less than 2 log decline in WT1 transcripts after induction therapy enhanced the significant risk of relapse in patients with AML [51]. While another research group, Ido et al. showed association between expression levels of WT1 before and after Allo-HSCT and the risk of fatality among AML. In patients with AML who underwent Allo-HSCT after two years, the fatality rate was significantly lower in AML patients who are having low expression levels of WT1 when compared with cases having high expression level of WT1. Furthermore, in the whole cohort of AML patients, WT1 mRNA ≥5000 copies/μg RNA before Allo-HSCT was significantly associated with an increased risk of mortality [84]. These studies are suggesting that the WT1 mRNA level might reflect tumor burden.
Future research on WT immunotherapy in AML
The virtue of being a pan leukemia markers makes WT1 as a potential target for immunotherapy. Both higher expression of WT1 and mutation in WT1 gene is involved in AML has lead to both clinical and preclinical therapeutic strategies in hematological and solid malignancies such as uterine sarcoma [85-87]. In cases where raised WT1 expression has been estimated, WT1 based immunotherapy combined with standard neoadjuvant therapy induces T cell recognition of tumor antigens by vaccination and induces immune response to produce tumor antibodies in breast cancer cases [88]. WT1 peptide vaccination can induce cells of WT1-specific cytotoxic T lymphocytes, prevent relapse, and sustains long-term, complete remission in AML [89]. After WT1 vaccination, most studies showed that the number of granulocytes, lymphocytes, and leukemia blast cells were reduced. Phase I study conducted mice vaccinated with Mycobacterium Bovis bacillus Calmette-Guerin cell-wall skeleton (BCG-CWS) with WT1 peptide survived significantly longer when compared with a non-vaccinated mice. Thus, adjuvant like BCG-CWS may prove to enhance the clinical efficacy of WT1 vaccine for humans [90]. Currently WT1 vaccine based immunotherapy research are in early trials and phase I studies have generated positive inclinations. This has promoted phase II trials to evaluate it further, once phase II clinical trial has been carried out on AML cases. The treatment with WT1 peptide was combined with treatment with granulocyte-macrophage colony-stimulating factor [GM-CSF]. This cytokine is used as an adjuvant and functions as a white blood cell growth factor. The treatment with this vaccine was well-tolerated, blast reduction and hematological improvement were seen in some patients; the results were overall promising [91,92]. Such in vitro findings have led to design of the human WT1 vaccine. Recently several studies (Table 1) have reported the safety and efficacy with favorable results concerning the use of the WT1 peptide vaccine in patients with AML. These results showed that the WT1 vaccine is well tolerated, stimulates a specific immune response, and can improve the prognosis of patients.
Conclusion
The newer advancement in field of molecular genetics and therapeutic researches has drastically improved the overall survival figure for a complex heterogeneous disease such as AML. Our understanding on the impact of mutation and expression of WT1 gene on AML has greatly improved. This has facilitated application of WT1 as as potential biological marker for diagnosis, clinical management, monitoring of therapy, detection of MRD and Immunotherapy. The current article has tried to emphasize the upcoming application of WT1 based interventions in AML. WT1 encodes a transcription factor that plays a regulatory role in normal and malignant haematopoiesis. Frequent monitoring of the WT1 gene expression level during follow-ups in AML patients is useful as a marker for residual blast populations or even to predict the risk of relapse following allogeneic SCT. The current review validates WT1 as promising potential biomarker for AML on the basis of available published medical literature.
Acknowledgements
We are thankful to Science and Engineering Research Board, Department of Science and Technology, Government of India through Grant No. EEQ/2016/000318 for supporting resources in this study.
Disclosure of conflict of interest
None.
References
- 1.Goryainova NV. The clinical significance of genetic mutations in acute myeloid leukemia. Lik Sprava. 2014;12:10–18. [PubMed] [Google Scholar]
- 2.Siegel RL, Miller KD, Jemal A. Cancer statistics 2020. CA Cancer J Clin. 2020;1:7–30. doi: 10.3322/caac.21590. [DOI] [PubMed] [Google Scholar]
- 3.Appelbaum FR, Gundacker H, Head DR, Slovak ML, Willman CL, Godwin JE, Anderson JE, Petersdorf SH. Age and acute myeloid leukemia. Blood. 2006;9:3481–3485. doi: 10.1182/blood-2005-09-3724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lagunas-Rangel FA, Chavez-Valencia V, Gomez-Guijosa MA, Cortes-Penagos C. Acute myeloid leukemia-genetic alterations and their clinical prognosis. Int J Hematol Oncol Stem Cell Res. 2017;4:328–339. [PMC free article] [PubMed] [Google Scholar]
- 5.De Kouchkovsky I, Abdul-Hay M. Acute myeloid leukemia: a comprehensive review and 2016 update. Blood Cancer J. 2016;7:e441. doi: 10.1038/bcj.2016.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Stadhouders R, Filion GJ, Graf T. Transcription factors and 3D genome conformation in cell-fate decisions. Nature. 2019;7756:345–354. doi: 10.1038/s41586-019-1182-7. [DOI] [PubMed] [Google Scholar]
- 7.Janin M, Coll-SanMartin L, Esteller M. Disruption of the RNA modifications that target the ribosome translation machinery in human cancer. Mol Cancer. 2020;1:70. doi: 10.1186/s12943-020-01192-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Lindstedt I, Lindgren MA, Andersson E, Engstrom W. The WT1 gene-its role in tumourigenesis and prospects for immunotherapeutic advances. In Vivo. 2014;5:675–681. [PubMed] [Google Scholar]
- 9.Toska E, Roberts SG. Mechanisms of transcriptional regulation by WT1 (Wilms’ tumour 1) Biochem J. 2014;1:15–32. doi: 10.1042/BJ20131587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lv L, Chen G, Zhou J, Li J, Gong J. WT1-AS promotes cell apoptosis in hepatocellular carcinoma through down-regulating of WT1. J Exp Clin Cancer Res. 2015;34:119. doi: 10.1186/s13046-015-0233-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Ariyaratana S, Loeb DM. The role of the Wilms tumour gene (WT1) in normal and malignant haematopoiesis. Expert Rev Mol Med. 2007;14:1–17. doi: 10.1017/S1462399407000336. [DOI] [PubMed] [Google Scholar]
- 12.Loeb DM. WT1 influences apoptosis through transcriptional regulation of Bcl-2 family members. Cell Cycle. 2006;12:1249–1253. doi: 10.4161/cc.5.12.2807. [DOI] [PubMed] [Google Scholar]
- 13.Belluti S, Rigillo G, Imbriano C. Transcription factors in cancer: when alternative splicing determines opposite cell fates. Cells. 2020;3:760. doi: 10.3390/cells9030760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Boublikova L, Kalinova M, Ryan J, Quinn F, O’Marcaigh A, Smith O, Browne P, Stary J, McCann SR, Trka J, Lawler M. Wilms’ tumor gene 1 (WT1) expression in childhood acute lymphoblastic leukemia: a wide range of WT1 expression levels, its impact on prognosis and minimal residual disease monitoring. Leukemia. 2006;2:254–263. doi: 10.1038/sj.leu.2404047. [DOI] [PubMed] [Google Scholar]
- 15.Sakamoto Y, Yoshida M, Semba K, Hunter T. Inhibition of the DNA-binding and transcriptional repression activity of the Wilms’ tumor gene product, WT1, by cAMP-dependent protein kinase-mediated phosphorylation of Ser-365 and Ser-393 in the zinc finger domain. Oncogene. 1997;17:2001–2012. doi: 10.1038/sj.onc.1201391. [DOI] [PubMed] [Google Scholar]
- 16.Yang L, Han Y, Suarez Saiz F, Minden MD. A tumor suppressor and oncogene: the WT1 story. Leukemia. 2007;5:868–876. doi: 10.1038/sj.leu.2404624. [DOI] [PubMed] [Google Scholar]
- 17.Hashimoto H, Zhang X, Zheng Y, Wilson GG, Cheng X. Denys-Drash syndrome associated WT1 glutamine 369 mutants have altered sequence-preferences and altered responses to epigenetic modifications. Nucleic Acids Res. 2016;21:10165–10176. doi: 10.1093/nar/gkw766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Mrowka C, Schedl A. Wilms’ tumor suppressor gene WT1: from structure to renal pathophysiologic features. J Am Soc Nephrol. 2000;(Suppl 16):S106–S115. [PubMed] [Google Scholar]
- 19.Miller-Hodges E, Hohenstein P. WT1 in disease: shifting the epithelial-mesenchymal balance. J Pathol. 2012;2:229–240. doi: 10.1002/path.2977. [DOI] [PubMed] [Google Scholar]
- 20.Scholz H, Kirschner KM. A role for the Wilms’ tumor protein WT1 in organ development. Physiology (Bethesda) 2005;1:54–59. doi: 10.1152/physiol.00048.2004. [DOI] [PubMed] [Google Scholar]
- 21.Park J, Kim DH, Shah SR, Kim HN, Kshitiz , Kim P, Quinones-Hinojosa A, Levchenko A. Switch-like enhancement of epithelial-mesenchymal transition by YAP through feedback regulation of WT1 and Rho-family GTPases. Nat Commun. 2019;1:2797. doi: 10.1038/s41467-019-10729-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Stoll R, Lee BM, Debler EW, Laity JH, Wilson IA, Dyson HJ, Wright PE. Structure of the Wilms tumor suppressor protein zinc finger domain bound to DNA. J Mol Biol. 2007;5:1227–1245. doi: 10.1016/j.jmb.2007.07.017. [DOI] [PubMed] [Google Scholar]
- 23.Ullmark T, Montano G, Gullberg U. DNA and RNA binding by the Wilms’ tumour gene 1 (WT1) protein +KTS and -KTS isoforms-from initial observations to recent global genomic analyses. Eur J Haematol. 2018;3:229–240. doi: 10.1111/ejh.13010. [DOI] [PubMed] [Google Scholar]
- 24.Bor YC, Swartz J, Morrison A, Rekosh D, Ladomery M, Hammarskjold ML. The Wilms’ tumor 1 (WT1) gene (+KTS isoform) functions with a CTE to enhance translation from an unspliced RNA with a retained intron. Genes Dev. 2006;12:1597–1608. doi: 10.1101/gad.1402306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Wells J, Rivera MN, Kim WJ, Starbuck K, Haber DA. The predominant WT1 isoform (+KTS) encodes a DNA-binding protein targeting the planar cell polarity gene Scribble in renal podocytes. Mol Cancer Res. 2010;7:975–985. doi: 10.1158/1541-7786.MCR-10-0033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Baudry D, Faussillon M, Cabanis MO, Rigolet M, Zucker JM, Patte C, Sarnacki S, Boccon-Gibod L, Junien C, Jeanpierre C. Changes in WT1 splicing are associated with a specific gene expression profile in Wilms’ tumour. Oncogene. 2002;36:5566–5573. doi: 10.1038/sj.onc.1205752. [DOI] [PubMed] [Google Scholar]
- 27.Haber DA, Buckler AJ, Glaser T, Call KM, Pelletier J, Sohn RL, Douglass EC, Housman DE. An internal deletion within an 11p13 zinc finger gene contributes to the development of Wilms’ tumor. Cell. 1990;7:1257–1269. doi: 10.1016/0092-8674(90)90690-g. [DOI] [PubMed] [Google Scholar]
- 28.Han Y, San-Marina S, Yang L, Khoury H, Minden MD. The zinc finger domain of Wilms’ tumor 1 suppressor gene (WT1) behaves as a dominant negative, leading to abrogation of WT1 oncogenic potential in breast cancer cells. Breast Cancer Res. 2007;4:R43. doi: 10.1186/bcr1743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lee KY, Jeon YJ, Kim HG, Ryu J, Lim DY, Jung SK, Yu DH, Chen H, Bode AM, Dong Z. The CUG-translated WT1, not AUG-WT1, is an oncogene. Carcinogenesis. 2017;12:1228–1240. doi: 10.1093/carcin/bgx108. [DOI] [PubMed] [Google Scholar]
- 30.Kim J, Lee K, Pelletier J. The DNA binding domains of the WT1 tumor suppressor gene product and chimeric EWS/WT1 oncoprotein are functionally distinct. Oncogene. 1998;8:1021–1030. doi: 10.1038/sj.onc.1201616. [DOI] [PubMed] [Google Scholar]
- 31.Ravasi T, Huber T, Zavolan M, Forrest A, Gaasterland T, Grimmond S, Hume DA RIKEN GER Group; GSL Members. Systematic characterization of the zinc-finger-containing proteins in the mouse transcriptome. Genome Res. 2003;13:1430–1442. doi: 10.1101/gr.949803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Morris JF, Madden SL, Tournay OE, Cook DM, Sukhatme VP, Rauscher FJ 3rd. Characterization of the zinc finger protein encoded by the WT1 Wilms’ tumor locus. Oncogene. 1991;12:2339–2348. [PubMed] [Google Scholar]
- 33.Maugeri G, D’Amico AG, Rasa DM, Reitano R, Saccone S, Federico C, Parenti R, Magro G, D’Agata V. Expression profile of Wilms Tumor 1 (WT1) isoforms in undifferentiated and all-trans retinoic acid differentiated neuroblastoma cells. Genes Cancer. 2016;7:47–58. doi: 10.18632/genesandcancer.94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Laity JH, Dyson HJ, Wright PE. Molecular basis for modulation of biological function by alternate splicing of the Wilms’ tumor suppressor protein. Proc Natl Acad Sci U S A. 2000;22:11932–11935. doi: 10.1073/pnas.97.22.11932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yang Z, Hong SH, Privalsky ML. Transcriptional anti-repression. Thyroid hormone receptor beta-2 recruits SMRT corepressor but interferes with subsequent assembly of a functional corepressor complex. J Biol Chem. 1999;52:37131–37138. doi: 10.1074/jbc.274.52.37131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Jensen HA, Yourish HB, Bunaciu RP, Varner JD, Yen A. Induced myelo-monocytic differentiation in leukemia cells is accompanied by noncanonical transcription factor expression. FEBS Open Bio. 2015;5:789–800. doi: 10.1016/j.fob.2015.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lamond AI. RNA processing. Wilms’ tumour-the splicing connection? Curr Biol. 1995;8:862–865. doi: 10.1016/s0960-9822(95)00174-6. [DOI] [PubMed] [Google Scholar]
- 38.Sampson VB, David JM, Puig I, Patil PU, de Herreros AG, Thomas GV, Rajasekaran AK. Wilms’ tumor protein induces an epithelial-mesenchymal hybrid differentiation state in clear cell renal cell carcinoma. PLoS One. 2014;9:e102041. doi: 10.1371/journal.pone.0102041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Moriya S, Takiguchi M, Seki N. Expression of the WT1 gene -KTS domain isoforms suppresses the invasive ability of human lung squamous cell carcinoma cells. Int J Oncol. 2008;2:349–356. [PubMed] [Google Scholar]
- 40.Bourkoula K, Englert C, Giaisi M, Köhler R, Krammer PH, Li-Weber M. The Wilms’ tumor suppressor WT1 enhances CD95L expression and promotes activation-induced cell death in leukemic T cells. Int J Cancer. 2014;2:291–300. doi: 10.1002/ijc.28379. [DOI] [PubMed] [Google Scholar]
- 41.Rampal R, Figueroa ME. Wilms tumor 1 mutations in the pathogenesis of acute myeloid leukemia. Haematologica. 2016;6:672–9. doi: 10.3324/haematol.2015.141796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.King-Underwood L, Pritchard-Jones K. Wilms’ tumor (WT1) gene mutations occur mainly in acute myeloid leukemia and may confer drug resistance. Blood. 1998;8:2961–2968. [PubMed] [Google Scholar]
- 43.Gaidzik VI, Schlenk RF, Moschny S, Becker A, Bullinger L, Corbacioglu A, Krauter J, Schlegelberger B, Ganser A, Dohner H, Dohner K. Prognostic impact of WT1 mutations in cytogenetically normal acute myeloid leukemia: a study of the German-Austrian AML Study Group. Blood. 2009;19:4505–4511. doi: 10.1182/blood-2008-10-183392. [DOI] [PubMed] [Google Scholar]
- 44.Wang D, Horton JR, Zheng Y, Blumenthal RM, Zhang X, Cheng X. Role for first zinc finger of WT1 in DNA sequence specificity: Denys-Drash syndrome-associated WT1 mutant in ZF1 enhances affinity for a subset of WT1 binding sites. Nucleic Acids Res. 2018;8:3864–3877. doi: 10.1093/nar/gkx1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Aref S, El Sharawy S, Sabry M, Azmy E, Raouf DA. Prognostic relevance of Wilms tumor 1 (WT1) gene Exon 7 mutations in-patient with cytogenetically normal acute myeloid leukemia. Indian J Hematol Blood Transfus. 2014;4:226–230. doi: 10.1007/s12288-013-0288-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Miyagi T, Ahuja H, Kubota T, Kubonishi I, Koeffler HP, Miyoshi I. Expression of the candidate Wilms tumor gene, WT1, in human leukemia cells. Leukemia. 1993;7:970–977. [PubMed] [Google Scholar]
- 47.Sinha S, Thomas D, Yu L, Gentles AJ, Jung N, Corces-Zimmerman MR, Chan SM, Reinisch A, Feinberg AP, Dill DL, Majeti R. Mutant WT1 is associated with DNA hypermethylation of PRC2 targets in AML and responds to EZH2 inhibition. Blood. 2015;2:316–26. doi: 10.1182/blood-2014-03-566018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wang Y, Xiao M, Chen X, Chen L, Xu Y, Lv L, Wang P, Yang H, Ma S, Lin H, Jiao B, Ren R, Ye D, Guan KL, Xiong Y. WT1 recruits TET2 to regulate its target gene expression and suppress leukemia cell proliferation. Mol Cell. 2015;4:662–673. doi: 10.1016/j.molcel.2014.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Rampal R, Alkalin A, Madzo J, Vasanthakumar A, Pronier E, Patel J, Li Y, Ahn J, Abdel-Wahab O, Shih A, Lu C, Ward PS, Tsai JJ, Hricik T, Tosello V, Tallman JE, Zhao X, Daniels D, Dai Q, Ciminio L, Aifantis I, He C, Fuks F, Tallman MS, Ferrando A, Nimer S, Paietta E, Thompson CB, Licht JD, Mason CE, Godley LA, Melnick A, Figueroa ME, Levine RL. DNA hydroxymethylation profiling reveals that WT1 mutations result in loss of TET2 function in acute myeloid leukemia. Cell Rep. 2014;5:1841–1855. doi: 10.1016/j.celrep.2014.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Lu KH, Li W, Liu XH, Sun M, Zhang ML, Wu WQ, Xie WP, Hou YY. Long non-coding RNA MEG3 inhibits NSCLC cells proliferation and induces apoptosis by affecting p53 expression. BMC Cancer. 2013;13:461. doi: 10.1186/1471-2407-13-461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Liu H, Wang X, Zhang H, Wang J, Chen Y, Ma T, Shi J, Kang Y, Xi J, Wang M, Zhang M. Dynamic changes in the level of WT1 as an MRD marker to predict the therapeutic outcome of patients with AML with and without allogeneic stem cell transplantation. Mol Med Rep. 2019;3:2426–2432. doi: 10.3892/mmr.2019.10440. [DOI] [PubMed] [Google Scholar]
- 52.Rautenberg C, Germing U, Pechtel S, Lamers M, Fischermanns C, Jager P, Geyh S, Haas R, Kobbe G, Schroeder T. Prognostic impact of peripheral blood WT1-mRNA expression in patients with MDS. Blood Cancer J. 2019;11:86. doi: 10.1038/s41408-019-0248-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Nomdedeu JF, Hoyos M, Carricondo M, Bussaglia E, Estivill C, Esteve J, Tormo M, Duarte R, Salamero O, de Llano MP, Garcia A, Bargay J, Heras I, Marti-Tutusaus JM, Llorente A, Ribera JM, Gallardo D, Aventin A, Brunet S, Sierra J CETLAM Group. Bone marrow WT1 levels at diagnosis, post-induction and post-intensification in adult de novo AML. Leukemia. 2013;11:2157–2164. doi: 10.1038/leu.2013.111. [DOI] [PubMed] [Google Scholar]
- 54.Zhao XS, Jin S, Zhu HH, Xu LP, Liu DH, Chen H, Liu KY, Huang XJ. Wilms’ tumor gene 1 expression: an independent acute leukemia prognostic indicator following allogeneic hematopoietic SCT. Bone Marrow Transplant. 2012;4:499–507. doi: 10.1038/bmt.2011.121. [DOI] [PubMed] [Google Scholar]
- 55.Cilloni D, Renneville A, Hermitte F, Hills RK, Daly S, Jovanovic JV, Gottardi E, Fava M, Schnittger S, Weiss T, Izzo B, Nomdedeu J, van der Heijden A, van der Reijden BA, Jansen JH, van der Velden VH, Ommen H, Preudhomme C, Saglio G, Grimwade D. Real-time quantitative polymerase chain reaction detection of minimal residual disease by standardized WT1 assay to enhance risk stratifi cation in acute myeloid leukemia: a European Leukemia Net study. J. Clin. Oncol. 2009;31:5195–5201. doi: 10.1200/JCO.2009.22.4865. [DOI] [PubMed] [Google Scholar]
- 56.Galimberti S, Ghio F, Guerrini F, Ciabatti E, Grassi S, Ferreri MI, Petrini M. WT1 expression levels at diagnosis could predict long-term time-to-progression in adult patients affected by acute myeloid leukaemia and myelodysplastic syndromes. Br J Haematol. 2010;3:451–454. doi: 10.1111/j.1365-2141.2009.08063.x. [DOI] [PubMed] [Google Scholar]
- 57.Maslak PG, Dao T, Bernal Y, Chanel SM, Zhang R, Frattini M, Rosenblat T, Jurcic JG, Brentjens RJ, Arcila ME, Rampal R, Park JH, Douer D, Katz L, Sarlis N, Tallman MS, Scheinberg DA. Phase 2 trial of a multivalent WT1 peptide vaccine (galinpepimut-S) in acute myeloid leukemia. Blood Adv. 2018;3:224–234. doi: 10.1182/bloodadvances.2017014175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Nakata J, Nakae Y, Kawakami M, Morimoto S, Motooka D, Hosen N, Fujiki F, Nakajima H, Hasegawa K, Nishida S, Tsuboi A, Oji Y, Oka Y, Kumanogoh A, Sugiyama H. Wilms tumour 1 peptide vaccine as a cure-oriented post-chemothe rapy strategy for patients with acute myeloid leukaemia at high risk of relapse. Br J Haematol. 2018;2:287–290. doi: 10.1111/bjh.14768. [DOI] [PubMed] [Google Scholar]
- 59.Liu H, Zha Y, Choudhury N, Malnassy G, Fulton N, Green M, Park JH, Nakamura Y, Larson RA, Salazar AM, Odenike O, Gajewski TF, Stock W. WT1 peptide vaccine in Montanide in contrast to poly ICLC, is able to induce WT1-specific immune response with TCR clonal enrichment in myeloid leukemia. Exp Hematol Oncol. 2018;7:1. doi: 10.1186/s40164-018-0093-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Anguille S, Van de Velde AL, Smits EL, Van Tendeloo VF, Juliusson G, Cools N, Nijs G, Stein B, Lion E, Van Driessche A, Vandenbosch I, Verlinden A, Gadisseur AP, Schroyens WA, Muylle L, Vermeulen K, Maes MB, Deiteren K, Malfait R, Gostick E, Lammens M, Couttenye MM, Jorens P, Goossens H, Price DA, Ladell K, Oka Y, Fujiki F, Oji Y, Sugiyama H, Berneman ZN. Dendritic cell vaccination as post remission treatment to prevent or delay relapse in acute myeloid leukemia. Blood. 2017;15:1713–1721. doi: 10.1182/blood-2017-04-780155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kobayashi Y, Sakura T, Miyawaki S, Toga K, Sogo S, Heike Y. A new peptide vaccine OCV-501: in vitro pharmacology and phase 1 study in patients with acute myeloid leukemia. Cancer Immunol Immunother. 2017;7:851–863. doi: 10.1007/s00262-017-1981-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Brayer J, Lancet JE, Powers J, List A, Balducci L, Komrokji R, Pinilla-Ibarz J. WT1 vaccination in AML and MDS: a pilot trial with synthetic analog peptides. Am J Hematol. 2015;7:602–607. doi: 10.1002/ajh.24014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Rezvani K, Yong AS, Mielke S, Jafarpour B, Savani BN, Le RQ, Eniafe R, Musse L, Boss C, Kurlander R, Barrett AJ. Repeated PR1 and WT1 peptide vaccination in Montanide-adjuvant fails to induce sustained high-avidity, epitope-specific CD8+ T cells in myeloid malignancies. Haematologica. 2011;3:432–440. doi: 10.3324/haematol.2010.031674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Maslak PG, Dao T, Krug LM, Chanel S, Korontsvit T, Zakhaleva V, Zhang R, Wolchok JD, Yuan J, Pinilla-Ibarz J, Berman E, Weiss M, Jurcic J, Frattini MG, Scheinberg DA. Vaccination with synthetic analog peptides derived from WT1 oncoprotein induces T-cell responses in patients with complete remission from acute myeloid leukemia. Blood. 2010;2:171–179. doi: 10.1182/blood-2009-10-250993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Rezvani K, Yong AS, Mielke S, Savani BN, Musse L, Superata J, Jafarpour B, Boss C, Barrett AJ. Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies. Blood. 2008;1:236–242. doi: 10.1182/blood-2007-08-108241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Mailander V, Scheibenbogen C, Thiel E, Letsch A, Blau IW, Keilholz U. Complete remission in a patient with recurrent acute myeloid leukemia induced by vaccination with WT1 peptide in the absence of hematological or renal toxicity. Leukemia. 2004;1:165–166. doi: 10.1038/sj.leu.2403186. [DOI] [PubMed] [Google Scholar]
- 67.Ravandi F, Walter RB, Freeman SD. Evaluating measurable residual disease in acute myeloid leukemia. Blood Adv. 2018;11:1356–1366. doi: 10.1182/bloodadvances.2018016378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Rossi G, Minervini MM, Carella AM, Melillo L, Cascavilla N. Wilms’ Tumor Gene (WT1) Expression and Minimal Residual Disease in Acute Myeloid Leukemia. In: van den Heuvel-Eibrink MM, editor. Wilms Tumor. Brisbane: Exon Publications; 2016. pp. 273–285. [PubMed] [Google Scholar]
- 69.Bergmann L, Maurer U, Weidmann E. Wilms tumor gene expression in acute myeloid leukemias. Leuk Lymphoma. 1997;25:435–443. doi: 10.3109/10428199709039030. [DOI] [PubMed] [Google Scholar]
- 70.Ward C, Cauchy P, Garcia P, Frampton J, Esteban MA, Volpe G. High WBP5 expression correlates with elevation of HOX genes levels and is associated with inferior survival in patients with acute myeloid leukaemia. Sci Rep. 2020;1:3505. doi: 10.1038/s41598-020-60480-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Roboz GJ, Guzman M. Acute myeloid leukemia stem cells: seek and destroy. Expert Rev Hematol. 2009;6:663–672. doi: 10.1586/ehm.09.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;7:730–737. doi: 10.1038/nm0797-730. [DOI] [PubMed] [Google Scholar]
- 73.Hope KJ, Jin L, Dick JE. Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in self-renewal capacity. Nat Immunol. 2004;7:738–743. doi: 10.1038/ni1080. [DOI] [PubMed] [Google Scholar]
- 74.LovvikJuul-Dam K, Guldborg Nyvold C, Valerhaugen H, Zeller B, Lausen B, Hasle H, Beier Ommen H. Measurable residual disease monitoring using Wilms tumor gene 1 expression in childhood acute myeloid leukemia based on child-specific reference values. Pediatr Blood Cancer. 2019;6:e27671. doi: 10.1002/pbc.27671. [DOI] [PubMed] [Google Scholar]
- 75.Ho PA, Zeng R, Alonzo TA, Gerbing RB, Miller KL, Pollard JA, Stirewalt DL, Heerema NA, Raimondi SC, Hirsch B, Franklin JL, Lange B, Meshinchi S. Prevalence and prognostic implications of WT1 mutations in pediatric acute myeloid leukemia (AML): a report from the Children’s Oncology Group. Blood. 2010;5:702–10. doi: 10.1182/blood-2010-02-268953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Brieger J, Weidmann E, Fenchel K, Mitrou PS, Hoelzer D, Bergmann L. The expression of the Wilms’ tumor gene in acute myelocytic leukemias as a possible marker for leukemic blast cells. Leukemia. 1994;12:2138–2143. [PubMed] [Google Scholar]
- 77.Karakas T, Miething CC, Maurer U, Weidmann E, Ackermann H, Hoelzer D, Bergmann L. The coexpression of the apoptosis-related genes bcl-2 and wt1 in predicting survival in adult acute myeloid leukemia. Leukemia. 2002;5:846–854. doi: 10.1038/sj.leu.2402434. [DOI] [PubMed] [Google Scholar]
- 78.Shah MV, Jorgensen JL, Saliba RM, Wang SA, Alousi AM, Andersson BS, Bashir Q, Ciurea SO, Kebriaei P, Marin D, Patel KP, Popat UR, Rezvani K, Rondon G, Shpall EJ, Champlin RE, Oran B. Early post-transplant minimal residual disease assessment improves risk stratification in acute myeloid leukemia. Biol Blood Marrow Transplant. 2018;7:1514–1520. doi: 10.1016/j.bbmt.2018.02.003. [DOI] [PubMed] [Google Scholar]
- 79.Medinger M, Lengerke C, Passweg J. Novel therapeutic options in acute myeloid leukemia. Leuk Res Rep. 2016;6:39–49. doi: 10.1016/j.lrr.2016.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Qin YZ, Wang Y, Zhu HH, Gale RP, Zhang MJ, Jiang Q, Jiang H, Xu LP, Chen H, Zhang XH, Liu YR, Lai YY, Jiang B, Liu KY, Huang XJ. Low WT1 transcript levels at diagnosis predicted poor outcomes of acute myeloid leukemia patients with t(8;21) who received chemotherapy or allogeneic hematopoietic stem cell transplantation. Chin J Cancer. 2016;35:46. doi: 10.1186/s40880-016-0110-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Voso MT, Ottone T, Lavorgna S, Venditti A, Maurillo L, Lo-Coco F, Buccisano F. MRD in AML: the role of new techniques. Front Oncol. 2019;9:655. doi: 10.3389/fonc.2019.00655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ogawa H, Tamaki H, Ikegame K, Soma T, Kawakami M, Tsuboi A, Kim EH, Hosen N, Murakami M, Fujioka T, Masuda T, Taniguchi Y, Nishida S, Oji Y, Oka Y, Sugiyama H. The usefulness of monitoring WT1 gene transcripts for the prediction and management of relapse following allogeneic stem cell transplan tation in acute type leukemia. Blood. 2003;5:1698–1704. doi: 10.1182/blood-2002-06-1831. [DOI] [PubMed] [Google Scholar]
- 83.Weisser M, Kern W, Rauhut S, Schoch C, Hiddemann W, Haferlach T, Schnittger S. Prognostic impact of RT-PCR-based quantification of WT1 gene expression during MRD monitoring of acute myeloid leukemia. Leukemia. 2005;8:1416–1423. doi: 10.1038/sj.leu.2403809. [DOI] [PubMed] [Google Scholar]
- 84.Ido K, Nakamae M, Koh H, Okamura H, Nanno S, Nishimoto M, Takeoka Y, Hirose A, Nakashima Y, Hashimoto Y, Nakane T, Hino M, Nakamae H. The proportional relationship between pretransplant WT1 mRNA levels and risk of mortality after allogeneic hematopoietic cell transplantation in acute myeloid leukemia not in remission. Transplantation. 2019;10:2201–2210. doi: 10.1097/TP.0000000000002662. [DOI] [PubMed] [Google Scholar]
- 85.Xu J, Zhang Y, Hu J, Ren Y, Wang H. Clinical features and prognosis of normal karyotype acute myeloid leukemia pediatric patients with WT1 mutations: an analysis based on TCGA database. Hematology. 2020;1:79–84. doi: 10.1080/16078454.2020.1720102. [DOI] [PubMed] [Google Scholar]
- 86.Coosemans A. Wilms’ Tumour gene 1 (WT1) as an immunotherapeutic target. Facts Views Vis Obgyn. 2011;2:89–99. [PMC free article] [PubMed] [Google Scholar]
- 87.Oka Y, Tsuboi A, Nakata J, Nishida S, Hosen N, Kumanogoh A, Oji Y, Sugiyama H. Wilms’ Tumor gene 1 (WT1) peptide vaccine therapy for hematological malignancies: from CTL epitope identification to recent progress in clinical studies including a cure-oriented strategy. Oncol Res Treat. 2017;11:682–690. doi: 10.1159/000481353. [DOI] [PubMed] [Google Scholar]
- 88.Higgins M, Curigliano G, Dieras V, Kuemmel S, Kunz G, Fasching PA, Campone M, Bachelot T, Krivorotko P, Chan S, Ferro A, Schwartzberg L, Gillet M, De Sousa Alves PM, Wascotte V, Lehmann FF, Goss P. Safety and immunoge nicity of neoadjuvant treatment using WT1-immunotherapeutic in combination with standard therapy in patients with WT1-positive Stage II/III breast cancer: a randomized Phase I study. Breast Cancer Res Treat. 2017;3:479–488. doi: 10.1007/s10549-017-4130-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Di Stasi A, Jimenez AM, Minagawa K, Al-Obaidi M, Rezvani K. Review of the results of WT1 peptide vaccination strategies for myelodysplastic syndromes and acute myeloid leukemia from nine different studies. Front Immunol. 2015;6:36. doi: 10.3389/fimmu.2015.00036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Nishida S, Tsuboi A, Tanemura A, Ito T, Nakajima H, Shirakata T, Morimoto S, Fujiki F, Hosen N, Oji Y, Kumanogoh A, Kawase I, Oka Y, Azuma I, Morita S, Sugiyama H. Immune adjuvant therapy using Bacillus Calmette-Guérin cell wall skeleton (BCG-CWS) in advanced malignancies: a phase 1 study of safety and immunogenicity assessments. Medicine (Baltimore) 2019;33:e16771. doi: 10.1097/MD.0000000000016771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Zhao L, Zhang M, Cong H. Advances in the study of HLA-restricted epitope vaccines. Hum Vaccin Immunother. 2013;12:2566–2577. doi: 10.4161/hv.26088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ohno S, Okuyama R, Aruga A, Sugiyama H, Yamamoto M. Phase I trial of Wilms’ Tumor 1 (WT1) peptide vaccine with GM-CSF or CpG in patients with solid malignancy. Anticancer Res. 2012;6:2263–2269. [PubMed] [Google Scholar]