ArticlePDF AvailableLiterature Review

GD2-targeted immunotherapy and potential predictive value of circulating microRNAs in neuroblastoma

Authors:

Abstract and Figures

Neuroblastoma (NB) with various clinical presentation is a known childhood malignancy. Despite significant progress in treatment of NB afflicted patients, high risk disease is usually associated with poor outcome, resulting in long-term survival of less that 50%. Known as a disease most commonly originated form the nerve roots, the variants involved in NB imitation and progression remain to be elucidated. The outcome of low to intermediate risk disease is favorable whereas the high risk NB disease with dismal prognosis, positing the necessity of novel approaches for early detection and prognostication of advanced disease. Tailored immunotherapy approaches have shown significant improvement in highrisk NB patients. It has found a link between Gangliosides and progression of NB. The vast majority of neuroblastoma tumors express elevated levels of GD2, opening new insight into using anti-GD2 drugs as potential treatments for NBs. Implication of anti-GD2 monoclonal antibodies for treatment of high risk NBs triggers further investigation to unearth novel biomarkers as prognostic and response biomarker to guide additional multimodal tailored treatment approaches. A growing body of evidence supports the usefulness of miRNAs to evaluate high risk NBs response to anti-GD2 drugs and further prevent drug-related toxicities in refractory or recurrent NBs. miRNAs and circulating proteins in body fluids (plasma, and serum) present as potential biomarkers in early detection of NBs. Here, we summarize various biomarkers involved in diagnosis, prognosis and response to treatment in patients with NB. We further attempted to overview prognostic biomarkers in response to treatment with anti-GD2 drugs.
Content may be subject to copyright.
This article is protected by copyright. All rights reserved 1
REVIEW ARTICLE
GD2-targeted immunotherapy and potential value of circulating microRNAs in
neuroblastoma
Sharareh Gholamin1,2,*, Hamed Miezaei3,*, Seyed-Mostafa Razavi4,*, Seyed Mahdi
Hassanian5,6,*,Leila Saadatpour7, Aria Masoudifar11, Soodabeh ShahidSales8,# Amir Avan9,10,#
(1) Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United
States
(2)Department of Bioengineering at California Institute of Technology, Pasadena, CA, United states.
(3) Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences,
Mashhad, Iran
(4) Department of Neurosurgery, Stanford University, Stanford, CA, United States.
(5) Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences,
Mashhad, Iran.
(6) Microanatomy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
(7) Department of Neurology, University of Florida College of Medicine, Gainesville, FL, United States.
(8) Cancer Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
(9) Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences,
Mashhad, Iran.
(10) Molecular Medicine group, Department of Modern Sciences and Technologies, Mashhad University of
Medical Sciences, Mashhad, Iran.
(11) Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for
Biotechnology, ACECR, Isfahan, Iran
#Corresponding Authors:
Amir Avan, PhD; Metabolic Syndrome Research Center, School of Medicine, Mashhad University
of Medical Sciences, Mashhad, Iran., Tel:+9851138002298, Fax: +985118002298; Email:
avana@mums.ac.ir & amir_avan@yahoo.com
Soudabeh ShahidSales MD; Cancer Research Center, School of Medicine, Mashhad University of
Medical Sciences, Mashhad, Iran. Tel:+9851138002298, Fax: +985118002298; Email:
ShahidSaless@mums.ac.ir
* equally contributed as first author to this study.
Running title: anti-GD2 therapy in Neuroblastoma
Grant: This study was supported by grant awarded (Amir Avan) by the Mashhad University of
Medical Sciences.
Conflict of interest: The authors have no conflict of interest to disclose
This article has been accepted for publication and undergone full peer review but has not been
through the copyediting, typesetting, pagination and proofreading process, which may lead to
differences between this version and the Version of Record. Please cite this article as doi:
[10.1002/jcp.25793]
Received 9 January 2017; Accepted 10 January 2017 Journal of Cellular Physiology
This article is protected by copyright. All rights reserved DOI 10.1002/jcp.25793
This article is protected by copyright. All rights reserved 2
Abstract
Neuroblastoma (NB) with various clinical presentation is a known childhood malignancy. Despite
significant progress in treatment of NB afflicted patients, high risk disease is usually associated
with poor outcome, resulting in long-term survival of less that 50%. Known as a disease most
commonly originated form the nerve roots, the variants involved in NB imitation and progression
remain to be elucidated. The outcome of low to intermediate risk disease is favorable whereas the
high risk NB disease with dismal prognosis, positing the necessity of novel approaches for early
detection and prognostication of advanced disease. Tailored immunotherapy approaches have
shown significant improvement in high-risk NB patients. It has found a link between Gangliosides
and progression of NB. The vast majority of neuroblastoma tumors express elevated levels of GD2,
opening new insight into using anti-GD2 drugs as potential treatments for NBs. Implication of anti-
GD2 monoclonal antibodies for treatment of high risk NBs triggers further investigation to unearth
novel biomarkers as prognostic and response biomarker to guide additional multimodal tailored
treatment approaches. A growing body of evidence supports the usefulness of miRNAs to evaluate
high risk NBs response to anti-GD2 drugs and further prevent drug-related toxicities in refractory or
recurrent NBs. miRNAs and circulating proteins in body fluids (plasma, and serum) present as
potential biomarkers in early detection of NBs. Here, we summarize various biomarkers involved
in diagnosis, prognosis and response to treatment in patients with NB. We further attempted to
overview prognostic biomarkers in response to treatment with anti-GD2 drugs. This article is
protected by copyright. All rights reserved
Key words: Neuroblastoma, miRNAs, anti-GD2 drugs
This article is protected by copyright. All rights reserved 3
Introduction
Neuroblastoma (NB) is usually derived from neoplastic neuroblasts of the sympathoadrenal lineage
of the neural crest. The tumor has the propensity to arise from sympathetic ganglia and the adrenal
medulla. NB is a most common solid tumor in children. The current treatment approaches
compromise surgical resection, radiotherapy and immunotherapy (9). With grim outcome in nearly
12% of children younger than 15 years of age, high-risk NB remains as a significant disease burden
in pediatric neuro-oncology. Several studies showed that NB is a heterogeneous and complex
disease and a lot of factors such as stage of disease, age and genetic alterations involved in
progression of this malignancy (1-7). Beside genetic heterogeneity and aberrant epigenetic
regulation, abnormal expression of small non-coding RNAs (miRNAs) has revealed indispensable
role in NB establishment and resistance to current standard treatments (8-10). Different expression
of miRNAs between MYC amplified and non-amplified human neuroblastoma tumors corroborated
the role of miRNAs in neurblastoma pathogenesis (10-13). Identification of such factors opens new
insight into their implication as biomarkers in diagnosis, prognosis and therapy in NB (3).
In recent years, immunotherapy has emerged as a promising approach in treatment of multiple
cancers. Recent effective implication of monoclonal antibodies for specific targeted antigens on
cancer cells in human trials provided the grounds to further investigate the therapeutic outcome by
optimizing treatment strategies. Several reports support the ubiquitous expression of gangliosides
such as disialoganglioside GD2 on NB tumor cells, conferring its important role in tumor growth
through different mechanisms, usually executing the function as a receptor for different molecules
such as viruses, toxins, hormones and growth factors(14, 15). Monoclonal antibodies against
disialoganglioside GD2 (anti-GD2 mAbs) have shown therapeutic effects on patients with NB,
usually preventing further relapse in afflicted patients (14). To extend the therapeutic effect and
improve additional therapeutic strategies of anti-GD2 mAbs with on-time administration of the drug
and monitoring on disease progression, identification of highly sensitive diagnostic and predictive
biomarkers are of paramount importance .
This article is protected by copyright. All rights reserved 4
Together, defining novel screening paradigms for early detection of clinical disease or to enhance
the therapeutic outcomes would benefit neuroblastoma patients, particularly patients with advanced-
stage disease.
Neuroblastoma and anti-GD2 drugs
Neuroblastoma cells express high level of gangliosides on their surface(15) . Apart from high
expression of gangliosides on tumor cells surface, gangliosides are also found in microenvirment of
tumors. A number of studies has also demonstrated that gangliosides circulate in the blood stream
of patients (14-16). Several biological effects are, at least in part, linked to the presence of
gangliosides on NB cells such as: modulating cell signaling, acting as receptors, participating in
intercellular communication, tumor cell surface markers, cell cycling, and cell motility (14, 15).
Additionally, these molecules are also involved in various biological pathways that are implicated
in pathogenesis and progression of tumors (15). GD2 has a high expression on the surface of tumor
cells originating from neuroectoderm, such as NB and melanoma, while has limited expression on
normal tissues, principally in the cerebellum and peripheral nerves (14-17). Several reports
revealed that GD2 contribute to the attachment of NB cells to the extracellular matrix (15, 18, 19).
Figure 1 illustrates the structure of GD2 ganglioside. Chemically, GD2 ganglioside is: GalNAcβ1
4 (Neu5Acα2 → 8) (Neu5Acα2 → 3) Galβ1 → 4Glcβ1 → 1’-ceramide. Expression of GD2 in
NB cells is more than benign tumors such as ganglioneuroblastoma and ganglioneuroma. Hence,
GD2 is known as sensitive diagnostic biomarker that contributes to distinguish NB from other
tumors (20-22). Czaplicki et al, presented GD2 as a biomarker in progression of NB where high
plasma levels of GD2 correlated with a more rapid disease progression among afflicted patients.
Besides, their results revealed that the level of GD2 ganglioside in the plasma of NB patients
declined substantially in the course of treatment (23). Because of an evident expression of GD2 on
NB cells surface and role in NB pathogenesis and progression, it potentially serves as a target in NB
therapy (16, 24). GD2 has been utilized as a target in monoclonal antibody therapy and has been the
primary target of antibody recognition in NB (20, 21, 25). Various researches demonstrated that
This article is protected by copyright. All rights reserved 5
normal tissue in brain only expression GD2 where is inaccessible to circulating antibodies. Hence,
peripheral nerves and melanocytes can be suitable target for systemic tumor immunotherapy (24,
26). Various anti-GD2 antibodies with proven safety and efficacy have been assessed in clinical
trials for NB for several years (15, 27). The application of anti-GD2 antibodies is associated with
some limitations such as inability of antibodies to treat bulky tumor and acute pain toxicity related
to GD2 expression on peripheral nerve fibers (1, 27). Researchers have tried to overcome to these
limitations through implementing several strategies such as using blocking antibodies, bypassing
complement activation, targeting of O-acetyl-GD2 derivative that is not expressed on peripheral
nerves surface in order to reduce pain toxicity (28). on the other hand, to improve and enhance anti-
tumor efficacy, the studies have designed new anti-GD2 monoclonal antibodies and fragments
associated with immunotoxins, bispecific T-cell engaging antibodies, radiolabeled antibodies,
immunocytokines, antibody drug conjugates, targeted nanoparticles and chimeric antigen receptors
(15, 29). Table 1 illustrates various anti-GD2 antibodies that are used in clinical for NB treatment.
Several reports indicated that the application of monoclonal antibodies specific to GD2 such as
chimeric 14.18 and 3F8 induce anti-tumor response via monocyte-macrophage mediated
cytotoxicity (phagocytosis), granulocyte-mediated ADCC (granulocyte-ADCC); complement-
mediated cytotoxicity (CMC) by binding to C1q; NK cell-mediated antibody-dependent cell-
mediated cytotoxicity (NK-ADCC).
Associated biomarkers which are modulated in response to the treatment
Of the reported biomarkers modulated by anti-GD2 antibodies, miRNAs have the the potential to
assist prognosis, diagnosis and therapy effect prediction in NB (36-39). Table 3 shows biomarkers
that can regulate some pathways in NB and can serve as novel biomarkers in treatment and response
to therapy in NB. As described by Tivnan et. al, nanoparticles encapsulating miR-34a and
conjugated to a GD2 antibody facilitated tumor-specific delivery following systemic administration
into tumor bearing mice. Their results demonstrated that a significantly increased apoptosis,
decreased tumor growth and a reduction in vascularisation. They demonstrated that miR-34a with
This article is protected by copyright. All rights reserved 6
multi-targets as a novel therapy led to increased levels of the tissue inhibitor metallopeptidase 2
precursor (TIMP2) proteins, accounted for the highly decreased vascularization noted in miR-34a-
treated tumors (40). Another study by Tarek et al showed that there are different response between
NB patients who lack one or more HLA ligands compared with patients who possess all HLA KIR
ligands when treated with anti-GD2 mAbs . NB patients without one or more HLA ligands had
respons better to treatment with lower rates of relapse [Reference]. Their results revealed that the
unlicensed NK cells expresses KIRs for nonself-HLA ligands in these patients can be more
effective in tumor elimination than the licensed NK cells expresses KIRs for self HLA (41). Hence,
KIR-HLA immunogenetics may be acceptable biomarkers in response to treatment and therefore
survival in patients with NB.
One of other biomarkers is minimal residual disease (MDR) that may be the utilized to response to
drugs in NB patient. Several studies demonstrated that to provide more sensitive detection of MRD
and overcome tumor heterogeneity several sets of real-time RT-PCR markers have been assessed
for MDR monitoring in NB patients. Cheung et al, revealed that bone marrow (BM) MRD may be
an early-response biomarker and a consistent independent predictor of survival after anti-GD2
therapy. In this study, BM MRD was mastered before 3F8 treatment and after cycle 2 with utilizing
assessment of four-marker panel (B4GALNT1, PHOX2B, CCND1, and ISL1) by quantitative
reverse transcription polymerase chain reaction ( qRT-PCR) (42). In other study, Hartomo et al
reported a panel including 14 genes for detecting MDR in NB patients. They validated the ability
of 14 commonly used biomarkers to detect MRD based on their expression in NB . They revealed
that the samples as MRD-positive when the expression of one of the 11 biomarkers (CHRNA3,
CRMP1, DBH, DCX, DDC, GABRB3, GAP43, ISL1, KIF1A, PHOX2B and TH) exceeded the
normal range (43). Their result clearly manifested that the MRD detection protocol based on the
expression of a set of 11 real-time RT-PCR biomarkers in NB provide sensitive MRD monitoring
in NB patients. Further studies revealed epigenetic, molecular and genetic factors involved in NB
This article is protected by copyright. All rights reserved 7
pathogenesis and progression, presenting them as strong candidates to predict treatment response to
anti-GD2 drugs (22, 44, 45)
This article is protected by copyright. All rights reserved 8
Circulating biomarkers
The majority of NB patients have been diagnosed in advanced stages of this disease. Fast and easy
detection in NB patients can contribute to diagnosis and monitoring of therapeutic response in these
patients, further on paving the road to early detection of the disease (1). Moreoever, improved
detection methods and identification of powerful biomarkers can increase survival and response to
different drugs. Circulating biomarkers present in body fluids such as serum, plasma and urine are
known as powerful tools in diagnosis, prognosis and therapy in several solid tumors such as NB
(105-107). These biomarkers have a lot of advantages such as non-invasive, easy and fast detection.
Some studies revealed that there are several circulating biomarkers in NB (108, 109). Circulating
miRNA is one of them. miRNAs have significant biology roles in signaling pathways and may
induce or inhibit progression of various cancers such as NB (7, 109-115). Ramraj et al, assessed
miRNAs present in serum that involve in aggressive NB in mouse models of non-metastatic. In this
study, they examined circulating miRNAs signatures and alteration of profile them in mice bearing
favorable or high-risk disease utilizing whole genome human miRNA profiling approach. Their
result revealed that 33 miRNAs of 42 miRNAs showed a significant up regulation (>2 fold) in
animals with high-risk NB. Preferential exclusion of mouse miRNA with sequence homology blast
indicated 11 (off 33 up-regulation) unique human miRNA. Consistently, homology blasting
identified 13 (off 42 down-regulation) miRNAs (116). Proteomics is other fields that contribute to
identifying new biomarkers and help to their clinical application. The utility of proteomics-based
methodologies is such that it offers the potential of identifying signature patterns of multiple
proteins specific to a particular cancer (117, 118). Using of these techniques, previous works
showed mass spectrometry-based protein profiles/pattern differences between serum samples
among children with NB and non-cancer patients (119). In one study, identification of plasma
Complement C3 as a potential biomarker for NB using a quantitative proteomic approach. They
have demonstrated the suitability of the TH-MYCN+/+mouse model of NB for identification of novel
disease biomarkers in humans, and have identified Complement C3 as a candidate plasma
This article is protected by copyright. All rights reserved 9
biomarker for measuring disease state in NB patients (9). In other study, Sandoval et al, the
utilizing of proteomics approaches for investigating other proteins profiles as new candidate serum
biomarkers in an established animal model of advance stage human NB. Their investigations
indicated that some serum proteins differentially expressed in NB model than normal groups.
Hence, these serum proteins may be used as new biomarkers in NB patients. They showed that
investigation serum proteins profile by proteomics approaches can be utilized for monitoring in
early high-risk NB. Their results indicated that up-regulation of five mouse serum proteins 1-
antitrypsin, α2-macroglobulin, serum amyloid A, α1-acid glycoprotein and serum amyloid P-
component) were identified in animal model of NB. Down-regulation of immunoglobulin kappa
chain constant region (Ig κ-C) was found in the serum of animal model of NB as compared to
controls (2.5-fold, p≤ 0.05). In NB patients, apolipoprotein A-IV, α1-acid glycoprotein, serum
amyloid A and haptoglobin were observed to be overexpressed (120). In all studies have not been
used anti-GD2 drugs until obtaining these biomarkers as predictive biomarkers to response to these
drugs. Hence, we suggest that these biomarkers can be used as novel biomarkers after treatment
with anti-GD2 drugs.
Drug-resistant mechanisms
Several studies revealed that treatment failure in patients with poor prognosis lead to resistance to a
diverse range of functionally and structurally unrelated cytotoxic drugs. Amplification of the N-
myc oncogene is one of main powerful biomarkers of poor outcome in NB patients (125). Various
studies approved that appearance multiple drug resistance in patients associated with amplification
of the N- myc oncogene (125-127). Mechanisms involve in amplification that effect on the
phenotype of NB are unclear. Several laboratories have investigated multidrug resistance (MDR)
properties in various malignancies (128, 129). Some reports indicated that MDR1 gene is one of
genes involve in MDR . This gene encodes P-glycoprotein(130). These studies suggested that
MDR1 have main key roles in MDR in various cancers such as NB (130, 131). MRP is another
gene involve in MDR in NB patients. This gene locates on chromosome 16p13.1 and encodes a
This article is protected by copyright. All rights reserved 10
glycoprotein . MRP functions is like P-glycoprotein and observed that this protein show multiple
drug resistance properties in several malignancies (132, 133). Some researches approved that
change expression this gene, led to observed multiple drug resistance properties in NB cells in vitro
(125, 126, 134, 135). In other study, Norris et al, revealed that there is relation between MRP
expression with poor outcome and MRP expression outcome in patients with NB (134). In another
research, Lu et al, was examined effect of the expression of p-glycoprotein (P-gp), multidrug
resistance-associated protein (MRP), and lung resistance protein (LRP) in NB patients. Their results
revealed that multidrug resistance of NB involves the combined effects of MRP, P-gp and LRP.
MRP expression may be a main factor determining prognosis in NB patients (136). In other hands,
there are not any reports that indicate pathways and genes involve in resistance of anti-GD2 drugs
in NB. Hence, these genes and new genes may be used as new options for obtaining anti-GD2 drugs
resistance markers in NB.
Exosome and NB
Exsosomes are known as one of main players in cell-to-cell communications. A number of cells
including tumor cells, stem cells, and some normal cells could release exosomes (137-139). These
nano-carriers are capable to carry a variety of molecules such as proteins, mRNAs, and miRNAs. It
has been shown that these cargos could affect on a sequence of targets in recipient cells which could
lead to change behavior cells (137-139). A large number evidences indicating exosomes released
from tumor cells could contribute to progression of tumor in various stages. These vehicles and
their cargos could have critical roles in main processes including apoptosis, angiogenesis, tumor
growth and resistance to chemotherapy present in beginning and progression of tumors (137, 138).
Various studies showed that these nanoparticles and their cargos affect on progression of NB (140).
In a study by Challagundla and et al, assessed role of exosomes containing miRNAs in resistance to
chemotherapy for NB cells (140). Their results indicated that some exosomal miRNAs such as miR-
21 and miR-155 had main roles in chemotherapy resistance by targeting TLR8-NF-кB and TERF1
This article is protected by copyright. All rights reserved 11
signaling pathways. They showed that up regulation of miR-155 associated with low levels of
TERF1 proteins. These data suggested that exosomes containing various miRNAs could be used as
diagnostic and therapeutic biomarkers in NB patients (140).
Another study, Haug and colleagues assessed potential oncogenic roles of exosomes release from
NB cells in progression of NB (141). They showed that MYCN-amplified NB cell lines released a
variety of exosome-like vesicular particles containing miRNAs . These exosomes carried various
cargos such as proteins ( e.g. CD9, CD63, and TSG101) and miRNAs ( e.g. miR-16, miR-125b,
miR-21, miR-23a, miR-24,miR-25, miR-27b, miR-218, miR-320a, miR-320b and miR-92a). These
exosomal miRNAs act oncomiRs, which could affect on a sequence of targets (e.g. TLR8, NF-kB,
STAT3 and p53) present in NB pathogenesis. These results suggested that exososomal miRNAs might
play a critical roles in progression of NB by targeting a variety of cellular and molecular targets
present in NB pathogenesis (141).
Proteins are other important cargos, which could carry with exosomes. In a study by Marimpietri et
al., indicated that exosomes released from NB cells could carry a number of proteins including
etraspanins, fibronectin, heat shock proteins, MVB proteins, cytoskeleton-related proteins,
prominin-1, basigin and B7-H3, CD9 and CD63 (142). They showed that these proteins could
anticipate in various processes such as defense response, cell differentiation, and cell proliferation.
Therefore, exosomal proteins are other options, which may be used as diagnostic and therapeutic
biomarkers in NB patients (142).
Conclusion
NB is known as a heterogenetic disease in children. In spite of obtaining many achievements in the
finding and understanding of molecular pathways and biological heterogeneity of this disease, NB
has remained a growing malignancy in children. GD2 ganglioside is known as an agent involved in
carcinogenesis and has important biology roles in NB development. Various drugs can block this
agent such as anti-GD2 drugs. The recognition of new biomarkers that associated with prognosis,
This article is protected by copyright. All rights reserved 12
diagnosis, therapy, survival, response to treatment and relapse after treatment with anti-GD2 drugs
treatment may contribute to improve and monitor of disease progression in NB afflicated patients.
This article is protected by copyright. All rights reserved 13
References
1. Schleiermacher G, Janoueix-Lerosey I, Delattre O. Recent insights into the biology of
neuroblastoma. International journal of cancer Journal international du cancer. 2014;135(10):2249-61. Epub
2014/08/16.
2. Louis CU, Shohet JM. Neuroblastoma: molecular pathogenesis and therapy. Annual review of
medicine. 2015;66:49-63. Epub 2014/11/12.
3. Maris JM. Recent advances in neuroblastoma. New England Journal of Medicine.
2010;362(23):2202-11.
4. Brodeur GM, Bagatell R. Mechanisms of neuroblastoma regression. Nature Reviews Clinical
Oncology. 2014;11(12):704-13.
5. Mirzaei H, Khataminfar S, Mohammadparast S, Shahid Sales S, Maftouh M, Mohammadi M, et al.
Circulating microRNAs as Potential Diagnostic Biomarkers and Therapeutic Targets in Gastric Cancer:
Current Status and Future Perspectives. Current medicinal chemistry. 2016;23(36):4135-50.
6. Fathullahzadeh S, Mirzaei H, Honardoost M, Sahebkar A, Salehi M. Circulating microRNA-192 as a
diagnostic biomarker in human chronic lymphocytic leukemia. Cancer gene therapy. 2016;23(10):327-32.
7. Simonian M, Mosallayi M, Mirzaei H. Circulating miR-21 as novel biomarker in gastric cancer:
Diagnostic and prognostic biomarker. 2016.
8. McCLURE M, Shirataki S. Child psychiatry in Japan. Journal of the American Academy of Child &
Adolescent Psychiatry. 1989;28(4):488-92.
9. Kim PY, Tan O, Diakiw SM, Carter D, Sekerye EO, Wasinger VC, et al. Identification of plasma
Complement C3 as a potential biomarker for neuroblastoma using a quantitative proteomic approach. Journal
of proteomics. 2014;96:1-12.
10. Chen Y, Stallings RL. Differential patterns of microRNA expression in neuroblastoma are correlated
with prognosis, differentiation, and apoptosis. Cancer research. 2007;67(3):976-83.
11. Schulte J, Horn S, Samans B, Heukamp L, Eilers U-C, Otto T, et al. N-myc Regulates Oncogenic
microRNAs in neuroblastoma. Cancer Research. 2007;67(9 Supplement):4512-.
12. Beckers A, Van Peer G, Carter DR, Gartlgruber M, Herrmann C, Agarwal S, et al. MYCN-driven
regulatory mechanisms controlling LIN28B in neuroblastoma. Cancer letters. 2015;366(1):123-32.
13. Buechner J, Einvik C. N-myc and noncoding RNAs in neuroblastoma. Molecular Cancer Research.
2012;10(10):1243-53.
14. Suzuki M, Cheung NK. Disialoganglioside GD2 as a therapeutic target for human diseases. Expert
opinion on therapeutic targets. 2015;19(3):349-62. Epub 2015/01/22.
15. Ahmed M, Cheung NK. Engineering anti-GD2 monoclonal antibodies for cancer immunotherapy.
FEBS letters. 2014;588(2):288-97. Epub 2013/12/04.
16. Saito M, Yu RK, Cheung NK. Ganglioside GD2 specificity of monoclonal antibodies to human
neuroblastoma cell. Biochemical and biophysical research communications. 1985;127(1):1-7. Epub
1985/02/28.
17. Berois N, Osinaga E. Glycobiology of neuroblastoma: impact on tumor behavior, prognosis, and
therapeutic strategies. Frontiers in oncology. 2014;4.
18. Cheung N-KV, Cheung IY, Kushner BH, Ostrovnaya I, Chamberlain E, Kramer K, et al. Murine
anti-GD2 monoclonal antibody 3F8 combined with granulocyte-macrophage colony-stimulating factor and
13-cis-retinoic acid in high-risk patients with stage 4 neuroblastoma in first remission. Journal of Clinical
Oncology. 2012:JCO. 2011.41. 3807.
19. Kushner BH, Kramer K, Modak S, Cheung N-KV. Successful multifold dose escalation of anti-GD2
monoclonal antibody 3F8 in patients with neuroblastoma: a phase I study. Journal of Clinical Oncology.
2011;29(9):1168-74.
20. Kushner B, Kramer K, Modak S, Cheung N, editors. Anti-GD2 anitbody 3F8 plus granulocyte-
macrophage colony stimulating factor (GM-CSF) for primary refractory neuroblastoma (NB) in the bone
marrow (BM). Proc Amer Soc Clin Oncol; 2007.
21. Thurin J, Thurin M, Kimoto Y, Herlyn M, Lubeck MD, Elder DE, et al. Monoclonal antibody-
defined correlations in melanoma between levels of GD2 and GD3 antigens and antibody-mediated
cytotoxicity. Cancer research. 1987;47(5):1229-33.
22. Imber B, Olow A, Karski E, Allen I, DuBois S, Haas-Kogan D. Biomarker Modulations in Children
With Neuroblastoma Following Treatment With 131I-mIBG. International Journal of Radiation Oncology•
Biology• Physics. 2014;90(1):S729-S30.
This article is protected by copyright. All rights reserved 14
23. Czaplicki D, Horwacik I, Kowalczyk A, Wieczorek A, Bolek-Marzec K, Balwierz W, et al. New
method for quantitative analysis of GD2 ganglioside in plasma of neuroblastoma patients. Acta biochimica
Polonica. 2009;56(3):423-31. Epub 2009/09/03.
24. Yang RK, Sondel PM. Anti-GD2 strategy in the treatment of neuroblastoma. Drugs of the future.
2010;35(8):665.
25. Brown BS. Anti-GD2 Etoposide-Loaded Immunoliposomes for the Treatment of GD2 Positive
Tumors. 2014.
26. Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, et al. Anti-GD2
antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. The New England journal of
medicine. 2010;363(14):1324-34. Epub 2010/10/01.
27. Matthay KK, George RE, Alice LY. Promising therapeutic targets in neuroblastoma. Clinical Cancer
Research. 2012;18(10):2740-53.
28. Alvarez-Rueda N, Desselle A, Cochonneau D, Chaumette T, Clemenceau B, Leprieur S, et al. A
monoclonal antibody to O-acetyl-GD2 ganglioside and not to GD2 shows potent anti-tumor activity without
peripheral nervous system cross-reactivity. PLoS One. 2011;6(9):e25220.
29. Dobrenkov K, Cheung N-KV, editors. GD2-targeted immunotherapy and radioimmunotherapy.
Seminars in oncology; 2014: Elsevier.
30. Cheung NK, Guo H, Hu J, Tassev DV, Cheung IY. Humanizing murine IgG3 anti-GD2 antibody
m3F8 substantially improves antibody-dependent cell-mediated cytotoxicity while retaining targeting in
vivo. Oncoimmunology. 2012;1(4):477-86. Epub 2012/07/04.
31. Kushner BH, Kramer K, Modak S, Cheung NK. Successful multifold dose escalation of anti-GD2
monoclonal antibody 3F8 in patients with neuroblastoma: a phase I study. Journal of clinical oncology :
official journal of the American Society of Clinical Oncology. 2011;29(9):1168-74. Epub 2011/02/24.
32. Cheung NK, Modak S. Oral (1-->3),(1-->4)-beta-D-glucan synergizes with antiganglioside GD2
monoclonal antibody 3F8 in the therapy of neuroblastoma. Clinical cancer research : an official journal of
the American Association for Cancer Research. 2002;8(5):1217-23. Epub 2002/05/15.
33. Cheung NK, Cheung IY, Kushner BH, Ostrovnaya I, Chamberlain E, Kramer K, et al. Murine anti-
GD2 monoclonal antibody 3F8 combined with granulocyte-macrophage colony-stimulating factor and 13-
cis-retinoic acid in high-risk patients with stage 4 neuroblastoma in first remission. Journal of clinical
oncology : official journal of the American Society of Clinical Oncology. 2012;30(26):3264-70. Epub
2012/08/08.
34. Ozkaynak MF, Sondel PM, Krailo MD, Gan J, Javorsky B, Reisfeld RA, et al. Phase I study of
chimeric human/murine anti-ganglioside G(D2) monoclonal antibody (ch14.18) with granulocyte-
macrophage colony-stimulating factor in children with neuroblastoma immediately after hematopoietic stem-
cell transplantation: a Children's Cancer Group Study. Journal of clinical oncology : official journal of the
American Society of Clinical Oncology. 2000;18(24):4077-85. Epub 2000/12/19.
35. Terme M, Dorvillius M, Cochonneau D, Chaumette T, Xiao W, Diccianni MB, et al. Chimeric
antibody c.8B6 to O-acetyl-GD2 mediates the same efficient anti-neuroblastoma effects as therapeutic
ch14.18 antibody to GD2 without antibody induced allodynia. PLoS One. 2014;9(2):e87210. Epub
2014/02/13.
36. Annibali D, Gioia U, Savino M, Laneve P, Caffarelli E, Nasi S. A new module in neural
differentiation control: two microRNAs upregulated by retinoic acid, miR-9 and -103, target the
differentiation inhibitor ID2. PLoS One. 2012;7(7):e40269. Epub 2012/08/01.
37. Bolkar ST, Ghadge MS, Raste AS. Biochemical parameters in neuroblastoma. Indian journal of
clinical biochemistry : IJCB. 2008;23(3):293-5. Epub 2008/07/01.
38. Cheung IY, Farazi TA, Ostrovnaya I, Xu H, Tran H, Mihailovic A, et al. Deep MicroRNA
sequencing reveals downregulation of miR-29a in neuroblastoma central nervous system metastasis. Genes,
chromosomes & cancer. 2014;53(10):803-14. Epub 2014/06/06.
39. Cheng L, Yang T, Kuang Y, Kong B, Yu S, Shu H, et al. MicroRNA-23a promotes neuroblastoma
cell metastasis by targeting CDH1. Oncology letters. 2014;7(3):839-45. Epub 2014/02/13.
40. Tivnan A, Tracey L, Buckley PG, Alcock LC, Davidoff AM, Stallings RL. MicroRNA-34a is a
potent tumor suppressor molecule in vivo in neuroblastoma. BMC cancer. 2011;11:33. Epub 2011/01/27.
41. Tarek N, Le Luduec JB, Gallagher MM, Zheng J, Venstrom JM, Chamberlain E, et al. Unlicensed
NK cells target neuroblastoma following anti-GD2 antibody treatment. The Journal of clinical investigation.
2012;122(9):3260-70. Epub 2012/08/07.
This article is protected by copyright. All rights reserved 15
42. Cheung IY, Feng Y, Gerald W, Cheung NK. Exploiting gene expression profiling to identify novel
minimal residual disease markers of neuroblastoma. Clinical cancer research : an official journal of the
American Association for Cancer Research. 2008;14(21):7020-7. Epub 2008/11/05.
43. Hartomo TB, Kozaki A, Hasegawa D, Van Huyen Pham T, Yamamoto N, Saitoh A, et al. Minimal
residual disease monitoring in neuroblastoma patients based on the expression of a set of real-time RT-PCR
markers in tumor-initiating cells. Oncology reports. 2013;29(4):1629-36. Epub 2013/02/19.
44. Lodrini M, Oehme I, Schroeder C, Milde T, Schier MC, Kopp-Schneider A, et al. MYCN and
HDAC2 cooperate to repress miR-183 signaling in neuroblastoma. Nucleic acids research.
2013;41(12):6018-33. Epub 2013/04/30.
45. Hsu WM, Hsieh FJ, Jeng YM, Kuo ML, Tsao PN, Lee H, et al. GRP78 expression correlates with
histologic differentiation and favorable prognosis in neuroblastic tumors. International journal of cancer
Journal international du cancer. 2005;113(6):920-7. Epub 2004/10/30.
46. Pandey GK, Mitra S, Subhash S, Hertwig F, Kanduri M, Mishra K, et al. The risk-associated long
noncoding RNA NBAT-1 controls neuroblastoma progression by regulating cell proliferation and neuronal
differentiation. Cancer cell. 2014;26(5):722-37. Epub 2014/12/18.
47. Nozato M, Kaneko S, Nakagawara A, Komuro H. Epithelial-mesenchymal transition-related gene
expression as a new prognostic marker for neuroblastoma. International journal of oncology.
2013;42(1):134-40. Epub 2012/11/09.
48. Caren H, Djos A, Nethander M, Sjoberg RM, Kogner P, Enstrom C, et al. Identification of
epigenetically regulated genes that predict patient outcome in neuroblastoma. BMC cancer. 2011;11:66.
Epub 2011/02/15.
49. Mestdagh P, Bostrom AK, Impens F, Fredlund E, Van Peer G, De Antonellis P, et al. The miR-17-
92 microRNA cluster regulates multiple components of the TGF-beta pathway in neuroblastoma. Molecular
cell. 2010;40(5):762-73. Epub 2010/12/15.
50. Zhao Z, Ma X, Shelton SD, Sung DC, Li M, Hernandez D, et al. A combined gene expression and
functional study reveals the crosstalk between N-Myc and differentiation-inducing microRNAs in
neuroblastoma cells. Oncotarget. 2016;15(10):12676.
51. Xin C, Buhe B, Hongting L, Chuanmin Y, Xiwei H, Hong Z, et al. MicroRNA-15a promotes
neuroblastoma migration by targeting reversion-inducing cysteine-rich protein with Kazal motifs (RECK)
and regulating matrix metalloproteinase-9 expression. The FEBS journal. 2013;280(3):855-66. Epub
2012/11/28.
52. Wang Z, Lei H, Sun Q. MicroRNA-141 and its associated gene FUS modulate proliferation,
migration and cisplatin chemosensitivity in neuroblastoma cell lines. Oncol Rep. 2016;35(5):2943-51.
53. He XY, Tan ZL, Mou Q, Liu FJ, Liu S, Yu CW, et al. microRNA-221 enhances MYCN via targeting
nemo-like kinase, and functions as an oncogene related to poor prognosis in neuroblastoma. Clin Cancer
Res. 2016;21(10):1078-0432.
54. Patil KS, Basak I, Pal R, Ho HP, Alves G, Chang EJ, et al. A Proteomics Approach to Investigate
miR-153-3p and miR-205-5p Targets in Neuroblastoma Cells. PLoS One. 2015;10(12).
55. Zhang H, Qi M, Li S, Qi T, Mei H, Huang K, et al. microRNA-9 targets matrix metalloproteinase 14
to inhibit invasion, metastasis, and angiogenesis of neuroblastoma cells. Molecular cancer therapeutics.
2012;11(7):1454-66. Epub 2012/05/09.
56. Qu H, Zheng L, Song H, Jiao W, Li D, Fang E, et al. microRNA-558 facilitates the expression of
hypoxia-inducible factor 2 alpha through binding to 5'-untranslated region in neuroblastoma. Oncotarget.
2016;7(26):40657-73.
57. Li Z, Xu Z, Xie Q, Gao W, Xie J, Zhou L. miR-1303 promotes the proliferation of neuroblastoma
cell SH-SY5Y by targeting GSK3beta and SFRP1. Biomed Pharmacother. 2016;83:508-13.
58. Trager C, Kogner P, Lindskog M, Ponthan F, Kullman A, Kagedal B. Quantitative analysis of
tyrosine hydroxylase mRNA for sensitive detection of neuroblastoma cells in blood and bone marrow.
Clinical chemistry. 2003;49(1):104-12. Epub 2003/01/01.
59. Das S, Bryan K, Buckley PG, Piskareva O, Bray IM, Foley N, et al. Modulation of neuroblastoma
disease pathogenesis by an extensive network of epigenetically regulated microRNAs. Oncogene.
2013;32(24):2927-36. Epub 2012/07/17.
60. Oltra S, Martinez F, Orellana C, Grau E, Fernandez JM, Canete A, et al. The doublecortin gene, a
new molecular marker to detect minimal residual disease in neuroblastoma. Diagnostic molecular pathology
: the American journal of surgical pathology, part B. 2005;14(1):53-7. Epub 2005/02/17.
61. Osman J, Galli S, Hanafy M, Tang X, Ahmed A. Identification of novel biomarkers in
neuroblastoma associated with the risk for bone marrow metastasis: a pilot study. Clinical & translational
This article is protected by copyright. All rights reserved 16
oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer
Institute of Mexico. 2013;15(11):953-8. Epub 2013/04/05.
62. Buechner J, Tomte E, Haug BH, Henriksen JR, Lokke C, Flaegstad T, et al. Tumour-suppressor
microRNAs let-7 and mir-101 target the proto-oncogene MYCN and inhibit cell proliferation in MYCN-
amplified neuroblastoma. British journal of cancer. 2011;105(2):296-303. Epub 2011/06/10.
63. Naraparaju K, Kolla V, Zhuang T, Higashi M, Iyer R, Kolla S, et al. Role of microRNAs in
epigenetic silencing of the CHD5 tumor suppressor gene in neuroblastomas. Oncotarget. 2016;7(13):15977-
85.
64. Brodeur GM, Nakagawara A, Yamashiro DJ, Ikegaki N, Liu XG, Azar CG, et al. Expression of
TrkA, TrkB and TrkC in human neuroblastomas. Journal of neuro-oncology. 1997;31(1-2):49-55. Epub
1997/01/01.
65. Li YG, He JH, Yu L, Hang ZP, Li W, Shun WH, et al. microRNA202 suppresses MYCN expression
under the control of E2F1 in the neuroblastoma cell line LAN5. Molecular medicine reports. 2014;9(2):541-
6. Epub 2013/12/18.
66. Nakamura K, Martin KC, Jackson JK, Beppu K, Woo CW, Thiele CJ. Brain-derived neurotrophic
factor activation of TrkB induces vascular endothelial growth factor expression via hypoxia-inducible factor-
1alpha in neuroblastoma cells. Cancer Res. 2006;66(8):4249-55. Epub 2006/04/19.
67. Ryan J, Tivnan A, Fay J, Bryan K, Meehan M, Creevey L, et al. MicroRNA-204 increases sensitivity
of neuroblastoma cells to cisplatin and is associated with a favourable clinical outcome. British journal of
cancer. 2012;107(6):967-76. Epub 2012/08/16.
68. Bray I, Bryan K, Prenter S, Buckley PG, Foley NH, Murphy DM, et al. Widespread dysregulation of
MiRNAs by MYCN amplification and chromosomal imbalances in neuroblastoma: association of miRNA
expression with survival. PLoS One. 2009;4(11):e7850. Epub 2009/11/20.
69. Zhao D, Tian Y, Li P, Wang L, Xiao A, Zhang M, et al. MicroRNA-203 inhibits the malignant
progression of neuroblastoma by targeting Sam68. Mol Med Rep. 2015;12(4):5554-60.
70. Zhang L, Yeger H, Das B, Irwin MS, Baruchel S. Tissue microenvironment modulates CXCR4
expression and tumor metastasis in neuroblastoma. Neoplasia (New York, NY). 2007;9(1):36-46. Epub
2007/02/28.
71. Zhang H, Liu T, Yi S, Gu L, Zhou M. Targeting MYCN IRES in MYCN-amplified neuroblastoma
with miR-375 inhibits tumor growth and sensitizes tumor cells to radiation. Molecular oncology. 2015. Epub
2015/04/14.
72. Creevey L, Ryan J, Harvey H, Bray IM, Meehan M, Khan AR, et al. MicroRNA-497 increases
apoptosis in MYCN amplified neuroblastoma cells by targeting the key cell cycle regulator WEE1.
Molecular cancer. 2013;12:23. Epub 2013/03/28.
73. Hsu WM, Lee H, Juan HF, Shih YY, Wang BJ, Pan CY, et al. Identification of GRP75 as an
independent favorable prognostic marker of neuroblastoma by a proteomics analysis. Clinical cancer
research : an official journal of the American Association for Cancer Research. 2008;14(19):6237-45. Epub
2008/10/03.
74. Kitanaka C, Kato K, Ijiri R, Sakurada K, Tomiyama A, Noguchi K, et al. Increased Ras expression
and caspase-independent neuroblastoma cell death: possible mechanism of spontaneous neuroblastoma
regression. Journal of the National Cancer Institute. 2002;94(5):358-68. Epub 2002/03/07.
75. Singh A, Rokes C, Gireud M, Fletcher S, Baumgartner J, Fuller G, et al. Retinoic acid induces REST
degradation and neuronal differentiation by modulating the expression of SCF(beta-TRCP) in neuroblastoma
cells. Cancer. 2011;117(22):5189-202. Epub 2011/04/28.
76. Xiang X, Mei H, Zhao X, Pu J, Li D, Qu H, et al. miRNA-337-3p suppresses neuroblastoma
progression by repressing the transcription of matrix metalloproteinase 14. Oncotarget. 2015;6(26):22452-
66.
77. Rubie H, Hartmann O, Michon J, Frappaz D, Coze C, Chastagner P, et al. N-Myc gene amplification
is a major prognostic factor in localized neuroblastoma: results of the French NBL 90 study. Neuroblastoma
Study Group of the Societe Francaise d'Oncologie Pediatrique. Journal of clinical oncology : official journal
of the American Society of Clinical Oncology. 1997;15(3):1171-82. Epub 1997/03/01.
78. Pedram M, Heidari M, Keikhaei B, Malamiri RA, Poopak B, Fekri K. Impact of N-myc
Amplification on Median Survival in Children With Neuroblastoma. Journal of Comprehensive Pediatrics.
2012;3(1):29-33.
79. Yoshimoto M, Caminada De Toledo SR, Monteiro Caran EM, de Seixas MT, de Martino Lee ML,
de Campos Vieira Abib S, et al. MYCN gene amplification. Identification of cell populations containing
This article is protected by copyright. All rights reserved 17
double minutes and homogeneously staining regions in neuroblastoma tumors. The American journal of
pathology. 1999;155(5):1439-43. Epub 1999/11/07.
80. Hiyama E, Hiyama K, Yokoyama T, Fukuba I, Yamaoka H, Shay JW, et al. Rapid detection of
MYCN gene amplification and telomerase expression in neuroblastoma. Clinical cancer research : an official
journal of the American Association for Cancer Research. 1999;5(3):601-9. Epub 1999/04/01.
81. Laneve P, Di Marcotullio L, Gioia U, Fiori ME, Ferretti E, Gulino A, et al. The interplay between
microRNAs and the neurotrophin receptor tropomyosin-related kinase C controls proliferation of human
neuroblastoma cells. Proceedings of the National Academy of Sciences of the United States of America.
2007;104(19):7957-62. Epub 2007/05/08.
82. Mandili G, Marini C, Carta F, Zanini C, Prato M, Khadjavi A, et al. Identification of
phosphoproteins as possible differentiation markers in all-trans-retinoic acid-treated neuroblastoma cells.
PLoS One. 2011;6(5):e18254. Epub 2011/05/17.
83. Cheng M, Liu L, Lao Y, Liao W, Liao M, Luo X, et al. MicroRNA-181a suppresses parkin-mediated
mitophagy and sensitizes neuroblastoma cells to mitochondrial uncoupler-induced apoptosis. Oncotarget.
2016;7(27):42274-87.
84. Rihani A, Van Goethem A, Ongenaert M, De Brouwer S, Volders PJ, Agarwal S, et al. Genome
wide expression profiling of p53 regulated miRNAs in neuroblastoma. Scientific reports. 2015;5:9027. Epub
2015/03/13.
85. Wu K, Yang L, Chen J, Zhao H, Wang J, Xu S, et al. miR-362-5p inhibits proliferation and
migration of neuroblastoma cells by targeting phosphatidylinositol 3-kinase-C2beta. FEBS Lett.
2015;589(15):1911-9.
86. Fabbri E, Montagner G, Bianchi N, Finotti A, Borgatti M, Lampronti I, et al. MicroRNA miR-93-5p
regulates expression of IL-8 and VEGF in neuroblastoma SK-N-AS cells. Oncol Rep. 2016;35(5):2866-72.
87. Venstrom JM, Zheng J, Noor N, Danis KE, Yeh AW, Cheung IY, et al. KIR and HLA genotypes are
associated with disease progression and survival following autologous hematopoietic stem cell
transplantation for high-risk neuroblastoma. Clinical cancer research : an official journal of the American
Association for Cancer Research. 2009;15(23):7330-4. Epub 2009/11/26.
88. Gao SL, Wang LZ, Liu HY, Liu DL, Xie LM, Zhang ZW. miR-200a inhibits tumor proliferation by
targeting AP-2gamma in neuroblastoma cells. Asian Pacific journal of cancer prevention : APJCP.
2014;15(11):4671-6. Epub 2014/06/28.
89. Evangelisti C, Florian MC, Massimi I, Dominici C, Giannini G, Galardi S, et al. MiR-128 up-
regulation inhibits Reelin and DCX expression and reduces neuroblastoma cell motility and invasiveness.
FASEB journal : official publication of the Federation of American Societies for Experimental Biology.
2009;23(12):4276-87. Epub 2009/08/29.
90. Qiao J, Lee S, Paul P, Theiss L, Tiao J, Qiao L, et al. miR-335 and miR-363 regulation of
neuroblastoma tumorigenesis and metastasis. Surgery. 2013;154(2):226-33. Epub 2013/06/29.
91. Lynch J, Fay J, Meehan M, Bryan K, Watters KM, Murphy DM, et al. MiRNA-335 suppresses
neuroblastoma cell invasiveness by direct targeting of multiple genes from the non-canonical TGF-beta
signalling pathway. Carcinogenesis. 2012;33(5):976-85. Epub 2012/03/03.
92. Tong Q, Zheng L, Tang S, Li S, Jiang G, Cai J, et al. Expression of Fas and FasL in human
neuroblastoma and its clinical significance. World J Pediatr. 2007;3(3):209-13.
93. Decock A, Ongenaert M, Hoebeeck J, De Preter K, Van Peer G, Van Criekinge W, et al. Genome-
wide promoter methylation analysis in neuroblastoma identifies prognostic methylation biomarkers. Genome
biology. 2012;13(10):R95. Epub 2012/10/05.
94. Liu G, Xu Z, Hao D. MicroRNA451 inhibits neuroblastoma proliferation, invasion and migration by
targeting macrophage migration inhibitory factor. Mol Med Rep. 2016;13(3):2253-60.
95. Foley NH, Bray IM, Tivnan A, Bryan K, Murphy DM, Buckley PG, et al. MicroRNA-184 inhibits
neuroblastoma cell survival through targeting the serine/threonine kinase AKT2. Molecular cancer.
2010;9:83. Epub 2010/04/23.
96. Trager C, Vernby A, Kullman A, Ora I, Kogner P, Kagedal B. mRNAs of tyrosine hydroxylase and
dopa decarboxylase but not of GD2 synthase are specific for neuroblastoma minimal disease and predicts
outcome for children with high-risk disease when measured at diagnosis. International journal of cancer
Journal international du cancer. 2008;123(12):2849-55. Epub 2008/09/25.
97. Cheung IY, Vickers A, Cheung NK. Sialyltransferase STX (ST8SiaII): a novel molecular marker of
metastatic neuroblastoma. International journal of cancer Journal international du cancer. 2006;119(1):152-6.
Epub 2006/02/02.
This article is protected by copyright. All rights reserved 18
98. Inamori K, Gu J, Ohira M, Kawasaki A, Nakamura Y, Nakagawa T, et al. High expression of N-
acetylglucosaminyltransferase V in favorable neuroblastomas: Involvement of its effect on apoptosis. FEBS
letters. 2006;580(2):627-32. Epub 2006/01/13.
99. Yanez Y, Grau E, Rodriguez-Cortez VC, Hervas D, Vidal E, Noguera R, et al. Two independent
epigenetic biomarkers predict survival in neuroblastoma. Clinical epigenetics. 2015;7(1):16. Epub
2015/03/15.
100. Berois N, Blanc E, Ripoche H, Mergui X, Trajtenberg F, Cantais S, et al. ppGalNAc-T13: a new
molecular marker of bone marrow involvement in neuroblastoma. Clinical chemistry. 2006;52(9):1701-12.
Epub 2006/07/29.
101. Berois N, Gattolliat CH, Barrios E, Capandeguy L, Douc-Rasy S, Valteau-Couanet D, et al.
GALNT9 gene expression is a prognostic marker in neuroblastoma patients. Clinical chemistry.
2013;59(1):225-33. Epub 2012/11/09.
102. Ho WL, Che MI, Chou CH, Chang HH, Jeng YM, Hsu WM, et al. B3GNT3 expression suppresses
cell migration and invasion and predicts favorable outcomes in neuroblastoma. Cancer science.
2013;104(12):1600-8. Epub 2013/10/15.
103. Chang HH, Chen CH, Chou CH, Liao YF, Huang MJ, Chen YH, et al. beta-1,4-
Galactosyltransferase III enhances invasive phenotypes via beta1-integrin and predicts poor prognosis in
neuroblastoma. Clinical cancer research : an official journal of the American Association for Cancer
Research. 2013;19(7):1705-16. Epub 2013/02/28.
104. Hsu WM, Che MI, Liao YF, Chang HH, Chen CH, Huang YM, et al. B4GALNT3 expression
predicts a favorable prognosis and suppresses cell migration and invasion via beta(1) integrin signaling in
neuroblastoma. The American journal of pathology. 2011;179(3):1394-404. Epub 2011/07/12.
105. Combaret V, Audoynaud C, Iacono I, Favrot M-C, Schell M, Bergeron C, et al. Circulating MYCN
DNA as a tumor-specific marker in neuroblastoma patients. Cancer research. 2002;62(13):3646-8.
106. Ghossein RA, Bhattacharya S, Rosai J. Molecular detection of micrometastases and circulating
tumor cells in solid tumors. Clinical Cancer Research. 1999;5(8):1950-60.
107. Ramraj SK, Pandian V, Khan FH, Aravindan S, Herman TS, Natarajan M, et al. Profiling of
circulating miRNAs is a critical prognostic biomarker tool for high-risk neuroblastoma. Cancer Research.
2014;74(19 Supplement):5229-.
108. Allegra A, Alonci A, Campo S, Penna G, Petrungaro A, Gerace D, et al. Circulating microRNAs:
new biomarkers in diagnosis, prognosis and treatment of cancer (review). International journal of oncology.
2012;41(6):1897-912.
109. Ma R, Jiang T, Kang X. Circulating microRNAs in cancer: origin, function and application. J Exp
Clin Cancer Res. 2012;31(1):38.
110. Mirzaei H, Gholamin S, Shahidsales S, Sahebkar A, Jafaari MR, Mirzaei HR, et al. MicroRNAs as
potential diagnostic and prognostic biomarkers in melanoma. European Journal of Cancer. 2016;53:25-32.
111. Gholamin S, Pasdar A, Sadegh Khorrami M, Mirzaei H, Reza Mirzaei H, Salehi R, et al. The
potential for circulating microRNAs in the diagnosis of myocardial infarction: A novel approach to disease
diagnosis and treatment. Current pharmaceutical design. 2016;22(3):397-403.
112. Reza Mirzaei H. Circulating microRNAs in hepatocellular carcinoma: potential diagnostic and
prognostic biomarkers. Current Pharmaceutical Design.22.
113. Salarini R, Sahebkar A, Mirzaei H, Jaafari M, Riahi M, Hadjati J, et al. Epi-drugs and Epi-miRs:
Moving beyond current cancer therapies. Current cancer drug targets. 2015.
114. Mirzaei H, Naseri G, Rezaee R, Mohammadi M, Banikazemi Z, Reza Mirzaei H, et al. Curcumin: A
new candidate for melanoma therapy? International Journal of Cancer. 2016.
115. Mohammadi M, Goodarzi M, Jaafari MR, Mirzaei HR, Mirzaei H. Circulating microRNA: a new
candidate for diagnostic biomarker in neuroblastoma. Cancer Gene Ther. 2016;23(11):371-2.
116. Ramraj SK, Aravindan S, Somasundaram DB, Herman TS, Natarajan M, Aravindan N. Serum-
circulating miRNAs predict neuroblastoma progression in mouse model of high-risk metastatic disease.
Oncotarget. 2016;7(14):18605-19.
117. Combaret V, Bergeron C, Bréjon S, Iacono I, Perol D, Négrier S, et al. Protein chip array profiling
analysis of sera from neuroblastoma patients. Cancer letters. 2005;228(1):91-6.
118. Hsu W-M, Lee H, Juan H-F, Shih Y-Y, Wang B-J, Pan C-Y, et al. Identification of GRP75 as an
independent favorable prognostic marker of neuroblastoma by a proteomics analysis. Clinical Cancer
Research. 2008;14(19):6237-45.
119. Diamandis EP. Mass spectrometry as a diagnostic and a cancer biomarker discovery tool
opportunities and potential limitations. Molecular & Cellular Proteomics. 2004;3(4):367-78.
This article is protected by copyright. All rights reserved 19
120. Sandoval JA, Turner KE, Hoelz DJ, Rescorla FJ, Hickey RJ, Malkas LH. Serum protein profiling to
identify high-risk neuroblastoma: preclinical relevance of blood-based biomarkers. Journal of Surgical
Research. 2007;142(2):268-74.
121. Kim PY, Tan O, Diakiw SM, Carter D, Sekerye EO, Wasinger VC, et al. Identification of plasma
complement C3 as a potential biomarker for neuroblastoma using a quantitative proteomic approach. J
Proteomics. 2014;96:1-12. Epub 2013/11/10.
122. Sandoval JA, Turner KE, Hoelz DJ, Rescorla FJ, Hickey RJ, Malkas LH. Serum protein profiling to
identify high-risk neuroblastoma: preclinical relevance of blood-based biomarkers. The Journal of surgical
research. 2007;142(2):268-74. Epub 2007/08/31.
123. Smith SJ, Diehl NN, Smith BD, Mohney BG. Urine catecholamine levels as diagnostic markers for
neuroblastoma in a defined population: implications for ophthalmic practice. Eye (London, England).
2010;24(12):1792-6. Epub 2010/09/25.
124. Helson L, Ghavimi F, Wu CJ, Fieisher M, Schwartz MK. Carcinoembryonic antigen in children with
neuroblastoma. Journal of the National Cancer Institute. 1976;57(3):725-6. Epub 1976/09/01.
125. Alisi A, Cho WC, Locatelli F, Fruci D. Multidrug resistance and cancer stem cells in neuroblastoma
and hepatoblastoma. International journal of molecular sciences. 2013;14(12):24706-25.
126. Bordow SB, Haber M, Madafiglio J, Cheung B, Marshall GM, Norris MD. Expression of the
multidrug resistance-associated protein (MRP) gene correlates with amplification and overexpression of the
N-myc oncogene in childhood neuroblastoma. Cancer research. 1994;54(19):5036-40.
127. Seeger RC, Brodeur GM, Sather H, Dalton A, Siegel SE, Wong KY, et al. Association of multiple
copies of the N-myc oncogene with rapid progression of neuroblastomas. New England Journal of Medicine.
1985;313(18):1111-6.
128. Donnenberg VS, Donnenberg AD. Multiple drug resistance in cancer revisited: the cancer stem cell
hypothesis. The Journal of Clinical Pharmacology. 2005;45(8):872-7.
129. Ozben T. Mechanisms and strategies to overcome multiple drug resistance in cancer. FEBS letters.
2006;580(12):2903-9.
130. Haber M, Bordow S, Haber P, Marshall G, Stewart B, Norris M. The prognostic value of MDR1
gene expression in primary untreated neuroblastoma. European Journal of Cancer. 1997;33(12):2031-6.
131. Minemura M, Tanimura H, Tabor E. Overexpression of multidrug resistance genes MDR1 and
cMOAT in human hepatocellular carcinoma and hepatoblastoma cell lines. International journal of oncology.
1999;15(3):559-622.
132. Manohar CF, Bray JA, Salwen HR, Madafiglio J, Cheng A, Flemming C, et al. MYCN-mediated
regulation of the MRP1 promoter in human neuroblastoma. Oncogene. 2004;23(3):753-62.
133. Matsunaga T, Shirasawa H, Hishiki T, Enomoto H, Kouchi K, Ohtsuka Y, et al. Expression of MRP
and cMOAT in childhood neuroblastomas and malignant liver tumors and its relevance to clinical behavior.
Cancer science. 1998;89(12):1276-83.
134. Norris M, Burkhart C, Marshall G, Weiss W, Haber M. Expression of Nmyc and MRP genes and
their relationship to Nmyc gene dosage and tumor formation in a murine neuroblastoma model. Medical and
pediatric oncology. 2000;35(6):585-9.
135. Sun Y, Li Q, Xu Y, Pu C, Zhao L, Guo Z, et al. Study of the mechanisms underlying the reversal of
multidrug resistance of human neuroblastoma multidrug-resistant cell line SK-N-SH/MDR1 by low-intensity
pulsed ultrasound. Oncology reports. 2013;29(5):1939-45.
136. Lu QJ, Dong F, Zhang JH, Li XH, Ma Y, Jiang WG. Expression of multidrug resistance-related
markers in primary neuroblastoma. Chinese medical journal. 2004;117(9):1358-63. Epub 2004/09/21.
137. Mirzaei H, Sahebkar A, Jaafari MR, Goodarzi M, Mirzaei HR. Diagnostic and Therapeutic Potential
of Exosomes in Cancer: The Beginning of a New Tale? Journal of Cellular Physiology. 2016.
138. Saadatpour L, Fadaee E, Fadaei S, Mansour RN, Mohammadi M, Mousavi S, et al. Glioblastoma:
exosome and microRNA as novel diagnosis biomarkers. Cancer Gene Therapy. 2016;23(12):415-8.
139. Nedaeinia R, Manian M, Jazayeri M, Ranjbar M, Salehi R, Sharifi M, et al. Circulating exosomes
and exosomal microRNAs as biomarkers in gastrointestinal cancer. Cancer Gene Therapy. 2016.
140. Challagundla KB, Wise PM, Neviani P, Chava H, Murtadha M, Xu T, et al. Exosome-mediated
transfer of microRNAs within the tumor microenvironment and neuroblastoma resistance to chemotherapy. J
Natl Cancer Inst. 2015;107(7).
141. Haug BH, Hald OH, Utnes P, Roth SA, Lokke C, Flaegstad T, et al. Exosome-like Extracellular
Vesicles from MYCN-amplified Neuroblastoma Cells Contain Oncogenic miRNAs. Anticancer Res.
2015;35(5):2521-30.
This article is protected by copyright. All rights reserved 20
142. Marimpietri D, Petretto A, Raffaghello L, Pezzolo A, Gagliani C, Tacchetti C, et al. Proteome
profiling of neuroblastoma-derived exosomes reveal the expression of proteins potentially involved in tumor
progression. PLoS One. 2013;8(9).
This article is protected by copyright. All rights reserved 21
Fig 1. Schematic chemical structure of GD2 ganglioside.
Fig 2. Schematic diagram of various miRNAs network in NB tumorigenesis and metastasis
This article is protected by copyright. All rights reserved 22
Table 1. Current clinical trials testing various GD2 antibodies for treatment of NB
Antibody
Kind of
Antibody
Comments
Clinical Status
Ref
hu14.18
Humanized
Naked antibody
Phase I (NCT00743496)
(15)
hu14.18 +IL-2 + GM-CSF + NK Cells
Humanized
Naked antibody
Phase I (NCT01576692)
(15)
hu14.18+IL-2 + G-CSF + GM-CSF
Humanized
Naked antibody
Phase II (NCT01857934)
(15)
hu3F8
Humanized
Naked antibody
Phase I (NCT01419834)
(30)
hu3F8 + GM-CSF
Humanized
Naked antibody
Phase I (NCT01418495)
(15)
hu3F8 + IL-2
Humanized
Naked antibody
Phase I (NCT01662804)
(15)
3F8 + allogenic NK cells
Murine
Naked antibody
Phase I (NCT00877110)
(15)
3F8 + heat modified 3F8 + GM-CSF
Murine
Naked antibody
Phase I (NCT00450307)
(31)
3F8 + beta-glucan
Murine
Naked antibody
Phase I (NCT00492167)
(32)
3F8 + GM-CSF
Murine
Naked antibody
Phase II (NCT00072358)
(20)
3F8
Murine
Naked antibody
Phase II (NCT00002458)
(16)
3F8 + GM-CSF + isotretinoin
Murine
Naked antibody
Phase II (NCT01183897)
(33)
3F8 + GM-CSF + beta-glucan +
isotretinoin
Murine
Naked antibody
Phase II (NCT00089258)
(15)
ch14.18 + GM-CSF
Chimeric
Naked antibody
Phase I (NCT01418495)
(34)
ch14.18/CHO + isotretinoin + IL-2
Chimeric
Naked antibody
Phase I/II
(NCT01701479)
(15)
ch14.18 + lenalidomide + isotretinoin
Chimeric
Naked antibody
Phase I (NCT01711554)
(15)
ch14.18 + GM-CSF + IL-2
Chimeric
Naked antibody
Phase III
(NCT00026312)
(15)
ch14.18/CHO + isotretinoin + IL-2 +
G-CSF
Chimeric
Naked antibody
Phase III
(NCT01704716)
(26)
8B6
Chimeric
Naked antibody, anti-O-
acetyl-GD2
Pre-clinical
(35)
MORAb028
Human
Naked antibody, human IgM
Phase I (NCT01123304)
(15)
131I-3F8 + GM-CSF + bevacizumab
Murine
Radiolabeled antibody
Phase I (NCT00450827)
(15)
131I-3F8 and 124 I-3F8
Murine
Radiolabeled antibody
Phase II (NCT00445965)
(15)
hu3F8 multistep targeting
Murine
Radiolabeled antibody
Pre-clinical
(15)
hu3F8 multistep targeting
Murine
Radiolabeled antibody
Pre-clinical
(15)
hu3F8xhuOKT3 BsAb
Murine
Antibody for pre-targeting
Pre-clinical
(15)
iC9-GD2 CAR transduced T cells
Murine
Chimeric antigen receptor
Phase I (NCT01822652)
(15)
14G2a-CAR in EBV-CTL
Murine
Chimeric antigen receptor
Phase I (NCT00085930)
(15)
hu3F8-CAR
Humanized
Chimeric antigen receptor
Pre-clinical
(15)
iC9-GD2 CAR transduced VZV
specific T cell + VZV vaccine
Murine
Chimeric antigen receptor
Phase I (NCT01953900)
(15)
This article is protected by copyright. All rights reserved 23
Table 2. Predictive biomarkers in NB
This article is protected by copyright. All rights reserved 24
This article is protected by copyright. All rights reserved 25
This article is protected by copyright. All rights reserved 26
Table 3. Circulating biomarkers in NB
This article is protected by copyright. All rights reserved 27
1 erugiF
This article is protected by copyright. All rights reserved 28
2 erugiF
... angiogenesis, growth, and differentiation [60]. Hence, different miRNAs could be employed as diagnostic, prognostic, and therapeutic biomarkers through their expression levels for several malignancies, for example, melanoma [61,62]. There is evidence that many miRNAs like miR-148, miR-155, miR-182, miR-200c, miR-211, miR-214, miR-221, and miR-222 could influence the origination and growth of melanoma by targeting different melanoma-related genes (i.e. ...
Article
Full-text available
Melanoma is an aggressive tumor that exhibits an increased occurrence and poor prognosis if metastasized. Early diagnosis and effective management are mainstays for melanoma management. Effective management includes selection of effective treatment plan, supportive care and training of patients for early diagnosis. Various diagnostic options are available i.e. biopsy assessment, imaging procedures and different biomarkers like liquid biopsy, various proteins and polymorphism. As for the biomarkers, assessment of circulating tumor cells, microRNAs (miRNAs) and cell-free DNAs and RNAs are effectively used in the diagnosis of melanoma cases; the dysfunction of these molecules give rise to pathogenesis of melanoma. Many treatment modalities could be used potentially for melanoma management i.e. radiation therapy, surgery, systemic therapy, immunotherapy and antibodies therapy. Proper care and training to melanoma patients contributes in enhanced monitoring of the cases, in response to different treatment modalities. In this review, we have summarized details keeping in view malignant melanoma: its epidemiology, classification, diagnosis and various treatment modalities.
... Normal and malignant tissues have signi cantly different miRNA expression pro les, and particular clinicobiological characteristics are linked to particular miRNA signatures [4]. It has also been proved that dysregulation of miRNAs can affect the initiation and progression of several malignancies, including breast cancer [6][7][8][9]. According to research, miR-99a expression in breast cancer tissues is reduced in comparison to adjacent normal tissues, and breast cancer progression is associated with miR-99a downregulation. ...
Preprint
Full-text available
Introduction: Breast cancer (BC) is the most common cancer type and the fifth leading cause of cancer-related death. The primary goals of BC treatment are to remove the tumor and prevent metastasis. Despite advances in BC treatment, more effective therapies are required. miRNAs can regulate many targets involved in biological processes and tumor progression; these molecules have emerged as a promising cancer treatment strategy. In the present study, we investigated the effects of miR-99a and miR-143 in based single expression plasmids for BC inhibition. Methods: In this study, the precursor structure of miRNAs in the expression vector pEGFP-N1 entered single and double states, and MCF7 and T47D cells were transfected. The miRNAs expression level after transfection was then measured using qPCR. The MultiMiR package was used to obtain predicted and validated miRNA targets. MTT assay, qRT-PCR, migration test, and flow cytometry were used to assess the effect of miRNA and gene modulation. Results: The qPCR results revealed that miRNA constructs were significantly expressed after the transfection of both cell lines. The biological function of miRNAs showed that upregulation of miR-99a and miR-143 in any of the two selected BC cells inhibited their proliferation and migration rate, significantly inducing apoptosis (p<0.01). Also, miR-99a/-143 co-treatment has a synergistic anticancer effect in cancer cells via Akt1 and CDK6 targeting. Conclusion: These findings suggest that miR-99a/-143 plays synergistic regulatory roles in BC, possibly via a shared signaling pathway, providing a therapeutic strategy for BC treatment.
... However, the molecular mechanisms underlying TAM transformation in the complex TME are not yet fully understood. miRNAs are principal regulators of gene expression that function through the post-transcriptional regulation of target genes in several immune cells, including macrophages (44,45). A variety of miRNAs have been confirmed to be involved in macrophage polarization. ...
Article
Full-text available
Macrophages are principal immune cells with a high plasticity in the human body that can differentiate under different conditions in the tumor microenvironment to adopt two polarized phenotypes with opposite functions. Therefore, converting macrophages from the immunosuppressive phenotype (M2) to the inflammatory phenotype (M1) is considered a promising therapeutic strategy for cancer. However, the molecular mechanisms underlying this conversion process have not yet been completely elucidated. In recent years, microRNAs (miRNAs or miRs) have been shown to play key roles in regulating macrophage polarization through their ability to modulate gene expression. In the present study, it was found that miR‑382 expression was significantly downregulated in tumor‑associated macrophages (TAMs) and M2‑polarized macrophages in breast cancer. In vitro, macrophage polarization toward the M2 phenotype and M2‑type cytokine release were inhibited by transfection with miR‑382‑overexpressing lentivirus. Similarly, the overexpression of miR‑382 inhibited the ability of TAMs to promote the malignant behaviors of breast cancer cells. In addition, peroxisome proliferator‑activated receptor γ coactivator‑1α (PGC‑1α) was identified as the downstream target of miR‑382 and it was found that PGC‑1α affected macrophage polarization by altering the metabolic status. The ectopic expression of PGC‑1α restored the phenotype and cytokine secretion of miR‑382‑overexpressing macrophages. Furthermore, PGC‑1α expression reversed the miR‑382‑induced changes in the metabolic state of TAMs and the effects of TAMs on breast cancer cells. Of note, the in vivo growth and metastasis of 4T1 cells were inhibited by miR‑382‑overexpressing TAMs. Taken together, the results of the present study suggest that miR‑382 may alter the metabolic status of macrophages by targeting PGC‑1α, thereby decreasing the proportion of TAMs with the M2 phenotype, and inhibiting the progression and metastasis of breast cancer.
... binding to mRNAs and thereby promoting degradation of the target mRNA or blocking its translation into protein (19,(68)(69)(70)(71)(72)(73)(74)(75)(76)(77)(78)(79). MiRNA biogenesis is initiated from the nucleus similar to other RNAs (80)(81)(82)(83). These non-coding RNAs are primarily synthesized as primary transcripts or pri-miRNAs by RNA polymerase II (RNA pol II), which are processed by the RNAse III, Drosha into pre-miRNAs with long hairpin precursors of ∼70-100 nucleotides. ...
Article
Full-text available
Oral cancer remains a major public concern with considerable socioeconomic impact in the world. Despite substantial advancements have been made in treating oral cancer, the five-year survival rate for oral cancer remained undesirable, and the molecular mechanisms underlying OSCC carcinogenesis have not been fully understood. Noncoding RNAs (ncRNAs) include transfer RNAs (tRNAs), as well as small RNAs such as microRNAs, and the long ncRNAs such as HOTAIR are a large segment of the transcriptome that do not have apparent protein-coding roles, but they have been verified to play important roles in diverse biological processes, including cancer cell development. Cell death, such as apoptosis, necrosis, and autophagy, plays a vital role in the progression of cancer. A better understanding of the regulatory relationships between ncRNAs and these various types of cancer cell death is therefore urgently required. The occurrence and development of oral cancer can be controlled by increasing or decreasing the expression of ncRNAs, a method which confers broad prospects for oral cancer treatment. Therefore, it is urgent for us to understand the influence of ncRNAs on the development of different modes of oral tumor death, and to evaluate whether ncRNAs have the potential to be used as biological targets for inducing cell death and recurrence of chemotherapy. The purpose of this review is to describe the impact of ncRNAs on cell apoptosis and autophagy in oral cancer in order to explore potential targets for oral cancer therapy.
Article
Breast cancer (BC) is the most common cancer type and the fifth leading cause of cancer‐related deaths. The primary goals of BC treatment are to remove the tumor and prevent metastasis. Despite advances in BC treatment, more effective therapies are required. miRNAs can regulate many targets involved in biological processes and tumor progression; these molecules have emerged as a promising cancer treatment strategy. In the present study, we investigated the effects of miR‐99a and miR‐143 in single expression plasmids for BC inhibition. In this study, the precursor structure of miRNAs in the expression vector pEGFP‐N1 entered single and double states, and MCF7 and T47D cells were transfected. The miRNAs expression level after transfection was then measured using qPCR. The MultiMiR package was used to obtain predicted and validated miRNA targets. MTT assay, qRT‐PCR, migration test, and flow cytometry were used to assess the effect of miRNA and gene modulation. The qPCR results revealed that miRNA constructs were significantly expressed after the transfection of both cell lines. The biological function of miRNAs showed that upregulation of miR‐99a and miR‐143 in any of the two selected BC cells inhibited their proliferation and migration rate, significantly inducing apoptosis ( p < 0.01). Also, miR‐99a/miR‐143 co‐treatment has a synergistic anticancer effect in cancer cells via Akt1 and CDK6 targeting. These findings suggest that miR‐99a/miR‐143 plays synergistic regulatory roles in BC, possibly via a shared signaling pathway, providing a therapeutic strategy for BC treatment.
Article
Background Circulating miRNAs have been implicated in various aspects of diabetic wound healing, including inflammation, angiogenesis, and extracellular matrix remodeling. Thus, in alternative herbal medicine strategies, miRNAs will be potential therapeutic molecular targets in nonhealing wounds. These could be valuable elements for understanding the molecular basis of diabetic wound healing and could be used as good elements in bioinformatics. Objectives To elucidate the molecular mechanisms of microRNAs in association with apoptosis-inducing genes in controlling skin wound healing in diabetic wounds treated with green tea polyphenols (GTPs). Methods Green tea hydro extract (GTE) at doses of100–200 mg/ml was topically applied to the skin tissues of rats with T1DM induced by a single dose of streptozotocin (STZ; 100 mg/kg, in 0.01 M sodium citrate, pH 4.3–4.5) injected intraperitoneally for seven consecutive days to induce T1DM. The rats were treated with green tea for three weeks. A sterile surgical blade was used to inflict a circular wound approximately 2 cm in diameter on the anterior-dorsal side of previously anesthetized rats by a combination of ketamine hydrochloride (50 mg/kg, i.e., body weight) and xylazine hydrochloride. Afterward, the molecular roles of the circulating miRNAs miR-21, miR-23a, miR-146a, and miR-29b and apoptotic genes were determined by quantitative real-time PCR to evaluate Bax, Caspase-3, and Bcl-2 in wound healing. In addition, HPLC analysis was also performed to estimate the active polyphenols (GTPs) present in the hydro extract of green tea leaves. Results Wound healing was improved in diabetic skin wounds following treatment with GTE at doses of 100–200 mg/dl for three weeks. The wound parameters contraction, epithelialization, and scar formation significantly improved in a short time (14 days) compared to the longer periods identified in diabetic non-treated rats (20 days) and the standard control (15.5 days). Molecular analyses reported a significant increase in the levels of miR-21, miR-23a, and miR-146a and a decrease in the levels of miR-29b in green tea-treated diabetic rats compared to those in the standard control and STZ-diabetic non-treated rats. In addition, the molecular apoptotic genes Bax and caspase-3 significantly increased, and the BcL-2 gene significantly decreased following treatment with green tea polyphenols. Conclusions The data showed that active green tea polyphenols (GTPs) present in GTE significantly improved diabetic wound healing by controlling apoptotic genes and the circulating microRNAs miR-21, miR-23a, miR-146a, and miR-29b, which might be involved in cellular apoptosis and angiogenesis processes. Thus, to establish a future model for the treatment of diabetic wounds, further studies are needed to understand the potential association of these biological parameters with the wound-healing process in diabetic wounds.
Article
Full-text available
Cancer is caused by defects in coding and non-coding RNAs. In addition, duplicated biological pathways diminish the efficacy of mono target cancer drugs. MicroRNAs (miRNAs) are short, endogenous, non-coding RNAs that regulate many target genes and play a crucial role in physiological processes such as cell division, differentiation, cell cycle, proliferation, and apoptosis, which are frequently disrupted in diseases such as cancer. MiR-766, one of the most adaptable and highly conserved microRNAs, is notably overexpressed in several diseases, including malignant tumors. Variations in miR-766 expression are linked to various pathological and physiological processes. Additionally, miR- 766 promotes therapeutic resistance pathways in various types of tumors. Here, we present and discuss evidence implicating miR-766 in the development of cancer and treatment resistance. In addition, we discuss the potential applications of miR-766 as a therapeutic cancer target, diagnostic biomarker, and prognostic indicator. This may shed light on the development of novel therapeutic strategies for cancer therapy.
Article
Prostate cancer (PCa) is known as one of the most prevalent malignancies globally and is not yet curable owing to its progressive nature. It has been well documented that Genetic and epigenetic alterations maintain mandatory roles in PCa development. Apoptosis, a form of programmed cell death, has been shown to be involved in a number of physiological processes. Apoptosis disruption is considered as one of the main mechanism involved in lots of pathological conditions, especially malignancy. There is ample of evidence in support of the fact that microRNAs (miRNAs) have crucial roles in several cellular biological processes, including apoptosis. Escaping from apoptosis is a common event in malignancy progression. Emerging evidence revealed miRNAs capabilities to act as apoptotic or anti-apoptotic factors by altering the expression levels of tumor inhibitor or oncogene genes. In the present narrative review, we described in detail how apoptosis dysfunction could be involved in PCa processes and additionally, the mechanisms behind miRNAs affect the apoptosis pathways in PCa. Identifying the mechanisms behind the effects of miRNAs and their targets on apoptosis can provide scientists new targets for PCa treatment.
Article
Full-text available
Incidence of Malignant Melanoma has become the 5th in the UK. To date, the major anticancer therapeutics include cell therapy, immunotherapy, gene therapy and nanotechnology‐based strategies. Recently, extracellular vesicles, especially exosomes, have been highlighted for their therapeutic benefits in numerous chronic diseases. Exosomes display multifunctional properties, including inhibition of cancer cell proliferation and initiation of apoptosis. In the present in vitro study, the antitumour effect of cord blood stem cell (CBSC)‐derived exosomes was confirmed by the CCK‐8 assay (p < 0.05) on CHL‐1 melanoma cells and improve the repair mechanism on lymphocytes from melanoma patients. Importantly, no significant effect was observed in healthy lymphocytes when treated with the exosome concentrations at 24, 48 and 72 h. Comet assay results (OTM and %Tail DNA) demonstrated that the optimal exosome concentration showed a significant impact (p < 0.05) in lymphocytes from melanoma patients whilst causing no significant DNA damage in lymphocytes of healthy volunteers was 300 μg/ml. Similarly, the Comet assay results depicted significant DNA damage in a melanoma cell line (CHL‐1 cells) treated with CBSC‐derived exosomes, both the cytotoxicity of CHL‐1 cells treated with CBSC‐derived exosomes exhibited a significant time‐dependent decrease in cell survival. Sequencing analysis of CBSC exosomes showed the presence of the let‐7 family of miRNAs, including let‐7a‐5p, let‐7b‐5p, let‐7c‐5p, let‐7d‐3p, let‐7d‐5p and two novel miRNAs. The potency of CBSC exosomes in inhibiting cancer progression in lymphocytes from melanoma patients and CHL‐1 cells whilst causing no harm to the healthy lymphocytes makes it a potential candidate as an anticancer therapy.
Article
Full-text available
The most important biological function of exosomes is their possible use as biomarkers in clinical diagnosis. Compared with biomarkers identified in conventional specimens such as serum or urine, exosomal biomarkers provide the highest amount of sensitivity and specificity, which can be attributed to their excellent stability. Exosomes, which harbor different types of proteins, nucleic acids and lipids, are present in almost all bodily fluids. The molecular constituents of exosomes, especially exosomal proteins and microRNAs (miRNAs), are promising as biomarkers in clinical diagnosis. This discovery that exosomes also contain messenger RNAs and miRNAs shows that they could be carriers of genetic information. Although the majority of RNAs found in exosomes are degraded RNA fragments with a length of <200 nucleotides, some full-length RNAs might be present that may affect protein production in the recipient cell. In addition, exosomal miRNAs have been found to be associated with certain diseases. Several studies have pointed out miRNA contents of circulating exosomes that are similar to those of originating cancer cells. In this review, the recent advances in circulating exosomal miRNAs as biomarkers in gastrointestinal cancers are discussed. These studies indicated that miRNAs can be detected in exosomes isolated from body fluids such as saliva, which suggests potential advantages of using exosomal miRNAs as noninvasive novel biomarkers.Cancer Gene Therapy advance online publication, 16 December 2016; doi:10.1038/cgt.2016.77.
Article
Full-text available
Glioblastoma (GBM) is known as a tumor type, which arises from astrocytes. Several studies indicated that GBM tumor cells are malignant. This is because of the fact that they consist of different cell types, which are reproducing very quickly and are also supported by a large network of blood vessels. The correct identification of various stages of GBM could help to better treat the patients with this disease. Therefore, new biomarkers such as exosomes and microRNAs (miRNAs) may help us to learn more about GBM and they may also lead to a more effective treatment for patients with GBM. Exosomes have emerged as biological vehicles, which can perform various tasks in carcinogenesis pathways such as PI3K/AKT, SOX2, PTEN, ERK, and STAT3. The miRNAs are known as small noncoding RNAs that are involved in several GBM pathogenic events. These molecules have key roles in various biological processes such as angiogenesis, metastasis and tumor growth. In this study, we highlighted various exosomes and miRNAs that could be used for diagnosis and/or prognosis biomarkers in patients with GBM.Cancer Gene Therapy advance online publication, 11 November 2016; doi:10.1038/cgt.2016.48.
Article
Full-text available
MYCN amplification is the most common genetic alteration in neuroblastoma and plays a critical role in neuroblastoma tumorigenesis. MYCN regulates neuroblastoma cell differentiation, which is one of the mechanisms underlying its oncogenic function. We recently identified a group of differentiation-inducing microRNAs. Given the demonstrated inter-regulation between MYCN and microRNAs, we speculated that MYCN and the differentiation-inducing microRNAs might form an interaction network to control the differentiation of neuroblastoma cells. In this study, we found that eight of the thirteen differentiation-inducing microRNAs, miR-506-3p, miR-124-3p, miR-449a, miR-34a-5p, miR-449b-5p, miR-103a-3p, miR-2110 and miR-34b-5p, inhibit N-Myc expression by either directly targeting the MYCN 3'UTR or through indirect regulations. Further investigation showed that both MYCN-dependent and MYCN-independent pathways play roles in mediating the differentiation-inducing function of miR-506-3p and miR-449a, two microRNAs that dramatically down-regulate MYCN expression. On the other hand, we found that N-Myc inhibits the expression of multiple differentiation-inducing microRNAs, suggesting that these miRNAs play a role in mediating the function of MYCN. In examining the published dataset collected from clinical neuroblastoma specimens, we found that expressions of two miRNAs, miR-137 and miR-2110, were significantly anti-correlated with MYCN mRNA levels, suggesting their interactions with MYCN play a clinically-relevant role in maintaining the MYCN and miRNA expression levels in neuroblastoma. Our findings altogether suggest that MYCN and differentiation-inducing miRNAs form an interaction network that play an important role in neuroblastoma tumorigenesis through regulating cell differentiation.
Article
9502 Background: Despite high-dose chemotherapy, NB in BM often persists and forebodes death. Methods: 3F8/GM- CSF was used in 63 patients (pts) with NB in BM by morphology and/or metaiodobenzylguanidine (MIBG) scan, no prior progressive disease or immunotherapy, and no soft tissue NB; 35 (56%) of the pts had received second-line therapy and 24 (38%) were post-transplant. Treatment was repeated if human anti-mouse antibody (HAMA) titer was low. Results: Of 30 pts with NB in BM but normal MIBG scans (Group 1), 25 (83%) had complete response (CR) after cycle 1 (n=17), cycle 2 (n=5), or cycle 3, 6, or 7 (one pt each), including 13/16 post-transplant pts. Among 15 pts with NB in BM and abnormal MIBG scans (Group 2), 12 (80%) had CR in BM after cycle 1 (n=5), cycle 2 (n=4), cycle 4 (n=2), or cycle 9 (n=1); MIBG scans normalized in 5/11 pts who had multiple abnormal MIBG(+) sites and in 4/4 pts who had one abnormal MIBG(+) site (irradiated in three pts). Of 18 pts who had abnormal MIBG scans but no NB seen in BM tests (Group 3), 14 (78%) had CR or near CR, including eight whose MIBG(+) sites were irradiated. Early HAMA limited treatment in 19 pts, but was prevented by high-dose cyclophosphamide. CR continues in 12 pts (five never transplanted) with long follow-up (20+ -to- 146+ months) and in 10 pts with short follow-up. The only common toxicities of this outpatient treatment were pain and hives; there were no long-term toxicities. Conclusions: 3F8/GM-CSF is well tolerated, achieves a high CR rate against primary refractory NB in BM (including post-transplant), and may prolong disease control in non- transplanted pts. Further experience will show whether it ought to be used for consolidative therapy in place of myeloablative cytoreduction. No significant financial relationships to disclose.
Article
Plasma carcinoembryonic antigen (CEA) was assayed with a radioimmune procedure in 27 healthy control children. The upper limit of plasma CEA (mean+2 sd) was derived from healthy controls and was 3.35 ng/ml.This value was compared with those obtained from 15 children with active neuroblastoma, 7 with active embryonal rhabdomyosarcoma, 16 with treated neuroblastoma and without evidence of disease, 14 disease-free patients with embryonal rhabdomyosarcoma, and 17 patients still on therapy. The neuroblastoma and embryonal rhabdomyosarcoma patients with active disease had higher CEA values than did the successfully treated neuroblastoma and embryonal rhabdomyosarcoma patients. CEA plasma values greater than 3.35 ng/ml were found in 35% and 24% of patients with neuroblastoma and embryonal rhabdomyosarcoma, respectively.
Article
Purpose: MYCN is one of the most well-characterized genetic markers of neuroblastoma (NB). However, the mechanisms as to how MYCN mediate NB tumorigenesis is not fully clear. Increasing evidence has confirmed that the dysregulation of miRNAs is involved in MYCN-mediated NB tumorigenesis, supporting their potential as therapeutic targets for NB. Though miR-221 has been reported as one of the upregulated miRNAs, the interplay between miR-221 and MYCN-mediated NB progression remains largely elusive. Experimental design: The expression of miR-221 in the formalin-fixed, paraffin-embedded tissues from 31 confirmed NB patients was detected by locked nucleic acid-in situ hybridization and qRT-PCR. The correlation between miR-221 expression and clinical features in NB patients were assessed. The mechanisms as to how miR-221 regulate MYCN in NB were addressed. The effect of miR-221 on cellular proliferation in NB were determined both in vitro and in vivo. Results: miR-221 was significantly upregulated in NB tumor cells and tissues that overexpress MYCN, and high expression of miR-221 was positively associated with poor survival in NB patients. Nemo-like kinase (NLK) as a direct target of miR-221 in NB were verified. In addition, overexpression of miR-221 decreased LEF1 phosphorylation, but increased the expression of MYCN via targeting of NLK and further regulated cell cycle, particularly in S-phase, promoting the growth of NB cells. Conclusions: This study provides a novel insight for miR-221 in the control of NB cell proliferation and tumorigenesis, suggesting potentials of miR-221 as a prognosis marker and therapeutic target for patients with MYCN overexpressing NB.
Article
Exosomes have emerged as one of the main players in intercellular communication. These small nano-sized particles have many roles in various physiological pathways in normal and abnormal cells. Exosomes can carry various cargos such as proteins, mRNAs and miRNAs to recipient cells. Uptake of exosomes and their cargo can induce and/or inhibit different cellular and molecular pathways that lead to the alteration of cell behavior. Multiple lines of evidence have indicated that exosomes released from cancer cells can effect development of cancer in different stages. These particles and their cargo could regulate different processes such as tumor growth, metastasis, drug resistance, angiogenesis and immune system functioning. It has been observed that exosomes can be used as potential diagnostic biomarkers in various cancer types. Moreover, some studies have used these particles as biological vehicles for delivery of various drugs such as doxorubicin, siRNAs and miRNAs. Here, we summarized the findings on the role of exosomes in different pathological processes involved in cancer. Moreover, application of these particles as diagnostic and therapeutic biomarkers in different types of cancers is discussed.
Article
Neuroblastoma (NB) is known as a pediatric neoplasm that is associated with variable histopathological features. The use of biomarkers contributes to the monitoring and treatment of various malignancies such as NB. The identification of novel biomarkers such as (epi)genetic biomarkers and microRNAs (miRNAs) in NB has led to better treatments of NB. Among them, miRNAs have emerged as powerful tools in diagnosis, prognosis and therapeutic biomarkers for patients with NB. Circulating biomarkers such as circulating miRNAs present in body fluids (for example, plasma, serum and urine) provided an interesting field of study in NB treatment. The miRNAs have central roles in different pathogenic events in various malignancies such as NB. Hence, these molecules can be a suitable candidate for monitoring and treating NB patients. Here, we summarize some miRNAs as potential prognosis, diagnosis and therapeutic biomarkers in NB.
Article
Chronic lymphocytic leukemia (CLL) is known as the most common lymphoid malignancy in the Western world. MicroRNAs (miRNAs) are a class of small noncoding RNAs with pivotal roles in cellular and molecular processes related to different malignancies including CLL. Recently, some studies have shown that miR-192 plays a key role in CLL pathogenesis through increasing CDKN1A/p21 levels, suppression of Bcl-2 and enhancement of wild-type P53 and cell cycle arrest. Forty samples, including 20 patients with CLL, diagnosed in Omid hospital (Isfahan, Iran) and 20 healthy controls were sampled during a period of 4 months. Using real-time PCR method, expression of miR-192 was analyzed in peripheral blood mononuclear cells (PBMCs) of CLL patients in comparison with healthy subjects. In silico molecular signaling pathway enrichment analysis was also performed on validated and predicted targets (targetome) of miR-192 in DAVID database to explore possible role of miR-192 in some pathways. The expression of miR-192 was found to be significantly reduced (~2.5-folds) in CLL patients compared with healthy subjects (P=0.002). In silico molecular signaling pathway enrichment analysis detected cell indicated signaling pathway as one of the most statistically relevant pathway with miR-192 targetome. Our findings showed that miR-192 could be a biomarker for early diagnosis of CLL.Cancer Gene Therapy advance online publication, 23 September 2016; doi:10.1038/cgt.2016.34.