Next Article in Journal
Whole-Genome Characterization of Rotavirus G9P[6] and G9P[4] Strains That Emerged after Rotavirus Vaccine Introduction in Mozambique
Previous Article in Journal
Useful Clinical Criteria for Identifying Neonates with Congenital Cytomegalovirus Infection at Birth in the Context of an Expanded Targeted Screening Program
Previous Article in Special Issue
Multimodal Therapy Approaches for NUT Carcinoma by Dual Combination of Oncolytic Virus Talimogene Laherparepvec with Small Molecule Inhibitors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oncolytic Viral Therapy in Osteosarcoma

1
Morsani College of Medicine, University of South Florida Health, Tampa, FL 33602, USA
2
Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA
*
Author to whom correspondence should be addressed.
Viruses 2024, 16(7), 1139; https://doi.org/10.3390/v16071139
Submission received: 2 July 2024 / Revised: 13 July 2024 / Accepted: 14 July 2024 / Published: 16 July 2024
(This article belongs to the Special Issue Oncolytic Viruses as Immunotherapeutic Agents)

Abstract

:
Primary bone malignancies, including osteosarcoma (OS), are rare but aggressive. Current OS treatment, involving surgical resection and chemotherapy, has improved survival for non-metastatic cases but remains ineffective for recurrent or metastatic OS. Oncolytic viral therapy (OVT) is a promising alternative, using naturally occurring or genetically modified viruses to selectively target and lyse cancer cells and induce a robust immune response against remaining OS cells. Various oncolytic viruses (OVs), such as adenovirus, herpes simplex virus, and measles virus, have demonstrated efficacy in preclinical OS models. Combining OVT with other therapeutics, such as chemotherapy or immunotherapy, may further improve outcomes. Despite these advances, challenges in reliability of preclinical models, safety, delivery, and immune response must be addressed to optimize OVT for clinical use. Future research should focus on refining delivery methods, exploring combination treatments, and clinical trials to ensure OVT’s efficacy and safety for OS. Overall, OVT represents a novel approach with the potential to drastically improve survival outcomes for patients with OS.

1. Introduction

Primary malignancies of bone are rare, representing only 0.2% of all cancer diagnoses and accounting for 5% of childhood cancer diagnoses [1]. The most common of these is osteosarcoma (OS), a bone tumor in which the tumor cells produce immature bone, known as osteoid [2]. Despite its rarity, osteosarcoma is particularly aggressive with 10 to 20 percent of patients exhibiting evidence of macroscopic metastatic disease at presentation. Metastases most commonly occur in the lungs, followed by other bones [3].
While a range of treatment strategies exist for OS, conventional treatment for localized disease involves the combination of wide surgical resection with neoadjuvant and/or adjuvant chemotherapy. The chemotherapy regimen typically includes doxorubicin and cisplatin, with or without subsequent high-dose methotrexate [4]. For nonmetastatic OS, the introduction of multiagent chemotherapy and improvement of surgical techniques initially increased 5-year survival three-fold to 65–70%, making radical resection with limb salvage possible in greater than 90% of cases [5,6]. In stark contrast, survival outcomes for patients with recurrent or metastatic osteosarcoma remain poor. Despite the initial success of these treatment advances, overall survival rates for OS have not improved appreciably in nearly four decades, necessitating the exploration of novel therapies to advance survival outcomes in this disease [7].
Both targeted gene therapy and immunotherapy have shown recent promise for the treatment of cancer. However, the genetic heterogeneity and immunosuppressive microenvironment of OS have made the utilization of these treatment modalities extremely difficult [7]. Oncolytic viral therapy (OVT) is a new cancer immunotherapy that utilizes naturally occurring or genetically modified viruses that specifically target malignant cells. The role of oncolytic viruses (OVs) in the treatment of cancer is two-fold: these viruses induce oncolysis and activate immune system cells, triggering both innate and adaptive immune responses [8]. OVs have been shown to modify the tumor microenvironment and target tumor vasculature [9,10]. A wide range of OVTs are currently being evaluated for the treatment of various cancers including adenovirus, vaccinia virus, measles virus, parvovirus, and several others [11]. To date, only Talimogene laherparepvec (T-VEC), a herpes simplex virus that is delivered through an intralesional injection, has been granted FDA approval for the treatment of metastatic melanoma [12]. The diverse and robust immune response elicited by OVs highlights their role as a powerful therapeutic tool. Our knowledge on the use of OVT in osteosarcoma is currently sparse but has garnered increasing attention as a treatment option in recent years. This review seeks to compile and summarize the current findings regarding OVT within the context of OS.

Proposed Mechanism of Action of OVT in Osteosarcoma

The general mechanism by which OVT works is via selective tumor cell infection and lysis with subsequent formation of antitumor immunity (see Figure 1 below). OSs are characterized by aberrant cellular proteins and pathways, such as retinoblastoma tumor suppressor (Rb) telomerase, thymidine kinase, Ras, and many others. Additionally, OSs have been shown to modify the local environment to evade the immune system [13]. These features contribute to the aggressiveness and pathologic fitness of OS by facilitating unchecked cell growth. Alternatively, these same features allow unchecked viral replication, which is the basis for the efficacy and selectivity of OVT. Once infected, OVs replicate and lyse tumor cells, inducing immunogenic cell death (ICD). ICD corresponds to the release of damage-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs), and tumor antigens. These molecules induce cytokines which trigger a strong immune response towards tumor cells locally, leading to their destruction. OS-specific antigens can also induce a systemic adaptive immune response, priming the immune system against all OS cells in the body [14,15]. Additionally, OVs can be further modified to alter the OS tumor microenvironment in a way that promotes local immune infiltration and limit tumor growth [13].

2. Therapeutic Potential of OVT in OS

Multiple studies have explored the application of OVT for the treatment of OS, both as a monotherapy and in combination with other cancer treatments. These studies include promising preclinical evaluations of OVT’s effects in various treatment settings (see Table 1 below).

2.1. Monotherapies

Regarding OVT monotherapy for OS, there have been multiple viruses studied to date. Delta-24-ACT is an oncolytic adenovirus (OAd) that is modified to express immune costimulatory ligand 4-1BB (4-1BBL). This ligand, when expressed on virally infected cells, interacts with T cells to promote the survival and expansion of activated T cells, improving targeting of local OS and potentiating a systemic immune response [16]. In a human OS cell line, Delta-24-ACT killed OS cells and triggered the production of damage-associated molecular patterns (DAMPs), a proxy for immunogenic cell death (ICD). In vivo in a murine model, Delta-24-ACT had antitumor effects against both the primary tumor and metastases, and significantly increased survival time. Most notably, all long-term responders developed an anti-OS memory immune response that was effective against rechallenge with the OS cell line. Finally, this OVT exhibited no toxicity [16].
Another OAd is VCN-01, with multiple modifications to improve cancer cell infectivity and tumor microenvironment penetrance. The first is a RGDK motif in the heparin sulphate-glycosaminoglycans (HSG)–binding domain KKTK of the fiber shaft, which readily binds HSGs and integrins that are highly expressed on malignant cells, resulting in greater affinity and specificity for cancer cells. The second is hyaluronidase, which degrades extracellular matrix hyaluronic acid, disturbing the tumor microenvironment and allowing greater immune infiltration. Past research has supported VCN-1 as being a potent antitumor virus both in human OS cell lines and in mice with tibial OS or metastatic OS with no toxicity [17].
Shifting to another OVT for OS, vaccine strain measles virus (MV) has been shown to be a promising species for OS viral therapy. Vaccine strain MV enters cells primarily through CD46, which is highly expressed in tumors, making it specific for cancer cells. MV also has potent anti-OS effects in human OS cell lines as well as decreasing tumor size and extending median survival time in mice with tibial and metastatic OS [21].
An additional virus that has been studied for OVT in OS is protoparvovirus H-1 (H-1PV). This virus selectively infects highly proliferative cells, such as OS cancer cells, because its life cycle depends on host cell factors involved in proliferation and replication [27]. In the context of OS in human cell lines, H-1PV had strong anti-OS effects, with no cytotoxicity to normal cells [22]. Although H-1PV has not been explored for OS in vivo, in mice with Ewing’s sarcoma, H-1PV also showed an antiproliferative effect, but did not improve survival [28].
Reovirus (RV) is another virus that has been trialed in OVT for OS. RV is a naturally occurring virus with preferential replication in cancer cells with activated Ras signaling, such as in OS. RV inhibits growth and viability of human OS cells in vitro and prevents progression of OS tumors in mice [23]. The only other setting in which RV has been studied for OS is in canine cell lines, which were minimally susceptible to RV infection [24]. In the context of cancer patients, RV has been shown to be safe and well tolerated [25].
A further option for OVT in the context of OS is vesicular stomatitis virus (VSV). Prior research has shown a significant oncolytic effect of VSV both in vitro and in vivo in mice with improvements in short-term but not long-term survival [29,30]. This has prompted modification of VSV to encode antitumor micro-RNA143, which is downregulated in OS and may contribute to its pathogenicity. Injection of miRNA143 decreases metastatic potential of OS and VSV expressing miRNA143 improves specificity in other cancers [31,32]. In OS models in vitro, VSV-miRNA143 had significant cytotoxic and antimigration effects, and these effects were much greater than unmodified VSV [26]. Additionally, VSV is a relatively benign virus in humans and is unlikely to have severe cytotoxic effects on normal cells [33].

2.2. Combination Therapies

OVT may not be able to fully eliminate OS when applied alone. Therefore, combining OVs with other therapies, such as chemotherapy and immunotherapy, offers a compelling strategy to further improve oncologic outcomes. One such example of combinatory therapy combines an OAd (dlE102) with granulocyte-colony stimulating factor (G-CSF). OAds target cells with a defective Rb pathway, which is commonly impaired in OS tumors and can be administered via virus-infected human mesenchymal stem cells (MSCs) with suggestion of improved tumor tropism [34]. G-CSF is traditionally used to manage neutropenia in patients receiving chemotherapy [35,36] and has been reported to enhance immune cell infiltration into tumors and to provide more effective responses to immunotherapies [37,38,39]. In vitro and in vivo studies utilizing OAd dlE102-MSCs to treat OS have shown favorable results. Murine models of OS treated with OAds experience reduced tumor size, as well as increased survival when combined with G-CSF. The immunologic profiles of mice treated with OAd-MSCs were comparable to OS patients with good prognoses [18]. Additionally, OAd-MSCs that have been previously trialed for neuroblastoma and other relapsed/refractory solid tumors demonstrated an excellent safety profile [40,41,42].
Another combination therapy that has been studied is doxorubicin and OAd that is modified to express p53 (OBP-702), theorized to produce a more potent oncolytic effect through the p53-apoptotic pathway [43,44]. A previous phase I clinical trial of a similar OAd without p53 expression showed promising safety and antitumor effects in the context of other malignancies [45,46,47]. In human OS cell lines, with a set dosage of DOX, addition of OAd OBP-702 decreased the number and viability of both DOX-sensitive and resistant OS cells in a dose-dependent manner. Furthermore, using a DOX-resistant OS tumor model in mice, combination DOX and OBP-702 significantly mitigated tumor growth, while treatment with either treatment alone did not have this effect [19].
Reovirus (RV), described earlier under monotherapies, has also been looked at in combination with cisplatin. In mice with OS, RV prevents growth of tumors when applied alone, but when combined with cisplatin, these mice showed a significant reduction in tumor size [23].
An additional combination therapy involves use of a modified herpes simplex virus, OV HSV1716, plus anti-PD-1. HSV1716 (Seprehvir) is an HSV-1 OV with deletions in both copies of the neurovirulence gene RL1 (encoding virulence factor γ134.5), making it selectively target cancer cells [20]. PD-1 is an immune checkpoint receptor highly expressed on T cells. The ligand to this receptor, PD-1-ligand (PD-L1), is expressed on a variety of cells throughout the body. Binding of these molecules leads to T cell inactivation, thereby preventing tissue damage and maintaining self-tolerance. Although necessary to keep immune responses in check, malignant cancers, including OS, hijack this pathway by expressing high levels of PD-L1 to evade the immune system [48]. Many immunotherapies such as anti-PD-1 and anti-PD-L1 target this pathway but are ineffective in treating OS alone [49]. In vivo, HSV1716 plus anti-PD-1 has differential survival effects dependent on the line of OS. In mice with less-immunogenic OS tumors, HSV1716 and anti-PD-1 alone or in combination did not affect tumor growth or survival. However, in mice with highly immunoreactive OS tumors, combination therapy delayed tumor growth and prolonged survival. Finally, HSV1716 (Seprehvir) and anti-PD-1 (Pembrolizumab) have independently been shown to be safe in prior studies, although further studies are necessary to validate their safety in a combined approach [50,51,52].

3. Current Challenges and Barriers to Implementation

Despite positive preclinical and clinical data supporting OVT, various challenges regarding the utility of preclinical trials, clinical safety, efficacy, delivery, and regulation must be addressed prior to the adoption of OVT as a mainstay treatment of OS.

3.1. Preclinical Challenges

Regarding the preclinical evaluation of OVs, many viruses, such as varicella-zoster virus, have restrictive species tropism, limiting the utility of in vivo animal models [53]. Additionally, viruses such as herpes simplex virus type 1 infect and propagate through different species in a distinct manner, undermining the results from animal models [54,55,56,57]. Immunocompromised mice can host human OS, overcoming some limitations of syngeneic models. However, this also compromises the applicability of these studies, as OVT relies on an immunocompetent host [58]. In addition, most preclinical OV studies plant tumors superficially for ease of administration, but this can result in deviations from the typical tumor microenvironment seen in patients with OS [59]. Orthotopic mouse models are more ideal for recreating the microenvironment of clinical cases but are more labor intensive and time consuming for tumor implantation and virus administration. Animal models are incredibly useful for determining efficacy and safety of OVT, but these barriers highlight the limitations of such models for extrapolation of data to humans [12].

3.2. Safety: Off-Target Effects, Toxicities, Mutation, Viral Transmission

As live viruses, OVs present concerns for off-target effects, unexpected toxicities, mutation, recombination with wild-type viruses in the environment, and transmission to others [12,14]. Additionally, administration of some OVs is associated with fever, chills, nausea, vomiting, arthralgia, among many other symptoms resulting from a systemic immune response [60]. These adverse outcomes are difficult to predict, making the ability to eliminate OVs a crucial safety component of therapy [16,40,41,42,50]. This feature makes oncolytic HSVs, such as HSV1716, an attractive option for OS. These OVs retain the enzyme thymidine kinase, making them easily neutralizable and limiting the potential for serious adverse events [42,50]. The risk of transmission to healthy individuals has been mitigated through the development of multiple protocols to aid clinicians and patients in proper storage, handling, administration, and sterilization techniques [61,62,63]. Additionally, there has been no evidence of transmission of the FDA-approved OV T-VEC to healthy individuals [12]. Nonetheless, short- and long-term safety with OVT remains a concern, reflected in clinical trials where immunocompromised patients and those with active viral infections are excluded [15].

3.3. Host vs. Virus

Another challenge of OVT is the immune system’s ability to recognize and neutralize the OV before it can produce a therapeutic effect. First, the innate immune system is exceptional at targeting viruses and may quickly eliminate the virus before it can reach OS cells in significant quantities [64,65,66]. Additionally, the adaptive immune system’s neutralizing antibodies also diminishes the efficacy of OVs. More than half of humans already have antibodies against HSV-1, measles, and reovirus, and in response to OVT, antibodies are rapidly generated, limiting the window and dose range in which therapy is effective [67,68,69,70,71,72]. Due to these factors, high and frequent administered viral doses required to overcome these antibodies lead to administration and safety concerns [13,68]. Immunosuppressive agents that dampen the immune response, such as cyclophosphamide [73], or engineering viruses to evade immune detection may combat this. However, these approaches come with their own pitfalls such as increased susceptibility to infection, treatment toxicity, and potential negation of the immunogenic and antitumor effect of OVTs [12]. This necessitates a delicate immunologic balance to ensure the efficacy of OVT for OS while retaining patient safety.

3.4. Delivery Method

The delivery mechanism of OVs presents another challenge to its application in OS. Due to a systemic immune response limiting viral spread, intratumoral injection is the most advantageous route for administering OVT, and this is the route currently being used for the only FDA-approved OVT, T-VEC [74]. Intratumoral injections are ideal for superficial and small tumors, but this method comes with the risk of damaging implicated structures with large tumors or deep metastases, such as to the lungs [13,75]. Other routes, such as intravenous administration can reach virtually any metastasis; however, the viral load at these locations is often decreased due to significant viral destruction by the host. This has prompted exploration of specific viruses, such as vaccinia, that when enveloped is not neutralized by antibodies, and can reach distant organs with high viral loads [76,77]. Additionally, a delivery system utilizing nanoparticles is a strategy that shows potential in addressing the obstacles present in the administration of OVT [78].

4. Future Directions

As evidenced by preclinical studies, OVT is a promising development that may improve the way OS is managed. As a budding therapy, there are several opportunities for investigating treatment optimization. Establishing a safe and effective delivery method is imperative. As noted above, effective OVT delivery must address the challenges of difficult access for deep intratumoral injection, premature neutralization by the immune system, and the safety profile of increased viral loads necessary to combat pre-existing antibodies.
Combination therapies involving OVs may also warrant further exploration. The existing data on combination therapies in OS focus on only a subset of the available therapeutics. Further investigations on combination with other forms of chemotherapy and radiation therapy are needed, especially since this is standard care for unresectable OS.
Lastly, the progression of OVT to clinical trials for OS will provide valuable data that may be more widely applicable to human subjects. While murine models have shown promising results, clinical trials will highlight patient response to OVT and validate its safety and efficacy. These trials will hopefully establish OVT as a viable treatment option for OS, improving patient outcomes.

5. Conclusions

OVT represents a novel approach to the treatment with OS with the ability to selectively target and destroy cancer cells while sparing normal tissues. The synergistic potential of combining OVT with existing therapies represents a novel avenue for more effective treatment of OS. By addressing current challenges, this therapy could revolutionize the management of OS and overcome the seemingly inertial stagnation of survival rates for OS exhibited by the last few decades.

Author Contributions

Conceptualization, A.L. and G.Z.; methodology, A.L., T.K. and T.H.H.; investigation, T.K. and T.H.H.; writing—original draft preparation, T.K., T.H.H. and C.C.; writing—review and editing, T.K., T.H.H., C.C., G.Z., B.C., S.L., D.J., G.D.L., J.M., O.B., J.E.M. and A.L.; visualization, T.H.H.; supervision, A.L.; project administration, A.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. British Sarcoma Group; Gerrand, C.; Athanasou, N.; Brennan, B.; Grimer, R.; Judson, I.; Morland, B.; Peake, D.; Seddon, B.; Whelan, J. UK Guidelines for the Management of Bone Sarcomas. Clin. Sarcoma Res. 2016, 6, 7. [Google Scholar] [CrossRef] [PubMed]
  2. Klein, M.J.; Siegal, G.P. Osteosarcoma: Anatomic and Histologic Variants. Am. J. Clin. Pathol. 2006, 125, 555–581. [Google Scholar] [CrossRef] [PubMed]
  3. Miller, B.J.; Cram, P.; Lynch, C.F.; Buckwalter, J.A. Risk Factors for Metastatic Disease at Presentation with Osteosarcoma: An Analysis of the SEER Database. J. Bone Jt. Surg. 2013, 95, e89. [Google Scholar] [CrossRef] [PubMed]
  4. Chou, A.J.; Gorlick, R. Chemotherapy Resistance in Osteosarcoma: Current Challenges and Future Directions. Expert Rev. Anticancer Ther. 2006, 6, 1075–1085. [Google Scholar] [CrossRef] [PubMed]
  5. Kim, M.S.; Bolia, I.K.; Iglesias, B.; Sharf, T.; Roberts, S.I.; Kang, H.; Christ, A.B.; Menendez, L.R. Timing of Treatment in Osteosarcoma: Challenges and Perspectives—A Scoping Review. BMC Cancer 2022, 22, 970. [Google Scholar] [CrossRef] [PubMed]
  6. Wan, J.; Zhang, X.; Liu, T.; Zhang, X. Strategies and Developments of Immunotherapies in Osteosarcoma. Oncol. Lett. 2016, 11, 511–520. [Google Scholar] [CrossRef] [PubMed]
  7. Kansara, M.; Teng, M.W.; Smyth, M.J.; Thomas, D.M. Translational Biology of Osteosarcoma. Nat. Rev. Cancer 2014, 14, 722–735. [Google Scholar] [CrossRef] [PubMed]
  8. Jafari, M.; Kadkhodazadeh, M.; Shapourabadi, M.B.; Goradel, N.H.; Shokrgozar, M.A.; Arashkia, A.; Abdoli, S.; Sharifzadeh, Z. Immunovirotherapy: The Role of Antibody Based Therapeutics Combination with Oncolytic Viruses. Front. Immunol. 2022, 13, 1012806. [Google Scholar] [CrossRef]
  9. Everts, A.; Bergeman, M.; McFadden, G.; Kemp, V. Simultaneous Tumor and Stroma Targeting by Oncolytic Viruses. Biomedicines 2020, 8, 474. [Google Scholar] [CrossRef]
  10. Breitbach, C.J.; De Silva, N.S.; Falls, T.J.; Aladl, U.; Evgin, L.; Paterson, J.; Sun, Y.Y.; Roy, D.G.; Rintoul, J.L.; Daneshmand, M.; et al. Targeting Tumor Vasculature with an Oncolytic Virus. Mol. Ther. 2011, 19, 886–894. [Google Scholar] [CrossRef]
  11. 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]
  12. Shalhout, S.Z.; Miller, D.M.; Emerick, K.S.; Kaufman, H.L. Therapy with Oncolytic Viruses: Progress and Challenges. Nat. Rev. Clin. Oncol. 2023, 20, 160–177. [Google Scholar] [CrossRef] [PubMed]
  13. Wedekind, M.F.; Cripe, T.P. Oncolytic Viruses and Their Potential as a Therapeutic Opportunity in Osteosarcoma. In Current Advances in the Science of Osteosarcoma; Kleinerman, E.S., Gorlick, R., Eds.; Springer International Publishing: Cham, Switzerland, 2020; Volume 1258, pp. 77–89. ISBN 9783030430849. [Google Scholar]
  14. Santos Apolonio, J.; Lima De Souza Gonçalves, V.; Cordeiro Santos, M.L.; Silva Luz, M.; Silva Souza, J.V.; Rocha Pinheiro, S.L.; De Souza, W.R.; Sande Loureiro, M.; De Melo, F.F. Oncolytic Virus Therapy in Cancer: A Current Review. WJV 2021, 10, 229–255. [Google Scholar] [CrossRef] [PubMed]
  15. Lawler, S.E.; Speranza, M.-C.; Cho, C.-F.; Chiocca, E.A. Oncolytic Viruses in Cancer Treatment: A Review. JAMA Oncol. 2017, 3, 841. [Google Scholar] [CrossRef]
  16. Martinez-Velez, N.; Laspidea, V.; Zalacain, M.; Labiano, S.; García-Moure, M.; Puigdelloses, M.; Marrodan, L.; Gonzalez-Huarriz, M.; Herrador, G.; De La Nava, D.; et al. Local Treatment of a Pediatric Osteosarcoma Model with a 4-1BBL Armed Oncolytic Adenovirus Results in an Antitumor Effect and Leads to Immune Memory. Mol. Cancer Ther. 2022, 21, 471–480. [Google Scholar] [CrossRef] [PubMed]
  17. Martínez-Vélez, N.; Xipell, E.; Vera, B.; Acanda De La Rocha, A.; Zalacain, M.; Marrodán, L.; Gonzalez-Huarriz, M.; Toledo, G.; Cascallo, M.; Alemany, R.; et al. The Oncolytic Adenovirus VCN-01 as Therapeutic Approach against Pediatric Osteosarcoma. Clin. Cancer Res. 2016, 22, 2217–2225. [Google Scholar] [CrossRef] [PubMed]
  18. Morales-Molina, A.; Gambera, S.; Leo, A.; García-Castro, J. Combination Immunotherapy Using G-CSF and Oncolytic Virotherapy Reduces Tumor Growth in Osteosarcoma. J. Immunother. Cancer 2021, 9, e001703. [Google Scholar] [CrossRef]
  19. Sugiu, K.; Tazawa, H.; Hasei, J.; Yamakawa, Y.; Omori, T.; Komatsubara, T.; Mochizuki, Y.; Kondo, H.; Osaki, S.; Fujiwara, T.; et al. Oncolytic Virotherapy Reverses Chemoresistance in Osteosarcoma by Suppressing MDR1 Expression. Cancer Chemother. Pharmacol. 2021, 88, 513–524. [Google Scholar] [CrossRef] [PubMed]
  20. Wedekind, M.F.; Miller, K.E.; Chen, C.-Y.; Wang, P.-Y.; Hutzen, B.J.; Currier, M.A.; Nartker, B.; Roberts, R.D.; Boon, L.; Conner, J.; et al. Endogenous Retrovirus Envelope as a Tumor-Associated Immunotherapeutic Target in Murine Osteosarcoma. iScience 2021, 24, 102759. [Google Scholar] [CrossRef]
  21. Domingo-Musibay, E.; Allen, C.; Kurokawa, C.; Hardcastle, J.J.; Aderca, I.; Msaouel, P.; Bansal, A.; Jiang, H.; DeGrado, T.R.; Galanis, E. Measles Edmonston Vaccine Strain Derivatives Have Potent Oncolytic Activity against Osteosarcoma. Cancer Gene Ther. 2014, 21, 483–490. [Google Scholar] [CrossRef]
  22. Geiss, C.; Kis, Z.; Leuchs, B.; Frank-Stöhr, M.; Schlehofer, J.; Rommelaere, J.; Dinsart, C.; Lacroix, J. Preclinical Testing of an Oncolytic Parvovirus: Standard Protoparvovirus H-1PV Efficiently Induces Osteosarcoma Cell Lysis In Vitro. Viruses 2017, 9, 301. [Google Scholar] [CrossRef] [PubMed]
  23. Hingorani, P.; Zhang, W.; Lin, J.; Liu, L.; Guha, C.; Kolb, E.A. Systemic Administration of Reovirus (Reolysin) Inhibits Growth of Human Sarcoma Xenografts. Cancer 2011, 117, 1764–1774. [Google Scholar] [CrossRef] [PubMed]
  24. Igase, M.; Hwang, C.C.; Coffey, M.; Okuda, M.; Noguchi, S.; Mizuno, T. The Oncolytic Effects of Reovirus in Canine Solid Tumor Cell Lines. J. Vet. Med. Sci. 2015, 77, 541–548. [Google Scholar] [CrossRef] [PubMed]
  25. Kolb, E.A.; Sampson, V.; Stabley, D.; Walter, A.; Sol-Church, K.; Cripe, T.; Hingorani, P.; Ahern, C.H.; Weigel, B.J.; Zwiebel, J.; et al. A Phase I Trial and Viral Clearance Study of Reovirus (Reolysin) in Children with Relapsed or Refractory Extra-cranial Solid Tumors: A Children’s Oncology Group Phase I Consortium Report. Pediatr. Blood Cancer 2015, 62, 751–758. [Google Scholar] [CrossRef] [PubMed]
  26. Sakuda, T.; Kubo, T.; Johan, M.P.; Furuta, T.; Sakaguchi, T.; Adachi, N. Development of an Oncolytic Recombinant Vesicular Stomatitis Virus Encoding a Tumor-Suppressor MicroRNA. Anticancer Res. 2020, 40, 6319–6325. [Google Scholar] [CrossRef] [PubMed]
  27. Marchini, A.; Bonifati, S.; Scott, E.M.; Angelova, A.L.; Rommelaere, J. Oncolytic Parvoviruses: From Basic Virology to Clinical Applications. Virol. J. 2015, 12, 6. [Google Scholar] [CrossRef] [PubMed]
  28. Lacroix, J.; Kis, Z.; Josupeit, R.; Schlund, F.; Stroh-Dege, A.; Frank-Stöhr, M.; Leuchs, B.; Schlehofer, J.R.; Rommelaere, J.; Dinsart, C. Preclinical Testing of an Oncolytic Parvovirus in Ewing Sarcoma: Protoparvovirus H-1 Induces Apoptosis and Lytic Infection In Vitro but Fails to Improve Survival In Vivo. Viruses 2018, 10, 302. [Google Scholar] [CrossRef]
  29. Kubo, T.; Shimose, S.; Matsuo, T.; Fujimori, J.; Sakaguchi, T.; Yamaki, M.; Shinozaki, K.; Woo, S.L.C.; Ochi, M. Oncolytic Vesicular Stomatitis Virus Administered by Isolated Limb Perfusion Suppresses Osteosarcoma Growth. J. Orthop. Res. 2011, 29, 795–800. [Google Scholar] [CrossRef]
  30. Johan, M.P.; Kubo, T.; Furuta, T.; Sakuda, T.; Sakaguchi, T.; Nakanishi, M.; Ochi, M.; Adachi, N. Metastatic Tumor Cells Detection and Anti-metastatic Potential with Vesicular Stomatitis Virus in Immunocompetent Murine Model of Osteosarcoma. J. Orthop. Res. 2018, 36, 2562–2569. [Google Scholar] [CrossRef]
  31. Osaki, M.; Takeshita, F.; Sugimoto, Y.; Kosaka, N.; Yamamoto, Y.; Yoshioka, Y.; Kobayashi, E.; Yamada, T.; Kawai, A.; Inoue, T.; et al. MicroRNA-143 Regulates Human Osteosarcoma Metastasis by Regulating Matrix Metalloprotease-13 Expression. Mol. Ther. 2011, 19, 1123–1130. [Google Scholar] [CrossRef]
  32. Edge, R.E.; Falls, T.J.; Brown, C.W.; Lichty, B.D.; Atkins, H.; Bell, J.C. A Let-7 MicroRNA-Sensitive Vesicular Stomatitis Virus Demonstrates Tumor-Specific Replication. Mol. Ther. 2008, 16, 1437–1443. [Google Scholar] [CrossRef] [PubMed]
  33. Letchworth, G.J.; Rodriguez, L.L.; Del Cbarrera, J. Vesicular Stomatitis. Vet. J. 1999, 157, 239–260. [Google Scholar] [CrossRef] [PubMed]
  34. Mahasa, K.J.; De Pillis, L.; Ouifki, R.; Eladdadi, A.; Maini, P.; Yoon, A.-R.; Yun, C.-O. Mesenchymal Stem Cells Used as Carrier Cells of Oncolytic Adenovirus Results in Enhanced Oncolytic Virotherapy. Sci. Rep. 2020, 10, 425. [Google Scholar] [CrossRef] [PubMed]
  35. Lyman, G.H.; Yau, L.; Nakov, R.; Krendyukov, A. Overall Survival and Risk of Second Malignancies with Cancer Chemotherapy and G-CSF Support. Ann. Oncol. 2018, 29, 1903–1910. [Google Scholar] [CrossRef] [PubMed]
  36. Lyman, G.H.; Dale, D.C.; Culakova, E.; Poniewierski, M.S.; Wolff, D.A.; Kuderer, N.M.; Huang, M.; Crawford, J. The Impact of the Granulocyte Colony-Stimulating Factor on Chemotherapy Dose Intensity and Cancer Survival: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Ann. Oncol. 2013, 24, 2475–2484. [Google Scholar] [CrossRef] [PubMed]
  37. Adams, S.; Goldstein, L.J.; Sparano, J.A.; Demaria, S.; Badve, S.S. Tumor Infiltrating Lymphocytes (TILs) Improve Prognosis in Patients with Triple Negative Breast Cancer (TNBC). OncoImmunology 2015, 4, e985930. [Google Scholar] [CrossRef] [PubMed]
  38. Mina, M.; Boldrini, R.; Citti, A.; Romania, P.; D’Alicandro, V.; De Ioris, M.; Castellano, A.; Furlanello, C.; Locatelli, F.; Fruci, D. Tumor-Infiltrating T Lymphocytes Improve Clinical Outcome of Therapy-Resistant Neuroblastoma. Oncol. Immunol. 2015, 4, e1019981. [Google Scholar] [CrossRef]
  39. Vassilakopoulou, M.; Avgeris, M.; Velcheti, V.; Kotoula, V.; Rampias, T.; Chatzopoulos, K.; Perisanidis, C.; Kontos, C.K.; Giotakis, A.I.; Scorilas, A.; et al. Evaluation of PD-L1 Expression and Associated Tumor-Infiltrating Lymphocytes in Laryngeal Squamous Cell Carcinoma. Clin. Cancer Res. 2016, 22, 704–713. [Google Scholar] [CrossRef]
  40. García-Castro, J.; Alemany, R.; Cascalló, M.; Martínez-Quintanilla, J.; Del Mar Arriero, M.; Lassaletta, Á.; Madero, L.; Ramírez, M. Treatment of Metastatic Neuroblastoma with Systemic Oncolytic Virotherapy Delivered by Autologous Mesenchymal Stem Cells: An Exploratory Study. Cancer Gene Ther. 2010, 17, 476–483. [Google Scholar] [CrossRef]
  41. Melen, G.J.; Franco-Luzón, L.; Ruano, D.; González-Murillo, Á.; Alfranca, A.; Casco, F.; Lassaletta, Á.; Alonso, M.; Madero, L.; Alemany, R.; et al. Influence of Carrier Cells on the Clinical Outcome of Children with Neuroblastoma Treated with High Dose of Oncolytic Adenovirus Delivered in Mesenchymal Stem Cells. Cancer Lett. 2016, 371, 161–170. [Google Scholar] [CrossRef]
  42. Ruano, D.; López-Martín, J.A.; Moreno, L.; Lassaletta, Á.; Bautista, F.; Andión, M.; Hernández, C.; González-Murillo, Á.; Melen, G.; Alemany, R.; et al. First-in-Human, First-in-Child Trial of Autologous MSCs Carrying the Oncolytic Virus Icovir-5 in Patients with Advanced Tumors. Mol. Ther. 2020, 28, 1033–1042. [Google Scholar] [CrossRef] [PubMed]
  43. Yamasaki, Y.; Tazawa, H.; Hashimoto, Y.; Kojima, T.; Kuroda, S.; Yano, S.; Yoshida, R.; Uno, F.; Mizuguchi, H.; Ohtsuru, A.; et al. A Novel Apoptotic Mechanism of Genetically Engineered Adenovirus-Mediated Tumour-Specific P53 Overexpression through E1A-Dependent P21 and MDM2 Suppression. Eur. J. Cancer 2012, 48, 2282–2291. [Google Scholar] [CrossRef] [PubMed]
  44. Hasei, J.; Sasaki, T.; Tazawa, H.; Osaki, S.; Yamakawa, Y.; Kunisada, T.; Yoshida, A.; Hashimoto, Y.; Onishi, T.; Uno, F.; et al. Dual Programmed Cell Death Pathways Induced by P53 Transactivation Overcome Resistance to Oncolytic Adenovirus in Human Osteosarcoma Cells. Mol. Cancer Ther. 2013, 12, 314–325. [Google Scholar] [CrossRef] [PubMed]
  45. Osaki, S.; Tazawa, H.; Hasei, J.; Yamakawa, Y.; Omori, T.; Sugiu, K.; Komatsubara, T.; Fujiwara, T.; Sasaki, T.; Kunisada, T.; et al. Ablation of MCL1 Expression by Virally Induced microRNA-29 Reverses Chemoresistance in Human Osteosarcomas. Sci. Rep. 2016, 6, 28953. [Google Scholar] [CrossRef] [PubMed]
  46. Nemunaitis, J.; Tong, A.W.; Nemunaitis, M.; Senzer, N.; Phadke, A.P.; Bedell, C.; Adams, N.; Zhang, Y.-A.; Maples, P.B.; Chen, S.; et al. A Phase I Study of Telomerase-Specific Replication Competent Oncolytic Adenovirus (Telomelysin) for Various Solid Tumors. Mol. Ther. 2010, 18, 429–434. [Google Scholar] [CrossRef] [PubMed]
  47. Sasaki, T.; Tazawa, H.; Hasei, J.; Kunisada, T.; Yoshida, A.; Hashimoto, Y.; Yano, S.; Yoshida, R.; Uno, F.; Kagawa, S.; et al. Preclinical Evaluation of Telomerase-Specific Oncolytic Virotherapy for Human Bone and Soft Tissue Sarcomas. Clin. Cancer Res. 2011, 17, 1828–1838. [Google Scholar] [CrossRef] [PubMed]
  48. Blank, C.; Brown, I.; Peterson, A.C.; Spiotto, M.; Iwai, Y.; Honjo, T.; Gajewski, T.F. PD-L1/B7H-1 Inhibits the Effector Phase of Tumor Rejection by T Cell Receptor (TCR) Transgenic CD8+ T Cells. Cancer Res. 2004, 64, 1140–1145. [Google Scholar] [CrossRef] [PubMed]
  49. Zhang, Z.; Tan, X.; Jiang, Z.; Wang, H.; Yuan, H. Immune Checkpoint Inhibitors in Osteosarcoma: A Hopeful and Challenging Future. Front. Pharmacol. 2022, 13, 1031527. [Google Scholar] [CrossRef] [PubMed]
  50. Streby, K.A.; Geller, J.I.; Currier, M.A.; Warren, P.S.; Racadio, J.M.; Towbin, A.J.; Vaughan, M.R.; Triplet, M.; Ott-Napier, K.; Dishman, D.J.; et al. Intratumoral Injection of HSV1716, an Oncolytic Herpes Virus, Is Safe and Shows Evidence of Immune Response and Viral Replication in Young Cancer Patients. Clin. Cancer Res. 2017, 23, 3566–3574. [Google Scholar] [CrossRef]
  51. Tawbi, H.A.; Burgess, M.; Bolejack, V.; Van Tine, B.A.; Schuetze, S.M.; Hu, J.; D’Angelo, S.; Attia, S.; Riedel, R.F.; Priebat, D.A.; et al. Pembrolizumab in Advanced Soft-Tissue Sarcoma and Bone Sarcoma (SARC028): A Multicentre, Two-Cohort, Single-Arm, Open-Label, Phase 2 Trial. Lancet Oncol. 2017, 18, 1493–1501. [Google Scholar] [CrossRef]
  52. Streby, K.A.; Currier, M.A.; Triplet, M.; Ott, K.; Dishman, D.J.; Vaughan, M.R.; Ranalli, M.A.; Setty, B.; Skeens, M.A.; Whiteside, S.; et al. First-in-Human Intravenous Seprehvir in Young Cancer Patients: A Phase 1 Clinical Trial. Mol. Ther. 2019, 27, 1930–1938. [Google Scholar] [CrossRef]
  53. Zerboni, L.; Sen, N.; Oliver, S.L.; Arvin, A.M. Molecular Mechanisms of Varicella Zoster Virus Pathogenesis. Nat. Rev. Microbiol. 2014, 12, 197–210. [Google Scholar] [CrossRef]
  54. Subramanian, G.; LeBlanc, R.A.; Wardley, R.C.; Fuller, A.O. Defective Entry of Herpes Simplex Virus Types 1 and 2 into Porcine Cells and Lack of Infection in Infant Pigs Indicate Species Tropism. J. Gen. Virol. 1995, 76, 2375–2379. [Google Scholar] [CrossRef] [PubMed]
  55. Montgomery, R.I.; Warner, M.S.; Lum, B.J.; Spear, P.G. Herpes Simplex Virus-1 Entry into Cells Mediated by a Novel Member of the TNF/NGF Receptor Family. Cell 1996, 87, 427–436. [Google Scholar] [CrossRef]
  56. Mata, M.; Zhang, M.; Hu, X.; Fink, D.J. HveC (Nectin-1) Is Expressed at High Levels in Sensory Neurons, but Not in Motor Neurons, of the Rat Peripheral Nervous System. J. Neurovirol. 2001, 7, 476–480. [Google Scholar] [CrossRef] [PubMed]
  57. Taylor, J.M.; Lin, E.; Susmarski, N.; Yoon, M.; Zago, A.; Ware, C.F.; Pfeffer, K.; Miyoshi, J.; Takai, Y.; Spear, P.G. Alternative Entry Receptors for Herpes Simplex Virus and Their Roles in Disease. Cell Host Microbe 2007, 2, 19–28. [Google Scholar] [CrossRef]
  58. Lemos De Matos, A.; Franco, L.S.; McFadden, G. Oncolytic Viruses and the Immune System: The Dynamic Duo. Mol. Ther.—Methods Clin. Dev. 2020, 17, 349–358. [Google Scholar] [CrossRef]
  59. Ottaviani, G.; Jaffe, N. The Epidemiology of Osteosarcoma. In Pediatric and Adolescent Osteosarcoma; Jaffe, N., Bruland, O.S., Bielack, S., Eds.; Springer: Boston, MA, USA, 2009; Volume 152, pp. 3–13. ISBN 9781441902832. [Google Scholar]
  60. Li, Z.; Jiang, Z.; Zhang, Y.; Huang, X.; Liu, Q. Efficacy and Safety of Oncolytic Viruses in Randomized Controlled Trials: A Systematic Review and Meta-Analysis. Cancers 2020, 12, 1416. [Google Scholar] [CrossRef] [PubMed]
  61. Harrington, K.J.; Michielin, O.; Malvehy, J.; Pezzani Grüter, I.; Grove, L.; Frauchiger, A.L.; Dummer, R. A Practical Guide to the Handling and Administration of Talimogene Laherparepvec in Europe. Oncol. Targets Ther. 2017, 10, 3867–3880. [Google Scholar] [CrossRef]
  62. Hom, V.; Karonis, E.; Sigidi, T.; Cawley, K. Development of a Nursing Policy for the Administration of an Oncolytic Virus in the Outpatient Setting. Semin. Oncol. Nurs. 2019, 35, 150928. [Google Scholar] [CrossRef]
  63. Gutzmer, R.; Harrington, K.J.; Hoeller, C.; Lebbé, C.; Malvehy, J.; Öhrling, K.; Downey, G.; Dummer, R. Practical Clinical Guide on the Use of Talimogene Laherparepvec Monotherapy in Patients with Unresectable Melanoma in Europe. Eur. J. Dermatol. 2018, 28, 736–749. [Google Scholar] [CrossRef] [PubMed]
  64. 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]
  65. Fulci, G.; Dmitrieva, N.; Gianni, D.; Fontana, E.J.; Pan, X.; Lu, Y.; Kaufman, C.S.; Kaur, B.; Lawler, S.E.; Lee, R.J.; et al. Depletion of Peripheral Macrophages and Brain Microglia Increases Brain Tumor Titers of Oncolytic Viruses. Cancer Res. 2007, 67, 9398–9406. [Google Scholar] [CrossRef] [PubMed]
  66. 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] [PubMed]
  67. Gong, J.; Sachdev, E.; Mita, A.C.; Mita, M.M. Clinical Development of Reovirus for Cancer Therapy: An Oncolytic Virus with Immune-Mediated Antitumor Activity. WJM 2016, 6, 25. [Google Scholar] [CrossRef] [PubMed]
  68. Russell, S.J.; Federspiel, M.J.; Peng, K.-W.; Tong, C.; Dingli, D.; Morice, W.G.; Lowe, V.; O’Connor, M.K.; Kyle, R.A.; Leung, N.; et al. Remission of Disseminated Cancer After Systemic Oncolytic Virotherapy. Mayo Clin. Proc. 2014, 89, 926–933. [Google Scholar] [CrossRef] [PubMed]
  69. Friedrich, N.; Poethko-Müller, C.; Kuhnert, R.; Matysiak-Klose, D.; Koch, J.; Wichmann, O.; Santibanez, S.; Mankertz, A. Seroprevalence of Measles-, Mumps-, and Rubella-Specific Antibodies in the German Adult Population—Cross-Sectional Analysis of the German Health Interview and Examination Survey for Adults (DEGS1). Lancet Reg. Health—Eur. 2021, 7, 100128. [Google Scholar] [CrossRef] [PubMed]
  70. Quach, H.Q.; Ovsyannikova, I.G.; Grill, D.E.; Warner, N.D.; Poland, G.A.; Kennedy, R.B. Seroprevalence of Measles Antibodies in a Highly MMR-Vaccinated Population. Vaccines 2022, 10, 1859. [Google Scholar] [CrossRef] [PubMed]
  71. Perfetto, B.; Paduano, G.; Grimaldi, E.; Sansone, V.; Donnarumma, G.; Di Giuseppe, G. Seroprevalence for Measles, Varicella, Mumps and Rubella in the Trainee Obstetric Population: A Survey in Southern Italy. Vaccines 2024, 12, 335. [Google Scholar] [CrossRef]
  72. Xu, F.; Sternberg, M.R.; Kottiri, B.J.; McQuillan, G.M.; Lee, F.K.; Nahmias, A.J.; Berman, S.M.; Markowitz, L.E. Trends in Herpes Simplex Virus Type 1 and Type 2 Seroprevalence in the United States. JAMA 2006, 296, 964. [Google Scholar] [CrossRef]
  73. 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]
  74. Monga, V.; Miller, B.J.; Tanas, M.; Boukhar, S.; Allen, B.; Anderson, C.; Stephens, L.; Hartwig, S.; Varga, S.; Houtman, J.; et al. Intratumoral Talimogene Laherparepvec Injection with Concurrent Preoperative Radiation in Patients with Locally Advanced Soft-Tissue Sarcoma of the Trunk and Extremities: Phase IB/II Trial. J. Immunother. Cancer 2021, 9, e003119. [Google Scholar] [CrossRef] [PubMed]
  75. Sheth, R.A.; Murthy, R.; Hong, D.S.; Patel, S.; Overman, M.J.; Diab, A.; Hwu, P.; Tam, A. Assessment of Image-Guided Intratumoral Delivery of Immunotherapeutics in Patients with Cancer. JAMA Netw. Open 2020, 3, e207911. [Google Scholar] [CrossRef] [PubMed]
  76. Vanderplasschen, A.; Hollinshead, M.; Smith, G.L. Antibodies against Vaccinia Virus Do Not Neutralize Extracellular Enveloped Virus but Prevent Virus Release from Infected Cells and Comet Formation. J. Gen. Virol. 1997, 78, 2041–2048. [Google Scholar] [CrossRef] [PubMed]
  77. Breitbach, C.J.; Burke, J.; Jonker, D.; Stephenson, J.; Haas, A.R.; Chow, L.Q.M.; Nieva, J.; Hwang, T.-H.; Moon, A.; Patt, R.; et al. Intravenous Delivery of a Multi-Mechanistic Cancer-Targeted Oncolytic Poxvirus in Humans. Nature 2011, 477, 99–102. [Google Scholar] [CrossRef]
  78. Chen, C.; Wang, S.; Wang, J.; Yao, F.; Tang, X.; Guo, W. Nanosized Drug Delivery Strategies in Osteosarcoma Chemotherapy. APL Bioeng. 2023, 7, 011501. [Google Scholar] [CrossRef]
Figure 1. Proposed mechanism of action of OVT: oncolytic viruses selectively infect and lyse tumor cells, releasing damage-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs), tumor antigens, and cytokines. These molecules are recognized by members of the innate immune system, such as natural killer cells and antigen-presenting cells, and by members of the adaptive immune system, such as helper (CD4) and cytotoxic (CD8) T cells. This results in a robust immune response, leading to the infiltration of the tumor microenvironment by additional immune cells.
Figure 1. Proposed mechanism of action of OVT: oncolytic viruses selectively infect and lyse tumor cells, releasing damage-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs), tumor antigens, and cytokines. These molecules are recognized by members of the innate immune system, such as natural killer cells and antigen-presenting cells, and by members of the adaptive immune system, such as helper (CD4) and cytotoxic (CD8) T cells. This results in a robust immune response, leading to the infiltration of the tumor microenvironment by additional immune cells.
Viruses 16 01139 g001
Table 1. Overview of oncolytic viral therapies in osteosarcoma.
Table 1. Overview of oncolytic viral therapies in osteosarcoma.
Virus (+Additional Therapy)ModelKey Findings
Adenovirus
Delta-24-ACT [16]Human cellsPotent anti-OS effect; triggered release of damage-associated molecular patterns
MiceEffective against primary cancer and metastases; increased survival time; safe, no toxicity
VCN-01 [17]Human cellsPotent anti-OS effect
MiceEffective against primary cancer and metastases; safe, no toxicity
dlE102 + G-CSF [18]MiceReduced tumor size; Increased survival with G-CSF
OBP-702 + Doxorubicin [19]Human cellsPotent anti-OS effect with doxorubicin in a dose-dependent manner
MiceCombination treatment mitigated tumor growth; monotherapies were ineffective
Herpes Simplex Virus
HSV1716 (Seprehvir) + Anti-PD-1 [20]MiceIneffective in less immunogenic OS; Combination therapy delayed growth and prolonged survival in highly immunogenic OS
Measles Virus
MV [21]Human cellsPotent anti-OS effect
MiceEffective against primary cancer and metastases; increased survival
Protoparvovirus
H-1PV [22]Human cellsPotent anti-OS effect; no toxicity to normal cells
Reovirus
RV (Reolysin) + Cisplatin 1 [23,24,25]HumanSafe with little toxicity; no tumor response
Human cellsPotent anti-OS effect
Mice 1Potent anti-OS effect; prevented growth; reduced size with cisplatin
Canine cellsNo anti-OS effect
Vesicular Stomatitis Virus
VSV-miRNA143 [26]Human cellsPotent anti-OS effect, greater than unmodified VSV
1 For RV, combination therapy was only analyzed in mice.
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

Karadimas, T.; Huynh, T.H.; Chose, C.; Zervoudakis, G.; Clampitt, B.; Lapp, S.; Joyce, D.; Letson, G.D.; Metts, J.; Binitie, O.; et al. Oncolytic Viral Therapy in Osteosarcoma. Viruses 2024, 16, 1139. https://doi.org/10.3390/v16071139

AMA Style

Karadimas T, Huynh TH, Chose C, Zervoudakis G, Clampitt B, Lapp S, Joyce D, Letson GD, Metts J, Binitie O, et al. Oncolytic Viral Therapy in Osteosarcoma. Viruses. 2024; 16(7):1139. https://doi.org/10.3390/v16071139

Chicago/Turabian Style

Karadimas, Thomas, Thien Huong Huynh, Chloe Chose, Guston Zervoudakis, Bryan Clampitt, Sean Lapp, David Joyce, George Douglas Letson, Jonathan Metts, Odion Binitie, and et al. 2024. "Oncolytic Viral Therapy in Osteosarcoma" Viruses 16, no. 7: 1139. https://doi.org/10.3390/v16071139

APA Style

Karadimas, T., Huynh, T. H., Chose, C., Zervoudakis, G., Clampitt, B., Lapp, S., Joyce, D., Letson, G. D., Metts, J., Binitie, O., Mullinax, J. E., & Lazarides, A. (2024). Oncolytic Viral Therapy in Osteosarcoma. Viruses, 16(7), 1139. https://doi.org/10.3390/v16071139

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