Next Article in Journal
Association between Vitamin D Deficiency and Serologic Response to Hepatitis B Virus Vaccination among Heavy Industry Workers
Previous Article in Journal
Buccal Administration of a Zika Virus Vaccine Utilizing 3D-Printed Oral Dissolving Films in a Mouse Model
Previous Article in Special Issue
Developing Effective Cancer Vaccines Using Rendered-Inactive Tumor Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Natural Killer Cells in Oncolytic Virotherapy: Friends or Foes?

by
Michael L. Franks
1,2,†,
Ju-Hyun An
1,† and
Jianmei W. Leavenworth
1,3,4,*
1
Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
2
Graduate Biomedical Sciences Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA
3
The O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
4
Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Vaccines 2024, 12(7), 721; https://doi.org/10.3390/vaccines12070721
Submission received: 31 May 2024 / Revised: 26 June 2024 / Accepted: 26 June 2024 / Published: 28 June 2024
(This article belongs to the Special Issue Cancer Immunotherapy: Therapeutics and Mechanisms)

Abstract

:
Oncolytic virotherapy (OVT) has emerged as a promising cancer immunotherapy, and is capable of potentiating other immunotherapies due to its capacity to increase tumor immunogenicity and to boost host antitumor immunity. Natural killer (NK) cells are a critical cellular component for mediating the antitumor response, but hold a mixed reputation for their role in mediating the therapeutic efficacy of OVT. This review will discuss the pros and cons of how NK cells impact OVT, and how to harness this knowledge for the development of effective strategies that could modulate NK cells to improve OVT-based therapeutic outcomes.

1. Introduction

Immunotherapies, such as immune checkpoint blockade (ICB) and chimeric antigen receptor (CAR)-engineered cellular therapy, have shown promise in cancer patients. However, the immunosuppressive tumor microenvironment (TME) inherent in many types of cancer often represents a huge hurdle to favorable therapeutic outcomes [1]. Oncolytic virotherapy (OVT) has been proposed as a strategy to address this barrier. OVT utilizes viruses that preferentially infect and kill cancer cells while promoting a subsequent antitumor immune response. This process reshapes the TME and increases the immunogenicity of a tumor, converting immunosuppressive “cold” tumors into immunostimulatory “hot” tumors. The immune-associated mechanisms underlying this conversion have been extensively reviewed elsewhere [2,3,4]. Interestingly, studies of the impact of OVT on the host immune response have revealed that natural killer (NK) cells play a complicated role in OVT-based therapeutic outcomes. NK cells are integral to the innate immune response against both viral infection and tumors. Unfortunately, the antiviral response of NK cells has been shown to limit the spread of oncolytic viruses (OVs), often impeding their therapeutic efficacy. On the other hand, in some OVTs, NK cells have demonstrably contributed to their therapeutic efficacy, and several studies have successfully utilized the potent antitumor response of NK cells by combining OVT with other immunotherapies. This review will focus on the multifaceted aspects of NK cells in OVT, and the strategies that have successfully utilized NK cells to improve OVT efficacy against cancer.

2. Immune Response to Cancer

The immune surveillance mechanism against cancer is often explained by the cancer-immunity (CI) cycle [5,6,7]. The CI cycle was first proposed in the context of the T-cell-mediated antitumor response, illustrating the cyclic processes starting from the cancer’s antigen release to the acquisition of adaptive immunity against cancer [7]. The T-cell-focused CI cycle is initiated by antigen-presenting cells (APCs), such as dendritic cells (DCs), which uptake and present tumor antigens, thereby priming and activating T-cells. The activated T-cells then migrate, infiltrate the tumor and kill the cancer cells, consequently leading to the release of more tumor antigens. Since the T-cell CI cycle requires the presence of initial antitumor immune responses, its initiation is highly dependent on the immunological milieu of the tumor parenchyma, referred to as tumor immunotypes, including the immune desert, immune excluded, and immune inflamed [6]. Immune-desert tumors are defined as tumors devoid of tumor-infiltrating lymphocytes (TILs), with prevalent WNT/β-catenin signaling and include many neuroendocrine tumors [8,9,10]. Immune-excluded tumors are characterized by abundant immune-suppressive cell populations located within or proximal to the tumor site, including tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and regulatory T-cells (Tregs). These cells release immunosuppressive cytokines, such as interleukin (IL)-10, IL-35, and transforming growth factor (TGF)-β [11,12,13,14,15], consequentially leading to the exhaustion and poor tumor infiltration of the immune effector cells and failure of tumor clearance. In contrast, immune-inflamed tumors are characterized by a TME rich in interferon-gamma (IFN-γ), a high tumor mutational burden, a large population of TILs, and an elevated expression of major histocompatibility complex (MHC) class I/II in tumor cells [16,17,18,19,20]. Recent findings have revealed that NK cells play a pivotal role in shaping the immune-inflamed immunotype by not only directly killing and debulking the cancer mass, but also by secreting proinflammatory cytokines [5]. Most importantly, IFN-γ promotes the activation and maturation of APCs, invigorating the intervention of the adaptive immune system against cancers and amplifying the CI cycle [21,22,23,24].
The failure of immune surveillance that is involved in each step of the CI cycle often leads to cancer growth and progression, which proceeds through three sequential phases, including elimination, equilibrium, and escape, referred to as cancer immunoediting [25]. During elimination, the growing tumor stresses the tissues surrounding it, creating a proinflammatory environment, and prompting an innate immune response. NK cells are cytotoxic lymphocytes that are integral to the innate immune response to cancer. They detect and kill tumor cells as well as release IFN-γ, which promotes APCs to upregulate MHCI/II expression [22,26]. APCs migrate to the draining lymph node, where the selection and recruitment of adaptive immune cells, such as CD4+ or CD8+ T-cells, specific to tumor antigens can begin. During the elimination phase, cancer cells are effectively killed by NK and cytotoxic T-cells, and the cancer can be resolved at this point [25]. The equilibrium phase is entered into if the immune system is not able to eradicate the cancer entirely. During this phase, the growth of the cancer is controlled by the immune system, and the cancer cells undergo a natural selection process, acquiring resistance to antitumor immunity [25,27]. The escape phase occurs when the cancer cells have acquired enough resistance to grow and spread beyond the immune surveillance [25,27]. The methods by which cancer cells attain resistance to antitumor immunity are varied, including the release of suppressive cytokines to inhibit immune cell activity, the downregulation of antigen presentation, the upregulation of inhibitory ligands, and the attraction of immune-suppressive regulatory cells, which have been extensively described [28].

3. Cancer Immunotherapies

Cancer immunotherapies have been developed to modulate the immune response to cancer, facilitating re-entry into the elimination phase and amplifying the CI cycle [5,6,7]. ICB, cancer-targeting monoclonal antibodies, and CAR-engineered cellular therapy are the major classes of cancer immunotherapies. ICBs are antibodies that target either the inhibitory receptors on immune effector cells, or the inhibitory ligands expressed on tumor cells and, in some cases, immune cells. Inhibitory receptors, such as programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4), are often upregulated in activated lymphocytes to prevent hyperactivation, or are expressed on immunosuppressive cells, like Tregs. Many cancers have been reported to upregulate inhibitory ligands, such as programmed cell death ligand 1 (PD-L1) and PD-L2, facilitating their escape from immune surveillance [29]. ICBs competitively inhibit the binding of inhibitory receptors to their cognate ligands, enabling cytotoxic immune cells to attack cancer cells [9,10]. In addition to ICBs, monoclonal antibodies designed to specifically target cancer cells have also been shown to promote antitumor responses. Cancer-bound antibodies facilitate the antibody-dependent cellular cytotoxicity (ADCC) of macrophages and NK cells that express CD16, also known as FcγRIII [30,31,32]. CAR-T and CAR-NK cells are cytotoxic lymphocytes armed with CARs specific to the tumor antigens expressed by their target cancer cells. CARs ensure the engagement of cytotoxic lymphocytes against cancer cells, launching an antigen-specific response with an emphasis on cell-to-cell cytotoxic killing [33,34].
Immunotherapies have changed the paradigm for the treatment of cancer patients with advanced disease stages. However, their efficacy is highly dependent upon the immune phenotype of the tumor. Immunotherapies, such as ICBs, have been most successful in immune-inflamed tumors. In such tumors, ICBs block the inhibitory signals, enhancing the tumor-killing efficacy of tumor-infiltrating immune cells. In contrast, ICBs have been less effective on tumors with immune-excluded and immune-desert immunotypes, as in these tumors, the effector cells are already exhausted or can barely infiltrate the tumor site. To overcome this barrier, strategies that are designed to shift the immune phenotype of a cancer towards immune-inflamed status to improve the efficacy of cancer immunotherapies are being developed, and one such strategy is OVT.

4. Oncolytic Virotherapy

4.1. Introduction to Oncolytic Viruses

OVT utilizes viruses that preferentially infect and replicate in cancer cells compared to non-cancer cells [35]. The clearance of cancer cells during viral infections was noted as far back as the early 20th century. In 1912, tumor regression was noted in a cervical cancer patient after receiving the rabies vaccine, which contains an attenuated version of the rabies virus [36]. Tumor regression has also been seen in response to measles, polio, smallpox, adenovirus, vestibular stomatitis virus (VSV), and herpes simplex virus 1 (HSV-1), among others [37,38,39,40,41,42]. Cancer cells are favorable hosts for viral infection because their cellular machinery for DNA replication is highly upregulated to support rapid proliferation, which facilitates viral DNA replication upon infection. Additionally, many cancer cells exhibit impaired protein kinase R (PKR) pathway activity, which has antiviral defense mechanisms, by limiting protein synthesis and inducing apoptosis upon viral genome detection, making tumor cells more vulnerable to viral infection [43]. In addition, some cancers are known to have dysfunctional type 1 interferon pathways, increasing their susceptibility to viruses [44].
OVs kill cancer cells and promote antitumor immune responses. The replication of OVs in cancer cells induces the lysis of tumor cells, leading to the release of pathogen-associated molecular pattern molecules (PAMPs), damage-associated molecular pattern molecules (DAMPs), type 1 interferons, viral antigens, and tumor antigens into the TME. All these factors subsequently provoke a robust immune response [45,46,47]. OVT-mediated innate immune responses prompt the recruitment and maturation of DCs [48,49,50,51,52]. As professional APCs, DCs uptake, process, and present tumor antigens to T-cells, facilitating adaptive immunity against cancer, in a process called epitope spreading [47,53,54]. Interestingly, recent research has demonstrated that pre-existing immunity to viruses also contributes to developing antitumor adaptive immunity [55,56]. In the murine colon adenocarcinoma MC38 model, the bystander T-cells specific to the lymphocytic choriomeningitis virus (LCMV) within the TME exhibited the specific killing of infected tumor cells, substantially contributing to DC-mediated cancer epitope spreading and acquisition of antitumor immunity [56]. Similarly, another study also demonstrated that the therapeutic efficacy of oncolytic Newcastle disease virus (NDV) can be potentiated by pre-existing immunity to NDV [55]. Taken together, these studies suggest that antiviral immune responses to OVT potentially potentiate antitumor responses.

4.2. Oncolytic Viruses for Cancer Immunotherapy

Several different viruses have exhibited prominent anticancer efficacy, including the adenovirus [57], reovirus [58], VSV [41], vaccinia virus (VV) [59], and HSV [60]. The use of oncolytic adenoviruses (oAds) is one of the earliest forms of OVT to enter clinical trials and has undergone extensive research. oAds can infect a wide range of cancer cells and release highly immunogenic viral components [61]. Cancer cells killed by oAds exhibit features of necrosis and autophagy [62], and release potent immunostimulants, classified as DAMPs, such as ATP, calreticulin, and heat shock protein 70 [62,63,64]. Reovirus is a non-encapsulated double-stranded RNA virus, and its lytic cycle can be accelerated by Ras signaling activation. This property makes the reovirus ideal for targeting cancers harboring constitutively active Ras mutations [65]. Additionally, DCs exposed to reovirus have been shown to drive potent antitumor immune responses [66]. VSV is a non-segmented RNA virus in the family Rhabdoviridae. Due to the absence of an immunological memory against VSV in the majority of human population, oncolytic recombinant VSVs have exhibited high tolerability when administered [67]. Alongside this property, the high viral titer and tumor trophism enable the systemic administration of VSV, making it a promising therapeutic option [67,68,69]. Oncolytic VV is distinguished by its hypoxic TME tolerability and antigenic variety [70,71,72]. The properties of each OV have been extensively discussed elsewhere [2]. We will focus on oncolytic HSV (oHSV) in the following sections.

4.3. Genetic Engineering in OVT

OVs have been genetically engineered to improve their safety and efficacy. For example, HSV-1 has been genetically engineered to improve its tumor-killing efficacy and safety profile. HSV-1 is a double-stranded DNA virus, characterized by a large linear genome (~25 kb), which allows HSV-1 to accommodate relatively large genetic modifications, such as transgene insertions for improving its anticancer efficacy [73]. Arming oHSV-1 through the insertion of proinflammatory cytokines, including granulocyte–macrophage colony-stimulating factor (GM-CSF) [74], IL-12 [75], and IL-15 [76,77], has improved its antitumor efficacy, and lacZ insertion has allowed for tracking viral distribution and infection [78]. Likewise, OVs can be armed with inhibitors targeting immune checkpoint receptors, such as anti-PD-1 single-chain variable fragment (scFv) [79,80] and anti-CTLA-4 scFv [81], to reduce the immunosuppressive milieu of a tumor. In addition, to facilitate the engagement of the adaptive immune system, DNA viruses can be modified to express tumor-associated antigens (TAAs), such as melanoma-derived tumor antigen gp100 [82], carcinoembryonic antigen (CEA) [83], glycoprotein oncofetal tumor antigen (5T4) [84], and lung cancer antigen (EphA2) [85].
In terms of the safety features of OVs, the initial roadblock for OVT is the off-target effects that deteriorate normal tissues. An example of genetic engineering that has improved the safety of OVT is the deletion of thymidine kinase in oHSV. Thymidine kinase plays a pivotal role in de novo synthesis of nucleic acids for viral genome replication, making it indispensable for oHSV multiplication [86]. While oHSVs with thymidine kinase mutations cannot replicate in non-proliferative cells, which are deficient in DNA replication machinery, they are able to readily infect and multiply in proliferative cells by exploiting the thymidine kinase of host cells [73]. In the context of OVT against brain tumors, employing the thymidine kinase knockout strategy mitigates the risk of infecting healthy neural tissues that are typically quiescent, while enabling oHSVs to selectively target highly proliferative cancer cells [87]. The γ34.5 gene is another common target for deletion in oHSV. The γ34.5 gene is a key factor of HSV-1-induced neurovirulence and is known to inhibit the aforementioned PKR pathway, thereby facilitating viral replication in host cells. The deletion of this gene substantially reduces the risk of oHSV-mediated neurovirulence by preventing viral protein synthesis in healthy cells, while maintaining its competence in cells with impaired PKR function, such as cancer cells [88,89].
The above-described genetic modifications have given oHSV a superior safety profile, making it an ideal candidate for treating neuronal brain cancers. A genetically engineered oHSV, which incorporates the deletion of thymidine kinase and ectopic IL-12 expression, has exhibited beneficial therapeutic outcomes against a variety of brain malignancies in preclinical trials [90,91,92]. IL-12 promotes anticancer immune responses by stimulating immune effector cells to directly target cancer cells via cytotoxicity or by secreting inflammatory cytokines [93,94,95,96,97]. Furthermore, genetically engineered oHSVs have shown superior therapeutic outcomes in clinical trials, including talimogene laherparepvec (T-VEC) for melanoma, the first OVT approved by the FDA [40]. T-VEC is a genetically engineered oHSV harboring the deletion of both γ34.5 and Herpes_US12 genes. The latter is known to inhibit antigenic peptide transportation to MHCI in infected cells [89,98]. In addition, proinflammatory cytokine GM-CSF has been introduced to facilitate DC recruitment and T-cell priming [40,99,100]. In clinical trials of T-VEC against cutaneous head and neck melanoma, 16% of the T-VEC-treated patients achieved a durable response rate (DRR), while the control group treated with GM-CSF alone only had a 2% DRR in a landmark phase 3 clinical trial [101,102]. In Austria, Switzerland, and Germany, T-VEC therapy has reported a complete response rate of 43.2% in melanoma patients [103]. G47Δ, another genetically engineered oHSV, has also shown encouraging therapeutic outcomes in clinical trials. The deletion of the α47 gene in G47Δ is designed to increase MHCI expression in infected cells [104]. In a phase 2 clinical trial, G47Δ achieved a superior 1-year survival rate in patients with recurring glioblastoma multiforme (GBM) of 92.3%, which is a substantial improvement over the 14% seen in patients who received standard chemo–radio therapy [105,106]. This result led to the approval of G47Δ as a treatment for malignant glioma in Japan. While not yet approved, clinical trials for another engineered oHSV, named G207, have demonstrated promising therapeutic outcomes, with a median survival of 12.2 months versus 5.6 months for the current standard of care received by pediatric patients with high-grade glioma [60]. G207 has also been shown to improve immune infiltration into GBM tumors [107] and is able to further extend survival in adult patients with recurring GBM when combined with radiotherapy [108]. Lastly, CAN-3110, an engineered oHSV designed to express the γ34.5 gene under the tumor-specific promoter, has shown enhanced viral replication upon infection and the improved survival of high-grade glioma patients [109]. In summary, genetic engineering technologies in the past decades have significantly improved the safety and efficacy of oncolytic viruses, thereby leading to the development of promising OVs against cancer.

4.4. The Controversial Role of the Host Immune System in OVT

As described above, the host immune response to OVT is crucial to its therapeutic efficacy. The infection and subsequent lysis of tumor cells by oncolytic viruses lead to the release of proinflammatory molecule cascades into the TME, including type 1 interferons, DAMPs, PAMPs, IL-18, IL-1β, and also tumor antigens [53,110,111,112,113,114], generating robust antiviral and antitumor immune responses. In the context of OVT, antiviral responses have been believed to be detrimental, as they limit the spread of OVs. However, further studies have demonstrated that the antiviral response is necessary to prime both the innate and adaptive antitumor responses [47]. As part of the innate immune system, the antiviral responses mediated by NK cells, compared to other cytotoxic lymphocytes, occur in the early phase of OVT [115], and have been shown to impede OVT efficacy by killing infected cells at early timepoints, limiting the reservoirs of viral replication [115] (Figure 1A). In contrast, NK cells have been shown to contribute to the OV-mediated antitumor response in a variety of cancers [116,117,118], likely when the antiviral response is controlled (Figure 1B). Collectively, NK cells could possibly lead to controversial therapeutic outcomes, depending on the balance between their antiviral and antitumor properties (Figure 1). Understanding both antitumor and antiviral immune responses in the context of OVT will shed light on how to design OVTs that utilize the maximal potential of NK cells.

5. NK Cell Biology

5.1. NK Cell Antitumor Responses

NK cells play a pivotal role in immune surveillance and are the primary defense against abnormal cells in the body, including infected, stressed, and neoplastic cells. Unlike cytotoxic T-cells, NK cells are not antigen specific; instead, they detect abnormal cells via a set of inhibitory and activating receptors. The functional activity of NK cells is determined by the balance between the signals emanating from these receptors, which are clustered in the immunological synapses of the NK cell, and its target [119]. Under homeostatic conditions, NK cells maintain basal levels of activation through the presentation of natural cytotoxicity receptors (NCRs) and other activation receptors on their surface, including NKp46, NKp30, NKp44, NKG2D (an NKG2 family member of C-type lectin-like receptor), and toll-like receptors (TLRs). This baseline receptor repertoire enables them to promptly respond to abnormal cells [120,121,122,123,124]. NKG2D and NCRs are the receptors that play an important role in the NK cell-mediated antitumor response. These receptors have been shown to be crucial in preventing early tumor formation by recognizing stress-induced ligands that are expressed on different types of cancer cells [125,126,127], such as MHC class I-related polypeptide sequence A (MICA) [127,128,129], natural killer cell cytotoxicity receptor 3 ligand 1 (NCR3LG1), and BCL2-associated athanogene 6 (BAG6) [130,131]. Inhibitory receptors present on NK cells, including killer immunoglobulin-like receptors (KIR), bind to MHCI molecules, thereby sustaining immune tolerance for healthy cells and preventing an autoimmune response. Tumor cells often downregulate MHCI expression, making them vulnerable to NK cell-mediated cytotoxicity. Another major mechanism of cancer recognition by NK cells is the detection of heat shock proteins (HSPs). HSPs are highly conserved molecular chaperones that prevent protein misfolding and aggregation under stressful conditions, and help transport antigenic peptides to MHCI/II molecules [132,133,134]. For example, 70 kDa heat shock proteins (HSP70s), which are highly upregulated in cancer, support tumor growth and metastasis by disrupting various pathways, leading to reduced apoptosis and increased proliferation and angiogenesis [135,136]. Membrane-bound HSP70s on cancer cells are recognized by the C-type lectin receptor CD94 on NK cells, exerting their antitumor activities [137,138,139]. Additionally, HSP70s upregulate MICA expression, facilitating NK cell-mediated tumor clearance via NKG2D [140].
The killing capacity of NK cells against cancer cells within the TME is impeded by a variety of immune-suppressive mechanisms that drive the NK cell signaling balance towards inhibition (Figure 2A). NK cell gene expression is rapidly altered upon entering a tumor, resulting in impaired cytokine and chemokine production, as well as granzyme secretion. This alteration impedes the antitumor functionality of NK cells and their capacity to recruit and activate DCs, which is thought to be caused, in part, by immunosuppressive cytokines and hypoxic conditions within the TME [141]. The TAMs and MDSCs often present within the TME secrete immune-suppressive cytokines, such as IL-10 and TGF-β, which downregulate the expression of activation receptors and suppress the lytic granule release of NK cells [142]. Additionally, interactions between NK cells and extracellular matrix (ECM) components, such as collagen, fibronectin, laminin, and elastin, promote the differentiation of NK cells, downregulating NK cell cytotoxicity and upregulating cytokine production [143,144]. This functional transition impairs NK cell-mediated tumor clearance [143,144,145]. Fibronectin was recently identified as a ligand of leukocyte immunoglobulin-like receptor subfamily B member 4 (LILRB4), one of NK cells inhibitory receptors, and their interaction attenuates NK cell killing of Lewis lung carcinoma cells [145]. The ECM is also a structural barrier to NK cell infiltration [146]. Treating melanoma-bearing mice with losartan and dihydroxybenzoic acid (DHB), which impair collagen synthesis and assembly, restores NK cell antitumor functionality and overall mice survival [143]. One strategy designed to cope with the ECM barrier is the oVV-Hyal1, an oncolytic VV-expressing hyaluronidase, which substantially degrades ECM components, such as collagen I/III, elastin, fibronectin, and hyaluronic acid, improving lymphocyte infiltration and antitumor efficacy [147]. Also, indoleamine 2,3-dioxygenase (IDO) in the TME leads to the depletion of tryptophan and the accumulation of L-kynurenine, inducing NK cell exhaustion and dysfunction [148], accompanied by the downregulation of NKG2D and NKp30 [148]. Interestingly, NKG2D ligands often shed into the extracellular milieu of the TME, which can be facilitated by various proteases, including A disintegrin and metalloprotease (ADAM)-10, ADAM-17, endoplasmic reticulum protein 5 (Erp5), and matrix metallopeptidase 14 (MMP14) [149,150,151]. Interactions with the soluble forms of NKG2D ligands leads to the internalization of NKG2D, thereby impairing NK cell antitumor activity, which is another mechanism underlying NK cell paralysis in a tumor [152,153,154,155].

5.2. NK Cell Antiviral Responses

NK cells also play a critical role in controlling viral infections, as the loss of NK cell functionality results in severe and even fatal infection from relatively benign viruses, such as Epstein Barr virus (EBV), HSV, and varicella zoster virus (VZV). There is an increased incidence of human papilloma virus (HPV)- and EBV-induced cancers [156,157,158], and influenza infection has exhibited higher lethality in a mouse model bearing a loss of function of Ncr1, which encodes NKp46 [120]. Similar to the way that NK cells target tumor cells, the detection and killing of infected cells are governed by the balance between the activating and inhibitory signals received by NK cells. The strength of these signals is determined by the following mechanisms: (1) the interaction of PAMPs and DAMPs with TLRs [159,160], (2) proinflammatory cytokine stimulation [161] and, finally, (3) the interplay of inhibitory and activation receptors on NK cells with their cognate ligands [120,162,163]. While crucial for host defense against viral infections, NK cell antiviral responses can impede the spread and efficacy of OVs [115], which will be discussed in detail in the sections below.

5.3. NK Cell Memory Responses

NK cells were conventionally thought of as short lived and incapable of “memory” responses, which is typically associated with the adaptive immune system, but there is now considerable evidence supporting the existence of long-lived memory NK cells that develop in response to viral infection and cytokine stimulation [164,165,166,167], i.e., these NK cells exhibit quick and strong activation and cytotoxicity after a challenge from the same stimuli that they encountered before [164,167,168,169]. NK cell memory in response to viral infection is best understood using murine cytomegalovirus (MCMV) infection models. The long-lived NK cells, mainly a subset expressing the Ly49H receptor, exhibit rapid recall responses to an MCMV rechallenge. These NK cells undergo clonal expansion upon exposure to m157, a virally encoded protein, and are capable of controlling peripheral viremia [164]. Human NK cells can also develop memory. For example, the memory response against human cytomegalovirus (HCMV) is mediated by CD94/NKG2C+ NK cells and is dependent on CD2 co-stimulation. These NK cells are epigenetically distinct from naïve NK cells and quickly respond to an HCMV rechallenge, undergoing clonal expansion [170,171,172]. While the exact ligand promoting NK cell memory to HCMV infection remains unclear, various UL40-encoded peptides contribute to the recognition of HCMV-infected cells by NKG2C+ adaptive memory-like NK cells [173]. Antigen-specific NK cell responses to HIV and influenza have also been documented, with CD94/NKG2C+ NK cells recognizing peptides presented by HLA-E on infected cells [174].
The molecular mechanisms that regulate NK cell memory are not entirely unclear, and are, most likely, experimental-model dependent. However, recent studies have revealed some similarities between memory NK cells and memory T-cells. For instance, T-cell factor 1(Tcf-1) is a transcription factor which is associated with stemness and crucial for the development of T-cell memory, as well as progenitor exhausted T-cells [175,176,177,178,179,180,181]. Although its role in NK cells is known to limit granzyme B expression to prevent self-destruction [182], a recent study has shown that tissue-resident Tcf-1+ NK cells can promote enhanced recall responses against secondary infections [183]. Additionally, NK cells positive for Tcf-1 and Bach2, another transcription factor associated with stemness, exhibit memory-like responses in the context of zika virus and HIV infection [184,185]. These studies have demonstrated an expanded role of NK cells with stem-like features in response to infection, especially secondary infection.
In addition to memory response to infection, the simultaneous stimulation of NK cells with IL-12, IL-15, and IL-18 can lead to the generation of cytokine-induced memory-like (CIML) NK cells. CIML NK cells exhibit enhanced antitumor efficacy compared to naïve NK cells, and maintain improved functionality for up to four months post-stimulation [167,186]. CIML NK cells are highly effective as a cellular therapy for acute myeloid leukemia (AML), with promising results from multiple clinical trials. In a phase 1 clinical trial with the transfer of ex vivo IL-12/IL-15/IL-18-stimulated NK cells against pediatric AML patients, four out of eight patients were in complete remission within 28 days of treatment. Moreover, donor memory NK cells remained functional for 3 months post-implantation, contributing to durable remission in two patients [187]. Adoptive CIML NK cell-based therapies have been similarly effective in adult patients with relapsing/recurring AML, with four out of nine patients achieving complete remission [187,188,189,190]. Additionally, CIML NK cells have shown enhanced efficacy against solid tumors in both clinical and preclinical studies, indicating improved tumor control in patients with head and neck cancer, and ovarian cancer-bearing mice [191,192]. Intriguingly, tumor-induced memory-like NK cells (TIML) are formed during the tumor priming of NK cells. TIML NK cells display enhanced survival, more efficient immune synapse formation, and increased synthesis of perforin. TIML NK cells primed with leukemic cells of B cell precursor acute lymphoblastic leukemia and AML exhibited enhanced cytotoxicity against their respective cancers both in vitro and in vivo [193]. Collectively, NK cell memory holds the potential to enhance both antitumor and antiviral activity, while their role remains poorly elucidated in the context of cancer immunotherapy, particularly OVT.

6. NK Cell Biology in OVT

6.1. NK Cell Antitumor and Antiviral Responses in OVT

Several studies have demonstrated that OVs convert the immune-suppressive TME into an immune stimulatory condition, thereby improving the NK cell-mediated killing of tumor cells [116,117,118,194,195,196] (Table 1). Upon administration of OVs, NK cells effectively kill OV-infected cancer cells through their antiviral immune response. Additionally, OVT-mediated TME conversion reshapes the NK cell receptor profile, thereby increasing cytotoxicity against uninfected tumor cells [197] (Figure 2B). NK cells have also been shown to improve the efficacy of OVT through facilitating replication and localization of OVs. In the context of the oncolytic reovirus model, hyperactive NK cells in mice preconditioned with IL-2 and Treg-depleting antibody display increased extravasation, transmigration, and tumor infiltration, which led to increased reovirus localization [198]. Concordantly, oncolytic VSV replication and spreading in both in vitro and in vivo settings using B16 melanoma cells are facilitated by activated NK cells. Interestingly, NK cells are able to mediate the localization of systemically delivered oncolytic VSV to the sites of primary and metastasized tumors [199], as this is dampened by NK cell depletion [198,199]. The overall body of research on OVT suggests that NK cells have a substantial role in promoting OVT therapeutic efficacy.
Controversially, NK cells have also been reported to impede therapeutic outcomes of OVTs through their potent antiviral response [115,200] (Table 2). OVs upregulate the expression of ligands for NK cell activation receptors in infected cells, such as NKG2D ligands, MICA, and retinoic acid early transcript 1 alpha (RAE1α) [201,202], and many NCR ligands, including hemagglutinin and hemagglutinin-neuraminidase (HN) [203,204]. Additionally, OVs can lower MHCI expression in infected cells, increasing their vulnerability to NK cells [163]. NK cells have been reported to preferentially target virus-infected tumor cells over uninfected tumor cells through NKp30 and NKp46, causing early viral clearance, limiting the reservoirs of viral replication, and consequentially compromising therapeutic outcomes [115,205,206,207]. Notably, in experimental GBM mouse models, the antiviral responses of macrophage and microglia in the brain depend on NK cell activation, and NK cell depletion confers a significant survival advantage over a non-depleted group [115]. Similarly, NK cells dampen the efficacy of oncolytic VSV therapy against hepatocellular carcinoma [205,208]. The improved antitumoral efficacy of oncolytic VSV therapy is accomplished by equipping OVs with the equine herpes virus-1 glycoprotein G (gGEHV-1), which binds to a broad range of chemokines, thereby suppressing the chemotaxis of NK cells to the site of infection [205]. Furthermore, the beneficial effect of NK cell depletion on the antitumor efficacy of oHSV against GBM tumors has also been reported [200].
Although the premature clearance of OVs by NK cells possibly undermines the therapeutic outcomes of OVTs (Figure 1A), it is noteworthy that the antiviral responses of NK cells also serve a vital role in priming the adaptive immune system against tumors by promoting the recruitment and maturation of DCs [47,50,51,209,210,211,212]. The activated NK cells facilitate the recruitment of DCs to the TME through the release of chemokines, such as C-C motif chemokine ligand 5 (CCL5) and X-C motif chemokine ligand 1 (XCL1), which direct DC localization [212]. NK cells also release tumor necrosis factor alpha (TNF-α) and IFN-γ, promoting maturation of DCs and upregulating their TLR and MHCII expression [213,214,215,216], which consequently activate T-cells to initiate the adaptive antitumor response [217,218,219].
Based on the previous studies, mathematical models have been established to predict the therapeutic outcomes resulting from alterations to parameters, including initial OV infection, NK cell activation, and tumor burden [220,221]. These models extrapolate how NK cell activation affects the efficacy of OVT, either in a beneficial or detrimental way. High viral cytopathy and early antiviral NK cell responses are linked to the impediment of OVT, while the addition of combination therapies is linked to the enhancement of OVT efficacy. Although the role of NK cells in OVT is controversial and context-dependent, several studies have sought to enhance the potential of NK cells in OVT by either inhibiting antiviral NK cells responses or promoting antitumor responses.

6.2. Enhancing NK Cell Antitumor Responses

Although the role of NK cells in OVT has shown mixed outcomes, their potential to enhance the effectiveness of OVs has prompted the development of NK cell immunotherapies that can synergize with OVTs (Figure 3). The C-C chemokine receptor 5 (CCR5)–CCL5 chemotaxis pathway has been identified as a mechanism for NK cell infiltration into a tumor [222]. To capitalize on this interaction, Li et al. engineered NK cells to overexpress CCR5 and the oncolytic VV to express CCL5 [223]. These genetic modifications facilitate NK cell migration towards OV-infected tumor cells, but do not alter their tumor-killing efficacy their production of cytokines, such as IFN-γ and TNF-α. In in vivo settings, CCR5-engineered NK cells have reduced tumor size and prolonged mice survival in combination with CCL5-expressing OVs.
A combination of oHSV and epidermal growth factor receptor (EGFR)-specific CAR-NK92 cells has shown increased tumor-killing efficacy against breast cancer and brain metastatic cells, leading to better survival outcomes [224]. Similarly, an oHSV armed with the IL-15/IL-15Rα complex, which activates NK cells more efficiently than IL-15 alone [225,226,227,228], has been shown to increase the effectiveness of EGFR CAR-NK antitumor activity against GBM cell lines. The IL-15/IL-15Rα complex secreted from virus-infected tumor cells not only facilitates the recruitment of CAR-NK cells to the tumor site, but also promotes NK cell survival and priming [77,226,229,230]. In a study designed to test the effects of reovirus on DCs, NK cells co-cultured with DCs exposed to reovirus (reo-DCs) displayed enhanced tumor-killing efficacy compared to NK cells treated with IL-2 or NK cells co-cultured with untreated DCs. NK cells co-cultured with reo-DCs showed elevated IFN-γ secretion and, of note, reovirus exposure alone did not lead to the improved antitumor functionality of NK cells [66]. Treatment with bortezomib increased GBM sensitivity to adoptive NK cell therapy via upregulation of death receptor (DR)-5 [198]. This sensitivity was further enhanced by the addition of oHSV. The triple combination therapy of bortezomib, oHSV, and adoptive NK cell transfer has achieved superior responses against GBM in vitro and in vivo compared to bortezomib and NK cell adoptive transfer combination therapy [199]. Interestingly, the efficacy of oHSV and bortezomib treatment alone can be improved by endogenous NK depletion in GBM-bearing mice. This result indicates that the host NK cells may impede tumor clearance, and the adoptive transfer of NK cells is required for better therapeutic outcomes [200]. Therefore, a better understanding of the mechanisms of NK cell antitumor responses in the context of OVT has the potential to formulate combination therapies to improve the efficacy of OVT.

6.3. Inhibiting NK Cell Antiviral Responses

Several studies have aimed to prevent the premature clearance of viruses by NK cells while preserving their antitumor functionality (Figure 3). One such approach is to generate an oHSV expressing the cadherin 1 (CDH1) gene which encodes E-cadherin [231]. E-cadherin is a ligand of killer cell lectin-like receptor subfamily G, member 1 (KLRG1), expressed on terminally differentiated NK cells, and the binding of E-cadherin to KLRG1 impedes NK cell cytotoxicity [232,233]. In preclinical GBM models, CDH1-expressing OVs inhibit the cytotoxicity of KLRG1+ NK cells, which constitute more than 50% of the total NK cells within the TME. Compared to their parental oHSV, CDH1-expressing OVs have superior viral spread and are capable of improving overall survival in GBM-bearing mice [231]. Another approach has utilized “trojan horse” cell delivery strategies. These strategies utilize non-immunogenic cells as a reservoir of OV replication, functioning as a cellular therapy. For example, one study demonstrated that adipose-derived stem cells (ADSCs) inhibit NK cell-mediated antiviral responses, and yield superior viral spread when used as a delivery system for oncolytic VV [234]. However, for this strategy, MHC-matched donors are required to avoid graft rejection. A recent study demonstrated that it is possible to redirect the NK cell immune response towards the tumor. This is achieved by using an oncolytic measles virus that is genetically modified to express a bispecific killer engager (BiKE). BiKEs are capable of binding to two different targets. A BiKE targeting the carcinoembryonic antigen on the tumor cells and CD16 on the NK cells enables the formation of an immunological synapse between these cells, facilitating ADCC-mediated cancer cell clearance [235]. This approach increases NK cell reactivity against non-infected bystander tumor cells, preventing the early clearance of virus-infected cells.

7. Conclusions

In conclusion, the role of NK cells in OVT is pivotal. They have the potential to enhance OVT efficacy through their robust antitumor responses and have the capacity to prime the adaptive antitumor response. Conversely, they may impede OVT therapeutic outcomes by limiting viral replication and spread via their antiviral responses. The research addressing the optimal balance between these responses to maximize OVT efficacy is only just beginning, but the progress made thus far is promising. With ongoing research on new aspects of NK cell biology, such as NK cell memory, the synergy between OVT and NK cells will continue to improve.

Author Contributions

M.L.F., J-H.A. and J.W.L. drafted the manuscript, revised it critically, and approved this final version for publication. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the University of Alabama at Birmingham faculty start-up funds, DoD W81XWH-18-1-0315, and HT9425-23-1-0792 (J.W.L.). J.W.L was also supported by NIH R01AI148711, R01AI148711-03S1/04S1 (via NIA), R01CA276190, R21CA278853, the American Cancer Society RSG-23-1038722-01-IBCD, and the Breast Cancer Research Foundation of Alabama.

Acknowledgments

The figures were created by BioRender.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ADAMa disintegrin and metalloprotease
ADCCantibody-dependent cellular cytotoxicity
ADSCsadipose-derived stem cells
AMLacute myeloid leukemia
APCsantigen-presenting cells
BAG6BCL2-associated athanogene 6
BiKEbispecific killer engager
CARchimeric antigen receptor
CCL5C-C motif chemokine ligand 5
CCR5C-C chemokine receptor 5
CDH1cadherin 1
CI cyclecancer-immunity cycle
CIML NK cellscytokine-induced memory-like NK cells
CTLA-4cytotoxic T lymphocyte-associated protein 4
DAMPsdamage-associated molecular pattern molecules
DCsdendritic cells
DRRdurable response rate
EBVEpstein Barr virus
ECMextracellular matrix
EGFRepidermal growth factor receptor
Erp5endoplasmic reticulum protein 5
GBMglioblastoma multiforme
gGEHV-1equine herpes virus-1 glycoprotein G
GM-CSFgranulocyte–macrophage colony-stimulating factor
HCMVhuman cytomegalovirus
HSPheat shock protein
HNhemagglutinin–neuraminidase
HPVhuman papilloma virus
HSVherpes simplex virus
ICBimmune checkpoint blockade
IDOindoleamine 2:3-dioxygenase
IFN-γinterferon-gamma
ILinterleukin
KIRkiller immunoglobulin-like receptors
KLRG1killer cell lectin-like receptor subfamily G member 1
LILRB4leukocyte immunoglobulin-like receptor subfamily B member 4
LCMVlymphocytic choriomeningitis virus
MCMVmurine cytomegalovirus
MDSCsmyeloid-derived suppressor cells
MHCmajor histocompatibility complex
MICAMHC class I-related polypeptide sequence A
MMP14matrix metallopeptidase 14
NCRsnatural cytotoxicity receptors
NCR3LG1natural killer cell cytotoxicity receptor 3 ligand 1
NDVNewcastle disease virus
NK cellsnatural killer cells
oAdsoncolytic adenoviruses
oHSVoncolytic herpes simplex virus
OVsoncolytic viruses
OVToncolytic virotherapy
PAMPspathogen-associated molecular pattern molecules
PD-1programmed cell death protein 1
PD-L1programmed cell death ligand 1
PD-L2programmed cell death ligand 2
PKRprotein kinase R
RAE1αretinoic acid early transcript 1 alpha
scFvsingle-chain variable fragment
TAAstumor-associated antigens
TAMstumor-associated macrophages
Tcf-1T-cell factor 1
TGFtransforming growth factor
TILtumor-infiltrating lymphocytes
TLRstoll-like receptors
TMEtumor microenvironment
Tregsregulatory T-cells
TNF-αtumor necrosis factor alpha
T-VECtalimogene laherparepvec
VSVvestibular stomatitis virus
VVvaccinia virus
VZVvaricella zoster virus
XCL1X-C motif chemokine ligand 1

References

  1. Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef] [PubMed]
  2. Ma, R.; Li, Z.; Chiocca, E.A.; Caligiuri, M.A.; Yu, J. The emerging field of oncolytic virus-based cancer immunotherapy. Trends Cancer 2023, 9, 122–139. [Google Scholar] [CrossRef] [PubMed]
  3. Nguyen, H.M.; Saha, D. The Current State of Oncolytic Herpes Simplex Virus for Glioblastoma Treatment. Oncolytic Virother. 2021, 10, 1–27. [Google Scholar] [CrossRef] [PubMed]
  4. Yin, J.; Markert, J.M.; Leavenworth, J.W. Modulation of the Intratumoral Immune Landscape by Oncolytic Herpes Simplex Virus Virotherapy. Front. Oncol. 2017, 7, 136. [Google Scholar] [CrossRef] [PubMed]
  5. Huntington, N.D.; Cursons, J.; Rautela, J. The cancer–natural killer cell immunity cycle. Nat. Rev. Cancer 2020, 20, 437–454. [Google Scholar] [CrossRef] [PubMed]
  6. Mellman, I.; Chen, D.S.; Powles, T.; Turley, S.J. The cancer-immunity cycle: Indication, genotype, and immunotype. Immunity 2023, 56, 2188–2205. [Google Scholar] [CrossRef]
  7. Chen, D.S.; Mellman, I. Oncology Meets Immunology: The Cancer-Immunity Cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef] [PubMed]
  8. Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef] [PubMed]
  9. Carvajal-Hausdorf, D.; Altan, M.; Velcheti, V.; Gettinger, S.N.; Herbst, R.S.; Rimm, D.L.; Schalper, K.A. Expression and clinical significance of PD-L1, B7-H3, B7-H4 and TILs in human small cell lung Cancer (SCLC). J. Immunother. Cancer 2019, 7, 65. [Google Scholar] [CrossRef]
  10. Hansen, A.R.; Massard, C.; Ott, P.A.; Haas, N.B.; Lopez, J.S.; Ejadi, S.; Wallmark, J.M.; Keam, B.; Delord, J.P.; Aggarwal, R.; et al. Pembrolizumab for advanced prostate adenocarcinoma: Findings of the KEYNOTE-028 study. Ann. Oncol. 2018, 29, 1807–1813. [Google Scholar] [CrossRef]
  11. Kim, O.-H.; Kang, G.-H.; Noh, H.; Cha, J.-Y.; Lee, H.-J.; Yoon, J.-H.; Mamura, M.; Nam, J.-S.; Lee, D.H.; Kim, Y.A.; et al. Proangiogenic TIE2+/CD31+ macrophages are the predominant population of tumor-associated macrophages infiltrating metastatic lymph nodes. Mol. Cells 2013, 36, 432–438. [Google Scholar] [CrossRef]
  12. Jarnicki, A.G.; Lysaght, J.; Todryk, S.; Mills, K.H.G. Suppression of Antitumor Immunity by IL-10 and TGF-β-Producing T Cells Infiltrating the Growing Tumor: Influence of Tumor Environment on the Induction of CD4+ and CD8+ Regulatory T Cells1. J. Immunol. 2006, 177, 896–904. [Google Scholar] [CrossRef]
  13. Collison, L.W.; Workman, C.J.; Kuo, T.T.; Boyd, K.; Wang, Y.; Vignali, K.M.; Cross, R.; Sehy, D.; Blumberg, R.S.; Vignali, D.A. The inhibitory cytokine IL-35 contributes to regulatory T-cell function. Nature 2007, 450, 566–569. [Google Scholar] [CrossRef]
  14. Yaseen, M.M.; Abuharfeil, N.M.; Darmani, H.; Daoud, A. Mechanisms of immune suppression by myeloid-derived suppressor cells: The role of interleukin-10 as a key immunoregulatory cytokine. Open Biol. 2020, 10, 200111. [Google Scholar] [CrossRef]
  15. Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel Iii, E.E.; Koeppen, H.; Astarita, J.L.; Cubas, R. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
  16. Shen, J.; Choi, Y.L.; Lee, T.; Kim, H.; Chae, Y.K.; Dulken, B.W.; Bogdan, S.; Huang, M.; Fisher, G.A.; Park, S.; et al. Inflamed immune phenotype predicts favorable clinical outcomes of immune checkpoint inhibitor therapy across multiple cancer types. J. Immunother. Cancer 2024, 12, e008339. [Google Scholar] [CrossRef] [PubMed]
  17. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S. Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef]
  18. Paijens, S.T.; Vledder, A.; de Bruyn, M.; Nijman, H.W. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol. Immunol. 2021, 18, 842–859. [Google Scholar] [CrossRef]
  19. Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Kaufman, D.R.; Albright, A.; Cheng, J.D.; Kang, S.P.; Shankaran, V.; et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Investig. 2017, 127, 2930–2940. [Google Scholar] [CrossRef]
  20. Kawase, K.; Kawashima, S.; Nagasaki, J.; Inozume, T.; Tanji, E.; Kawazu, M.; Hanazawa, T.; Togashi, Y. High Expression of MHC Class I Overcomes Cancer Immunotherapy Resistance Due to IFNγ Signaling Pathway Defects. Cancer Immunol. Res. 2023, 11, 895–908. [Google Scholar] [CrossRef]
  21. Bian, Y.; Walter, D.L.; Zhang, C. Efficiency of Interferon-γ in Activating Dendritic Cells and Its Potential Synergy with Toll-like Receptor Agonists. Viruses 2023, 15, 1198. [Google Scholar] [CrossRef] [PubMed]
  22. Wijdeven, R.H.; van Luijn, M.M.; Wierenga-Wolf, A.F.; Akkermans, J.J.; van den Elsen, P.J.; Hintzen, R.Q.; Neefjes, J. Chemical and genetic control of IFNγ-induced MHCII expression. EMBO Rep. 2018, 19, e45553. [Google Scholar] [CrossRef] [PubMed]
  23. Fenton, S.E.; Saleiro, D.; Platanias, L.C. Type I and II Interferons in the Anti-Tumor Immune Response. Cancers 2021, 13, 1037. [Google Scholar] [CrossRef]
  24. Gao, Q.; Li, F.; Wang, S.; Shen, Z.; Cheng, S.; Ping, Y.; Qin, G.; Chen, X.; Yang, L.; Cao, L. A cycle involving HMGB1, IFN-γ and dendritic cells plays a putative role in anti-tumor immunity. Cell. Immunol. 2019, 343, 103850. [Google Scholar] [CrossRef]
  25. Lasek, W. Cancer immunoediting hypothesis: History, clinical implications and controversies. Cent. Eur. J. Immunol. 2022, 47, 168–174. [Google Scholar] [CrossRef]
  26. Zhou, F. Molecular mechanisms of IFN-gamma to up-regulate MHC class I antigen processing and presentation. Int. Rev. Immunol. 2009, 28, 239–260. [Google Scholar] [CrossRef]
  27. Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef]
  28. Zitvogel, L.; Tesniere, A.; Kroemer, G. Cancer despite immunosurveillance: Immunoselection and immunosubversion. Nat. Rev. Immunol. 2006, 6, 715–727. [Google Scholar] [CrossRef]
  29. Wu, Y.; Biswas, D.; Swanton, C. Impact of cancer evolution on immune surveillance and checkpoint inhibitor response. Semin. Cancer Biol. 2022, 84, 89–102. [Google Scholar] [CrossRef]
  30. Sulica, A.; Morel, P.; Metes, D.; Herberman, R.B. Ig-binding receptors on human NK cells as effector and regulatory surface molecules. Int. Rev. Immunol. 2001, 20, 371–414. [Google Scholar] [CrossRef]
  31. Steplewski, Z.; Lubeck, M.D.; Koprowski, H. Human macrophages armed with murine immunoglobulin G2a antibodies to tumors destroy human cancer cells. Science 1983, 221, 865–867. [Google Scholar] [CrossRef]
  32. Adams, G.P.; Weiner, L.M. Monoclonal antibody therapy of cancer. Nat. Biotechnol. 2005, 23, 1147–1157. [Google Scholar] [CrossRef] [PubMed]
  33. Gong, Y.; Klein Wolterink, R.G.J.; Wang, J.; Bos, G.M.J.; Germeraad, W.T.V. Chimeric antigen receptor natural killer (CAR-NK) cell design and engineering for cancer therapy. J. Hematol. Oncol. 2021, 14, 73. [Google Scholar] [CrossRef]
  34. Graham, C.; Hewitson, R.; Pagliuca, A.; Benjamin, R. Cancer immunotherapy with CAR-T cells—Behold the future. Clin. Med. 2018, 18, 324–328. [Google Scholar] [CrossRef]
  35. Lin, D.; Shen, Y.; Liang, T. Oncolytic virotherapy: Basic principles, recent advances and future directions. Signal Transduct. Target. Ther. 2023, 8, 156. [Google Scholar] [CrossRef] [PubMed]
  36. De Pace, N. Sulla scomparsa di un enorme cancro vegetante del callo dell’utero senza cura chirurgica. Ginecologia 1912, 9, 82–88. [Google Scholar]
  37. Russell, S.J.; Peng, K.W. Measles virus for cancer therapy. Curr. Top. Microbiol. Immunol. 2009, 330, 213–241. [Google Scholar] [CrossRef] [PubMed]
  38. Toyoda, H.; Yin, J.; Mueller, S.; Wimmer, E.; Cello, J. Oncolytic treatment and cure of neuroblastoma by a novel attenuated poliovirus in a novel poliovirus-susceptible animal model. Cancer Res. 2007, 67, 2857–2864. [Google Scholar] [CrossRef]
  39. Tsang, E.S.; Munster, P.N. Vaccinia (Smallpox) for the Treatment of Ovarian Cancer—Turning an Old Foe Into a Friend? JAMA Oncol. 2023, 9, 894–896. [Google Scholar] [CrossRef]
  40. Conry, R.M.; Westbrook, B.; McKee, S.; Norwood, T.G. Talimogene laherparepvec: First in class oncolytic virotherapy. Hum. Vaccin. Immunother. 2018, 14, 839–846. [Google Scholar] [CrossRef]
  41. Hastie, E.; Grdzelishvili, V.Z. Vesicular stomatitis virus as a flexible platform for oncolytic virotherapy against cancer. J. Gen. Virol. 2012, 93, 2529–2545. [Google Scholar] [CrossRef]
  42. Boozari, B.; Mundt, B.; Woller, N.; Strüver, N.; Gürlevik, E.; Schache, P.; Kloos, A.; Knocke, S.; Manns, M.P.; Wirth, T.C.; et al. Antitumoural immunity by virus-mediated immunogenic apoptosis inhibits metastatic growth of hepatocellular carcinoma. Gut 2010, 59, 1416–1426. [Google Scholar] [CrossRef]
  43. Cejalvo, J.M.; Falato, C.; Villanueva, L.; Tolosa, P.; González, X.; Pascal, M.; Canes, J.; Gavilá, J.; Manso, L.; Pascual, T.; et al. Oncolytic viruses: A new immunotherapeutic approach for breast cancer treatment? Cancer Treat. Rev. 2022, 106, 102392. [Google Scholar] [CrossRef]
  44. Matveeva, O.V.; Chumakov, P.M. Defects in interferon pathways as potential biomarkers of sensitivity to oncolytic viruses. Rev. Med. Virol. 2018, 28, e2008. [Google Scholar] [CrossRef]
  45. Stojdl, D.F.; Lichty, B.; Knowles, S.; Marius, R.; Atkins, H.; Sonenberg, N.; Bell, J.C. Exploiting tumor-specific defects in the interferon pathway with a previously unknown oncolytic virus. Nat. Med. 2000, 6, 821–825. [Google Scholar] [CrossRef]
  46. Krysko, D.V.; Garg, A.D.; Kaczmarek, A.; Krysko, O.; Agostinis, P.; Vandenabeele, P. Immunogenic cell death and DAMPs in cancer therapy. Nat. Rev. Cancer 2012, 12, 860–875. [Google Scholar] [CrossRef]
  47. Gujar, S.; Pol, J.G.; Kim, Y.; Lee, P.W.; Kroemer, G. Antitumor Benefits of Antiviral Immunity: An Underappreciated Aspect of Oncolytic Virotherapies. Trends Immunol. 2018, 39, 209–221. [Google Scholar] [CrossRef]
  48. Burke, S.; Shergold, A.; Elder, M.J.; Whitworth, J.; Cheng, X.; Jin, H.; Wilkinson, R.W.; Harper, J.; Carroll, D.K. Oncolytic Newcastle disease virus activation of the innate immune response and priming of antitumor adaptive responses in vitro. Cancer Immunol. Immunother. 2020, 69, 1015–1027. [Google Scholar] [CrossRef]
  49. Gujar, S.A.; Pan, D.A.; Marcato, P.; Garant, K.A.; Lee, P.W. Oncolytic virus-initiated protective immunity against prostate cancer. Mol. Ther. 2011, 19, 797–804. [Google Scholar] [CrossRef]
  50. Prestwich, R.J.; Errington, F.; Steele, L.P.; Ilett, E.J.; Morgan, R.S.; Harrington, K.J.; Pandha, H.S.; Selby, P.J.; Vile, R.G.; Melcher, A.A. Reciprocal human dendritic cell-natural killer cell interactions induce antitumor activity following tumor cell infection by oncolytic reovirus. J. Immunol. 2009, 183, 4312–4321. [Google Scholar] [CrossRef]
  51. van Vloten, J.P.; Matuszewska, K.; Minow, M.A.A.; Minott, J.A.; Santry, L.A.; Pereira, M.; Stegelmeier, A.A.; McAusland, T.M.; Klafuric, E.M.; Karimi, K.; et al. Oncolytic Orf virus licenses NK cells via cDC1 to activate innate and adaptive antitumor mechanisms and extends survival in a murine model of late-stage ovarian cancer. J. Immunother. Cancer 2022, 10, e004335. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, J.; Tai, L.H.; Ilkow, C.S.; Alkayyal, A.A.; Ananth, A.A.; de Souza, C.T.; Wang, J.; Sahi, S.; Ly, L.; Lefebvre, C.; et al. Maraba MG1 virus enhances natural killer cell function via conventional dendritic cells to reduce postoperative metastatic disease. Mol. Ther. 2014, 22, 1320–1332. [Google Scholar] [CrossRef]
  53. Kanerva, A.; Nokisalmi, P.; Diaconu, I.; Koski, A.; Cerullo, V.; Liikanen, I.; Tähtinen, S.; Oksanen, M.; Heiskanen, R.; Pesonen, S.; et al. Antiviral and antitumor T-cell immunity in patients treated with GM-CSF-coding oncolytic adenovirus. Clin. Cancer Res. 2013, 19, 2734–2744. [Google Scholar] [CrossRef]
  54. Lai, J.; Mardiana, S.; House, I.G.; Sek, K.; Henderson, M.A.; Giuffrida, L.; Chen, A.X.Y.; Todd, K.L.; Petley, E.V.; Chan, J.D.; et al. Adoptive cellular therapy with T cells expressing the dendritic cell growth factor Flt3L drives epitope spreading and antitumor immunity. Nat. Immunol. 2020, 21, 914–926. [Google Scholar] [CrossRef] [PubMed]
  55. Ricca, J.M.; Oseledchyk, A.; Walther, T.; Liu, C.; Mangarin, L.; Merghoub, T.; Wolchok, J.D.; Zamarin, D. Pre-existing Immunity to Oncolytic Virus Potentiates Its Immunotherapeutic Efficacy. Mol. Ther. 2018, 26, 1008–1019. [Google Scholar] [CrossRef] [PubMed]
  56. Chen, X.; Zhao, J.; Yue, S.; Li, Z.; Duan, X.; Lin, Y.; Yang, Y.; He, J.; Gao, L.; Pan, Z.; et al. An oncolytic virus delivering tumor-irrelevant bystander T cell epitopes induces anti-tumor immunity and potentiates cancer immunotherapy. Nat. Cancer 2024, 1–19. [Google Scholar] [CrossRef] [PubMed]
  57. Xie, D.; Tian, Y.; Hu, D.; Wang, Y.; Yang, Y.; Zhou, B.; Zhang, R.; Ren, Z.; Liu, M.; Xu, J.; et al. Oncolytic adenoviruses expressing checkpoint inhibitors for cancer therapy. Signal Transduct. Target. Ther. 2023, 8, 436. [Google Scholar] [CrossRef]
  58. Samson, A.; Bentham, M.J.; Scott, K.; Nuovo, G.; Bloy, A.; Appleton, E.; Adair, R.A.; 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, 562–573. [Google Scholar] [CrossRef] [PubMed]
  59. Guse, K.; Cerullo, V.; Hemminki, A. Oncolytic vaccinia virus for the treatment of cancer. Expert. Opin. Biol. Ther. 2011, 11, 595–608. [Google Scholar] [CrossRef]
  60. Friedman, G.K.; Johnston, J.M.; Bag, A.K.; Bernstock, J.D.; Li, R.; Aban, I.; Kachurak, K.; Nan, L.; Kang, K.-D.; Totsch, S. Oncolytic HSV-1 G207 immunovirotherapy for pediatric high-grade gliomas. N. Engl. J. Med. 2021, 384, 1613–1622. [Google Scholar] [CrossRef]
  61. Peter, M.; Kühnel, F. Oncolytic Adenovirus in Cancer Immunotherapy. Cancers 2020, 12, 3354. [Google Scholar] [CrossRef] [PubMed]
  62. Ito, H.; Aoki, H.; Kühnel, F.; Kondo, Y.; Kubicka, S.; Wirth, T.; Iwado, E.; Iwamaru, A.; Fujiwara, K.; Hess, K.R.; et al. Autophagic cell death of malignant glioma cells induced by a conditionally replicating adenovirus. J. Natl. Cancer Inst. 2006, 98, 625–636. [Google Scholar] [CrossRef] [PubMed]
  63. Liikanen, I.; Ahtiainen, L.; Hirvinen, M.L.; Bramante, S.; Cerullo, V.; Nokisalmi, P.; Hemminki, O.; Diaconu, I.; Pesonen, S.; Koski, A.; et al. Oncolytic adenovirus with temozolomide induces autophagy and antitumor immune responses in cancer patients. Mol. Ther. 2013, 21, 1212–1223. [Google Scholar] [CrossRef] [PubMed]
  64. Di Somma, S.; Iannuzzi, C.A.; Passaro, C.; Forte, I.M.; Iannone, R.; Gigantino, V.; Indovina, P.; Botti, G.; Giordano, A.; Formisano, P.; et al. The Oncolytic Virus dl922-947 Triggers Immunogenic Cell Death in Mesothelioma and Reduces Xenograft Growth. Front. Oncol. 2019, 9, 564. [Google Scholar] [CrossRef] [PubMed]
  65. Marcato, P.; Shmulevitz, M.; Pan, D.; Stoltz, D.; Lee, P.W. Ras transformation mediates reovirus oncolysis by enhancing virus uncoating, particle infectivity, and apoptosis-dependent release. Mol. Ther. 2007, 15, 1522–1530. [Google Scholar] [CrossRef] [PubMed]
  66. Errington, F.; Steele, L.; Prestwich, R.; Harrington, K.J.; Pandha, H.S.; Vidal, L.; de Bono, J.; Selby, P.; Coffey, M.; Vile, R.; et al. Reovirus activates human dendritic cells to promote innate antitumor immunity. J. Immunol. 2008, 180, 6018–6026. [Google Scholar] [CrossRef] [PubMed]
  67. Lyles, D.; Kuzmin, I.; Rupprecht, C. Rhabdoviridae. Fields Virology; Wolters Kluwer: Philadelphia, PA, USA, 2013; pp. 885–2456. [Google Scholar]
  68. Naik, S.; Nace, R.; Federspiel, M.J.; Barber, G.N.; Peng, K.W.; Russell, S.J. Curative one-shot systemic virotherapy in murine myeloma. Leukemia 2012, 26, 1870–1878. [Google Scholar] [CrossRef] [PubMed]
  69. Nakashima, H.; Kaur, B.; Chiocca, E.A. Directing systemic oncolytic viral delivery to tumors via carrier cells. Cytokine Growth Factor. Rev. 2010, 21, 119–126. [Google Scholar] [CrossRef] [PubMed]
  70. Pipiya, T.; Sauthoff, H.; Huang, Y.Q.; Chang, B.; Cheng, J.; Heitner, S.; Chen, S.; Rom, W.N.; Hay, J.G. Hypoxia reduces adenoviral replication in cancer cells by downregulation of viral protein expression. Gene Ther. 2005, 12, 911–917. [Google Scholar] [CrossRef]
  71. Hiley, C.T.; Yuan, M.; Lemoine, N.R.; Wang, Y. Lister strain vaccinia virus, a potential therapeutic vector targeting hypoxic tumours. Gene Ther. 2010, 17, 281–287. [Google Scholar] [CrossRef]
  72. Pütz, M.M.; Midgley, C.M.; Law, M.; Smith, G.L. Quantification of antibody responses against multiple antigens of the two infectious forms of Vaccinia virus provides a benchmark for smallpox vaccination. Nat. Med. 2006, 12, 1310–1315. [Google Scholar] [CrossRef]
  73. Peters, C.; Rabkin, S.D. Designing Herpes Viruses as Oncolytics. Mol. Ther.-Oncolytics 2015, 2, 15010. [Google Scholar] [CrossRef] [PubMed]
  74. Kim, K.-J.; Moon, D.; Kong, S.J.; Lee, Y.S.; Yoo, Y.; Kim, S.; Kim, C.; Chon, H.J.; Kim, J.-H.; Choi, K.-J. Antitumor effects of IL-12 and GM-CSF co-expressed in an engineered oncolytic HSV-1. Gene Ther. 2021, 28, 186–198. [Google Scholar] [CrossRef]
  75. Parker, J.N.; Gillespie, G.Y.; Love, C.E.; Randall, S.; Whitley, R.J.; Markert, J.M. Engineered herpes simplex virus expressing IL-12 in the treatment of experimental murine brain tumors. Proc. Natl. Acad. Sci. USA 2000, 97, 2208–2213. [Google Scholar] [CrossRef]
  76. Gaston, D.C.; Odom, C.I.; Li, L.; Markert, J.M.; Roth, J.C.; Cassady, K.A.; Whitley, R.J.; Parker, J.N. Production of bioactive soluble interleukin-15 in complex with interleukin-15 receptor alpha from a conditionally-replicating oncolytic HSV-1. PLoS ONE 2013, 8, e81768. [Google Scholar] [CrossRef]
  77. Ma, R.; Lu, T.; Li, Z.; Teng, K.-Y.; Mansour, A.G.; Yu, M.; Tian, L.; Xu, B.; Ma, S.; Zhang, J.; et al. An Oncolytic Virus Expressing IL15/IL15Rα Combined with Off-the-Shelf EGFR-CAR NK Cells Targets Glioblastoma. Cancer Res. 2021, 81, 3635–3648. [Google Scholar] [CrossRef]
  78. Mineta, T.; Rabkin, S.D.; Martuza, R.L. Treatment of malignant gliomas using ganciclovir-hypersensitive, ribonucleotide reductase-deficient herpes simplex viral mutant. Cancer Res. 1994, 54, 3963–3966. [Google Scholar] [PubMed]
  79. Ju, F.; Luo, Y.; Lin, C.; Jia, X.; Xu, Z.; Tian, R.; Lin, Y.; Zhao, M.; Chang, Y.; Huang, X.; et al. Oncolytic virus expressing PD-1 inhibitors activates a collaborative intratumoral immune response to control tumor and synergizes with CTLA-4 or TIM-3 blockade. J. Immunother. Cancer 2022, 10, e004762. [Google Scholar] [CrossRef]
  80. Wang, G.; Kang, X.; Chen, K.S.; Jehng, T.; Jones, L.; Chen, J.; Huang, X.F.; Chen, S.-Y. An engineered oncolytic virus expressing PD-L1 inhibitors activates tumor neoantigen-specific T cell responses. Nat. Commun. 2020, 11, 1395. [Google Scholar] [CrossRef] [PubMed]
  81. Hamilton, J.R.; Vijayakumar, G.; Palese, P. A Recombinant Antibody-Expressing Influenza Virus Delays Tumor Growth in a Mouse Model. Cell Rep. 2018, 22, 1–7. [Google Scholar] [CrossRef]
  82. Wongthida, P.; Diaz, R.M.; Pulido, C.; Rommelfanger, D.; Galivo, F.; Kaluza, K.; Kottke, T.; Thompson, J.; Melcher, A.; Vile, R. Activating systemic T-cell immunity against self tumor antigens to support oncolytic virotherapy with vesicular stomatitis virus. Hum. Gene Ther. 2011, 22, 1343–1353. [Google Scholar] [CrossRef]
  83. Galanis, E.; Dooley, K.E.; Keith Anderson, S.; Kurokawa, C.B.; Carrero, X.W.; Uhm, J.H.; Federspiel, M.J.; Leontovich, A.A.; Aderca, I.; Viker, K.B.; et al. Carcinoembryonic antigen-expressing oncolytic measles virus derivative in recurrent glioblastoma: A phase 1 trial. Nat. Commun. 2024, 15, 493. [Google Scholar] [CrossRef] [PubMed]
  84. Li, M.; Zhang, M.; Ye, Q.; Liu, Y.; Qian, W. Preclinical and clinical trials of oncolytic vaccinia virus in cancer immunotherapy: A comprehensive review. Cancer Biol. Med. 2023, 20, 646–661. [Google Scholar] [CrossRef] [PubMed]
  85. Yu, F.; Wang, X.; Guo, Z.S.; Bartlett, D.L.; Gottschalk, S.M.; Song, X.T. T-cell engager-armed oncolytic vaccinia virus significantly enhances antitumor therapy. Mol. Ther. 2014, 22, 102–111. [Google Scholar] [CrossRef] [PubMed]
  86. Field, H.J.; Wildy, P. The pathogenicity of thymidine kinase-deficient mutants of herpes simplex virus in mice. J. Hyg. 1978, 81, 267–277. [Google Scholar] [CrossRef] [PubMed]
  87. Martuza, R.L.; Malick, A.; Markert, J.M.; Ruffner, K.L.; Coen, D.M. Experimental therapy of human glioma by means of a genetically engineered virus mutant. Science 1991, 252, 854–856. [Google Scholar] [CrossRef] [PubMed]
  88. Kanai, R.; Zaupa, C.; Sgubin, D.; Antoszczyk, S.J.; Martuza, R.L.; Wakimoto, H.; Rabkin, S.D. Effect of γ34.5 deletions on oncolytic herpes simplex virus activity in brain tumors. J. Virol. 2012, 86, 4420–4431. [Google Scholar] [CrossRef]
  89. Wilcox, D.R.; Longnecker, R. The Herpes Simplex Virus Neurovirulence Factor γ34.5: Revealing Virus-Host Interactions. PLoS Pathog. 2016, 12, e1005449. [Google Scholar] [CrossRef] [PubMed]
  90. Megison, M.L.; Gillory, L.A.; Stewart, J.E.; Nabers, H.C.; Mroczek-Musulman, E.; Waters, A.M.; Coleman, J.M.; Kelly, V.; Markert, J.M.; Gillespie, G.Y.; et al. Preclinical Evaluation of Engineered Oncolytic Herpes Simplex Virus for the Treatment of Pediatric Solid Tumors. PLoS ONE 2014, 9, e86843. [Google Scholar] [CrossRef]
  91. Gillory, L.A.; Megison, M.L.; Stewart, J.E.; Mroczek-Musulman, E.; Nabers, H.C.; Waters, A.M.; Kelly, V.; Coleman, J.M.; Markert, J.M.; Gillespie, G.Y.; et al. Preclinical Evaluation of Engineered Oncolytic Herpes Simplex Virus for the Treatment of Neuroblastoma. PLoS ONE 2013, 8, e77753. [Google Scholar] [CrossRef]
  92. Markert, J.M.; Cody, J.J.; Parker, J.N.; Coleman, J.M.; Price, K.H.; Kern, E.R.; Quenelle, D.C.; Lakeman, A.D.; Schoeb, T.R.; Palmer, C.A.; et al. Preclinical Evaluation of a Genetically Engineered Herpes Simplex Virus Expressing Interleukin-12. J. Virol. 2012, 86, 5304–5313. [Google Scholar] [CrossRef] [PubMed]
  93. Smyth, M.J.; Taniguchi, M.; Street, S.E.A. The Anti-Tumor Activity of IL-12: Mechanisms of Innate Immunity That Are Model and Dose Dependent1. J. Immunol. 2000, 165, 2665–2670. [Google Scholar] [CrossRef] [PubMed]
  94. Oka, N.; Markova, T.; Tsuzuki, K.; Li, W.; El-Darawish, Y.; Pencheva-Demireva, M.; Yamanishi, K.; Yamanishi, H.; Sakagami, M.; Tanaka, Y.; et al. IL-12 regulates the expansion, phenotype, and function of murine NK cells activated by IL-15 and IL-18. Cancer Immunol. Immunother. 2020, 69, 1699–1712. [Google Scholar] [CrossRef] [PubMed]
  95. Trinchieri, G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat. Rev. Immunol. 2003, 3, 133–146. [Google Scholar] [CrossRef]
  96. Roth, J.C.; Cassady, K.A.; Cody, J.J.; Parker, J.N.; Price, K.H.; Coleman, J.M.; Peggins, J.O.; Noker, P.E.; Powers, N.W.; Grimes, S.D.; et al. Evaluation of the Safety and Biodistribution of M032, an Attenuated Herpes Simplex Virus Type 1 Expressing hIL-12, After Intracerebral Administration to Aotus Nonhuman Primates. Hum. Gene Ther. Clin. Dev. 2014, 25, 16–27. [Google Scholar] [CrossRef] [PubMed]
  97. Saha, D.; Martuza, R.L.; Rabkin, S.D. Macrophage Polarization Contributes to Glioblastoma Eradication by Combination Immunovirotherapy and Immune Checkpoint Blockade. Cancer Cell 2017, 32, 253–267.e5. [Google Scholar] [CrossRef] [PubMed]
  98. Aisenbrey, C.; Sizun, C.; Koch, J.; Herget, M.; Abele, R.; Bechinger, B.; Tampé, R. Structure and dynamics of membrane-associated ICP47, a viral inhibitor of the MHC I antigen-processing machinery. J. Biol. Chem. 2006, 281, 30365–30372. [Google Scholar] [CrossRef]
  99. Kaufman, H.L.; Ruby, C.E.; Hughes, T.; Slingluff, C.L., Jr. Current status of granulocyte-macrophage colony-stimulating factor in the immunotherapy of melanoma. J. Immunother. Cancer 2014, 2, 11. [Google Scholar] [CrossRef]
  100. Kaufman, H.L.; Kim, D.W.; DeRaffele, G.; Mitcham, J.; Coffin, R.S.; 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, 718–730. [Google Scholar] [CrossRef]
  101. Andtbacka, R.H.; Agarwala, S.S.; Ollila, D.W.; Hallmeyer, S.; Milhem, M.; Amatruda, T.; Nemunaitis, J.J.; Harrington, K.J.; Chen, L.; Shilkrut, M.; et al. Cutaneous head and neck melanoma in OPTiM, a randomized phase 3 trial of talimogene laherparepvec versus granulocyte-macrophage colony-stimulating factor for the treatment of unresected stage IIIB/IIIC/IV melanoma. Head Neck 2016, 38, 1752–1758. [Google Scholar] [CrossRef]
  102. Andtbacka, R.H.; Kaufman, H.L.; Collichio, F.; Amatruda, T.; Senzer, N.; Chesney, J.; Delman, K.A.; Spitler, L.E.; Puzanov, I.; Agarwala, S.S.; et al. Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. J. Clin. Oncol. 2015, 33, 2780–2788. [Google Scholar] [CrossRef] [PubMed]
  103. Ressler, J.M.; Karasek, M.; Koch, L.; Silmbrod, R.; Mangana, J.; Latifyan, S.; Aedo-Lopez, V.; Kehrer, H.; Weihsengruber, F.; Koelblinger, P.; et al. Real-life use of talimogene laherparepvec (T-VEC) in melanoma patients in centers in Austria, Switzerland and Germany. J. Immunother. Cancer 2021, 9, e001701. [Google Scholar] [CrossRef]
  104. Todo, T.; Martuza, R.L.; Rabkin, S.D.; Johnson, P.A. Oncolytic herpes simplex virus vector with enhanced MHC class I presentation and tumor cell killing. Proc. Natl. Acad. Sci. USA 2001, 98, 6396–6401. [Google Scholar] [CrossRef] [PubMed]
  105. Todo, T.; Ito, H.; Ino, Y.; Ohtsu, H.; Ota, Y.; Shibahara, J.; Tanaka, M. Intratumoral oncolytic herpes virus G47∆ for residual or recurrent glioblastoma: A phase 2 trial. Nat. Med. 2022, 28, 1630–1639. [Google Scholar] [CrossRef]
  106. Ballman, K.V.; Buckner, J.C.; Brown, P.D.; Giannini, C.; Flynn, P.J.; LaPlant, B.R.; Jaeckle, K.A. The relationship between six-month progression-free survival and 12-month overall survival end points for phase II trials in patients with glioblastoma multiforme. Neuro Oncol. 2007, 9, 29–38. [Google Scholar] [CrossRef]
  107. Miller, K.E.; Cassady, K.A.; Roth, J.C.; Clements, J.; Schieffer, K.M.; Leraas, K.; Miller, A.R.; Prasad, N.; Leavenworth, J.W.; Aban, I.B.; et al. Immune Activity and Response Differences of Oncolytic Viral Therapy in Recurrent Glioblastoma: Gene Expression Analyses of a Phase IB Study. Clin. Cancer Res. 2022, 28, 498–506. [Google Scholar] [CrossRef]
  108. Markert, J.M.; Razdan, S.N.; Kuo, H.C.; Cantor, A.; Knoll, A.; Karrasch, M.; Nabors, L.B.; Markiewicz, M.; Agee, B.S.; Coleman, J.M.; et al. A phase 1 trial of oncolytic HSV-1, G207, given in combination with radiation for recurrent GBM demonstrates safety and radiographic responses. Mol. Ther. 2014, 22, 1048–1055. [Google Scholar] [CrossRef]
  109. Ling, A.L.; Solomon, I.H.; Landivar, A.M.; Nakashima, H.; Woods, J.K.; Santos, A.; Masud, N.; Fell, G.; Mo, X.; Yilmaz, A.S.; et al. Clinical trial links oncolytic immunoactivation to survival in glioblastoma. Nature 2023, 623, 157–166. [Google Scholar] [CrossRef]
  110. Fujioka, N.; Akazawa, R.; Ohashi, K.; Fujii, M.; Ikeda, M.; Kurimoto, M. Interleukin-18 protects mice against acute herpes simplex virus type 1 infection. J. Virol. 1999, 73, 2401–2409. [Google Scholar] [CrossRef] [PubMed]
  111. Sergerie, Y.; Rivest, S.; Boivin, G. Tumor necrosis factor-alpha and interleukin-1 beta play a critical role in the resistance against lethal herpes simplex virus encephalitis. J. Infect. Dis. 2007, 196, 853–860. [Google Scholar] [CrossRef]
  112. Ma, Y.; He, B. Recognition of herpes simplex viruses: Toll-like receptors and beyond. J. Mol. Biol. 2014, 426, 1133–1147. [Google Scholar] [CrossRef] [PubMed]
  113. Jiang, H.; Fueyo, J. Healing after death: Antitumor immunity induced by oncolytic adenoviral therapy. Oncoimmunology 2014, 3, e947872. [Google Scholar] [CrossRef] [PubMed]
  114. Bridle, B.W.; Stephenson, K.B.; Boudreau, J.E.; Koshy, S.; Kazdhan, N.; Pullenayegum, E.; Brunellière, J.; Bramson, J.L.; Lichty, B.D.; Wan, Y. Potentiating cancer immunotherapy using an oncolytic virus. Mol. Ther. 2010, 18, 1430–1439. [Google Scholar] [CrossRef] [PubMed]
  115. Alvarez-Breckenridge, C.A.; Yu, J.; Price, R.; Wojton, J.; Pradarelli, J.; Mao, H.; Wei, M.; Wang, Y.; He, S.; Hardcastle, J.; et al. NK cells impede glioblastoma virotherapy through NKp30 and NKp46 natural cytotoxicity receptors. Nat. Med. 2012, 18, 1827–1834. [Google Scholar] [CrossRef] [PubMed]
  116. Allen, C.; Vongpunsawad, S.; Nakamura, T.; James, C.D.; Schroeder, M.; Cattaneo, R.; Giannini, C.; Krempski, J.; Peng, K.W.; Goble, J.M.; et al. Retargeted oncolytic measles strains entering via the EGFRvIII receptor maintain significant antitumor activity against gliomas with increased tumor specificity. Cancer Res. 2006, 66, 11840–11850. [Google Scholar] [CrossRef] [PubMed]
  117. Miller, C.G.; Fraser, N.W. Role of the immune response during neuro-attenuated herpes simplex virus-mediated tumor destruction in a murine intracranial melanoma model. Cancer Res. 2000, 60, 5714–5722. [Google Scholar] [PubMed]
  118. Heiber, J.F.; Barber, G.N. Vesicular stomatitis virus expressing tumor suppressor p53 is a highly attenuated, potent oncolytic agent. J. Virol. 2011, 85, 10440–10450. [Google Scholar] [CrossRef] [PubMed]
  119. Verron, Q.; Forslund, E.; Brandt, L.; Leino, M.; Frisk, T.W.; Olofsson, P.E.; Önfelt, B. NK cells integrate signals over large areas when building immune synapses but require local stimuli for degranulation. Sci. Signal. 2021, 14, eabe2740. [Google Scholar] [CrossRef] [PubMed]
  120. Gazit, R.; Gruda, R.; Elboim, M.; Arnon, T.I.; Katz, G.; Achdout, H.; Hanna, J.; Qimron, U.; Landau, G.; Greenbaum, E.; et al. Lethal influenza infection in the absence of the natural killer cell receptor gene Ncr1. Nat. Immunol. 2006, 7, 517–523. [Google Scholar] [CrossRef] [PubMed]
  121. Werner, J.M.; Heller, T.; Gordon, A.M.; Sheets, A.; Sherker, A.H.; Kessler, E.; Bean, K.S.; Stevens, M.; Schmitt, J.; Rehermann, B. Innate immune responses in hepatitis C virus-exposed healthcare workers who do not develop acute infection. Hepatology 2013, 58, 1621–1631. [Google Scholar] [CrossRef]
  122. Draghi, M.; Pashine, A.; Sanjanwala, B.; Gendzekhadze, K.; Cantoni, C.; Cosman, D.; Moretta, A.; Valiante, N.M.; Parham, P. NKp46 and NKG2D Recognition of Infected Dendritic Cells Is Necessary for NK Cell Activation in the Human Response to Influenza Infection1. J. Immunol. 2007, 178, 2688–2698. [Google Scholar] [CrossRef] [PubMed]
  123. Vivier, E.; Raulet, D.H.; Moretta, A.; Caligiuri, M.A.; Zitvogel, L.; Lanier, L.L.; Yokoyama, W.M.; Ugolini, S. Innate or adaptive immunity? The example of natural killer cells. Science 2011, 331, 44–49. [Google Scholar] [CrossRef] [PubMed]
  124. Brandt, C.S.; Baratin, M.; Yi, E.C.; Kennedy, J.; Gao, Z.; Fox, B.; Haldeman, B.; Ostrander, C.D.; Kaifu, T.; Chabannon, C. The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans. J. Exp. Med. 2009, 206, 1495–1503. [Google Scholar] [CrossRef] [PubMed]
  125. Armeanu, S.; Bitzer, M.; Lauer, U.M.; Venturelli, S.; Pathil, A.; Krusch, M.; Kaiser, S.; Jobst, J.; Smirnow, I.; Wagner, A.; et al. Natural Killer Cell–Mediated Lysis of Hepatoma Cells via Specific Induction of NKG2D Ligands by the Histone Deacetylase Inhibitor Sodium Valproate. Cancer Res. 2005, 65, 6321. [Google Scholar] [CrossRef] [PubMed]
  126. Guerra, N.; Tan, Y.X.; Joncker, N.T.; Choy, A.; Gallardo, F.; Xiong, N.; Knoblaugh, S.; Cado, D.; Greenberg, N.R.; Raulet, D.H. NKG2D-Deficient Mice Are Defective in Tumor Surveillance in Models of Spontaneous Malignancy. Immunity 2008, 28, 571–580. [Google Scholar] [CrossRef] [PubMed]
  127. Vieillard, V.; Baychelier, F.; Debré, P. NKp44L: A new tool for fighting cancer. Oncoimmunology 2014, 3, e27988. [Google Scholar] [CrossRef] [PubMed]
  128. Wu, S.-Y.; Fu, T.; Jiang, Y.-Z.; Shao, Z.-M. Natural killer cells in cancer biology and therapy. Mol. Cancer 2020, 19, 120. [Google Scholar] [CrossRef] [PubMed]
  129. Bauer, S.; Groh, V.; Wu, J.; Steinle, A.; Phillips, J.H.; Lanier, L.L.; Spies, T. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 1999, 285, 727–729. [Google Scholar] [CrossRef]
  130. Textor, S.; Bossler, F.; Henrich, K.-O.; Gartlgruber, M.; Pollmann, J.; Fiegler, N.; Arnold, A.; Westermann, F.; Waldburger, N.; Breuhahn, K.; et al. The proto-oncogene Myc drives expression of the NK cell-activating NKp30 ligand B7-H6 in tumor cells. Oncoimmunology 2016, 5, e1116674. [Google Scholar] [CrossRef]
  131. Guyton, D.; Caobi, A.; Singh, S.K.; Peyser, M.; Tevfik Dorak, M. 14-OR: HLA REGION AND LUNG CANCER SUSCEPTIBILITY: CONFIRMATION OF BAT3/BAG6 ASSOCIATION AND FUNCTIONAL REPLICATION. Hum. Immunol. 2012, 73, 12. [Google Scholar] [CrossRef]
  132. Hu, C.; Yang, J.; Qi, Z.; Wu, H.; Wang, B.; Zou, F.; Mei, H.; Liu, J.; Wang, W.; Liu, Q. Heat shock proteins: Biological functions, pathological roles, and therapeutic opportunities. MedComm 2022, 3, e161. [Google Scholar] [CrossRef] [PubMed]
  133. Ichiyanagi, T.; Imai, T.; Kajiwara, C.; Mizukami, S.; Nakai, A.; Nakayama, T.; Udono, H. Essential role of endogenous heat shock protein 90 of dendritic cells in antigen cross-presentation. J. Immunol. 2010, 185, 2693–2700. [Google Scholar] [CrossRef] [PubMed]
  134. Udono, H.; Ichiyanagi, T.; Mizukami, S.; Imai, T. Heat shock proteins in antigen trafficking—Implications on antigen presentation to T cells. Int. J. Hyperth. 2009, 25, 617–625. [Google Scholar] [CrossRef] [PubMed]
  135. Albakova, Z.; Armeev, G.A.; Kanevskiy, L.M.; Kovalenko, E.I.; Sapozhnikov, A.M. HSP70 Multi-Functionality in Cancer. Cells 2020, 9, 587. [Google Scholar] [CrossRef] [PubMed]
  136. Guliy, O.I.; Staroverov, S.A.; Dykman, L.A. Heat Shock Proteins in Cancer Diagnostics. Appl. Biochem. Microbiol. 2023, 59, 395–407. [Google Scholar] [CrossRef]
  137. Linder, M.; Pogge von Strandmann, E. The Role of Extracellular HSP70 in the Function of Tumor-Associated Immune Cells. Cancers 2021, 13, 4721. [Google Scholar] [CrossRef] [PubMed]
  138. Multhoff, G. Activation of natural killer cells by heat shock protein 70. Int. J. Hyperth. 2002, 18, 576–585. [Google Scholar] [CrossRef] [PubMed]
  139. Specht, H.M.; Ahrens, N.; Blankenstein, C.; Duell, T.; Fietkau, R.; Gaipl, U.S.; Günther, C.; Gunther, S.; Habl, G.; Hautmann, H.; et al. Heat Shock Protein 70 (Hsp70) Peptide Activated Natural Killer (NK) Cells for the Treatment of Patients with Non-Small Cell Lung Cancer (NSCLC) after Radiochemotherapy (RCTx)—From Preclinical Studies to a Clinical Phase II Trial. Front. Immunol. 2015, 6, 162. [Google Scholar] [CrossRef] [PubMed]
  140. Elsner, L.; Muppala, V.; Gehrmann, M.; Lozano, J.; Malzahn, D.; Bickeböller, H.; Brunner, E.; Zientkowska, M.; Herrmann, T.; Walter, L.; et al. The heat shock protein HSP70 promotes mouse NK cell activity against tumors that express inducible NKG2D ligands. J. Immunol. 2007, 179, 5523–5533. [Google Scholar] [CrossRef]
  141. Dean, I.; Lee, C.Y.C.; Tuong, Z.K.; Li, Z.; Tibbitt, C.A.; Willis, C.; Gaspal, F.; Kennedy, B.C.; Matei-Rascu, V.; Fiancette, R.; et al. Rapid functional impairment of natural killer cells following tumor entry limits anti-tumor immunity. Nat. Commun. 2024, 15, 683. [Google Scholar] [CrossRef]
  142. Wu, Y.; Kuang, D.M.; Pan, W.D.; Wan, Y.L.; Lao, X.M.; Wang, D.; Li, X.F.; Zheng, L. Monocyte/macrophage-elicited natural killer cell dysfunction in hepatocellular carcinoma is mediated by CD48/2B4 interactions. Hepatology 2013, 57, 1107–1116. [Google Scholar] [CrossRef] [PubMed]
  143. Bunting, M.D.; Vyas, M.; Requesens, M.; Langenbucher, A.; Schiferle, E.B.; Manguso, R.T.; Lawrence, M.S.; Demehri, S. Extracellular matrix proteins regulate NK cell function in peripheral tissues. Sci. Adv. 2022, 8, eabk3327. [Google Scholar] [CrossRef] [PubMed]
  144. Rossi, G.R.; Trindade, E.S.; Souza-Fonseca-Guimaraes, F. Tumor Microenvironment-Associated Extracellular Matrix Components Regulate NK Cell Function. Front. Immunol. 2020, 11, 73. [Google Scholar] [CrossRef] [PubMed]
  145. Itagaki, F.; Nakatsuka, K.; Sakai, H.; Endo, S.; Su, M.-T.; Takai, T. Fibronectin on target cells attenuates natural cytotoxicity of NK cells via myeloid immune checkpoint ILT3/LILRB4/gp49B. Int. Immunol. 2023, 35, 339–348. [Google Scholar] [CrossRef] [PubMed]
  146. Mai, Z.; Lin, Y.; Lin, P.; Zhao, X.; Cui, L. Modulating extracellular matrix stiffness: A strategic approach to boost cancer immunotherapy. Cell Death Dis. 2024, 15, 307. [Google Scholar] [CrossRef] [PubMed]
  147. Wang, S.; Li, Y.; Xu, C.; Dong, J.; Wei, J. An oncolytic vaccinia virus encoding hyaluronidase reshapes the extracellular matrix to enhance cancer chemotherapy and immunotherapy. J. Immunother. Cancer 2024, 12, e008431. [Google Scholar] [CrossRef] [PubMed]
  148. Zhang, J.; Han, X.; Hu, X.; Jin, F.; Gao, Z.; Yin, L.; Qin, J.; Yin, F.; Li, C.; Wang, Y. IDO1 impairs NK cell cytotoxicity by decreasing NKG2D/NKG2DLs via promoting miR-18a. Mol. Immunol. 2018, 103, 144–155. [Google Scholar] [CrossRef] [PubMed]
  149. Liu, G.; Atteridge, C.L.; Wang, X.; Lundgren, A.D.; Wu, J.D. The membrane type matrix metalloproteinase MMP14 mediates constitutive shedding of MHC class I chain-related molecule A independent of A disintegrin and metalloproteinases. J. Immunol. 2010, 184, 3346–3350. [Google Scholar] [CrossRef] [PubMed]
  150. Zocchi, M.R.; Catellani, S.; Canevali, P.; Tavella, S.; Garuti, A.; Villaggio, B.; Zunino, A.; Gobbi, M.; Fraternali-Orcioni, G.; Kunkl, A.; et al. High ERp5/ADAM10 expression in lymph node microenvironment and impaired NKG2D ligands recognition in Hodgkin lymphomas. Blood 2012, 119, 1479–1489. [Google Scholar] [CrossRef] [PubMed]
  151. Chitadze, G.; Lettau, M.; Bhat, J.; Wesch, D.; Steinle, A.; Fürst, D.; Mytilineos, J.; Kalthoff, H.; Janssen, O.; Oberg, H.H.; et al. Shedding of endogenous MHC class I-related chain molecules A and B from different human tumor entities: Heterogeneous involvement of the “a disintegrin and metalloproteases” 10 and 17. Int. J. Cancer 2013, 133, 1557–1566. [Google Scholar] [CrossRef]
  152. Lee, J.-C.; Lee, K.-M.; Kim, D.-W.; Heo, D.S. Elevated TGF-β1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients. J. Immunol. 2004, 172, 7335–7340. [Google Scholar] [CrossRef] [PubMed]
  153. Groh, V.; Wu, J.; Yee, C.; Spies, T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 2002, 419, 734–738. [Google Scholar] [CrossRef] [PubMed]
  154. Baragaño Raneros, A.; Suarez Álvarez, B.; López Larrea, C. Secretory pathways generating immunosuppressive NKG2D ligands: New targets for therapeutic intervention. Oncoimmunology 2014, 3, e28497. [Google Scholar] [CrossRef] [PubMed]
  155. Vyas, M.; Reinartz, S.; Hoffmann, N.; Reiners, K.S.; Lieber, S.; Jansen, J.M.; Wagner, U.; Müller, R.; von Strandmann, E.P. Soluble NKG2D ligands in the ovarian cancer microenvironment are associated with an adverse clinical outcome and decreased memory effector T cells independent of NKG2D downregulation. Oncoimmunology 2017, 6, e1339854. [Google Scholar] [CrossRef] [PubMed]
  156. Orange, J.S. Natural killer cell deficiency. J. Allergy Clin. Immunol. 2013, 132, 515–525. [Google Scholar] [CrossRef] [PubMed]
  157. Gineau, L.; Cognet, C.; Kara, N.; Lach, F.P.; Dunne, J.; Veturi, U.; Picard, C.; Trouillet, C.; Eidenschenk, C.; Aoufouchi, S.; et al. Partial MCM4 deficiency in patients with growth retardation, adrenal insufficiency, and natural killer cell deficiency. J. Clin. Investig. 2012, 122, 821–832. [Google Scholar] [CrossRef] [PubMed]
  158. Shaw, R.K.; Issekutz, A.C.; Fraser, R.; Schmit, P.; Morash, B.; Monaco-Shawver, L.; Orange, J.S.; Fernandez, C.V. Bilateral adrenal EBV-associated smooth muscle tumors in a child with a natural killer cell deficiency. Blood 2012, 119, 4009–4012. [Google Scholar] [CrossRef] [PubMed]
  159. Schmidt, K.N.; Leung, B.; Kwong, M.; Zarember, K.A.; Satyal, S.; Navas, T.A.; Wang, F.; Godowski, P.J. APC-independent activation of NK cells by the Toll-like receptor 3 agonist double-stranded RNA. J. Immunol. 2004, 172, 138–143. [Google Scholar] [CrossRef] [PubMed]
  160. Hart, O.M.; Athie-Morales, V.; O’Connor, G.M.; Gardiner, C.M. TLR7/8-mediated activation of human NK cells results in accessory cell-dependent IFN-gamma production. J. Immunol. 2005, 175, 1636–1642. [Google Scholar] [CrossRef]
  161. Nguyen, K.B.; Salazar-Mather, T.P.; Dalod, M.Y.; Van Deusen, J.B.; Wei, X.Q.; Liew, F.Y.; Caligiuri, M.A.; Durbin, J.E.; Biron, C.A. Coordinated and distinct roles for IFN-alpha beta, IL-12, and IL-15 regulation of NK cell responses to viral infection. J. Immunol. 2002, 169, 4279–4287. [Google Scholar] [CrossRef]
  162. Gumá, M.; Angulo, A.; López-Botet, M. NK cell receptors involved in the response to human cytomegalovirus infection. Curr. Top. Microbiol. Immunol. 2006, 298, 207–223. [Google Scholar] [CrossRef]
  163. Petersen, J.L.; Morris, C.R.; Solheim, J.C. Virus Evasion of MHC Class I Molecule Presentation1. J. Immunol. 2003, 171, 4473–4478. [Google Scholar] [CrossRef]
  164. Sun, J.C.; Beilke, J.N.; Lanier, L.L. Adaptive immune features of natural killer cells. Nature 2009, 457, 557–561. [Google Scholar] [CrossRef]
  165. Nabekura, T.; Kanaya, M.; Shibuya, A.; Fu, G.; Gascoigne, N.R.J.; Lanier, L.L. Costimulatory Molecule DNAM-1 Is Essential for Optimal Differentiation of Memory Natural Killer Cells during Mouse Cytomegalovirus Infection. Immunity 2014, 40, 225–234. [Google Scholar] [CrossRef]
  166. Romee, R.; Schneider, S.E.; Leong, J.W.; Chase, J.M.; Keppel, C.R.; Sullivan, R.P.; Cooper, M.A.; Fehniger, T.A. Cytokine activation induces human memory-like NK cells. Blood 2012, 120, 4751–4760. [Google Scholar] [CrossRef]
  167. Cooper, M.A.; Elliott, J.M.; Keyel, P.A.; Yang, L.; Carrero, J.A.; Yokoyama, W.M. Cytokine-induced memory-like natural killer cells. Proc. Natl. Acad. Sci. USA 2009, 106, 1915–1919. [Google Scholar] [CrossRef]
  168. Min-Oo, G.; Bezman, N.A.; Madera, S.; Sun, J.C.; Lanier, L.L. Proapoptotic Bim regulates antigen-specific NK cell contraction and the generation of the memory NK cell pool after cytomegalovirus infection. J. Exp. Med. 2014, 211, 1289–1296. [Google Scholar] [CrossRef]
  169. Sun, J.C.; Beilke, J.N.; Bezman, N.A.; Lanier, L.L. Homeostatic proliferation generates long-lived natural killer cells that respond against viral infection. J. Exp. Med. 2011, 208, 357–368. [Google Scholar] [CrossRef]
  170. Lee, J.; Zhang, T.; Hwang, I.; Kim, A.; Nitschke, L.; Kim, M.; Scott, J.M.; Kamimura, Y.; Lanier, L.L.; Kim, S. Epigenetic modification and antibody-dependent expansion of memory-like NK cells in human cytomegalovirus-infected individuals. Immunity 2015, 42, 431–442. [Google Scholar] [CrossRef]
  171. Liu, L.L.; Landskron, J.; Ask, E.H.; Enqvist, M.; Sohlberg, E.; Traherne, J.A.; Hammer, Q.; Goodridge, J.P.; Larsson, S.; Jayaraman, J.; et al. Critical Role of CD2 Co-stimulation in Adaptive Natural Killer Cell Responses Revealed in NKG2C-Deficient Humans. Cell Rep. 2016, 15, 1088–1099. [Google Scholar] [CrossRef]
  172. Rückert, T.; Lareau, C.A.; Mashreghi, M.F.; Ludwig, L.S.; Romagnani, C. Clonal expansion and epigenetic inheritance of long-lasting NK cell memory. Nat. Immunol. 2022, 23, 1551–1563. [Google Scholar] [CrossRef]
  173. Hammer, Q.; Rückert, T.; Borst, E.M.; Dunst, J.; Haubner, A.; Durek, P.; Heinrich, F.; Gasparoni, G.; Babic, M.; Tomic, A.; et al. Peptide-specific recognition of human cytomegalovirus strains controls adaptive natural killer cells. Nat. Immunol. 2018, 19, 453–463. [Google Scholar] [CrossRef]
  174. Jost, S.; Lucar, O.; Lee, E.; Yoder, T.; Kroll, K.; Sugawara, S.; Smith, S.; Jones, R.; Tweet, G.; Werner, A.; et al. Antigen-specific memory NK cell responses against HIV and influenza use the NKG2/HLA-E axis. Sci. Immunol. 2023, 8, eadi3974. [Google Scholar] [CrossRef]
  175. Escobar, G.; Mangani, D.; Anderson, A.C. T cell factor 1: A master regulator of the T cell response in disease. Sci. Immunol. 2020, 5, eabb9726. [Google Scholar] [CrossRef]
  176. Raghu, D.; Xue, H.H.; Mielke, L.A. Control of Lymphocyte Fate, Infection, and Tumor Immunity by TCF-1. Trends Immunol. 2019, 40, 1149–1162. [Google Scholar] [CrossRef]
  177. Weber, B.N.; Chi, A.W.; Chavez, A.; Yashiro-Ohtani, Y.; Yang, Q.; Shestova, O.; Bhandoola, A. A critical role for TCF-1 in T-lineage specification and differentiation. Nature 2011, 476, 63–68. [Google Scholar] [CrossRef]
  178. Zhou, X.; Yu, S.; Zhao, D.M.; Harty, J.T.; Badovinac, V.P.; Xue, H.H. Differentiation and persistence of memory CD8(+) T cells depend on T cell factor 1. Immunity 2010, 33, 229–240. [Google Scholar] [CrossRef]
  179. Jeannet, G.; Boudousquié, C.; Gardiol, N.; Kang, J.; Huelsken, J.; Held, W. Essential role of the Wnt pathway effector Tcf-1 for the establishment of functional CD8 T cell memory. Proc. Natl. Acad. Sci. USA 2010, 107, 9777–9782. [Google Scholar] [CrossRef]
  180. Connolly, K.A.; Kuchroo, M.; Venkat, A.; Khatun, A.; Wang, J.; William, I.; Hornick, N.I.; Fitzgerald, B.L.; Damo, M.; Kasmani, M.Y.; et al. A reservoir of stem-like CD8(+) T cells in the tumor-draining lymph node preserves the ongoing antitumor immune response. Sci. Immunol. 2021, 6, eabg7836. [Google Scholar] [CrossRef]
  181. Im, S.J.; Hashimoto, M.; Gerner, M.Y.; Lee, J.; Kissick, H.T.; Burger, M.C.; Shan, Q.; Hale, J.S.; Lee, J.; Nasti, T.H.; et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 2016, 537, 417–421. [Google Scholar] [CrossRef]
  182. Jeevan-Raj, B.; Gehrig, J.; Charmoy, M.; Chennupati, V.; Grandclément, C.; Angelino, P.; Delorenzi, M.; Held, W. The Transcription Factor Tcf1 Contributes to Normal NK Cell Development and Function by Limiting the Expression of Granzymes. Cell Rep. 2017, 20, 613–626. [Google Scholar] [CrossRef]
  183. Torcellan, T.; Friedrich, C.; Doucet-Ladevèze, R.; Ossner, T.; Solé, V.V.; Riedmann, S.; Ugur, M.; Imdahl, F.; Rosshart, S.P.; Arnold, S.J.; et al. Circulating NK cells establish tissue residency upon acute infection of skin and mediate accelerated effector responses to secondary infection. Immunity 2024, 57, 124–140.e127. [Google Scholar] [CrossRef]
  184. Kujur, W.; Murillo, O.; Adduri, R.S.R.; Vankayalapati, R.; Konduru, N.V.; Mulik, S. Memory like NK cells display stem cell like properties after Zika virus infection. PLoS Pathog. 2021, 16, e1009132. [Google Scholar] [CrossRef]
  185. Wang, Y.; Lifshitz, L.; Gellatly, K.; Vinton, C.L.; Busman-Sahay, K.; McCauley, S.; Vangala, P.; Kim, K.; Derr, A.; Jaiswal, S.; et al. HIV-1-induced cytokines deplete homeostatic innate lymphoid cells and expand TCF7-dependent memory NK cells. Nat. Immunol. 2020, 21, 274–286. [Google Scholar] [CrossRef]
  186. Keppel, M.P.; Yang, L.; Cooper, M.A. Murine NK cell intrinsic cytokine-induced memory-like responses are maintained following homeostatic proliferation. J. Immunol. 2013, 190, 4754–4762. [Google Scholar] [CrossRef]
  187. Bednarski, J.J.; Zimmerman, C.; Berrien-Elliott, M.M.; Foltz, J.A.; Becker-Hapak, M.; Neal, C.C.; Foster, M.; Schappe, T.; McClain, E.; Pence, P.P.; et al. Donor memory-like NK cells persist and induce remissions in pediatric patients with relapsed AML after transplant. Blood 2022, 139, 1670–1683. [Google Scholar] [CrossRef]
  188. Fehniger, T.A.; Cooper, M.A. Harnessing NK Cell Memory for Cancer Immunotherapy. Trends Immunol. 2016, 37, 877–888. [Google Scholar] [CrossRef]
  189. Parihar, R. Memory NK cells to forget relapsed AML. Blood 2022, 139, 1607–1608. [Google Scholar] [CrossRef]
  190. Romee, R.; Rosario, M.; Berrien-Elliott, M.M.; Wagner, J.A.; Jewell, B.A.; Schappe, T.; Leong, J.W.; Abdel-Latif, S.; Schneider, S.E.; Willey, S.; et al. Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia. Sci. Transl. Med. 2016, 8, 357ra123. [Google Scholar] [CrossRef]
  191. Hanna, G.J.; Coleman, K.; Birch, G.; Redd, R.A.; Alonso, A.; Bednarz, S.; Daley, H.; Hernandez Rodriguez, D.E.; Shaw, K.L.; Haddad, R.I.; et al. Abstract CT540: A phase 1 trial of cytokine-induced memory-like (CIML) natural killer (NK) cell therapy with IL-15 superagonist in advanced head and neck cancer: Part 1 results. Cancer Res. 2022, 82, CT540. [Google Scholar] [CrossRef]
  192. Uppendahl, L.D.; Felices, M.; Bendzick, L.; Ryan, C.; Kodal, B.; Hinderlie, P.; Boylan, K.L.M.; Skubitz, A.P.N.; Miller, J.S.; Geller, M.A. Cytokine-induced memory-like natural killer cells have enhanced function, proliferation, and in vivo expansion against ovarian cancer cells. Gynecol. Oncol. 2019, 153, 149–157. [Google Scholar] [CrossRef]
  193. Pal, M.; Schwab, L.; Yermakova, A.; Mace, E.M.; Claus, R.; Krahl, A.C.; Woiterski, J.; Hartwig, U.F.; Orange, J.S.; Handgretinger, R.; et al. Tumor-priming converts NK cells to memory-like NK cells. Oncoimmunology 2017, 6, e1317411. [Google Scholar] [CrossRef]
  194. El-Sherbiny, Y.M.; Holmes, T.D.; Wetherill, L.F.; Black, E.V.; Wilson, E.B.; Phillips, S.L.; Scott, G.B.; Adair, R.A.; Dave, R.; Scott, K.J.; et al. Controlled infection with a therapeutic virus defines the activation kinetics of human natural killer cells in vivo. Clin. Exp. Immunol. 2015, 180, 98–107. [Google Scholar] [CrossRef]
  195. Miyamoto, S.; Inoue, H.; Nakamura, T.; Yamada, M.; Sakamoto, C.; Urata, Y.; Okazaki, T.; Marumoto, T.; Takahashi, A.; Takayama, K.; et al. Coxsackievirus B3 Is an Oncolytic Virus with Immunostimulatory Properties That Is Active against Lung Adenocarcinoma. Cancer Res. 2012, 72, 2609–2621. [Google Scholar] [CrossRef]
  196. Ahmed, J.; Chard, L.S.; Yuan, M.; Wang, J.; Howells, A.; Li, Y.; Li, H.; Zhang, Z.; Lu, S.; Gao, D.; et al. A new oncolytic Vacciniavirus augments antitumor immune responses to prevent tumor recurrence and metastasis after surgery. J. Immunother. Cancer 2020, 8, e000415. [Google Scholar] [CrossRef]
  197. Leung, E.Y.L.; Ennis, D.P.; Kennedy, P.R.; Hansell, C.; Dowson, S.; Farquharson, M.; Spiliopoulou, P.; Nautiyal, J.; McNamara, S.; Carlin, L.M.; et al. NK Cells Augment Oncolytic Adenovirus Cytotoxicity in Ovarian Cancer. Mol. Ther.-Oncolytics 2020, 16, 289–301. [Google Scholar] [CrossRef]
  198. Kottke, T.; Thompson, J.; Diaz, R.M.; Pulido, J.; Willmon, C.; Coffey, M.; Selby, P.; Melcher, A.; Harrington, K.; Vile, R.G. Improved systemic delivery of oncolytic reovirus to established tumors using preconditioning with cyclophosphamide-mediated Treg modulation and interleukin-2. Clin. Cancer Res. 2009, 15, 561–569. [Google Scholar] [CrossRef]
  199. Kottke, T.; Galivo, F.; Wongthida, P.; Maria Diaz, R.; Thompson, J.; Jevremovic, D.; Barber, G.N.; Hall, G.; Chester, J.; Selby, P.; et al. Treg Depletion–enhanced IL-2 Treatment Facilitates Therapy of Established Tumors Using Systemically Delivered Oncolytic Virus. Mol. Ther. 2008, 16, 1217–1226. [Google Scholar] [CrossRef]
  200. Kim, Y.; Yoo, J.Y.; Lee, T.J.; Liu, J.; Yu, J.; Caligiuri, M.A.; Kaur, B.; Friedman, A. Complex role of NK cells in regulation of oncolytic virus-bortezomib therapy. Proc. Natl. Acad. Sci. USA 2018, 115, 4927–4932. [Google Scholar] [CrossRef] [PubMed]
  201. Aurelian, L.; Bollino, D.; Colunga, A. The oncolytic virus ÃÂPK has multimodal anti-tumor activity. Pathog. Dis. 2016, 74, ftw050. [Google Scholar] [CrossRef] [PubMed]
  202. Wongthida, P.; Diaz, R.M.; Galivo, F.; Kottke, T.; Thompson, J.; Pulido, J.; Pavelko, K.; Pease, L.; Melcher, A.; Vile, R. Type III IFN interleukin-28 mediates the antitumor efficacy of oncolytic virus VSV in immune-competent mouse models of cancer. Cancer Res. 2010, 70, 4539–4549. [Google Scholar] [CrossRef] [PubMed]
  203. Jarahian, M.; Watzl, C.; Fournier, P.; Arnold, A.; Djandji, D.; Zahedi, S.; Cerwenka, A.; Paschen, A.; Schirrmacher, V.; Momburg, F. Activation of natural killer cells by newcastle disease virus hemagglutinin-neuraminidase. J. Virol. 2009, 83, 8108–8121. [Google Scholar] [CrossRef] [PubMed]
  204. Jarahian, M.; Fiedler, M.; Cohnen, A.; Djandji, D.; Hämmerling, G.J.; Gati, C.; Cerwenka, A.; Turner, P.C.; Moyer, R.W.; Watzl, C. Modulation of NKp30-and NKp46-mediated natural killer cell responses by poxviral hemagglutinin. PLoS Pathog. 2011, 7, e1002195. [Google Scholar] [CrossRef] [PubMed]
  205. Altomonte, J.; Wu, L.; Chen, L.; Meseck, M.; Ebert, O.; García-Sastre, A.; Fallon, J.; Woo, S.L.C. Exponential enhancement of oncolytic vesicular stomatitis virus potency by vector-mediated suppression of inflammatory responses in vivo. Mol. Ther. 2008, 16, 146–153. [Google Scholar] [CrossRef] [PubMed]
  206. Han, J.; Chen, X.; Chu, J.; Xu, B.; Meisen, W.H.; Chen, L.; Zhang, L.; Zhang, J.; He, X.; Wang, Q.E.; et al. TGFβ Treatment Enhances Glioblastoma Virotherapy by Inhibiting the Innate Immune Response. Cancer Res. 2015, 75, 5273–5282. [Google Scholar] [CrossRef] [PubMed]
  207. Fulci, G.; Breymann, L.; Gianni, D.; Kurozomi, K.; Rhee, S.S.; Yu, J.; Kaur, B.; Louis, D.N.; Weissleder, R.; Caligiuri, M.A.; et al. Cyclophosphamide enhances glioma virotherapy by inhibiting innate immune responses. Proc. Natl. Acad. Sci. USA 2006, 103, 12873–12878. [Google Scholar] [CrossRef] [PubMed]
  208. Altomonte, J.; Wu, L.; Meseck, M.; Chen, L.; Ebert, O.; Garcia-Sastre, A.; Fallon, J.; Mandeli, J.; Woo, S.L.C. Enhanced oncolytic potency of vesicular stomatitis virus through vector-mediated inhibition of NK and NKT cells. Cancer Gene Ther. 2009, 16, 266–278. [Google Scholar] [CrossRef]
  209. Westwood, J.A.; Kelly, J.M.; Tanner, J.E.; Kershaw, M.H.; Smyth, M.J.; Hayakawa, Y. Cutting Edge: Novel Priming of Tumor-Specific Immunity by NKG2D-Triggered NK Cell-Mediated Tumor Rejection and Th1-Independent CD4+ T Cell Pathway1. J. Immunol. 2004, 172, 757–761. [Google Scholar] [CrossRef] [PubMed]
  210. Kelly, J.M.; Darcy, P.K.; Markby, J.L.; Godfrey, D.I.; Takeda, K.; Yagita, H.; Smyth, M.J. Induction of tumor-specific T cell memory by NK cell–mediated tumor rejection. Nat. Immunol. 2002, 3, 83–90. [Google Scholar] [CrossRef]
  211. Mocikat, R.; Braumüller, H.; Gumy, A.; Egeter, O.; Ziegler, H.; Reusch, U.; Bubeck, A.; Louis, J.; Mailhammer, R.; Riethmüller, G. Natural killer cells activated by MHC class Ilow targets prime dendritic cells to induce protective CD8 T cell responses. Immunity 2003, 19, 561–569. [Google Scholar] [CrossRef]
  212. Böttcher, J.P.; Bonavita, E.; Chakravarty, P.; Blees, H.; Cabeza-Cabrerizo, M.; Sammicheli, S.; Rogers, N.C.; Sahai, E.; Zelenay, S.; Reis e Sousa, C. NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control. Cell 2018, 172, 1022–1037.e14. [Google Scholar] [CrossRef] [PubMed]
  213. Moretta, A.; Marcenaro, E.; Sivori, S.; Chiesa, M.D.; Vitale, M.; Moretta, L. Early liaisons between cells of the innate immune system in inflamed peripheral tissues. Trends Immunol. 2005, 26, 668–675. [Google Scholar] [CrossRef]
  214. Frasca, L.; Nasso, M.; Spensieri, F.; Fedele, G.; Palazzo, R.; Malavasi, F.; Ausiello, C.M. IFN-γ Arms Human Dendritic Cells to Perform Multiple Effector Functions1. J. Immunol. 2008, 180, 1471–1481. [Google Scholar] [CrossRef] [PubMed]
  215. Rock, K.L.; Reits, E.; Neefjes, J. Present Yourself! By MHC Class I and MHC Class II Molecules. Trends Immunol. 2016, 37, 724–737. [Google Scholar] [CrossRef] [PubMed]
  216. Schoenborn, J.R.; Wilson, C.B. Regulation of interferon-gamma during innate and adaptive immune responses. Adv. Immunol. 2007, 96, 41–101. [Google Scholar] [CrossRef]
  217. Théry, C.; Amigorena, S. The cell biology of antigen presentation in dendritic cells. Curr. Opin. Immunol. 2001, 13, 45–51. [Google Scholar] [CrossRef]
  218. Colbert, J.D.; Cruz, F.M.; Rock, K.L. Cross-presentation of exogenous antigens on MHC I molecules. Curr. Opin. Immunol. 2020, 64, 1–8. [Google Scholar] [CrossRef] [PubMed]
  219. Schuster, P.; Lindner, G.; Thomann, S.; Haferkamp, S.; Schmidt, B. Prospect of Plasmacytoid Dendritic Cells in Enhancing Anti-Tumor Immunity of Oncolytic Herpes Viruses. Cancers 2019, 11, 651. [Google Scholar] [CrossRef] [PubMed]
  220. Kim, D.; Shin, D.-H.; Sung, C.K. The Optimal Balance between Oncolytic Viruses and Natural Killer Cells: A Mathematical Approach. Mathematics 2022, 10, 3370. [Google Scholar] [CrossRef]
  221. Senekal, N.S.; Mahasa, K.J.; Eladdadi, A.; de Pillis, L.; Ouifki, R. Natural Killer Cells Recruitment in Oncolytic Virotherapy: A Mathematical Model. Bull. Math. Biol. 2021, 83, 75. [Google Scholar] [CrossRef]
  222. Mgrditchian, T.; Arakelian, T.; Paggetti, J.; Noman, M.Z.; Viry, E.; Moussay, E.; Van Moer, K.; Kreis, S.; Guerin, C.; Buart, S.; et al. Targeting autophagy inhibits melanoma growth by enhancing NK cells infiltration in a CCL5-dependent manner. Proc. Natl. Acad. Sci. USA 2017, 114, e9271–e9279. [Google Scholar] [CrossRef] [PubMed]
  223. Li, F.; Sheng, Y.; Hou, W.; Sampath, P.; Byrd, D.; Thorne, S.; Zhang, Y. CCL5-armed oncolytic virus augments CCR5-engineered NK cell infiltration and antitumor efficiency. J. Immunother. Cancer 2020, 8, e000131. [Google Scholar] [CrossRef] [PubMed]
  224. Chen, X.; Han, J.; Chu, J.; Zhang, L.; Zhang, J.; Chen, C.; Chen, L.; Wang, Y.; Wang, H.; Yi, L.; et al. A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases. Oncotarget 2016, 7, 27764–27777. [Google Scholar] [CrossRef] [PubMed]
  225. Lee, G.A.; Liou, Y.-H.; Wang, S.-W.; Ko, K.-L.; Jiang, S.-T.; Liao, N.-S. Different NK Cell Developmental Events Require Different Levels of IL-15 Trans-Presentation. J. Immunol. 2011, 187, 1212–1221. [Google Scholar] [CrossRef] [PubMed]
  226. Lucas, M.; Schachterle, W.; Oberle, K.; Aichele, P.; Diefenbach, A. Dendritic cells prime natural killer cells by trans-presenting interleukin 15. Immunity 2007, 26, 503–517. [Google Scholar] [CrossRef] [PubMed]
  227. Mortier, E.; Woo, T.; Advincula, R.; Gozalo, S.; Ma, A. IL-15Ralpha chaperones IL-15 to stable dendritic cell membrane complexes that activate NK cells via trans presentation. J. Exp. Med. 2008, 205, 1213–1225. [Google Scholar] [CrossRef] [PubMed]
  228. Stoklasek, T.A.; Schluns, K.S.; Lefrançois, L. Combined IL-15/IL-15Ralpha immunotherapy maximizes IL-15 activity in vivo. J. Immunol. 2006, 177, 6072–6080. [Google Scholar] [CrossRef] [PubMed]
  229. Huntington, N.D.; Legrand, N.; Alves, N.L.; Jaron, B.; Weijer, K.; Plet, A.; Corcuff, E.; Mortier, E.; Jacques, Y.; Spits, H.; et al. IL-15 trans-presentation promotes human NK cell development and differentiation in vivo. J. Exp. Med. 2008, 206, 25–34. [Google Scholar] [CrossRef] [PubMed]
  230. Huntington, N.D.; Puthalakath, H.; Gunn, P.; Naik, E.; Michalak, E.M.; Smyth, M.J.; Tabarias, H.; Degli-Esposti, M.A.; Dewson, G.; Willis, S.N.; et al. Interleukin 15-mediated survival of natural killer cells is determined by interactions among Bim, Noxa and Mcl-1. Nat. Immunol. 2007, 8, 856–863. [Google Scholar] [CrossRef]
  231. Xu, B.; Ma, R.; Russell, L.; Yoo, J.Y.; Han, J.; Cui, H.; Yi, P.; Zhang, J.; Nakashima, H.; Dai, H.; et al. An oncolytic herpesvirus expressing E-cadherin improves survival in mouse models of glioblastoma. Nat. Biotechnol. 2019, 37, 45–54. [Google Scholar] [CrossRef]
  232. Ito, M.; Maruyama, T.; Saito, N.; Koganei, S.; Yamamoto, K.; Matsumoto, N. Killer cell lectin-like receptor G1 binds three members of the classical cadherin family to inhibit NK cell cytotoxicity. J. Exp. Med. 2006, 203, 289–295. [Google Scholar] [CrossRef] [PubMed]
  233. Li, Y.; Hofmann, M.; Wang, Q.; Teng, L.; Chlewicki, L.K.; Pircher, H.; Mariuzza, R.A. Structure of natural killer cell receptor KLRG1 bound to E-cadherin reveals basis for MHC-independent missing self recognition. Immunity 2009, 31, 35–46. [Google Scholar] [CrossRef] [PubMed]
  234. Draganov, D.D.; Santidrian, A.F.; Minev, I.; Nguyen, D.; Kilinc, M.O.; Petrov, I.; Vyalkova, A.; Lander, E.; Berman, M.; Minev, B.; et al. Delivery of oncolytic vaccinia virus by matched allogeneic stem cells overcomes critical innate and adaptive immune barriers. J. Transl. Med. 2019, 17, 100. [Google Scholar] [CrossRef] [PubMed]
  235. Floerchinger, A.; Klein, J.E.; Finkbeiner, M.S.C.; Schäfer, T.E.; Fuchs, G.; Doerner, J.; Zirngibl, H.; Ackermann, M.; Kvasnicka, H.M.; Chester, K.A.; et al. A vector-encoded bispecific killer engager to harness virus-activated NK cells as anti-tumor effectors. Cell Death Dis. 2023, 14, 104. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Premature OV clearance dampens long-term therapeutic outcomes of OVT. (A) Antiviral response-mediated early OV clearance leads to poor tumor rejection and tumor recurrence. (B) Controlled antiviral response allows OV to spread within a tumor, resulting in improved therapeutic outcomes.
Figure 1. Premature OV clearance dampens long-term therapeutic outcomes of OVT. (A) Antiviral response-mediated early OV clearance leads to poor tumor rejection and tumor recurrence. (B) Controlled antiviral response allows OV to spread within a tumor, resulting in improved therapeutic outcomes.
Vaccines 12 00721 g001
Figure 2. OVT promotes NK cell infiltration and tumor killing. (A) The immune-suppressive TME upregulates checkpoint receptors and ligands on NK and tumor cells, respectively, thereby inducing NK cell dysfunction and exhaustion. (B) OVT converts the immunologically “cold” TME to the “hot” condition, promoting NK cell infiltration and tumor lysis.
Figure 2. OVT promotes NK cell infiltration and tumor killing. (A) The immune-suppressive TME upregulates checkpoint receptors and ligands on NK and tumor cells, respectively, thereby inducing NK cell dysfunction and exhaustion. (B) OVT converts the immunologically “cold” TME to the “hot” condition, promoting NK cell infiltration and tumor lysis.
Vaccines 12 00721 g002
Figure 3. Strategies for improving OVT therapeutic efficacy. Improved therapeutic outcomes for OVT can be achieved by facilitating OV spread and/or promoting tumor-killing capacity of NK cells in different ways.
Figure 3. Strategies for improving OVT therapeutic efficacy. Improved therapeutic outcomes for OVT can be achieved by facilitating OV spread and/or promoting tumor-killing capacity of NK cells in different ways.
Vaccines 12 00721 g003
Table 1. NK cells contribute to therapeutic outcomes of OVTs in preclinical studies.
Table 1. NK cells contribute to therapeutic outcomes of OVTs in preclinical studies.
OVTumor ModelCombination TherapyStudy/References
(Measles virus) MV-GFP-HSNS-EGFRvIIIGlioma (GBM39)Not applicable (N/A)[116]
(Herpes simplex virus-1) gamma34.5 mutantMelanoma lines (Syngeneic DBA/2)N/A[117]
(Vesicular stomatitis virus) VSV-ΔM-mp53Melanoma lines (TS/A and B16)N/A[118]
(Reovirus) ReolysinLeukemia cell line (K562)N/A[194]
Coxsackievirus B3Non-small-cell lung carcinoma line (A549)N/A[195]
(Vaccinia virus) VVΔTKΔN1L-IL12Lewis lung carcinomaN/A[196]
(Adenovirus)
dl922-947, Ad3/Ad11p
Ovarian cancer (TOV21G and OVCAR4)N/A[197]
(Reovirus) WT type 3 Dearing strainMelanoma line (B16)Treg depletion and IL-2 treatment[198]
(Vesicular stomatitis virus) VSV-GFPMelanoma lines (B16 and YAC-1)Treg depletion and IL-2 treatment[199]
Table 2. NK cells impede therapeutic outcomes of OVTs in preclinical studies.
Table 2. NK cells impede therapeutic outcomes of OVTs in preclinical studies.
OVTumor ModelCombination TherapyStudy/References
(Herpes simplex virus-1) rQNestin34.5Human glioma (U87dEGFR)
Syngeneic mouse glioma
(KR158dEGFR)
N/A[115]
(Vesicular stomatitis virus) rVSV-gG and rVSV-fHepatocellular carcinoma lines (BHK-21, McA-RH7777)N/A[205]
(Herpes simplex virus-1) rQNestin34.5Human GBM xenograft (GB30-FFL)
Syngeneic mouse GBM (4C8)
TGF-β[206]
(Herpes simplex virus-1) OV hrR3Rat glioma line (D74/HveC)cyclophosphamide (CPA)[207]
(Vesicular stomatitis virus) rVSV-UL141Hepatocellular carcinoma line (McA-RH7777)N/A[208]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Franks, M.L.; An, J.-H.; Leavenworth, J.W. The Role of Natural Killer Cells in Oncolytic Virotherapy: Friends or Foes? Vaccines 2024, 12, 721. https://doi.org/10.3390/vaccines12070721

AMA Style

Franks ML, An J-H, Leavenworth JW. The Role of Natural Killer Cells in Oncolytic Virotherapy: Friends or Foes? Vaccines. 2024; 12(7):721. https://doi.org/10.3390/vaccines12070721

Chicago/Turabian Style

Franks, Michael L., Ju-Hyun An, and Jianmei W. Leavenworth. 2024. "The Role of Natural Killer Cells in Oncolytic Virotherapy: Friends or Foes?" Vaccines 12, no. 7: 721. https://doi.org/10.3390/vaccines12070721

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop