Next Article in Journal
Pattern Recognition of the Multiple Sclerosis Syndrome
Next Article in Special Issue
4SC-202 as a Potential Treatment for the Pediatric Brain Tumor Medulloblastoma
Previous Article in Journal
Metacognitive Precursors: An Analysis in Children with Different Disabilities
Previous Article in Special Issue
Changes in Cognition and Decision Making Capacity Following Brain Tumour Resection: Illustrated with Two Cases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunotherapy for Pediatric Brain Tumors

University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lilian S. Wells Department of Neurosurgery, 1149 South Newell Drive, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
*
Author to whom correspondence should be addressed.
Brain Sci. 2017, 7(10), 137; https://doi.org/10.3390/brainsci7100137
Submission received: 16 August 2017 / Revised: 20 September 2017 / Accepted: 18 October 2017 / Published: 21 October 2017
(This article belongs to the Special Issue Advances in Adult and Pediatric Brain Tumor Management)

Abstract

:
Malignant brain tumors are the most common cause of solid cancer death in children. New targeted therapies are vital to improve treatment outcomes, but must be developed to enable trafficking across the blood brain barrier (BBB). Since activated T cells cross the BBB, cancer immunotherapy can be harnessed to unlock the cytotoxic potential of the immune system. However, standard of care treatments (i.e., chemotherapy and radiation) applied concomitant to pediatric brain tumor immunotherapy may abrogate induction of immunotherapeutic responses. This review will discuss the development of immunotherapies within this paradigm using emerging approaches being investigated in phase I/II trials in children with refractory brain tumors, including checkpoint inhibitors, vaccine immunotherapy, and adoptive cell therapy.

1. Introduction

Malignant brain tumors are the most common solid tumor in children [1,2,3]. Therapy is generally confined to maximal surgical resection, followed by adjuvant radiotherapy, and combination chemotherapy [4,5,6]. Despite improvements in outcomes, survivors often suffer toxicities from off-target effects, including incapacitating neurological deficits and cognitive decline [7]. Due to the debilitating side effects of surgery, radiation, and chemotherapy, new therapies are vital [7]. However, pediatric brain cancers are particularly challenging due to the blood brain barrier (BBB) and a lack of approved novel agents [8]. Most chemotherapeutic agents have limited penetration of the BBB [9,10,11,12,13]. For drugs that do penetrate (i.e., temozolomide), systemic levels typically dwarf therapeutic levels in the central nervous system (CNS), which compounds their secondary side effects [9,10,11,12,13]. New agents, such as tyrosine kinase inhibitors, may have limited penetration of the capillary tight junctions formed by endothelial cells of the BBB [14,15]. This endothelial barrier is reinforced by an epithelial barrier formed by the choroid plexus at the interface between the blood and cerebrospinal fluid [16]. Hence, translocation across these barriers is difficult to achieve from pharmacologic therapies, and direct intratumoral injection is encumbered by need for surgical access and indwelling catheters that are prone to increased edema, bleeding, and infection [14,17]. Given these limitations, new agents are necessary for specific targeting of brain tumor cells without the bystander damage synonymous with traditional chemotherapeutic agents.

2. Immunotherapy and Chemoradiation

Cancer immunotherapy is a burgeoning field of targeted therapy that harnesses the cytotoxic potential of the immune system against cancer cells. T cells are the effector arm responsible for flushing out foreign pathogens that pretend to be ‘self’. Viral infections typically hijack cell machinery, but major histocompatibility complex (MHC) class presented epitopes allow T cells to recognize and reject viral antigens on infected host cells [18]. Similarly, the preponderance of cancer cells are composed of normal antigens, but can be riddled with neoeptiopes sensitive to immunotherapeutic targeting [19,20,21]. Unlike traditional pharmacologic agents that require increased titration for full effect (often with dose limiting toxicities that preclude rampant dose escalation), immunotherapy is a biologic agent that may not necessitate the same dose-dependent increases for maximal impact [22]. These biologic agents require a catalyst to activate/reactivate the endogenous host response responsible for unlocking adaptive immunity and perpetuating self-sustaining immunologic T cell memory against malignant brain tumors [23]. Despite the BBB, the concept of the CNS being ‘immune-privileged’ is no longer adequate [24,25,26,27,28]. Recently, lymphatic drainage networks have been described within the CNS that communicate with deep cervical draining lymph nodes [24,25,26,27,28]. Activated T cells can be primed against tumor-specific antigens and traverse the BBB through adhesion markers (i.e., VLA-4), allowing them to penetrate the tumor microenvironment and induce their effector functions against pediatric cancer cells [24,25,26,27,28].
For children with malignant brain tumors, immunotherapy may be employed in conjunction with standard of care therapy. Since standard treatment typically involves surgery, radiation, and chemotherapy, the timing of immunotherapy must be reconciled to mitigate against the detrimental effects associated with these traditional modalities. Radiation may promote angiogenesis/wound healing and confound Th1 type responses by decreasing CD8+ dendritic cells [29,30]. However, radiation can synergize with immunotherapy through the release of damage associated molecular patterns (DAMPs) and tumor antigens that can be processed by endogenous antigen presenting cells (APCs) for presentation to T cells in local draining lymph nodes [31]. Chemotherapy, like radiation, can have both positive and negative effects when used in conjunction with immunotherapy [31]. Chemotherapy can be directly toxic to activated T cells, but may eliminate regulatory populations (i.e., regulatory T cells, myeloid derived suppressor cells) that compete for essential cytokines [32,33,34]. Removal of these cell compartments acting as cytokine sinks allows immunotherapeutic agents to induce their effector responses without competition [32,33,34]. To engender T cell or NK cell survival, adoptive cellular therapy is best utilized after radiation and chemotherapy; however, vaccine immunotherapy may synergize in conjunction with these treatment modalities [35,36]. Vaccines prime innate immunity for polarization into an adaptive T cell response and may synergize with the inflammatory milieu induced by radiation and chemotherapy [31,35,36]. Preclinical and clinical investigations have shown that vaccination through repeated cycles of chemotherapy enhances antigen specific T cell immunity [36,37,38]. Based on these data, future immunotherapy trials for children with malignant brain tumors can be developed in conjunction with standard of care chemoradiation. Vaccines may be optimal soon after surgical resection for continuation through cycles of adjuvant chemotherapy. Adoptive cellular therapy, if also utilized, can follow after completion of chemotherapy with continued vaccination cycles to maintain immunologic memory.

3. Antigen Selection

To incite an immune response against pediatric brain tumors, appropriate tumor antigens have to be selected and targeted. While nonsynonymous mutations that generate neoantigens are optimal targets (since they are not expressed on normal tissue), these are likely personalized epitopes that differ between patients, and are less prevalent in tumors with low mutational burdens (i.e., childhood CNS malignancies) [25]. Alternative targets include differentiation antigens or overexpressed normal antigens, but immunologic targeting of epitopes expressed on normal tissue may elicit autoimmunity [39]. Although most pediatric brain tumors have scant mutations at the genomic level, they are characterized by a plethora of epigenetic dysregulatory mechanisms, which may lead to the re-expression of developmental fetal antigens in malignancies, such as medulloblastoma and primitive neural ectodermal tumors (PNETs) [40,41,42]. In these malignancies, developmental antigens may be identifiable and targetable as tumor specific epitopes not expressed on mature normal tissue. Due to clustering of many pediatric brain tumors into distinct subgroups (based on expression profiles, medulloblastomas can be clustered into four distinct subgroups [43], and recently have been further subdivided into seven subgroups [44], while ependymomas can be stratified based on supratentorial or posterior fossa location [45]), there may be conserved targets unique to each subtype. Interestingly, diffuse intrinsic pontine gliomas (DIPGs) typically have a conserved mutation in histone 3.3 or 3.1 (K27M) [46]. This mutation may allow targeted immunotherapy in a surgically unresectable tumor against novel epitopes spanning the region of the K27M mutation [6,47,48]. Currently, H3.3K27M specific cancer vaccines are being evaluated in early phase studies for children with H3.3K27M positive DIPGs (clinicaltrials.gov: NCT02960230).

4. Immunotherapeutic Strategies

The immune system is one of the prime defense mechanisms against cancer cells, and works to actively survey and remove cells that are undergoing oncologic transformation [49,50]. Endogenous immunity is responsible for eradicating infectious etiologies and removing dysregulated pre-cancerous cells, but can also be remodeled (by malignancies) into an immunoregulatory state [51,52,53,54,55] When a critical mass of cancer cells develops, the immune system mounts a response, but consequently, selects immune resistant tumor cells to re-populate the mass [49,50]. As malignancies gain mutations they evolve to resist the immune system, while actively subverting it from inducing anti-tumor activity [56,57]. This new tumor microenvironment becomes comprised of immunoregulatory populations, including FoxP3+ regulatory T cells, tumor associated macrophages, and myeloid derived suppressor cells, which actively work to suppress effector T cells from mediating their cytolytic functions [58,59,60]. Immunotherapy can be employed to effectively reprogram the host immune response via immune checkpoint blockade, cancer vaccines, and adoptive cell therapy.
Immune checkpoint blockade antagonizes inhibitory receptor/ligand interactions (i.e., programmed death-ligand 1 (PD-L1) on APCs and tumor cells/programmed death-1 (PD-1) receptor on T cells) designed to inhibit activated T cells [61]. Blocking these inhibitory axes with monoclonal antibodies (mAbs) allows activated T cells to ‘stay on’ and mediate their anti-tumor effector functions [61]. Checkpoint inhibitors have shown remarkable results against adult cancers (i.e., skin and lung), but pediatric malignancies are distinct [39,62]. Unlike melanoma and lung cancer, which are characterized by high mutational burdens typically acquired via environmental mutagens (i.e., ultraviolet rays predisposing skin cancer and cigarette smoke predisposing lung cancer), pediatric cancers have a low mutational burden [21,61,62]. Responsiveness of melanoma to immune checkpoint blockade has been strongly associated with mutational burdens from non-synonymous changes and intratumoral enrichment of PD-1+ CD8+ T cells [21,54,61,63,64,65,66]. Although PD-1 can serve as an exhaustion marker, it is expressed on activated T cells, which are typically stymied by the regulatory microenvironment endemic to refractory solid tumors [65]. Checkpoint inhibitors unlock these activated T cells allowing them to exert their effector functions against neoantigen tumor epitopes [21,65]. Since pediatric brain tumors have low mutational burdens, they may be devoid of these endogenous PD-1+ T cells. Moreover, immune checkpoint mAbs are not expected to cross the BBB; however, in the presence of chemotherapy and radiation, this barrier may be perturbed, allowing these molecules to translocate [25,67,68]. Clinical trials are currently underway in children with high grade gliomas (HGGs) and DIPGs (phase I, clinicaltrials.gov: NCT02359565) to determine the utility of these agents against pediatric CNS malignancies.
Since childhood brain cancer is considered as a rare disease population, the development of novel therapies in clinical trials that can be studied in large data sets is necessary. While checkpoint inhibitors (i.e., PD-1 and PD-L1 mAbs) are promising candidates that can be leveraged for all patients, they appear to require tumors with high mutational burdens and PD-1+ intratumoral T cells (or high expressing PD-L1+ tumors) [61,62]. CD47, an immune evasion marker on the surface of many solid cancers, may represent a more relevant tumor immunosuppressive target [69]. CD47 is overexpressed by malignant cells to evade the innate immune response and may be particularly enriched in pediatric brain tumors including medulloblastoma, acute teratoid rhabdoid tumor, pediatric glioblastoma, and DIPG [69]. By antagonizing CD47 (which binds to activating signal regulatory protein-alpha (SIRPalpha) on myeloid cells) with a humanized mAb, pre-clinical investigations have demonstrated that macrophages can be unlocked to phagocytize tumor cells in several pediatric brain tumor models [69]. In addition to CD47 targeting, tumor immunosuppression can be overcome through targeting indoleamine 2,3-dioxygenase (IDO) [70,71]. IDO is an enzyme enriched in malignant brain tumors and functions to deplete tryptophan, an essential amino acid, from effector T cells [70,71]. Tryptophan is critical for DC maturation and for maintaining effector T cell survival/longevity [70,71]. Tumors that utilize IDO deplete tryptophan from T cells forming kynurenines, which predispose an exhausted and regulatory T cell phenotype [70,71]. IDO blockers have been developed with promising pre-clinical results in murine brain tumor models and are currently being evaluated in children in early phase studies (phase I, clinical trials.gov: NCT02502708) [55,59].
While several strategies exist for overcoming tumor-mediated immunosuppression, in the absence of robust preexisting endogenous immunity, targeting immunosuppression may be insufficient to induce meaningful anti-tumor responses. Alternatively, strategies have been employed to activate immunologic responses de novo against pediatric brain tumors. These include cancer vaccines, oncolytic viral therapy, and chimeric antigen receptor modified T cells. Cancer vaccines seek to deliver tumor antigens to APCs allowing them to process and present these epitopes on the surface of their MHC class I and II molecules for presentation to T cells [72]. Glioma associated peptide vaccines with poly-I/C have been investigated in HLA-A2 positive patients with DIPG and HGGs [73]. These peptides (commonly expressed in childhood gliomas) included EphA2, interleukin-13 receptor alpha, and survivin [73]. The peptides were emulsified in montanide-ISA-51 and given in conjunction with poly I/C (every three weeks x 8 cycles followed by booster administrations every six weeks) [73]. Thirteen of twenty-one children treated with these peptide vaccines had an immune response to the antigens [73]. Although these results are promising, HLA-A2 restricted vaccines constrain eligibility to patients that express this haplotype. Alternatively, DNA or RNA based vaccines bypass HLA haplotype restriction, allowing an individual’s cell machinery to transcribe/translate nucleic acids into proteins for personalized processing [74,75,76,77]. Our group has employed the use of RNA loaded ex vivo activated autologous dendritic cells from patients with medulloblastoma [78]. DCs are matured ex vivo from peripheral blood mononuclear cells (obtained via leukapheresis) and activated in the presence of inflammatory cytokines [78]. These cells are loaded with a personalized cohort of total tumor messenger RNA amplified from a personalized cDNA library representing a tumor specific transcriptome [78]. Autologous DCs are cultured with T cells (also obtained via leukapheresis) to activate them before both products (RNA-loaded DCs and ex vivo activated T cells) are re-administered to patients [78]. In patients with medulloblastoma and primitive neural ectodermal tumors, investigations are currently underway in phase I and II studies (clinicaltrials.gov NCT01326104) [78].
Like vaccines, oncolytic viral therapy intends to induce endogenous immune responses [79,80,81,82,83]. Oncolytic viruses are attenuated to preferentially propagate in tumor cells (which lack normal cellular defense mechanisms) until they are recognized by the immune system for rejection [79,80,81,82,83]. As these infected cancer cells lyse, a broader immune response is engendered more globally against shed tumor antigens, in a process known as epitope spreading [79,80,81,82,83,84]. Oncolytic viral therapy has been employed in several cancers with promising results and is currently being evaluated in phase I studies in children with recurrent supratentorial tumors (using herpes simplex virus; clinicaltrials.gov: NCT02457845) [85].
To mediate tumor regression, cancer vaccines and oncolytic viral therapy stimulate innate immune responses for polarization of an antigen specific T cell response. For these strategies to be successful, appropriate orchestration of an innate stimulus, APC activation/trafficking, and presentation to T cells must occur [72]. Subsequently, T cells must penetrate the notoriously immunosuppressive microenvironment characteristic of many CNS malignancies, and induce their effector functions against tumor cells that can downregulate MHC class I, which is requisite for T cell recognition [86]. To bypass this hurdle, engineered chimeric antigen receptor (CAR) modified T cells can be utilized to target surface antigens [87,88,89,90,91,92]. Since they recognize and reject surface antigens (i.e., HER2), CAR T cells overcome MHC downregulation by tumor cells [87,88,89,90,91,92]. Recently, CAR T cells have been shown to mediate significant anti-tumor activity against adult glioblastoma by targeting HER2 or IL13Ra2, which can be employed against pediatric high-grade gliomas (phase I, clinicaltrials.gov: NCT02208362) [93,94]. CAR T cells embed the specificity of an antibody with the cytotoxicity of a T cell, but remain limited by concerns of off-target effects [87,88,89,90,91,92]. CAR T cell targeting of normal antigens in the brain may lead to CNS inflammation, neurotoxicity, and risks for an increased intracranial pressure and herniation. Future development of CAR T cell therapies that activate only after binding to multiple tumor surface antigens may limit these off-target effects [95].
Other types of adoptive cell therapies for pediatric brain tumors include the transplantation of autologous/allogeneic tumor-specific cytotoxic T lymphocytes and natural killer (NK) cells. In children with posterior fossa tumors, autologous NK cells are being be expanded ex vivo before forth ventricular infusion (via ommaya reservoir) (phase I, clinicaltrials.gov: NCT02271711). Allogeneic T and NK cells are also being administered after non-myeloablative haploidentical transplant (reduced intensity conditioning regimens) to enable graft versus tumor killing against high risk solid tumors (i.e., pediatric brain tumors) (phase II, clinicaltrials.gov: NCT01804634 and NCT02100891). While these early phase studies are important to better understand the optimal dose, safety, and efficacy of these investigational agents, combinatorial approaches are likely requisite to fully exploit the benefits of adoptive cell therapy. By antagonizing tumor induced immunosuppression and activating endogenous immunity (via vaccine/viral immunotherapy and adoptive T cell therapy), combination immunotherapy promises to synergize for maximal therapeutic benefit.

5. Conclusions

While aggressive surgical resection, radiation, and chemotherapy have improved the cure rates for many pediatric patients with malignant brain tumors, these cancers remain the most common cause of solid cancer death in children [96]. New targeted therapies are vital to improve treatment outcomes, but must be developed to enable trafficking across the BBB. Cancer immunotherapy is a promising treatment modality with encouraging results in melanoma, leukemia/lymphoma, and lung cancer [62,91,97]. Although immunotherapy has shown promise against peripheral cancers, it remains challenged by the perceived lack of immunogenic/targetable antigens in pediatric brain tumors. However, given the emergence of monoclonal antibodies targeting checkpoint molecules and ‘don’t eat me’ signals (i.e., CD47), we can now unlock innate and adaptive immunity in a non-specific manner. Although the BBB remains a significant hurdle, activated T cells can cross this barricade. Tumor specific T cells can be activated de novo through cancer vaccines, oncolytic viral therapy, and ex vivo engineered cell therapies. As new targets are uncovered from subgroup speciation of pediatric brain tumors, novel therapies are expected to follow with improved outcomes.

Acknowledgments

E.J.S. is supported by the U.S. Department of Defense (W81XWH-17-1-0510) National Institutes of Health (NCI K08CA199224) and St. Baldrick’s Foundation (Hannah’s Heroes Scholar Award). D.A.M. is supported by the National Institutes of Health (NCI R01-CA195563-01, 1R01-CA175517-01), Department of Defense (W81XWH-10-1-0089) and Hyundai Hope on Wheels.

Conflicts of Interest

D.A.M. has patented immunotherapy related technology that have been licensed by Annias Immunotherapeutics, Inc., and Celldex Therapeutics, Inc.

References

  1. Hennika, T.; Becher, O.J. Diffuse Intrinsic Pontine Glioma: Time for Cautious Optimism. J. Child Neurol. 2015. [Google Scholar] [CrossRef] [PubMed]
  2. Graham, M.L.; Herndon, J.E., II; Casey, J.R.; Chaffee, S.; Ciocci, G.H.; Krischer, J.P.; Kurtzberg, J.; Laughlin, M.J.; Longee, D.C.; Olson, J.F.; et al. High-dose chemotherapy with autologous stem-cell rescue in patients with recurrent and high-risk pediatric brain tumors. J. Clin. Oncol. 1997, 15, 1814–1823. [Google Scholar] [CrossRef] [PubMed]
  3. Brandao, L.A.; Young Poussaint, T. Posterior Fossa Tumors. Neuroimaging Clin. N. Am. 2017, 27, 1–37. [Google Scholar] [CrossRef] [PubMed]
  4. Peres, E.; Wood, G.W.; Poulik, J.; Baynes, R.; Sood, S.; Abidi, M.H.; Klein, J.; Bhambhani, K.; Dansey, R.; Abella, E. High-dose chemotherapy and adoptive immunotherapy in the treatment of recurrent pediatric brain tumors. Neuropediatrics 2008, 39, 151–156. [Google Scholar] [CrossRef] [PubMed]
  5. Ruggiero, A.; Cefalo, G.; Garre, M.L.; Massimino, M.; Colosimo, C.; Attina, G.; Lazzareschi, I.; Maurizi, P.; Ridola, V.; Mazzarella, G.; et al. Phase II trial of temozolomide in children with recurrent high-grade glioma. J. Neuro-Oncol. 2006, 77, 89–94. [Google Scholar] [CrossRef] [PubMed]
  6. Solomon, D.A.; Wood, M.D.; Tihan, T.; Bollen, A.W.; Gupta, N.; Phillips, J.J.; Perry, A. Diffuse Midline Gliomas with Histone H3-K27M Mutation: A Series of 47 Cases Assessing the Spectrum of Morphologic Variation and Associated Genetic Alterations. Brain Pathol. 2015. [Google Scholar] [CrossRef] [PubMed]
  7. Finlay, J.L.; Goldman, S.; Wong, M.C.; Cairo, M.; Garvin, J.; August, C.; Cohen, B.H.; Stanley, P.; Zimmerman, R.A.; Bostrom, B.; et al. Pilot study of high-dose thiotepa and etoposide with autologous bone marrow rescue in children and young adults with recurrent CNS tumors. The Children’s Cancer Group. J. Clin. Oncol. 1996, 14, 2495–2503. [Google Scholar] [CrossRef] [PubMed]
  8. Ransohoff, R.M.; Kivisakk, P.; Kidd, G. Three or more routes for leukocyte migration into the central nervous system. Nat. Rev. Immunol. 2003, 3, 569–581. [Google Scholar] [CrossRef] [PubMed]
  9. Finlay, J.L.; Dhall, G.; Boyett, J.M.; Dunkel, I.J.; Gardner, S.L.; Goldman, S.; Yates, A.J.; Rosenblum, M.K.; Stanley, P.; Zimmerman, R.A.; et al. Myeloablative chemotherapy with autologous bone marrow rescue in children and adolescents with recurrent malignant astrocytoma: Outcome compared with conventional chemotherapy: A report from the Children’s Oncology Group. Pediatr. Blood Cancer 2008, 51, 806–811. [Google Scholar] [CrossRef] [PubMed]
  10. Bouffet, E.; Mottolese, C.; Jouvet, A.; Philip, I.; Frappaz, D.; Carrie, C.; Brunat-Mentigny, M. Etoposide and thiotepa followed by ABMT (autologous bone marrow transplantation) in children and young adults with high-grade gliomas. Eur. J. Cancer 1997, 33, 91–95. [Google Scholar] [CrossRef]
  11. Chen, B.; Ahmed, T.; Mannancheril, A.; Gruber, M.; Benzil, D.L. Safety and efficacy of high-dose chemotherapy with autologous stem cell transplantation for patients with malignant astrocytomas. Cancer 2004, 100, 2201–2207. [Google Scholar] [CrossRef] [PubMed]
  12. Sengupta, S.; Marrinan, J.; Frishman, C.; Sampath, P. Impact of temozolomide on immune response during malignant glioma chemotherapy. Clin. Dev. Immunol. 2012, 2012, 831090. [Google Scholar] [CrossRef] [PubMed]
  13. Wolff, J.E.; Finlay, J.L. High-dose chemotherapy in childhood brain tumors. Onkologie 2004, 27, 239–245. [Google Scholar] [CrossRef] [PubMed]
  14. Miranda, A.; Blanco-Prieto, M.; Sousa, J.; Pais, A.; Vitorino, C. Breaching barriers in glioblastoma. Part I: Molecular pathways and novel treatment approaches. Int. J. Pharm. 2017, 531, 372–388. [Google Scholar] [CrossRef] [PubMed]
  15. Zhou, K.; Bostrom, M.; Ek, C.J.; Li, T.; Xie, C.; Xu, Y.; Sun, Y.; Blomgren, K.; Zhu, C. Radiation induces progenitor cell death, microglia activation, and blood-brain barrier damage in the juvenile rat cerebellum. Sci. Rep. 2017, 7, 46181. [Google Scholar] [CrossRef] [PubMed]
  16. Vandenbroucke, R.E. A Hidden Epithelial Barrier in the Brain with a Central Role in Regulating Brain Homeostasis. Implications for Aging. Ann. Am. Thorac. Soc. 2016, 13, S407–S410. [Google Scholar] [CrossRef] [PubMed]
  17. Shahar, T.; Ram, Z.; Kanner, A.A. Convection-enhanced delivery catheter placements for high-grade gliomas: Complications and pitfalls. J. Neuro-Oncol. 2012, 107, 373–378. [Google Scholar] [CrossRef] [PubMed]
  18. Fruh, K.; Picker, L. CD8+ T cell programming by cytomegalovirus vectors: Applications in prophylactic and therapeutic vaccination. Curr. Opin. Immunol. 2017, 47, 52–56. [Google Scholar] [CrossRef] [PubMed]
  19. Lu, Y.C.; Robbins, P.F. Targeting neoantigens for cancer immunotherapy. Int. Immunol. 2016, 28, 365–370. [Google Scholar] [CrossRef] [PubMed]
  20. Gros, A.; Parkhurst, M.R.; Tran, E.; Pasetto, A.; Robbins, P.F.; Ilyas, S.; Prickett, T.D.; Gartner, J.J.; Crystal, J.S.; Roberts, I.M.; et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat. Med. 2016, 22, 433–438. [Google Scholar] [CrossRef] [PubMed]
  21. Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S.; et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 2014, 371, 2189–2199. [Google Scholar] [CrossRef] [PubMed]
  22. Kreiter, S.; Vormehr, M.; van de Roemer, N.; Diken, M.; Lower, M.; Diekmann, J.; Boegel, S.; Schrors, B.; Vascotto, F.; Castle, J.C.; et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 2015, 520, 692–696. [Google Scholar] [CrossRef] [PubMed]
  23. Ponsaerts, P.; Van Tendeloo, V.F.; Berneman, Z.N. Cancer immunotherapy using RNA-loaded dendritic cells. Clin. Exp. Immunol. 2003, 134, 378–384. [Google Scholar] [CrossRef] [PubMed]
  24. Sampson, J.H.; Heimberger, A.B.; Archer, G.E.; Aldape, K.D.; Friedman, A.H.; Friedman, H.S.; Gilbert, M.R.; Herndon, J.E., II; McLendon, R.E.; Mitchell, D.A.; et al. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J. Clin. Oncol. 2010, 28, 4722–4729. [Google Scholar] [CrossRef] [PubMed]
  25. Sampson, J.H.; Maus, M.V.; June, C.H. Immunotherapy for Brain Tumors. J. Clin. Oncol. 2017, 35, 2450–2456. [Google Scholar] [CrossRef] [PubMed]
  26. Schlager, C.; Korner, H.; Krueger, M.; Vidoli, S.; Haberl, M.; Mielke, D.; Brylla, E.; Issekutz, T.; Cabanas, C.; Nelson, P.J.; et al. Effector T-cell trafficking between the leptomeninges and the cerebrospinal fluid. Nature 2016, 530, 349–353. [Google Scholar] [CrossRef] [PubMed]
  27. Aspelund, A.; Antila, S.; Proulx, S.T.; Karlsen, T.V.; Karaman, S.; Detmar, M.; Wiig, H.; Alitalo, K. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J. Exp. Med. 2015, 212, 991–999. [Google Scholar] [CrossRef] [PubMed]
  28. Louveau, A.; Smirnov, I.; Keyes, T.J.; Eccles, J.D.; Rouhani, S.J.; Peske, J.D.; Derecki, N.C.; Castle, D.; Mandell, J.W.; Lee, K.S.; et al. Structural and functional features of central nervous system lymphatic vessels. Nature 2015, 523, 337–341. [Google Scholar] [CrossRef] [PubMed]
  29. Haubner, F.; Ohmann, E.; Pohl, F.; Strutz, J.; Gassner, H.G. Wound healing after radiation therapy: Review of the literature. Radiat. Oncol. 2012, 7, 162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Liu, H.; Li, B.; Jia, X.; Ma, Y.; Gu, Y.; Zhang, P.; Wei, Q.; Cai, J.; Cui, J.; Gao, F.; et al. Radiation-induced decrease of CD8+ dendritic cells contributes to Th1/Th2 shift. Int. Immunopharmacol. 2017, 46, 178–185. [Google Scholar] [CrossRef] [PubMed]
  31. Vandenabeele, P.; Vandecasteele, K.; Bachert, C.; Krysko, O.; Krysko, D.V. Immunogenic Apoptotic Cell Death and Anticancer Immunity. Adv. Exp. Med. Biol. 2016, 930, 133–149. [Google Scholar] [CrossRef] [PubMed]
  32. Dunkel, I.J.; Gardner, S.L.; Garvin, J.H., Jr.; Goldman, S.; Shi, W.; Finlay, J.L. High-dose carboplatin, thiotepa, and etoposide with autologous stem cell rescue for patients with previously irradiated recurrent medulloblastoma. Neuro-Oncology 2010, 12, 297–303. [Google Scholar] [CrossRef] [PubMed]
  33. Welters, M.J.; van der Sluis, T.C.; van Meir, H.; Loof, N.M.; van Ham, V.J.; van Duikeren, S.; Santegoets, S.J.; Arens, R.; de Kam, M.L.; Cohen, A.F.; et al. Vaccination during myeloid cell depletion by cancer chemotherapy fosters robust T cell responses. Sci. Transl. Med. 2016, 8, 334ra352. [Google Scholar] [CrossRef] [PubMed]
  34. Gattinoni, L.; Finkelstein, S.E.; Klebanoff, C.A.; Antony, P.A.; Palmer, D.C.; Spiess, P.J.; Hwang, L.N.; Yu, Z.; Wrzesinski, C.; Heimann, D.M.; et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J. Exp. Med. 2005, 202, 907–912. [Google Scholar] [CrossRef] [PubMed]
  35. Sanchez-Perez, L.A.; Choi, B.D.; Archer, G.E.; Cui, X.; Flores, C.; Johnson, L.A.; Schmittling, R.J.; Snyder, D.; Herndon, J.E., II; Bigner, D.D.; et al. Myeloablative temozolomide enhances CD8(+) T-cell responses to vaccine and is required for efficacy against brain tumors in mice. PLoS ONE 2013, 8, e59082. [Google Scholar] [CrossRef] [PubMed]
  36. Mitchell, D.A.; Cui, X.; Schmittling, R.J.; Sanchez-Perez, L.; Snyder, D.J.; Congdon, K.L.; Archer, G.E.; Desjardins, A.; Friedman, A.H.; Friedman, H.S.; et al. Monoclonal antibody blockade of IL-2 receptor alpha during lymphopenia selectively depletes regulatory T cells in mice and humans. Blood 2011, 118, 3003–3012. [Google Scholar] [CrossRef] [PubMed]
  37. Mitchell, D.A.; Batich, K.A.; Gunn, M.D.; Huang, M.N.; Sanchez-Perez, L.; Nair, S.K.; Congdon, K.L.; Reap, E.A.; Archer, G.E.; Desjardins, A.; et al. Tetanus toxoid and CCL3 improve dendritic cell vaccines in mice and glioblastoma patients. Nature 2015, 519, 366–369. [Google Scholar] [CrossRef] [PubMed]
  38. Batich, K.A.; Swartz, A.M.; Sampson, J.H. Preconditioning Vaccine Sites for mRNA-Transfected Dendritic Cell Therapy and Antitumor Efficacy. Methods mol. Biol. 2016, 1403, 819–838. [Google Scholar] [CrossRef] [PubMed]
  39. Hao, C.; Tian, J.; Liu, H.; Li, F.; Niu, H.; Zhu, B. Efficacy and safety of anti-PD-1 and anti-PD-1 combined with anti-CTLA-4 immunotherapy to advanced melanoma: A systematic review and meta-analysis of randomized controlled trials. Medicine 2017, 96, e7325. [Google Scholar] [CrossRef] [PubMed]
  40. Alizadeh, A.A.; Aranda, V.; Bardelli, A.; Blanpain, C.; Bock, C.; Borowski, C.; Caldas, C.; Califano, A.; Doherty, M.; Elsner, M.; et al. Toward understanding and exploiting tumor heterogeneity. Nat. Med. 2015, 21, 846–853. [Google Scholar] [CrossRef] [PubMed]
  41. Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Borresen-Dale, A.L.; et al. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef] [PubMed]
  42. Fontebasso, A.M.; Gayden, T.; Nikbakht, H.; Neirinck, M.; Papillon-Cavanagh, S.; Majewski, J.; Jabado, N. Epigenetic dysregulation: A novel pathway of oncogenesis in pediatric brain tumors. Acta Neuropathol. 2014, 128, 615–627. [Google Scholar] [CrossRef] [PubMed]
  43. Northcott, P.A.; Korshunov, A.; Witt, H.; Hielscher, T.; Eberhart, C.G.; Mack, S.; Bouffet, E.; Clifford, S.C.; Hawkins, C.E.; French, P.; et al. Medulloblastoma comprises four distinct molecular variants. J. Clin. Oncol. 2011, 29, 1408–1414. [Google Scholar] [CrossRef] [PubMed]
  44. Schwalbe, E.C.; Lindsey, J.C.; Nakjang, S.; Crosier, S.; Smith, A.J.; Hicks, D.; Rafiee, G.; Hill, R.M.; Iliasova, A.; Stone, T.; et al. Novel molecular subgroups for clinical classification and outcome prediction in childhood medulloblastoma: A cohort study. Lancet Oncol. 2017, 18, 958–971. [Google Scholar] [CrossRef]
  45. Pajtler, K.W.; Mack, S.C.; Ramaswamy, V.; Smith, C.A.; Witt, H.; Smith, A.; Hansford, J.R.; von Hoff, K.; Wright, K.D.; Hwang, E.; et al. The current consensus on the clinical management of intracranial ependymoma and its distinct molecular variants. Acta Neuropathol. 2017, 133, 5–12. [Google Scholar] [CrossRef] [PubMed]
  46. Castel, D.; Philippe, C.; Calmon, R.; Le Dret, L.; Truffaux, N.; Boddaert, N.; Pages, M.; Taylor, K.R.; Saulnier, P.; Lacroix, L.; et al. Histone H3F3A and HIST1H3B K27M mutations define two subgroups of diffuse intrinsic pontine gliomas with different prognosis and phenotypes. Acta Neuropathol. 2015, 130, 815–827. [Google Scholar] [CrossRef] [PubMed]
  47. Hou, Y.F.; Kohanbash, G.; Okada, K.; Shrivastav, S.; Smith-Cohn, M.; Nicolaides, T.; Mueller, S.; Carcaboso, A.M.; Pollack, I.F.; Okada, H. Novel and shared neoantigen for glioma T cell therapy derived from histone 3 variant H3.3 K27M mutation. Cancer Immunol. Res. 2016, 4, P445. [Google Scholar] [CrossRef]
  48. Nguyen, A.T.; Colin, C.; Nanni-Metellus, I.; Padovani, L.; Maurage, C.A.; Varlet, P.; Miquel, C.; Uro-Coste, E.; Godfraind, C.; Lechapt-Zalcman, E.; et al. Evidence for BRAF V600E and H3F3A K27M double mutations in paediatric glial and glioneuronal tumours. Neuropathol. Appl. Neurobiol. 2015, 41, 403–408. [Google Scholar] [CrossRef] [PubMed]
  49. Koebel, C.M.; Vermi, W.; Swann, J.B.; Zerafa, N.; Rodig, S.J.; Old, L.J.; Smyth, M.J.; Schreiber, R.D. Adaptive immunity maintains occult cancer in an equilibrium state. Nature 2007, 450, 903–907. [Google Scholar] [CrossRef] [PubMed]
  50. Kozlowska, A.; Mackiewicz, J.; Mackiewicz, A. Therapeutic gene modified cell based cancer vaccines. Gene 2013, 525, 200–207. [Google Scholar] [CrossRef] [PubMed]
  51. Sampson, J.H.; Aldape, K.D.; Archer, G.E.; Coan, A.; Desjardins, A.; Friedman, A.H.; Friedman, H.S.; Gilbert, M.R.; Herndon, J.E.; McLendon, R.E.; et al. Greater chemotherapy-induced lymphopenia enhances tumor-specific immune responses that eliminate EGFRvIII-expressing tumor cells in patients with glioblastoma. Neuro-Oncology 2011, 13, 324–333. [Google Scholar] [CrossRef] [PubMed]
  52. Bloch, O.; Crane, C.A.; Kaur, R.; Safaee, M.; Rutkowski, M.J.; Parsa, A.T. Gliomas promote immunosuppression through induction of B7-H1 expression in tumor-associated macrophages. Clin. Cancer Res. 2013, 19, 3165–3175. [Google Scholar] [CrossRef] [PubMed]
  53. Highfill, S.L.; Cui, Y.; Giles, A.J.; Smith, J.P.; Zhang, H.; Morse, E.; Kaplan, R.N.; Mackall, C.L. Disruption of CXCR2-mediated MDSC tumor trafficking enhances anti-PD1 efficacy. Sci. Transl. Med. 2014, 6, 237ra267. [Google Scholar] [CrossRef] [PubMed]
  54. John, L.B.; Devaud, C.; Duong, C.P.; Yong, C.S.; Beavis, P.A.; Haynes, N.M.; Chow, M.T.; Smyth, M.J.; Kershaw, M.H.; Darcy, P.K. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin. Cancer Res. 2013, 19, 5636–5646. [Google Scholar] [CrossRef] [PubMed]
  55. Wainwright, D.A.; Balyasnikova, I.V.; Chang, A.L.; Ahmed, A.U.; Moon, K.S.; Auffinger, B.; Tobias, A.L.; Han, Y.; Lesniak, M.S. IDO expression in brain tumors increases the recruitment of regulatory T cells and negatively impacts survival. Clin. Cancer Res. 2012, 18, 6110–6121. [Google Scholar] [CrossRef] [PubMed]
  56. Van der Burg, S.H.; Arens, R.; Ossendorp, F.; van Hall, T.; Melief, C.J. Vaccines for established cancer: Overcoming the challenges posed by immune evasion. Nat. Rev. Cancer 2016, 16, 219–233. [Google Scholar] [CrossRef] [PubMed]
  57. Zitvogel, L.; Tesniere, A.; Kroemer, G. Cancer despite immunosurveillance: Immunoselection and immunosubversion. Nat. Rev. Immunol. 2006, 6, 715–727. [Google Scholar] [CrossRef] [PubMed]
  58. Klepsch, V.; Hermann-Kleiter, N.; Baier, G. Beyond CTLA-4 and PD-1: Orphan nuclear receptor NR2F6 as T cell signaling switch and emerging target in cancer immunotherapy. Immunol. Lett. 2016, 178, 31–36. [Google Scholar] [CrossRef] [PubMed]
  59. Lob, S.; Konigsrainer, A.; Rammensee, H.G.; Opelz, G.; Terness, P. Inhibitors of indoleamine-2,3-dioxygenase for cancer therapy: Can we see the wood for the trees? Nat. Rev. Cancer 2009, 9, 445–452. [Google Scholar] [CrossRef] [PubMed]
  60. Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef] [PubMed]
  61. Duraiswamy, J.; Kaluza, K.M.; Freeman, G.J.; Coukos, G. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res. 2013, 73, 3591–3603. [Google Scholar] [CrossRef] [PubMed]
  62. Borghaei, H.; Paz-Ares, L.; Horn, L.; Spigel, D.R.; Steins, M.; Ready, N.E.; Chow, L.Q.; Vokes, E.E.; Felip, E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 1627–1639. [Google Scholar] [CrossRef] [PubMed]
  63. Ansell, S.M.; Lesokhin, A.M.; Borrello, I.; Halwani, A.; Scott, E.C.; Gutierrez, M.; Schuster, S.J.; Millenson, M.M.; Cattry, D.; Freeman, G.J.; et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N. Engl. J. Med. 2015, 372, 311–319. [Google Scholar] [CrossRef] [PubMed]
  64. Badoual, C.; Hans, S.; Merillon, N.; Van Ryswick, C.; Ravel, P.; Benhamouda, N.; Levionnois, E.; Nizard, M.; Si-Mohamed, A.; Besnier, N.; et al. PD-1-expressing tumor-infiltrating T cells are a favorable prognostic biomarker in HPV-associated head and neck cancer. Cancer Res. 2013, 73, 128–138. [Google Scholar] [CrossRef] [PubMed]
  65. Gros, A.; Robbins, P.F.; Yao, X.; Li, Y.F.; Turcotte, S.; Tran, E.; Wunderlich, J.R.; Mixon, A.; Farid, S.; Dudley, M.E.; et al. PD-1 identifies the patient-specific CD8(+) tumor-reactive repertoire infiltrating human tumors. J. Clin. Investig. 2014, 124, 2246–2259. [Google Scholar] [CrossRef] [PubMed]
  66. Karyampudi, L.; Lamichhane, P.; Scheid, A.D.; Kalli, K.R.; Shreeder, B.; Krempski, J.W.; Behrens, M.D.; Knutson, K.L. Accumulation of memory precursor CD8 T cells in regressing tumors following combination therapy with vaccine and anti-PD-1 antibody. Cancer Res. 2014, 74, 2974–2985. [Google Scholar] [CrossRef] [PubMed]
  67. Baumann, B.C.; Kao, G.D.; Mahmud, A.; Harada, T.; Swift, J.; Chapman, C.; Xu, X.; Discher, D.E.; Dorsey, J.F. Enhancing the efficacy of drug-loaded nanocarriers against brain tumors by targeted radiation therapy. Oncotarget 2013, 4, 64–79. [Google Scholar] [CrossRef] [PubMed]
  68. Sattiraju, A.; Xiong, X.; Pandya, D.N.; Wadas, T.J.; Xuan, A.; Sun, Y.; Jung, Y.; Solingapuram Sai, K.K.; Dorsey, J.F.; Li, K.C.; et al. Alpha particle enhanced Blood Brain/Tumor Barrier permeabilization in glioblastomas using integrin alpha-v beta-3 targeted liposomes. Mol. Cancer Ther. 2017. [Google Scholar] [CrossRef] [PubMed]
  69. Gholamin, S.; Mitra, S.S.; Feroze, A.H.; Liu, J.; Kahn, S.A.; Zhang, M.; Esparza, R.; Richard, C.; Ramaswamy, V.; Remke, M.; et al. Disrupting the CD47-SIRPalpha anti-phagocytic axis by a humanized anti-CD47 antibody is an efficacious treatment for malignant pediatric brain tumors. Sci. Transl. Med. 2017, 9, eaaf2968. [Google Scholar] [CrossRef] [PubMed]
  70. Hwang, S.L.; Chung, N.P.; Chan, J.K.; Lin, C.L. Indoleamine 2,3-dioxygenase (IDO) is essential for dendritic cell activation and chemotactic responsiveness to chemokines. Cell Res. 2005, 15, 167–175. [Google Scholar] [CrossRef] [PubMed]
  71. Mbongue, J.C.; Nicholas, D.A.; Torrez, T.W.; Kim, N.S.; Firek, A.F.; Langridge, W.H. The Role of Indoleamine 2,3-Dioxygenase in Immune Suppression and Autoimmunity. Vaccines 2015, 3, 703–729. [Google Scholar] [CrossRef] [PubMed]
  72. Jahnisch, H.; Fussel, S.; Kiessling, A.; Wehner, R.; Zastrow, S.; Bachmann, M.; Rieber, E.P.; Wirth, M.P.; Schmitz, M. Dendritic cell-based immunotherapy for prostate cancer. Clin. Dev. Immunol. 2010, 2010, 517493. [Google Scholar] [CrossRef] [PubMed]
  73. Pollack, I.F.; Jakacki, R.I.; Butterfield, L.H.; Hamilton, R.L.; Panigrahy, A.; Potter, D.M.; Connelly, A.K.; Dibridge, S.A.; Whiteside, T.L.; Okada, H. Antigen-specific immune responses and clinical outcome after vaccination with glioma-associated antigen peptides and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in children with newly diagnosed malignant brainstem and nonbrainstem gliomas. J. Clin. Oncol. 2014, 32, 2050–2058. [Google Scholar] [CrossRef] [PubMed]
  74. Holtkamp, S.; Kreiter, S.; Selmi, A.; Simon, P.; Koslowski, M.; Huber, C.; Tureci, O.; Sahin, U. Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells. Blood 2006, 108, 4009–4017. [Google Scholar] [CrossRef] [PubMed]
  75. Kranz, L.M.; Diken, M.; Haas, H.; Kreiter, S.; Loquai, C.; Reuter, K.C.; Meng, M.; Fritz, D.; Vascotto, F.; Hefesha, H.; et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 2016, 534, 396–401. [Google Scholar] [CrossRef] [PubMed]
  76. Kreiter, S.; Selmi, A.; Diken, M.; Koslowski, M.; Britten, C.M.; Huber, C.; Tureci, O.; Sahin, U. Intranodal vaccination with naked antigen-encoding RNA elicits potent prophylactic and therapeutic antitumoral immunity. Cancer Res. 2010, 70, 9031–9040. [Google Scholar] [CrossRef] [PubMed]
  77. Coban, C.; Kobiyama, K.; Aoshi, T.; Takeshita, F.; Horii, T.; Akira, S.; Ishii, K.J. Novel strategies to improve DNA vaccine immunogenicity. Curr. Gene Ther. 2011, 11, 479–484. [Google Scholar] [CrossRef] [PubMed]
  78. Nair, S.K.; Driscoll, T.; Boczkowski, D.; Schmittling, R.; Reynolds, R.; Johnson, L.A.; Grant, G.; Fuchs, H.; Bigner, D.D.; Sampson, J.H.; et al. Ex vivo generation of dendritic cells from cryopreserved, post-induction chemotherapy, mobilized leukapheresis from pediatric patients with medulloblastoma. J. Neuro-Oncol. 2015, 125, 65–74. [Google Scholar] [CrossRef] [PubMed]
  79. Aurelian, L. Oncolytic viruses as immunotherapy: Progress and remaining challenges. OncoTargets Ther. 2016, 9, 2627–2637. [Google Scholar] [CrossRef] [PubMed]
  80. Kaufman, H.L.; Kohlhapp, F.J.; Zloza, A. Oncolytic viruses: A new class of immunotherapy drugs. Nat. Rev. Drug Discov. 2015, 14, 642–662. [Google Scholar] [CrossRef] [PubMed]
  81. Killock, D. Skin cancer: T-VEC oncolytic viral therapy shows promise in melanoma. Nat. Rev. Clin. Oncol. 2015, 12, 438. [Google Scholar] [CrossRef] [PubMed]
  82. Kohlhapp, F.J.; Kaufman, H.L. Molecular Pathways: Mechanism of Action for Talimogene Laherparepvec, a New Oncolytic Virus Immunotherapy. Clin. Cancer Res. 2016, 22, 1048–1054. [Google Scholar] [CrossRef] [PubMed]
  83. Yu, Y.A.; Galanis, C.; Woo, Y.; Chen, N.; Zhang, Q.; Fong, Y.; Szalay, A.A. Regression of human pancreatic tumor xenografts in mice after a single systemic injection of recombinant vaccinia virus GLV-1h68. Mol. Cancer Ther. 2009, 8, 141–151. [Google Scholar] [CrossRef] [PubMed]
  84. Kanerva, A.; Nokisalmi, P.; Diaconu, I.; Koski, A.; Cerullo, V.; Liikanen, I.; Tahtinen, S.; Oksanen, M.; Heiskanen, R.; Pesonen, S.; et al. Antiviral and antitumor T-cell immunity in patients treated with GM-CSF-coding oncolytic adenovirus. Clin. Cancer Res. 2013, 19, 2734–2744. [Google Scholar] [CrossRef] [PubMed]
  85. Saha, D.; Martuza, R.L.; Rabkin, S.D. Macrophage Polarization Contributes to Glioblastoma Eradication by Combination Immunovirotherapy and Immune Checkpoint Blockade. Cancer Cell 2017, 32, 253–267. [Google Scholar] [CrossRef] [PubMed]
  86. Zaretsky, J.M.; Garcia-Diaz, A.; Shin, D.S.; Escuin-Ordinas, H.; Hugo, W.; Hu-Lieskovan, S.; Torrejon, D.Y.; Abril-Rodriguez, G.; Sandoval, S.; Barthly, L.; et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N. Engl. J. Med. 2016, 375, 819–829. [Google Scholar] [CrossRef] [PubMed]
  87. Garfall, A.L.; Maus, M.V.; Hwang, W.T.; Lacey, S.F.; Mahnke, Y.D.; Melenhorst, J.J.; Zheng, Z.; Vogl, D.T.; Cohen, A.D.; Weiss, B.M.; et al. Chimeric Antigen Receptor T Cells against CD19 for Multiple Myeloma. N. Engl. J. Med. 2015, 373, 1040–1047. [Google Scholar] [CrossRef] [PubMed]
  88. Grupp, S.A.; Kalos, M.; Barrett, D.; Aplenc, R.; Porter, D.L.; Rheingold, S.R.; Teachey, D.T.; Chew, A.; Hauck, B.; Wright, J.F.; et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 2013, 368, 1509–1518. [Google Scholar] [CrossRef] [PubMed]
  89. Kalos, M.; Levine, B.L.; Porter, D.L.; Katz, S.; Grupp, S.A.; Bagg, A.; June, C.H. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci. Transl. Med. 2011, 3, 95ra73. [Google Scholar] [CrossRef] [PubMed]
  90. Louis, C.U.; Savoldo, B.; Dotti, G.; Pule, M.; Yvon, E.; Myers, G.D.; Rossig, C.; Russell, H.V.; Diouf, O.; Liu, E.; et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 2011, 118, 6050–6056. [Google Scholar] [CrossRef] [PubMed]
  91. Maude, S.L.; Frey, N.; Shaw, P.A.; Aplenc, R.; Barrett, D.M.; Bunin, N.J.; Chew, A.; Gonzalez, V.E.; Zheng, Z.; Lacey, S.F.; et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 2014, 371, 1507–1517. [Google Scholar] [CrossRef] [PubMed]
  92. Porter, D.L.; Hwang, W.T.; Frey, N.V.; Lacey, S.F.; Shaw, P.A.; Loren, A.W.; Bagg, A.; Marcucci, K.T.; Shen, A.; Gonzalez, V.; et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci. Transl. Med. 2015, 7, 303ra139. [Google Scholar] [CrossRef] [PubMed]
  93. Brown, C.E.; Alizadeh, D.; Starr, R.; Weng, L.; Wagner, J.R.; Naranjo, A.; Ostberg, J.R.; Blanchard, M.S.; Kilpatrick, J.; Simpson, J.; et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N. Engl. J. Med. 2016, 375, 2561–2569. [Google Scholar] [CrossRef] [PubMed]
  94. Ahmed, N.; Brawley, V.; Hegde, M.; Bielamowicz, K.; Kalra, M.; Landi, D.; Robertson, C.; Gray, T.L.; Diouf, O.; Wakefield, A.; et al. HER2-Specific Chimeric Antigen Receptor-Modified Virus-Specific T Cells for Progressive Glioblastoma: A Phase 1 Dose-Escalation Trial. JAMA Oncol. 2017, 3, 1094–1101. [Google Scholar] [CrossRef] [PubMed]
  95. Hegde, M.; Mukherjee, M.; Grada, Z.; Pignata, A.; Landi, D.; Navai, S.A.; Wakefield, A.; Fousek, K.; Bielamowicz, K.; Chow, K.K.; et al. Tandem CAR T cells targeting HER2 and IL13Ralpha2 mitigate tumor antigen escape. J. Clin. Investig. 2016, 126, 3036–3052. [Google Scholar] [CrossRef] [PubMed]
  96. Di Rosa, M.; Sanfilippo, C.; Libra, M.; Musumeci, G.; Malaguarnera, L. Different pediatric brain tumors are associated with different gene expression profiling. Acta Histochem. 2015, 117, 477–485. [Google Scholar] [CrossRef] [PubMed]
  97. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Sayour, E.J.; Mitchell, D.A. Immunotherapy for Pediatric Brain Tumors. Brain Sci. 2017, 7, 137. https://doi.org/10.3390/brainsci7100137

AMA Style

Sayour EJ, Mitchell DA. Immunotherapy for Pediatric Brain Tumors. Brain Sciences. 2017; 7(10):137. https://doi.org/10.3390/brainsci7100137

Chicago/Turabian Style

Sayour, Elias J., and Duane A. Mitchell. 2017. "Immunotherapy for Pediatric Brain Tumors" Brain Sciences 7, no. 10: 137. https://doi.org/10.3390/brainsci7100137

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop