Abstract
Medulloblastoma (MB) is a cancer of the cerebellum and the most common primary pediatric malignancy of the central nervous system. Classified as a primitive neural ectoderm tumor; it is thought to arise from granule cell precursors in the cerebellum. The standard of care consists of surgery, chemotherapy and age-dependent radiation therapy. Despite aggressive multimodality therapy; approximately 30% of MB patients remain incurable. Moreover, for long-term survivors, the treatment related sequelae are often debilitating. Side effects include cerebellar mutism, sterility, neurocognitive deficits, and a substantial risk of developing secondary cancers. In a quest for more effective and targeted therapies, scientists have begun to investigate the biological events that not only initiate but also sustain the malignant phenotype in MB. Of particular interest is, the role of the tumor microenvironment in tumor pathogenesis. This review seeks to highlight several key processes observed in cancer biology, particularly the involvement of the tumor microenvironment, with relevant examples from MB.
Keywords: brain, medulloblastoma, microenvironment, review, therapy
BIOLOGY
TUMORIGENESIS
Tumorigenesis refers to the steps involved in the initiation and formation of a tumor. It is commonly accepted that cancer is a multistep process, yet the steps resulting in tumorigenesis may vary from one tumor to another. Ultimately, these events culminate in the inability of the cells to maintain a balance between proliferation and cell death. Often this imbalance is achieved via activation of an oncogene or loss of a tumor suppressor gene.
ONCOGENE ACTIVATION
Proto-oncogenes frequently code for proteins involved in cellular proliferation and differentiation. Three common mechanisms of oncogene activation are: (A) chromosomal abnormalities, (B) viruses, and (C) mutations in the DNA sequence of these genes.
Chromosomal Abnormalities
Amplification of proto-oncogenes can result in an increased amount of protein within a cell, thereby converting a proto-oncogene into an oncogene. Examples observed in human cancers that have been shown to result in consequent protein overexpression include amplification of HER2, MYC, MYL1, MYCN, AKT2, and REL15 [1].
MYC and Medulloblastoma (MB)
Myc family oncogenes have been implicated in the development of several human tumors and are the most commonly detected amplifications in MB [2–4]. Perhaps, the most remarkable observation of the Myc proteins is their pleiotropic nature. Amplification and subsequent overexpression of Myc family proteins have been attributed to a host of cellular processes involved in tumorigenesis, including the ability to promote cellular proliferation, growth, and inhibition of cellular differentiation [5]. More recently, the role of Myc proteins has extended beyond tumor initiation to tumor progression and an invasive phenotype, with aberrant expression resulting in angiogenesis [6], increased invasiveness [7], and genomic instability [5, 8]. In a large study of pediatric, MB patients (n=292), both MYCC and MYCN amplification were independently and significantly associated with a poor prognosis [2].
Isochromosome 17q and MB
The loss of chromosome arm 17p and subsequent gain of 17q, resulting in isochromosome 17q is the most common chromosomal abnormality in MB, occurring in approximately 40 percent of cases [9–11]. Presence of isochromosome 17q serves as a negative prognosis indicator [12]. The mechanism by which this abnormality plays a role in tumorigenesis has not been clearly elucidated. Chromosome arm17p contains several tumor supressor genes, and thus it is postulated that deletion of this gene region may be the culprit. One of these genes is the tumor suppressor p53 [9, 12–14].
Other copy number abnormalities known to be observed in MB include losses on 6q, 8p, 9q, 10q, 11q, 20, X, and Y and gains on 1q, 2p, 4q, 6q, 9p, 13q, and 14q [15].
Viral oncogenesis
Viruses are yet another source of oncogene activation. While viruses in of themselves are relatively inert, viruses are capable of hijacking the host cellular machinery and inducing damage in the host organism. One of the earliest links between viruses and cancer was Peyton Rous’ discovery that a virus could induce tumor in chickens [16–18]. Howard Temin and Harry Rubin would later expound upon Rous’ discovery demonstrating that cultured cells upon infection with the Rous Sarcoma Virus were transformed into tumor cells [19, 20]. This transformation would be discovered to be the result of a viral version of the src protein, v-src, acting as an oncogene [21]. For those viruses that do not contain oncogenes, oncogenic activation may also occur via insertional mutagenesis of the viral genome adjacent to a proto-oncogene in which the proto-oncogene is placed under the control of the transcription promoter.
John Cunningham (JC) Virus and MB
JC virus is a neurotropic polyomavirus and the causative agent of progressive multi-focal leukoencephalopathy (PML) [22]. JC virus is thought to promote tumorigenesis via the inactivation of tumor suppressors (pRb, p107 and p53) and deregulation of signaling pathways, such as the Wnt signaling pathway by the viral early proteins, particularly T antigen [23–29]. Within the past 35 years a body of literature has developed surrounding the JC virus and MB [30, 31]. Inoculation of hamsters with JC virus, greatly increased their propensity to develop cerebellar tumors [32, 33]. In a transgenic model, using JC virus T antigen, mice developed cerebellar tumors mimicking MB [34]. Despite compelling data from animal model research, human detection of JC virus in MB is controversial [35, 36]. Studies have reported detection of JC virus in MB patients [37, 38]. However, a few groups have challenged this claim indicating that JC virus detection was minimal to absent in large cohorts of MB patients [35, 36].
Human cytomegalovirus (HCMV) and MB
HCMV is a member of the herpes virus family. It is estimated that approximately 50–80% of the population has been exposed to the virus [39]. In most individuals, this virus remains latent, with no clinical signs of infection. Animal model research has implicated HCMV in tumorigenesis, capable of promoting cellular differentiation, proliferation, migration, and angiogenesis while inducing inflammation yet employing immune evasion strategies [40, 41]. Studies have shown that transgenic mice expressing the HCMV protein, US28 in epithelial cells, resulted in hyperplastic intestinal epithelium and tumor development [42]. The HCMV protein, IE72 has been demonstrated to induce telomerase activity, an event associated with tumor transformation. HCMV DNA and protein have been detected in several tumors, including glioblastoma, prostate, breast, and colon cancer [43]. Recently, it was discovered that both primary MB and MB cell lines contain detectable amounts of HCMV DNA and protein [44].
Mutations in Proto-oncogenes
Mutations in the DNA sequence of a proto-oncogene resulting in a gain of function mutation, such as increased enzymatic activity, could result in deregulated proliferation and/or differentiation, thus increasing the propensity for cancer to develop. A classic example observed in human cancers is the activating BRAF mutation in which valine is substituted for glutamate at the 599th position. Under normal conditions, BRAF kinase is controlled via phosphorylation at neighboring threonine or serine residues (Thr598 and Ser601) [45]. The activated form of BRAF kinase is able to override this checkpoint and renders the enzyme constitutively active [45]. This results in the phosphorylation of downstream signaling events and ultimately abnormal growth [45]. Based on the literature, mutations in proto-oncogenes resulting in activating mutations, do not appear to be highly implicated in the pathogenesis of MB. However, in sporadic cases of MB, oncogenic mutations in the wingless (Wnt) family proteins, axin and beta-catenin have been reported [46].
TUMOR SUPPRESSOR LOSS
To ensure that these processes are regulated the body is equipped with genes whose functions counteract proto-oncogene function. These genes are referred to as tumor suppressor genes. The proteins encoded by tumor suppressor genes serve to repress the cell cycle, promote apoptosis, or aid in DNA repair. Many tumor suppressors carry out these functions by acting as transcription factors.
Unlike oncogenes, tumor suppressors are often recessive, loss-of-function mutations, following Knudson’s “two-hit hypothesis” of tumorigenesis, in which two mutant alleles are required to display a cancerous phenotype. A classic example of this phenomenon is observed in hereditary retinoblastoma, in which the child inherits one mutated copy of the pRb gene [47]. A second mutation or inactivation of the second functional copy, an event known as loss of heterozygosity most always results in the development of retinoblastoma in the child.
TP53 and MB
Not all tumor suppressors adhere to the “two hit hypothesis”; mutations in the p53 gene can result in a dominant negative/antimorphmutant have been observed in several human cancers. Tumor protein 53 (TP53) is a tumor suppressor gene, involved in cell cycle regulation. TP53 is perhaps one of the most notorious tumor suppressor genes. According to a report published in 2002 by the International Agency for Research on Cancer (IACR), TP53 is mutated in 30–50% of common human cancers. A majority of these mutations are missense, resulting in an amino acid substitution versus nonsense mutations. Li-Fraumeni syndrome, in which patients have a predisposition to develop a variety of tumors, including MB, more than half have mutations in TP53 [48–52].
PTCH1 and MB
Haplo-insufficiency is another mechanism by which tumor suppressors may be unable to maintain homeostasis, resulting in cancer formation. An example of this is the PTCH1mutation in MB [53]. PTCH1 is a member of the patched gene family. Its product, Patched (Ptch1) is a transmembrane receptor involved in the hedgehog signaling pathway. Members of the sonic hedgehog signaling pathway (SHH) function to regulate growth-inducing signal transduction for normal cerebellar development. In the absence of ligand, Ptch1 serves as an inhibitor of downstream signaling in the hedgehog pathway [54]. The association between MB and PTCH1 was uncovered through studying the biology of familial nevoid basal cell carcinoma, also known as Gorlin’s syndrome. Patients with Gorlin’s syndrome possess germ line inactivating mutations in the PTCH1 gene and are predisposed to developing a variety of tumors, including MB. Roughly 20% of sporadic, nonfamilial cases of MB have also been shown to contain PTCH1 or other SHH pathway activating mutations. In further studying individuals with these mutations, it was discovered that these patients also possess wildtype copies of the PTCH1 gene. In mice, knocking out PTCH1 results in an embryonic lethal phenotype. PTCH1 heterozygotes survive, yet are predisposed to developing cerebellar tumors, resembling MB, in further support of haploinsufficiency [51, 55, 56].
APC and MB
The Wingless (Wnt) signaling pathway consists of a network of proteins involved in embryogenesis via gene regulation and cell–cell control. Among the target genes of this pathway include the proto-oncogenes MYCC and MYCN. The tumor suppressor gene product, Adenomatous polyposis coli (Apc) is a member of the Wnt signaling pathway, whose function is to regulate activity of beta-catenin. Interestingly, patients with Turcot’s syndrome, a syndrome characterized by intestinal polyp formation and brain tumors, such as MB, possess a germ line mutation in the APC gene [57].
EPIGENETIC MODIFICATIONS
In recent years, there has been great emphasis placed on studying heritable information other than the underlying DNA sequence capable of altering gene expression or cellular phenotype, the epigenome. Evidence of epigenetic influence on tumorigenesis has been observed in several cancers. One mechanism that is gaining increasing credibility is promoter hypermethylation in tumor suppressor gene silencing. The tumor suppressor genes RASSF1A, HIC1, and CASP8 have been shown to exhibit increased methylation when compared to normal cerebellum [58].This methylation was associated with transcriptional silencing and expression was induced upon treatment with DNA methyl transferase inhibitors [58].
MAINTENANCE OF THE CANCEROUS PHENOTYPE
The aforementioned processes serve to describe key events in the initiation of tumor formation. In order to maintain a cancerous phenotype often other mechanisms must be employed. With continuous growth comes an increased nutritional demand. A key mechanism through which tumor development is achieved is via cell surface receptor signaling.
RECEPTOR TYROSINE KINASES AND MB
Among the most notable receptors in this class are receptor tyrosine kinases. Receptor tyrosine kinases serve as cell surface receptors for growth factors, cytokines, and hormones. Upon ligand binding receptor tyrosine kinases cluster, activating the protein’s cytoplasmic domains; autophosphorylation of tyrosine residues within the receptor form binding sites for Src homology 2 (SH2) and phosphotyrosine binding (PTB) domains. Phosphorylation of these proteins then results in the activation of downstream signaling pathways involved in diverse cellular responses, such as cell division, differentiation and motility. Members of this family, specifically neurotrophin-3 receptor (TrkC), human epidermal growth factor receptor 2 (HER2/erbb-2), platelet derived growth factor receptor (PDGFR), insulin-like growth factor receptor (IGFR), and nerve growth factor receptor (NGFR) have been associated with MB pathogenesis [59].
HER2
Of the erbb family members, HER2, is most highly expressed by MB and is associated with a poor prognosis. In a study of 81MB, 40% of tumors tested positive forHER2 [60]. Of these tumors, the large cell anaplastic variant, characterized by a more aggressive clinical course, expressed HER2 with the highest frequency [60]. In this study, HER2 overexpression was found to associate with a poor prognosis independent of histological variant [60].
In contrast to breast cancer where overexpression of the protein HER2 is associated with HER2 gene amplification; this same mechanism of overexpression has not been demonstrated in MB. In a study of 70 primary MB out of those who tested positive for HER2, expression amplification of the gene HER2 was not detected [61]. In addition to this finding, work from our group has demonstrated that when compared to HER2 expression in breast cancer, MB express HER2 at low levels [62]. Furthermore, HER2 is not expressed in the normal cerebellum [61]. Taken together, these observations implicate HER2 as a key player in MB pathology. However, further investigation needs to be conducted in an attempt to better understand the mechanism behind HER2 overexpression in MB.
Notch Signaling
Other signaling pathways that have associated with MB pathogenesis include the Notch and CXCR4 signaling pathways. Both of these pathways, like many others listed in this review, play an integral role in embryogenesis. The receptor Notch2 has been detected in proliferating progenitor cells in MB and is associated with a poor prognosis [59, 63].
CANCER STEM CELLS
An active area of research within the past decade has been to identify the cancer stem cell populations of various tumors. Cancer “stem cells” (cscs) or cancer initiating cells describe a population of cells capable of not only self-renewal but also producing progenitor cells capable of differentiating into more terminally differentiated cell types, such as endothelial cells. In order to identify such a population, several xenograft studies have been conducted in mice in which human tumors are serially transplanted into mice and the phenotype of the cells that are able to establish a tumor are assayed. Brain cells expressing the glycoprotein CD133 are thought to be enriched for cscs. Work from our lab has demonstrated that the CD133 positive population of MB cell lines also expresses higher levels of the receptor tyrosine kinase, HER2, which is associated with a poor prognosis in MB [62]. This finding could provide a link between stemness and the HER2 pathway in MB. Although there is still a considerable amount of work to be done in the field of cscs evidence of such a population may contribute to our understanding of epithelial-mesenchymal transition and tumor invasion and metastasis.
MICROENVIRONMENT
In 1889, English surgeon, Stephen Paget proposed that the ability of tumor cells, “the seed,” to metastasize to distant organs was dependent upon a compatible environment in those organs, “the soil.” [64, 65] This theory was supported by work conducted by Paget in which he analyzed the autopsy records of over 700 women with breast cancer [64]. From his analyses he was able to conclude that metastasis to visceral organs and bone was not a random occurrence [64]. Since then there has been a wealth of literature published in support of Paget’s initial observations indicating that the ability of cancer cells to flourish is dependent on the presence of a niche that favors its survival and progression. This niche most often referred to as the “tumor microenvironment” exists as a complex of several biological systems, consisting of stromal tissue (nerves, immune cells, fibroblasts, smooth muscle cells, and blood vessels), extracellular matrix and the cancerous cells [66–68].
Most cancers are not merely a collective body of homogenous cells running the same aberrant program. Instead most often cancers are heterogeneous in nature that includes in addition to the cancer cell a myriad of different cell types that collectively constitute the tumor microenvironment. Greater than 80% of malignancies are classified as carcinomas, having derived from epithelial origin. Consequently much of what we know about the tumor microenvironment is based on research on carcinomas. In order to conduct such research, it is crucial to understand the structure and function of normal epithelium in order to better understand the biology of cancerous epithelium.
THE TUMOR STROMA
The stroma or surrounding connective tissue of an organ serves as a support system for that organ; acting as a reservoir of cells, capable of differentiating upon insult or injury to that organ. In the case of cancer, homeostasis is lost and the tissue resembles a wound incapable of repairing, with fibrosis and chronic inflammation. A once highly structured tissue now displays a loss of membrane integrity, polarity, and contact inhibition. These changes result in an increased secretion and mixing of soluble factors and the line between the cancerous cells of the epithelium and the underlying stroma is blurred.
Cancer Associated Fibroblasts (CAF)
Fibroblasts are the most abundant cell type of the stromal compartment. Perhaps this is due to their critical role in maintaining the structural integrity of the connective tissues. Fibroblasts synthesize the components of the extracellular matrix, the formation of which is important in growth and wound healing [69]. Fibroblasts, similar to other cell types found in the stromal compartment are of mesenchymal origin. The expression of the intermediate filament vimentin serves as a marker for cells of mesenchymal origin. In cancer, fibroblasts often develop into myofibroblasts coexpressing vimentin and alpha smooth muscle actin. Cells that follow this pattern are commonly referred to as cancer-associated fibroblasts (cafs). Cafs or myofibroblasts, although helpful in wound repair under conditions of chronic inflammation, have proven deleterious resulting in aberrant fibrosis. The aptly named desmoplastic variant of MB is characterized by densely packed cells, with a collagen-rich stroma. The fibrotic nature of this variant implicates aberrant fibrosis as a causative agent in a subset of MB.
Transforming growth factor beta (TGF-β) secreted by platelets upon damage to the tissues, is a key mediator of tissue homeostasis. In cancer, TGF-beta is released not only by platelets, but is overexpressed also by both the cancer cells and surrounding stromal cells. This secretion in turn serves as a proliferative signal for both the cancer cells and its stromal compartment. TGF-β has also been shown to induce angiogenesis and dampen the immune response supporting amore invasive phenotype [70].
The chemokine interleukin-8 (IL-8) also referred to as CXCL8 functions similarly to TGF-beta to promote tumor progression. It is secreted by several cell types in the tumor microenvironment and promotes angiogenesis. Acting as a chemoattractant IL-8 is capable of recruiting cells expressing the receptors CXCR1 and CXCR2. Cells that express one or more of these receptors include neutrophils recruited to sites of inflammation.
Both TGF-beta and IL-8 have been shown to be overexpressed in primary MB and MB cell lines [59, 71].
THE TUMOR VASCULAR ENDOTHELIUM
Normal epithelium is avascular; dependent upon the vascular network of the underlying stroma for its nutrients. With an increasing demand for oxygen and nutrients by these rapidly dividing cells, angiogenesis is often a rate-limiting factor in sustaining tumor growth and promoting tumor progression. This process is carried out in large part by endothelial cells, which make up the inner linings of arteries, veins and capillaries. This cell type can be induced to generate new vessels from pre-existing vessels in a process known as angiogenesis.
The bulk of angiogenesis occurs during embryogenesis. In adults angiogenesis is likely to occur as a repair process, as observed in wound healing. In order for angiogenesis to occur pericytes need to be removed from the branching vessel [72], after which the basement membrane of the vessel must be degraded and restructured via matrix metalloproteinases (mmps) [72, 73]. Once the matrix has been remodeled, growth signals then act to recruit endothelial cells and induce endothelial cell proliferation [72]. These cells then form tube-like structures and depending on the size of the vessel recruit either pericytes or smooth muscle cells for added support [72].
Tumors, similar to normal tissues, require an adequate supply of oxygen and nutrients as well as a mechanism to export metabolic waste. This is often provided by expanding the network of blood vessels generated during angiogenesis. Despite many similarities between normal and tumor-associated vasculature, tumor-associated vasculature is often abnormal. In contrast to normal vasculature, tumor-associated vasculature frequently lacks pericytes and is dilated, containing large fenestrations and having a tortuous morphology [74]. In some instances, the vasculaturemay be an admixture of both endothelial cells and tumor cells [72].
Vascular endothelial growth factor (VEGF) has been shown to increase endothelial cell permeability to metabolites, while also stimulating plasminogen activators and interstitial collagenase [72, 75]. Collectively, VEGF acts as a regulator of proteolytic activity required for matrix remodeling and subsequent endothelial cell tube formation [72]. Production of VEGF is mediated by local oxygen concentration. Hypoxia induces VEGF expression via binding of the hypoxia-inducible factor 1a (HIF-1alpha) to the VEGF promoter [72,76]. The tumor-associated vasculature with its leaky nature and increasing demand for nutrients as it grows and expands induces a hypoxic environment. This in turn signals an increase in VEGF production thereby promoting angiogenesis.
Functional VEGF and VEGF receptors have been detected in primary MB and MB cell lines [77]. However, whether VEGF expression is correlated with a different prognosis in MB is still under investigation. In one study seven of 32 tumors expressed VEGF; there was no detectable difference in the 5-year survival rate on the basis of VEGF expression [78].
IMMUNE CELLS IN THE TUMOR MICROENVIRONMENT
The mammalian immune system is a complex network of specialized cell types and factors that serve to protect against disease. To do so the immune system must be able to identify self from non-self. Through this mechanism the immune system is able to target foreign invaders, while maintaining tolerance to self-antigen. The function of the immune system is not limited to fighting infection. Like other components found in the stromal compartment it also plays an essential role in maintaining tissue homeostasis.
In the case of cancer, aberrant signaling may result in chronic inflammation thereby inducing immunosuppression and promoting tumor growth. In cancer an increased innate immune response at the site of the tumor microenvironment is associated with angiogenesis and in many cases a poor prognosis [79].
Tumor Infiltrating Lymphocytes (TILs)
TILs have been reported in a variety of cancers [80–82]. However, whether these cells function primarily in an antitumor or protumor capacity remains controversial [80,82,83]. In this review, we focus on tumor-infiltrating T-lymphocytes. However, it would be remiss if we failed to mention, the “other” TILs [82], B cells (for more on B-TILs please refer to Nelson BH. J Immunol 2010).
Emerging data suggest that T cells and their impact on tumor growth may depend heavily upon the particular TIL subset. In the literature, it is the growing consensus that the majority of CD4 + T cells, with the exception of the Th1 subset favor tumor progression, while CD8 + T cells (cytotoxic T cells) favor tumor rejection [84]. However, there are several reports that counter this claim [85–87].
In 2003, Wakabayashi et al. reported that CD4+T cells in cancer stroma, not CD8+ T cells in cancer nests were associated with a favorable prognosis in human nonsmall cell lung cancers [85]. Also in 2003, Cho et al. reported that in patients with esophageal squamous carcinoma both the CD4+and the CD8+T cells work together to improve prognosis [86]. There have also been reports indicating that a high density of Tumor-Infiltrating FOXP3 T Regulatory cells were associated with improved survival in patients with colorectal cancer [87].
TILs in MB
Work from our lab has shown that T lymphocytes consistently migrate to and infiltrate primary MB [71]. Our experimental model revealed that MB cell lines interact with the tumor endothelium to recruit T cells to the MB microenvironment [71]. In particular, Macrophage Migration Inhibitory Factor (MIF) is the key chemokine molecule secreted by MB cells which induces the endothelial cells within the MB microenvironment to secrete the potent T lymphocyte attractant “Regulated upon Activation, Normal T-cell Expressed, and Secreted (RANTES).”This in turn creates a chemotactic gradient for RANTES-receptor bearing T lymphocytes [71]. Regarding the role of T lymphocytes in the MB microenvironment, in a clinical study comparing patients with MB to healthy controls, MB patients had a higher proportion of Th17 cells at the site of the tumor and in their peripheral blood [88]. In a recent report by Weigering et al., high expression of IFN-γ and tnfα in T cells in the peripheral blood of MB patients in the early post-transplant period correlated with a better prognosis [89].
CONCLUSION
Overall a substantial body of work focused on the biology of MB has been conducted. Results of this work have implicated disrupted pathways involved in embryogenesis and cellular proliferation and differentiation as key mediators of MB pathogenesis. Chromosomal abnormalities in the form of amplifications and deletions have been identified as a common culprit. Often these modifications result in the upregulation of growth factor signaling and inactivation of tumor suppressor genes to contribute to tumor formation. We also cannot forget the increasing evidence of the association of viral infection, particularly JC virus and HCMV, and MB. All together, with an emphasis in understanding not only the biology of cancer cells, but the surrounding tumor microenvironment it is likely that our understanding of MB pathogenesis will increase. The next logical step is to apply our increasing knowledge regarding MB pathogenesis towards the development of new therapeutic options. Our lab has previously validated HER2/erbb-2 as a MB (and glioblastoma) restricted target antigen, demonstrated in preclinical models the efficacy of targeting this antigen [62, 90] the product of which is currently being used in a clinical trial for patients with Glioblastoma (ClinicalTrials. gov identifier: NCT01109095). Perhaps small molecule inhibitors in conjunction with immunotherapy targeting viral and tumor co-associated antigens may prove advantageous in accessing the brain and targeting tumors like MB.
ACKNOWLEDGMENTS
The authors would like to thank Dr. Helen Heslop for her mentoring and guidance. This work was supported in part by the NIH Training grant, Training in Cell and Gene Therapy: T32HL092332. The authors would also like to thank Kevin Shahbahrami for helping to edit this manuscript.
Footnotes
Declaration of Interest
The authors hereby report no conflicts of interest (no competing financial interests) in the publication of this manuscript.
REFERENCES
- 1.Santarius T, Shipley J, Brewer D, Stratton MR, Cooper CS. A census of amplified and overexpressed human cancer genes. Nat Rev Cancer. 2010 Jan;10(1):59–64. doi: 10.1038/nrc2771. [DOI] [PubMed] [Google Scholar]
- 2.Ryan SL, Schwalbe EC, Cole M, et al. MYC family amplification and clinical risk-factors interact to predict an extremely poor prognosis in childhood medulloblastoma. Acta Neuropathol. 2012;123:501–513. doi: 10.1007/s00401-011-0923-y. [DOI] [PubMed] [Google Scholar]
- 3.Korshunov A, Remk M, Kool M, et al. Biological and clinical heterogeneity of MYCN-amplified medulloblastoma. Acta Neuropathol. 2012;123:515–527. doi: 10.1007/s00401-011-0918-8. [DOI] [PubMed] [Google Scholar]
- 4.Lindberg E, Persson A, Ora I, et al. Concurrent gain of 17q and the MYC oncogene in a medullomyoblastoma. Neuropathology. 2007 Dec;27(6):556–560. doi: 10.1111/j.1440-1789.2007.00810.x. [DOI] [PubMed] [Google Scholar]
- 5.Whitfield JR, Soucek L. Tumor microenvironment: becoming sick of Myc. Cell Mol Life Sci. 2012 Oct 28;69:931–934. doi: 10.1007/s00018-011-0860-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Breit S, Ashman K, Wilting J, et al. The N-myc oncogene in human neuroblastoma cells: down-regulation of an angiogenesis inhibitor identified as activin A. Cancer Res. 2000 Aug 15;60(16):4596–4601. [PubMed] [Google Scholar]
- 7.Noujaim D, van Golen CM, van Golen KL, et al. N-Myc and Bcl-2 coexpression induces MMP-2 secretion and activation in human neuroblastoma cells. Oncogene. 2002 Jul 4;21(29):4549–4557. doi: 10.1038/sj.onc.1205552. [DOI] [PubMed] [Google Scholar]
- 8.Oikawa M, Yoshiura K, Kondo H, et al. Significance of genomic instability in breast cancer in atomic bomb survivors: analysis of microarray-comparative genomic hybridization. Radiat Oncol. 2011;6:168. doi: 10.1186/1748-717X-6-168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Castellino RC, De Bortoli M, Lu X, et al. Medulloblastomas overexpress the p53-inactivating oncogene WIP1/PPM1D. J Neurooncol. 2008 Feb;86(3):245–256. doi: 10.1007/s11060-007-9470-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Fuller C, Fouladi M, Gajjar A, et al. Chromosome 17 abnormalities in pediatric neuroblastic tumor with abundant neuropil and true rosettes. Am J Clin Pathol. 2006 Aug;126(2):277–283. doi: 10.1309/TFBX-1LWQ-93MX-QBAW. [DOI] [PubMed] [Google Scholar]
- 11.Biegel JA, Burk CD, Barr FG, Emanuel BS. Evidence for a 17p tumor related locus distinct from p53 in pediatric primitive neuroectodermal tumors. Cancer Res. 1992 Jun 15;52(12):3391–3395. [PubMed] [Google Scholar]
- 12.Pan E, Pellarin M, Holmes E, et al. Isochromosome 17q is a negative prognostic factor in poor-risk childhood medulloblastoma patients. Clin Cancer Res. 2005 Jul 1;11(13):4733–4740. doi: 10.1158/1078-0432.CCR-04-0465. [DOI] [PubMed] [Google Scholar]
- 13.Nam DH, Wang KC, Kim YM, et al. The effect of isochromosome17q presence, proliferative and apoptotic indices, expression of c-erbB-2, bcl-2 and p53 proteins on the prognosis of medulloblastoma. J Korean Med Sci. 2000 Aug;15(4):452–456. doi: 10.3346/jkms.2000.15.4.452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Loda M, Giangaspero F, Badiali M, et al. p53 gene expression in medulloblastoma by quantitative polymerase chain reaction. DiagnMol Pathol. 1992 Mar;1(1):36–44. doi: 10.1097/00019606-199203000-00006. [DOI] [PubMed] [Google Scholar]
- 15.Gilbertson RJ. Medulloblastoma: signalling a change in treatment. Lancet Oncol. 2004 Apr;5(4):209–218. doi: 10.1016/S1470-2045(04)01424-X. [DOI] [PubMed] [Google Scholar]
- 16.Rous P. A Transmissible avian neoplasm. (Sarcoma of the common fowl.) J Exp Med. 1910 Sep 1;12(5):696–705. doi: 10.1084/jem.12.5.696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Rous P. A sarcoma of the fowl transmissible by an agent separable from the tumor cells. J Exp Med. 1911 Apr 1;13(4):397–411. doi: 10.1084/jem.13.4.397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Murphy JB, Rous P. The behavior of Chicken Sarcoma implanted in the developing embryo. J Exp Med. 1912 Feb 1;15(2):119–132. doi: 10.1084/jem.15.2.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Rubin H, Temin HM. Infection with the Rous sarcoma virus in vitro. Fed Proc. 1958 Dec;17(4):994–1003. [PubMed] [Google Scholar]
- 20.Temin HM, Rubin H. Characteristics of an assay for Rous sarcoma virus and Rous sarcoma cells in tissue culture. Virology. 1958 Dec;6(3):669–688. doi: 10.1016/0042-6822(58)90114-4. [DOI] [PubMed] [Google Scholar]
- 21.Tarpley WG, Temin HM. The location of v-src in a retrovirus vector determines whether the virus is toxic or transforming. Mol Cell Biol. 1984 Dec;4(12):2653–2660. doi: 10.1128/mcb.4.12.2653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Garcia De Viedma D, Diaz Infantes M, Miralles P, et al. JC virus load in progressive multifocal leukoencephalopathy: analysis of the correlation between the viral burden in cerebrospinal fluid, patient survival, and the volume of neurological lesions. Clin Infect Dis. 2002 Jun 15;34(12):1568–1575. doi: 10.1086/340535. [DOI] [PubMed] [Google Scholar]
- 23.Caracciolo V, Reiss K, Khalili K, et al. Role of the interaction between large T antigen and Rb family members in the oncogenicity of JC virus. Oncogene. 2006 Aug 28;25(38):5294–5301. doi: 10.1038/sj.onc.1209681. [DOI] [PubMed] [Google Scholar]
- 24.Felsani A, Mileo AM, Paggi MG. Retinoblastoma family proteins as key targets of the small DNA virus oncoproteins. Oncogene. 2006 Aug 28;25(38):5277–5285. doi: 10.1038/sj.onc.1209621. [DOI] [PubMed] [Google Scholar]
- 25.Bollag B, Prins C, Snyder EL, Frisque RJ. Purified JC virus T and T’ proteins differentially interact with the retinoblastoma family of tumor suppressor proteins. Virology. 2000 Aug 15;274(1):165–178. doi: 10.1006/viro.2000.0451. [DOI] [PubMed] [Google Scholar]
- 26.Howard CM, Claudio PP, De Luca A, et al. Inducible pRb2/p130 expression and growth-suppressive mechanisms: evidence of a pRb2/p130, p27Kip1, and cyclin E negative feedback regulatory loop. Cancer Res. 2000 May 15;60(10):2737–2744. [PubMed] [Google Scholar]
- 27.White MK, Khalili K. Interaction of retinoblastoma protein family members with large T-antigen of primate polyomaviruses. Oncogene. 2006 Aug 28;25(38):5286–5293. doi: 10.1038/sj.onc.1209618. [DOI] [PubMed] [Google Scholar]
- 28.Caracciolo V, Reiss K, Crozier-Fitzgerald C, et al. Interplay between the retinoblastoma related pRb2/p130 and E2F-4 and -5 in relation to JCV-TAg. J Cell Physiol. 2007 Jul;212(1):96–104. doi: 10.1002/jcp.21005. [DOI] [PubMed] [Google Scholar]
- 29.Del Valle L, Baehring J, Lorenzana C, et al. Expression of a human polyomavirus oncoprotein and tumour suppressor proteins in medulloblastomas. Mol Pathol. 2001 Oct;54(5):331–337. doi: 10.1136/mp.54.5.331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Takakura K, Inoya H, Nagashima K, et al. Viral neurooncogenesis. Prog Exp Tumor Res. 1987;30:10–20. [PubMed] [Google Scholar]
- 31.Caracciolo V, Macaluso M, D’Agostino L, et al. Cross-talk between T-Ag presence and pRb family and p53/p73 signaling in mouse and human medulloblastoma. J Cell Biochem. 2010 May;110(1):182–190. doi: 10.1002/jcb.22525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zu Rhein GM. Studies of JC virus-induced nervous system tumors in the Syrian hamster: a review. Prog Clin Biol Res. 1983;105:205–221. [PubMed] [Google Scholar]
- 33.Padgett BL, Walker DL, ZuRhein GM, Varakis JN. Differential neurooncogenicity of strains of JC virus, a human polyoma virus, in newborn Syrian hamsters. Cancer Res. 1977 Mar;37(3):718–720. [PubMed] [Google Scholar]
- 34.Krynska B, Otte J, Franks R, et al. Human ubiquitous JCV(CY) T-antigen gene induces brain tumors in experimental animals. Oncogene. 1999 Jan 7;18(1):39–46. doi: 10.1038/sj.onc.1202278. [DOI] [PubMed] [Google Scholar]
- 35.Munoz-Marmol AM, Mola G, Ruiz-Larroya T, et al. Rarity of JC virus DNA sequences and early proteins in human gliomas and medulloblastomas: the controversial role of JC virus in human neurooncogenesis. Neuropathol Appl Neurobiol. 2006 Apr;32(2):131–140. doi: 10.1111/j.1365-2990.2006.00711.x. [DOI] [PubMed] [Google Scholar]
- 36.Vasishta RK, Pasricha N, Nath A, Sehgal S. The absence of JC virus antigens in Indian children with medulloblastomas. Indian J Pathol Microbiol. 2009 Jan-Mar;52(1):42–45. doi: 10.4103/0377-4929.44961. [DOI] [PubMed] [Google Scholar]
- 37.Krynska B, Del Valle L, Croul S, et al. Detection of human neurotropic JC virus DNA sequence and expression of the viral oncogenic protein in pediatric medulloblastomas. Proc Natl Acad Sci U S A. 1999 Sep 28;96(20):11519–11524. doi: 10.1073/pnas.96.20.11519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Weggen S, Bayer TA, von Deimling A, et al. Low frequency of SV40, JC and BK polyomavirus sequences in human medulloblastomas, meningiomas and ependymomas. Brain Pathol. 2000 Jan;10(1):85–92. doi: 10.1111/j.1750-3639.2000.tb00245.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Pereira L, Maidji E, Fisher SJ, et al. HCMV persistence in the population: potential transplacental transmission. In: Arvin A, Campadelli-Fiume G, Mocarski E, et al., editors. Human Herpesviruses: Biology, Therapy, and Immunoprophylaxis. Cambridge: Cambridge University Press; 2007. [PubMed] [Google Scholar]
- 40.Shimamura M, Murphy-Ullrich JE, Britt WJ. Human cytomegalovirus induces TGF-beta1 activation in renal tubular epithelial cells after epithelial-to-mesenchymal transition. PLoS Pathog. 2010;6(11):e1001170. doi: 10.1371/journal.ppat.1001170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Cobbs CS, Soroceanu L, Denham S, et al. Human cytomegalovirus induces cellular tyrosine kinase signaling and promotes glioma cell invasiveness. J Neurooncol. 2007 Dec;85(3):271–280. doi: 10.1007/s11060-007-9423-2. [DOI] [PubMed] [Google Scholar]
- 42.Zhu Y, Huang L, Anders DG. Human cytomegalovirus oriLyt sequence requirements. J Virol. 1998 Jun;72(6):4989–4996. doi: 10.1128/jvi.72.6.4989-4996.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Straat K, Liu C, Rahbar A, et al. Activation of telomerase by human cytomegalovirus. J Natl Cancer Inst. 2009 Apr 1;101(7):488–497. doi: 10.1093/jnci/djp031. [DOI] [PubMed] [Google Scholar]
- 44.Baryawno N, Rahbar A, Wolmer-Solberg N, et al. Detection of human cytomegalovirus in medulloblastomas reveals a potential therapeutic target. J Clin Invest. 2011 Oct;121(10):4043–4055. doi: 10.1172/JCI57147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Karbowniczek M, Cash T, Cheung M, et al. Regulation of B-Raf kinase activity by tuberin and Rheb is mammalian target of rapamycin (mTOR)-independent. J Biol Chem. 2004 Jul 16;279(29):29930–29937. doi: 10.1074/jbc.M402591200. [DOI] [PubMed] [Google Scholar]
- 46.Koch A, Hrychyk A, Hartmann W, et al. Mutations of the Wnt antagonist AXIN2 (Conductin) result in TCF-dependent transcription in medulloblastomas. Int J Cancer. 2007 Jul 15;121(2):284–291. doi: 10.1002/ijc.22675. [DOI] [PubMed] [Google Scholar]
- 47.Rubenfeld M, Abramson DH, Ellsworth RM, Kitchin FD. Unilateral vs. bilateral retinoblastoma. Correlations between age at diagnosis and stage of ocular disease. Ophthalmology. 1986 Aug;93(8):1016–1019. doi: 10.1016/s0161-6420(86)33629-7. [DOI] [PubMed] [Google Scholar]
- 48.Rausch T, Jones DT, Zapatka M, et al. Genome Sequencing of Pediatric Medulloblastoma Links Catastrophic DNA Rearrangements with TP53Mutations. Cell. 2012 Jan 20;148(1–2):59–71. doi: 10.1016/j.cell.2011.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Pfaff E, RemkeM, Sturm D, et al. TP53 mutation is frequently associated with CTNNB1 mutation or MYCN amplification and is compatible with long-term survival in medulloblastoma. J Clin Oncol. 2010 Dec 10;28(35):5188–5196. doi: 10.1200/JCO.2010.31.1670. [DOI] [PubMed] [Google Scholar]
- 50.Rieber J, Remke M, Hartmann C, et al. Novel oncogene amplifications in tumors from a family with Li-Fraumeni syndrome. Genes Chromosomes Cancer. 2009 Jul;48(7):558–568. doi: 10.1002/gcc.20665. [DOI] [PubMed] [Google Scholar]
- 51.Eichenmuller M, Bauer R, Von Schweinitz D, et al. Hedgehog-independent overexpression of transforming growth factor-beta1 in rhabdomyosarcoma of Patched1 mutant mice. Int J Oncol. 2007 Aug;31(2):405–412. [PubMed] [Google Scholar]
- 52.Rice JM. Inducible and transmissible genetic events and pediatric tumors of the nervous system. J Radiat Res (Tokyo) 2006;47(Suppl B):B1–B11. doi: 10.1269/jrr.47.b1. [DOI] [PubMed] [Google Scholar]
- 53.Cowan R, Hoban P, Kelsey A, et al. The gene for the naevoid basal cell carcinoma syndrome acts as a tumour-suppressor gene in medulloblastoma. Br J Cancer. 1997;76(2):141–145. doi: 10.1038/bjc.1997.354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Calzada-Wack J, Kappler R, Schnitzbauer U, et al. Unbalanced overexpression of the mutant allele in murine Patched mutants. Carcinogenesis. 2002 May;23(5):727–733. doi: 10.1093/carcin/23.5.727. [DOI] [PubMed] [Google Scholar]
- 55.Wetmore C, Eberhart DE, Curran T. The normal patched allele is expressed in medulloblastomas from mice with heterozygous germ-line mutation of patched. Cancer Res. 2000 Apr 15;60(8):2239–2246. [PubMed] [Google Scholar]
- 56.Zurawel RH, Allen C, Wechsler-Reya R, et al. Evidence that haploinsufficiency of Ptch leads to medulloblastoma in mice. Genes Chromosomes Cancer. 2000 May;28(1):77–81. [PubMed] [Google Scholar]
- 57.Hamilton SR, Liu B, Parsons RE, et al. The molecular basis of Turcot’s syndrome. N Engl J Med. 1995 Mar 30;332(13):839–847. doi: 10.1056/NEJM199503303321302. [DOI] [PubMed] [Google Scholar]
- 58.Lindsey JC, Lusher ME, Anderton JA, et al. Identification of tumour-specific epigenetic events in medulloblastoma development by hypermethylation profiling. Carcinogenesis. 2004 May;25(5):661–668. doi: 10.1093/carcin/bgh055. [DOI] [PubMed] [Google Scholar]
- 59.Crawford JR, MacDonald TJ, Packer RJ. Medulloblastoma in childhood: new biological advances. Lancet Neurol. 2007 Dec;6(12):1073–1085. doi: 10.1016/S1474-4422(07)70289-2. [DOI] [PubMed] [Google Scholar]
- 60.Gajjar A, Hernan R, Kocak M, et al. Clinical, histopathologic, and molecular markers of prognosis: toward a new disease risk stratification system for medulloblastoma. J Clin Oncol. 2004 Mar 15;22(6):984–993. doi: 10.1200/JCO.2004.06.032. [DOI] [PubMed] [Google Scholar]
- 61.Gilbertson RJ. ERBB2 in pediatric cancer: innocent until proven guilty. Oncologist. 2005 Aug;10(7):508–517. doi: 10.1634/theoncologist.10-7-508. [DOI] [PubMed] [Google Scholar]
- 62.Ahmed N, Ratnayake M, Savoldo B, et al. Regression of experimental medulloblastoma following transfer of HER2-specific T cells. Cancer Res. 2007 Jun 15;67(12):5957–5964. doi: 10.1158/0008-5472.CAN-06-4309. [DOI] [PubMed] [Google Scholar]
- 63.Gulino A, Arcella A, Giangaspero F. Pathological and molecular heterogeneity of medulloblastoma. Curr Opin Oncol. 2008 Nov;20(6):668–675. doi: 10.1097/CCO.0b013e32831369f4. [DOI] [PubMed] [Google Scholar]
- 64.Paget S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 1989 Aug;8(2):98–101. [PubMed] [Google Scholar]
- 65.Fokas E, Engenhart-Cabillic R, Daniilidis K, et al. Metastasis: the seed and soil theory gains identity. Cancer Metastasis Rev. 2007 Dec;26(3–4):705–715. doi: 10.1007/s10555-007-9088-5. [DOI] [PubMed] [Google Scholar]
- 66.Weber CE, Kuo PC. The tumor microenvironment. Surg Oncol. 2011 Sep 29; doi: 10.1016/j.suronc.2011.09.001. [DOI] [PubMed] [Google Scholar]
- 67.Witz IP. The tumor microenvironment: the making of a paradigm. Cancer Microenviron. 2009 Sep;2(Suppl 1):9–17. doi: 10.1007/s12307-009-0025-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009 Apr;9(4):239–252. doi: 10.1038/nrc2618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011 Mar 4;144(5):646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 70.Bates RC, Mercurio AM. The epithelial-mesenchymal transition (EMT) and colorectal cancer progression. Cancer Biol Ther. 2005 Apr;4(4):365–370. doi: 10.4161/cbt.4.4.1655. [DOI] [PubMed] [Google Scholar]
- 71.Salsman VS, Chow KK, Shaffer DR, et al. Crosstalk between medulloblastoma cells and endothelium triggers a strong chemotactic signal recruiting T lymphocytes to the tumor microenvironment. PLoS One. 2011;6(5):e20267. doi: 10.1371/journal.pone.0020267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Papetti M, Herman IM. Mechanisms of normal and tumor-derived angiogenesis. Am J Physiol Cell Physiol. 2002 May;282(5):C947–C970. doi: 10.1152/ajpcell.00389.2001. [DOI] [PubMed] [Google Scholar]
- 73.Moses MA. The regulation of neovascularization of matrix metalloproteinases and their inhibitors. Stem Cells. 1997;15(3):180–189. doi: 10.1002/stem.150180. [DOI] [PubMed] [Google Scholar]
- 74.Xian X, Hakansson J, Stahlberg A, et al. Pericytes limit tumor cell metastasis. J Clin Invest. 2006 Mar;116(3):642–651. doi: 10.1172/JCI25705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Unemori EN, Ferrara N, Bauer EA, Amento EP. Vascular endothelial growth factor induces interstitial collagenase expression in human endothelial cells. J Cell Physiol. 1992 Dec;153(3):557–562. doi: 10.1002/jcp.1041530317. [DOI] [PubMed] [Google Scholar]
- 76.Kimura H, Weisz A, Ogura T, et al. Identification of hypoxia-inducible factor 1 ancillary sequence and its function in vascular endothelial growth factor gene induction by hypoxia and nitric oxide. J Biol Chem. 2001 Jan 19;276(3):2292–2298. doi: 10.1074/jbc.M008398200. [DOI] [PubMed] [Google Scholar]
- 77.Slongo ML, Molena B, Brunati AM, et al. Functional VEGF and VEGF receptors are expressed in human medulloblastomas. Neuro Oncol. 2007 Oct;9(4):384–392. doi: 10.1215/15228517-2007-032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Tural S, Gercek A, Konya D, et al. Microvessel density and vascular endothelial growth factor expression as predictors of childrens’ survival from cerebellar medulloblastoma. J Clin Neurosci. 2009 Sep;16(9):1199–1202. doi: 10.1016/j.jocn.2008.10.026. [DOI] [PubMed] [Google Scholar]
- 79.de Visser KE, Eichten A, Coussens LM. Paradoxical roles of the immune system during cancer development. Nat Rev Cancer. 2006 Jan;6(1):24–37. doi: 10.1038/nrc1782. [DOI] [PubMed] [Google Scholar]
- 80.Deschoolmeester V, Baay M, VanMarck E, et al. Tumor infiltrating lymphocytes: an intriguing player in the survival of colorectal cancer patients. BMC Immunol. 2010;11:19. doi: 10.1186/1471-2172-11-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ruffell B, Au A, Rugo HS, et al. Leukocyte composition of human breast cancer. Proc Natl Acad Sci U S A. 2012 Aug 8;109:2796–2801. doi: 10.1073/pnas.1104303108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Nelson BH. CD20+ B cells: the other tumor-infiltrating lymphocytes. J Immunol. 2010 Nov 1;185(9):4977–4982. doi: 10.4049/jimmunol.1001323. [DOI] [PubMed] [Google Scholar]
- 83.Gonzalez-Martin A, Gomez L, Lustgarten J, et al. Maximal T cell-mediated antitumor responses rely upon CCR5 expression in both CD4(+) and CD8(+) T cells. Cancer Res. 2011 Aug 15;71(16):5455–5466. doi: 10.1158/0008-5472.CAN-11-1687. [DOI] [PubMed] [Google Scholar]
- 84.Ostrand-Rosenberg S. Immune surveillance: a balance between protumor and antitumor immunity. Curr Opin Genet Dev. 2008 Feb;18(1):11–18. doi: 10.1016/j.gde.2007.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Wakabayashi O, Yamazaki K, Oizumi S, et al. CD4+T cells in cancer stroma, not CD8+T cells in cancer cell nests, are associated with favorable prognosis in human non-small cell lung cancers. Cancer Sci. 2003 Nov;94(11):1003–1009. doi: 10.1111/j.1349-7006.2003.tb01392.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Vincent S, Mirshari M, Nicolas C, et al. Large-cell medulloblastoma with arrestin-like protein expression. Clin Neuropathol. 2003 Jan-Feb;22(1):1–9. [PubMed] [Google Scholar]
- 87.Salama P, Phillips M, Grieu F, et al. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J Clin Oncol. 2009 Jan 10;27(2):186–192. doi: 10.1200/JCO.2008.18.7229. [DOI] [PubMed] [Google Scholar]
- 88.Zhou P, Sha H, Zhu J. The role of T-helper 17 (Th17) cells in patients with medulloblastoma. J Int Med Res. 2010 Mar-Apr;38(2):611–619. doi: 10.1177/147323001003800223. [DOI] [PubMed] [Google Scholar]
- 89.Wiegering V, Eyrich M, Rutkowski S, et al. TH1 predominance is associated with improved survival in pediatric medulloblastoma patients. Cancer Immunol Immunother. 2011 May;60(5):693–703. doi: 10.1007/s00262-011-0981-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Ahmed N, Salsman VS, Kew Y, et al. HER2-specific T cells target primary glioblastoma stem cells and induce regression of autologous experimental tumors. Clin Cancer Res. 2010 Jan 15;16(2):474–485. doi: 10.1158/1078-0432.CCR-09-1322. [DOI] [PMC free article] [PubMed] [Google Scholar]