51
|
Gilboa E. A quantum leap in cancer vaccines? J Immunother Cancer 2016; 4:87. [PMID: 28018600 PMCID: PMC5168708 DOI: 10.1186/s40425-016-0192-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 11/11/2016] [Indexed: 11/10/2022] Open
Affiliation(s)
- Eli Gilboa
- University of Miami School of Medicine, Miami, USA
| |
Collapse
|
52
|
Pastor F. Aptamers: A New Technological Platform in Cancer Immunotherapy. Pharmaceuticals (Basel) 2016; 9:E64. [PMID: 27783034 PMCID: PMC5198039 DOI: 10.3390/ph9040064] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 09/29/2016] [Accepted: 10/19/2016] [Indexed: 12/12/2022] Open
Abstract
The renaissance of cancer immunotherapy is, nowadays, a reality. In the near future, it will be very likely among the first-line treatments for cancer patients. There are several different approaches to modulate the immune system to fight against tumor maladies but, so far, monoclonal antibodies may currently be the most successful immuno-tools used to that end. The number of ongoing clinical trials with monoclonal antibodies has been increasing exponentially over the last few years upon the Food and Drug Administration (FDA) approval of the first immune-checkpoint blockade antibodies. In spite of the proved antitumor effect of these reagents, the unleashing of the immune system to fight cancer cells has a cost, namely auto-inflammatory toxicity. Additionally, only a small fraction of all patients treated with immune-checkpoint antibodies have a clinical benefit. Taking into account all this, it is urgent new therapeutic reagents are developed with a contained toxicity that could facilitate the combination of different immune-modulating pathways to broaden the antitumor effect in most cancer patients. Based on preclinical data, oligonucleotide aptamers could fulfill this need. Aptamers have not only been successfully used as antagonists of immune-checkpoint receptors, but also as agonists of immunostimulatory receptors in cancer immunotherapy. The simplicity of aptamers to be engineered for the specific delivery of different types of cargos to tumor cells and immune cells so as to harvest an efficient antitumor immune response gives aptamers a significant advantage over antibodies. In this review all of the recent applications of aptamers in cancer immunotherapy will be described.
Collapse
Affiliation(s)
- Fernando Pastor
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
- Program of Molecular Therapies, Aptamer Unit, Centro de Investigación Medica Aplicada (CIMA), Pamplona 31008, Spain.
| |
Collapse
|
53
|
Gilboa E, Berezhnoy A, Schrand B. Reducing Toxicity of Immune Therapy Using Aptamer-Targeted Drug Delivery. Cancer Immunol Res 2016; 3:1195-200. [PMID: 26541880 DOI: 10.1158/2326-6066.cir-15-0194] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Modulating the function of immune receptors with antibodies is ushering in a new era in cancer immunotherapy. With the notable exception of PD-1 blockade used as monotherapy, immune modulation can be associated with significant toxicities that are expected to escalate with the development of increasingly potent immune therapies. A general way to reduce toxicity is to target immune potentiating drugs to the tumor or immune cells of the patient. This Crossroads article discusses a new class of nucleic acid-based immune-modulatory drugs that are targeted to the tumor or to the immune system by conjugation to oligonucleotide aptamer ligands. Cell-free chemically synthesized short oligonucleotide aptamers represent a novel and emerging platform technology for generating ligands with desired specificity that offer exceptional versatility and feasibility in terms of development, manufacture, and conjugation to an oligonucleotide cargo. In proof-of-concept studies, aptamer ligands were used to target immune-modulatory siRNAs or aptamers to induce neoantigens in the tumor cells, limit costimulation to the tumor lesion, or enhance the persistence of vaccine-induced immunity. Using increasingly relevant murine models, the aptamer-targeted immune-modulatory drugs engendered protective antitumor immunity that was superior to that of current "gold-standard" therapies in terms of efficacy and lack of toxicity or reduced toxicity. To overcome immune exhaustion aptamer-targeted siRNA conjugates could be used to downregulate intracellular mediators of exhaustion that integrate signals from multiple inhibitory receptors. Recent advances in aptamer development and second-generation aptamer-drug conjugates suggest that we have only scratched the surface.
Collapse
Affiliation(s)
- Eli Gilboa
- Department of Microbiology and Immunology, Dodson Interdisciplinary Immunotherapy Institute, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida.
| | - Alexey Berezhnoy
- Department of Microbiology and Immunology, Dodson Interdisciplinary Immunotherapy Institute, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Brett Schrand
- Department of Microbiology and Immunology, Dodson Interdisciplinary Immunotherapy Institute, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| |
Collapse
|
54
|
Toy R, Roy K. Engineering nanoparticles to overcome barriers to immunotherapy. Bioeng Transl Med 2016; 1:47-62. [PMID: 29313006 PMCID: PMC5689503 DOI: 10.1002/btm2.10005] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/14/2016] [Accepted: 04/15/2016] [Indexed: 12/14/2022] Open
Abstract
Advances in immunotherapy have led to the development of a variety of promising therapeutics, including small molecules, proteins and peptides, monoclonal antibodies, and cellular therapies. Despite this wealth of new therapeutics, the efficacy of immunotherapy has been limited by challenges in targeted delivery and controlled release, that is, spatial and temporal control on delivery. Particulate carriers, especially nanoparticles have been widely studied in drug delivery and vaccine research and are being increasingly investigated as vehicles to deliver immunotherapies. Nanoparticle-mediated drug delivery could provide several benefits, including control of biodistribution and transport kinetics, the potential for site-specific targeting, immunogenicity, tracking capability using medical imaging, and multitherapeutic loading. There are also a unique set of challenges, which include nonspecific uptake by phagocytic cells, off-target biodistribution, permeation through tissue (transport limitation), nonspecific immune-activation, and poor control over intracellular localization. This review highlights the importance of understanding the relationship between a nanoparticle's size, shape, charge, ligand density and elasticity to its vascular transport, biodistribution, cellular internalization, and immunogenicity. For the design of an effective immunotherapy, we highlight the importance of selecting a nanoparticle's physical characteristics (e.g., size, shape, elasticity) and its surface functionalization (e.g., chemical or polymer modifications, targeting or tissue-penetrating peptides) with consideration of its reactivity to the targeted microenvironment (e.g., targeted cell types, use of stimuli-sensitive biomaterials, immunogenicity). Applications of this rational nanoparticle design process in vaccine development and cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Randall Toy
- Wallace H. Coulter Dept. of Biomedical Engineering Georgia Institute of Technology, and Emory University Atlanta GA 30332
| | - Krishnendu Roy
- Wallace H. Coulter Dept. of Biomedical Engineering Georgia Institute of Technology, and Emory University Atlanta GA 30332
| |
Collapse
|
55
|
Lieberman NAP, Moyes KW, Crane CA. Developing immunotherapeutic strategies to target brain tumors. Expert Rev Anticancer Ther 2016; 16:775-88. [PMID: 27253692 DOI: 10.1080/14737140.2016.1192470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Recent years have seen rapid growth in cancer treatments that enhance the anti-tumor activities of the immune system. Collectively known as immunotherapy, modulation of the immune system has shown success treating some hematological malignancies, but has yet to be successfully applied to the treatment of patients with brain tumors. AREAS COVERED This review highlights mechanistic insights from murine studies and compiled recent clinical trial data, focusing on the most aggressive brain tumor, glioblastoma (GBM). The field has recently accumulated a critical mass of data, and we discuss past treatment failures in the context of newly developed approaches now entering clinical trials. This article provides an overview of the immunotherapeutic armamentarium currently in development for the treatment of patients with GBM, who are in dire need of safe and effective therapies. Expert commentary: Themes that emerge include the importance of mitigating the effects of an immunosuppressive tumor microenvironment and the potential for innate immune cell activation to enhance cytotoxic anti-tumor activity. Consideration of these studies as a collective may inform the design of new immunotherapies, as well as the immune monitoring protocols for patients participating in clinical trials.
Collapse
Affiliation(s)
- Nicole A P Lieberman
- a Seattle Children's Research Institute, Ben Towne Center for Childhood Cancer Research , Seattle , WA , USA
| | - Kara White Moyes
- a Seattle Children's Research Institute, Ben Towne Center for Childhood Cancer Research , Seattle , WA , USA
| | - Courtney A Crane
- a Seattle Children's Research Institute, Ben Towne Center for Childhood Cancer Research , Seattle , WA , USA.,b Department of Neurological Surgery , University of Washington School of Medicine , Seattle , WA , USA
| |
Collapse
|
56
|
Hodges TR, Ferguson SD, Heimberger AB. Immunotherapy in glioblastoma: emerging options in precision medicine. CNS Oncol 2016; 5:175-86. [PMID: 27225028 DOI: 10.2217/cns-2016-0009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy for glioblastoma (GBM) provides a unique opportunity for targeted therapies for each patient, addressing individual variability in genes, tumor biomarkers and clinical profile. As immunotherapy has the potential to specifically target tumor cells with minimal risk to normal tissue, several immunotherapeutic strategies are currently being evaluated in clinical trials in GBM. With the Precision Medicine Initiative being announced in the President's State of the Union Address in 2016, GBM immunotherapy provides a useful platform for changing the landscape in treating patients with difficult disease.
Collapse
Affiliation(s)
- Tiffany R Hodges
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sherise D Ferguson
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
57
|
Benaduce AP, Brenneman R, Schrand B, Pollack A, Gilboa E, Ishkanian A. 4-1BB Aptamer-Based Immunomodulation Enhances the Therapeutic Index of Radiation Therapy in Murine Tumor Models. Int J Radiat Oncol Biol Phys 2016; 96:458-461. [PMID: 27598810 DOI: 10.1016/j.ijrobp.2016.05.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/31/2016] [Accepted: 05/13/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE To report a novel strategy using oligonucleotide aptamers to 4-1BB as an alternate method for costimulation, and show that combinatorial therapy with radiation improves the therapeutic ratio over equivalent monoclonal antibodies. METHODS AND MATERIALS Subcutaneous 4T1 (mouse mammary carcinoma) tumors were established (approximately 100 mm(3)), and a radiation therapy (RT) dose/fractionation schedule that optimally synergizes with 4-1BB monoclonal antibody (mAb) was identified. Comparable tumor control and animal survival was observed when either 4-1BB antibody or aptamer were combined with RT using models of breast cancer and melanoma (4T1 and B16-F10). Off-target CD8(+) T-cell toxicity was evaluated by quantification of CD8(+) T cells in livers and spleens of treated animals. RESULTS When combined with 4-1BB mAb, significant differences in tumor control were observed by varying RT dose and fractionation schedules. Optimal synergy between RT and 4-1BB mAb was observed at 5 Gy × 6. Testing 4-1BB mAb and aptamer independently using the optimal RT (5 Gy × 6 for 4T1/Balb/c and 12 Gy × 1 for B16/C57BL6J mouse models) revealed equivalent tumor control using 4-1BB aptamer and 4-1BB mAb. 4-1BB mAb, but not 4-1BB aptamer-treated animals, exhibited increased lymphocytic liver infiltrates and increased splenic and liver CD8(+) T cells. CONCLUSIONS Radiation therapy synergizes with 4-1BB mAb, and this effect is dependent on RT dose and fractionation. Tumor control by 4-1BB aptamer is equivalent to 4-1BB mAb when combined with optimal RT dose, without eliciting off-target liver and spleen CD8(+) expansion. 4-1BB aptamer-based costimulation affords a comparable and less toxic strategy to augment RT-mediated tumor control.
Collapse
Affiliation(s)
- Ana Paula Benaduce
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Randall Brenneman
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Brett Schrand
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Alan Pollack
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Eli Gilboa
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Adrian Ishkanian
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida.
| |
Collapse
|
58
|
Patel MA, Kim JE, Theodros D, Tam A, Velarde E, Kochel CM, Francica B, Nirschl TR, Ghasemzadeh A, Mathios D, Harris-Bookman S, Jackson CC, Jackson C, Ye X, Tran PT, Tyler B, Coric V, Selby M, Brem H, Drake CG, Pardoll DM, Lim M. Agonist anti-GITR monoclonal antibody and stereotactic radiation induce immune-mediated survival advantage in murine intracranial glioma. J Immunother Cancer 2016; 4:28. [PMID: 27190629 PMCID: PMC4869343 DOI: 10.1186/s40425-016-0132-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 04/26/2016] [Indexed: 12/29/2022] Open
Abstract
Background Glioblastoma (GBM) is a poorly immunogenic neoplasm treated with focused radiation. Immunotherapy has demonstrated synergistic survival effects with stereotactic radiosurgery (SRS) in murine GBM. GITR is a co-stimulatory molecule expressed constitutively on regulatory T-cells and by effector T-cells upon activation. We tested the hypothesis that anti-GITR monoclonal antibody (mAb) and SRS together would confer an immune-mediated survival benefit in glioma using the orthotopic GL261 glioma model. Methods Mice received SRS and anti-GITR 10 days after implantation. The anti-GITR mAbs tested were formatted as mouse IgG1 D265A (anti-GITR (1)) and IgG2a (anti-GITR (2a)) isotypes. Mice were randomized to four treatment groups: (1) control; (2) SRS; (3) anti-GITR; (4) anti-GITR/SRS. SRS was delivered to the tumor in one fraction, and mice were treated with mAb thrice. Mice were euthanized on day 21 to analyze the immunologic profile of tumor, spleen, and tumor draining lymph nodes. Results Anti-GITR (1)/SRS significantly improved survival over either treatment alone (p < .0001) with a cure rate of 24 % versus 0 % in a T-lymphocyte-dependent manner. There was elevated intratumoral CD4+ effector cell infiltration relative to Treg infiltration in mice treated with anti-GITR (1)/SRS, as well as significantly elevated IFNγ and IL-2 production by CD4+ T-cells and elevated IFNγ and TNFα production by CD8+ T-cells. There was increased mRNA expression of M1 markers and decreased expression of M2 markers in tumor infiltrating mononuclear cells. The anti-GITR (2a)/SRS combination did not improve survival, induce tumor regression, or result in Treg depletion. Conclusions These findings provide preclinical evidence for the use of anti-GITR (1) non-depleting antibodies in combination with SRS in GBM. Electronic supplementary material The online version of this article (doi:10.1186/s40425-016-0132-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mira A Patel
- The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Jennifer E Kim
- The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Debebe Theodros
- The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Ada Tam
- Department of Oncology, Baltimore, USA
| | | | | | | | | | | | - Dimitrios Mathios
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | - Sarah Harris-Bookman
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | - Christopher C Jackson
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | - Christina Jackson
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | - Xiaobu Ye
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | - Phuoc T Tran
- Department of Oncology, Baltimore, USA.,Department Radiation Oncology, Baltimore, USA.,and the Brady Urological Institute, Baltimore, USA
| | - Betty Tyler
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | | | - Mark Selby
- Bristol-Myers Squibb Company, San Francisco, CA USA
| | - Henry Brem
- The Johns Hopkins University School of Medicine, Baltimore, USA.,Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | | | | | - Michael Lim
- The Johns Hopkins University School of Medicine, Baltimore, USA.,Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| |
Collapse
|
59
|
Dharmadhikari B, Wu M, Abdullah NS, Rajendran S, Ishak ND, Nickles E, Harfuddin Z, Schwarz H. CD137 and CD137L signals are main drivers of type 1, cell-mediated immune responses. Oncoimmunology 2016; 5:e1113367. [PMID: 27141396 PMCID: PMC4839363 DOI: 10.1080/2162402x.2015.1113367] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/22/2015] [Accepted: 10/22/2015] [Indexed: 10/22/2022] Open
Abstract
CD137 is expressed on activated T cells and NK cells, among others, and is a potent co-stimulator of antitumor immune responses. CD137 ligand (CD137L) is expressed by antigen presenting cells (APC), and CD137L reverse signaling into APC enhances their activity. CD137-CD137L interactions as main driver of type 1, cell-mediated immune responses explains the puzzling observation that CD137 agonists which enhance antitumor immune responses also ameliorate autoimmune diseases. Upon co-stimulation by CD137, Th1 CD4+ T cells together with Tc1 CD8+ T cells and NK cells inhibit other T cell subsets, thereby promoting antitumor responses and mitigating non-type 1 auto-immune diseases.
Collapse
Affiliation(s)
- Bhushan Dharmadhikari
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
| | - Meihui Wu
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
| | - Nur Sharalyn Abdullah
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
| | - Sakthi Rajendran
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
| | - Nur Diana Ishak
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
| | - Emily Nickles
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
| | - Zulkarnain Harfuddin
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
60
|
Tabarzad M, Jafari M. Trends in the Design and Development of Specific Aptamers Against Peptides and Proteins. Protein J 2016; 35:81-99. [PMID: 26984473 DOI: 10.1007/s10930-016-9653-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Aptamers are single stranded oligonucleotides, comparable to monoclonal antibodies (mAbs) in selectivity and affinity and have significant strategic properties in design, development and applications more than mAbs. Ease of design and development, simple chemical modification and the attachment of functional groups, easily handling and more adaptability with analytical methods, small size and adaptation with nanostructures are the valuable characteristics of aptamers in comparison to large protein based ligands. Among a broad range of targets that their specific aptamers developed, proteins and peptides have significant position according to the number of related studies performed so far. Since proteins control many of important physiological and pathological incidents in the living organisms, particularly human beings and because of the benefits of aptamers in clinical and analytical applications, aptamer related technologies in the field of proteins and peptides are under progress, exclusively. Currently, there is only one FDA approved therapeutic aptamer in the pharmaceutical market, which is specific to vascular endothelial growth factor and is prescribed for age related macular degenerative disease. Additionally, there are several aptamers in the different phases of clinical trials. Almost all of these aptamers are specific to clinically important peptide or protein targets. In addition, the application of protein specific aptamers in the design and development of targeted drug delivery systems and diagnostic biosensors is another interesting field of aptamer technology. In this review, significant efforts related to development and applications of aptamer technologies in proteins and peptides sciences were considered to emphasis on the importance of aptamers in medicinal and clinical applications.
Collapse
Affiliation(s)
- Maryam Tabarzad
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Vali Asr Avenue, Niayesh Junction, Tehran, PO Box: 14155-6153, Iran.
| | - Marzieh Jafari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
61
|
Abstract
Aptamers are chemically synthesized oligonucleotides that can be easily engineered for cancer immunotherapy use. So far, most of the therapeutic aptamers described are antagonistic and block the function of a receptor or its soluble ligand. Recently, aptamers have been modified to act as agonists by multimerization, with a direct application in cancer immunotherapy. Several agonistic aptamers against costimulatory receptors have been described. However, systemic costimulation, though potentially a very potent antitumor immune strategy, is not devoid of auto-inflammatory side effects. In a quest to reduce toxicity and improve efficacy – reducing the therapeutic index – the first bi-specific aptamers to target the costimulatory ligand to the tumor have been described, showing very promising results in different preclinical tumor models.
Collapse
Affiliation(s)
- Fernando Pastor
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Pamplona 31008, Spain.,Program of Molecular Therapies, Aptamer Unit, Centro de Investigación Medica Aplicada (CIMA), Pamplona, 31008, Spain
| |
Collapse
|
62
|
Zhou G, Wilson G, Hebbard L, Duan W, Liddle C, George J, Qiao L. Aptamers: A promising chemical antibody for cancer therapy. Oncotarget 2016; 7:13446-13463. [PMID: 26863567 PMCID: PMC4924653 DOI: 10.18632/oncotarget.7178] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 01/24/2016] [Indexed: 12/20/2022] Open
Abstract
Aptamers, also known as chemical antibodies, are single-stranded nucleic acid oligonucleotides which bind to their targets with high specificity and affinity. They are typically selected by repetitive in vitro process termed systematic evolution of ligands by exponential enrichment (SELEX). Owing to their excellent properties compared to conventional antibodies, notably their smaller physical size and lower immunogenicity and toxicity, aptamers have recently emerged as a new class of agents to deliver therapeutic drugs to cancer cells by targeting specific cancer-associated hallmarks. Aptamers can also be structurally modified to make them more flexible in order to conjugate other agents such as nano-materials and therapeutic RNA agents, thus extending their applications for cancer therapy. This review presents the current knowledge on the practical applications of aptamers in the treatment of a variety of cancers.
Collapse
Affiliation(s)
- Gang Zhou
- Storr Liver Centre, Westmead Millennium Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, Australia
| | - George Wilson
- Storr Liver Centre, Westmead Millennium Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, Australia
| | - Lionel Hebbard
- Discipline of Molecular and Cell Biology, James Cook University, Townsville, QLD, Australia
| | - Wei Duan
- School of Medicine, Deakin University, Waurn Ponds, VIC, Australia
| | - Christopher Liddle
- Storr Liver Centre, Westmead Millennium Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Millennium Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, Australia
| | - Liang Qiao
- Storr Liver Centre, Westmead Millennium Institute for Medical Research, University of Sydney and Westmead Hospital, Westmead, NSW, Australia
| |
Collapse
|
63
|
Hodges TR, Ferguson SD, Caruso HG, Kohanbash G, Zhou S, Cloughesy TF, Berger MS, Poste GH, Khasraw M, Ba S, Jiang T, Mikkelson T, Yung WKA, de Groot JF, Fine H, Cantley LC, Mellinghoff IK, Mitchell DA, Okada H, Heimberger AB. Prioritization schema for immunotherapy clinical trials in glioblastoma. Oncoimmunology 2016; 5:e1145332. [PMID: 27471611 DOI: 10.1080/2162402x.2016.1145332] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/12/2016] [Accepted: 01/16/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Emerging immunotherapeutic strategies for the treatment of glioblastoma (GBM) such as dendritic cell (DC) vaccines, heat shock proteins, peptide vaccines, and adoptive T-cell therapeutics, to name a few, have transitioned from the bench to clinical trials. With upcoming strategies and developing therapeutics, it is challenging to critically evaluate the practical, clinical potential of individual approaches and to advise patients on the most promising clinical trials. METHODS The authors propose a system to prioritize such therapies in an organized and data-driven fashion. This schema is based on four categories of factors: antigenic target robustness, immune-activation and -effector responses, preclinical vetting, and early evidence of clinical response. Each of these categories is subdivided to focus on the most salient elements for developing a successful immunotherapeutic approach for GBM, and a numerical score is generated. RESULTS The Score Card reveals therapeutics that have the most robust data to support their use, provides a reference prioritization score, and can be applied in a reiterative fashion with emerging data. CONCLUSIONS The authors hope that this schema will give physicians an evidence-based and rational framework to make the best referral decisions to better guide and serve this patient population.
Collapse
Affiliation(s)
- Tiffany R Hodges
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Sherise D Ferguson
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Hillary G Caruso
- The Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Gary Kohanbash
- Department of Neurosurgery, the University of California at San Francisco , San Francisco, USA
| | - Shouhao Zhou
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Timothy F Cloughesy
- Department of Neuro-Oncology, the University of California at Los Angeles , Los Angeles, CA, USA
| | - Mitchel S Berger
- Department of Neurosurgery, the University of California at San Francisco , San Francisco, USA
| | | | | | - Sujuan Ba
- The National Foundation for Cancer Research, Bethesda, MD, USA, Asian Fund for Cancer Research , Hong Kong, People's Republic of China
| | - Tao Jiang
- Department of Neurosurgery, Tiantan Hospital, Capital Medical University , Beijing, China
| | - Tom Mikkelson
- Department of Neurosurgery, Henry Ford Health System , Detroit, MI, USA
| | - W K Alfred Yung
- Department of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - John F de Groot
- Department of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Howard Fine
- Division of Neuro-Oncology, Weill Cornell Medical College , New York, NY, USA
| | - Lewis C Cantley
- Department of Systems Biology, Harvard Medical School , Boston, MA, USA
| | - Ingo K Mellinghoff
- Department of Neurology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Duane A Mitchell
- Department of Neurosurgery, University of Florida , Gainesville, FL, USA
| | - Hideho Okada
- Department of Neurosurgery, the University of California at San Francisco , San Francisco, USA
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| |
Collapse
|
64
|
Abstract
Gliomas are the most common primary brain tumors of the central nervous system, and carry a grim prognosis. Novel approaches utilizing the immune system as adjuvant therapy are quickly emerging as viable and effective options. Immunotherapeutic strategies being investigated to treat glioblastoma include: vaccination therapy targeted against either specific tumor antigens or whole tumor lysate, adoptive cellular therapy with cytotoxic T lymphocytes, chimeric antigen receptors and bi-specific T-cell engaging antibodies allowing circumvention of major histocompatibility complex restriction, aptamer therapy with aims for more efficient target delivery, and checkpoint blockade in order to release the tumor-mediated inhibition of the immune system. Given the heterogeneity of glioblastoma and its ability to gain mutations throughout the disease course, multifaceted treatment strategies utilizing multiple forms of immunotherapy in combination with conventional therapy will be most likely to succeed moving forward.
Collapse
Affiliation(s)
- Brandon D Liebelt
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Houston Methodist Neurological Institute, Houston, TX, USA
| | - Gaetano Finocchiaro
- Department of Neuro-oncology, IRCCS Istituto Neurologico Besta, Milan, Italy
| | - Amy B Heimberger
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
65
|
Kong LY, Wei J, Fuller GN, Schrand B, Gabrusiewicz K, Zhou S, Rao G, Calin G, Gilboa E, Heimberger AB. Tipping a favorable CNS intratumoral immune response using immune stimulation combined with inhibition of tumor-mediated immune suppression. Oncoimmunology 2015; 5:e1117739. [PMID: 27467917 DOI: 10.1080/2162402x.2015.1117739] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 10/22/2022] Open
Abstract
High-grade gliomas are notoriously heterogeneous regarding antigen expression, effector responses, and immunosuppressive mechanisms. Therefore, combinational immune therapeutic approaches are more likely to impact a greater number of patients and result in longer, durable responses. We have previously demonstrated the monotherapeutic effects of miR-124, which inhibits the signal transducer and activator of transcription 3 (STAT3) immune suppressive pathway, and immune stimulatory 4-1BB aptamers against a variety of malignancies, including genetically engineered immune competent high-grade gliomas. To evaluate potential synergy, we tested an immune stimulatory aptamer together with microRNA-124 (miRNA-124), which blocks tumor-mediated immune suppression, and found survival to be markedly enhanced, including beyond that produced by monotherapy. The synergistic activity appeared to be not only secondary to enhanced CD3(+) cell numbers but also to reduced macrophage immune tumor trafficking, indicating that a greater therapeutic benefit can be achieved with approaches that both induce immune activation and inhibit tumor-mediated immune suppression within the central nervous system (CNS) tumors.
Collapse
Affiliation(s)
- Ling-Yuan Kong
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Jun Wei
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Gregory N Fuller
- Neuropathology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Brett Schrand
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine , Miami, FL, USA
| | - Konrad Gabrusiewicz
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Shouhao Zhou
- Biostatistics, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Ganesh Rao
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - George Calin
- Experimental Therapeutics, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Eli Gilboa
- Department of Microbiology & Immunology, University of Miami Miller School of Medicine , Miami, FL, USA
| | - Amy B Heimberger
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| |
Collapse
|
66
|
Tavaré R, Escuin-Ordinas H, Mok S, McCracken MN, Zettlitz KA, Salazar FB, Witte ON, Ribas A, Wu AM. An Effective Immuno-PET Imaging Method to Monitor CD8-Dependent Responses to Immunotherapy. Cancer Res 2015; 76:73-82. [PMID: 26573799 DOI: 10.1158/0008-5472.can-15-1707] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/16/2015] [Indexed: 12/31/2022]
Abstract
The rapidly advancing field of cancer immunotherapy is currently limited by the scarcity of noninvasive and quantitative technologies capable of monitoring the presence and abundance of CD8(+) T cells and other immune cell subsets. In this study, we describe the generation of (89)Zr-desferrioxamine-labeled anti-CD8 cys-diabody ((89)Zr-malDFO-169 cDb) for noninvasive immuno-PET tracking of endogenous CD8(+) T cells. We demonstrate that anti-CD8 immuno-PET is a sensitive tool for detecting changes in systemic and tumor-infiltrating CD8 expression in preclinical syngeneic tumor immunotherapy models including antigen-specific adoptive T-cell transfer, agonistic antibody therapy (anti-CD137/4-1BB), and checkpoint blockade antibody therapy (anti-PD-L1). The ability of anti-CD8 immuno-PET to provide whole body information regarding therapy-induced alterations of this dynamic T-cell population provides new opportunities to evaluate antitumor immune responses of immunotherapies currently being evaluated in the clinic.
Collapse
Affiliation(s)
- Richard Tavaré
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, California. Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California.
| | - Helena Escuin-Ordinas
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles, Los Angeles, California
| | - Stephen Mok
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California
| | - Melissa N McCracken
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California
| | - Kirstin A Zettlitz
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, California. Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California
| | - Felix B Salazar
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, California. Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California
| | - Owen N Witte
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California. Howard Hughes Medical Institute, University of California Los Angeles, Los Angeles, California. Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, California. Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, California
| | - Antoni Ribas
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California. Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California. Surgery, Division of Surgical Oncology, University of California Los Angeles, Los Angeles, California. Institute for Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Anna M Wu
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, California. Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California. Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California.
| |
Collapse
|
67
|
Manipulating the in vivo immune response by targeted gene knockdown. Curr Opin Immunol 2015; 35:63-72. [PMID: 26149459 DOI: 10.1016/j.coi.2015.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/09/2015] [Accepted: 06/19/2015] [Indexed: 02/06/2023]
Abstract
Aptamers, nucleic acids selected for high affinity binding to proteins, can be used to activate or antagonize immune mediators or receptors in a location and cell-type specific manner and to enhance antigen presentation. They can also be linked to other molecules (other aptamers, siRNAs or miRNAs, proteins, toxins) to produce multifunctional compounds for targeted immune modulation in vivo. Aptamer-siRNA chimeras (AsiCs) that induce efficient cell-specific knockdown in immune cells in vitro and in vivo can be used as an immunological research tool or potentially as an immunomodulating therapeutic.
Collapse
|
68
|
Sanmamed MF, Pastor F, Rodriguez A, Perez-Gracia JL, Rodriguez-Ruiz ME, Jure-Kunkel M, Melero I. Agonists of Co-stimulation in Cancer Immunotherapy Directed Against CD137, OX40, GITR, CD27, CD28, and ICOS. Semin Oncol 2015; 42:640-55. [PMID: 26320067 DOI: 10.1053/j.seminoncol.2015.05.014] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
T and natural killer (NK) lymphocytes are considered the main effector players in the immune response against tumors. Full activation of T and NK lymphocytes requires the coordinated participation of several surface receptors that meet their cognate ligands through structured transient cell-to-cell interactions known as immune synapses. In the case of T cells, the main route of stimulation is driven by antigens as recognized in the form of short polypeptides associated with major histocompatibility complex (MHC) antigen-presenting molecules. However, the functional outcome of T-cell stimulation towards clonal expansion and effector function acquisition is contingent on the contact of additional surface receptor-ligand pairs and on the actions of cytokines in the milieu. While some of those interactions are inhibitory, others are activating and are collectively termed co-stimulatory receptors. The best studied belong to either the immunoglobulin superfamily or the tumor necrosis factor-receptor (TNFR) family. Co-stimulatory receptors include surface moieties that are constitutively expressed on resting lymphocytes such as CD28 or CD27 and others whose expression is induced upon recent previous antigen priming, ie, CD137, GITR, OX40, and ICOS. Ligation of these glycoproteins with agonist antibodies actively conveys activating signals to the lymphocyte. Those signals, acting through a potentiation of the cellular immune response, give rise to anti-tumor effects in mouse models. Anti-CD137 antibodies are undergoing clinical trials with evidence of clinical activity and anti-OX40 monoclonal antibodies (mAbs) induce interesting immunomodulation effects in humans. Antibodies anti-CD27 and GITR have recently entered clinical trials. The inherent dangers of these immunomodulation strategies are the precipitation of excessive systemic inflammation or/and invigorating silent autoimmunity. Agonist antibodies, recombinant forms of the natural ligands, and polynucleotide-based aptamers constitute the pharmacologic tools to manipulate such receptors. Preclinical data suggest that the greatest potential of these agents is achieved in combined treatment strategies.
Collapse
Affiliation(s)
- Miguel F Sanmamed
- Department of Immunobiology, Yale School of Medicine, New Haven, CT.
| | - Fernando Pastor
- Centro de investigación médica aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Alfonso Rodriguez
- Centro de investigación médica aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | | | | | | | - Ignacio Melero
- Centro de investigación médica aplicada (CIMA), Universidad de Navarra, Pamplona, Spain; Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
69
|
Bartkowiak T, Curran MA. 4-1BB Agonists: Multi-Potent Potentiators of Tumor Immunity. Front Oncol 2015; 5:117. [PMID: 26106583 PMCID: PMC4459101 DOI: 10.3389/fonc.2015.00117] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/11/2015] [Indexed: 01/12/2023] Open
Abstract
Immunotherapy is a rapidly expanding field of oncology aimed at targeting, not the tumor itself, but the immune system combating the cancerous lesion. Of the many approaches currently under study to boost anti-tumor immune responses; modulation of immune co-receptors on lymphocytes in the tumor microenvironment has thus far proven to be the most effective. Antibody blockade of the T cell co-inhibitory receptor cytotoxic T lymphocyte antigen-4 (CTLA-4) has become the first FDA approved immune checkpoint blockade; however, tumor infiltrating lymphocytes express a diverse array of additional stimulatory and inhibitory co-receptors, which can be targeted to boost tumor immunity. Among these, the co-stimulatory receptor 4-1BB (CD137/TNFSF9) possesses an unequaled capacity for both activation and pro-inflammatory polarization of anti-tumor lymphocytes. While functional studies of 4-1BB have focused on its prominent role in augmenting cytotoxic CD8 T cells, 4-1BB can also modulate the activity of CD4 T cells, B cells, natural killer cells, monocytes, macrophages, and dendritic cells. 4-1BB’s expression on both T cells and antigen presenting cells, coupled with its capacity to promote survival, expansion, and enhanced effector function of activated T cells, has made it an alluring target for tumor immunotherapy. In contrast to immune checkpoint blocking antibodies, 4-1BB agonists can both potentiate anti-tumor and anti-viral immunity, while at the same time ameliorating autoimmune disease. Despite this, 4-1BB agonists can trigger high grade liver inflammation which has slowed their clinical development. In this review, we discuss how the underlying immunobiology of 4-1BB activation suggests the potential for therapeutically synergistic combination strategies in which immune adverse events can be minimized.
Collapse
Affiliation(s)
- Todd Bartkowiak
- Department of Immunology, University of Texas MD Anderson Cancer Center , Houston, TX , USA ; The University of Texas Graduate School of Biomedical Sciences at Houston , Houston, TX , USA
| | - Michael A Curran
- Department of Immunology, University of Texas MD Anderson Cancer Center , Houston, TX , USA ; The University of Texas Graduate School of Biomedical Sciences at Houston , Houston, TX , USA
| |
Collapse
|
70
|
Schrand B, Berezhnoy A, Brenneman R, Williams A, Levay A, Gilboa E. Reducing toxicity of 4-1BB costimulation: targeting 4-1BB ligands to the tumor stroma with bi-specific aptamer conjugates. Oncoimmunology 2015; 4:e970918. [PMID: 25949891 DOI: 10.4161/21624011.2014.970918] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 09/25/2014] [Indexed: 11/19/2022] Open
Abstract
Systemic administration of immune modulatory antibodies to cancer patients is associated with autoimmune pathologies. We have developed a clinically feasible and broadly applicable approach to limit immune stimulation to disseminated tumor lesions using a bi-specific agonistic 4-1BB oligonucleotide aptamer targeted to a broadly expressed stromal product (e.g., VEGF or osteopontin). The stroma-targeted aptamer conjugates engendered potent antitumor immunity against unrelated tumors and exhibited a superior therapeutic index compared to non-targeted agonistic 4-1BB antibody.
Collapse
Affiliation(s)
- B Schrand
- Department of Microbiology & Immunology; Dodson Interdisciplinary Immunotherapy Institute; Sylvester Comprehensive Cancer Center; University of Miami ; Miami, FL, USA
| | - A Berezhnoy
- Department of Microbiology & Immunology; Dodson Interdisciplinary Immunotherapy Institute; Sylvester Comprehensive Cancer Center; University of Miami ; Miami, FL, USA
| | - R Brenneman
- Department of Microbiology & Immunology; Dodson Interdisciplinary Immunotherapy Institute; Sylvester Comprehensive Cancer Center; University of Miami ; Miami, FL, USA
| | - A Williams
- Department of Pathology; Miller School of Medicine; University of Miami ; Miami, FL, USA
| | - A Levay
- Department of Microbiology & Immunology; Dodson Interdisciplinary Immunotherapy Institute; Sylvester Comprehensive Cancer Center; University of Miami ; Miami, FL, USA
| | - E Gilboa
- Department of Microbiology & Immunology; Dodson Interdisciplinary Immunotherapy Institute; Sylvester Comprehensive Cancer Center; University of Miami ; Miami, FL, USA
| |
Collapse
|