Elsevier

Biochimie

Volume 145, February 2018, Pages 34-44
Biochimie

Review
Aptamers as potential therapeutic agents for ovarian cancer

https://doi.org/10.1016/j.biochi.2017.12.001Get rights and content

Highlights

  • Current ovarian cancer treatments do not convey acceptable survival advantages and is plagued by drug resistance.

  • Ovarian oncology has put considerable effort into understanding tumour characteristics to develop targeted treatments.

  • Antibodies have made significant progress towards targeted therapies but there is a need for novel targeting agents.

  • EpCAM has been identified as a suitable ovarian cancer biomarker and its targeting has been investigated with varied success.

  • Aptamers are attractive alternatives to antibodies as targeting ligands and use in ovarian cancer is rapidly emerging.

Abstract

Current therapy for ovarian cancer typically involves indiscriminate chemotherapies that can have severe off target effects on healthy tissue and are still plagued by aggressive recurrence. Recent shifts towards targeted therapies offer the possibility of circumventing the obstacles experienced by these traditional treatments. While antibodies are the pioneering agents in targeted therapies, clinical experience has demonstrated that their antitumor efficacy is limited due to their high immunogenicity, large molecular size, and costly and laborious production. In contrast, nucleic acid based chemical antibodies, also known as aptamers, are ideal for this application given their small size, lack of immunogenicity and in vitro production. As aptamers have begun to demonstrate their promise through targeting Epithelial Cell Adhesion Molecule (EpCAM), as well as a number of ovarian cancer biomarkers, in in vivo and in vitro models, their clinical applicability is slowly being realised. This review explores some of the current progress of aptamers targeting cancer biomarkers and their potential role as ovarian cancer therapeutics.

Introduction

Ovarian cancer can be considered the most lethal amongst any gynaecologic malignancy, causing more fatalities annually than any other female reproductive system cancer [1]. Of the estimated 240,000 new ovarian cancer diagnoses each year, close to two thirds are fatal, and the poor 5-year survival rates have remained stagnant since 2008 despite prolific drug development [1], [2], [3]. The reasons for the lower survival outcomes for ovarian cancer have been postulated to be due to the relatively high proportion of diagnoses made at an advanced stage, attributable to the non-specific nature of the symptoms of this cancer [4]. Nearly all ovarian tumours originate from one of three cell types: epithelial cells, sex cord stromal cells and germ cells, though most are epithelial [4]. It is important to note that ovarian cancers reflect a heterogeneous group of diseases, each with their own histologic subtypes that differ in cellular origin, the molecular alterations that mark their initiation and progression, and subsequently their reactions to common treatments. Moreover, the aetiology of this lethal disease is poorly understood and research to identify strategies for early detection and treatment are needed.

Current treatment options for ovarian cancer are relatively non-specific. Many studies stress the importance of debulking surgery, though this can cause menopause and infertility, especially in younger patients. Additionally, surgery places a life-threatening stress to the patient and has been found to have several associated risks such as the triggering of metastasis [5]. The current gold standard chemotherapy is intravenous platinum based carboplatin or cisplatin and paclitaxel combination therapies [6], [7], [8], [9]. The platinum analogues cisplatin, carboplatin and oxaliplatin (amongst others) have been investigated for therapeutic efficacy and toxicity [10], [11]. Carboplatin has now assumed widespread adoption as it was found to exhibit less toxicity with similar efficacy as cisplatin and therefore suitable for more aggressive high-dose chemotherapy [12]. Another example is the renewed interest of anthracycline class drugs such as doxorubicin (DOX), which was originally used in first line treatment in the 1970s [10], [13]. Today, a new formulation of DOX known as PEGylated liposomal DOX, is being preferentially used for its favourable pharmacokinetic properties when compared with the conventional form, resulting in different and more convenient toxicity and efficacy profiles [10], [13]. Unfortunately, even with these minor improvements to their formulations, these conventional forms of chemotherapy are still notorious for the poor quality of life associated with the non-specific cytotoxic side effects targeting the normal healthy cells as well as the intended cancer cells. Furthermore, despite the extensive research that has gone into the best combination/application of chemotherapies, only marginal progress has been made to improving their non-specific toxicity profiles and overall survival.

In addition to the problems associated with the non-specific side effects, drug resistance is a major problem that contributes to limited efficacy. This resistance can be defined as either pre-existent (intrinsic) or drug induced (acquired), depending on whether most the tumour cells already possess resistance-mediating factors, or whether the tumour cells develop resistance during treatment [14].

Acquired resistance is the result of mutations arising during treatment in combination with adaptive evolutionary responses. Inadequate amounts of therapeutic agents reaching the target site places a high risk of selection of drug resistant cell clones. A diverse range of molecular resistance mechanisms have been identified, some of which include drug efflux, and enhanced DNA damage repair. These factors, as well as other factors such as abnormal blood and lymphatic vessels, elevated interstitial fluid pressure, high tumour cell density, and acidic environment, affect the delivery of chemotherapeutic agents to solid tumours and present numerous obstacles to overcome with conventional chemotherapeutic agents [15]. Successful treatment very much depends on the ability to deliver sufficient doses of therapeutic agents to the tumour cells while overcoming these obstacles, as inadequate concentration at the tumour site can contribute to acquired drug resistance [15].

To compound this issue of drug resistance, should a sufficient amount of therapeutic agent reach the tumour, the presence of transporter proteins on cancer cell membranes which promotes drug efflux across the plasma membrane can pump drugs out of the cell (Fig. 1). This process is largely regulated by members of the ATP-binding cassette (ABC) transporter family of proteins [16]. Consisting of 49 members in total, each transporter is characterised by a highly-conserved nucleotide binding domain and a variable transmembrane domain, though each family member's structure varies from protein to protein [16], [17]. Upon substrate binding to the transmembrane domain, a change in conformation at the nucleotide binding site results in the substrate being fluxed out of the cell [16]. The overexpression of these transporters in cancer cell lines and tumours results in multidrug resistance and is one of the major contributors to the failure of chemotherapy [18]. Much of the research into these transporters and their role in chemo-resistance has been focused on multi-drug resistance protein 1 (MDR1) and multidrug resistance-associated protein 1 (MRP1). With each transporter having broad overlapping specificities, these transporters eliminate a number of chemotherapeutics employed in the treatment of ovarian cancer (Table 1) [14]. Recent progress in the understanding of the characteristics of the disease, tumour biomarkers and microenvironment however, has facilitated the development of various targeted agents with mechanisms of action that can circumvent these efflux pumps [13].

The field of cancer research and the knowledge of the disease has been advancing in recent years, revealing the complex and dynamic nature of the disease progression and development. These advancements include the discoveries of the various capabilities of cancerous cells and the knowledge of the various cell types that form a tumour. Beginning in the year 2000, there have been a number of capabilities of cancerous cells discovered that are acquired during the multistep development of human tumours. These have evolved with our understanding of tumorigenesis to include sustaining proliferative signalling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis and activating invasion and metastasis, deregulation cellular energetics and avoiding immune destruction, as well as genome instability and inflammation [22], [23]. In addition, the other area of cancer research that has advanced greatly is the formation of tumours and the cells that take part in forming a “tumour microenvironment”. By conceptualising the mechanisms that enable tumour survival, new research provides targets for treatments specific and essential to cancer pathogenesis, and translating to more effective and robust therapies with fewer off target effects and less non-specific toxicity.

Considerable effort in the past two decades has focussed on combining these hallmarks with biological indicators associated with ovarian cancer to not only understand the aetiology of the disease, but also to develop a ‘personalised’ treatment for this cancer. While biomarkers can take many forms, such as metabolites, karyotypes, methylation status, DNA, mRNA and protein expression, it is the cell surface proteins that have become synonymous with biomarkers and have been the main choice for targeting agents [24], [25]. Membrane proteins have been found to function in many biological process including; cell to cell interaction, adhesion, migration and signal transduction, and their expression has been associated to tumorigenesis. In 2006, Yildirim et al. demonstrated that membrane bound proteins were the target of 69% of US Food and Drug Administration (FDA) approved drugs, and while that number dropped to 36% after 2014, it remains a major site of drug targeting [26], [27], [28]. With these proteins being the target of choice, modalities with high specificity and sensitivity to these targets have been highly sought after.

The production and use of monoclonal antibodies (mAbs) to selectively target tumours became a reality with the advent of Hybridoma technology in 1975 [29]. These mAbs were typically immunogenic in humans due to their origins in mice and had poor abilities to induce human immune effector responses, which limited their clinical applicability. However, with the advances in antibody engineering, solutions to address these problems were devised and the development of chimeric, humanized and fully human mAbs emerged [30], [31], [32].

The success of mAbs is exemplified by Rituximab in the treatment of non- Hodgkin's lymphoma, which was the first mAb to get FDA approval in 1997. Rituximab is a chimeric mAb which targets the CD20 cell surface marker, expressed on 90% of B-cell lymphomas. Its function is to mediate complement and antibody-dependent cell-mediated cytotoxicity (ADCC) and has direct antiproliferative effects against malignant B-cell lines in vitro. Its success has been demonstrated in several clinical trials when used in combination with CHOP (cyclophosphamide, DOX, vincristine, and prednisone) chemotherapy. The addition of rituximab to the CHOP regimen increases the complete-response rate and prolongs event-free and overall survival with diffuse large-B-cell lymphoma, without a clinically significant increase in toxicity [33], [34], [35].

Increasingly, mAbs have been used for the effective treatment of cancer over the past ten years, with tumour expressed antigens being the specific target of these mAbs. Monoclonal antibodies conjugated to radioactive isotopes or chemotherapeutic drugs have shown therapeutic efficacy mainly in haematological malignancies, and mAbs targeting growth factor receptors, such as the epidermal growth factor receptor and human epidermal growth factor receptor 2, are regularly used for the treatment of non-leukaemic cancers.

To be an effective therapeutic for ovarian cancer, mAbs need to penetrate tissues and the extracellular matrix to reach their target cells. Typically, mAbs have limited tissue penetration due to their large size and molecular weight and is considered a major limiting factor [15], [36], [37], [38]. As tumours are characterised with heterogeneous and tortuous vasculature, high interstitial fluid pressure and high viscosity of the tumour blood supply, a major determinant of speed of diffusion through tumours is molecular size. The rate of diffusion is approximately inversely proportional to the cube root of molecular weight. Consequently, large macromolecules such as mAbs diffuse poorly explaining why larger tumour masses may be more difficult to treat by mAb therapy [39]. It has been demonstrated in murine xenograft models, that mAbs directed against tumour-specific antigens largely remain in the blood and no more than 20% of the administered dose typically interacts with the tumour [39]. In addition, achieving optimal affinity is critical for tolerability and cytotoxicity, which is not always easy to engineer as these properties depend on several factors, including antigen density, internalisation, association and dissociation rates.

In addition to mAbs that target tumour antigens, mAbs that target the tumour microenvironment slow tumour growth by enhancing host immune responses to tumour-associated antigens or by curtailing pro-tumorigenic factors produced in the tumour stroma [30]. This ‘enhancing host immune responses’ relies on a functional immune response for treatment to be effective [40]. As described in 2011 by Hanahan and Weinberg, an emerging hallmark is the ability of tumours to evade immune destruction and that a competent immune system will routinely eliminate highly immunogenic cancer cell clones [22]. This process has been referred to as “immunoediting” which will select for weakly immunogenic variants to develop into solid tumours [22]. Tumours may also supress immune response by disabling components of the immune system. For example, cancer cells may paralyse infiltrating cytotoxic T lymphocytes and natural killer cells, by secreting TGF-β or other immunosuppressive factors or through the recruitment of inflammatory cells that are actively immunosuppressive, including regulatory T cells and myeloid-derived suppressor cells [41], [42], [43], [44]. This suggests that this type of therapy is limited due to the known hallmarks of cancer.

However, when considering the hallmarks of cancer, inducing angiogenesis is one target that has shown promising effects in ovarian cancer. Targeting vascular endothelial growth factor (VEGF) has provided the most success for the treatment of ovarian cancer, and bevacizumab has been adopted in combination therapy. Bevacizumab is an anti-angiogenesis antibody targeting VEGF and has demonstrated abilities to improve progression-free survival in women with ovarian cancer [45], [46], [47], [48]. While some believed that effective inhibition of angiogenesis could result in tumour dormancy and even remission, anti-angiogenesis therapies have demonstrated only transitory clinical responses and can lead to other off target effects [49], [50], [51], [52]. While mAbs have demonstrated efficacy in several cancers and for many patients, and we have gained invaluable insight into the mechanism of action of these therapeutic agents, it is evident that further investigation into other targeted therapies are required to improved long term survival in patients.

Section snippets

Aptamers – overcoming challenges

An alternative targeting system that is smaller in size and reliably chemically synthesised could be highly advantageous. The generation of chemical antibodies, also known as aptamers, as an alternative to conventional antibodies is an area of research that shows promise in expanding the diagnostic and clinical arsenal in cancer diagnosis and treatment.

Aptamers, nucleic acid-based ligands, are small single-stranded DNA or RNA oligonucleotides that are produced in vitro via a process known as

Future perspectives and challenges

While investigation into ovarian cancer specific aptamers is still in its infancy, it is envisaged that the aptamers could have therapeutic applications. Ovarian cancer aptamers have only begun to prove their efficacy in in vitro clonal cell lines and in vivo pre-clinical animal models. It is suggested that the following areas are given attention by aptamer researchers:

  • Tumour sphere models: By enriching for ovarian cancer stem cells by growing clonal cell lines in non-adherent conditions to

Conclusions

Through functioning by molecular recognition aptamers can be developed for therapeutic applications with the same intended function as antibodies, such as drug delivery vehicles. Though analogous to their protein counterpart in regards to target recognition and application, aptamers retain numerous key advantages, the main being their size, production process and nucleic acid structure. The significantly smaller size of aptamers (6–10 kDa vs. 150 kDa) allows for greater tissue penetration and

Conflicts of interest

The authors declare no conflict of interest.

Acknowledgements

This review was supported by the School of Medicine Postgraduate Research Scholarship tenable at Deakin University.

References (152)

  • L. Yang et al.

    TGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progression

    Trends Immunol.

    (2010)
  • D. Mougiakakos et al.

    Regulatory T cells in cancer

    Adv. Cancer Res.

    (2010)
  • E.S. Han et al.

    What is the risk of bowel perforation associated with bevacizumab therapy in ovarian cancer?

    Gynecol. Oncol.

    (2007)
  • F. Azam et al.

    Mechanisms of resistance to antiangiogenesis therapy

    Europ. J. Cancer (Oxford, England : 1990)

    (2010)
  • D. Musumeci et al.

    Polyvalent nucleic acid aptamers and modulation of their activity: a focus on the thrombin binding aptamer

    Pharmacol. Therapeut.

    (2012)
  • R. Stoltenburg et al.

    SELEX—a (r)evolutionary method to generate high-affinity nucleic acid ligands

    Biomol. Eng.

    (2007)
  • G. Spizzo et al.

    Overexpression of epithelial cell adhesion molecule (Ep-CAM) is an independent prognostic marker for reduced survival of patients with epithelial ovarian cancer

    Gynecol. Oncol.

    (2006)
  • P.T.H. Went et al.

    Frequent EpCam protein expression in human carcinomas

    Hum. Pathol.

    (2004)
  • M. Trzpis et al.

    Epithelial cell adhesion molecule: more than a carcinoma marker and adhesion molecule

    Am. J. Pathol.

    (2007)
  • H.F. Sears et al.

    Phase-I clinical trial of monoclonal antibody in treatment of gastrointestinal tumours

    Lancet

    (1982)
  • G. Riethmüller et al.

    A.S.G. German Cancer Aid 17–, Randomised trial of monoclonal antibody for adjuvant therapy of resected Dukes' C colorectal carcinoma

    Lancet

    (1994)
  • D. Seimetz et al.

    Development and approval of the trifunctional antibody catumaxomab (anti-EpCAM x anti-CD3) as a targeted cancer immunotherapy

    Cancer Treat Rev.

    (2010)
  • R.L. Coleman et al.

    Latest research and treatment of advanced-stage epithelial ovarian cancer

    Nat. Rev. Clin. Oncol.

    (2013)
  • J. Ferlay et al.

    GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC Cancer Base No. 11

    (2013)
  • A. Jemal et al.

    Global cancer statistics

    CA A Cancer J. Clin.

    (2011)
  • Australian Institute of Health and Welfare & Cancer Australia Cancer

    Gynaecological Cancers in Australia: an Overview

    (2012)
  • S. Tohme et al.

    Surgery for cancer: a trigger for metastases

    Cancer Res.

    (2017)
  • W.P. McGuire et al.

    Cyclophosphamide and cisplatin compared with paclitaxel and cisplatin in patients with stage III and stage IV ovarian cancer

    N. Engl. J. Med.

    (1996)
  • N. Katsumata, M. Yasuda, F. Takahashi, S. Isonishi, T. Jobo, D. Aoki, H. Tsuda, T. Sugiyama, S. Kodama, E. Kimura, K....
  • D.K. Armstrong et al.

    Intraperitoneal cisplatin and paclitaxel in ovarian cancer

    N. Engl. J. Med.

    (2006)
  • S. Dilruba et al.

    Platinum-based drugs: past, present and future

    Cancer Chemother. Pharmacol.

    (2016)
  • R.F. Ozols et al.

    Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: a gynecologic oncology group study

    J. Clin. Oncol.

    (2003)
  • R. Lokadasan et al.

    Targeted agents in epithelial ovarian cancer: review on emerging therapies and future developments

    ecancermedicalscience

    (2016)
  • C. Holohan et al.

    Cancer drug resistance: an evolving paradigm

    Nat. Rev. Cancer

    (2013)
  • D. Xiang et al.

    Nucleic acid aptamer-guided cancer therapeutics and diagnostics: the next generation of cancer medicine

    Theranostics

    (2015)
  • G. Housman et al.

    Drug resistance in cancer: an overview

    Cancers

    (2014)
  • G. Chang et al.

    Structure of MsbA from E. coli: a homolog of the multidrug resistance ATP binding cassette (ABC) transporters

    Science

    (2001)
  • Y.-L. Sun et al.

    Role of ABC transporters in cancer chemotherapy

    Chin. J. Cancer

    (2012)
  • J.F. Deeken et al.

    The blood-brain barrier and cancer: transporters, treatment, and Trojan horses

    Clin. Cancer Res.

    (2007)
  • N. Rifai et al.

    Protein biomarker discovery and validation: the long and uncertain path to clinical utility

    Nat. Biotechnol.

    (2006)
  • M. Köbel et al.

    Ovarian carcinoma subtypes are different diseases: implications for biomarker studies

    PLoS Med.

    (2008)
  • F. Barneh et al.

    Updates on drug–target network; facilitating polypharmacology and data integration by growth of DrugBank database

    Briefings Bioinf.

    (2016)
  • M.A. Yildirim et al.

    Drug-target network

    Nat. Biotechnol.

    (2007)
  • G. Kohler et al.

    Continuous cultures of fused cells secreting antibody of predefined specificity

    Nature

    (1975)
  • L.M. Weiner et al.

    Monoclonal antibodies: versatile platforms for cancer immunotherapy

    Nat. Rev. Immunol.

    (2010)
  • B. Coiffier et al.

    CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma

    N. Engl. J. Med.

    (2002)
  • P. McLaughlin et al.

    Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program

    J. Clin. Oncol.

    (1998)
  • C. Zahnd et al.

    Efficient tumor targeting with high-affinity designed ankyrin repeat proteins: effects of affinity and molecular size

    Cancer Res.

    (2010)
  • I.-K. Choi et al.

    Strategies to increase drug penetration in solid tumors

    Front. Oncol.

    (2013)
  • D. Xiang et al.

    Superior performance of aptamer in tumor penetration over antibody: implication of aptamer-based theranostics in solid tumors

    Theranostics

    (2015)
  • Cited by (18)

    • Cytotoxic effects of aptamer-doxorubicin conjugates in an ovarian cancer cell line

      2023, Biochimie
      Citation Excerpt :

      To improve targeted delivery of DOX, we conjugated it to a ligand specific for Epithelial Cell Adhesion Molecule (EpCAM). EpCAM is a well-characterised cell surface biomarker in OC and ovarian cancer stem cells (CSCs) [35,36]. We have previously developed an aptamer as a ligand specific for EpCAM [37].

    • Future of PD-1/PD-L1 axis modulation for the treatment of triple-negative breast cancer

      2022, Pharmacological Research
      Citation Excerpt :

      Aptamers are nano-sized structures weighing 5–30 kDa and therefore are recognised as nanomedicines [66–70]. Aptamers have found a great application in cancer treatment studies and especially due to their nano-scale size and good tissue penetration, are an interesting modality in brain tumour studies [71–76]. Table 3 compares some of the characteristics of aptamers and mAbs.

    • Molecular dynamics simulation study of gold nanosheet as drug delivery vehicles for anti-HIV-1 aptamers

      2021, Computational Biology and Chemistry
      Citation Excerpt :

      There is relatively little understanding of the mechanisms of ligand binding to aptamers, but it is essential for the full utilization of aptamers as a therapeutic tool (Lin et al., 2010) and other applications. The application of RNA aptamers as a drug for the treatment of various types viral and cancer diseases and other diseases have been studied by researchers (Yu et al., 2018; Romanucci et al., 2018; Bala et al., 2018; Henri et al., 2018; Bouvier-Müller and Ducongé, 2018; Jung et al., 2018; Chakravarthy et al., 2018). Recently, a molecular dynamics investigation of the interactions between the anticancer aptamer and boron nitride/graphene monoxide nanosheets were reported by Mashatooki et al. (2019).

    • Structural optimization and anti-allergic activity of nucleotide aptamers target to Cε3-Cε4

      2019, Biochemical Pharmacology
      Citation Excerpt :

      Macugen is the first commercially available nucleic acid aptamer drug, it can specifically bind and block the function of VEG165, and is extremely used for the treatment of senile retinal macular dystrophy in 2004. The nucleic acid aptamer drug became a milestone in the field of nucleic acid aptamers, subsequently, aptamer drugs targeting various diseases have entered the laboratory and clinical research phases, pushing aptamer drug research to a new climax [3–6]. It is commonly known that aptamer drug research needs to search for key sites for binding to target substances, identify chemically modified and optimized structures after optimally binding bases, in order to improve the stability of nucleic acid aptamers and solving three problems at the same time.

    View all citing articles on Scopus
    View full text