Skip to main content
Erschienen in: BMC Immunology 1/2018

Open Access 01.12.2018 | Methodology article

Oncolytic herpes simplex virus and immunotherapy

verfasst von: Wenqing Ma, Hongbin He, Hongmei Wang

Erschienen in: BMC Immunology | Ausgabe 1/2018

Abstract

Background

Oncolytic viruses have been proposed to be employed as a potential treatment of cancer. Well targeted, they will serve the purpose of cracking tumor cells without causing damage to normal cells. In this category of oncolytic viral drugs human pathogens herpes simplex virus (HSV) is especially suitable for the cause. Although most viral infection causes antiviral reaction in the host, HSV has multiple mechanisms to evade those responses. Powerful anti-tumor effect can thus be achieved via genetic manipulation of the HSV genes involved in this evading mechanism, namely deletions or mutations that adapt its function towards a tumor microenvironment. Currently, oncolytic HSV (oHSV) is widely use in clinical; moreover, there’s hope that its curative effect will be further enhanced through the combination of oHSV with both traditional and emerging therapeutics.

Results

In this review, we provide a summary of the HSV host antiviral response evasion mechanism, HSV expresses immune evasion genes such as ICP34.5, ICP0, Us3, which are involved in inducing and activating host responses, so that the virus can evade the immune system and establish effective long-term latent infection; we outlined details of the oHSV strains generated by removing genes critical to viral replication such as ICP34.5, ICP0, and inserting therapeutic genes such as LacZ, granulocyte macrophage colony-stimulating factor (GM-CSF); security and limitation of some oHSV such G207, 1716, OncoVEX, NV1020, HF10, G47 in clinical application; and the achievements of oHSV combined with immunotherapy and chemotherapy.

Conclusion

We reviewed the immunotherapy mechanism of the oHSV and provided a series of cases. We also pointed out that an in-depth study of the application of oHSV in cancer treatment will potentially benefits cancer patients more.
Abkürzungen
APC
antigen presenting cell
DC
Dendritic cell
DNAM1
DNAX accessory molecule 1
EIF2a
eukaryotic Initiation Factor 2
GM-CSF
granulocyte macrophage colony-stimulating factor
IFN
interferon
oHSV
oncolytic herpes simplex virus
OPTN
optineurin
PKR
protein kinase R
RR
ribonucleotide reductase
TAP
transporter associated with antigen presentation
TK
thymidine kinase
TL
T lymphocyte
TNF-α
tumor necrosis factor
Vhs
virion host shutoff protein

Review

Introduction

For the past few years,despite constant new attempts finding phenomenal cancer treatments, chemotherapy, radiation and targeted drugs therapy are still the main therapeutic method in clinical practice. However, many problems remain in these methods, such as incompleteness, severe side effects, easy development of drug resistance, and lack of control in tumor recurrence and metastasis, etc., all of which lead to unsatisfactory result in treating tumor. The shortcomings of these major therapies call for new strategies in the field of cancer [1].
The Oncolytic virus is a subtype of a lytic virus that selectively replicates and kills cancer cells and spreads within the tumor without damaging normal tissue. The activities of oncolytic virus reflect the basic biological principles of the virus and the interaction of host-virus in the fight between pathogenesis and the immune system [2].
HSV, a member of the alpha-herpesviruses subfamily, shares many similarities with pseudorabies virus, varicella-zoster virus and infectious bovine rhinotracheitis virus [3]. The virus contains double stranded DNA genomes of at least 120 kb, encoding for 70 or more genes. At present, lysotype HSV is the first virus to be developed into a recombinant oncolytic viral therapeutic vector, and the first oncolytic virus to fight cancer. As a cytolytic virus HSV possesses the following advantages: (1) HSV replicate quickly in cells and has capability to infect multiple types of cancer cells; (2) HSV has a large genome, which can be easily modified and be inserted with multiple additional transgenes [4, 5]; (3) HSV can be prevented with antiviral drugs when the dose start to impose threat to the patients’ lives [68]; (4) Modifying the glycoprotein of HSV can improve the targeting of tumor cells [9].
As efficient OVs, HSV has some ability to escape the host’s immune response including: To complement and incapacitate immunoglobulins via viral glycoproteins; to inhibit the production of cytokine/chemokine from infected cells [10]; to block the antigen presenting cells’ (APCs) maturation [11]; to evade host immunological surveillance via negative-regulation of the expression of MHC class I [12] and to inhibit the apoptosis and cell death induced by cytotoxic T lymphocyte(TL) [13]. For deletion or mutation of those genes that were involved in HSV’s escape through its host’ immune defense will prohibit its replication in normal cells. Tumor microenvironment is often in an immunosuppressive state, which may allow the virus’ entry and replication, which in turns eventually leads to the dissolution and death of tumor cells (Fig. 1). oHSV can also reverse the immune suppression of tumor microenvironment, enhance tumor immunogenicity, promote the infiltration of inflammatory cells, and play an effective anti-tumor effect.
This review elaborates on how the HSV surmounts the anti-viral defense mechanism of the host; the oHSVs’ involvements in deletion or modification of viral gene and the clinical development of oHSV. A better understanding of the complex pattern of the interaction between HSV and host, and combination with current clinical oHSV is essential to the refinements the strategy of oHSV, thus to improve the therapeutic effects and to comprehend the oHSV immune imperfection.

The mechanism of HSV confronted the host immune response

The infection of HSV causes to a cascade reaction of host anti-viral immunity responses. As a successful pathogen, HSV expresses proteins which are involved in inducing and activating host responses, so that the virus can escape from the immune system and set up effective long-term latent infection. HSV has various mechanisms to escape the host reactions (Fig. 2):

Envelope glycoprotein

The envelope glycoproteins of HSV can escape humoral immunity mechanisms, such as Glycoprotein C binding and inactivating properdin and complement protein C3b, C5 to protect the virus from virus neutralization mediated by complement induced via natural IgM and antibody-independent complement neutralization [14, 15]. gE and gI encode Fc receptors, which can attach to IgG [16]. This binding inactivation of complement mediated via antibody and cytotoxicity of antibody dependence conduce pathogenicity [17]. gD inhibit the expression of CD112, which binds to the natural killer (NK) cells excitating receptors DNAX accessory molecule 1 (DNAM1), leading to a noneffective binding and lysis to HSV-infected or gD-transfected cells via NK [18]. HSV inhibits the expression of CD1d, surface molecules of APCs, and thus reduces NKT cells stimulate [19].

Immune evasion genes

A series of genes encoded via HSV escape the host antiviral immune monitoring mechanism [10, 20]. In cancer cells, some of these pathways are flawed [21]. Interferon (IFN) 1 signaling pathway, crucial for antiviral innate immunity [22], relates to genes that are involved in pathways such as the TLR signaling pathway: Us3 inhibit the signal transmission of TLR3 and TLR2 to TRAF6 [23], deubiquitinase for UL36 down-regulate the expression of TRAF3 [20], ICP0 inhibite the expression of P50 and P65, the subunits of MyD88 and NF-κB [24], UL42 and Us3 inhibit the process of nuclear translocation via NF-κB [25, 26], and ICP27 bind to IκBa inhibits NF-κB [27]. In the RLR signaling pathway: the Us11 can combine with RIG-I and MDA-5, thus inhibits the integration with MAVS [28, 29]. ICP34.5 inhibit the phosphorylation of IRF3 by binding TBK1 [30], US3 inhibit production of IFN-β via hyperphosphorylation of IFN regulatory factor 3 (IRF3) [31], ICP0 prevents IRF3 sustained activation, thus inhibiting translocation from the nucleus to cytoplasm [32]. Influence of the DNA sensor signaling pathway is also present: IFI16, involved in sensing pathogen DNA and initiating signaling pathways, inhibits via ICP0 in the nucleus [33]. In addition, IFN-stimulated genes, UL41 the virion host shutoff protein (Vhs), inhibits viperin, ZAP, and tetherin via degraded mRNA [20, 34], Us11 inhibits OAS [35] and protein kinase R (PKR) [36]. Eukaryotic Initiation Factor 2 (eIF2a) phosphorylated via PKR, shuts down the synthesis of protein. ICP27 blocks the phosphorylation and activation of STAT-1 [37]. Due to the defection of cancer cells in IFN signaling [38], deletion or mutation in these immune evasion genes such as ICP34.5, ICP0, Us3 in oHSV will avail ourselves of a cancer therapy [21].

Block dendritic cells function

Dendritic cells (DCs) are polymorphisms and heterogeneous antigen presenting cells, it has vital function for the recognition of pathogens at the site of infection and the initiation of protective HSV-specific T cells [39]. HSV has numerous mechanisms to inhibit DCs function [11]. ICP34.5 binds to TBK1 and IKKα/β, impeding the maturation of DCs and inhibits autophagy by interfering with antigen presentation [40, 41]. ICP0 induces CD83 degradation as a DCs maturation marker, leading to a decrease in T cell stimulation [42] ICP47 blocks transporter associated with antigen presentation (TAP), inhibits MHC I-peptide presentation, and thus leads to a mediated by MHC I to CD8+ T-cells to escapes immunological surveillance in host cells without antigen presentation. Pourchet A et al. showed that oHSV express UL49.5 from BHV-1 has a high efficacy treating cancer models, which is rely on CD8+ T cells [43]. gB binds to HLA-DM and HLA-DR, which yields a negtive-regulation of MHC II pathway in CD4+ T cells [44]. ICP34.5 and UL41 interfere with antigen presentation of CD4+ T cells via down-regulated MHC II accumulation on the surface of glioblastoma cells [45].
T cells are also influenced via HSV infection. Firstly Us3 inhibits LAT, which is the linker activating T cells, and in turns blocks TCR signaling [24]. Secondly oHSV infection has an impact on the pathway in T cells, such as inhibiting NF-κB, activating STAT3, JNK and MAPK p38 pathways, and suppressing the pro-inflammatory cytokines synthesis, such as IL-2, TNF-a and increasing IL-10 synthesis [46].

Inhibition of autophagy

Autophagy is an important cellular degradative pathway [47], which exerts on cellular pathogens like oHSV with a process similar to the MHC I and II presented in APCs [48]. ICP34.5 targets Beclin1 and interacts with PPP1CA, blocking the formation of autophagosome [4951]. HSV induced EIF2AK2 activation down-regulates the Beclin1-mediated autophagy [49, 51]. In additional, ICP34.5 directly inhibits TBK1, which can regulated the phosphorylation of autophagic receptors SQSTM1/p62 and optineurin (OPTN) to mediate substances recruitment into phagophores for degradation [52]. Us11 interacts with EIF2AK2, inhibiting the phosphorylation of EIF2S1, mediated via EIF2AK2, to block autophagy [53].

Inhibition of apoptosis

Apoptosis, the programmed cell death, can clear up the infected cells. HSV encoding anti-apoptotic virulence factors to suppress apoptosis then gives the virus enough time to replicate after infection [13, 54]. After HSV infection, some genes like ICP6, Us3, gD and Us5 (gJ) play role in the suppression of cells apoptosis. Us3 suppresses the expression of cytochrome c and the activation of caspase-3 [54], Us3 protein kinase activates the proapoptotic proteins Bad and Bid [55]. Us5 (gJ) antagonizes Fas/UV-induced apoptosis and weakens the granzyme B-mediated pathways of CTL-induced apoptosis [56]. Compared to gJ, Us6 (gD) blocks apoptosis at different stages of the viral life cycle. Necroptosis, another programmed cell death, which absence of caspases. ICP6 suppress apoptosis by blocking caspase 8 mediated via TNF-α and Fas ligand [57], also blocking necroptosis induced by TNF via inhibits the binding of RIP1 and RIP3 [58]. However, ICP6 had the reverse effect, in mice [13, 59].
The interactions of these host viruses are crucial to harmonize the OV activity, regulate the OVs anti-viral immune responses, and inducing anti-tumor immunity.

Genetically engineered oHSV

The key to eradicate tumors in this new therapy is to improve the precision when targeting oHSV for tumors, and enhance the suppression. Genetic engineering can affect many aspects of how viruses’ work. To enhance tumor selectivity by removing key genes in healthy cells that replicate with the virus. Table 1 summarizes the modified viral genes in oHSV and the functions of viral proteins encoded by these genes.
Table 1
Immune evasion genes of HSV
Gene
Protein
Function
oHSV name
UL27
gB
Part of initial attachment of the virus to the cell by binding to heparan sulfate. With gH/gL, enables fusion of the envelope with the cell membrane. Down-regulation of MHC II processing pathway in CD4+ cells.
R5141; KNE
UL44
gC
Forms the initial attachment of the virus to the cell by binding to heparan sulfate. Inactivates serum complement proteins.
R5141
US6
gD
Binds to HVeM and/or nectin-1, leading to a conformation change that initiates fusion. Down-regulates NK receptor ligand and NK-mediated lysis; inhibition of apoptosis.
R5141; R-LM249; HSV1716EGFR; KNE
RL1
ICP34.5
Major neurovirulence gene. Suppression of PKR/eIF-2a signaling pathway and IFN-induced anti-viral mechanisms; Inhibits DC maturation and antigen. Presentation; Blocks MHC class II accumulation on the cell surface; Binds to Beclin-1, inhibiting autophagy.
HSV1716; R3616; OncoVexGMCSF; G47; ΔG207; DM33;
RL2
ICP0
Blocks NF-κB-mediated transcription of immunomodulatory cytokines, and IRF3-induced and IRF7-induced anti-viral signaling pathways; inhibits IRF3 translocation to the nucleus; inhibits IFI16; degradation of mature DC marker (CD83). Involved in transcription of viral genes. Has ubiquitin ligase activity. Inhibits interferon response. Alters the cellular environment to promote viral replication.
R7020 (NV1020);
UL39
ICP6
Major subunit of ribonucleotide reductase. Blocks TNF-a-mediated and Fas ligand-mediated apoptosis through interacting with caspase 8 and necroptosis.
hrR3; G47Δ
UL54
ICP27
Inhibits cellular mRNA splicing. Recruits necessary proteins involved in viral transcription and translation. Activates cellular pathways to promote viral replication. Blocks NF-κB and IRF3 signaling pathways; blocks STAT1 activation and its translocation to the nucleus.
HF10
US12
ICP47
Down-regulates MHC class I by inhibiting TAP.
G47Δ; OncoVexGMCSF
US11
US11
Binds to and is phosphorylated by PKR, preventing cellular inhibition of protein synthesis and autophagy; blocks OAS.
G47Δ
US3
US3
Inhibits NF-κB activation and reduces cytokine expression, such as IL-8; inhibits induction of apoptosis; hyperphosphorylates IRF3 to block activation of RLR signaling pathway.
R7041
UL48
VP16
Initiates transcription of immediate early genes. Inhibits NF-κB activation and blocks IRF3 pathway and IFN-β production.
KM100
Dlsptk, the first type 1 herpes simplex virus mutants, includes a mutation within the UL23 gene that encodes the thymidine kinase (TK) gene. Dlsptk can inhibit the growth of glioma in nude rat brain [60, 61]. Nevertheless, high doses of Dlsptk can cause fatal encephalitis. For this reason, it is necessary to look for other engineered herpes mutants with low toxicity [62].
HrR3, the recombinant HSV-1, insert a LacZ in HSV-1 UL39 (encode ICP6), alternatively replicates in cancer cells, which has remarkable anticancer activity [63, 64].
HSV1716 was isolated from HSV-1 (17+) strain, deletes two copies of the main neurotoxic determinant generepeat RL1, which encodes neurotoxic determinant ICP34.5). PKR phosphorylates eIF2a, thus inhibits protein translation and induces cell apoptosis and kills the virus. ICP34.5 mediated dephosphorylation of eIF2a prevent cell apoptosis and protect the survival and reproduction of virus [65]. 1716 targets cancer cells that uncontrollable protein synthesis [66].
R3616, isolated from HSV-1 (F) strain, deletes the two replicas of ICP34.5 genes. R3616 can effectively induce host anti-tumor immune response by inducing a series of immune cells [67]. Kanzaki et al. showed that R3616 infects tumor antigen-specific lymphocytes; this not only effect on primary tumors, but also regulates multiple metastases [68].
NV1020 is an attenuated HSV that contains a diploid gene (RL1, RL2 and s1), with UL56 in the genome deleted [69]. Moreover, NV1020 attenuated via delete the TK gene and the UL24 genes promoter, and then inserts an exogenous copy of TK gene. These changes allow NV1020 highly attenuated and only proliferates in tumor cells.
G207, the first oHSV to be tested in clinical trials, deletes the ICP34.5 and inserted the LacZ gene, so the virus can selectively spread in tumor cells [70]. The deletion mutants ICP34.5 induced the down-regulate of late viral genes including US11 via PKR [9]. G207 can induce systemic anti-tumor immunity, which is related to the activation of cytotoxic T lymphocytes [8].
G47Δ derived from G207, contain two of the mutations in the RL1 and ICP47 genes, and insert the LacZ in ICP6 gene (coding ribonucleic acid reductase large subunit) area cause its inactivation. Inactivation of ICP6 then induces the oHSV’s only replicate in proliferating cells. Furthermore, the ICP47 mutation can effectively activate the host’s anti-tumor immune response via enhanced MHC-I expression [71]. Due to the three remoulds in the genome, G47Δ may be less toxic and more secure than G207 and T-Vec.
DM33 includes deletions of ICP34.5 and LAT gene. Unlike Dlsptk, DM33 was isolated from the McKrae strain, which promotes viral growth and kills cancer cells [72, 73].
HF10, remove the 3.9 kb connection point between the right end of UL and UL/IRL, which caused the loss expression of UL56, and reproduction of UL53 (gK), UL54 (ICP27) and UL55 [9]. HF10 enhances angiogenesis and induces acytotoxic T lymphocytes anti-tumor response [62].
OncovexGM-CSF, ICP34.5 and ICP47 genes in HSV-1 were strike out, and the integration of human GM-CSF was step in the ICP34.5 site. A series of cytokines such as IL12, GM-CSF, IFN-α and tumor necrosis factor (TNF-α) used with oHSV can modify and enhance the anti-tumor immunity. The GM-CSF shows the most effective results. TNF-α, IL-12 and IFN-α preclinical cancer studies have also show promising contributions [74, 75]. OncovexGM-CSF enhanced antigen-specific T cell response and decreased inhibitory CD4+ regulation of T cell expression, with a specific antitumor effects achieved in CD8+ T cells [76].

Clinical development and limitations of oHSV

Oncolytic viruses have assessment for treatment of a series of mlignation tumors. The first clinical trials of engineered virus was conducted in the 1990s [77]. Several different oHSV have been or will be tested worldwide for various cancers; some have been developed to phase II/III trials, such as G207, 1716, OncoVEX, NV1020, HF10, G47Δ (Table 2) [78].
Table 2
oHSVs of genetic engineering and its clinical application
oHSV name
Genetic modification
Descrption
Clinical application
Dlsptk
TK(UL23)
Internal deletion within UL23
Malignant human gliomas
hrR3
UL39
Insertion of LacZ (encodes β-galactosidase) in UL39
Pancreatic cancer; colon carcinoma; liver cancers
HSV1716
ICP34.5
Deletion in both copies of ICP34.5
Glioblastoma multiforme; anaplastic astrocytoma; oral squamous cell carcinoma
R3616
ICP34.5
Deletion of two copies of ICP34.5
Pancreatic cancer; colon carcinoma
G207
ICP34.5
Deletions of two copies of the ICP34.5; insertion of an Escherichia coli LacZ
Prostate adenocarcinoma; glioblastoma; hepatocellular carcinoma; colorectal cancer
R7020 (NV1020)
UL23, UL55, UL56, RL1, RL2, RS1
Deletion of UL23, as well as the region encoding UL55, UL56, and one copy of RL1, RL2, and RS1 (though not the RS1 promoter)
Pancreatic cancer; colon carcinoma; bladder cancer; pleural cancer
G47Δ
RL1, UL39, US11, US12
Deletion of the overlapping US11 promoter/US12 region, putting expression of the normally late US11 gene under the immediate early US12 promoter
Prostate adenocarcinoma; glioblastoma; rectal cancer; nasopharyngeal carcinoma; breast cancer
OncoVexGM-CSF
ICP34.5 and ICP47
Deletion of two copies of ICP34.5 gene and the viral ICP47 genes; insertion of GM-CSF
Breast cancer; head and neck cancer; gastrointestinal cancers; malignant melanoma
HF10
UL53, UL54, UL55, UL56
Spontaneous deletion of UL56 as well as duplication of UL53, UL54, and UL55
Breast cancer; malignant melanoma; pancreatic cancer
DM33
ICP34.5
Deletions of γ-34.5 and LAT gene
Human gliomas and glioma cell line
Initially, oHSVs lay emphasis on security vectors, which included the deletes ICP34.5 gene, such as HSV1716. HSV1716 was first demonstrated to be safe and toxic in patients with pleomorphic glioblastoma and intersex astrocytoma [79]. The results showed that HSV1716 had good tolerance and no adverse reactions occurred after high dose of 1 × 105 PFU treatment. HSV1716 has been used in the treatment of glioma and oral squamous cell carcinoma [8082].
Then strains with additional multiple deletions or mutations in case of the reversion of wild type virus, like the G207 [83], became the first oHSVs used in clinical trial. G207 has been used for recurrent malignant glioma in phase I studies, untoward effect have been moderated to slight fever and local erythema/inflammation reactions at the sites of injection [84].
G47Δ enhances the anti-tumor efficacy while retaining the safety characteristics of G207 [8587]. G47Δ showed efficacy in all solid tumor models tested in vivo, such as hepatocellular carcinoma [88], schwannoma [89], prostate cancer [87, 90, 91], nasopharyngeal carcinoma [71], glioma, thyroid carcinoma [92], colorectal cancer, breast cancer [93] and malignant peripheral nerve sheath tumor. G47Δ has the ability to killing cancer stem cells [94]. At present, G47 Δ is the only third generation oHSVs tested on humans [85].
NV1020 safety and efficacy have been demonstrated in some cancer diseases, such as colon carcinoma, pleural cancer, bladder cancer and pancreatic cancer [9598]. Kemeny N et.al investigated the safety and tolerance of NV1020 in liver metastasis of colorectal cancer in a phase I clinical trial [99]. NV1020 was also tested for liver metastasis from colorectal cancer in phase II trials. The results show that NV1020 is safe and effective in anti-tumor therapy [100]. In phase III trials, NV1020 was used in combination with cytotoxic and targeted drugs [62].
OncovexGM-CSF is the first type of oncolytic virus. After genetic engineering, oncogm-csf can selectively replicate in tumor cells, directly inject into the lesion, express GM-CSF, and enhance systemic anti-tumor immune response [68, 101]. GM-CSF insertion can promote complementary anti-tumor immune response by recruiting APCs [102]. After intratumoral injection of oncovexGM-CSF, the lesions of 8/50 patients with metastatic malignant melanoma disappeared completely [103]. The safety of an oncovexGM-CSF has been determined in phase I studies [104]. Direct injection of oncovexGM-CSF into melanoma lesions yielded an objective response rate of 28% in phase II clinical trials. Phase III clinical trials are ongoing [62].
However, oncolytic viruses all have the peculiarity of parental viruses and have some defects. Although HSV-1 is transmitted between cells and does not cause viremia, the most effective method for oncolytic HSV-1 is intracellular administration, which may not be suitable for intravenous infusion [105]. Because there are some drawbacks in intravenous administration, circulating antibodies may reduce the efficacy [106]. Viremia naturally causes viruses to be easily neutralized by antibodies; so the antineoplastic effect of intravenous administration of such viruses is limited in patients who have been treated or vaccinated. Clinical trials using oncolytic measles virus in the treatment of multiple myeloma fully demonstrated the adverse effects of circulating antibodies [107]. In dose-increasing studies, intravenous measles virus injection showed efficacy only when the dose reached a high dose of 1011 TCID50. In mice with transplanted tumors, intravenous injection of reovirus (REV) for 3 weeks after initial inhibition of tumor growth resulted in tumor regeneration, while the serum titer of anti- REVs antibodies increased [108]. Phase I study showed that 12 of 33 patients (36%) reached the maximum neutralizing REV antibody titer on the 7th day and 20 patients (61%) reached the maximum neutralizing REV antibody titer on the 14th day [109]. Hence, in the first week of treatment, speediness, repetitive, high-dose administration should be given before serum neutralizing antibodies rise, and should be combined with other anticancer therapies [106].

Increase the efficacy of oHSV deliver to tumor cells

At present, in the application of ohsv, there are some problems that limit its therapeutic effect, whether intratumoral or intravenous injection, there are some defects. Intratumoral injection can ensure that virus particles reach the lesion directly, but it is difficult to spread to the lesion area outside the injection area. Intravenous injection provides an opportunity for the virus to infect all cancer cells, and is particularly effective in the treatment of metastatic lesions [110]. Nevertheless, viral particles injected into veins are bound to suffer innate immune responses from the host [111], which may result in the virus particles being neutralized by antibodies before reaching the target cells.
The receptors that bind oHSV to cells are repositioned to ensure that the virus is more readily accessible to cancer cells. The co-injection of oHSV and collagenase can degrade the extracellular matrix of tumors by collagenase, and make the region outside the injection site of virus particles diffuse. The method for delivery of the oHSV can be enhanced or reduced by pretreatment with antiangiogenesis molecules. When oHSV was administered by direct injection, prior injection of cyclic RGD peptide, an antiangiogenic agent, reduced tumor vascular permeability and infiltration of leukocytes [112]. When oHSV is injected intravenously, the blood-brain barrier will increase the difficulty of injection. In order to solve this problem, it has been proved that destruction of the blood-brain barrier through hypertonic solution of mannitol can increase the number of viruses reaching tumors [110]. Ultrasound technique is also used to enhance the permeability of cell membrane and the efficacy of chemotherapeutic drugs anti-cancer [113]. Shintani et al. showed that effective use of ultrasound technology to help oHSV-1 enter squamous cell carcinomas [114]. Combination with key immunoregulatory inhibitors can improve the efficacy of oncolytic virus. For example, a study showed that intravenous injection of anti-PD-1 antibodies combined with Reolysin was significantly more effective in treating subcutaneous melanoma in mice than intravenous injection of Reolysin or anti-PD-1 alone [115]. Combination of anti-PD-1 antibody therapy can improve NK cells’ effective lysis of REV infected malignant cells by reducing the activity of regulatory T cells. Phase I study of combined therapy of oncolytic virus T-Vec and pembrolizumab (anti-PD-1) for head and neck cancer has been completed [105].
Besides, the oHSV combination with chemotherapy is also an effective strategy for tumor treatment. Toyoizumi et al. showed that combining the HSV1716 with chemotherapeutic drug MMC to treat the human non-small cell lung cancer yielded profound efficacy [116]. This study showed that chemotherapy and oHSV can work together treat cancer, and this synergistic effect will strengthen anti-tumor ability. Co-administrated with cyclophosphamide, the anticancer activity of HrR3 improved effectively [117]. Cyclophosphamide can enhance the replication of oncolytic virus by inhibiting the immune response of the system and has better anti-cancer effect [118]. However, in all oncolytic theatmets, the long range of side effects of inducing via body anti-tumor immunity, such as the emergence of AIDs, requires close study.

Conclusion

Oncolytic therapy, successes or failure hangs on the interaction of antiviral and antitumor immune responses between virus and host. HSV has been shown to be a site virus gets for oncolytic treatment because it is susceptible to genetic changes, deletions or mutations in genes with immunoregulatory function like ICP0, ICP 34.5, ICP 27, Us3 and UL39. This genetic alteration may result in an enhanced innate immune response, weakening viral replication and spreading in tumors.
At present, oHSV applied in clinical trials have not experienced serious adverse result and has achieved some effectiveness. For example, HSV1716 has been used for the treatment of oral squamous cell carcinoma and gloma [8587]; G47Δ showed efficacy in glioma, breast cancer [93], malignant peripheral nerve sheath tumor [92], schwannoma [89], nasopharyngeal carcinoma [71], hepatocellular carcinoma [88], prostate cancer [87, 90, 91], colorectal cancer and thyroid carcinoma; NV1020 can effectively control liver metastasis and prolong survival via re-sensitizing to chemotherapy [100].
Although the deleted or mutated genes confer safety and selectivity to oHSV in the treatments of tumor cells, efficacy has been attenuated. Direct injection of oHSVs is usually preferred during treatment, but this procedure limits the delivery to the sites where the tumor actually occurs. Physical factors like the extracellular matrix can limit the initial distribution and external diffusion of oHSV in the tumors [119]. The inborn and acquired anti-virus immunity can limit the replication and spread of oHSV [120]. In oncolytic virotherapy, these are only some examples of the many hurdles to be overcome. This made it necessary to combine oHSV with other therapies. The expectations are to develop a combination therapy regimen that produces synergic action against tumor cells without overlapping side effects. For examples, the combinations of oHSV with collagenase can degrade the extracellular matrix of tumors by collagenase, and make the region outside the injection site of virus particles diffuse; Injection of circulating RGD peptide before oHSV infection can reduce the permeability of blood vessels and infiltration of leukocytes in tumors [112]; and many published combination joint research tested the efficacy of oHSV combined with immunotherapies and chemotherapies in vitro. These identified combinations have achieved some good results.
With the development of preclinical research into clinical application, it is more likely to achieve greater success in understanding the combination of oHSV and other treatments. In a word, there are many areas to be researched in development of oHSV combined with other therapies. But hopefully all would join hand to cure cancer patients.

Acknowledgements

Not applicable.

Funding

This work was partially supported by National Key Research and Development Program of China (2018YFD0501600), Shandong province Key Research and Development program Fund (2018GNC113011), National Natural Science Fund of China (31872490, 31672556, 31502064), Taishan Scholar and Distinguished Experts (H. H.).

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analysed during the current study.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Sanchala DS, Bhatt LK, Prabhavalkar KS. Oncolytic Herpes Simplex viral therapy: a stride toward selective targeting of Cancer cells. Front Pharmacol. 2017;8:270.PubMedPubMedCentralCrossRef Sanchala DS, Bhatt LK, Prabhavalkar KS. Oncolytic Herpes Simplex viral therapy: a stride toward selective targeting of Cancer cells. Front Pharmacol. 2017;8:270.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Hamid O, Hoffner B, Gasal E, Hong J, Carvajal RD. Oncolytic immunotherapy: unlocking the potential of viruses to help target cancer. Cancer Immunol Immunother. 2017;66(10):1249–64.PubMedPubMedCentralCrossRef Hamid O, Hoffner B, Gasal E, Hong J, Carvajal RD. Oncolytic immunotherapy: unlocking the potential of viruses to help target cancer. Cancer Immunol Immunother. 2017;66(10):1249–64.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Hou P, Wang H, Zhao G, He C, He H. Rapid detection of infectious bovine Rhinotracheitis virus using recombinase polymerase amplification assays. BMC Vet Res. 2017;13(1):386.PubMedPubMedCentralCrossRef Hou P, Wang H, Zhao G, He C, He H. Rapid detection of infectious bovine Rhinotracheitis virus using recombinase polymerase amplification assays. BMC Vet Res. 2017;13(1):386.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Dai MH, Zamarin D, Gao SP, Chou TC, Gonzalez L, Lin SF, Fong Y. Synergistic action of oncolytic herpes simplex virus and radiotherapy in pancreatic cancer cell lines. Br J Surg. 2010;97(9):1385–94.PubMedCrossRef Dai MH, Zamarin D, Gao SP, Chou TC, Gonzalez L, Lin SF, Fong Y. Synergistic action of oncolytic herpes simplex virus and radiotherapy in pancreatic cancer cell lines. Br J Surg. 2010;97(9):1385–94.PubMedCrossRef
5.
Zurück zum Zitat Eisenberg DP, Adusumilli PS, Hendershott KJ, Yu Z, Mullerad M, Chan MK, Chou TC, Fong Y. 5-fluorouracil and gemcitabine potentiate the efficacy of oncolytic herpes viral gene therapy in the treatment of pancreatic cancer. J Gastrointest Surg. 2005;9(8):1068–77 discussion 1077-1069.PubMedPubMedCentralCrossRef Eisenberg DP, Adusumilli PS, Hendershott KJ, Yu Z, Mullerad M, Chan MK, Chou TC, Fong Y. 5-fluorouracil and gemcitabine potentiate the efficacy of oncolytic herpes viral gene therapy in the treatment of pancreatic cancer. J Gastrointest Surg. 2005;9(8):1068–77 discussion 1077-1069.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Hartkopf AD, Fehm T, Wallwiener D, Lauer U. Oncolytic virotherapy of gynecologic malignancies. Gynecol Oncol. 2011;120(2):302–10.PubMedCrossRef Hartkopf AD, Fehm T, Wallwiener D, Lauer U. Oncolytic virotherapy of gynecologic malignancies. Gynecol Oncol. 2011;120(2):302–10.PubMedCrossRef
7.
8.
Zurück zum Zitat Todo T. “Armed” oncolytic herpes simplex viruses for brain tumor therapy. Cell Adhes Migr. 2008;2(3):208–13.CrossRef Todo T. “Armed” oncolytic herpes simplex viruses for brain tumor therapy. Cell Adhes Migr. 2008;2(3):208–13.CrossRef
9.
Zurück zum Zitat Sokolowski NA, Rizos H, Diefenbach RJ. Oncolytic virotherapy using herpes simplex virus: how far have we come? Oncolytic Virother. 2015;4:207–19.PubMedPubMedCentral Sokolowski NA, Rizos H, Diefenbach RJ. Oncolytic virotherapy using herpes simplex virus: how far have we come? Oncolytic Virother. 2015;4:207–19.PubMedPubMedCentral
10.
Zurück zum Zitat Suazo PA, Ibanez FJ, Retamal-Diaz AR, Paz-Fiblas MV, Bueno SM, Kalergis AM, Gonzalez PA. Evasion of early antiviral responses by herpes simplex viruses. Mediat Inflamm. 2015;2015:593757.CrossRef Suazo PA, Ibanez FJ, Retamal-Diaz AR, Paz-Fiblas MV, Bueno SM, Kalergis AM, Gonzalez PA. Evasion of early antiviral responses by herpes simplex viruses. Mediat Inflamm. 2015;2015:593757.CrossRef
11.
12.
Zurück zum Zitat Rombout JH, Yang G, Kiron V. Adaptive immune responses at mucosal surfaces of teleost fish. Fish Shellfish Immunol. 2014;40(2):634–43.PubMedCrossRef Rombout JH, Yang G, Kiron V. Adaptive immune responses at mucosal surfaces of teleost fish. Fish Shellfish Immunol. 2014;40(2):634–43.PubMedCrossRef
13.
Zurück zum Zitat Yu X, He S. The interplay between human herpes simplex virus infection and the apoptosis and necroptosis cell death pathways. Virol J. 2016;13:77.PubMedPubMedCentralCrossRef Yu X, He S. The interplay between human herpes simplex virus infection and the apoptosis and necroptosis cell death pathways. Virol J. 2016;13:77.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Hook LM, Lubinski JM, Jiang M, Pangburn MK, Friedman HM. Herpes simplex virus type 1 and 2 glycoprotein C prevents complement-mediated neutralization induced by natural immunoglobulin M antibody. J Virol. 2006;80(8):4038–46.PubMedPubMedCentralCrossRef Hook LM, Lubinski JM, Jiang M, Pangburn MK, Friedman HM. Herpes simplex virus type 1 and 2 glycoprotein C prevents complement-mediated neutralization induced by natural immunoglobulin M antibody. J Virol. 2006;80(8):4038–46.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Wang X, Ju Z, Huang J, Hou M, Zhou L, Qi C, Zhang Y, Gao Q, Pan Q, Li G, Zhong J, Wang C. The relationship between the variants of the bovine MBL2 gene and milk production traits, mastitis, serum MBL-C levels and complement activity. Vet Immunol Immunopathol. 2012;148(3–4):311–9.PubMedCrossRef Wang X, Ju Z, Huang J, Hou M, Zhou L, Qi C, Zhang Y, Gao Q, Pan Q, Li G, Zhong J, Wang C. The relationship between the variants of the bovine MBL2 gene and milk production traits, mastitis, serum MBL-C levels and complement activity. Vet Immunol Immunopathol. 2012;148(3–4):311–9.PubMedCrossRef
16.
Zurück zum Zitat Dubin G, Frank I, Friedman HM. Herpes simplex virus type 1 encodes two fc receptors which have different binding characteristics for monomeric immunoglobulin G (IgG) and IgG complexes. J Virol. 1990;64(6):2725–31. Dubin G, Frank I, Friedman HM. Herpes simplex virus type 1 encodes two fc receptors which have different binding characteristics for monomeric immunoglobulin G (IgG) and IgG complexes. J Virol. 1990;64(6):2725–31.
17.
Zurück zum Zitat Lubinski JM, Lazear HM, Awasthi S, Wang F, Friedman HM. The herpes simplex virus 1 IgG fc receptor blocks antibody-mediated complement activation and antibody-dependent cellular cytotoxicity in vivo. J Virol. 2011;85(7):3239–49.PubMedPubMedCentralCrossRef Lubinski JM, Lazear HM, Awasthi S, Wang F, Friedman HM. The herpes simplex virus 1 IgG fc receptor blocks antibody-mediated complement activation and antibody-dependent cellular cytotoxicity in vivo. J Virol. 2011;85(7):3239–49.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Grauwet K, Cantoni C, Parodi M, De Maria A, Devriendt B, Pende D, Moretta L, Vitale M, Favoreel HW. Modulation of CD112 by the alphaherpesvirus gD protein suppresses DNAM-1-dependent NK cell-mediated lysis of infected cells. Proc Natl Acad Sci U S A. 2014;111(45):16118–23.PubMedPubMedCentralCrossRef Grauwet K, Cantoni C, Parodi M, De Maria A, Devriendt B, Pende D, Moretta L, Vitale M, Favoreel HW. Modulation of CD112 by the alphaherpesvirus gD protein suppresses DNAM-1-dependent NK cell-mediated lysis of infected cells. Proc Natl Acad Sci U S A. 2014;111(45):16118–23.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Yuan W, Dasgupta A, Cresswell P. Herpes simplex virus evades natural killer T cell recognition by suppressing CD1d recycling. Nat Immunol. 2006;7(8):835–42.PubMedCrossRef Yuan W, Dasgupta A, Cresswell P. Herpes simplex virus evades natural killer T cell recognition by suppressing CD1d recycling. Nat Immunol. 2006;7(8):835–42.PubMedCrossRef
21.
Zurück zum Zitat Peters C, Rabkin SD. Designing Herpes viruses as Oncolytics. Mol Ther Oncolytics. 2015;2:15010. Peters C, Rabkin SD. Designing Herpes viruses as Oncolytics. Mol Ther Oncolytics. 2015;2:15010.
22.
Zurück zum Zitat Shan S, Qi C, Zhu Y, Li H, An L, Yang G. Expression profile of carp IFN correlate with the up-regulation of interferon regulatory factor-1 (IRF-1) in vivo and in vitro: the pivotal molecules in antiviral defense. Fish Shellfish Immunol. 2016;52:94–102.PubMedCrossRef Shan S, Qi C, Zhu Y, Li H, An L, Yang G. Expression profile of carp IFN correlate with the up-regulation of interferon regulatory factor-1 (IRF-1) in vivo and in vitro: the pivotal molecules in antiviral defense. Fish Shellfish Immunol. 2016;52:94–102.PubMedCrossRef
23.
Zurück zum Zitat Yang Y, Wu S, Wang Y, Pan S, Lan B, Liu Y, Zhang L, Leng Q, Chen D, Zhang C, He B, Cao Y. The Us3 protein of Herpes Simplex virus 1 inhibits T cell signaling by confining linker for activation of T cells (LAT) activation via TRAF6 protein. J Biol Chem. 2015;290(25):15670–8.PubMedPubMedCentralCrossRef Yang Y, Wu S, Wang Y, Pan S, Lan B, Liu Y, Zhang L, Leng Q, Chen D, Zhang C, He B, Cao Y. The Us3 protein of Herpes Simplex virus 1 inhibits T cell signaling by confining linker for activation of T cells (LAT) activation via TRAF6 protein. J Biol Chem. 2015;290(25):15670–8.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat van Lint AL, Murawski MR, Goodbody RE, Severa M, Fitzgerald KA, Finberg RW, Knipe DM, Kurt-Jones EA. Herpes simplex virus immediate-early ICP0 protein inhibits toll-like receptor 2-dependent inflammatory responses and NF-kappaB signaling. J Virol. 2010;84(20):10802–11.PubMedPubMedCentralCrossRef van Lint AL, Murawski MR, Goodbody RE, Severa M, Fitzgerald KA, Finberg RW, Knipe DM, Kurt-Jones EA. Herpes simplex virus immediate-early ICP0 protein inhibits toll-like receptor 2-dependent inflammatory responses and NF-kappaB signaling. J Virol. 2010;84(20):10802–11.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Zhang J, Wang S, Wang K, Zheng C. Herpes simplex virus 1 DNA polymerase processivity factor UL42 inhibits TNF-alpha-induced NF-kappaB activation by interacting with p65/RelA and p50/NF-kappaB1. Med Microbiol Immunol. 2013;202(4):313–25.PubMedCrossRef Zhang J, Wang S, Wang K, Zheng C. Herpes simplex virus 1 DNA polymerase processivity factor UL42 inhibits TNF-alpha-induced NF-kappaB activation by interacting with p65/RelA and p50/NF-kappaB1. Med Microbiol Immunol. 2013;202(4):313–25.PubMedCrossRef
26.
Zurück zum Zitat Wang K, Ni L, Wang S, Zheng C. Herpes simplex virus 1 protein kinase US3 hyperphosphorylates p65/RelA and dampens NF-kappaB activation. J Virol. 2014;88(14):7941–51.PubMedPubMedCentralCrossRef Wang K, Ni L, Wang S, Zheng C. Herpes simplex virus 1 protein kinase US3 hyperphosphorylates p65/RelA and dampens NF-kappaB activation. J Virol. 2014;88(14):7941–51.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Kim JC, Lee SY, Kim SY, Kim JK, Kim HJ, Lee HM, Choi MS, Min JS, Kim MJ, Choi HS, Ahn JK. HSV-1 ICP27 suppresses NF-kappaB activity by stabilizing IkappaBalpha. FEBS Lett. 2008;582(16):2371–6.PubMedCrossRef Kim JC, Lee SY, Kim SY, Kim JK, Kim HJ, Lee HM, Choi MS, Min JS, Kim MJ, Choi HS, Ahn JK. HSV-1 ICP27 suppresses NF-kappaB activity by stabilizing IkappaBalpha. FEBS Lett. 2008;582(16):2371–6.PubMedCrossRef
28.
Zurück zum Zitat Xing J, Wang S, Lin R, Mossman KL, Zheng C. Herpes simplex virus 1 tegument protein US11 downmodulates the RLR signaling pathway via direct interaction with RIG-I and MDA-5. J Virol. 2012;86(7):3528–40.PubMedPubMedCentralCrossRef Xing J, Wang S, Lin R, Mossman KL, Zheng C. Herpes simplex virus 1 tegument protein US11 downmodulates the RLR signaling pathway via direct interaction with RIG-I and MDA-5. J Virol. 2012;86(7):3528–40.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Zhu YY, Xing WX, Shan SJ, Zhang SQ, Li YQ, Li T, An L, Yang GW. Characterization and immune response expression of the rig-I-like receptor mda5 in common carp Cyprinus carpio. J Fish Biol. 2016;88(6):2188–202.PubMedCrossRef Zhu YY, Xing WX, Shan SJ, Zhang SQ, Li YQ, Li T, An L, Yang GW. Characterization and immune response expression of the rig-I-like receptor mda5 in common carp Cyprinus carpio. J Fish Biol. 2016;88(6):2188–202.PubMedCrossRef
30.
Zurück zum Zitat Ma Y, Jin H, Valyi-Nagy T, Cao Y, Yan Z, He B. Inhibition of TANK binding kinase 1 by herpes simplex virus 1 facilitates productive infection. J Virol. 2012;86(4):2188–96.PubMedPubMedCentralCrossRef Ma Y, Jin H, Valyi-Nagy T, Cao Y, Yan Z, He B. Inhibition of TANK binding kinase 1 by herpes simplex virus 1 facilitates productive infection. J Virol. 2012;86(4):2188–96.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Wang S, Wang K, Lin R, Zheng C. Herpes simplex virus 1 serine/threonine kinase US3 hyperphosphorylates IRF3 and inhibits beta interferon production. J Virol. 2013;87(23):12814–27.PubMedPubMedCentralCrossRef Wang S, Wang K, Lin R, Zheng C. Herpes simplex virus 1 serine/threonine kinase US3 hyperphosphorylates IRF3 and inhibits beta interferon production. J Virol. 2013;87(23):12814–27.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Paladino P, Collins SE, Mossman KL. Cellular localization of the herpes simplex virus ICP0 protein dictates its ability to block IRF3-mediated innate immune responses. PLoS One. 2010;5(4):e10428.PubMedPubMedCentralCrossRef Paladino P, Collins SE, Mossman KL. Cellular localization of the herpes simplex virus ICP0 protein dictates its ability to block IRF3-mediated innate immune responses. PLoS One. 2010;5(4):e10428.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Everett RD. Dynamic response of IFI16 and Promyelocytic leukemia nuclear body components to Herpes Simplex virus 1 infection. J Virol. 2015;90(1):167–79.PubMedPubMedCentralCrossRef Everett RD. Dynamic response of IFI16 and Promyelocytic leukemia nuclear body components to Herpes Simplex virus 1 infection. J Virol. 2015;90(1):167–79.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Zenner HL, Mauricio R, Banting G, Crump CM. Herpes simplex virus 1 counteracts tetherin restriction via its virion host shutoff activity. J Virol. 2013;87(24):13115–23.PubMedPubMedCentralCrossRef Zenner HL, Mauricio R, Banting G, Crump CM. Herpes simplex virus 1 counteracts tetherin restriction via its virion host shutoff activity. J Virol. 2013;87(24):13115–23.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Sanchez R, Mohr I. Inhibition of cellular 2′-5′ oligoadenylate synthetase by the herpes simplex virus type 1 Us11 protein. J Virol. 2007;81(7):3455–64.PubMedPubMedCentralCrossRef Sanchez R, Mohr I. Inhibition of cellular 2′-5′ oligoadenylate synthetase by the herpes simplex virus type 1 Us11 protein. J Virol. 2007;81(7):3455–64.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Poppers J, Mulvey M, Khoo D, Mohr I. Inhibition of PKR activation by the proline-rich RNA binding domain of the herpes simplex virus type 1 Us11 protein. J Virol. 2000;74(23):11215–21.PubMedPubMedCentralCrossRef Poppers J, Mulvey M, Khoo D, Mohr I. Inhibition of PKR activation by the proline-rich RNA binding domain of the herpes simplex virus type 1 Us11 protein. J Virol. 2000;74(23):11215–21.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Johnson KE, Song B, Knipe DM. Role for herpes simplex virus 1 ICP27 in the inhibition of type I interferon signaling. Virology. 2008;374(2):487–94.PubMedCrossRef Johnson KE, Song B, Knipe DM. Role for herpes simplex virus 1 ICP27 in the inhibition of type I interferon signaling. Virology. 2008;374(2):487–94.PubMedCrossRef
39.
40.
Zurück zum Zitat Ma Y, He B. Recognition of herpes simplex viruses: toll-like receptors and beyond. J Mol Biol. 2014;426(6):1133–47.PubMedCrossRef Ma Y, He B. Recognition of herpes simplex viruses: toll-like receptors and beyond. J Mol Biol. 2014;426(6):1133–47.PubMedCrossRef
41.
Zurück zum Zitat Gobeil PA, Leib DA. Herpes simplex virus gamma34.5 interferes with autophagosome maturation and antigen presentation in dendritic cells. MBio. 2012;3(5):e00267–12.PubMedPubMedCentralCrossRef Gobeil PA, Leib DA. Herpes simplex virus gamma34.5 interferes with autophagosome maturation and antigen presentation in dendritic cells. MBio. 2012;3(5):e00267–12.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Heilingloh CS, Muhl-Zurbes P, Steinkasserer A, Kummer M. Herpes simplex virus type 1 ICP0 induces CD83 degradation in mature dendritic cells independent of its E3 ubiquitin ligase function. J Gen Virol. 2014;95(Pt 6):1366–75.PubMedCrossRef Heilingloh CS, Muhl-Zurbes P, Steinkasserer A, Kummer M. Herpes simplex virus type 1 ICP0 induces CD83 degradation in mature dendritic cells independent of its E3 ubiquitin ligase function. J Gen Virol. 2014;95(Pt 6):1366–75.PubMedCrossRef
43.
Zurück zum Zitat Pourchet A, Fuhrmann SR, Pilones KA, Demaria S, Frey AB, Mulvey M, Mohr I. CD8(+) T-cell immune evasion enables oncolytic virus immunotherapy. EBioMed. 2016;5:59–67.CrossRef Pourchet A, Fuhrmann SR, Pilones KA, Demaria S, Frey AB, Mulvey M, Mohr I. CD8(+) T-cell immune evasion enables oncolytic virus immunotherapy. EBioMed. 2016;5:59–67.CrossRef
44.
Zurück zum Zitat Neumann J, Eis-Hubinger AM, Koch N. Herpes simplex virus type 1 targets the MHC class II processing pathway for immune evasion. J Immunol. 2003;171(6):3075–83.PubMedCrossRef Neumann J, Eis-Hubinger AM, Koch N. Herpes simplex virus type 1 targets the MHC class II processing pathway for immune evasion. J Immunol. 2003;171(6):3075–83.PubMedCrossRef
45.
Zurück zum Zitat Trgovcich J, Johnson D, Roizman B. Cell surface major histocompatibility complex class II proteins are regulated by the products of the gamma(1)34.5 and U(L)41 genes of herpes simplex virus 1. J Virol. 2002;76(14):6974–86.PubMedPubMedCentralCrossRef Trgovcich J, Johnson D, Roizman B. Cell surface major histocompatibility complex class II proteins are regulated by the products of the gamma(1)34.5 and U(L)41 genes of herpes simplex virus 1. J Virol. 2002;76(14):6974–86.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Sloan DD, Jerome KR. Herpes simplex virus remodels T-cell receptor signaling, resulting in p38-dependent selective synthesis of interleukin-10. J Virol. 2007;81(22):12504–14.PubMedPubMedCentralCrossRef Sloan DD, Jerome KR. Herpes simplex virus remodels T-cell receptor signaling, resulting in p38-dependent selective synthesis of interleukin-10. J Virol. 2007;81(22):12504–14.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Xie W, Zhou J. Aberrant regulation of autophagy in mammalian diseases. Biol Lett. 2018;14(1):20170540. Xie W, Zhou J. Aberrant regulation of autophagy in mammalian diseases. Biol Lett. 2018;14(1):20170540.
49.
Zurück zum Zitat Orvedahl A, Alexander D, Talloczy Z, Sun Q, Wei Y, Zhang W, Burns D, Leib DA, Levine B. HSV-1 ICP34.5 confers neurovirulence by targeting the Beclin 1 autophagy protein. Cell Host Microbe. 2007;1(1):23–35.PubMedCrossRef Orvedahl A, Alexander D, Talloczy Z, Sun Q, Wei Y, Zhang W, Burns D, Leib DA, Levine B. HSV-1 ICP34.5 confers neurovirulence by targeting the Beclin 1 autophagy protein. Cell Host Microbe. 2007;1(1):23–35.PubMedCrossRef
51.
Zurück zum Zitat Kanai R, Zaupa C, Sgubin D, Antoszczyk SJ, Martuza RL, Wakimoto H, Rabkin SD. Effect of gamma34.5 deletions on oncolytic herpes simplex virus activity in brain tumors. J Virol. 2012;86(8):4420–31.PubMedPubMedCentralCrossRef Kanai R, Zaupa C, Sgubin D, Antoszczyk SJ, Martuza RL, Wakimoto H, Rabkin SD. Effect of gamma34.5 deletions on oncolytic herpes simplex virus activity in brain tumors. J Virol. 2012;86(8):4420–31.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Pilli M, Arko-Mensah J, Ponpuak M, Roberts E, Master S, Mandell MA, Dupont N, Ornatowski W, Jiang S, Bradfute SB, Bruun JA, Hansen TE, Johansen T, Deretic V. TBK-1 promotes autophagy-mediated antimicrobial defense by controlling autophagosome maturation. Immunity. 2012;37(2):223–34.PubMedPubMedCentralCrossRef Pilli M, Arko-Mensah J, Ponpuak M, Roberts E, Master S, Mandell MA, Dupont N, Ornatowski W, Jiang S, Bradfute SB, Bruun JA, Hansen TE, Johansen T, Deretic V. TBK-1 promotes autophagy-mediated antimicrobial defense by controlling autophagosome maturation. Immunity. 2012;37(2):223–34.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Lussignol M, Queval C, Bernet-Camard MF, Cotte-Laffitte J, Beau I, Codogno P, Esclatine A. The herpes simplex virus 1 Us11 protein inhibits autophagy through its interaction with the protein kinase PKR. J Virol. 2013;87(2):859–71.PubMedPubMedCentralCrossRef Lussignol M, Queval C, Bernet-Camard MF, Cotte-Laffitte J, Beau I, Codogno P, Esclatine A. The herpes simplex virus 1 Us11 protein inhibits autophagy through its interaction with the protein kinase PKR. J Virol. 2013;87(2):859–71.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat You Y, Cheng AC, Wang MS, Jia RY, Sun KF, Yang Q, Wu Y, Zhu D, Chen S, Liu MF, Zhao XX, Chen XY. The suppression of apoptosis by alpha-herpesvirus. Cell Death Dis. 2017;8(4):e2749.PubMedPubMedCentralCrossRef You Y, Cheng AC, Wang MS, Jia RY, Sun KF, Yang Q, Wu Y, Zhu D, Chen S, Liu MF, Zhao XX, Chen XY. The suppression of apoptosis by alpha-herpesvirus. Cell Death Dis. 2017;8(4):e2749.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Cartier A, Broberg E, Komai T, Henriksson M, Masucci MG. The herpes simplex virus-1 Us3 protein kinase blocks CD8T cell lysis by preventing the cleavage of bid by granzyme B. Cell Death Differ. 2003;10(12):1320–8.PubMedCrossRef Cartier A, Broberg E, Komai T, Henriksson M, Masucci MG. The herpes simplex virus-1 Us3 protein kinase blocks CD8T cell lysis by preventing the cleavage of bid by granzyme B. Cell Death Differ. 2003;10(12):1320–8.PubMedCrossRef
56.
Zurück zum Zitat Jerome KR, Chen Z, Lang R, Torres MR, Hofmeister J, Smith S, Fox R, Froelich CJ, Corey L. HSV and glycoprotein J inhibit caspase activation and apoptosis induced by granzyme B or Fas. J Immunol. 2001;167(7):3928–35.PubMedCrossRef Jerome KR, Chen Z, Lang R, Torres MR, Hofmeister J, Smith S, Fox R, Froelich CJ, Corey L. HSV and glycoprotein J inhibit caspase activation and apoptosis induced by granzyme B or Fas. J Immunol. 2001;167(7):3928–35.PubMedCrossRef
57.
Zurück zum Zitat Dufour F, Sasseville AM, Chabaud S, Massie B, Siegel RM, Langelier Y. The ribonucleotide reductase R1 subunits of herpes simplex virus types 1 and 2 protect cells against TNFalpha- and FasL-induced apoptosis by interacting with caspase-8. Apoptosis. 2011;16(3):256–71.PubMedCrossRef Dufour F, Sasseville AM, Chabaud S, Massie B, Siegel RM, Langelier Y. The ribonucleotide reductase R1 subunits of herpes simplex virus types 1 and 2 protect cells against TNFalpha- and FasL-induced apoptosis by interacting with caspase-8. Apoptosis. 2011;16(3):256–71.PubMedCrossRef
58.
Zurück zum Zitat Guo H, Omoto S, Harris PA, Finger JN, Bertin J, Gough PJ, Kaiser WJ, Mocarski ES. Herpes simplex virus suppresses necroptosis in human cells. Cell Host Microbe. 2015;17(2):243–51.PubMedPubMedCentralCrossRef Guo H, Omoto S, Harris PA, Finger JN, Bertin J, Gough PJ, Kaiser WJ, Mocarski ES. Herpes simplex virus suppresses necroptosis in human cells. Cell Host Microbe. 2015;17(2):243–51.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Wang X, Li Y, Liu S, Yu X, Li L, Shi C, He W, Li J, Xu L, Hu Z, Yu L, Yang Z, Chen Q, Ge L, Zhang Z, Zhou B, et al. Direct activation of RIP3/MLKL-dependent necrosis by herpes simplex virus 1 (HSV-1) protein ICP6 triggers host antiviral defense. Proc Natl Acad Sci U S A. 2014;111(43):15438–43.PubMedPubMedCentralCrossRef Wang X, Li Y, Liu S, Yu X, Li L, Shi C, He W, Li J, Xu L, Hu Z, Yu L, Yang Z, Chen Q, Ge L, Zhang Z, Zhou B, et al. Direct activation of RIP3/MLKL-dependent necrosis by herpes simplex virus 1 (HSV-1) protein ICP6 triggers host antiviral defense. Proc Natl Acad Sci U S A. 2014;111(43):15438–43.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Martuza RL, Malick A, Markert JM, Ruffner KL, Coen DM. Experimental therapy of human glioma by means of a genetically engineered virus mutant. Science. 1991;252(5007):854–6.PubMedCrossRef Martuza RL, Malick A, Markert JM, Ruffner KL, Coen DM. Experimental therapy of human glioma by means of a genetically engineered virus mutant. Science. 1991;252(5007):854–6.PubMedCrossRef
61.
Zurück zum Zitat Markert JM, Coen DM, Malick A, Mineta T, Martuza RL. Expanded spectrum of viral therapy in the treatment of nervous system tumors. J Neurosurg. 1992;77(4):590–4.PubMedCrossRef Markert JM, Coen DM, Malick A, Mineta T, Martuza RL. Expanded spectrum of viral therapy in the treatment of nervous system tumors. J Neurosurg. 1992;77(4):590–4.PubMedCrossRef
62.
Zurück zum Zitat Liu S, Dai M, You L, Zhao Y. Advance in herpes simplex viruses for cancer therapy. Sci China Life Sci. 2013;56(4):298–305.PubMedCrossRef Liu S, Dai M, You L, Zhao Y. Advance in herpes simplex viruses for cancer therapy. Sci China Life Sci. 2013;56(4):298–305.PubMedCrossRef
63.
Zurück zum Zitat D J. GOLDSTEIN. And WELLER. SK. Herpes Simplex virus type 1-induced ribonucleotide reductase activity is dispensable for virus growth and DNA synthesis: isolation and characterization of an ICP6 lacZ insertion mutant. J Virol. 1988;62(1):196–205. D J. GOLDSTEIN. And WELLER. SK. Herpes Simplex virus type 1-induced ribonucleotide reductase activity is dispensable for virus growth and DNA synthesis: isolation and characterization of an ICP6 lacZ insertion mutant. J Virol. 1988;62(1):196–205.
64.
Zurück zum Zitat Preston GV, Palfreyman WJ, Dutia MB. Identification of a Herpes Simplex virus type 1 polypeptide which is a component of the virus-induced ribonucleotide reductase. J Gen Virol. 1984;65(9):1457 1466.CrossRef Preston GV, Palfreyman WJ, Dutia MB. Identification of a Herpes Simplex virus type 1 polypeptide which is a component of the virus-induced ribonucleotide reductase. J Gen Virol. 1984;65(9):1457 1466.CrossRef
65.
Zurück zum Zitat He B, Gross M, Roizman B. The gamma(1)34.5 protein of herpes simplex virus 1 complexes with protein phosphatase 1alpha to dephosphorylate the alpha subunit of the eukaryotic translation initiation factor 2 and preclude the shutoff of protein synthesis by double-stranded RNA-activated protein kinase. Proc Natl Acad Sci U S A. 1997;94(3):843–8.PubMedPubMedCentralCrossRef He B, Gross M, Roizman B. The gamma(1)34.5 protein of herpes simplex virus 1 complexes with protein phosphatase 1alpha to dephosphorylate the alpha subunit of the eukaryotic translation initiation factor 2 and preclude the shutoff of protein synthesis by double-stranded RNA-activated protein kinase. Proc Natl Acad Sci U S A. 1997;94(3):843–8.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Chou J, Roizman B. The gamma 1(34.5) gene of herpes simplex virus 1 precludes neuroblastoma cells from triggering total shutoff of protein synthesis characteristic of programed cell death in neuronal cells. Proc Natl Acad Sci U S A. 1992;89(8):3266–70.PubMedPubMedCentralCrossRef Chou J, Roizman B. The gamma 1(34.5) gene of herpes simplex virus 1 precludes neuroblastoma cells from triggering total shutoff of protein synthesis characteristic of programed cell death in neuronal cells. Proc Natl Acad Sci U S A. 1992;89(8):3266–70.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Shirota T, Kasuya H, Kodera Y, Nishikawa Y, Shikano T, Sahin TT, Gewen T, Yamamura K, Fukuda S, Kanzaki A, Yamada S, Fujii T, Sugimoto H, Nomoto S, Takeda S, Nakao A. Oncolytic herpes virus induces effective anti-cancer immunity against murine colon cancer. Hepato-Gastroenterology. 2011;58(110–111):1482–9.PubMed Shirota T, Kasuya H, Kodera Y, Nishikawa Y, Shikano T, Sahin TT, Gewen T, Yamamura K, Fukuda S, Kanzaki A, Yamada S, Fujii T, Sugimoto H, Nomoto S, Takeda S, Nakao A. Oncolytic herpes virus induces effective anti-cancer immunity against murine colon cancer. Hepato-Gastroenterology. 2011;58(110–111):1482–9.PubMed
68.
Zurück zum Zitat Kanzaki A, Kasuya H, Yamamura K, Sahin TT, Nomura N, Shikano T, Shirota T, Tan G, Fukuda S, Misawa M, Nishikawa Y, Yamada S, Fujii T, Sugimoto H, Nomoto S, Takeda S, et al. Antitumor efficacy of oncolytic herpes simplex virus adsorbed onto antigen-specific lymphocytes. Cancer Gene Ther. 2012;19(4):292–8.PubMedCrossRef Kanzaki A, Kasuya H, Yamamura K, Sahin TT, Nomura N, Shikano T, Shirota T, Tan G, Fukuda S, Misawa M, Nishikawa Y, Yamada S, Fujii T, Sugimoto H, Nomoto S, Takeda S, et al. Antitumor efficacy of oncolytic herpes simplex virus adsorbed onto antigen-specific lymphocytes. Cancer Gene Ther. 2012;19(4):292–8.PubMedCrossRef
69.
Zurück zum Zitat Koshizuka T, Kawaguchi Y, Nishiyama Y. Herpes simplex virus type 2 membrane protein UL56 associates with the kinesin motor protein KIF1A. J Gen Virol. 2005;86(Pt 3):527–33.PubMedCrossRef Koshizuka T, Kawaguchi Y, Nishiyama Y. Herpes simplex virus type 2 membrane protein UL56 associates with the kinesin motor protein KIF1A. J Gen Virol. 2005;86(Pt 3):527–33.PubMedCrossRef
70.
Zurück zum Zitat Kramm CM, Chase M, Herrlinger U, Jacobs A, Pechan PA, Rainov NG, Sena-Esteves M, Aghi M, Barnett FH, Chiocca EA, Breakefield XO. Therapeutic efficiency and safety of a second-generation replication-conditional HSV1 vector for brain tumor gene therapy. Hum Gene Ther. 1997;8(17):2057–68.PubMedCrossRef Kramm CM, Chase M, Herrlinger U, Jacobs A, Pechan PA, Rainov NG, Sena-Esteves M, Aghi M, Barnett FH, Chiocca EA, Breakefield XO. Therapeutic efficiency and safety of a second-generation replication-conditional HSV1 vector for brain tumor gene therapy. Hum Gene Ther. 1997;8(17):2057–68.PubMedCrossRef
71.
Zurück zum Zitat Wang JN, Hu P, Zeng MS, Liu RB. Anti-tumor effect of oncolytic herpes simplex virus G47delta on human nasopharyngeal carcinoma. Chinese J Cancer. 2011;30(12):831–41.CrossRef Wang JN, Hu P, Zeng MS, Liu RB. Anti-tumor effect of oncolytic herpes simplex virus G47delta on human nasopharyngeal carcinoma. Chinese J Cancer. 2011;30(12):831–41.CrossRef
72.
Zurück zum Zitat Samoto K, Ehtesham M, Perng GC, Hashizume K, Wechsler SL, Nesburn AB, Black KL, Yu JS. A herpes simplex virus type 1 mutant with gamma 34.5 and LAT deletions effectively oncolyses human U87 glioblastomas in nude mice. Neurosurgery. 2002;50(3):599–605 discussion 605-596.PubMed Samoto K, Ehtesham M, Perng GC, Hashizume K, Wechsler SL, Nesburn AB, Black KL, Yu JS. A herpes simplex virus type 1 mutant with gamma 34.5 and LAT deletions effectively oncolyses human U87 glioblastomas in nude mice. Neurosurgery. 2002;50(3):599–605 discussion 605-596.PubMed
73.
Zurück zum Zitat Samoto K, Perng GC, Ehtesham M, Liu Y, Wechsler SL, Nesburn AB, Black KL, Yu JS. A herpes simplex virus type 1 mutant deleted for gamma34.5 and LAT kills glioma cells in vitro and is inhibited for in vivo reactivation. Cancer Gene Ther. 2001;8(4):269–77.PubMedCrossRef Samoto K, Perng GC, Ehtesham M, Liu Y, Wechsler SL, Nesburn AB, Black KL, Yu JS. A herpes simplex virus type 1 mutant deleted for gamma34.5 and LAT kills glioma cells in vitro and is inhibited for in vivo reactivation. Cancer Gene Ther. 2001;8(4):269–77.PubMedCrossRef
74.
Zurück zum Zitat Liu BL, Robinson M, Han ZQ, Branston RH, English C, Reay P, McGrath Y, Thomas SK, Thornton M, Bullock P, Love CA, Coffin RS. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther. 2003;10(4):292–303.PubMedCrossRef Liu BL, Robinson M, Han ZQ, Branston RH, English C, Reay P, McGrath Y, Thomas SK, Thornton M, Bullock P, Love CA, Coffin RS. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther. 2003;10(4):292–303.PubMedCrossRef
75.
Zurück zum Zitat Cui LL, Yang G, Pan J, Zhang C. Tumor necrosis factor alpha knockout increases fertility of mice. Theriogenology. 2011;75(5):867–76.PubMedCrossRef Cui LL, Yang G, Pan J, Zhang C. Tumor necrosis factor alpha knockout increases fertility of mice. Theriogenology. 2011;75(5):867–76.PubMedCrossRef
76.
Zurück zum Zitat Kaufman HL, Kim DW, DeRaffele G, Mitcham J, Coffin RS, Kim-Schulze S. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann Surg Oncol. 2010;17(3):718–30.PubMedCrossRef Kaufman HL, Kim DW, DeRaffele G, Mitcham J, Coffin RS, Kim-Schulze S. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann Surg Oncol. 2010;17(3):718–30.PubMedCrossRef
77.
Zurück zum Zitat Kirn D, Martuza RL, Zwiebel J. Replication-selective virotherapy for cancer: biological principles, risk management and future directions. Nat Med. 2001;7(7):781–7.PubMedCrossRef Kirn D, Martuza RL, Zwiebel J. Replication-selective virotherapy for cancer: biological principles, risk management and future directions. Nat Med. 2001;7(7):781–7.PubMedCrossRef
78.
Zurück zum Zitat Kanai R, Wakimoto H, Cheema T, Rabkin SD. Oncolytic herpes simplex virus vectors and chemotherapy: are combinatorial strategies more effective for cancer? Future Oncol. 2010;6(4):619–34.PubMedCrossRef Kanai R, Wakimoto H, Cheema T, Rabkin SD. Oncolytic herpes simplex virus vectors and chemotherapy: are combinatorial strategies more effective for cancer? Future Oncol. 2010;6(4):619–34.PubMedCrossRef
79.
Zurück zum Zitat Rampling R, Cruickshank G, Papanastassiou V, Nicoll J, Hadley D, Brennan D, Petty R, MacLean A, Harland J, McKie E, Mabbs R, Brown M. Toxicity evaluation of replication-competent herpes simplex virus (ICP 34.5 null mutant 1716) in patients with recurrent malignant glioma. Gene Ther. 2000;7(10):859–66.PubMedCrossRef Rampling R, Cruickshank G, Papanastassiou V, Nicoll J, Hadley D, Brennan D, Petty R, MacLean A, Harland J, McKie E, Mabbs R, Brown M. Toxicity evaluation of replication-competent herpes simplex virus (ICP 34.5 null mutant 1716) in patients with recurrent malignant glioma. Gene Ther. 2000;7(10):859–66.PubMedCrossRef
80.
Zurück zum Zitat Mace AT, Ganly I, Soutar DS, Brown SM. Potential for efficacy of the oncolytic Herpes simplex virus 1716 in patients with oral squamous cell carcinoma. Head Neck. 2008;30(8):1045–51.PubMedCrossRef Mace AT, Ganly I, Soutar DS, Brown SM. Potential for efficacy of the oncolytic Herpes simplex virus 1716 in patients with oral squamous cell carcinoma. Head Neck. 2008;30(8):1045–51.PubMedCrossRef
81.
Zurück zum Zitat Papanastassiou V, Rampling R, Fraser M, Petty R, Hadley D, Nicoll J, Harland J, Mabbs R, Brown M. The potential for efficacy of the modified (ICP 34.5(−)) herpes simplex virus HSV1716 following intratumoural injection into human malignant glioma: a proof of principle study. Gene Ther. 2002;9(6):398–406.PubMedCrossRef Papanastassiou V, Rampling R, Fraser M, Petty R, Hadley D, Nicoll J, Harland J, Mabbs R, Brown M. The potential for efficacy of the modified (ICP 34.5(−)) herpes simplex virus HSV1716 following intratumoural injection into human malignant glioma: a proof of principle study. Gene Ther. 2002;9(6):398–406.PubMedCrossRef
82.
Zurück zum Zitat Harrow S, Papanastassiou V, Harland J, Mabbs R, Petty R, Fraser M, Hadley D, Patterson J, Brown SM, Rampling R. HSV1716 injection into the brain adjacent to tumour following surgical resection of high-grade glioma: safety data and long-term survival. Gene Ther. 2004;11(22):1648–58.PubMedCrossRef Harrow S, Papanastassiou V, Harland J, Mabbs R, Petty R, Fraser M, Hadley D, Patterson J, Brown SM, Rampling R. HSV1716 injection into the brain adjacent to tumour following surgical resection of high-grade glioma: safety data and long-term survival. Gene Ther. 2004;11(22):1648–58.PubMedCrossRef
83.
Zurück zum Zitat Moore AE. Viruses with oncolytic properties and their adaptation to tumors. Ann N Y Acad Sci. 1952;54(6):945–52.PubMedCrossRef Moore AE. Viruses with oncolytic properties and their adaptation to tumors. Ann N Y Acad Sci. 1952;54(6):945–52.PubMedCrossRef
84.
Zurück zum Zitat Markert JM, Medlock MD, Rabkin SD, Gillespie GY, Todo T, Hunter WD, Palmer CA, Feigenbaum F, Tornatore C, Tufaro F, Martuza RL. Conditionally replicating herpes simplex virus mutant, G207 for the treatment of malignant glioma: results of a phase I trial. Gene Ther. 2000;7(10):867–74.PubMedCrossRef Markert JM, Medlock MD, Rabkin SD, Gillespie GY, Todo T, Hunter WD, Palmer CA, Feigenbaum F, Tornatore C, Tufaro F, Martuza RL. Conditionally replicating herpes simplex virus mutant, G207 for the treatment of malignant glioma: results of a phase I trial. Gene Ther. 2000;7(10):867–74.PubMedCrossRef
85.
Zurück zum Zitat Todo T, Martuza RL, Rabkin SD, Johnson PA. Oncolytic herpes simplex virus vector with enhanced MHC class I presentation and tumor cell killing. Proc Natl Acad Sci U S A. 2001;98(11):6396–401.PubMedPubMedCentralCrossRef Todo T, Martuza RL, Rabkin SD, Johnson PA. Oncolytic herpes simplex virus vector with enhanced MHC class I presentation and tumor cell killing. Proc Natl Acad Sci U S A. 2001;98(11):6396–401.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Passer BJ, Wu CL, Wu S, Rabkin SD, Martuza RL. Analysis of genetically engineered oncolytic herpes simplex viruses in human prostate cancer organotypic cultures. Gene Ther. 2009;16(12):1477–82.PubMedPubMedCentralCrossRef Passer BJ, Wu CL, Wu S, Rabkin SD, Martuza RL. Analysis of genetically engineered oncolytic herpes simplex viruses in human prostate cancer organotypic cultures. Gene Ther. 2009;16(12):1477–82.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat 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(21):7886–90.PubMedCrossRef 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(21):7886–90.PubMedCrossRef
88.
Zurück zum Zitat Wang J, Xu L, Zeng W, Hu P, Zeng M, Rabkin SD, Liu R. Treatment of human hepatocellular carcinoma by the oncolytic herpes simplex virus G47delta. Cancer Cell Int. 2014;14(1):83.PubMedPubMedCentralCrossRef Wang J, Xu L, Zeng W, Hu P, Zeng M, Rabkin SD, Liu R. Treatment of human hepatocellular carcinoma by the oncolytic herpes simplex virus G47delta. Cancer Cell Int. 2014;14(1):83.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Prabhakar S, Messerli SM, Stemmer-Rachamimov AO, Liu TC, Rabkin S, Martuza R, Breakefield XO. Treatment of implantable NF2 schwannoma tumor models with oncolytic herpes simplex virus G47Delta. Cancer Gene Ther. 2007;14(5):460–7.PubMedCrossRef Prabhakar S, Messerli SM, Stemmer-Rachamimov AO, Liu TC, Rabkin S, Martuza R, Breakefield XO. Treatment of implantable NF2 schwannoma tumor models with oncolytic herpes simplex virus G47Delta. Cancer Gene Ther. 2007;14(5):460–7.PubMedCrossRef
90.
Zurück zum Zitat Varghese S, Rabkin SD, Liu R, Nielsen PG, Ipe T, Martuza RL. Enhanced therapeutic efficacy of IL-12, but not GM-CSF, expressing oncolytic herpes simplex virus for transgenic mouse derived prostate cancers. Cancer Gene Ther. 2006;13(3):253–65.PubMedCrossRef Varghese S, Rabkin SD, Liu R, Nielsen PG, Ipe T, Martuza RL. Enhanced therapeutic efficacy of IL-12, but not GM-CSF, expressing oncolytic herpes simplex virus for transgenic mouse derived prostate cancers. Cancer Gene Ther. 2006;13(3):253–65.PubMedCrossRef
91.
Zurück zum Zitat Fukuhara H, Ino Y, Kuroda T, Martuza RL, Todo T. Triple gene-deleted oncolytic herpes simplex virus vector double-armed with interleukin 18 and soluble B7-1 constructed by bacterial artificial chromosome-mediated system. Cancer Res. 2005;65(23):10663–8.PubMedCrossRef Fukuhara H, Ino Y, Kuroda T, Martuza RL, Todo T. Triple gene-deleted oncolytic herpes simplex virus vector double-armed with interleukin 18 and soluble B7-1 constructed by bacterial artificial chromosome-mediated system. Cancer Res. 2005;65(23):10663–8.PubMedCrossRef
92.
Zurück zum Zitat Antoszczyk S, Spyra M, Mautner VF, Kurtz A, Stemmer-Rachamimov AO, Martuza RL, Rabkin SD. Treatment of orthotopic malignant peripheral nerve sheath tumors with oncolytic herpes simplex virus. Neuro-Oncology. 2014;16(8):1057–66.PubMedPubMedCentralCrossRef Antoszczyk S, Spyra M, Mautner VF, Kurtz A, Stemmer-Rachamimov AO, Martuza RL, Rabkin SD. Treatment of orthotopic malignant peripheral nerve sheath tumors with oncolytic herpes simplex virus. Neuro-Oncology. 2014;16(8):1057–66.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Liu R, Martuza RL, Rabkin SD. Intracarotid delivery of oncolytic HSV vector G47Delta to metastatic breast cancer in the brain. Gene Ther. 2005;12(8):647–54.PubMedCrossRef Liu R, Martuza RL, Rabkin SD. Intracarotid delivery of oncolytic HSV vector G47Delta to metastatic breast cancer in the brain. Gene Ther. 2005;12(8):647–54.PubMedCrossRef
94.
Zurück zum Zitat Cheema TA, Wakimoto H, Fecci PE, Ning J, Kuroda T, Jeyaretna DS, Martuza RL, Rabkin SD. Multifaceted oncolytic virus therapy for glioblastoma in an immunocompetent cancer stem cell model. Proc Natl Acad Sci U S A. 2013;110(29):12006–11.PubMedPubMedCentralCrossRef Cheema TA, Wakimoto H, Fecci PE, Ning J, Kuroda T, Jeyaretna DS, Martuza RL, Rabkin SD. Multifaceted oncolytic virus therapy for glioblastoma in an immunocompetent cancer stem cell model. Proc Natl Acad Sci U S A. 2013;110(29):12006–11.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat McAuliffe PF, Jarnagin WR, Johnson P, Delman KA, Federoff H, Fong Y. Effective treatment of pancreatic tumors with two multimutated herpes simplex oncolytic viruses. J Gastroint Surg. 2000;4(6):580–8.CrossRef McAuliffe PF, Jarnagin WR, Johnson P, Delman KA, Federoff H, Fong Y. Effective treatment of pancreatic tumors with two multimutated herpes simplex oncolytic viruses. J Gastroint Surg. 2000;4(6):580–8.CrossRef
96.
Zurück zum Zitat Gutermann A, Mayer E, von Dehn-Rothfelser K, Breidenstein C, Weber M, Muench M, Gungor D, Suehnel J, Moebius U, Lechmann M. Efficacy of oncolytic herpesvirus NV1020 can be enhanced by combination with chemotherapeutics in colon carcinoma cells. Hum Gene Ther. 2006;17(12):1241–53.PubMedCrossRef Gutermann A, Mayer E, von Dehn-Rothfelser K, Breidenstein C, Weber M, Muench M, Gungor D, Suehnel J, Moebius U, Lechmann M. Efficacy of oncolytic herpesvirus NV1020 can be enhanced by combination with chemotherapeutics in colon carcinoma cells. Hum Gene Ther. 2006;17(12):1241–53.PubMedCrossRef
97.
Zurück zum Zitat Cozzi PJ, Malhotra S, McAuliffe P, Kooby DA, Federoff HJ, Huryk B, Johnson P, Scardino PT, Heston WD, Fong Y. Intravesical oncolytic viral therapy using attenuated, replication-competent herpes simplex viruses G207 and Nv1020 is effective in the treatment of bladder cancer in an orthotopic syngeneic model. FASEB J. 2001;15(7):1306–8.PubMedCrossRef Cozzi PJ, Malhotra S, McAuliffe P, Kooby DA, Federoff HJ, Huryk B, Johnson P, Scardino PT, Heston WD, Fong Y. Intravesical oncolytic viral therapy using attenuated, replication-competent herpes simplex viruses G207 and Nv1020 is effective in the treatment of bladder cancer in an orthotopic syngeneic model. FASEB J. 2001;15(7):1306–8.PubMedCrossRef
98.
Zurück zum Zitat Ebright MI, Zager JS, Malhotra S, Delman KA, Weigel TL, Rusch VW, Fong Y. Replication-competent herpes virus NV1020 as direct treatment of pleural cancer in a rat model. J Thorac Cardiovasc Surg. 2002;124(1):123–9.PubMedCrossRef Ebright MI, Zager JS, Malhotra S, Delman KA, Weigel TL, Rusch VW, Fong Y. Replication-competent herpes virus NV1020 as direct treatment of pleural cancer in a rat model. J Thorac Cardiovasc Surg. 2002;124(1):123–9.PubMedCrossRef
99.
Zurück zum Zitat Kemeny N, Brown K, Covey A, Kim T, Bhargava A, Brody L, Guilfoyle B, Haag NP, Karrasch M, Glasschroeder B, Knoll A, Getrajdman G, Kowal KJ, Jarnagin WR, Fong Y. Phase I, open-label, dose-escalating study of a genetically engineered herpes simplex virus, NV1020, in subjects with metastatic colorectal carcinoma to the liver. Hum Gene Ther. 2006;17(12):1214–24.PubMedCrossRef Kemeny N, Brown K, Covey A, Kim T, Bhargava A, Brody L, Guilfoyle B, Haag NP, Karrasch M, Glasschroeder B, Knoll A, Getrajdman G, Kowal KJ, Jarnagin WR, Fong Y. Phase I, open-label, dose-escalating study of a genetically engineered herpes simplex virus, NV1020, in subjects with metastatic colorectal carcinoma to the liver. Hum Gene Ther. 2006;17(12):1214–24.PubMedCrossRef
100.
Zurück zum Zitat Geevarghese SK, Geller DA, de Haan HA, Horer M, Knoll AE, Mescheder A, Nemunaitis J, Reid TR, Sze DY, Tanabe KK, Tawfik H. Phase I/II study of oncolytic herpes simplex virus NV1020 in patients with extensively pretreated refractory colorectal cancer metastatic to the liver. Hum Gene Ther. 2010;21(9):1119–28.PubMedPubMedCentralCrossRef Geevarghese SK, Geller DA, de Haan HA, Horer M, Knoll AE, Mescheder A, Nemunaitis J, Reid TR, Sze DY, Tanabe KK, Tawfik H. Phase I/II study of oncolytic herpes simplex virus NV1020 in patients with extensively pretreated refractory colorectal cancer metastatic to the liver. Hum Gene Ther. 2010;21(9):1119–28.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat McGeoch DJ, Dalrymple MA, Davison AJ, Dolan A, Frame MC, McNab D, Perry LJ, Scott JE, Taylor P. The complete DNA sequence of the long unique region in the genome of herpes simplex virus type 1. J Gen Virol. 1988;69(Pt 7):1531–74.PubMedCrossRef McGeoch DJ, Dalrymple MA, Davison AJ, Dolan A, Frame MC, McNab D, Perry LJ, Scott JE, Taylor P. The complete DNA sequence of the long unique region in the genome of herpes simplex virus type 1. J Gen Virol. 1988;69(Pt 7):1531–74.PubMedCrossRef
102.
Zurück zum Zitat van de Laar L, Coffer PJ, Woltman AM. Regulation of dendritic cell development by GM-CSF: molecular control and implications for immune homeostasis and therapy. Blood. 2012;119(15):3383–93.PubMedCrossRef van de Laar L, Coffer PJ, Woltman AM. Regulation of dendritic cell development by GM-CSF: molecular control and implications for immune homeostasis and therapy. Blood. 2012;119(15):3383–93.PubMedCrossRef
103.
Zurück zum Zitat Senzer NN, Kaufman HL, Amatruda T, Nemunaitis M, Reid T, Daniels G, Gonzalez R, Glaspy J, Whitman E, Harrington K, Goldsweig H, Marshall T, Love C, Coffin R, Nemunaitis JJ. Phase II clinical trial of a granulocyte-macrophage colony-stimulating factor-encoding, second-generation oncolytic herpesvirus in patients with unresectable metastatic melanoma. J Clin Oncol. 2009;27(34):5763–71.PubMedCrossRef Senzer NN, Kaufman HL, Amatruda T, Nemunaitis M, Reid T, Daniels G, Gonzalez R, Glaspy J, Whitman E, Harrington K, Goldsweig H, Marshall T, Love C, Coffin R, Nemunaitis JJ. Phase II clinical trial of a granulocyte-macrophage colony-stimulating factor-encoding, second-generation oncolytic herpesvirus in patients with unresectable metastatic melanoma. J Clin Oncol. 2009;27(34):5763–71.PubMedCrossRef
104.
Zurück zum Zitat Hu JC, Coffin RS, Davis CJ, Graham NJ, Groves N, Guest PJ, Harrington KJ, James ND, Love CA, McNeish I, Medley LC, Michael A, Nutting CM, Pandha HS, Shorrock CA, Simpson J, et al. A phase I study of OncoVEXGM-CSF, a second-generation oncolytic herpes simplex virus expressing granulocyte macrophage colony-stimulating factor. Clin Cancer Res. 2006;12(22):6737–47.PubMedCrossRef Hu JC, Coffin RS, Davis CJ, Graham NJ, Groves N, Guest PJ, Harrington KJ, James ND, Love CA, McNeish I, Medley LC, Michael A, Nutting CM, Pandha HS, Shorrock CA, Simpson J, et al. A phase I study of OncoVEXGM-CSF, a second-generation oncolytic herpes simplex virus expressing granulocyte macrophage colony-stimulating factor. Clin Cancer Res. 2006;12(22):6737–47.PubMedCrossRef
106.
Zurück zum Zitat Gong J, Sachdev E, Mita AC, Mita MM. Clinical development of reovirus for cancer therapy: An oncolytic virus with immune-mediated antitumor activity. World J Methodol. 2016;6(1):25–42.PubMedPubMedCentralCrossRef Gong J, Sachdev E, Mita AC, Mita MM. Clinical development of reovirus for cancer therapy: An oncolytic virus with immune-mediated antitumor activity. World J Methodol. 2016;6(1):25–42.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Russell SJ, Federspiel MJ, Peng KW, Tong C, Dingli D, Morice WG, Lowe V, O'Connor MK, Kyle RA, Leung N, Buadi FK, Rajkumar SV, Gertz MA, Lacy MQ, Dispenzieri A. Remission of disseminated cancer after systemic oncolytic virotherapy. Mayo Clin Proc. 2014;89(7):926–33.PubMedCrossRef Russell SJ, Federspiel MJ, Peng KW, Tong C, Dingli D, Morice WG, Lowe V, O'Connor MK, Kyle RA, Leung N, Buadi FK, Rajkumar SV, Gertz MA, Lacy MQ, Dispenzieri A. Remission of disseminated cancer after systemic oncolytic virotherapy. Mayo Clin Proc. 2014;89(7):926–33.PubMedCrossRef
108.
Zurück zum Zitat Hirasawa K, Nishikawa SG, Norman KL, Coffey MC, Thompson BG, Yoon CS, Waisman DM, Lee PW. Systemic reovirus therapy of metastatic cancer in immune-competent mice. Cancer Res. 2003;63(2):348–53.PubMed Hirasawa K, Nishikawa SG, Norman KL, Coffey MC, Thompson BG, Yoon CS, Waisman DM, Lee PW. Systemic reovirus therapy of metastatic cancer in immune-competent mice. Cancer Res. 2003;63(2):348–53.PubMed
109.
Zurück zum Zitat White CL, Twigger KR, Vidal L, De Bono JS, Coffey M, Heinemann L, Morgan R, Merrick A, Errington F, Vile RG, Melcher AA, Pandha HS, Harrington KJ. Characterization of the adaptive and innate immune response to intravenous oncolytic reovirus (Dearing type 3) during a phase I clinical trial. Gene Ther. 2008;15(12):911–20.PubMedCrossRef White CL, Twigger KR, Vidal L, De Bono JS, Coffey M, Heinemann L, Morgan R, Merrick A, Errington F, Vile RG, Melcher AA, Pandha HS, Harrington KJ. Characterization of the adaptive and innate immune response to intravenous oncolytic reovirus (Dearing type 3) during a phase I clinical trial. Gene Ther. 2008;15(12):911–20.PubMedCrossRef
110.
Zurück zum Zitat Simpson GR, Han Z, Liu B, Wang Y, Campbell G, Coffin RS. Combination of a fusogenic glycoprotein, prodrug activation, and oncolytic herpes simplex virus for enhanced local tumor control. Cancer Res. 2006;66(9):4835–42.PubMedCrossRef Simpson GR, Han Z, Liu B, Wang Y, Campbell G, Coffin RS. Combination of a fusogenic glycoprotein, prodrug activation, and oncolytic herpes simplex virus for enhanced local tumor control. Cancer Res. 2006;66(9):4835–42.PubMedCrossRef
111.
Zurück zum Zitat Yang HT, Zou SS, Zhai LJ, Wang Y, Zhang FM, An LG, Yang GW. Pathogen invasion changes the intestinal microbiota composition and induces innate immune responses in the zebrafish intestine. Fish Shellfish Immunol. 2017;71:35–42.PubMedCrossRef Yang HT, Zou SS, Zhai LJ, Wang Y, Zhang FM, An LG, Yang GW. Pathogen invasion changes the intestinal microbiota composition and induces innate immune responses in the zebrafish intestine. Fish Shellfish Immunol. 2017;71:35–42.PubMedCrossRef
112.
Zurück zum Zitat Mullerad M, Bochner BH, Adusumilli PS, Bhargava A, Kikuchi E, Hui-Ni C, Kattan MW, Chou TC, Fong Y. Herpes simplex virus based gene therapy enhances the efficacy of mitomycin C for the treatment of human bladder transitional cell carcinoma. J Urol. 2005;174(2):741–6.PubMedPubMedCentralCrossRef Mullerad M, Bochner BH, Adusumilli PS, Bhargava A, Kikuchi E, Hui-Ni C, Kattan MW, Chou TC, Fong Y. Herpes simplex virus based gene therapy enhances the efficacy of mitomycin C for the treatment of human bladder transitional cell carcinoma. J Urol. 2005;174(2):741–6.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Fechheimer M, Boylan JF, Parker S, Sisken JE, Patel GL, Zimmer SG. Transfection of mammalian cells with plasmid DNA by scrape loading and sonication loading. Proc Natl Acad Sci U S A. 1987;84(23):8463–7.PubMedPubMedCentralCrossRef Fechheimer M, Boylan JF, Parker S, Sisken JE, Patel GL, Zimmer SG. Transfection of mammalian cells with plasmid DNA by scrape loading and sonication loading. Proc Natl Acad Sci U S A. 1987;84(23):8463–7.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Shintani M, Takahashi G, Hamada M, Okunaga S, Iwai S, Yura Y. Effect of ultrasound on herpes simplex virus infection in cell culture. Virol J. 2011;8:446.PubMedPubMedCentralCrossRef Shintani M, Takahashi G, Hamada M, Okunaga S, Iwai S, Yura Y. Effect of ultrasound on herpes simplex virus infection in cell culture. Virol J. 2011;8:446.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Rajani K, Parrish C, Kottke T, Thompson J, Zaidi S, Ilett L, Shim KG, Diaz RM, Pandha H, Harrington K, Coffey M, Melcher A, Vile R. Combination therapy with Reovirus and anti-PD-1 blockade controls tumor growth through innate and adaptive immune responses. Mol Ther. 2016;24(1):166–74.PubMedCrossRef Rajani K, Parrish C, Kottke T, Thompson J, Zaidi S, Ilett L, Shim KG, Diaz RM, Pandha H, Harrington K, Coffey M, Melcher A, Vile R. Combination therapy with Reovirus and anti-PD-1 blockade controls tumor growth through innate and adaptive immune responses. Mol Ther. 2016;24(1):166–74.PubMedCrossRef
116.
Zurück zum Zitat Toyoizumi T, Mick R, Abbas AE, Kang EH, Kaiser LR, Molnar-Kimber KL. Combined therapy with chemotherapeutic agents and herpes simplex virus type 1 ICP34.5 mutant (HSV-1716) in human non-small cell lung cancer. Hum Gene Ther. 1999;10(18):3013–29.PubMedCrossRef Toyoizumi T, Mick R, Abbas AE, Kang EH, Kaiser LR, Molnar-Kimber KL. Combined therapy with chemotherapeutic agents and herpes simplex virus type 1 ICP34.5 mutant (HSV-1716) in human non-small cell lung cancer. Hum Gene Ther. 1999;10(18):3013–29.PubMedCrossRef
117.
Zurück zum Zitat Wakimoto H, Fulci G, Tyminski E, Chiocca EA. Altered expression of antiviral cytokine mRNAs associated with cyclophosphamide's enhancement of viral oncolysis. Gene Ther. 2004;11(2):214–23.PubMedPubMedCentralCrossRef Wakimoto H, Fulci G, Tyminski E, Chiocca EA. Altered expression of antiviral cytokine mRNAs associated with cyclophosphamide's enhancement of viral oncolysis. Gene Ther. 2004;11(2):214–23.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Ikeda K, Ichikawa T, Wakimoto H, Silver JS, Deisboeck TS, Finkelstein D, GRt H, Louis DN, Bartus RT, Hochberg FH, Chiocca EA. Oncolytic virus therapy of multiple tumors in the brain requires suppression of innate and elicited antiviral responses. Nat Med. 1999;5(8):881–7.PubMedCrossRef Ikeda K, Ichikawa T, Wakimoto H, Silver JS, Deisboeck TS, Finkelstein D, GRt H, Louis DN, Bartus RT, Hochberg FH, Chiocca EA. Oncolytic virus therapy of multiple tumors in the brain requires suppression of innate and elicited antiviral responses. Nat Med. 1999;5(8):881–7.PubMedCrossRef
119.
Zurück zum Zitat Yun CO. Overcoming the extracellular matrix barrier to improve intratumoral spread and therapeutic potential of oncolytic virotherapy. Curr Opin Mol Ther. 2008;10(4):356–61.PubMed Yun CO. Overcoming the extracellular matrix barrier to improve intratumoral spread and therapeutic potential of oncolytic virotherapy. Curr Opin Mol Ther. 2008;10(4):356–61.PubMed
120.
Zurück zum Zitat Fukuhara H, Todo T. Oncolytic herpes simplex virus type 1 and host immune responses. Curr Cancer Drug Targets. 2007;7(2):149–55.PubMedCrossRef Fukuhara H, Todo T. Oncolytic herpes simplex virus type 1 and host immune responses. Curr Cancer Drug Targets. 2007;7(2):149–55.PubMedCrossRef
Metadaten
Titel
Oncolytic herpes simplex virus and immunotherapy
verfasst von
Wenqing Ma
Hongbin He
Hongmei Wang
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Immunology / Ausgabe 1/2018
Elektronische ISSN: 1471-2172
DOI
https://doi.org/10.1186/s12865-018-0281-9

Weitere Artikel der Ausgabe 1/2018

BMC Immunology 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.