14,278
Views
93
CrossRef citations to date
0
Altmetric
Review

Recent advances of oncolytic virus in cancer therapy

, , &
Pages 2389-2402 | Received 12 Oct 2019, Accepted 23 Jan 2020, Published online: 20 Feb 2020

ABSTRACT

Oncolytic viruses have been taking the front stage in biological therapy for cancer recently. The first and most potent virus to be used in oncolytic virotherapy is human adenovirus. Recently, ongoing extensive research has suggested that other viruses like herpes simplex virus (HSV) and measles virus can also be considered as potential candidates in cancer therapy. An HSV-based oncolytic virus, T-VEC, has completed phase Ш clinical trial and has been approved by the U.S. Food and Drug Administration (FDA) for use in biological cancer therapy. Moreover, the vaccine strain of the measles virus has shown impressive results in pre-clinical and clinical trials. Considering their therapeutic efficacy, safety, and reduced side effects, the use of such engineered viruses in biological cancer therapy has the potential to establish a milestone in cancer research. In this review, we summarize the recent clinical advances in the use of oncolytic viruses in biological therapy for cancer. Additionally, this review evaluates the potential viral candidates for their benefits and shortcomings and sheds light on the future prospects.

Introduction

Oncolytic virotherapy, a revolutionary tool for cancer treatment has shown promising results for the last two decades.Citation1 More than a century ago it was first observed that cancer patients underwent cancer regression if they were infected with certain viruses.Citation2 Revolutions in recombinant DNA technology have provided important tools to study the biology of viruses, thereby advancing biological therapy for cancer, resulting in the new generation of cancer therapeutics. While chemo and radiation therapies continue to be the chosen cancer treatment options, the serious side effects are a major drawback of these therapies. Biological therapy for cancer, although relatively complex and challenging, is the preferred treatment option owing to good efficacy, limited side effects and being less painful to cancer patients. So far clinical trials report no deaths or clinically serious adverse events attributed to oncolytic virotherapy. In cancer treatment the patient’s safety is of utmost importance and treatment, using oncolytic viruses seems to be the most promising in this aspect. Most of the oncolytic viruses chosen for cancer therapy are attenuated strains or strains that can infect and replicate in humans without causing any serious disease. It is also important that the viruses chosen must be capable of utilizing the host immune system to recognize and destroy the cancer cells.

Although oncolytic viruses are potentially powerful therapeutic agents for cancer treatment, a single type of oncolytic virus is not enough to destroy all the cancer cells due to the heterogeneity of cancer tissues and complexity of cancer cells. Some cancer cells and the non-transformed supporting cells may be resistant to certain oncolytic viruses, indicating that a single type of virotherapeutic agent may not be effective in all types of cancers. Therefore, the most challenging part of the oncolytic virotherapy is to identify the virus and the delivery method that best fits the patient’s system and activates the immune system against the tumor cells. Currently, several viruses including vaccinia virus, coxsackievirus, adenovirus, reovirus, herpes simplex virus, and measles virus are being extensively investigated and are undergoing clinical trials for use in the treatment of various types of advanced cancers. The recognition of the genetically engineered herpes simplex virus-Talimogene Laherparepvec (T-VEC) by the U.S. Food and Drug Administration (FDA) and the European commission for oncolytic virotherapy is a major leap in the advancement of the application of viruses in cancer treatment. To achieve more stable and long-lasting results in cancer therapy, virotherapy has been combined with chemotherapy and/or immunotherapy recently.

In this review, we summarize the recent advances in biological therapy for cancer and also evaluate the potential viral candidates for their benefits and shortcomings while indicating the future prospects.

HSV-based oncolytic viruses

HSV has been considered and developed as an oncolytic virus for cancer therapy since 1991. There are seven HSV-based oncolytic viruses among which T-VEC (Commercial name Imlygic) has been approved by US-FDA and European Medicine Agency for clinical use after the successful phase I, II, and III clinical trials. This oncolytic virus is manufactured by Amgen Inc. (Thousand Oaks, USA), and was generated by deleting the ICP34.5 and ICP47 genes, and inserting two copies of human granulocyte-macrophage colony-stimulating factor (GM-CSF) gene in place of ICP34.5.Citation3 In normal cells, viral replication is blocked by protein kinase R (PKR) activation and subsequent phosphorylation of eukaryotic initiation factor 2 (eIF2). In cancer cells, the disrupted PKR-eIF2 pathway causes uncontrolled cell proliferation and unlimited permissiveness to viral replication as well.Citation4,Citation5 ICP34.5 causes dephosphorylation of eIF2 and blocks PKR-induced disruption of protein synthesis. Deletion of ICP34.5 in T-VEC ensures abortive infection in normal cells thereby enabling its replication to be cancer cell-specific.Citation6 ICP47 decreases the immune destruction caused by the host cell thereby supporting HSV1 proliferation. Deleting ICP47 therefore allows immune destruction of the virus in normal cells while enhancing the cell surface expression of MHC1 in cancer cells and increasing the tumor antigen presentation by the infected cancer cells.Citation7The GM-CSF engineered into T-VEC enhances T-cell priming by dendritic cells, thereby stimulating the immune system.

A phase I trial of T-VEC consisted of 30 patients all affected with different types of cancers, of whom 9 patients were diagnosed with refractory metastatic melanoma. Intratumoral T-VEC injection led to remission in two patients with no detectable adverse effects. This encouraged the researchers to perform phase II clinical trial on patients with stage IIIc or IV melanoma. Among the 50 patients treated intratumorally, 8 patients achieved a complete response (CR), while 5 patients achieved partial response (PR). This success led to a phase III clinical trial with the enrollment of 436 patients with stage IIIb, IIIc, and IV unresectable melanoma treated with T-VEC. This trial also compared T-VEC with recombinant GM-CSF. In the T-VEC group, the overall survival rate was higher than in the GM-CSF group. Among the 436 patients, the double response rate was higher in patients treated with T-VEC compared to those treated with GM-CSF alone. T-VEC treatment also showed profound efficacy in stage IIIB, IIIC, or IV M1a patients. Moreover, adverse effects related to T-VEC treatment except fatigue, chills, and pyrexia were not observed. This success of the phase III trial played a significant role in the US-FDA approval of T-VEC.Citation8 More T-VEC clinical trials are currently in progress. One single-arm, phase II, single-center study is being conducted on the low-risk squamous cell carcinoma patients, where 28 patients were injected with T-VEC at the target lesions. The second injection was administered 3 weeks after the first injection and the third and fourth injection were administered 2 weeks after the second and third injections, respectively (NCT03714828) (). Another study currently in progress to evaluate the efficacy of T-VEC in squamous cell carcinoma patients is designed to test T-VEC alone and T-VEC combined with ipilimumab (NCT01740297) (). A phase I study is ongoing with 36 patients having a recurrent or metastatic head and neck squamous cell carcinomas to evaluate the dose-limiting toxicity of T-VEC in combination with pembrolizumab (NCT02626000) (). An ongoing phase II study comprising 112 patients with unresected stage IIIB to IV M1c melanoma is designed to evaluate the correlation between CD8+ cell density and objective response rate in the patients (NCT02366195) (). Even though there is an overall success in the clinical trials using T-VEC as mentioned above such as in the treatment of stage IIIc and IV unresectable melanomas, one study has reported chronic granulomatous dermatitis at the T-VEC injection site in melanoma patients.Citation9 However, this is not of much concern as in most of the patients the granulomatous dermatitis did not relapse after treatment discontinuation and spontaneous regression of the nodules occurred over several months.Citation9

Table 1. Clinical trials of the HSV-based oncolytic viruses

HF-10, another HSV1-based oncolytic virus naturally lacks the expression of several genes of the virus such as UL43, UL49.5, UL55, UL56, and LAT (latency-associated transcripts). This oncolytic virus, produced by Takara Bio Inc., JapanCitation10 efficiently replicates in tumor cells and induces an increased number of CD4+ and CD8+ T-cells and NK cells within the tumor leading to a reduction in the tumor size. Like T-VEC, HF-10 has also been reported to have potent anti-tumor efficacy against a range of malignancies. It has been shown to effectively destroy colon carcinoma, peritoneal cancer, and melanoma in immune competent murine models when used alone or in combination.Citation11-13 Several clinical trials with HF-10 have either been completed, or are ongoing, or have been planned. Two studies have been conducted in melanoma patients (NCT02272855) () in the USA: one is a monotherapyCitation14 and the other is a combination therapy with ipilimumab. Besides, a phase 2 study ongoing in Japan to treat resectable stage IIIB, IIIC, IV M1a melanomas has recruited seven patients treated with HF-10 in combination with nivolumab in a single-arm, open-label study (NCT03259425) ().Citation15 A combination therapy for 76 patients with unresectable pancreatic cancer is being evaluated in a phase I, open-label, multi-center study to determine the recommended dose of HF-10 (NCT03252808) ().Citation16 In February 2005, a study was carried out on HSV-seropositive patients with head and neck squamous cell carcinoma (HNSCC) to demonstrate the safety and efficacy of HF-10. These patients were treated with intratumoral injections of HF-10 with a dose of 1 × 105PFU for 3 days. However, no significant decrease in tumor size was observed, and patients reported low-grade fever after the injection. A new trial was then planned for testing higher doses of HF-1015.

Although HF-10 and T-VEC originate from different strains, they have many similarities such as both their genomes consist of linear double-stranded DNA which has two unique inverted sequences and a unique long sequence flanked by terminally repeated long sequence and internally repeated long sequence.Citation17,Citation18 T-VEC is obtained by modifying the JS1 strain to improve tumor-specific growth (deletion of ICP34.5 and ICP47) and immune response (insertion of hGM-CSF).Citation18 HF-10, on the other hand, has natural deletions of UL56 and LAT and increased expression of genes like UL53 which enhance its safety aspects.Citation19-21

HSV1716 is an oncolytic HSV containing the deletion of only a specific determinant of virulence ICP34.5. This replication-defective mutant of HSV1 originates from the wild-type strain 17, and is produced by Virttu Biologics, Glasgow, UK. In a phase I study, HSV1716 has been administered in pediatric patients with relapsed or refractory extracranial cancer via an image-guided intratumoral injection. A total of nine patients has been treated; eight of them have been given single doses while one has been given two doses. No dose-limiting toxicities have been observed, indicating that intratumoral HSV1716 is safe for children and young adults with late-stage aggressive cancers.Citation22 In a different phase I study, HSV1716 has been shown to be safe when injected at a dose of 105 PFU directly into human high-grade gliomas, and also when injected up to 106 PFU directly into brain tumors. A phase I dose-escalation study has recently been completed where HSV1716 was injected intratumoral or intravenous in patients with refractory non-central nervous system solid tumors (NCT00931931) (). In another phase I/II trial, patients with malignant pleural mesothelioma (MPM) were treated with an intra-pleural injection of HSV1716. This treatment was found to be safe and well tolerated by the patients. Adequate HSV1716 replication was seen resulting in an effective immune response in pleural fluid and blood (NCT01721018) (). Further studies indicating the use of HSV1716 in combination with immune checkpoint inhibitors in MPM patients have been suggested.Citation23

G207 is an HSV1 resulting from the deletion of ICP34.5 and the substitution of ICP6 with LacZ. The deletion of ICP34.5 ensures the lack of neurovirulence while the ICP6 attenuation results in specificity for tumor cells with P16 tumor suppressor defects.Citation24 This clinically safeCitation25 oncolytic virus is manufactured by Medigene, Germany. Intratumoral injections of this oncolytic virus at high doses of 3 × 109 PFU were seen to be well tolerated among patients with tumors growing in important and sensitive organs. However, due to the lack of clinical efficacy evidence, cancer biotherapy using this oncolytic virus has not entered the phase II trial yet. In a study comprising six glioblastoma patients, two 1.15 × 109 PFU doses of G207 were injected stereotactically via a catheter into a resection cavity after 2–5 days of tumor removal. This was the maximum achievable dose that was tolerated when administered two times.Citation26 However, serious adverse events were experienced by all six patients due to their underlying disease. Even though some of the adverse events were thought to be related to G207 administration, no patients discontinued study participation. Also, the patient’s saliva, serum, urine, and conjunctiva samples were tested and none of the samples showed pathogenic evidence of encephalitis caused by HSV.Citation26 Furthermore, in some patients, extensive distribution of the progeny virus in the treated tumors was not observed.

M032 is an HSV1-based second-generation oncolytic virus expressing IL-12, manufactured by Acttis Inc, Pennsylvania, USA. It causes the dying tumor cells to secrete IL-12 which promotes an anti-tumor immune response. Notably, IL-12 has anti-angiogenic effects, hence, preventing the tumor growth.Citation27 Currently, this HSV1-based oncolytic virus has entered phase I study for the treatment of recurrent malignant glioma (NCT02062827) ().

G47Δ, an HSV1-based third-generation oncolytic virus manufactured by Daiichi Sankyo Company, Tokyo Japan, is structurally quite similar to G207 except for an additional ICP47-deletion. One study showed that G47Δ can effectively kill different subtypes of breast cancer cells. It affected both paclitaxel-resistant cancer stem cells and non-cancer stem cells,Citation28 equally. In 2005, there was a report of G47Δ used in combination with androgen ablation for the treatment of human prostate cancer. The study done in vitro and in vivo xenograft tumor model using human prostate cancer cells demonstrated that G47Δ could be a good candidate for treating human prostate cancer.Citation29

Adenovirus-based oncolytic viruses

Oncorine (also known as H101) is the first recombinant oncolytic adenovirus approved by China Food and Drug Administration Department (CFDA) to be used in combination with chemotherapy for the treatment of nasopharyngeal carcinoma in late 2005.Citation30-32 The modifications resulting in this oncolytic virus were E1B deletion and E3 partial deletion. A phase III study has been conducted successfully in patients with head and neck squamous cell carcinoma (HNSCC) and esophageal squamous cell carcinoma (ESCC). The intratumoral injection of H101 combined with cisplatin and 5-fluorouracil (PF) or adriamycin and 5-fluorouracil (AF) was compared with PF or AF treatment alone. The study indicated the safety and efficacy of the H101 intratumoral injection. This study was a milestone leading to CFDA’s approval for the use of oncorine as combination therapy for combating cancer.Citation33 After it was launched in the market, clinical trials in China tested oncorine for four different types of cancers. The best results were observed for the oncorine-treated patients with malignant plural effusion, showing 38% complete recovery ().Citation34

Table 2. Oncorine’s clinical trials

Almost at the same time, another adenovirus-based oncolytic virus, Onyx-015 was developed by Onyx Pharmaceutical, South San Francisco, USA, in which E1B (55 kDa) was completely deleted. Clinical trials involving Onyx-015 were done for the patients with HNSCC, pancreatic cancer, ovarian cancer, colorectal cancer, and premalignant oral dysplasia. Charles et al. have proved that the topical application of Onyx-015 as mouthwash therapy has significant efficacy against oral dysplasia.Citation35 For pancreatic cancer, phase I trial results ensured the safety of Onyx-015 for its use in combination therapy with gemcitabine. Among the 21 patients comprising the trial, six patients showed stable disease (SD), two showed partial response (PR), and two had minor regression (MR). However, the major challenge for Onyx-015 treatment is the preexisting neutralizing antibodies.Citation36

DNX-2401 is another promising adenovirus-based oncolytic virus, produced by DNAtrix, Huston, USA. It consists of a deletion of 24 bps in the E1A region and the engineering of the RGD motif into the HI-loop of the fiber knob. While this insertion of the RGD-4C motif enhances the replication and infectivity of the adenovirus in cancer cells, it reduces the sequestration of adenovirus by CAR-expressing normal cellsCitation37 thus emphasizing the potency and safety of DNX-2401. In a treatment regimen, two groups of patients with recurrent malignant glioma were treated with DNX-2401. One group (group A) received a single dose while the other group (group B) received 2 doses, first dose into multiple sites via implanted catheters and the second dose on day 14 after tumor regression. In group A, 5 among the 25 patients survived for more than 3 years of whom 3 patients showed more than 95% regression in tumor size. The results were promising, indicating that DNX-2401 could be used as a potential therapeutic agent in combination therapy for gliomas.Citation37,Citation38 Currently, two phase I clinical trials is in progress for recurrent glioblastomas, one is in combination with temozolomide (NCT01956734) () and the other is in combination with interferon-γ (NCT02197169) (). This success for glioblastoma patients has encouraged another ongoing clinical trial where the diffuse intrinsic pontine gliomas (DIPG) in children is being treated intratumorally with DNX-2401, 3–4 weeks prior to radiotherapy or chemotherapy (NCT03178032) (). The results of this study are unpublished. In another ongoing study for recurring glioblastoma or gliosarcoma, DNX-2401 is delivered directly to the tumor following intravenous administration of pembrolizumab (NCT02798406) ().

Table 3. Clinical trials of the adenovirus-based oncolytic viruses

Table 4. Clinical trials of MV-NIS

Table 5. Clinical trials of reovirus

Table 6. Clinical trials of PVSRIPO

ONCOS-102, produced by Targovax, Oslo, NorwayCitation39 is similar to DNX-2401 except that it is additionally armed with a potent immunological stimulator GM-CSF. A phase I study employing ONCOS-102 in combination with low-dose oral cyclophosphamide has been conducted to treat solid tumors in 12 patients.Citation40 The treatment was deemed safe, was well tolerated, and induced a tumor-specific immune response (NCT01598129) (). Another phase I/II study employing ONCOS-102 in combination with pemetrexed/cisplatin is ongoing in 30 patients with mesothelioma (NCT02879669) (). In another regimen, 12 patients with advanced melanoma have been treated with ONCOS-102 and pembrolizumab combination (NCT03003676) (). All these studies primarily indicate that ONCOS-102 is well tolerated and can induce an anti-tumor immune response.

VCN-01 is an Rb pathway selective- and hyaluronidase-armed oncolytic adenovirus, manufactured by VCN Biosciences SL, Barcelona, Spain. This oncolytic virus is being currently employed in several clinical trials, including a phase I dose-escalation study on advanced solid tumor treatment with intravenous VCN01 alone and in combination with gemcitabine and abraxane (NCT02045602) (), a phase I safety and VCN-01 activity evaluation study for patients with refractory retinoblastoma (RTB) (NCT03284268) () and a phase I study assessing the safety, tolerability, and efficacy of VCN-01 in combination with durvalumab in patients with HNSCC (NCT03799744) (). VCN-01 has shown cytotoxic effects on glioma cells in vitro and in vivo.Citation41 Recently a team of researchers from SJD Barcelona children’s hospital treated pediatric chemo-resistant retinoblastoma patients with intravitreous injection of VCN-01, and the results indicated enough viral replication in tumor cells to induce anti-tumor activity in retinoblastoma vitreous seeds and no systemic inflammation tumor.Citation42

LOAd-703 is an oncolytic virus loaded with genes encoding CD40L and 4-1BBL, produced by Lokon pharma, Uppasala, Sweden. This oncolytic adenovirus is currently being evaluated for its safety in two Phase I/II clinical trials. One is for pancreatic cancer patients (NCT02705196) () and the other where LOAd-703 is used together with the standard of care chemotherapy or gemcitabine for patients having various types of cancers (pancreatic, biliary, colorectal, or ovarian) (NCT03225989) ().

ICOVIR-7 and ICOVIR-5 are two types of oncolytic adenoviruses generated by the Targovax and Catalonia Institute of Oncology, respectively. ICOVIR −7 has been evaluated in a clinical trial for 21 patients with advanced and refractory solid tumors. Anti-tumor activity was observed in this clinical trial, showing stabilization or reduction in tumor size. The results were 1PR, 2MR, and 2SD, indicating its eligibility for further clinical trials.Citation43 A dose-escalation phase I clinical trial on metastatic melanoma patients showed that the virus could be used for successful systemic administration. However, it failed to induce tumor regression.Citation44

Adenovirus-based oncolytic viruses have exploited the p53 inactivation in most cancer cells to continue replication. Oncorine and Onyx-015 contain a deletion of the E1B (55 kDa)Citation45 gene product which selectively binds to p53 protein causing disruption of apoptosis. The deletion of E1B (55 kDa) reduces the replication ability of adenovirus in normal cells. However, p53-deficient cancer cells are conducive for the replication of the reconstructed virus. The E1B (19 kDa) in both oncolytic viruses might prevent apoptosis by blocking the function of Bax. The oncolytic strategies of Onyx-015Citation46 and Oncorine are based on two factors, one is tumor cell-specific viral replication and the other is virus-induced cell death, while DNX-2401 emphasizes on improving the ability of infecting tumor cells. DNX-2401 has the RGD-4C motif inserted in the fiber which enables selective infectivity in cancer cells with Rb/p16 tumor suppressor pathway defects. Integrin specificity of the modified fiber causes receptor-independent infection of the cancer cells and the oncolysis resulting from viral replication. VCN-01 expresses hyaluronidase which degrades the extracellular matrix (ECM) that acts as a physical barrier for the tumor cells enabling viral spread within tumor cells.Citation47-49 This results in enhanced penetration and efficacy of chemotherapeutic agents.Citation50-52 LoAd703 was designed to modulate the tumor microenvironment and to simultaneously activate the immune system against the tumor cells.Citation53 Recently, a new oncolytic vector ORCA-010 has been reported to show enhanced oncolytic efficacy and safety in vivo. This oncolytic vector has a novel mutation (T1) which contains a single adenine-base insertion at position 445 within the ER retention domain of the E3/19K gene. This mutation significantly increases the oncolytic potential of adenovirus human serotype 5 (AdHu5)-based vectors. These vectors are estimated to be more potent than the licensed ONYX015.Citation54,Citation55

CG0070, produced by Cold Genesys is a conditionally replicating oncolytic adenovirus that is armed with GM-CSF. In vitro and in vivo studies with bladder transitional cell carcinoma (Bladder-TCC) models have shown promising results, suggesting that CG0070 could be a potential therapeutic agent for bladder cancer.Citation56 Intravesical treatment with CG0070 in patients with Bacille Calmette-Guerin (BCG) resistant high-grade non-muscle invasive bladder cancer (NMIBC) has also shown promising results. A phase II study has also been conducted with patients suffering from BCG-unresponsive NMIBC who refuse cystectomy (NCT02365818). A group of 45 patients with 24 pure carcinoma-in-situ (CIS) patients was treated with intravesical CG0070. Within 6 months of the treatment regimen, 47% of the total and 50% of the CIS patients showed complete response,Citation57 indicating that CG0700 could be a potential anti-cancer therapeutic agent. Recently, another clinical trial has been planned by Cold Genesys to evaluate the combination therapy of CG0070 with pembrolizumab in bladder cancer.

Most of the adenovirus-based oncolytic viruses showed success in clinical trials. However, the major drawback is the high levels of neutralizing antibodies to the vector itself that was observed in the patients, which may impair the therapeutic efficacy. It is reported that chimpanzee adenovirus-based oncolytic virus could overcome the problems of preexisting immunity to human adenovirus serotypes,Citation58,Citation59 which implies that chimpanzee adenovirus has the potential to be applied clinically. Other strategies that have been considered to avoid the problem of preexisting immunity includes: (1) using less seroprevalent adenovirus serotypes. Adenovirus subgroup DCitation60 has decreased intrinsic hepato-tropism due to low affinity to FX,Citation61 and adenovirus type 9 was found to be the most appealing alternate serotype for cancer therapeutic application;Citation62,Citation63 (2) capsid pseudotyping. The fiber knob or the fiber of the AdHu5 capsid could be modified or substituted by the corresponding part from the less seroprevalent or less immunogenic serotypes. For example, exchanging AdHu5 hexon HVR with a less seroprevalent Ad48 HVR resulted in altered vector immunogenicity;Citation63-66 (3) genetic masking. It is achieved by either short heterologous peptide insertion within the fiberCitation67,Citation68 or by fiber deknobbing. In this strategy, artificial peptide structures are used to remove or replace the fiber knob domain;Citation69 (4) chemical shielding. The viral vector is shielded using distinct carriers such as PEGylated polymeric carriers.Citation70 The covalent attachment of PEG with hexon and the fiber reduces the chance of an immune attack. Another form of chemical shielding can be achieved by bio-reducible (cationic) polymers,Citation71 with liposomes or with bi-specific adaptor molecules.Citation72-75

Measles-based oncolytic viruses

Measles virus (MV) belongs to the paramyxoviridae family which also consists of mumps and other viruses that cause respiratory tract infections.Citation76 The extremely safe live-attenuated MV vaccine was derived following multiple passages in human kidney cells, human amnion cells, and chicken embryos following its isolation from the Edmonstonstrain.Citation77,Citation78 With respect to safety, the MV vaccine is very promising as the risk of reverting back of the non-segmented genome into the pathological form is very unlikely.Citation79 This feature of the MV vaccine is therefore very reliable when it comes to the matter of oncolytic viruses where safety is of utmost importance. MV-Edm-Zagreb (MV-EZ) vaccine strain is the genetically unmodified measles virus strain that has been tested in an open-label dose-escalation phase I clinical trial conducted in Switzerland. This strain was used in the intratumoral treatment of five measles immune patients with CTLC. To control the MV-EZ spread in the normal tissues these patients were subcutaneously injected with interferon alpha (IFN- α) 72 or 24 h before treatment. Complete recovery was observed in one patient while the remaining four patients showed partial recovery with the recovery of distant-noninjected lesions in two patients. Two variants of oncolytic measles virus have been constructed by genetically expressing human carcinoembryonic antigen (MV-CEA) and human sodium iodide symporter (MV-NIS). Currently, the clinical trials that are ongoing aim at treatment of ovarian cancer, glioblastoma, multiple myeloma, mesothelioma, head, and neck cancer, breast cancer, and malignant peripheral nerve sheath tumors. While one study uses the measles oncolytic virus alone, the other which is still ongoing is a comparative study using MV-NIS with chemotherapy for ovarian, fallopian, or peritoneal cancer (NCT02364713) (). In some other ongoing trials, 21 measles immune patients with recurrent ovarian cancer were treated by intraperitoneal administration of MV-CEA, while MV-NIS was tested to treat multiple myeloma (intravenous) (NCT00450814, NCT02192775) (), mesothelioma (intrapleural) (NCT01503177) (), HNSCC (intratumoral) (NCT01846091) (), and malignant peripheral nerve sheath tumors (intratumoral) (NCT02700230) (). Completed studies have not shown any dose-limiting toxicity. The main drawback of measles-based oncolytic virotherapy is the preexisting anti-measles antibodies which may hamper the systemic administration of the virus.Citation80 Several strategies are being considered to overcome this problem: (1) delivery of the virus to its target using a cellular vehicle;Citation81 (2) suppression of the intracellular pathways associated with innate immunity by encoding one or two immune-suppressing genes of the wild-type MV into MV-EDM. However, the safety of this approach is questionable; (3) replacement of the H and F glycoproteins on the MV envelop with immunologically unreactive but structurally similar glycoproteins of the related animal virus;Citation82 (4) combination of the MV-based oncolytic therapy with immune-suppressive drugs.

Other oncolytic viruses

Reovirus is one among the naturally occurring oncolytic viruses. The natural strain of reovirus is capable of recognizing the altered signaling pathways in cancers.Citation83 Reovirus oncolysis occurs mainly through apoptosis combined with autophagy. In HCV-associated hepatocellular carcinoma (HCC), reovirus induced pro-inflammatory response efficiently suppresses the multiplication of HCV in host cells, and reovirus-based immune responses are also effective against HBV-based HCC.Citation84

Several clinical trials have demonstrated that reovirus type 3 is a promising therapeutic agent for cancer.Citation85-89 In one study 33 patients with advanced cancers received escalating-doses of reovirus for 4 weeks and no dose-limiting toxicity was observed.Citation89 Currently, several phase I and II trials are ongoing some among which are combination therapies. Wild-type reovirus has been used in combination with sargramostim to treat the patients with refractory brain tumors (NCT02444546) (). Pelareorep, a live replication-competent naturally occurring reovirus type 3 (Dearing strain)Citation90 has been used in combination with paclitaxel to treat patients with recurrent fallopian tube carcinoma, recurrent ovarian carcinoma, and recurrent primary peritoneal carcinoma (NCT01199263) (). In a phase I clinical trial, relapsed or refractory multiple myeloma was treated with pelareorep in combination with dexamethasone, carfilzomib, and nivolumab (NCT03605719) ().

Other naturally occurring oncolytic viruses include Newcastle disease virus (NDV) and parvovirus H-1 (ParvOryx).Citation91 NDV has been widely used and promising results have been observed in several clinical trials against a variety of cancers such as leukemia, lymphoma, melanoma, neuroblastoma, fibrosarcoma, colon carcinoma, mesothelioma, and head and neck carcinoma.Citation92-116 Phase II clinical trials with NDV on various cancer types including stage II and III melanoma,Citation117-120 colorectal cancer with liver metastasis,Citation121-123 resectable colorectal cancer,Citation124,Citation125 metastatic renal cell carcinomaCitation126 have shown improved overall survival. NDV has a natural specificity for the cancer cells that have defects in the activation of the antiviral signaling pathway,Citation127-129 type I IFN signaling pathway,Citation127,Citation130 defects in apoptotic pathwayCitation131,Citation132 etc. NDV armed with pro-apoptotic protein, apoptoninCitation133 or cytokine,Citation96 immunoglobulinCitation134 or tumor-associated antigens (TAA),Citation135 etc., is a better oncolytic virus.

Cavatak is a naturally occurring picornavirus-based oncolytic virus,Citation136 manufactured by Viralytics, Australia. PVSRIPO is another picornavirus-modified oncolytic virus that is CD155/Nec15-dependent, because the internal ribosome entry site (IRES) of the virus has been replaced with the IRES of human rhinovirus type 2 (HRV2). A phase I clinical trial has been conducted in which 61 participants with recurrent glioblastoma were treated with PVSRIPO (NCT01491893) (). Other trials are ongoing for patients with grade IV malignant glioma (NCT02986178) and pediatric patients with recurrent malignant glioma (NCT03043391) ().

JX-594 (known as Pexa-Vec) is an oncolytic vaccinia virus armed with the GM-CSF gene, produced by a France-based company, Transgene. The vector was designed to activate the host immune system against tumor cells. Pexa-Vec has been tested in a phase I/II study against refractory metastatic colorectal cancer to evaluate its safety, tolerability, and feasibility in combination with immune checkpoint inhibition (NCT03206073). Recently, a phase III PHOCUS clinical trial (NCT02562755) on advanced hepatocellular carcinoma patients was conducted by Transgene in partnership with South Korean biotech company SillaJen. However, it has been discontinued because the study failed to meet the primary objectives and did not show any significant clinical benefit, which has been attributed to the complexity of hepatocellular carcinoma.Citation137

GL-ONC1 is an oncolytic vaccinia virus equipped with the light-emitting fusion protein Renilla luciferase-Aequorea green fluorescent protein (RUC-GFP). It enables easy detection and monitoring of virus infected-tumor cells both in vitro and in vivo. While virus induced-cell lysis is responsible for the death of the cancer cells, the release of TAA activates the immune system against the tumor.Citation138 A clinical trial has been carried out on patients with advanced-stage cancers with no standard care options for treatment (NCT03420430); however, no further information has been released yet. A phase I/II study, with GL-ONC1 is currently ongoing consisting of patients with recurrent or refractory ovarian cancer (NCT02759588). The treatment was planned as a monotherapy using GL-ONC1 alone or in combination with chemotherapy with or without bevacizumab. vvDD-CDSR is a vaccinia virus-derived oncolytic virus with deletion of the thymidine kinase (TK) gene and the growth factor gene and addition of the cytosine deaminase (CD) gene and the somatostatin receptor (SR) gene. vvDD-CDSR shows potential oncolytic activity in treatment for refractory and metastatic pediatric solid tumor. A phase I study has been conducted in patients with refractory-advanced colorectal or other solid cancers. The observations showed no dose-limiting toxicity or other adverse events except grade ½ flu symptoms. A potent Th1-mediated immunity against the virus as well as the cancer cells were also reported.Citation139

Alpha virus M1 has natural oncolytic properties. It significantly suppresses a variety of cancer cells that have zinc finger antiviral protein (ZAP) deficiency. The initial study was carried out in non-human primates (cynomolgus macaques) to evaluate the safety of intravenous administration of the virus for human trials in future.Citation140,Citation141 As ZAP is commonly deficient in human cancers, the application prospects of M1 are very broad. It is reported that M1 kills cancer cells by inducing endoplasmic reticulum stress-mediated apoptosis.Citation141 However, the neutralizing antibodies are the major issues for in vivo treatment using M1. A recent study reported that liposome encapsulation of M1 can ensure the secure transportation of this virus and protect it from neutralizing antibodies.Citation142 Clinical trials for evaluation of M1 are awaiting the safety confirmation so far.Citation143

Future prospects

Oncolytic viruses as therapeutic agents for cancer biotherapy emerged when Oncorine was first approved by CFDA for the treatment of nasopharyngeal carcinoma. The US-FDA approval of T-VEC in 2015 also created a lot of interest in oncolytic virotherapy. Several viruses including vaccinia virus, reovirus, parvovirus, picornavirus have been assessed as potential candidates for oncolytic virotherapy. The major challenge in this therapy is the targeted delivery of the virus into the tumor. In most cases, systemic administration does not work well due to preexisting immunity. Therefore, virus delivery needs to be improved for effective systemic administration since intratumoral administration is expensive and difficult especially in cases of malignant gliomas. Some of the novel approaches involve the use of nanoparticles, complex viral particle ligands, and immuno-modulatory agents. Delivery of the virus into the tumor via nanoparticles uses a technologically complex image-guided delivery system. Alternatively, to secure the delivery of the oncolytic viruses via the blood stream, carrier cells that possess inherent tumor tropism have also been considered.Citation144 Another challenge is the optimization of combination therapies using oncolytic viruses along with the chemotherapeutic or immunotherapeutic drugs to get better and stable results. Immune response induction by oncolytic viruses after infection suppresses the replication of the virus thereby posing a hindrance to the effective functioning of the biotherapy intended to treat cancer. Presently, many oncolytic viruses are undergoing clinical trials for applications in single therapy or combination therapy, and most of them are safe and show almost no dose-limiting toxicities. Therefore, the use of oncolytic viruses in cancer biotherapy has the potential to be an ideal and painless therapeutic option for the cancer patients in future if the above-mentioned challenges are appropriately dealt with.

Disclosure of potential conflicts of interest

The authors declare no competing interests.

Author contributions

D.Z. conceived the study. M.M. and G.J. searched the literature regarding oncolytic viruses in cancer therapy. M.M. wrote the draft. D.Z., G.J. and P.H. edited the manuscript. All authors read and approved the final manuscript.

Additional information

Funding

This work was supported by grants from the National Natural Science Foundation [31670946, 31870922], and partly by the Strategic Priority Research Program of the Chinese Academy of Sciences [XBD29040000], and a postdoctoral fellowship from Guangzhou Women and Children’s Medical Center.

References

  • Bell J, McFadden G. Viruses for tumor therapy. Cell Host Microbe. 2014;15:260–65. doi:10.1016/j.chom.2014.01.002.
  • Kelly E, Russell SJ. History of oncolytic viruses: genesis to genetic engineering. Mol Ther. 2007;15:651–59. doi:10.1038/sj.mt.6300108.
  • Kaufman HL, Ruby CE, Hughes T, Slingluff CL Jr. Current status of granulocyte-macrophage colony-stimulating factor in the immunotherapy of melanoma. J Immunother Cancer. 2014;2:11. doi:10.1186/2051-1426-2-11.
  • Farassati F, Yang AD, Lee PW. Oncogenes in Ras signalling pathway dictate host-cell permissiveness to herpes simplex virus 1. Nat Cell Biol. 2001;3:745–50. doi:10.1038/35087061.
  • Smith KD, Mezhir JJ, Bickenbach K, Veerapong J, Charron J, Posner MC, Roizman B, Weichselbaum RR. Activated MEK suppresses activation of PKR and enables efficient replication and in vivo oncolysis by Deltagamma(1)34.5 mutants of herpes simplex virus 1. J Virol. 2006;80:1110–20. doi:10.1128/JVI.80.3.1110-1120.2006.
  • Kohlhapp FJ, Kaufman HL. Molecular pathways: mechanism of action for talimogene laherparepvec, a new oncolytic virus immunotherapy. Clin Cancer Res. 2016;22:1048–54. doi:10.1158/1078-0432.CCR-15-2667.
  • Tomazin R, van Schoot NE, Goldsmith K, Jugovic P, Sempe P, Fruh K, Johnson DC. Herpes simplex virus type 2 ICP47 inhibits human TAP but not mouse TAP. J Virol. 1998;72:2560–63. doi:10.1128/JVI.72.3.2560-2563.1998.
  • Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, Delman KA, Spitler LE, Puzanov I, Agarwala SS, et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. 2015;33:2780–88. doi:10.1200/JCO.2014.58.3377.
  • Everett AS, Pavlidakey PG, Contreras CM, De Los Santos JF, Kim JY, McKee SB, Kaufman HL, Conry RM. Chronic granulomatous dermatitis induced by talimogene laherparepvec therapy of melanoma metastases. J Cutan Pathol. 2018;45:48–53. doi:10.1111/cup.13048.
  • Esaki S, Goshima F, Kimura H, Murakami S, Nishiyama Y. Enhanced antitumoral activity of oncolytic herpes simplex virus with gemcitabine using colorectal tumor models. Int J Cancer. 2013;132:1592–601. doi:10.1002/ijc.27823.
  • Kim RD, Sarker D, Meyer T, Yau T, Macarulla T, Park JW, Choo SP, Hollebecque A, Sung MW, Lim HY, et al. First-in-human phase i study of fisogatinib (BLU-554) validates aberrant FGF19 Signaling as a driver event in hepatocellular carcinoma. Cancer Discovery. 2019;2019(9):1–12.
  • Shimoyama S, Goshima F, Teshigahara O, Kasuya H, Kodera Y, Nakao A, Nishiyama Y. Enhanced efficacy of herpes simplex virus mutant HF10 combined with paclitaxel in peritoneal cancer dissemination models. Hepatogastroenterology. 2007;54:1038–42.
  • Watanabe D, Goshima F, Mori I, Tamada Y, Matsumoto Y, Nishiyama Y. Oncolytic virotherapy for malignant melanoma with herpes simplex virus type 1 mutant HF10. J Dermatol Sci. 2008;50:185–96. doi:10.1016/j.jdermsci.2007.12.001.
  • Andtbacka RH, Ross M, Puzanov I, Milhem M, Collichio F, Delman KA, Amatruda T, Zager JS, Cranmer L, Hsueh E, et al. Patterns of clinical response with talimogene laherparepvec (T-VEC) in patients with melanoma treated in the OPTiM phase III clinical trial. Ann Surg Oncol. 2016;23:4169–77. doi:10.1245/s10434-016-5286-0.
  • Fujimoto Y, Mizuno T, Sugiura S, Goshima F, Kohno S, Nakashima T, Nishiyama Y. Intratumoral injection of herpes simplex virus HF10 in recurrent head and neck squamous cell carcinoma. Acta Otolaryngol. 2006;126:1115–17. doi:10.1080/00016480600702100.
  • Nakao A, Kasuya H, Sahin TT, Nomura N, Kanzaki A, Misawa M, Shirota T, Yamada S, Fujii T, Sugimoto H, et al. A phase I dose-escalation clinical trial of intraoperative direct intratumoral injection of HF10 oncolytic virus in non-resectable patients with advanced pancreatic cancer. Cancer Gene Ther. 2011;18:167–75. doi:10.1038/cgt.2010.65.
  • Ushijima Y, Luo C, Goshima F, Yamauchi Y, Kimura H, Nishiyama Y. Determination and analysis of the DNA sequence of highly attenuated herpes simplex virus type 1 mutant HF10, a potential oncolytic virus. Microbes Infect. 2007;9:142–49. doi:10.1016/j.micinf.2006.10.019.
  • Liu BL, Robinson M, Han ZQ, Branston RH, English C, Reay P, McGrath Y, Thomas SK, Thornton M, Bullock P, et al. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther. 2003;10:292–303. doi:10.1038/sj.gt.3301885.
  • Avitabile E, Lombardi G, Gianni T, Capri M, Campadelli-Fiume G. Coexpression of UL20p and gK inhibits cell-cell fusion mediated by herpes simplex virus glycoproteins gD, gH-gL, and wild-type gB or an endocytosis-defective gB mutant and downmodulates their cell surface expression. J Virol. 2004;78:8015–25. doi:10.1128/JVI.78.15.8015-8025.2004.
  • Avitabile E, Lombardi G, Campadelli-Fiume G. Herpes simplex virus glycoprotein K, but not its syncytial allele, inhibits cell-cell fusion mediated by the four fusogenic glycoproteins, gD, gB, gH, and gL. J Virol. 2003;77:6836–44. doi:10.1128/JVI.77.12.6836-6844.2003.
  • Perng GC, Dunkel EC, Geary PA, Slanina SM, Ghiasi H, Kaiwar R, Nesburn AB, Wechsler SL. The latency-associated transcript gene of herpes simplex virus type 1 (HSV-1) is required for efficient in vivo spontaneous reactivation of HSV-1 from latency. J Virol. 1994;68:8045–55. doi:10.1128/JVI.68.12.8045-8055.1994.
  • Streby KA, Geller JI, Currier MA, Warren PS, Racadio JM, Towbin AJ, Vaughan MR, Triplet M, Ott-Napier K, Dishman DJ, et al. Intratumoral injection of HSV1716, an oncolytic herpes virus, is safe and shows evidence of immune response and viral replication in young cancer patients. Clin Cancer Res. 2017;23:3566–74. doi:10.1158/1078-0432.CCR-16-2900.
  • Kim R, Sarker D, Macarulla T, Yau T, Choo SP, Meyer T, et al. 365OPhase 1 safety and clinical activity of BLU-554 in advanced hepatocellular carcinoma (HCC). Ann Oncol. 2017;28. doi:10.1093/annonc/mdx075.
  • Aghi MK, Chiocca EA. Phase ib trial of oncolytic herpes virus G207 shows safety of multiple injections and documents viral replication. Mol Ther. 2009;17:8–9. doi:10.1038/mt.2008.275.
  • Markert JM, Medlock MD, Rabkin SD, Gillespie GY, Todo T, Hunter WD, Palmer CA, Feigenbaum F, Tornatore C, Tufaro F, et al. Conditionally replicating herpes simplex virus mutant, G207 for the treatment of malignant glioma: results of a phase I trial. Gene Ther. 2000;7:867–74. doi:10.1038/sj.gt.3301205.
  • Markert JM, Liechty PG, Wang W, Gaston S, Braz E, Karrasch M, Nabors LB, Markiewicz M, Lakeman AD, Palmer CA, et al. Phase Ib trial of mutant herpes simplex virus G207 inoculated pre-and post-tumor resection for recurrent GBM. Mol Ther. 2009;17:199–207. doi:10.1038/mt.2008.228.
  • Patel DM, Foreman PM, Nabors LB, Riley KO, Gillespie GY, Markert JM. Design of a phase I clinical trial to evaluate M032, a genetically engineered HSV-1 expressing IL-12, in patients with recurrent/progressive glioblastoma multiforme, anaplastic astrocytoma, or gliosarcoma. Hum Gene Ther Clin Dev. 2016;27:69–78. doi:10.1089/humc.2016.031.
  • Zeng W, Hu P, Wu J, Wang J, Li J, Lei L, LIU R. The oncolytic herpes simplex virus vector G47 effectively targets breast cancer stem cells. Oncol Rep. 2013;29:1108–14. doi:10.3892/or.2012.2211.
  • Fukuhara H, Martuza RL, Rabkin SD, Ito Y, Todo T. Oncolytic herpes simplex virus vector g47delta in combination with androgen ablation for the treatment of human prostate adenocarcinoma. Clin Cancer Res. 2005;11:7886–90. doi:10.1158/1078-0432.CCR-05-1090.
  • Cerullo V, Koski A, Vaha-Koskela M, Hemminki A. Chapter eight–Oncolytic adenoviruses for cancer immunotherapy: data from mice, hamsters, and humans. Adv Cancer Res. 2012;115:265–318.
  • Liang M. Clinical development of oncolytic viruses in China. Curr Pharm Biotechnol. 2012;13:1852–57. doi:10.2174/138920112800958760.
  • Raty JK, Pikkarainen JT, Wirth T, Yla-Herttuala S. Gene therapy: the first approved gene-based medicines, molecular mechanisms and clinical indications. Curr Mol Pharmacol. 2008;1:13–23. doi:10.2174/1874467210801010013.
  • Xia ZJ, Chang JH, Zhang L, Jiang WQ, Guan ZZ, Liu JW, Zhang Y, Hu XH, Wu GH, Wang HQ. Phase III randomized clinical trial of intratumoral injection of E1B gene-deleted adenovirus (H101) combined with cisplatin-based chemotherapy in treating squamous cell cancer of head and neck or esophagus. Ai Zheng. 2004;23:1666–70.
  • Liang M. Oncorine, the world first oncolytic virus medicine and its update in China. Curr Cancer Drug Targets. 2018;18:171–76. doi:10.2174/1568009618666171129221503.
  • Rudin CM, Cohen EE, Papadimitrakopoulou VA, Silverman S Jr., Recant W, El-Naggar AK, Stenson K, Lippman SM, Hong WK, Vokes EE, et al. An attenuated adenovirus, ONYX-015, as mouthwash therapy for premalignant oral dysplasia. J Clin Oncol. 2003;21:4546–52. doi:10.1200/JCO.2003.03.544.
  • Hecht JR, Bedford R, Abbruzzese JL, Lahoti S, Reid TR, Soetikno RM, Kirn DH, Freeman SM. A phase I/II trial of intratumoral endoscopic ultrasound injection of ONYX-015 with intravenous gemcitabine in unresectable pancreatic carcinoma. Clin Cancer Res. 2003;9:555–61.
  • Fueyo J, Alemany R, Gomez-Manzano C, Fuller GN, Khan A, Conrad CA, Liu T-J, Jiang H, Lemoine MG, Suzuki K, et al. Preclinical characterization of the antiglioma activity of a tropism-enhanced adenovirus targeted to the retinoblastoma pathway. J Natl Cancer Inst. 2003;95:652–60. doi:10.1093/jnci/95.9.652.
  • Lang FF, Conrad C, Gomez-Manzano C, Yung WKA, Sawaya R, Weinberg JS, Prabhu SS, Rao G, Fuller GN, Aldape KD, et al. Phase I study of DNX-2401 (Delta-24-RGD) oncolytic adenovirus: replication and immunotherapeutic effects in recurrent malignant glioma. J Clin Oncol. 2018;36:1419–27. doi:10.1200/JCO.2017.75.8219.
  • Eissa IR, Bustos-Villalobos I, Ichinose T, Matsumura S, Naoe Y, Miyajima N, Morimoto D, Mukoyama N, Zhiwen W, Tanaka M, et al. The current status and future prospects of oncolytic viruses in clinical trials against melanoma, glioma, pancreatic, and breast cancers. Cancers (Basel). 2018;10:356–78. doi:10.3390/cancers10100356.
  • Ranki T, Pesonen S, Hemminki A, Partanen K, Kairemo K, Alanko T, Lundin J, Linder N, Turkki R, Ristimäki A, et al. Phase I study with ONCOS-102 for the treatment of solid tumors - an evaluation of clinical response and exploratory analyses of immune markers. J Immunother Cancer. 2016;4:17. doi:10.1186/s40425-016-0121-5.
  • Vera B, Martinez-Velez N, Xipell E, Acanda de la Rocha A, Patino-Garcia A, Saez-Castresana J, Gonzalez-Huarriz M, Cascallo M, Alemany R, Alonso MM, et al. Characterization of the antiglioma effect of the oncolytic adenovirus VCN-01. PLoS One. 2016;11:e0147211. doi:10.1371/journal.pone.0147211.
  • Pascual-Pasto G, Bazan-Peregrino M, Olaciregui NG, Restrepo-Perdomo CA, Mato-Berciano A, Ottaviani D, Weber K, Correa G, Paco S, Vila-Ubach M, et al. Therapeutic targeting of the RB1 pathway in retinoblastoma with the oncolytic adenovirus VCN-01. Sci Transl Med. 2019;11:eaat9321. doi:10.1126/scitranslmed.aat9321.
  • Nokisalmi P, Pesonen S, Escutenaire S, Sarkioja M, Raki M, Cerullo V, Laasonen L, Alemany R, Rojas J, Cascallo M, et al. Oncolytic adenovirus ICOVIR-7 in patients with advanced and refractory solid tumors. Clin Cancer Res. 2010;16:3035–43. doi:10.1158/1078-0432.CCR-09-3167.
  • Garcia M, Moreno R, Gil-Martin M, Cascallo M, de Olza MO, Cuadra C, Piulats JM, Navarro V, Domenech M, Alemany R, et al. A phase 1 trial of oncolytic adenovirus ICOVIR-5 administered intravenously to cutaneous and uveal melanoma patients. Hum Gene Ther. 2019;30:352–64. doi:10.1089/hum.2018.107.
  • ONCORINE. Recombinant human adenovirus type 5 Injection. [accessed 2019 September]. http://www.sunwaybio.com.cn/en/product.html
  • Khuri FR, Nemunaitis J, Ganly I, Arseneau J, Tannock IF, Romel L, Gore M, Ironside J, MacDougall RH, Heise C, et al. a controlled trial of intratumoral ONYX-015, a selectively-replicating adenovirus, in combination with cisplatin and 5-fluorouracil in patients with recurrent head and neck cancer. Nat Med. 2000;6:879–85. doi:10.1038/78638.
  • Parato KA, Senger D, Forsyth PA, Bell JC. Recent progress in the battle between oncolytic viruses and tumours. Nat Rev Cancer. 2005;5:965–76. doi:10.1038/nrc1750.
  • Smith E, Breznik J, Lichty BD. Strategies to enhance viral penetration of solid tumors. Hum Gene Ther. 2011;22:1053–60. doi:10.1089/hum.2010.227.
  • Strauss R, Lieber A. Anatomical and physical barriers to tumor targeting with oncolytic adenoviruses in vivo. Curr Opin Mol Ther. 2009;11:513–22.
  • Jacobetz MA, Chan DS, Neesse A, Bapiro TE, Cook N, Frese KK, Feig C, Nakagawa T, Caldwell ME, Zecchini HI, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013;62:112–20. doi:10.1136/gutjnl-2012-302529.
  • Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21:418–29. doi:10.1016/j.ccr.2012.01.007.
  • Thompson CB, Shepard HM, O’Connor PM, Kadhim S, Jiang P, Osgood RJ, Bookbinder LH, Li X, Sugarman BJ, Connor RJ, et al. Enzymatic depletion of tumor hyaluronan induces antitumor responses in preclinical animal models. Mol Cancer Ther. 2010;9:3052–64. doi:10.1158/1535-7163.MCT-10-0470.
  • Eriksson E, Milenova I, Wenthe J, Stahle M, Leja-Jarblad J, Ullenhag G, Dimberg A, Moreno R, Alemany R, Loskog A, et al. Shaping the tumor stroma and sparking immune activation by CD40 and 4-1BB signaling induced by an armed oncolytic virus. Clin Cancer Res. 2017;23:5846–57. doi:10.1158/1078-0432.CCR-17-0285.
  • Dong W, van Ginkel JW, Au KY, Alemany R, Meulenberg JJ, van Beusechem VW. ORCA-010, a novel potency-enhanced oncolytic adenovirus, exerts strong antitumor activity in preclinical models. Hum Gene Ther. 2014;25:897–904. doi:10.1089/hum.2013.229.
  • Gros A, Martinez-Quintanilla J, Puig C, Guedan S, Mollevi DG, Alemany R, Cascallo M. Bioselection of a gain of function mutation that enhances adenovirus 5 release and improves its antitumoral potency. Cancer Res. 2008;68:8928–37. doi:10.1158/0008-5472.CAN-08-1145.
  • Ramesh N, Ge Y, Ennist DL, Zhu M, Mina M, Ganesh S, Reddy PS, Yu DC. CG0070, a conditionally replicating granulocyte macrophage colony-stimulating factor–armed oncolytic adenovirus for the treatment of bladder cancer. Clin Cancer Res. 2006;12:305–13. doi:10.1158/1078-0432.CCR-05-1059.
  • Packiam VT, Lamm DL, Barocas DA, Trainer A, Fand B, Davis RL 3rd, Clark W, Kroeger M, Dumbadze I, Chamie K, et al. An open label, single-arm, phase II multicenter study of the safety and efficacy of CG0070 oncolytic vector regimen in patients with BCG-unresponsive non-muscle-invasive bladder cancer: interim results. Urol Oncol. 2018;36:440–47. doi:10.1016/j.urolonc.2017.07.005.
  • Cheng T, Song Y, Zhang Y, Zhang C, Yin J, Chi Y, Zhou D. A novel oncolytic adenovirus based on simian adenovirus serotype 24. Oncotarget. 2017;8:26871–85. doi:10.18632/oncotarget.15845.
  • Xing M, Wang X, Chi Y, Zhou D. Gene therapy for colorectal cancer using adenovirus-mediated full-length antibody, cetuximab. Oncotarget. 2016;7:28262–72. doi:10.18632/oncotarget.v7i19.
  • Abbink P, Lemckert AA, Ewald BA, Lynch DM, Denholtz M, Smits S, Holterman L, Damen I, Vogels R, Thorner AR, et al. Comparative seroprevalence and immunogenicity of six rare serotype recombinant adenovirus vaccine vectors from subgroups B and D. J Virol. 2007;81:4654–63. doi:10.1128/JVI.02696-06.
  • Waddington SN, McVey JH, Bhella D, Parker AL, Barker K, Atoda H, Pink R, Buckley SMK, Greig JA, Denby L, et al. Adenovirus serotype 5 hexon mediates liver gene transfer. Cell. 2008;132:397–409. doi:10.1016/j.cell.2008.01.016.
  • Uchino J, Curiel DT, Ugai H, Glorioso JC. Species D human adenovirus type 9 exhibits better virus-spread ability for antitumor efficacy among alternative serotypes. PLoS One. 2014;9:e87342. doi:10.1371/journal.pone.0087342.
  • Kaufmann JK, Nettelbeck DM. Virus chimeras for gene therapy, vaccination, and oncolysis: adenoviruses and beyond. Trends Mol Med. 2012;18:365–76. doi:10.1016/j.molmed.2012.04.008.
  • Schoggins JW, Nociari M, Philpott N, Falck-Pedersen E. Influence of fiber detargeting on adenovirus-mediated innate and adaptive immune activation. J Virol. 2005;79:11627–37. doi:10.1128/JVI.79.18.11627-11637.2005.
  • Parker AL, Waddington SN, Buckley SM, Custers J, Havenga MJ, van Rooijen N, Goudsmit J, McVey JH, Nicklin SA, Baker AH, et al. Effect of neutralizing sera on factor x-mediated adenovirus serotype 5 gene transfer. J Virol. 2009;83:479–83. doi:10.1128/JVI.01878-08.
  • Rogee S, Grellier E, Bernard C, Jouy N, Loyens A, Beauvillain JC, Fender P, Corjon S, Hong SS, Boulanger P, et al. Influence of chimeric human-bovine fibers on adenoviral uptake by liver cells and the antiviral immune response. Gene Ther. 2010;17:880–91. doi:10.1038/gt.2010.37.
  • Coughlan L, Alba R, Parker AL, Bradshaw AC, McNeish IA, Nicklin SA, Baker AH. Tropism-modification strategies for targeted gene delivery using adenoviral vectors. Viruses. 2010;2:2290–355. doi:10.3390/v2102290.
  • Majhen D, Calderon H, Chandra N, Fajardo CA, Rajan A, Alemany R, Custers J. Adenovirus-based vaccines for fighting infectious diseases and cancer: progress in the field. Hum Gene Ther. 2014;25:301–17. doi:10.1089/hum.2013.235.
  • Magnusson MK, Hong SS, Boulanger P, Lindholm L. Genetic retargeting of adenovirus: novel strategy employing “deknobbing” of the fiber. J Virol. 2001;75:7280–89. doi:10.1128/JVI.75.16.7280-7289.2001.
  • O’Riordan CR, Lachapelle A, Delgado C, Parkes V, Wadsworth SC, Smith AE, Francis GE. PEGylation of adenovirus with retention of infectivity and protection from neutralizing antibody in vitro and in vivo. Hum Gene Ther. 1999;10:1349–58. doi:10.1089/10430349950018021.
  • Lee YS, Kim SW. Bioreducible polymers for therapeutic gene delivery. J Control Release. 2014;190:424–39. doi:10.1016/j.jconrel.2014.04.012.
  • Douglas JT, Miller CR, Kim M, Dmitriev I, Mikheeva G, Krasnykh V, Curiel DT. A system for the propagation of adenoviral vectors with genetically modified receptor specificities. Nat Biotechnol. 1999;17:470–75. doi:10.1038/8647.
  • Rancourt C, Robertson MW 3rd, Wang M, Goldman CK, Kelly JF, Alvarez RD, Siegal GP, Curiel DT. Endothelial cell vehicles for delivery of cytotoxic genes as a gene therapy approach for carcinoma of the ovary. Clin Cancer Res. 1998;4:265–70.
  • Dmitriev I, Kashentseva E, Rogers BE, Krasnykh V, Curiel DT. Ectodomain of coxsackievirus and adenovirus receptor genetically fused to epidermal growth factor mediates adenovirus targeting to epidermal growth factor receptor-positive cells. J Virol. 2000;74:6875–84. doi:10.1128/JVI.74.15.6875-6884.2000.
  • van Beusechem VW, Mastenbroek DC, van den Doel PB, Lamfers ML, Grill J, Wurdinger T, Haisma HJ, Pinedo HM, Gerritsen WR. Conditionally replicative adenovirus expressing a targeting adapter molecule exhibits enhanced oncolytic potency on CAR-deficient tumors. Gene Ther. 2003;10:1982–91. doi:10.1038/sj.gt.3302103.
  • Bhattacharjee S, Yadava PK. Measles virus: background and oncolytic virotherapy. Biochem Biophys Rep. 2018;13:58–62. doi:10.1016/j.bbrep.2017.12.004.
  • Enders JF, Peebles TC. Propagation in tissue cultures of cytopathogenic agents from patients with measles. Proc Soc Exp Biol Med. 1954;86:277–86. doi:10.3181/00379727-86-21073.
  • Enders JF, Katz SL, Milovanovic MV, Holloway A. Studies on an attenuated measles-virus vaccine. I. Development and preparations of the vaccine: technics for assay of effects of vaccination. N Engl J Med. 1960;263:153–59. doi:10.1056/NEJM196007282630401.
  • Hashiguchi T, Kajikawa M, Maita N, Takeda M, Kuroki K, Sasaki K, Kohda D, Yanagi Y, Maenaka K. Crystal structure of measles virus hemagglutinin provides insight into effective vaccines. Proc Natl Acad Sci USA. 2007;104:19535–40. doi:10.1073/pnas.0707830104.
  • Russell SJ, Federspiel MJ, Peng KW, Tong C, Dingli D, Morice WG, Lowe V, O’Connor MK, Kyle RA, Leung N, et al. Remission of disseminated cancer after systemic oncolytic virotherapy. Mayo Clin Proc. 2014;89:926–33. doi:10.1016/j.mayocp.2014.04.003.
  • Mader EK, Maeyama Y, Lin Y, Butler GW, Russell HM, Galanis E, Russell SJ, Dietz AB, Peng K-W. Mesenchymal stem cell carriers protect oncolytic measles viruses from antibody neutralization in an orthotopic ovarian cancer therapy model. Clin Cancer Res. 2009;15:7246–55. doi:10.1158/1078-0432.CCR-09-1292.
  • Miest TS, Yaiw KC, Frenzke M, Lampe J, Hudacek AW, Springfeld C, von Messling V, Ungerechts G, Cattaneo R. Envelope-chimeric entry-targeted measles virus escapes neutralization and achieves oncolysis. Mol Ther. 2011;19:1813–20. doi:10.1038/mt.2011.92.
  • Thirukkumaran C, Shi ZQ, Thirukkumaran P, Luider J, Kopciuk K, Spurrell J, Elzinga K, Morris D. PUMA and NF-kB are cell signaling predictors of reovirus oncolysis of breast cancer. PLoS One. 2017;12:e0168233. doi:10.1371/journal.pone.0168233.
  • Samson A, Bentham MJ, Scott K, Nuovo G, Bloy A, Appleton E, Adair RA, Dave R, Peckham-Cooper A, Toogood G, et al. Oncolytic reovirus as a combined antiviral and anti-tumour agent for the treatment of liver cancer. Gut. 2018;67:562–73. doi:10.1136/gutjnl-2016-312009.
  • Morris DG, Feng X, DiFrancesco LM, Fonseca K, Forsyth PA, Paterson AH, Coffey MC, Thompson B. REO-001: A phase I trial of percutaneous intralesional administration of reovirus type 3 dearing (Reolysin(R)) in patients with advanced solid tumors. Invest New Drugs. 2013;31:696–706. doi:10.1007/s10637-012-9865-z.
  • Lolkema MP, Arkenau HT, Harrington K, Roxburgh P, Morrison R, Roulstone V, Twigger K, Coffey M, Mettinger K, Gill G, et al. A phase I study of the combination of intravenous reovirus type 3 Dearing and gemcitabine in patients with advanced cancer. Clin Cancer Res. 2011;17:581–88. doi:10.1158/1078-0432.CCR-10-2159.
  • Forsyth P, Roldan G, George D, Wallace C, Palmer CA, Morris D, Cairncross G, Matthews MV, Markert J, Gillespie Y, et al. A phase I trial of intratumoral administration of reovirus in patients with histologically confirmed recurrent malignant gliomas. Mol Ther. 2008;16:627–32. doi:10.1038/sj.mt.6300403.
  • Harrington KJ, Karapanagiotou EM, Roulstone V, Twigger KR, White CL, Vidal L, Beirne D, Prestwich R, Newbold K, Ahmed M, et al. Two-stage phase I dose-escalation study of intratumoral reovirus type 3 dearing and palliative radiotherapy in patients with advanced cancers. Clin Cancer Res. 2010;16:3067–77.
  • Vidal L, Pandha HS, Yap TA, White CL, Twigger K, Vile RG, Melcher A, Coffey M, Harrington KJ, DeBono JS, et al. A phase I study of intravenous oncolytic reovirus type 3 Dearing in patients with advanced cancer. Clin Cancer Res. 2008;14:7127–37. doi:10.1158/1078-0432.CCR-08-0524.
  • Chakrabarty R, Tran H, Selvaggi G, Hagerman A, Thompson B, Coffey M. The oncolytic virus, pelareorep, as a novel anticancer agent: a review. Invest New Drugs. 2015;33:761–74. doi:10.1007/s10637-015-0216-8.
  • ORYX announces positive phase I/IIa data for oncolytic virus parvoryx demonstrating safety, anti-tumor effects, partial responses and prolonged overall survival in metastatic, inoperable pancreatic cancer. [accessed 2019 September]. https://www.biospace.com/article/oryx-announces-positive-phase-i-iia-data-for-oncolytic-virus-parvoryx-demonstrating-safety-anti-tumor-effects-partial-responses-and-prolonged-overall-survival-in-metastatic-inoperable-pancreatic-cancer/
  • Adams WR, Prince AF. Cellular changes associated with infection of the Ehrlich ascites tumor with Newcastle disease virus. Ann N Y Acad Sci. 1959;81:89–100. doi:10.1111/j.1749-6632.1959.tb49298.x.
  • Mason EJ, Kaufman N. The toxic properties of massive inoculums of Newcastle disease virus and influenza virus (PR8) for cell strains derived from normal and neoplastic tissue. Am J Pathol. 1960;37:231–43.
  • Eaton MD, Levinthal JD, Scala AR. Contribution of antiviral immunity to oncolysis by Newcastle disease virus in a murine lymphoma. J Natl Cancer Inst. 1967;39:1089–97.
  • Eaton SA MD. Further observations on the inhibitory effect of myxoviruses on a transplantable murine leukemia. Proc Soc Exp Biol Med. 1969;132(1):20–26. doi:10.3181/00379727-132-34138.
  • Vigil A, Park MS, Martinez O, Chua MA, Xiao S, Cros JF, Martínez-Sobrido L, Woo SLC, García-Sastre A. Use of reverse genetics to enhance the oncolytic properties of Newcastle disease virus. Cancer Res. 2007;67:8285–92. doi:10.1158/0008-5472.CAN-07-1025.
  • Zamarin D, Martinez-Sobrido L, Kelly K, Mansour M, Sheng G, Vigil A, García-Sastre A, Palese P, Fong Y. Enhancement of oncolytic properties of recombinant newcastle disease virus through antagonism of cellular innate immune responses. Mol Ther. 2009;17:697–706. doi:10.1038/mt.2008.286.
  • Ahlert T, Schirrmacher V. Isolation of a human melanoma adapted Newcastle disease virus mutant with highly selective replication patterns. Cancer Res. 1990;50:5962–68.
  • Li P, Chen CH, Li S, Givi B, Yu Z, Zamarin D, Palese P, Fong Y, Wong RJ. Therapeutic effects of a fusogenic newcastle disease virus in treating head and neck cancer. Head Neck. 2011;33:1394–99. doi:10.1002/hed.v33.10.
  • Silberhumer GR, Brader P, Wong J, Serganova IS, Gonen M, Gonzalez SJ, Blasberg R, Zamarin D, Fong Y. Genetically engineered oncolytic Newcastle disease virus effectively induces sustained remission of malignant pleural mesothelioma. Mol Cancer Ther. 2010;9:2761–69. doi:10.1158/1535-7163.MCT-10-0090.
  • Song KY, Wong J, Gonzalez L, Sheng G, Zamarin D, Fong Y. Antitumor efficacy of viral therapy using genetically engineered Newcastle disease virus [NDV(F3aa)-GFP] for peritoneally disseminated gastric cancer. J Mol Med (Berl). 2010;88:589–96. doi:10.1007/s00109-010-0605-6.
  • Fabian Z, Torocsik B, Kiss K, Csatary LK, Bodey B, Tigyi J, Csatary C, Szeberényi J. Induction of apoptosis by a Newcastle disease virus vaccine (MTH-68/H) in PC12 rat phaeochromocytoma cells. Anticancer Res. 2001;21:125–35.
  • Szeberenyi J, Fabian Z, Torocsik B, Kiss K, Csatary LK. Newcastle disease virus-induced apoptosis in PC12 pheochromocytoma cells. Am J Ther. 2003;10:282–88. doi:10.1097/00045391-200307000-00008.
  • Lorence RMRP, Rood PA. Newcastle disease virus as an antineoplastic agent: induction of tumor necrosisfactor-a and augmentation of its cytotoxicity. J Natl Cancer Inst. 1988;80(16):1305–12. doi:10.1093/jnci/80.16.1305.
  • Zorn U, Dallmann I, Grosse J, Kirchner H, Poliwoda H, Atzpodien J. Induction of cytokines and cytotoxicity against tumor cells by Newcastle disease virus. Cancer Biother. 1994;9:225–35. doi:10.1089/cbr.1994.9.225.
  • Termeer CC, Schirrmacher V, Brocker EB, Becker JC. Newcastle disease virus infection induces B7-1/B7-2-independent T-cell costimulatory activity in human melanoma cells. Cancer Gene Ther. 2000;7:316–23. doi:10.1038/sj.cgt.7700109.
  • Lorence RM, Katubig BB, Reichard KW, Reyes HM, Phuangsab A, Sassetti MD, Walter RJ, Peeples ME. Complete regression of human fibrosarcoma xenografts after local Newcastle disease virus therapy. Cancer Res. 1994;54:6017–21.
  • Zamarin D, Vigil A, Kelly K, Garcia-Sastre A, Fong Y. Genetically engineered Newcastle disease virus for malignant melanoma therapy. Gene Ther. 2009;16:796–804. doi:10.1038/gt.2009.14.
  • Altomonte J, Marozin S, Schmid RM, Ebert O. Engineered newcastle disease virus as an improved oncolytic agent against hepatocellular carcinoma. Mol Ther. 2010;18:275–84. doi:10.1038/mt.2009.231.
  • Reichard KW, Lorence RM, Katubig BB, Peeples ME, Reyes HM. Retinoic acid enhances killing of neuroblastoma cells by Newcastle disease virus. J Pediatr Surg. 1993;28:1221–25. discussion 1225–1226. doi:10.1016/S0022-3468(05)80302-1.
  • Lorence RM, Reichard KW, Katubig BB, Reyes HM, Phuangsab A, Mitchell BR, Cascino CJ, Walter RJ, Peeples ME. Complete regression of human neuroblastoma xenografts in athymic mice after local Newcastle disease virus therapy. J Natl Cancer Inst. 1994;86:1228–33. doi:10.1093/jnci/86.16.1228.
  • Tzadok-David Y, Metzkin-Eizenberg M, Zakay-Rones Z. The effect of a mesogenic and a lentogenic Newcastle disease virus strain on Burkitt lymphoma Daudi cells. J Cancer Res Clin Oncol. 1995;121:169–74. doi:10.1007/BF01198099.
  • Bar-Eli N, Giloh H, Schlesinger M, Zakay-Rones Z. Preferential cytotoxic effect of Newcastle disease virus on lymphoma cells. J Cancer Res Clin Oncol. 1996;122:409–15. doi:10.1007/BF01212880.
  • Schirrmacher V, Griesbach A, Ahlert T. Antitumor effects of Newcastle disease virus in vivo: local versus systemic effects. Int J Oncol. 2001;18:945–52. doi:10.3892/ijo.18.5.945.
  • Washburn B, Schirrmacher V. Human tumor cell infection by Newcastle disease virus leads to upregulation of HLA and cell adhesion molecules and to induction of interferons, chemokines and finally apoptosis. Int J Oncol. 2002;21:85–93. doi:10.3892/ijo.21.1.85.
  • Zulkifli MM, Ibrahim R, Ali AM, Aini I, Jaafar H, Hilda SS, Alitheen NB, Abdullah JM. Newcastle diseases virus strain V4UPM displayed oncolytic ability against experimental human malignant glioma. Neurol Res. 2009;31:3–10. doi:10.1179/174313208X325218.
  • Cassel WA, Murray DR, Torbin AH, Olkowski ZL, Moore ME. Viral oncolysate in the management of malignant melanoma. I. Preparation of the oncolysate and measurement of immunologic responses. Cancer. 1977;40:672–79.
  • Murray DR, Cassel WA, Torbin AH, Olkowski ZL, Moore ME. Viral oncolysate in the management of malignant melanoma. II. Clinical studies. Cancer. 1977;40:680–86.
  • Cassel WA, Murray DR. Treatment of stage II malignant melanoma patients with a Newcastle disease virus oncolysate. Nat Immun Cell Growth Regul. 1988;7:351–52.
  • Cassel WA, Murray DR. A ten-year follow-up on stage II malignant melanoma patients treated postsurgically with Newcastle disease virus oncolysate. Med Oncol Tumor Pharmacother. 1992;9:169–71. doi:10.1007/bf02987752.
  • Liebrich W, Schlag P, Manasterski M, Lehner B, Stohr M, Moller P, Schirrmacher V. In vitro and clinical characterisation of a Newcastle disease virus-modified autologous tumour cell vaccine for treatment of colorectal cancer patients. Eur J Cancer. 1991;27:703–10. doi:10.1016/0277-5379(91)90170-I.
  • Schlag P, Manasterski M, Gerneth T, Hohenberger P, Dueck M, Herfarth C, Liebrich W, Schirrmacher V. Active specific immunotherapy with Newcastle-disease-virus-modified autologous tumor cells following resection of liver metastases in colorectal cancer. First evaluation of clinical response of a phase II-trial. Cancer Immunol Immunother. 1992;35:325–30. doi:10.1007/BF01741145.
  • Schulze T, Kemmner W, Weitz J, Wernecke KD, Schirrmacher V, Schlag PM. Efficiency of adjuvant active specific immunization with Newcastle disease virus modified tumor cells in colorectal cancer patients following resection of liver metastases: results of a prospective randomized trial. Cancer Immunol Immunother. 2009;58:61–69. doi:10.1007/s00262-008-0526-1.
  • Ockert D, Schirrmacher V, Beck N, Stoelben E, Ahlert T, Flechtenmacher J, Hagmüller E, Buchcik R, Nagel M, Saeger HD, et al. Newcastle disease virus-infected intact autologous tumor cell vaccine for adjuvant active specific immunotherapy of resected colorectal carcinoma. Clin Cancer Res. 1996;2:21–28.
  • Liang W, Wang H, Sun TM, Yao WQ, Chen LL, Jin Y, Li CL, Meng FJ. Application of autologous tumor cell vaccine and NDV vaccine in treatment of tumors of digestive tract. World J Gastroenterol. 2003;9:495–98. doi:10.3748/wjg.v9.i3.495.
  • Pomer S, Schirrmacher V, Thiele R, Lohrke H, Brkovic D, Staehler G. Tumor response and 4 year survival-data of patients with advanced renal-cell carcinoma treated with autologous tumor vaccine and subcutaneous R-IL-2 and IFN-alpha(2b). Int J Oncol. 1995;6:947–54. doi:10.3892/ijo.6.5.947.
  • Elankumaran S, Chavan V, Qiao D, Shobana R, Moorkanat G, Biswas M, Samal SK. Type I interferon-sensitive recombinant newcastle disease virus for oncolytic virotherapy. J Virol. 2010;84:3835–44. doi:10.1128/JVI.01553-09.
  • Wilden H, Fournier P, Zawatzky R, Schirrmacher V. Expression of RIG-I, IRF3, IFN-beta and IRF7 determines resistance or susceptibility of cells to infection by Newcastle disease virus. Int J Oncol. 2009;34:971–82. doi:10.3892/ijo_00000223.
  • Fiola C, Peeters B, Fournier P, Arnold A, Bucur M, Schirrmacher V. Tumor selective replication of Newcastle disease virus: association with defects of tumor cells in antiviral defence. Int J Cancer. 2006;119:328–38. doi:10.1002/(ISSN)1097-0215.
  • Krishnamurthy S, Takimoto T, Scroggs RA, Portner A. Differentially regulated interferon response determines the outcome of Newcastle disease virus infection in normal and tumor cell lines. J Virol. 2006;80:5145–55. doi:10.1128/JVI.02618-05.
  • Mansour M, Palese P, Zamarin D. Oncolytic specificity of Newcastle disease virus is mediated by selectivity for apoptosis-resistant cells. J Virol. 2011;85:6015–23. doi:10.1128/JVI.01537-10.
  • Lazar I, Yaacov B, Shiloach T, Eliahoo E, Kadouri L, Lotem M, Perlman R, Zakay-Rones Z, Panet A, Ben-Yehuda D, et al. The oncolytic activity of Newcastle disease virus NDV-HUJ on chemoresistant primary melanoma cells is dependent on the proapoptotic activity of the inhibitor of apoptosis protein Livin. J Virol. 2010;84:639–46. doi:10.1128/JVI.00401-09.
  • Wu Y, Zhang X, Wang X, Wang L, Hu S, Liu X, Meng S. Apoptin enhances the oncolytic properties of Newcastle disease virus. Intervirology. 2012;55:276–86. doi:10.1159/000328325.
  • Maamary J, Array F, Gao Q, Garcia-Sastre A, Steinman RM, Palese P, Nchinda G. Newcastle disease virus expressing a dendritic cell-targeted HIV gag protein induces a potent gag-specific immune response in mice. J Virol. 2011;85:2235–46. doi:10.1128/JVI.02036-10.
  • Puhler F, Willuda J, Puhlmann J, Mumberg D, Romer-Oberdorfer A, Beier R. Generation of a recombinant oncolytic Newcastle disease virus and expression of a full IgG antibody from two transgenes. Gene Ther. 2008;15:371–83. doi:10.1038/sj.gt.3303095.
  • About CAVATAK®. [accessed 2019 September]. https://www.viralytics.com/our-pipeline/cavatak/cavataktm/
  • [accessed 2019 September].https://www.cancertherapyadvisor.com/home/cancer-topics/general-oncology/phase-3-trial-for-oncolytic-viral-therapy-pexa-vec-in-advanced-liver-cancer-terminated-early/
  • light-emitting oncolytic vaccinia virus GL-ONC1. [accessed 2019 September]. https://www.cancer.gov/publications/dictionaries/cancer-drug/def/light-emitting-oncolytic-vaccinia-virus-gl-onc1.
  • Downs-Canner S, Guo ZS, Ravindranathan R, Breitbach CJ, O’Malley ME, Jones HL, Moon A, McCart JA, Shuai Y, Zeh HJ, et al. Phase 1 study of intravenous oncolytic poxvirus (vvDD) in patients with advanced solid cancers. Mol Ther. 2016;24:1492–501. doi:10.1038/mt.2016.101.
  • Zhang H, Lin Y, Li K, Liang J, Xiao X, Cai J, Tan Y, Xing F, Mai J, Li Y, et al. Naturally existing oncolytic virus M1 is nonpathogenic for the Nonhuman primates after multiple rounds of repeated intravenous injections. Hum Gene Ther. 2016;27:700–11. doi:10.1089/hum.2016.038.
  • Lin Y, Zhang H, Liang J, Li K, Zhu W, Fu L, Wang F, Zheng X, Shi H, Wu S, et al. Identification and characterization of alphavirus M1 as a selective oncolytic virus targeting ZAP-defective human cancers. Proc Natl Acad Sci USA. 2014;111:E4504–4512. doi:10.1073/pnas.1408759111.
  • Wang Y, Huang H, Zou H, Tian X, Hu J, Qiu P, Hu H, Yan G. Liposome encapsulation of oncolytic virus M1 to reduce immunogenicity and immune clearance in vivo. Mol Pharm. 2019;16:779–85. doi:10.1021/acs.molpharmaceut.8b01046.
  • Lundstrom K. Oncolytic alphaviruses in cancer immunotherapy. Vaccines (Basel). 2017;5:9. doi:10.3390/vaccines5020009.
  • Roy DG, Bell JC. Cell carriers for oncolytic viruses: current challenges and future directions. Oncolytic Virother. 2013;2:47–56. doi:10.2147/OV.S36623.

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.