Abstract
Circulating free nucleic acids; cell free DNA and circulating micro-RNA, are found in the plasma of patients with hematologic and solid malignancies at levels higher than that of healthy individuals. In patients with hematologic malignancy cell free DNA reflects the underlying tumor mutational profile, whilst micro-RNAs reflect genetic interference mechanisms within a tumor and potentially the surrounding microenvironment and immune effector cells. These circulating nucleic acids offer a potentially simple, non-invasive, repeatable analysis that can aid in diagnosis, prognosis and therapeutic decisions in cancer treatment.
Keywords: Circulating nucleic acids, DNA, miRNA, hematologic malignancy, biopsy
Introduction
The presence of circulating cell free nucleic acids (CFNA), more specifically circulating free DNA (cfDNA), was first recognized in 1948 1 but it was not until 1977 that it was identified that patients with malignancy had greater amounts of cfDNA than healthy individuals 2. The potential of this medium was first demonstrated in 1994 following the identification of tumor associated RAS gene mutations in the blood of patients with MDS/AML 3 thus confirming the capacity to identify cancer specific mutations in the peripheral blood of cancer patients. Another key discovery came in 2008 when it was shown for the first time that tumor associated microRNAs (miRNA) were detectable in the peripheral blood of lymphoma patients 4. These discoveries have led to an increasing number of studies investigating the utility of CFNA for the characterization, monitoring and therapeutic targeting of both hematologic and solid malignancies 5.
Current methods employed for the evaluation of cancer genomes requires tissue biopsy; either bone marrow (BM) biopsy or biopsy of affected nodal/soft tissue. These procedures are invasive, not without complications and are limited by being representative of only a single site of the disease under evaluation. The latter is of increasing importance with the recognition that cancers may exhibit spatial genomic heterogeneity, with genetically different cancer sub-clones being located at different sites of disease within an affected individual 6. Thus 'single-site' tissue biopsies may not truly reflect the entire mutational profile of an individual's disease and from a logistical perspective may not be suited to repeat biopsy over short periods of time. With this in mind, the potential for 'liquid' biopsies that are non-invasive may not only provide better representation of cancer genetic makeup but provides a strategy that is ideally suited to repeated sampling.
Thus, cfDNA offers the potential of a truly representative cancer genome 'biopsy' that may aid in the diagnosis, prognostication and treatment of cancer. Similarly, the study of circulating miRNA may provide insights into not only diagnosis and prognosis but also previously unrecognized inter-cellular communications aiding in our understanding of cancer biology. This review focuses on the currently available literature focusing on CFNA in hematologic malignancies.
Circulating free DNA (cfDNA)
The first identified cancer-related mutations in the peripheral blood were mutant RAS genes 3, 7, identified in 1994, with their discovery prompting further investigation in the field with numerous publications by the turn of the century 8. While cfDNA is universally found in the plasma of healthy people as well as those with benign diseases 9, it has been observed that patients with malignant disease (both solid tumors and hematologic) have higher levels of cfDNA in their plasma 2. The source of cfDNA is predominantly thought to be due to cells undergoing either spontaneous or chemotherapy induced apoptosis 10, 11. As this process takes place nuclear material is packaged into apoptotic bodies and engulfed by macrophages, however, proportional amounts of cfDNA are also released into the circulation 12. Additionally it has been postulated the presence of cfDNA may be due to active cellular release 13, 14.
As is the case with solid tumors cfDNA may also be more representative of the entire tumor genome in hematological cancers than the information derived from single BM or nodal biopsies, as emerging evidence supports the notion that a range of hematologic malignancies including multiple myeloma (MM) 15, chronic lymphocytic leukaemia (CLL) 16 and non-Hodgkin’s lymphomas (NHL) 17, are likely to harbour significant sub-clonal and spatial genetic heterogeneity. Thus, as novel platforms and techniques in the field continue to evolve 18, 19 the potential of the medium rises in parallel (see Table 1). Moreover, the interrogation of cfDNA can provide information not only in relation to point mutations but also chromosomal translocations, epigenetic modifications, including hypermethylation, and loss of heterozygosity (LOH).
Table1.
cfNA | Target | Technique | Comment |
DNA | Mutation specific (eg. KRAS/NRAS) | PCR | Tumor specific and sensitive; potential use in longitudinal monitoring; may not be practical for broad screening |
Loss of heterozygosity (LOH) | PCR | Issues of reproducibility; circulating measurements inconsistent with primary tumor findings | |
Epigenetic (eg. methylation) | Bisulfite sequencing,Methylation specific PCR | Specific and sensitive tool; requires knowledge of candidate genes for specific testing; complex process | |
Viral DNA | PCR | Potential in malignancies associated with specific viruses (DLBCL, Burkitts); benign viral infection may complicate results | |
Nucleosomes | ELISA | Useful for tumor with high turnover/rates of apoptosis; may also be elevated in non-malignant conditions | |
MicroRNA | - | Microarray | Broad application for miRNA identification; cannot quantify miRNA; issues of sensitivity and reproducibility; inter-array variability and inconsistency |
- | RT-PCR | Useful for monitoring single miRNA; issues with standardization | |
- | NGS | Potential for novel miRNA discovery; costly; highly specialized techniques |
Acute myeloid leukaemia (AML)
The first study evaluating cfDNA mutational profiles in hematologic malignancies was in patients with AML and MDS 3. In this seminal publication, 3 patients had paired cfDNA and BM samples, whereas 7 had paired cfDNA and DNA derived from peripheral blood (PB) blast cells. Five cases consistently demonstrated detectable mutations in cfDNA that were not necessarily found in the paired PB or BM DNA. The authors concluded that plasma could prove to be an easily accessible and useful material for the detection and monitoring of myeloid disorders. Importantly, their findings were recapitulated a decade later when it was shown that the peripheral blood of patients with AML/MDS is enriched for tumor DNA 20.
Circulating nucleosomal fragments represent another source of cfDNA 21 and it has been shown these are increased in the peripheral blood of patients newly diagnosed with AML 22. In a small study of 25 AML patients undergoing induction chemotherapy, irrespective of the remission status eventually achieved, all patients demonstrated an eventual drop in their circulating nucleosomal DNA fragments. But, in those patients who went on to achieve a CR, an initial rise on days 2 to 4 following the commencement of induction chemotherapy was seen, possibly reflecting increased apoptosis in response to treatment. Cytogenetics and specific molecular profiles (NPM1, c-Kit, FLT3, CEBPA) in AML are used to define prognosis and guide treatment decisions following induction 23. It has been shown in a recently published study that NPM mutations can be detected and quantified in the PB 24 at a rate commensurate with the rate of detection in historical BM samples 25. However, this study did not report parallel assessment of NPM mutations in paired cfDNA with BM samples and consequently conclusions about the sensitivity of cfDNA detection could not be drawn. While these studies provide proof of concept of the potential of interrogating cfDNA in AML, they are preliminary and many questions surrounding the ultimate utility of cfDNA analysis remain.
Myelodysplastic syndromes (MDS)
Epigenetic modification is a hallmark of the pathogenesis of MDS, particularly methylation 26. Therefore, the potential to study cfDNA methylation utilizing highly sensitive and novel technologies such as DREAMing (Discrimination of Rare EpiAlleles by Melt) presents an exciting opportunity. In a proof of concept study of 10 MDS and myeloproliferative neoplasm (MPN) patients specifically attempting to address the issue of intra-tumoral epigenetic heterogeneity, analysis of CpG methylation at BRCA1 in the PB was undertaken with bisulfite conversion followed by methylation specific PCR 27. The degree of epiallelic heterogeneity was then derived and confirmed by pyrosequencing. In a second study, methylation analysis with bisulfite pyrosequencing at specific CpG sites of the LINE1 promoter was serially analyzed in patients treated with azacytidine 28. Following treatment, an initial decline in DNA methylation was observed followed by a progressive rise until the next cycle of therapy. Similarly, TET2 mutations were measured in paired cfDNA and BM samples and showed concordance. These data, although preliminary, set the scene for further studies of cfDNA methylation.
Lymphoid neoplasms
Patients with NHL have higher levels of total cfDNA than healthy controls 29, 30. However, whether cfDNA levels at diagnosis correlate with baseline disease characteristics is still unclear. To evaluate lymphoma-specific cfDNA, several groups have analyzed circulating clonal IgH gene rearrangements utilizing PCR 31, high-throughput sequencing (HTS) 32, 33 or next-generation sequencing (NGS) 34, 35. Irrespective of the technique used the majority of patients had measurable rearrangements in the plasma and the detection of plasma cfDNA IgH gene rearrangements was shown to have greater sensitivity than HTS of circulating tumor cells 33. Importantly, higher disease burden correlated with higher levels of IgH gene rearrangements 32- 34 and clearance of IgH gene rearrangements was seen in patients who achieved a CR 32, 34. At progression or relapse detectable clonal IgH genes re-appeared within cfDNA in parallel with radiological relapse. More specifically, in the largest study to date, Roschewski 34 found 3 patterns of IgH gene rearrangements in the context of progressive disease-1-failure of clearance, 2-transient clearance followed by rapid reappearance or 3-clearance followed by clinical progression, then reappearance. Whilst the finding was not statistically significant, those who failed to clear cfDNA derived clonal IgH genes experienced the shortest survival, an observation that if confirmed by others may have significant utility as a MRD strategy to inform risk-adapted changes in therapy. The experience with alternative lymphoma-specific cfDNA targets is limited with preliminary published data describing the use of NGS for single nucleotide variants in CD79A/B, EZH2, CARD11 or MYD88 36 (present in 11 of 12 patients), p61 tumor suppressor gene methylation 37 (present in 73%) and TP53 mutations 38 (present in 30%).
Similarly, in pediatric patients with ALL two groups have shown the presence of higher levels of cfDNA at diagnosis compared with healthy controls, and the feasibility of cfDNA as a marker for MRD 39, 40. Whilst in multivariate analysis cfDNA PCR MRD demonstrated a prognostic significance for EFS/RFS, there was no correlation with bone marrow based FACS MRD, and in several cases FACS MRD was positive whilst cfDNA PCR MRD was negative 40. Thus whilst showing promise as an adjunct in disease assessment, lower sensitivity than currently employed methods may pose an issue.
EBV cfDNA has been detected in patients with various subtypes of NHL and NK/T-cell lymphomas 41- 44. Findings are consistent across all groups, with EBV associated lymphoma patients almost universally having detectable EBV cfDNA whereas paired healthy donor samples do not 43. Moreover, in almost all cases, where patients achieved a CR, EBV cDNA became undetectable but patients who achieved a radiologic CR but failed to clear EBV DNA, subsequently relapsed and died 43. Not surprisingly, high EBV cfDNA at presentation was predictive of poorer outcomes, with patients experiencing shorter overall survival (OS), even in those with early stage disease 43. For patients who achieved only a partial remission (PR) or were refractory to therapy, persistent or rising EBV cfDNA levels were seen 41- 43. These studies show clear potential for cfDNA biomarkers in the diagnosis and monitoring of patients with NHL. Further evaluation is warranted to see if increasing intensity of therapy or stem cell transplant can abrogate the negative impact of persistence of cfDNA.
In proof of concept studies in CLL Sonnenberg et al. 19, 45 evaluated the detection of clonal IgH gene rearrangements in cfDNA using a novel method of DNA isolation-dielectrophoretic isolation (DEP)-that allows isolation of DNA from unprocessed blood for subsequent PCR. This was compared with alternative conventional methods of DNA isolation from paired leukemic cell samples. DEP was found to be comparable for both DNA isolation 19 and clonal IgH gene detection. This technique may therefore provide a simpler, rapid and more cost effective approach of isolating and analyzing cfDNA in CLL.
Hodgkin's lymphoma is associated with EBV in up to 50% of cases 46. Several groups have shown that EBV cfDNA can be detected circulating in the plasma of HL patients 47- 50. However, to date, due to inadequate sensitivity it cannot be used as a surrogate marker for EBER-ISH on nodal tissue 47. This fact notwithstanding, higher levels are associated with more advanced disease and patients that are EBV+ prior to therapy experience inferior failure free survival (FFS) 49 and progression free survival (PFS) 47. Furthermore, response to therapy is associated with undetectable viral loads in the plasma, whilst a single patient who remained positive for EBV viral cfDNA relapsed five months later 48. These data raise the prospect of EBV cfDNA as both a marker of prognosis and for monitoring response to therapy in HL.
As in NHL clonal rearrangements of the IgH gene are present in MM and can be detected utilizing allele specific oligonucleotide (ASO) PCR with probes designed for individual patients. A study describing the use of ASO PCR to analyze IgH VDJ rearrangements in matched bone marrow mononuclear cells (BMMCs), PB mononuclear cells (PBMCs) and cfDNA from MM patients has been reported 51. A statistically significant correlation was observed between the BMMC and PBMC compartments with both levels declining in response to therapy. Using the same ASO primers cfDNA was amplified, confirming the source of the DNA as the malignant MM cells in the BM. However, the circulating DNA level failed to drop following treatment, suggesting the persistence of disease at perhaps spatially disparate sites not demonstrated in single bone marrow biopsy samples.
miRNA
MicroRNAs (miRNA) are small, up to 25 nucleotides long, single stranded non-coding RNAs that bind to specific mRNA. In doing so, miRNAs regulate gene expression by either promoting mRNA hydrolysis and degradation or inhibiting its translation 52. MiRNAs are found in virtually all fluid compartments of the body 53, 54 including blood where they circulate bound to proteins 55, high-density lipoproteins 56, within exosomes or apoptotic bodies 57- 60. This binding or encapsulation renders them resistant to RNases and thus highly stable 61.
In addition to physiologic circulating miRNA, increased levels of circulating miRNA have been observed in malignancy, immune and inflammatory diseases 62. In malignancy the origin of the deregulated levels of miRNA is questioned, whether they originate from tumor cells and/or non-malignant cells including the supporting micro-environment is an area of ongoing research 63. Additionally, whether these are passively released into the circulation during apoptosis or specifically secreted, within exosomes is not clear. If malignant tumor cells actively secrete miRNA, it may represent a potential intercellular communication network whereby tumors could influence the genetic machinery of target cells, potentially influencing and enhancing supporting stroma or possibly inhibiting immune responses. Indeed if this were the case, then it is easy to imagine how disease-related miRNA signatures might correlate with prognosis.
There are currently three methods typically employed to measure circulating miRNA: (i) micro-arrays (eg. Taq Low Density array, Agilent Microarray), (ii) RT-PCR, and (iii) NGS 64. Typically, microarrays are employed first to screen and identify miRNAs of interest, with subsequent confirmation and validation using RT-PCR, whereas NGS assays are more time consuming and expensive and are not routinely used ( Table 1). Unfortunately, none of these methodologies are as yet validated or standardized, complicating generalizability and inter-study comparisons. Many groups report using 'internal' controls by comparing the levels of miRNA of interest to other species of miRNA that appear to remain stable or unaffected over periods of time, however, there is no universal agreement on which miRNA might be best suited to this purpose. The potentially problematic nature of this strategy is exemplified by the frequent use of miR-16 4 for such purposes, as miR-16 has itself been observed to change expression over time in response to therapy. An alternative and perhaps more robust approach is absolute quantitation of target miRNA per unit serum/plasma 65 as is often done with cfDNA studies.
AML
A small number of groups have identified miRNA signatures that demonstrate elevated or decreased levels of specific miRNA species at diagnosis in AML when compared with healthy controls. However, despite using similar techniques and investigational protocols there is surprisingly little, if any, overlap between the different miRNAs identified by different groups of investigators. Two differing miRNA sets-let-7d, miR-150, miR-339, and miR-342 66, and 199b-5p, miR-301b, miR-326, miR-361-5p, miR-625, miR-655 67 have been shown to be elevated at diagnosis in two independent studies. Additionally, in one of these studies let7b and miR-523 have been shown to be reduced 66. Where agreement was seen, was that in patients achieving a CR the initially elevated miRNA levels normalized to that observed in healthy controls. In other studies miR-92a has been identified by two groups to be significantly reduced in the plasma of patients with newly diagnosed AML and in patients with ALL 68. In both instances miR-92a increased following therapy, however, this partial normalization was not statistically different between those who did, or did not, achieve a CR 69.
Zhi et al. 70 employed a genome-wide approach using Solexa sequencing, to define a set of miRNA that might differentiate AML from healthy controls. Utilizing training and verification sets from newly diagnosed AML ( n=140) and normal volunteers ( n=135), 6 miRNA were found to be differentially expressed at statistically significant levels-miR-10a-5p, miR-93-5p, miR-129- 5p, miR-155-5p, miR-181b-5p, miR-320d and validated with RT-PCR. Using cluster analysis of these miRNA to differentiate AML from healthy controls in the validation set (both n=95), 11% of controls were misclassified as AML and only 4% of AML patients misclassified as 'healthy'. The same investigators also examined the prognostic significance of miRNAs in AML and showed that those patients with lower levels of miR-181b-5b at diagnosis had longer OS compared to those with higher levels. As the morphologic diagnosis of AML is rarely complicated, except in specific circumstances e.g. hypoplastic marrows or borderline blast counts, it may be in these settings that miRNA may have diagnostic utility in AML but these questions remain to be answered, as does the prognostic implications of altered levels of miRNA at the time of diagnosis.
MDS
Several miRNA have been shown to be elevated in the peripheral blood of patients with MDS-miR-17-3p, miR-17-5p, miR-18a, miR-15, miR-21, miR-142-3p 71, or reduced let-7a, miR-16 71, 72 and miR-21 73, when compared with healthy controls. Additionally, elevated levels of miR-181a and miR-222 in the BM but not the PB are associated with post MDS AML 71. High levels of miR-16 correlated with low risk IPSS with the converse also holding true. With respect to correlation with survival, patients with higher levels (closer to healthy controls) of let-7a and miR16 experienced longer PFS and OS 72, whereas lower levels of miR-21 correlated with better responses to hypo-methylating agents and longer PFS, but not OS 73. In all of these studies patient numbers were small and the populations studied were heterogeneous, including a mixture of MDS sub-types such as RCMD, RAEB1 and 2, CMML and the 5q- syndrome. Larger studies enriched for specific MDS sub-types are needed before this kind of data may be extrapolated to clinical practice.
Lymphoid malignancies
In diffuse large B-cell lymphoma (DLBCL) circulating miR-21 has been shown to be elevated compared with healthy controls. However, counter intuitively, those with higher levels of miR-21 had earlier stage disease (stages I and II vs. III and IV) and experienced longer relapse free survival (RFS) but not OS 4. Additionally, higher miR-21 was associated with an activated B-cell subtype 74 (ABC) rather than a germinal center B-cell subtype (GCB), which again would appear contradictory to existing evidence of the adverse prognosis associated with an ABC subtype. Moreover, a possible explanation for the observations, that the study population was enriched for early stage ABC was proven to be not the case. A further panel of four miRNA have been shown to be significantly elevated in DLBCL compared to healthy controls 75, however, no clinical correlations with any of the individual miRNA were identified. In both DLBCL and follicular lymphoma (FL) circulating miR-92a has been shown to be reduced 76. Following treatment these levels trended towards normal, however, did not reach that of healthy individuals. In those achieving a CR, who did not undergo hemopoietic stem cell transplant, those with a lower miR-92a at six months were more likely to relapse than those with higher levels.
Elevated levels of miR-155 have been shown to differentiate between healthy controls and individuals with monoclonal B-cell lymphocytosis (MBL) or CLL 77. Importantly, patients with lower levels of miR-155 treated with FCR (Fludaribine, Cyclophosphamide, Rituximab) were more likely to achieve CR and experience longer OS 77, moreover, even when patients with high levels of miR-155 attained a CR their OS was shorter than those with lower miR-155. Similarly, elevated miR-150 can differentiate between healthy controls and CLL 78 and higher plasma miR-150 levels have been shown to correlate with a higher tumor burden and advanced disease stage, expression of the poor prognostic marker CD38 78, ZAP70 positivity 79 and patients are more likely to require treatment 78. Patients with lower miR-150 expression experienced both longer treatment free survival (TFS) and OS 78. A panel of 14 miRNA has been reported to be able to discriminate between CLL, hairy cell leukemia (HCL) and MM 79, however, this requires confirmation by others.
It is perhaps not surprising that two groups have independently demonstrated a correlation between miR-150 and the diagnosis and subsequent prognosis of CLL given its established role in both normal and malignant hemopoiesis 80. Furthermore, increased circulating miR-150 has also been observed in AML 66 suggesting that it may be a non-specific marker of malignant hemopioesis but this will only be clarified by further studies. It should also be noted that in two of the CLL studies 77, 78 miR-16 was used for the normalization of their RT-PCR experiments. MiR-16 is found on chromosome 13q, deleted in up to 50% of cases of CLL 81, whilst both groups state its expression was consistent in the patients evaluated, it again raises the question as to what may constitute the best method of standardization for the reporting or miRNA studies.
A single study has evaluated circulating miRNA in HL 65. MiRNA of interest were identified via comparative microarray of HL involved lymph nodes (not micro-dissected HRS cells) and non-malignant lymph nodes, with validation by RT-qPCR. Five of the 7 identified nodal miRNAs (miR-494, miR2861, miR-21, miR155 and miR-16) were also significantly elevated in the plasma of HL patients when compared with healthy controls and positively correlated with Hasenclever scores. Additionally, changes in miR-494, miR-1973 and miR-21 correlated with disease response, with the plasma levels of all three miRNA at six months post CR (as per CT/PET) comparable to those seen in healthy controls. Moreover, interim evaluation of miRNA did not show a statistically significant difference between interim PR and post-therapy CR. Again highlighting the issue of normalization for circulating miRNA estimation, in this study it was found that RNA-U6 was elevated in the plasma of HL patients and that miR16 fluctuated with disease response. Both these RNAs are frequently used as controls in RT-PCR quantitation but were not used in this study that perhaps more appropriately utilized absolute quantitation of miRNA per volume unit of plasma.
The largest body of miRNA research in relation to hematological malignancies is in MM where several different miRNA signatures have been identified to help to differentiate the continuum of plasma cell dyscrasias ranging from normal plasma cells through monoclonal gammopathy of undetermined significance (MGUS), MM, extra-medullary MM and to plasma cell leukaemia ( Table 2) 82- 90. Despite most groups employing similar strategies, screening disease groups (MGUS versus MM etc.) with micro-array platforms followed by RT-PCR validation, different miRNA panels have been identified with very little overlap. Of particular concern is the conflicting data regarding miR20a, with two papers showing a decreased level in MM whilst another showed an increase. Additionally, highlighting the caution required in over-interpreting significance of specific miRNA, the study of Besse et al. 82 on initial screening found both miR-130a and miR-34a to be elevated in patients with extra-medullary MM (EM-MM). However, on a second set of patients, marker validation showed the opposite, patients with EM-MM was shown to have reduced levels of miR-34a, thus raising very pertinent questions about the reproducibility of the methodologies employed.
Table2.
Author | MiRNA increased | MiRNA decreased | Comment |
Besse 82 | MiR-130a | Elevated in patients with extra-medullary disease over MM | |
Navarro 83 | - | MiR-16, miR-17, miR-19b, miR-20a, miR-660 | MiRNA levels increased in patients achieving CR but did not reach healthy normal |
Wang 84 | - | Let7a, let7b, let7i, miR-15a, miR-15b, miR-16, miR-20a, miR-21, miR-106b, miR-223, miR-361 | 4 of 11 reduced in MGUS,8 of 11 reduced in SMM, 11 of 11 reduced in MM - suggests progressive 'accumulation' of miRNA dysregulation with progressive disease |
Sevcikova 85 | MiR-142-5p, miR-660, miR-29a | ||
Huang 86 | MiR-148a, miR-181a, miR20a, miR221, miR-99b | MiR20a finding conflicts with other groups | |
Kubiczkova 87 | MiR-34a | Let7d, let7e, miR-130a, miR-744 | Combination of let7e and miR-744 provided best ROC characteristics to differentiate HD and either MGUS or MM |
Jones 88 | MiR-720, miR-1246 | MiR-1308 | Different combinations of the three required to differentiate HD from MGUS or MM |
Hao 89 | MiR-4254 | MiR-19a | Combination could distinguish HD or MM |
Lionetti 90 | 42 upregulated | 41 downregulated | Differentiate MM & PCL |
Limited data supports an association between the expression of specific miRNA and underlying cytogenetic abnormalities, for example, several groups have found that specific miRNA correlate with del13q. In the largest cohort of MM patients studied ( n=288), Rocci et al. 91 found low miR-16 and miR-19b were associated with del13q, whereas others have shown low let7e 87 and low miR-221 86 to be associated with del13q. Whilst one group found low miR-19a 89 expression in the context of the co-existence of 1q amplification and del13q. Only one group found any other cytogenetic correlation, with high miR-99b correlating with the presence of t(4;14) 86. It should be noted these were only correlations without proven pathologic relationship and that patients may have any of these miRNA phenotypes without an associated cytogenetic abnormality.
Various miRNA have shown correlation with survival outcomes in MM. Elevated miR-16, miR-30a and miR-25 are all independent markers for longer OS, with miR-25 retaining significance in a multivariate model 91. High levels of miR-19b and miR-331 were associated with longer PFS after a CR is achieved with autologous stem cell transplant 83. Conversely, high miR-148 and miR-20a 86, and low let7e and miR-744 87 are correlated with shorter RFS and OS. In a similar vein a low level of miR-19a was associated with shorter PFS and OS, however, this adverse prognostic association was overcome with bortezomib-based but not thalidomide-based treatment 89.
Conclusions and future directions
The analysis of CFNA clearly offers a potential for diagnostics, prognostics, management and pathologic insights into various hematologic malignancies. Despite longstanding knowledge of the existence of CFNA 1, 2, it is only in the past two decades that research has focused initially on cfDNA 3 and more recently miRNA 4. The science is nascent but rapidly evolving.
The blood of patients with hematologic malignancy contains higher levels of cfDNA 2 that harbors mutations, chromosomal translocations and epigenetic changes present in the tumor population and as such presents an opportunity for simple, non-invasive, repeatable analysis of these genetic profiles. Furthermore, with increasingly sensitive methods 32- 35 of interrogation this source of genetic material may be more sensitive than single biopsies of primary sites of disease or that obtained from circulating tumor cells 33 and this theoretically may be of particular importance in the case of spatially and genetically heterogeneous malignancies 15 whereby CFNA analysis may provide a more 'holistic' description of a tumor's mutational profile. Clearly larger studies are needed to compare mutational profiles identified in CFNA with those found in tissue biopsies from primary sites of disease and identify any correlation with patient outcomes. Current prognostic algorithms 23 have been established for mutations detected in primary tumors, but to date their significance when detected in CFNA is unclear. Simple but important questions such as the significance of differing mutational burdens in the peripheral and the issue of how the identification of a novel mutation in the blood that may otherwise not have been identified in primary tissues should be addressed remain unanswered.
The prospective role of circulating miRNA is less evolved and less clear than that of cfDNA. The source of circulating miRNA remains contentious, with studies frequently finding no correlation between that which is found in the peripheral blood and primary tumor cells 63, 92. Current micro-array platforms, whilst very sensitive, identify vastly different miRNA signatures with little or no overlap between independent studies of the same diseases (see section regarding miRNA in AML and MM). This raises questions of mere association as opposed to a true signal and a potentially significant role in disease pathogenesis. Further efforts investigating circulating miRNA may best be focused on specific miRNA known to play roles in normal and malignant hemopoiesis 80, 93, as well as those already recurrently identified in more that one disease. For instance miR-150 is elevated in both AML 66 and CLL 78, whilst miR-92a is reduced in both AML 68, 69 and NHL 76.
Many questions have emerged in relation to the possible pathologic effects exerted by dysregulated circulating miRNA. Does the prognostic significance associated with an elevated specific miRNA result from its ability to interfere with tumor suppressor genes, the theoretical possibility that it may act as an inter-cellular messenger potentially inhibiting activity of immune effector cells or is it simply an association? How might reduced levels of specific miRNAs exert a prognostic effect? Is it due to increased cellular uptake influencing tumor and/or the microenvironment or is it representative an associated underlying genetic lesion? To properly understand the significance of dysregulated miRNA, functional studies are required. These would include evaluating mechanisms of cellular release and uptake; 'genetic interference' in tumor cells, the supporting micro-environment and immune effector cells; followed by correlation with clinical outcomes. Albeit complex these would provide insight into the true significance of dysregulated miRNA.
Prospects for clinical application are wide ranging, as novel testing platforms become more sensitive, quicker and more affordable, validation and standardization will be urgently required not only for 'inter-study' comparison but for potential integration into standard pathology practice 64. Whilst controls for cfDNA analyses have largely been established, little or no agreement exists for controls and standardization of miRNA PCR and is urgently needed. The field of CFNA is rapidly evolving, with the prospects of liquid biopsy either complementing or even replacing BM and/or tissue biopsy a real possibility in the future. However, until platforms and practice are standardized, greater concordance in the data achieved, paired with an understanding of the mechanisms behind the observations, hematologists shouldn’t throw out the marrow needle just yet.
Conflict of interest statement
No potential conflicts of interest are disclosed.
References
- Mandel P, Metais P. Les acides nucléiques du plasma sanguin chez l'homme. C R Seances Soc Biol Fil. 1948;142:241–3. [PubMed] [Google Scholar]
- Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977;37:646–50. [PubMed] [Google Scholar]
- Vasioukhin V, Anker P, Maurice P, Lyautey J, Lederrey C, Stroun M. Point mutations of the N-ras gene in the blood plasma DNA of patients with myelodysplastic syndrome or acute myelogenous leukaemia. Br J Haematol. 1994;86:774–9. doi: 10.1111/j.1365-2141.1994.tb04828.x. [DOI] [PubMed] [Google Scholar]
- Lawrie CH, Gal S, Dunlop HM, Pushkaran B, Liggins AP, Pulford K, et al. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br J Haematol. 2008;141:672–5. doi: 10.1111/j.1365-2141.2008.07077.x. [DOI] [PubMed] [Google Scholar]
- Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11:426–37. doi: 10.1038/nrc3066. [DOI] [PubMed] [Google Scholar]
- Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–15. doi: 10.1038/nature10166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sorenson GD, Pribish DM, Valone FH, Memoli VA, Bzik DJ, Yao SL. Soluble normal and mutated DNA sequences from single-copy genes in human blood. Cancer Epidemiol Biomarkers Prev. 1994;3:67–71. [PubMed] [Google Scholar]
- Ziegler A, Zangemeister-Wittke U, Stahel RA. Circulating DNA: a new diagnostic gold mine. Cancer Treat Rev. 2002;28:255–71. doi: 10.1016/s0305-7372(02)00077-4. [DOI] [PubMed] [Google Scholar]
- Swaminathan R, Butt AN. Circulating nucleic acids in plasma and serum: recent developments. Ann N Y Acad Sci. 2006;1075:1–9. doi: 10.1196/annals.1368.001. [DOI] [PubMed] [Google Scholar]
- Fournie GJ, Courtin JP, Laval F, et al. Plasma DNA as a marker of cancerous cell death. Plasma DNA as a marker of cancerous cell death. Investigations in patients suffering from lung cancer and in nude mice bearing human tumours. Cancer Lett. 1995;91:221–7. doi: 10.1016/0304-3835(95)03742-f. [DOI] [PubMed] [Google Scholar]
- Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001;61:1659–65. [PubMed] [Google Scholar]
- Choi JJ, Reich CF, Pisetsky DS. The role of macrophages in the in vitro Generation of extracellular DNA from apoptotic and necrotic cells. Immunology. 2005;115:55–62. doi: 10.1111/j.1365-2567.2005.02130.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stroun M, Lyautey J, Lederrey C, Mulcahy HE, Anker P. Alu repeat sequences are present in increased proportions compared to a unique gene in plasma/serum DNA: evidence for a preferential release from viable cells. Ann N Y Acad Sci. 2001;945:258–64. doi: 10.1111/j.1749-6632.2001.tb03894.x. [DOI] [PubMed] [Google Scholar]
- Peters DL, Pretorius PJ. Origin, translocation and destination of extracellular occurring DNA--a new paradigm in genetic behaviour. Clin Chim Acta. 2011;412:806–11. doi: 10.1016/j.cca.2011.01.026. [DOI] [PubMed] [Google Scholar]
- Lohr JG, Stojanov P, Carter SL, Lawrence MS, Auclair DA, Knoechel B, et al. Widespread genetic heterogeneity in multiple myeloma: implications for targeted therapy. Cancer Cell. 2014;25:91–101. doi: 10.1016/j.ccr.2013.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guieze R, Wu CJ. Genomic and epigenomic heterogeneity in chronic lymphocytic leukemia. Blood. 2015;126:445–53. doi: 10.1182/blood-2015-02-585042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu F, Yoshida N, Suguro M, Kato HA, Arita K, Yamamoto K, et al. Clonal heterogeneity of mantle cell lymphoma revealed by array comparative genomic hybridization. Eur J Haematol. 2013;90:51–8. doi: 10.1111/ejh.12030. [DOI] [PubMed] [Google Scholar]
- Hudecova I. Digital PCR analysis of circulating nucleic acids. Clin Biochem. 2015;48:948–56. doi: 10.1016/j.clinbiochem.2015.03.015. [DOI] [PubMed] [Google Scholar]
- Sonnenberg A, Marciniak JY, Rassenti L, Ghia EM, Skowronski EA, Manouchehri S, et al. Rapid electrokinetic isolation of cancer-related circulating cell-free DNA directly from blood. Clin Chem. 2014;60:500–9. doi: 10.1373/clinchem.2013.214874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rogers A, Joe Y, Manshouri T, Dey A, Jilani I, Giles F, et al. Relative increase in leukemia-specific DNA in peripheral blood plasma from patients with acute myeloid leukemia and myelodysplasia. Blood. 2004;103:2799–801. doi: 10.1182/blood-2003-06-1840. [DOI] [PubMed] [Google Scholar]
- Jiang N, Reich CF, Pisetsky DS. Role of macrophages in the Generation of circulating blood nucleosomes from dead and dying cells. Blood. 2003;102:2243–50. doi: 10.1182/blood-2002-10-3312. [DOI] [PubMed] [Google Scholar]
- Mueller S, Holdenrieder S, Stieber P, et al. Early prediction of therapy response in patients with acute myeloid leukemia by nucleosomal DNA fragments. BMC Cancer 2006; 6: 143.
- Cornelissen JJ, Gratwohl A, Schlenk RF, Sierra J, Bornhäuser M, Juliusson G, et al. The European LeukemiaNet AML working party consensus statement on allogeneic HSCT for patients with AML in remission: an integrated-risk adapted approach. Nat Rev Clin Oncol. 2012;9:579–90. doi: 10.1038/nrclinonc.2012.150. [DOI] [PubMed] [Google Scholar]
- Quan J, Gao YJ, Yang ZL, Chen H, Xian JR, Zhang SS, et al. Quantitative detection of circulating nucleophosmin mutations DNA in the plasma of patients with acute myeloid leukemia. Int J Med Sci. 2015;12:17–22. doi: 10.7150/ijms.10144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Falini B, Mecucci C, Tiacci E, Alcalay M, Rosati R, Pasqualucci L, et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med. 2005;352:254–66. doi: 10.1056/NEJMoa041974. [DOI] [PubMed] [Google Scholar]
- Issa J. The myelodysplastic syndrome as a prototypical epigenetic disease. Blood. 2013;121:3811–7. doi: 10.1182/blood-2013-02-451757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pisanic TR 2nd, Athamanolap P, Poh W, Chen C, Hulbert A, Brock MV, et al. DREAMing: a simple and ultrasensitive method for assessing intratumor epigenetic heterogeneity directly from liquid biopsies. Nucleic Acids Res. 2015;43:e154. doi: 10.1093/nar/gkv795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iriyama C, Tomita A, Hoshino H, Adachi-Shirahata M, Furukawa-Hibi Y, Yamada K, et al. Using peripheral blood circulating DNAs to detect CpG global methylation status and genetic mutations in patients with myelodysplastic syndrome. Biochem Biophys Res Commun. 2012;419:662–9. doi: 10.1016/j.bbrc.2012.02.071. [DOI] [PubMed] [Google Scholar]
- Greystoke A, O'connor J, Linton K, Taylor MB, Cummings J, Ward T, et al. Assessment of circulating biomarkers for potential pharmacodynamic utility in patients with lymphoma. Br J Cancer. 2011;104:719–25. doi: 10.1038/sj.bjc.6606082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hohaus S, Giachelia M, Massini G, Mansueto G, Bozzoli V, Criscuolo M, et al. Cell-free circulating DNA in Hodgkin's and non-Hodgkin's lymphomas. Ann Oncol. 2009;20:1408–13. doi: 10.1093/annonc/mdp006. [DOI] [PubMed] [Google Scholar]
- Frickhofen N, Muller E, Sandherr M, Binder T, Bangerter M, Wiest C, et al. Rearranged Ig heavy chain DNA is detectable in cell-free blood samples of patients with B-cell neoplasia. Blood. 1997;90:4953–60. [PubMed] [Google Scholar]
- Armand P, Oki Y, Neuberg DS, Faham M, Cummings C, Klinger M, et al. Detection of circulating tumour DNA in patients with aggressive B-cell non-Hodgkin lymphoma. Br J Haematol. 2013;163:123–6. doi: 10.1111/bjh.12439. [DOI] [PubMed] [Google Scholar]
- Kurtz DM, Green MR, Bratman SV, Scherer F, Liu CL, Kunder CA, et al. Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing. Blood. 2015;125:3679–87. doi: 10.1182/blood-2015-03-635169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roschewski M, Dunleavy K, Pittaluga SA, Pepin F, Kong K, Shovlin M, et al. Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: a correlative biomarker study. Lancet Oncol. 2015;16:541–9. doi: 10.1016/S1470-2045(15)70106-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He J, Wu J, Jiao Y, Wagner-Johnston N, Ambinder RF, Diaz LA, et al. IgH gene rearrangements as plasma biomarkers in Non- Hodgkin's lymphoma patients. Oncotarget. 2011;2:178–85. doi: 10.18632/oncotarget.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bohers E, Viailly PJ, Dubois SA, Maingonnat C, Mareschal SA, Picquenot JM, et al. Somatic mutations of cell-free circulating DNA detected by next-generation sequencing reflect the genetic changes in both germinal center B-cell-like and activated B-cell-like diffuse large B-cell lymphomas at the time of diagnosis. Haematologica. 2015;100:E280–4. doi: 10.3324/haematol.2015.123612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deligezer U, Yaman F, Erten N, Dalay N. Frequent copresence of methylated DNA and fragmented nucleosomal DNA in plasma of lymphoma patients. Clin Chim Acta. 2003;335:89–94. doi: 10.1016/s0009-8981(03)00279-1. [DOI] [PubMed] [Google Scholar]
- Hosny G, Farahat N, Hainaut P. TP53 mutations in circulating free DNA from Egyptian patients with non-Hodgkin's lymphoma. Cancer Lett. 2009;275:234–9. doi: 10.1016/j.canlet.2008.10.029. [DOI] [PubMed] [Google Scholar]
- Schwarz AK, Stanulla M, Cario G, Flohr T, Sutton R, Möricke A, et al. Quantification of free total plasma DNA and minimal residual disease detection in the plasma of children with acute lymphoblastic leukemia. Ann Hematol. 2009;88:897–905. doi: 10.1007/s00277-009-0698-6. [DOI] [PubMed] [Google Scholar]
- Cheng SH, Lau KM, Li CK, Chan NP, Ip RK, Cheng CK, et al. Minimal residual disease-based risk stratification in Chinese childhood acute lymphoblastic leukemia by flow cytometry and plasma DNA quantitative polymerase chain reaction. PloS One. 2013;8:e69467. doi: 10.1371/journal.pone.0069467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Au WY, Pang A, Choy C, Chim CS, Kwong YL. Quantification of circulating Epstein-Barr virus (EBV) DNA in the diagnosis and monitoring of natural killer cell and EBV-positive lymphomas in immunocompetent patients. Blood. 2004;104:243–9. doi: 10.1182/blood-2003-12-4197. [DOI] [PubMed] [Google Scholar]
- Jones K, Nourse JP, Keane C, Crooks P, Gottlieb D, Ritchie DS, et al. Tumor-specific but not nonspecific cell-free circulating DNA can be used to monitor disease response in lymphoma. Am J Hematol. 2012;87:258–65. doi: 10.1002/ajh.22252. [DOI] [PubMed] [Google Scholar]
- Lei KI, Chan LY, Chan WY, Johnson PJ, Lo YM. Diagnostic and prognostic implications of circulating cell-free Epstein-Barr virus DNA in natural killer/T-cell lymphoma. Clin Cancer Res. 2002;8:29–34. [PubMed] [Google Scholar]
- Lei KI, Chan LY, Chan WY, Johnson PJ, Lo YM. Quantitative analysis of circulating cell-free Epstein-Barr virus (EBV) DNA levels in patients with EBV-associated lymphoid malignancies. Br J Haematol. 2000;111:239–46. doi: 10.1046/j.1365-2141.2000.02344.x. [DOI] [PubMed] [Google Scholar]
- Sonnenberg A, Marciniak JY, Skowronski EA, Manouchehri S, Rassenti L, Ghia EM, et al. Dielectrophoretic isolation and detection of cancer-related circulating cell-free DNA biomarkers from blood and plasma. Electrophoresis. 2014;35:1828–36. doi: 10.1002/elps.201400016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Glaser SL, Lin RJ, Stewart SL, Ambinder RF, Jarrett RF, Brousset P, et al. Epstein-Barr virus-associated Hodgkin's disease: epidemiologic characteristics in international data. Int J Cancer. 1997;70:375–82. doi: 10.1002/(sici)1097-0215(19970207)70:4<375::aid-ijc1>3.0.co;2-t. [DOI] [PubMed] [Google Scholar]
- Hohaus S, Santangelo R, Giachelia M, Vannata B, Massini G, Cuccaro A, et al. The viral load of Epstein-Barr virus (EBV) DNA in peripheral blood predicts for biological and clinical characteristics in Hodgkin lymphoma. Clin Cancer Res. 2011;17:2885–92. doi: 10.1158/1078-0432.CCR-10-3327. [DOI] [PubMed] [Google Scholar]
- Gandhi MK, Lambley E, Burrows J, Dua U, Elliott SA, Prince HM, et al. Plasma Epstein-Barr virus (EBV) DNA is a biomarker for EBV-positive hodgkin's lymphoma. Clin Cancer Res. 2006;12:460–4. doi: 10.1158/1078-0432.CCR-05-2008. [DOI] [PubMed] [Google Scholar]
- Kanakry JA, Li H, Gellert LL, Lemas MV, Hsieh WS, Hong F, et al. Plasma Epstein-Barr virus DNA predicts outcome in advanced hodgkin lymphoma: correlative analysis from a large North American cooperative group trial. Blood. 2013;121:3547–53. doi: 10.1182/blood-2012-09-454694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Musacchio JG, Carvalho Mda G, Morais JC, Silva NH, Scheliga A, Romano S, et al. Detection of free circulating Epstein-Barr virus DNA in plasma of patients with Hodgkin's disease. Sao Paulo Med J. 2006;124:154–7. doi: 10.1590/S1516-31802006000300009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sata H, Shibayama H, Maeda I, Habuchi Y, Nakatani E, Fukushima K, et al. Quantitative polymerase chain reaction analysis with allele-specific oligonucleotide primers for individual IgH VDJ regions to evaluate tumor burden in myeloma patients. Exp Hematol. 2015;43:374–81 e2. doi: 10.1016/j.exphem.2015.01.002. [DOI] [PubMed] [Google Scholar]
- Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–97. doi: 10.1016/s0092-8674(04)00045-5. [DOI] [PubMed] [Google Scholar]
- Weber JA, Baxter DH, Zhang SL, Huang DY, Huang KH, Lee MJ, et al. The MicroRNA spectrum in 12 body fluids. Clin Chem. 2010;56:1733–41. doi: 10.1373/clinchem.2010.147405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang K, Yuan Y, Cho JH, McClarty S, Baxter D, Galas DJ. Comparing the MicroRNA spectrum between serum and plasma. PloS One. 2012;7:e41561. doi: 10.1371/journal.pone.0041561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arroyo JD, Chevillet JR, Kroh EM, et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci U S A. 2011;108:5003–8. doi: 10.1073/pnas.1019055108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol. 2011;13:423–33. doi: 10.1038/ncb2210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mittelbrunn M, Gutiérrez-Vázquez C, Villarroya-Beltri C, González S, Sánchez-Cabo F, González MÁ, et al. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat Commun. 2011;2:282. doi: 10.1038/ncomms1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaiswal R, Luk F, Gong J, Mathys JM, Grau GE, Bebawy M. Microparticle conferred microRNA profiles--implications in the transfer and dominance of cancer traits. Mol Cancer. 2012;11:37. doi: 10.1186/1476-4598-11-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, Liu D, Chen X, Li J, Li L, Bian Z, et al. Secreted monocytic miR-150 enhances targeted endothelial cell migration. Mol Cell. 2010;39:133–44. doi: 10.1016/j.molcel.2010.06.010. [DOI] [PubMed] [Google Scholar]
- Nolte-'t Hoen EN, Buermans HP, Waasdorp M, Stoorvogel W, Wauben MH, t Hoen PA. Deep sequencing of RNA from immune cell-derived vesicles uncovers the selective incorporation of small non-coding RNA biotypes with potential regulatory functions. Nucleic Acids Res. 2012;40:9272–85. doi: 10.1093/nar/gks658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A. 2008;105:10513–8. doi: 10.1073/pnas.0804549105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, et al. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008;18:997–1006. doi: 10.1038/cr.2008.282. [DOI] [PubMed] [Google Scholar]
- Turchinovich A, Weiz L, Burwinkel B. Extracellular miRNAs: the mystery of their origin and function. Trends Biochem Sci. 2012;37:460–5. doi: 10.1016/j.tibs.2012.08.003. [DOI] [PubMed] [Google Scholar]
- Ono S, Lam S, Nagahara M, Hoon DS. Circulating microRNA biomarkers as liquid biopsy for cancer patients: Pros and Cons of current assays. J Clin Med. 2015;4:1890–907. doi: 10.3390/jcm4101890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones K, Nourse JP, Keane C, Bhatnagar A, Gandhi MK. Plasma microRNA are disease response biomarkers in classical Hodgkin lymphoma. Clin Cancer Res. 2014;20:253–64. doi: 10.1158/1078-0432.CCR-13-1024. [DOI] [PubMed] [Google Scholar]
- Fayyad-Kazan H, Bitar N, Najar M, Lewalle P, Fayyad-Kazan M, Badran R, et al. Circulating miR-150 and miR-342 in plasma are novel potential biomarkers for acute myeloid leukemia. J Transl Med. 2013;11:31. doi: 10.1186/1479-5876-11-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koutova L, Sterbova M, Pazourkova E, Pospisilova S, Svobodova I, Horinek A, et al. The impact of standard chemotherapy on miRNA signature in plasma in AML patients. Leuk Res. 2015;39:1389–95. doi: 10.1016/j.leukres.2015.08.009. [DOI] [PubMed] [Google Scholar]
- Tanaka M, Oikawa K, Takanashi M, Kudo M, Ohyashiki J, Ohyashiki K, et al. Down-regulation of miR-92 in human plasma is a novel marker for acute leukemia patients. PloS One. 2009;4:e5532. doi: 10.1371/journal.pone.0005532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- El-Halawani N, Eldafrawi M, Mourad Z, Sorour A, Ghallab O. Diagnostic and prognostic value of plasma level of microRNA-92a in acute myeloid leukemia. Am J Mol Biol. 2014;4:1–10. [Google Scholar]
- Zhi F, Cao XS, Xie XB, Wang B, Dong WM, Gu WY, et al. Identification of circulating MicroRNAs as potential biomarkers for detecting acute myeloid leukemia. PLoS One. 2013;8:e56718. doi: 10.1371/journal.pone.0056718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pons A, Nomdedeu B, Navarro A, Gaya AA, Diaz T, Valera S, et al. Hematopoiesis-related microRNA expression in myelodysplastic syndromes. Leuk Lymphoma. 2009;50:U163–1854. doi: 10.3109/10428190903147645. [DOI] [PubMed] [Google Scholar]
- Zuo Z, Calin GA, De Paula HM, Medeiros LJ, Fernandez MH, Shimizu MA, et al. Circulating microRNAs let-7a and miR-16 predict progression-free survival and overall survival in patients with myelodysplastic syndrome. Blood. 2011;118:413–5. doi: 10.1182/blood-2011-01-330704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim Y, Cheong JW, Kim YK, Eom JI, Jeung HK, Kim SJ, et al. Serum microRNA-21 as a potential biomarker for response to hypomethylating agents in myelodysplastic syndromes. PloS One. 2014;9:e86933. doi: 10.1371/journal.pone.0086933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen W, Wang H, Chen H, Liu S, Lu H, Kong D, et al. Clinical significance and detection of microRNA-21 in serum of patients with diffuse large B-cell lymphoma in Chinese population. Eur J Haematol. 2014;92:407–12. doi: 10.1111/ejh.12263. [DOI] [PubMed] [Google Scholar]
- Fang C, Zhu DX, Dong HJ, Zhou Z, Liu L, Fan L, et al. Serum microRNAs are promising novel biomarkers for diffuse large B cell lymphoma. Ann Hematol. 2012;91:553–9. doi: 10.1007/s00277-011-1350-9. [DOI] [PubMed] [Google Scholar]
- Ohyashiki K, Umezu T, Yoshizawa S, Ito Y, Ohyashiki M, Kawashima H, et al. Clinical impact of down-regulated plasma miR-92a levels in non-Hodgkin's lymphoma. PloS One. 2011;6:e16408. doi: 10.1371/journal.pone.0016408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferrajoli A, Shanafelt TD, Ivan C, Shimizu M, Rabe KG, Nouraee N, et al. Prognostic value of miR-155 in individuals with monoclonal B-cell lymphocytosis and patients with B chronic lymphocytic leukemia. Blood. 2013;122:1891–9. doi: 10.1182/blood-2013-01-478222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stamatopoulos B, Van Damme M, Crompot E, Dessars B, Housni HE, Mineur P, et al. Opposite prognostic significance of cellular and serum circulating MicroRNA-150 in patients with chronic lymphocytic leukemia. Mol Med. 2015;21:123–33. doi: 10.2119/molmed.2014.00214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moussay E, Wang K, Cho JH, et al. MicroRNA as biomarkers and regulators in B-cell chronic lymphocytic leukemia. Proc Natl Acad Sci U S A. 2011;108:6573–8. doi: 10.1073/pnas.1019557108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He Y, Jiang X, Chen J. The role of miR-150 in normal and malignant hematopoiesis. Oncogene. 2014;33:3887–93. doi: 10.1038/onc.2013.346. [DOI] [PubMed] [Google Scholar]
- Dohner H, Stilgenbauer S, Benner A, Leupolt E, Krober A, Bullinger L, et al. Genomic aberrations and survival in chronic lymphocytic leukemia N Engl J Med. 2000; 343: 1910-6.
- Besse L, Sedlarikova L, Kryukov F, Nekvindova J, Radova L, Slaby O, et al. Circulating Serum MicroRNA-130a as a Novel Putative Marker of Extramedullary Myeloma. PloS One 2015; 10: e0137294.
- Navarro A, Diaz T, Tovar N, Pedrosa F, Tejero R, Teresa Cibeira M, et al. A serum microRNA signature associated with complete remission and progression after autologous stem-cell transplantation in patients with multiple myeloma. Oncotarget. 2015;6:1874–83. doi: 10.18632/oncotarget.2761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang W, Corrigan-Cummins M, Barber EA, Saleh LM, Zingone A, Ghafoor A, et al. Aberrant levels of miRNAs in bone marrow microenvironment and peripheral blood of myeloma patients and disease progression. J Mol Diagn. 2015;17:669–78. doi: 10.1016/j.jmoldx.2015.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sevcikova S, Kubiczkova L, Sedlarikova LA, Hajek R. Serum miR-29a as a marker of multiple myeloma. Leuk Lymphoma. 2013;54:189–91. doi: 10.3109/10428194.2012.704030. [DOI] [PubMed] [Google Scholar]
- Huang JJ, Yu J, Li JY, Liu YT, Zhong RQ. Circulating microRNA expression is associated with genetic subtype and survival of multiple myeloma. Med Oncol. 2012;29:2402–8. doi: 10.1007/s12032-012-0210-3. [DOI] [PubMed] [Google Scholar]
- Kubiczkova L, Kryukov F, Slaby O, Dementyeva E, Jarkovsky J, Nekvindova J, et al. Circulating serum microRNAs as novel diagnostic and prognostic biomarkers for multiple myeloma and monoclonal gammopathy of undetermined significance. Haematologica. 2014;99:511–8. doi: 10.3324/haematol.2013.093500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones CI, Zabolotskaya MV, King AJ, Stewart HJ, Horne GA, Chevassut TJ, et al. Identification of circulating microRNAs as diagnostic biomarkers for use in multiple myeloma. Br J Cancer. 2012;107:1987–96. doi: 10.1038/bjc.2012.525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hao M, Zang MR, Wendlandt E, Xu Y, An G, Gong DS, et al. Low serum miR-19a expression as a novel poor prognostic indicator in multiple myeloma. Int J Cancer. 2015;136:1835–44. doi: 10.1002/ijc.29199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lionetti M, Musto P, Di Martino MT, Fabris S, Agnelli L, Todoerti K, et al. Biological and clinical relevance of miRNA expression signatures in primary plasma cell leukemia. Clin Cancer Res. 2013;19:3130–42. doi: 10.1158/1078-0432.CCR-12-2043. [DOI] [PubMed] [Google Scholar]
- Rocci A, Hofmeister CC, Geyer S, Stiff A, Cascione L, Guan J, et al. Circulating miRNA markers show promise as new prognosticators for multiple myeloma. Leukemia. 2014;28:1922–6. doi: 10.1038/leu.2014.155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohyashiki JH, Umezu T, Kobayashi C, Hamamura RS, Tanaka M, Kuroda M, et al. Impact on cell to plasma ratio of miR-92a in patients with acute leukemia: in vivo assessment of cell to plasma ratio of miR-92a. BMC Res Notes. 2010;3:347. doi: 10.1186/1756-0500-3-347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Troppan K, Wenzl K, Deutsch A, Ling H, Neumeister P, Pichler M. MicroRNAs in diffuse large B-cell lymphoma: implications for pathogenesis, diagnosis, prognosis and therapy. Anticancer Res. 2014;34:557–64. [PubMed] [Google Scholar]