2,563
Views
6
CrossRef citations to date
0
Altmetric
Editorial

Self-amplifying RNA virus vectors: clinical applications in cancer drug delivery

Pages 1027-1029 | Received 12 Apr 2019, Accepted 06 Aug 2019, Published online: 12 Aug 2019

1. Introduction

Self-amplifying RNA viruses share the common features of single-stranded RNA surrounded by a capsid structure and a protein envelope [Citation1]. However, alphaviruses and flaviviruses possess a genome of positive polarity, while measles viruses (MV) and rhabdoviruses contain a negative-sense RNA genome. Moreover, all self-amplifying RNA viruses carry in their genome the nonstructural genes (RNA replicon) responsible for extreme RNA replication in infected host cells, providing the potential for high-level expression of recombinant proteins from engineered expression vectors [Citation2]. Expression vectors have been developed for transfection of mammalian and non-mammalian cell lines, transfection of primary cells such as neuronal cells, and delivery in vivo [Citation3]. Alphaviruses have demonstrated great flexibility as expression vectors can be delivered as recombinant viral particles (in the form of replication-proficient, replication-deficient, and liposomal nanoparticles), RNA replicons, and DNA/RNA layered plasmids. In the context of therapeutic and prophylactic applications, self-amplifying RNA virus vectors have been utilized for vaccine development and gene therapy in various areas such as infectious diseases and different cancers. In many cases, immunization with self-amplifying RNA virus vectors has provided protection against challenges with lethal doses of infectious agents and tumor cells in several animal models [Citation1]. Moreover, tumor regression and prolonged survival have been established in rodents after administration of self-amplifying RNA viral vectors. In this Editorial the focus will be on cancer therapy. It is important to point out that both prophylactic and therapeutic approaches are presented.

2. Preclinical applications for cancer therapy

A large number of preclinical studies have been conducted with self-amplifying RNA virus vectors in animal tumor models as listed by examples in . For example, an MV vector was constructed with the CD46 and the signaling lymphocyte activation molecule (SLAM) incorporated in the hemagglutinin (HA) protein in combination with the display of a single-chain antibody against the epidermal growth factor receptor (EGFR) at the HA C-terminus. It was shown that intratumoral MV particle administration generated potential antitumor activity in glioblastoma cell lines and therapeutic efficacy by providing tumor regression and significantly prolonged survival in mice with glioblastoma xenografts [Citation4]. Neuronal targeting of micro-RNA miRT124 sequences introduced into an oncolytic Semliki Forest virus (SFV) strain demonstrated virus replication in tumors, therapeutic activity by significant inhibition of tumor growth, and improved survival of C57BL/6 mice carrying CT-2A orthotopic gliomas after a single intraperitoneal injection [Citation5]. In the context of RNA-based delivery, a single intramuscular administration of 0.1 µg SFV-LacZ RNA elicited antigen-specific antibody and CD8+ T cell responses and protected immunized mice from challenges with colon CT26.CL25 tumor cells [Citation6]. In addition to prophylactic activity, therapeutic immunization demonstrated prolonged survival of mice with established tumors. Related to breast cancer, immunization of mice with dendritic cells (DCs) transduced with Venezuelan equine encephalitis virus (VEE) replicon particles expressing a truncated form of the neu oncogene generated robust neu-specific CD8+ T cell and anti-neu IgG responses and therapeutic regression of large established tumors [Citation7].

Table 1. Examples of pre-clinical studies using self-amplifying RNA viral vectors.

Virus-based cervical cancer has been a promising target for prophylactic vaccine development illustrated by immunization of mice with VEE particles expressing the human papilloma virus-16 (HPV16) E7 protein resulting in class I-restricted CD8+ T cell responses and prevention of tumor development [Citation8]. Therapeutic elimination of established tumors was observed in 67% of tumor-bearing mice. Combination therapy with SFV-HPV E6,7, 40 mg/kg sunitib, and low-dose (14 Gy) irradiation dramatically changed the intratumoral compartment, resulting in enhanced immunotherapeutic antitumor activity, inhibition of tumor growth, and 100% tumor-free survival in mice with implanted xenografts [Citation9]. In another combination therapy, co-immunization of two SFV DNA vectors expressing vascular endothelial growth factor-2 (VEGFR2) and interleukin-12 (IL-12), and survivin and β-hCG antigens, respectively, elicited humoral and cellular immune responses against VEGFR2, survivin, and β-HCG [Citation10]. The combined DNA vaccine provided therapeutic activity by superior inhibition of tumor growth and prolonged survival in a B16 melanoma mouse model compared to immunization with individual SFV DNA vectors. Moreover, therapeutic efficacy was observed after intratumoral administration of an oncolytic pseudotyped vesicular stomatitis virus (VSV-GP), which generated long-term remission in murine prostate cancer models, also in subcutaneous tumors and bone metastases after intravenous administration [Citation11]. Additional studies have confirmed therapeutic tumor regression in lung (MV particles and SFV VLPs), ovarian (oncolytic VSV particles and Sindbis virus VLPs), and pancreatic cancers [Citation1].

3. Clinical applications for cancer therapy

Although not to the same extent as recorded for adenovirus vectors, self-amplifying RNA virus vectors have been subjected to clinical trials. Related to cancers several clinical trials have been conducted with self-amplifying RNA virus vectors (). For instance, VEE particles expressing the carcinoembryonic antigen (CEA) were intramuscularly injected four times at doses of 4 × 107 to 4 × 108 IU every 3 weeks in patients with advanced pancreatic cancer in a Phase I trial [Citation12]. Clinically relevant CEA-specific T cell and antibody responses were elicited after repeated immunization. The antibody-dependent cellular toxicity against human colorectal cancer cells was mediated by CEA-specific antibodies and patients showed therapeutic relevance as prolonged overall survival. In the context of castration resistant metastatic prostate cancer (CRPC), five doses of either 0.9 × 107 or 3.6 × 107 IU of propagation-defective VEE particles expressing the prostate-specific membrane antigen (PSMA) were administered to patients with CRPC metastatic to bone in a Phase I trial [Citation13]. Both doses were well tolerated, but only weak PSMA-specific signals were detected. As neither clinical benefit nor robust immune responses were achieved it is thought that the dosing was suboptimal. In a protocol for recurrent glioblastoma multiforme, patients were planned to be treated with MV particles expressing CEA at a starting dose of 1 × 105 TCID50 escalating to the maximum dose level of 2 × 107 TCID50 [Citation14]. So far, three patients have been injected with 1 × 105 TCID50 and three other individuals received 1 × 106 TCID50 in the resection cavity, which generated no dose-limiting toxicity. MV-CEA was subjected to intraperitoneal injections in a Phase I trial in patients with advanced ovarian cancer at doses of 103 to 109 TCID50, which confirmed no dose-limiting toxicity [Citation15]. Stable disease was observed in 14 patients with a median duration of 88 days and a range of 55 to 277 days. The higher doses (107–109 TCID50) provided stable disease in all patients, while stable disease was achieved in only 5 of 12 patients after vaccination with lower doses (103–106 TCID50). Oncolytic MV particles expressing the human sodium iodide symporter (NIS) were subjected to a Phase I trial in patients with relapsed refractory myeloma [Citation16]. As the maximum tolerated dose (MTD) was not achieved in a dose-escalation study (1 × 106 to 1 × 109 TCID50), doses of 1 × 1010 and 1 × 1011 TCID50 were applied showing a complete response in one patient with the higher dose. The response persisted for nine months and the isolated occurring relapse was treated with irradiation, which kept the patient disease-free for an additional 19 months. In another patient, MV-NIS treatment resulted in subjective softening and shrinking of extramedullary plasmacytomas in the back and thighs. The MV Edmonston strain was subjected to a Phase I open-label, non-randomized dose-escalation study in patients with cutaneous T cell lymphoma [Citation17]. Intratumoral administration in combination with subcutaneous injection of interferon-α (IFN-α) resulted in complete regression of CTCL lesions in one patient and in partial regression in other patients. In an approach of tumor targeting and protection against recognition by the host immune system, SFV particles expressing interleukin-12 (LipoVIL12) were encapsulated in liposomes [Citation18]. Intravenous administration of LipoVIL12 in melanoma and kidney carcinoma patients showed up to 10-fold increase in IL-12 plasma levels in a Phase I trial after five days with an MTD of 3 × 109 particles per m2. The encapsulation enhanced tumor targeting, and repeated injections resulted in no toxicity related to the treatment.

Table 2. Examples of clinical trials using self-amplifying RNA viral vectors.

4. Expert opinion

Self-amplifying RNA virus vectors have been engineered for both prophylactic and therapeutic applications [Citation1]. Prophylactic cancer prevention comprises primarily of targeting risk factors to avoid cancers such as carcinogens and tobacco smoke, which is outside the scope of application of viral vector-based vaccines. However, secondary cancer prevention aims at controling or reducing the growth of existing tumors and prevention of metastatic tumors [Citation19]. Although promising results have also been obtained for targeting infectious diseases, the focus here is on cancer treatments. In this context, several studies in animal models have demonstrated efficient tumor regression, significant prolonged survival rates, and even cure. So far, the number of clinical trials has been fairly limited, and mainly Phase I trials have been conducted albeit showing good safety profiles. The therapeutic efficacy has not been outstanding, which may relate to the need of dose optimization and delivery modes (targeting DCs and encapsulation of viral particles). Although not targeting cancer, the encouraging results from a Phase III vaccination trial, which provided protection against EBOV challenges, bodes well for developing strategies for immunization against various cancers [Citation20]. The flexibility of applying self-amplifying RNA viruses as recombinant particles, RNA replicons, plasmid DNA vectors, and LNPs is an additional asset. Moreover, due to the RNA replication mechanism the amounts of RNA and DNA used for immunizations can be reduced. For instance, comparison to synthetic mRNA, self-amplifying VEE RNA replicons expressing the influenza hemagglutinin (HA) required 1.25 µg (64-fold less material) to achieve protection of mice against influenza virus challenges [Citation21]. Similarly, a 200-fold lower equimolar dose of 0.05 µg SFV DNA replicon-based cervical cancer vaccine was sufficient to generate complete tumor regression in 85% of immunized mice [Citation22]. These findings make self-amplifying RNA viruses attractive as an alternative approach for future development of cancer drugs.

Declaration of interest

The author has no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was not funded.

References

  • Lundstrom K. Self-replicating RNA viruses for RNA therapeutics. Molecules. 2018;23:E3310. doi: 10.3390/molecules23123310
  • Strauss JH, Strauss EG. The alphaviruses: gene expression, replication and evolution. Micobiol Rev. 1994;58:491–562.
  • Lundstrom K. RNA viruses as tools in gene therapy and vaccines development. Genes (Basel). 2018;10:E189.
  • Paraskevakou G, Allen C, Nakamura T, et al. Epidermal growth factor receptor (EGFR) retargeted measles virus strains effectively target EGFR- or EGFRvIII expressing gliomas. Mol Ther. 2007;15:677–686.
  • Martikainen M, Niittykoski M, von Und Zu Frauenberg M, et al. MicroRNA-attenuated clone of virulent Semliki forest virus overcomes antiviral type I interferon in resistant mouse CT-2A glioma. J Virol. 2015;89:10637–10647.
  • Ying H, Zaks TZ, Wang R-F, et al. Cancer therapy using a self-replicating RNA vaccine. Nat Med. 1999;5:823–827.
  • Moran TP, Burgents JE, Long B, et al. Alphaviral vector-transduced dendritic cells are successful therapeutic vaccines against neu-overexpressing tumors in wild-type mice. Vaccine. 2007;25:6604–6612.
  • Velders MP, McElhiney S, Cassetti MC, et al. Eradication of established tumors by vaccination with Venezuelan equine encephalitis virus replicon particles delivering human papillomavirus 16 E7 RNA. Cancer Res. 2001;61:7861–7867.
  • Draghiciu O, Boerma A, Hoogeboom BN, et al. A rationally designed combined treatment with an alphavirus-based cancer vaccine, sunitinib and low-dose tumor irradiation completely blocks tumor development. Oncoimmunology. 2015;4:e1029699.
  • Yin X, Wang W, Zhu X, et al. Synergistic antitumor efficacy of combined DNA vaccines targeting tumor cells and angiogenesis. Biochem Biophys Res Comm. 2015;465:239–244.
  • Urbiola C, Santer FR, Petersson M, et al. Oncolytic activity of the rhabdovirus VSV-GP against prostate cancer. Int J Cancer. 2018 Apr 26;143:1786–1796. [ Epub ahead of print].
  • Morse MA, Hobelka AC, Osada T, et al. An alphavirus vector overcomes the presence of neutralizing antibodies and elevated numbers of Tregs to induce immune responses in humans with advanced cancer. J Clin Investig. 2010;120:3234–3241.
  • Slovin SF, Kehoe M, Durso R, et al. A phase I dose escalation trial of vaccine replicon particles (VRP) expressing prostate-specific membrane antigen (PSMA) in subjects with prostate cancer. Vaccine. 2013;31:943–949.
  • NCT00390299. Viral therapy in treating patients with recurrent glioblastoma multiforme. NIH, Bethesda, MD; USA, 2008. Available from November 21, 2018: http://www.clinicaltrials.gov/ct2/show/NCT00390299.
  • Galanis E, Hartmann LC, Cliby W, et al. Phase I trial of intraperitoneal (IP) administration of a measles virus (MV) derivative expressing the human carcinoembryonic antigen (CEA) in recurrent ovarian cancer (OvCa). J Clin Oncol. 2008;26(15S):Abs 5538.
  • Dispenzieri A, Tong C, La Plant B, et al. Phase I trial of systemic administration of Edmonston strain of measles virus genetically engineered to express the sodium iodide symporter in patients with recurrent or refractory multiple myeloma. Leukemia. 2017;31:2791–2798.
  • Heinzerling L, Künzi V, Oberholzer PA, et al. Oncolytic measles virus in cutaneous T-cell lymphomas mounts antitumor immune responses in vivo and targets interferon-resistant tumor cells. Blood. 2005;106:2287–2294.
  • Lundstrom K. Biology and application of alphaviruses in gene therapy. Gene Ther. 2005;12(Suppl. 1):S92–S97.
  • Lollini P-L, Cavallo F, Nanni P, et al. The promise of preventive cancer vaccines. Vaccines (Basel). 2015;3:467–489.
  • Henao-Restrepo AM, Camacho A, Longini IM, et al. Efficacy and effectiveness of an rVSV-vectored vaccine in preventing Ebola virus disease: final results from the Guinea ring vaccination, open-label, cluster-randomised trial (Ebola Ca Suffit!). Lancet. 2017;389:505–518.
  • Vogel AB, Lambert L, Kinnear E, et al. Self-amplifying RNA vaccines give equivalent protection against influenza to mRNA vaccines but at much lower doses. Mol Ther. 2018;26:446–455.
  • Van de Wall S, Ljungberg K, Ip PP, et al. Potent therapeutic efficacy of an alphavirus replicon DNA vaccine expressing human papilloma virus E6 and E7 antigens. Oncoimmunology. 2018;7:e1487913.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.