Cancer immunotherapy via nucleic acid aptamers

https://doi.org/10.1016/j.intimp.2015.10.013Get rights and content

Highlights

  • Addressed progress in cancer immunotherapy with nucleic acid aptamer.

  • Highlighted recent developments; in immune system targeting and in methods of aptamers immunotherapy.

  • We discussed aptamers used as therapeutic agent for cancer.

  • We suggested some ways to overcome aptamer limitations.

Abstract

Over the past decade, immune therapy has become a standard treatment for a variety of cancers. Monoclonal antibodies, immune adjuvants and vaccines against oncogenic viruses are now well-established cancer therapies. Immune modulation is a principal element of supportive care for many high-dose chemotherapy regimens. Aptamers are short nucleic acids that bind to defined targets with high affinity and specificity. The first aptamers have been selected around two decades ago by an in vitro process named SELEX (systematic evolution of ligands by exponential enrichment). Since then, numerous aptamers with specificities for a variety of targets from small molecules to proteins or even whole cells have been selected. Targeting immunomodulatory ligands in the progressive tumor lesions of the patients would be prophylactic or therapeutic and may reduce drug-associated toxicities. A new class of inhibitory and agonistic ligands composed of short oligonucleotide (ODN) aptamers was developed recently that exhibited bioactivities comparable or superior to that of antibodies. This paper addressed progress in cancer immunotherapy with nucleic acid aptamers and highlighted recent developments either in immune system targeting or in immunotherapy methods involved aptamers. We discussed aptamer limitations when used as therapeutic agents for cancer treatment and suggested ways to overcome those limitations.

Introduction

There are established therapies employing a variety of manipulations to activate antitumor immunity. One type constitutes passive immunization with monoclonal antibodies, the introduction of adjuvants into the tumor microenvironment, and the systemic injection of cytokines. However, active transfer of antigen-loaded dendritic cells, tumor-activated T cell, tumor antigens formulated with either adjuvant or included in different delivery systems comprise second approach. Currently, immune therapy has become a standard treatment for a variety of cancers, and in some instances, could be considered as a replacement for nonresponsive cases to chemotherapy and other traditional strategies for cancer treatment [1].

Investigational immune therapies for cancer, involve devising more efficacious and less toxic cancer therapies upon established treatment regimens. A wide variety of novel strategies have been developed based on our understanding of the interactions between tumors and the immune system. Collectively, these strategies attempt to augment protective antitumor immunity and to disrupt the immune regulatory circuits that are critical for maintaining tumor tolerance [1]. The immune system has three basic roles in the prevention of tumors. First, it can protect the host from virus-induced tumors by eliminating or suppressing viral infections. Second is the timely and effective elimination of pathogens and prompt clearance of inflammation preventing the establishment of an inflammatory microenvironment conducive to tumorigenesis. Finally, some tumor cell express some antigens including tumor-associated antigens (TAS) and tumor specific antigens (TSA) which immune system can specifically identify them resulting in the elimination of tumor cells [2].

Aptamers are short nucleic acids (almost 12–80 nucleotides long) capable of specific and tight binding to their target molecules. The term aptamer is originated from the Latin word aptus (to fit) and the Greek word meros (part) [3]. Aptamer selection or isolation was accomplished by a process called SELEX (systematic evolution of ligands by exponential enrichment), which was first applied independently by Ellington and Szostak [4], Tuerk and Gold [5], and Robertson and Joyce [6] in 1990. A typical SELEX experiment starts with a library of up to 1015 random oligonucleotide sequences which can be DNA, RNA, or modified RNA (e.g., 2-OMe or 2-F modified RNA). Some members of this rich library have strong affinity for binding to a desired target. Numerous aptamers (also termed Oligonucleotide Aptamer Ligands), have been developed during the past 20 years which can bind and inhibit the activity of many proteins. The concept that nucleic acid ligands could modulate the activity of proteins emerged from basic studies of viruses and early works in the field of gene therapy [7]. Research on HIV and adenoviruses in the 1980s, discovered that these viruses encode a number of small-structured RNAs that bind to viral or cellular proteins with high affinity and specificity. For example, the human immunodeficiency virus has evolved a short-structured RNA ligand (TAR). The HIV TAR element binds to the viral proteins such as tat, as well as the cellular protein cyclin T1. HIV uses the TAR element to control viral gene expression and replication. Adenovirus has evolved a short-structured RNA aptamer, termed VA-RNA, which inhibits interferon-induced PKR activity and thus blocks one of the antiviral strategies employed by mammalian cell's [7]. In the late 1980s, the observation that viruses utilize RNA ligands in their biological activities particularly for escaping from immune-responses suggested that RNA ligands might also be useful for therapeutic and diagnostic purposes. The first study in 1990 indicated that CD4 + T cells containing the TAR aptamer were highly resistant to viral replication and cytotoxicity. Thus, for the first time in these studies, it was demonstrated that nucleic acid aptamers could be used as an agent to directly bind and inhibit the activity of proteins, suggesting possible clinical outcome [8]. This approach has been widely used during the past two decades to generate RNA ligands for many proteins. In 2003, Gilboa et al. demonstrated that aptamers can be employed as a potential therapeutic agent for cancer treatment by targeting immune-receptors described below. His work offered a new strategy for cancer immunotherapy [9].

In this review, we describe aptamers which are implicated in the treatment of cancer, particularly those which target proteins involved in tumor immunotherapy thereby preventing tumor immune-escaping (Table 1). Aptamers are not only a new and promising alternative to antibodies in tumor diagnostics, but also can be used in direct tumor immunotherapy and delivery systems. Aptamer can be used directly for either therapeutic applications or delivering drug molecules to disease-related cells or tissues of interest thereby minimizing the exposure of these possibly harmful agents to surrounding healthy tissues (decrease bystander and adverse effect of the main drug). Aptamer immunotherapy can replace monoclonal antibody therapy. There are some promising examples of cancer antibody therapy which potentially can be replaced by aptamer therapy. Ipilumimab is a FDA approved antibody that blocks the inhibitory action of CTLA-4 [10], [11], and clinical trials targeting 4-1BB and PD-1 or PD-L1, have demonstrated the therapeutic potential of using immunomodulatory antibodies to stimulate protective immunity in human patients [12]. Nonetheless, systemic administration of immunomodulatory antibodies has been associated with dose-limiting autoimmune pathologies, conceivably reflecting also the activation of resident auto-reactive T cells [8]. Aptamers have notable advantages over antibodies which include, a) as a cell-free chemical synthesis is used for aptamer selection, they are more simple and cost-effective in comparison to cell-based products like antibodies, b) conjugation of aptamers to other entities such as toxins or drug carriers is easier compared to antibodies, and c) antibody-based therapeutic agents have at least one foreign motif which may promote immune response in receiving hosts, whereas aptamers due to their nucleic acid nature and size, initiate less or no immune response. However, translating aptamers into clinic faces some major limitations such as fast renal clearance and digestion by serum nuclease [13] which are explained in the next section.

In traditional approach of cancer therapy by aptamers, they were mainly developed for targeting tumor markers. These include aptamers targeting MUC-1 (CD227) [14], prostate specific membrane antigen (PSMA) [15], human epidermal growth factor receptor-3 (HER3), tenascin-c [14], [16] and some other receptors [17]. Among these, only aptamers specific for HER3 as one of the tumor markers, exhibited antagonistic activity [2]. Nowadays, aptamers are not only considered as a new and promising alternative to antibodies in tumor diagnostics tests, but also they can be used in direct tumor immunotherapy and drug delivery system [18]. There are several advantages for aptamers which make them suitable alternative to mAbs in clinical applications. Aptamers are not recognized by the body's immune system as foreign, and do not evoke a negative immune response. Although aptamers are small molecules and they are subject to kidney filtration, resulting in shortened half-lives but further chemical modifications to overcome the susceptibility to kidney filtration can be easily introduced during synthesis. Tumor cells are different quantitatively and qualitatively from healthy cells in their protein repertoire and surface antigens making them suitable for targeting by antibodies and aptamers [19]. SELEX is performed using two approaches including ligand or specific molecule SELEX and cell SELEX in which surface molecules on whole cell are targeted. Cell SELEX can be employed for the generation of aptamers against known or unknown target proteins displayed on live cells. Cell SELEX is a particularly promising selection strategy for various applications such as cancer research, cancer diagnosis and therapy. Highly specific aptamers can be developed against tumor cells differentiated from normal cells. Different cancer cells have been used in this process, and as a result, a significant number of aptamers have been generated for most cancers studied. Cell SELEX and its clinical application were well discussed in other reviews [20], [21].

Extracellular targets are divided in two types including, a) secretory targets such as antibodies, cytokines, tumor secreted immunosuppressive proteins (Fig. 1) and b) cell surface targets such as co-stimulatory molecules, adhesive molecules, tumor immunosuppressive induction molecules, activation and inhibitory receptors (for agonist and antagonist targeting) on T cells, NK cells, macrophages and dendritic cells. In the case of extracellular targets, aptamer stability becomes a major issue. Therefore, stabilized RNA (e.g., by 2-O-methyl or 2-fluoro modifications) or DNA is preferable when extracellular proteins are targeted [20]. Even then, degradation and renal clearance is inevitable, and repeated administration will be required until treatment is complete. In this paper, some strategies have been suggested to overcome rapid aptamer clearance and degradation. On the other hand, immunotropism aptamers can be classified into three categories on the basis of their application including (i) agonistic or (ii) antagonistic activity on target protein and (iii) aptamer as a drug delivery agent [22]. Two excellent reports have explained use of oligonucleotide aptamer ligands to modulate the function of immune receptors [15], [23].

Section snippets

Methods used for developing immunotherapeutic aptamers and technical challenges involved

Methods used for the isolation of potential aptamers to specifically target the immune system are dependent on the type of target and the purpose for which aptamer is developed. Collectively, target cells or proteins are incubated with a complex library consisting of up to 1015 individual single stranded oligonucleotide molecules (ssDNA or RNA library).

SELEX is an iterative in vitro process which includes three main steps, a) binding step in which the target molecule is incubated with a random

Immune costimulatory-based cancer therapy with aptamers

Costimulatory (co-stimulatory) molecules expressed on the surface of APCs are responsible for the second signal, known as costimulatory signal. The interactions between costimulatory molecules and cognate receptors on the surface of T cells result in clonal T-cell expansion and differentiation, as well as carrying out their effector functions [31]. The presence of an efficient costimulation is crucial for improving antitumor immunity [32]. In fact, one of the mechanisms through which tumors are

Aptamers against immune-suppressive cytokines

Tumors and tumor-associated immune cells can produce a range of cytokines with the ability to suppress tumor-immunity (Fig. 1). The anti-inflammatory cytokines, IL-10 and transforming growth factor-β (TGF-β), are among the most important immunosuppressive cytokines produced or induced by tumor cells. Strategies to reverse this immune suppression represent an attractive target for immune therapy [60], [61], [62]. Targeting TGF-β can substantially augment antitumor immunity in animal models [63],

Bi-specific aptamers for cancer immunotherapy

The first bi-specific cancer immunotherapy was accomplished by bi-specific antibodies (bsAb). The earliest bsAbs were generated by two methods including, chemical cross-linking of whole or parts of antibodies and fusion of two hybridomas resulting in hybrid hybridoma (quadroma). This quadroma secreted bi-specific IgG molecules [74]. However these bsAbs have two major drawbacks. The first problem is associated with the production approach. It was difficult to generate large and homogeneous

Aptamer-targeted antigen delivery in cancer immunotherapy

Aptamers can also serve as modules that selectively recognize and bind to defined cell types or tissues. Conjugation of aptamers to drug, toxin or nanoparticulate systems enable them to deliver cargo molecules to or into specific cells or tissues of interest [82]. Utilizing aptamers for specific targeting and directional antigen transfer into antigen presenting cells has currently emerged as a replacement for antibodies. Targeting immune cells for acquiring potent immune response as well as

Ex vivo and in vivo use of aptamers in cancer immunotherapy

Although cancer vaccines have not yet been successful in patients, an alternative approach of cell-transfer cancer therapy which involves sensitization, ex vivo expansion of autologous lymphocytes with the ability to recognize, reach, and destroy cancer can overcome some barriers to the development of an effective cancer treatment [90]. There are a couple of advantages to this approach including the possibility of administering a very large number of highly selected cells with high affinity for

Conclusion

Rare and sporadic regressions of progression tumors were observed in both cell-based immunotherapy strategy and peptide vaccine emulsified in immune adjuvants. The lack of clinical responses points to the need of interfering with the escape mechanisms of tumors. These strategies have failed because of the inability of immune lymphocytes for homing and infiltrating into tumor tissues, loss of antigen expression by the tumor, the local presence of immunosuppressive factors or cells, and the

Acknowledgments

The authors are very grateful to Eli Gilboa for his excellent critical comments during the preparation of this review. Financial supports from Mashhad University of Medical Sciences are appreciated.

References (110)

  • C. Robert et al.

    Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial

    Lancet

    (2014)
  • N.A. Rizvi et al.

    Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial

    Lancet Oncol.

    (2015)
  • A. Prodeus et al.

    Targeting the PD-1/PD-L1 immune evasion axis with DNA aptamers as a novel therapeutic strategy for the treatment of disseminated cancers

    Mol. Ther.–Nucleic Acids

    (2015)
  • D.H. Bunka et al.

    Development of aptamer therapeutics

    Curr. Opin. Pharmacol.

    (2010)
  • A. Boltz et al.

    Bi-specific aptamers mediating tumor cell lysis

    J. Biol. Chem.

    (2011)
  • L. Beigelman et al.

    Chemical modification of hammerhead ribozymes catalytic activity and nuclease resistance

    J. Biol. Chem.

    (1995)
  • C.E. Tucker et al.

    Detection and plasma pharmacokinetics of an anti-vascular endothelial growth factor oligonucleotide-aptamer (NX1838) in rhesus monkeys

    J. Chromatogr. B Biomed. Sci. Appl.

    (1999)
  • M. Dougan et al.

    Immunotherapy of cancer

  • M.D. Vesely et al.

    Natural innate and adaptive immunity to cancer

    Annu. Rev. Immunol.

    (2011)
  • G. Mayer

    The chemical biology of aptamers

    Angew. Chem. Int. Ed.

    (2009)
  • A.D. Ellington et al.

    In vitro selection of RNA molecules that bind specific ligands

    Nature

    (1990)
  • C. Tuerk et al.

    Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase

    Science

    (1990)
  • D.L. Robertson et al.

    Selection in vitro of an RNA enzyme that specifically cleaves single-stranded DNA

    Nature

    (1990)
  • S. Santulli-Marotto et al.

    Multivalent RNA aptamers that inhibit CTLA-4 and enhance tumor immunity

    Cancer Res.

    (2003)
  • J. Weber

    Review: anti-CTLA-4 antibody ipilimumab: case studies of clinical response and immune-related adverse events

    Oncologist

    (2007)
  • F.S. Hodi et al.

    Improved survival with ipilimumab in patients with metastatic melanoma

    N. Engl. J. Med.

    (2010)
  • O. Hamid et al.

    Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma

    N. Engl. J. Med.

    (2013)
  • T. Mairal et al.

    Aptamers: molecular tools for analytical applications

    Anal. Bioanal. Chem.

    (2008)
  • B.C. Wengerter et al.

    Aptamer-targeted Antigen Delivery

    Mol. Ther.

    (2014)
  • S.K.N. Sandra Santulli-Marotto et al.

    Multivalent RNA aptamers that inhibit CTLA-4 and enhance tumor immunity

    Cancer Res.

    (2003)
  • J.P. Overington et al.

    How many drug targets are there?

    Nat. Rev. Drug Discov.

    (2006)
  • Meyer C, Hahn U, Rentmeister A. Cell-specific aptamers as emerging therapeutics. J. Nucleic Acids...
  • S. Ohuchi

    Cell-SELEX technology

    BioResearch open access.

    (2012)
  • J.O. McNamara et al.

    Cell type-specific delivery of siRNAs with aptamer-siRNA chimeras

    Nat. Biotechnol.

    (2006)
  • C.M. Dollins et al.

    Aptamers in immunotherapy

    Hum. Gene Ther.

    (2008)
  • P. Sharma

    Immune Checkpoint Strategies Introduction

    (2012)
  • S.M. Shamah et al.

    Complex target SELEX

    Acc. Chem. Res.

    (2008)
  • K. Sefah et al.

    Development of DNA aptamers using cell-SELEX

    Nat. Protoc.

    (2010)
  • A. Berezhnoy et al.

    Isolation and optimization of murine IL-10 receptor blocking oligonucleotide aptamers using high-throughput sequencing

    Mol. Ther.

    (2012)
  • K. Palucka et al.

    Cancer immunotherapy via dendritic cells

    Nat. Rev. Cancer

    (2012)
  • P.J. Tacken et al.

    Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting

    Nat. Rev. Immunol.

    (2007)
  • J.-C.D. Schwartz et al.

    Structural mechanisms of costimulation

    Nat. Immunol.

    (2002)
  • A. Molckovsky et al.

    First-in-class, first-in-human phase I results of targeted agents: highlights of the 2008 American society of clinical oncology

    J. Hematol. Oncol.

    (2008)
  • E.J. Small et al.

    A pilot trial of CTLA-4 blockade with human anti-CTLA-4 in patients with hormone-refractory prostate cancer

    Clin. Cancer Res.

    (2007)
  • C. Robert et al.

    Ipilimumab plus dacarbazine for previously untreated metastatic melanoma

    N. Engl. J. Med.

    (2011)
  • J.O. McNamara et al.

    Multivalent 4-1BB binding aptamers costimulate CD8 + T cells and inhibit tumor growth in mice

    J. Clin. Invest.

    (2008)
  • H.-W. Lee et al.

    4-1BB promotes the survival of CD8 + T lymphocytes by increasing expression of Bcl-xL and Bfl-1

    J. Immunol.

    (2002)
  • I. Melero et al.

    Monoclonal antibodies against the 4-1BB T-cell activation molecule eradicate established tumors

    Nat. Med.

    (1997)
  • K.F. May et al.

    Anti-4-1BB monoclonal antibody enhances rejection of large tumor burden by promoting survival but not clonal expansion of tumor-specific CD8 + T cells

    Cancer Res.

    (2002)
  • M. Sznol et al.

    Phase I study of BMS-663513, a fully human anti-CD137 agonist monoclonal antibody, in patients (pts) with advanced cancer (CA)

    J. Clin. Oncol.

    (2008)
  • Cited by (45)

    • Aptamers against cancer drug resistance: Small fighters switching tactics in the face of defeat

      2023, Biochimica et Biophysica Acta - Molecular Basis of Disease
    • Cancer immunotherapy via nucleic acid aptamers

      2022, Aptamers Engineered Nanocarriers for Cancer Therapy
    • Combinatorial RNA therapies in cancer immunotherapy: Challenges and directions

      2022, RNA Therapeutics: The Evolving Landscape of RNA Therapeutics
    View all citing articles on Scopus
    View full text