Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1773 2022-06-27 13:56:07 |
2 layout Meta information modification 1773 2022-06-28 04:34:21 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Sitta, J.;  Claudio, P.P.;  Howard, C.M. Virus-Based Immuno-Oncology Models. Encyclopedia. Available online: https://encyclopedia.pub/entry/24514 (accessed on 15 May 2024).
Sitta J,  Claudio PP,  Howard CM. Virus-Based Immuno-Oncology Models. Encyclopedia. Available at: https://encyclopedia.pub/entry/24514. Accessed May 15, 2024.
Sitta, Juliana, Pier Paolo Claudio, Candace M. Howard. "Virus-Based Immuno-Oncology Models" Encyclopedia, https://encyclopedia.pub/entry/24514 (accessed May 15, 2024).
Sitta, J.,  Claudio, P.P., & Howard, C.M. (2022, June 27). Virus-Based Immuno-Oncology Models. In Encyclopedia. https://encyclopedia.pub/entry/24514
Sitta, Juliana, et al. "Virus-Based Immuno-Oncology Models." Encyclopedia. Web. 27 June, 2022.
Virus-Based Immuno-Oncology Models
Edit

Oncolytic virus (OV) presents a natural or genetically engineered tropism for tumor cells that can be further enhanced to increase both innate and adaptive immune responses. Viruses and other pathogens naturally stimulate stronger immune responses than over-expressed self-antigens normally encountered in solid tumors. 

immunotherapeutic immune-oncology oncolytic virus

1. Introduction

Immunotherapeutic targets have emerged in the past decade as a promising addition to the oncology treatment arsenal for selected types of cancers [1][2]. With the rapid development of new immune anticancer drugs and viruses, accurate and reliable preclinical validation including assessment of tumor growth and response, evaluation of complications, drug resistance, and mechanistic effects became of utmost importance to expedited drug development [3]. Recent advances, including the recognition by the Nobel Prize in 2018 of Dr. James P. Allison and Dr. Tasuku Honjo for their discoveries in immune-oncology treatment, highlight the current research focus on the development of new immunotherapeutic drugs [3][4].
With the advent of new immunotherapeutic targets and new developments in oncolytic virus (OV) immunotherapeutic applications, problems with preclinical validation and safety have emerged [5]. Classically, simple preclinical models often use orthotopic or syngeneic tumors, either in immunocompromised or immunocompetent mice [6]. Although they are essential to initial target and mechanism determination as well as time and cost-effective, these models have demonstrated limited reproducibility in clinical trials [7].
Along those lines, multiple animal models have been developed in an attempt to suffice the need for a reliable preclinical assessment of these drugs. Humanized mice, which comprise immune-deficient mice engrafted with human hematopoietic cells and therefore with a human immune system, are the most promising models [8].

2. Oncologic Immunomodulation

The relationship between tumor tissues and the immunologic system has been increasingly explored with a new concept of immunologic modulation in the fight against cancer cells. The thought that tumors escape immune surveillance is not recent, but evolved significantly in the past decade with the development of immune checkpoint inhibitors (ICI), specifically the ones targeting the cytotoxic T lymphocyte antigen 4 (CTLA4) and programmed cell death 1 (PD1), that showed encouraging results in initial clinical trials [9][10]. Ipilimumab and tremelimumab emerged as therapeutic strategies to augment anti-tumor immunity against cancer [11]. Although initial studies demonstrated improved outcomes with a combination of ICI therapy, long-term follow-up showed no significant clinical difference of combined nivolumab and ipilimumab survival compared to single therapies [12]. Furthermore, ICI’s high cost and immune-related adverse effects became a concern [13][14]. Thus, there is growing interest in complementary agents with safer toxicity profile.
CTLA4 intrinsic function is closely linked to T lymphocyte modulation and a major responsibility for immunologic tolerance [15]. CTLA4 is released from intracellular vesicles to the immunologic synapse following T cell receptor (TCR) activation [16]. CTLA4 attenuates T cells by competitive binding to ligands B7-1 and B7-2. CD28 interaction with ligands B7-1 and B7-2 is intrinsically positive through downstream signaling mediated by phosphoinositide 3-kinase (PIK3) and protein kinase B (AKT) [17][18]. Both co-stimulatory positive and negative interactions are similar in strength, allowing for modulation swift. CTLA4 acts primarily in sites of lymphocyte priming but may also be encountered in varying degrees in peripheric tissues where antigen-presenting cells and activated lymphocytes express B7 ligands [19].
In addition to intrinsic function, CTLA4 also modulates T lymphocyte activation extrinsic mechanisms through regulatory T cells (Tregs) modulation. Loss of extrinsic CTLA4 modulation is sufficient to induce aberrant T cell activation and autoimmunity. In this context, CTLA4 appears to modulate B7 ligand activation in antigen-presenting cells, possibly by a mechanism of trans-endocytosis [20]. CTLA4 modulation occurs by selective competitive attenuation of high strength TCR expression, allowing for medium-strength T cell activation [21].
The PD1-PD-ligand 1 (PD-L1) pathway has shown to significantly improve survival in preclinical and clinical trials and is currently in clinical use for many types of cancer. PD-L1 is expressed by many types of cells including tumor cells while PD-L2 is expressed mainly by normal dendritic cells [22]. Thus, the PD-1 mechanism primarily acts in the periphery. PD-1 signaling pathway is activated upon T and B cell activation and, different from CTLA4, appears to directly inhibit TCR activation through activation of the downstream signaling mediated by the tyrosine phosphatase SHP2 [23][24]. In addition, CD28 has also been demonstrated to confer a primary target for PD-1, indicating a common pathway with CTLA4. The PD-1—PD-L1 interaction attenuates T lymphocyte immune response, and in the setting of cancer, is a major contributor to impaired anti-cancer immune response [25].

3. Oncolytic Viruses and Immuno-Oncologic Modulation

OVs present a natural or genetically engineered tropism for tumor cells that can be further enhanced to increase both innate and adaptive immune responses. Over the years, OVs, particularly adenovirus, have been continuously modified to increase tumor selectivity and minimize toxicity. OV mechanism of action is thought to occur through three main mechanisms: the primary lysis of tumor cells caused by intracellular viral replication, gene modification delivery, and the secondary increase in antigen-presenting molecules leading to an increased adaptive immunologic response. One of the main advantages is that given the selective nature of OVs, these mechanisms may be used systemically to reach tumor metastatic tissue that was not directly inoculated.
Viruses and other pathogens naturally stimulate stronger immune responses than over-expressed self-antigens normally encountered in solid tumors. OVs have evolved through the years to express multiple cell receptors and lack intrinsic replication capabilities. OVs mechanism rely on virus-mediated cytolysis promoting the release of pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), which are cancer cell death sensors [26]. Tumor oncolysis and release of PAMPs and DAMPs, which are recognized by pattern recognition receptors (PRR), lead to DC activation and subsequent CD4 and CD8 T cell priming.
With a growing body of evidence in immunologic modulation for cancer therapy, OVs followed a similar trend by genetically adding immunologic capabilities. Talimogene laherparepvec (T-VEC) is a herpes-simplex virus encoding granulocyte-macrophage colony-stimulating factor (GM-CSF) and was the first OV-based immunotherapy to reach a phase III trial. It is currently FDA approved for the treatment of selected patients with metastatic melanoma that are not candidates for surgical resection. Following intratumoral administration, T-VEC induces cell lysis followed by the release of tumor antigen and subsequent circulation of tumor-specific effective T-cells. Thus, although the administration is local, the treatment effect reaches systemic levels. GM-CSF controls both myeloid differentiation and the function of mature presenting antigen cells. This effect has been replicated with adenovirus (Ad5-D24-GMCSF), which demonstrated complete tumor regression and tumor-specific cytotoxic T lymphocyte response [27].

Oncolytic Viruses and Immunotherapeutic Drugs

Sipuleucel-T autologous cell-based immunotherapy vaccine FDA-approved for the treatment of hormone-refractory prostate cancer. Production is individualized by leukapheresis of the patient’s peripheral blood antigen-presenting cells followed by ex vivo antigen loading with prostate acid phosphatase (PAP) and GM-CSF enhancement [28]. Approval was based upon the IMPACT trial, a multicenter phase 3 clinical trial comparing sipuleucel-T or placebo in men with asymptomatic metastatic castration-resistant prostate cancer. In this population, sipuleucel-T showed modest improvement in overall survival (25.8 months in the sipuleucel-T group vs. 21.7 months in the placebo group) but no effect in time to disease progression [29]. Based on the effects of cancer vaccines such as Sipuleucel-T, several OVs armed with GM-CSF (HSV T-Vec, VV JX-594, Ad Ad5/3-D24-GMCSF, and CG0070 have entered clinical trials [30].
The combination of immunotherapy drugs has been attempted as an approach to increase tumor cell response. However, the combination of immunotherapy drugs carries concerns with increased adverse effects [31]. The concept of utilizing a combination of oncolytic virus and immunotherapy emerged to enhance immunomodulation, particularly to overcome immunosuppressive effects within the TME while maintaining a safety profile. Along those lines, a recent phase II clinical trial demonstrated the improved response of T-VEC combined with ipilimumab with decreased visceral lesion decreases in 52% of patients in the combination arm and 23% of patients in the ipilimumab arm. This research included selected patients bearing advanced melanoma. There was no significant increase in adverse events with the combination approach [32].
With the same concept of vaccine cancer therapy, researchers recently reported on the use of cancer stem cells lysate-pulsed dendritic cell vaccine with induced tumor-specific humoral and cellular immunologic response [33][34][35]. Cancer stem cells are encountered in higher number in selected tumors subtypes and are known for their high differentiation capabilities, contributing to tumor heterogeneity and therapy resistance [36]. Although innovative, response with single cancer stem cell vaccine therapy was limited by the immunosuppressive tumor microenvironment that hampered immunologic response [37]. In a more recent article, researchers explored the use of combination dual or triple therapy with PD-L1 or CTLA4 inhibitors, demonstrating increased T cell proliferation, improved tumor-specific CD8 + T cell response, and inhibition of tumor necrosis factor-beta (TNFβ), resulting in a dramatic tumor response in animal models [38].
Chimeric antigen receptor-modified T cell therapy (CAR-T) is an adoptive autologous T cell therapy strategy targeting cells or TME. CAR-T synthetically generates personalized effector T cells with a high affinity for tumor antigens independent of MHC [39]. CAR-T therapy demonstrated dramatic clinical response in B cell malignancies [40][41][42][43]. However, CAR-T therapy in solid tumors is much less successful, likely secondary to immunosuppressive TME changes [44][45][46]. Li et al. evaluated the combination of CAR-T therapy and oncolytic adenovirus expressing TNFβ receptor II-Fc (rAd.sT) in a triple-negative breast cancer model [47]. This modified adenovirus targets and inhibits TNFβ signaling, decreasing immunosuppressive effects in the TME [48]. The researchers constructed CAR-T cells targeting mesothelin, a protein that is normally expressed by various mesothelial tissues but also over-expressed by a wide range of cancers [49]. The researchers demonstrated moderate mesothelin expression in the MDA-MB-231 cell line. The combination therapy elicited increased apoptosis and the researcher detected a synergistic effect in the expression of IL-2 and IL-6 cytokines, important immunogenic cytokines.
In a different work by Watanabe et al., an engineered oncolytic adenovirus expressing tumor necrosis-alpha (TNFα) and interleukin-2 (IL2) (Ad5/3-E2F-D24-TNFa-IRES-IL2 or Ad-mTNFa-mIL2) was administered to humanized mice bearing pancreatic adenocarcinoma xenografts. Pancreatic adenocarcinoma has a highly immunosuppressive TME. The researchers demonstrated the increased intensity of T cell infiltration with sustained tumor regression after combined meso-CAR-T cells and oncolytic virus therapy. Pancreatic adenocarcinoma was resistant to meso-CAR-T cells alone, but co-administration with Ad-mTNFa-mIL2 elicited tumor regression. Moreover, Ad-mTNFa-mIL2 demonstrated induced increased M1 polarization of macrophages and DC maturation compared to control adenovirus, again indicating that oncolytic therapy enhances innate and adaptive immunity [50].
TMAs and OV interplay, however, is not completely clear and often contradictory in the literature. Although it has been reported that tumor inflammation inhibits viral replication through interferon release, some tumors have demonstrated a response to virus-based anti-tumor immune activation [51][52]. This relationship is, however, not linear and the type of macrophage polarization (M1 or M2) appears to vary by tumor and type of virus [53]. Recently, researchers tested the anti-tumor effect of a recombinant Newcastle disease virus (MEDI5395) expressing GM-CSF and demonstrated enhanced immune-cell activation and pro-inflammatory cytokine release in vitro [54].

References

  1. Esfahani, K.; Roudaia, L.; Buhlaiga, N.; del Rincon, S.V.; Papneja, N.; Miller, W.H. A review of cancer immunotherapy: From the past, to the present, to the future. Curr. Oncol. 2020, 27 (Suppl. 2), S87–S97.
  2. Ventola, C.L. Cancer Immunotherapy, Part 3: Challenges and Future Trends. Pharm. Ther. 2017, 42, 514–521.
  3. Bernadic, M.; Duchon, R.; Aziri, R.; Mladosievicova, B. New principles of cancer therapy give new hope for oncological patients. Bratisl. Med. J. 2019, 120, 15–18.
  4. Guo, Z.S. The 2018 Nobel Prize in medicine goes to cancer immunotherapy (editorial for BMC cancer). BMC Cancer 2018, 18, 1086.
  5. Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35.
  6. Ireson, C.R.; Alavijeh, M.S.; Palmer, A.M.; Fowler, E.R.; Jones, H.J. The role of mouse tumour models in the discovery and development of anticancer drugs. Br. J. Cancer 2019, 121, 101–108.
  7. Guerin, E.; Man, S.; Xu, P.; Kerbel, R.S. A model of postsurgical advanced metastatic breast cancer more accurately replicates the clinical efficacy of antiangiogenic drugs. Cancer Res. 2013, 73, 2743–2748.
  8. Adigbli, G.; Ménoret, S.; Cross, A.R.; Hester, J.; Issa, F.; Anegon, I. Humanization of Immunodeficient Animals for the Modeling of Transplantation, Graft Versus Host Disease, and Regenerative Medicine. Transplantation 2020, 104, 2290–2306.
  9. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N. Engl. J. Med. 2010, 363, 711–723.
  10. Grosso, J.F.; Jure-Kunkel, M.N. CTLA-4 blockade in tumor models: An overview of preclinical and translational research. Cancer Immun. 2013, 13, 5.
  11. Ascierto, P.A.; Marincola, F.M.; Ribas, A. Anti-CTLA4 monoclonal antibodies: The past and the future in clinical application. J. Transl. Med. 2011, 9, 196.
  12. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356.
  13. Michot, J.; Bigenwald, C.; Champiat, S.; Collins, M.; Carbonnel, F.; Postel-Vinay, S.; Berdelou, A.; Varga, A.; Bahleda, R.; Hollebecque, A.; et al. Immune-related adverse events with immune checkpoint blockade: A comprehensive review. Eur. J. Cancer 2016, 54, 139–148.
  14. Andrews, A. Treating with Checkpoint Inhibitors-Figure $1 Million per Patient. Am. Health Drug Benefits 2015, 8, 9.
  15. Walunas, T.L.; Lenschow, D.J.; Bakker, C.Y.; Linsley, P.S.; Freeman, G.J.; Green, J.M.; Thompson, C.B.; Bluestone, J.A. CTLA-4 can function as a negative regulator of T cell activation. Immunity 1994, 1, 405–413.
  16. Brunner, M.C.; Chambers, C.A.; Chan, F.K.; Hanke, J.; Winoto, A.; Allison, J.P. CTLA-4-Mediated inhibition of early events of T cell proliferation. J. Immunol. 1999, 162, 5813–5820.
  17. Linsley, P.S.; Greene, J.L.; Brady, W.; Bajorath, J.; Ledbetter, J.A.; Peach, R. Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity 1994, 1, 793–801.
  18. van der Merwe, P.A.; Bodian, D.L.; Daenke, S.; Linsley, P.; Davis, S.J. CD80 (B7-1) Binds Both CD28 and CTLA-4 with a Low Affinity and Very Fast Kinetics. J. Exp. Med. 1997, 185, 393–404.
  19. Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. 2016, 39, 98–106.
  20. Jain, N.; Nguyen, H.; Chambers, C.; Kang, J. Dual function of CTLA-4 in regulatory T cells and conventional T cells to prevent multiorgan autoimmunity. Proc. Natl. Acad. Sci. USA 2010, 107, 1524–1528.
  21. Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018, 8, 1069–1086.
  22. Joalland, N.; Scotet, E. Emerging Challenges of Preclinical Models of Anti-tumor Immunotherapeutic Strategies Utilizing Vγ9Vδ2 T Cells. Front. Immunol. 2020, 11, 992.
  23. Mizuno, R.; Sugiura, D.; Shimizu, K.; Maruhashi, T.; Watada, M.; Okazaki, I.-M.; Okazaki, T. PD-1 Primarily Targets TCR Signal in the Inhibition of Functional T Cell Activation. Front. Immunol. 2019, 10, 630.
  24. Yokosuka, T.; Takamatsu, M.; Kobayashi-Imanishi, W.; Hashimoto-Tane, A.; Azuma, M.; Saito, T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J. Exp. Med. 2012, 209, 1201–1217.
  25. Wu, M.; Huang, Q.; Xie, Y.; Wu, X.; Ma, H.; Zhang, Y.; Xia, Y. Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation. J. Hematol. Oncol. 2022, 15, 24.
  26. Cerullo, V.; Koski, A.; Vähä-Koskela, M.; Hemminki, A. Oncolytic Adenoviruses for Cancer Immunotherapy. ScienceDirect 2012, 115, 265–318.
  27. Cerullo, V.; Pesonen, S.; Diaconu, I.; Escutenaire, S.; Arstila, P.T.; Ugolini, M.; Nokisalmi, P.; Raki, M.; Laasonen, L.; Särkioja, M.; et al. Oncolytic Adenovirus Coding for Granulocyte Macrophage Colony-Stimulating Factor Induces Antitumoral Immunity in Cancer Patients. Cancer Res. 2010, 70, 4297–4309.
  28. Keshavarz-Fathi, M.; Rezaei, N. Candidate Cancers for Vaccination. In Vaccines for Cancer Immunotherapy; Elsevier: Amsterdam, The Netherlands, 2019; pp. 145–152.
  29. Kantoff, P.W.; Higano, C.S.; Shore, N.D.; Berger, E.R.; Small, E.J.; Penson, D.F.; Redfern, C.H.; Ferrari, A.C.; Dreicer, R.; Sims, R.B.; et al. Sipuleucel-T Immunotherapy for Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2010, 363, 411–422.
  30. Chiocca, E.A.; Rabkin, S.D. Oncolytic Viruses and Their Application to Cancer Immunotherapy. Cancer Immunol. Res. 2014, 2, 295–300.
  31. Ott, P.A.; Hodi, F.S.; Kaufman, H.L.; Wigginton, J.M.; Wolchok, J.D. Combination immunotherapy: A road map. J. ImmunoTherapy Cancer 2017, 5, 16.
  32. Chesney, J.; Puzanov, I.; Collichio, F.; Singh, P.; Milhem, M.M.; Glaspy, J.; Hamid, O.; Ross, M.; Friedlander, P.; Garbe, C.; et al. Randomized, Open-Label Phase II Study Evaluating the Efficacy and Safety of Talimogene Laherparepvec in Combination With Ipilimumab Versus Ipilimumab Alone in Patients With Advanced, Unresectable Melanoma. J. Clin. Oncol. 2018, 36, 1658–1667.
  33. Lu, L.; Tao, H.; Chang, A.E.; Jian-Chuan, X.; Shu, G.; Chen, Q.; Egenti, M.; Owen, J.; Moyer, J.S.; Prince, M.E.; et al. Cancer stem cell vaccine inhibits metastases of primary tumors and induces humoral immune responses against cancer stem cells. Oncoimmunology 2015, 4, e990767.
  34. Pan, Q.; Li, Q.; Liu, S.; Ning, N.; Zhang, X.; Xu, Y.; Chang, A.E.; Wicha, M.S. Concise Review: Targeting Cancer Stem Cells Using Immunologic Approaches. Stem Cells 2015, 33, 2085–2092.
  35. Dashti, A.; Ebrahimi, M.; Hadjati, J.; Memarnejadian, A.; Moazzeni, S.M. Dendritic cell based immunotherapy using tumor stem cells mediates potent antitumor immune responses. Cancer Lett. 2016, 374, 175–185.
  36. Naz, F.; Shi, M.; Sajid, S.; Yang, Z.; Yu, C. Cancer stem cells: A major culprit of intra-tumor heterogeneity. Am. J. Cancer Res. 2021, 11, 5782–5811.
  37. Dillman, R.O. Is there a role for therapeutic cancer vaccines in the age of checkpoint inhibitors? Hum. Vaccin Immunother. 2017, 13, 528–532.
  38. Zheng, F.; Dang, J.; Zhang, H.; Xu, F.; Ba, D.; Zhang, B.; Cheng, F.; Chang, A.E.; Wicha, M.S.; Li, Q. Cancer Stem Cell Vaccination With PD-L1 and CTLA-4 Blockades Enhances the Eradication of Melanoma Stem Cells in a Mouse Tumor Model. J. Immunother. 2018, 41, 361–368.
  39. Feins, S.; Kong, W.; Williams, E.F.; Milone, M.C.; Fraietta, J.A. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am. J. Hematol. 2019, 94 (Suppl. 1), S3–S9.
  40. Porter, D.L.; Levine, B.L.; Kalos, M.; Bagg, A.; June, C.H. Chimeric Antigen Receptor–Modified T Cells in Chronic Lymphoid Leukemia. N. Engl. J. Med. 2011, 365, 725–733.
  41. Kalos, M.; Levine, B.L.; Porter, D.L.; Katz, S.; Grupp, S.A.; Bagg, A.; June, C.H. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci. Transl. Med. 2011, 3, 95ra73.
  42. Maude, S.L.; Frey, N.; Shaw, P.A.; Aplenc, R.; Barrett, D.M.; Bunin, N.J.; Chew, A.; Gonzalez, V.E.; Zheng, Z.; Lacey, S.F.; et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 2014, 371, 1507–1517.
  43. Park, J.H.; Rivière, I.; Gonen, M.; Wang, X.; Sénéchal, B.; Curran, K.J.; Sauter, C.; Wang, Y.; Santomasso, B.; Mead, E.; et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 449–459.
  44. Simula, L.; Corrado, M.; Accordi, B.; Di Rita, A.; Nazio, F.; Antonucci, Y.; Di Daniele, A.; Caicci, F.; Caruana, I.; Soriano, M.E.; et al. JNK1 and ERK1/2 modulate lymphocyte homeostasis via BIM and DRP1 upon AICD induction. Cell Death Differ. 2020, 27, 2749–2767.
  45. Ivica, N.A.; Young, C.M. Tracking the CAR-T Revolution: Analysis of Clinical Trials of CAR-T and TCR-T Therapies for the Treatment of Cancer (1997–2020). Healthcare 2021, 9, 1062.
  46. Huan, T.; Chen, D.; Liu, G.; Zhang, H.; Wang, X.; Wu, Z.; Wu, Y.; Xu, Q.; Yu, F. Activation-induced cell death in CAR-T cell therapy. Hum. Cell 2022, 35, 441–447.
  47. Li, Y.; Xiao, F.; Zhang, A.; Zhang, D.; Nie, W.; Xu, T.; Han, B.; Seth, P.; Wang, H.; Yang, Y.; et al. Oncolytic adenovirus targeting TGF-β enhances anti-tumor responses of mesothelin-targeted chimeric antigen receptor T cell therapy against breast cancer. Cell. Immunol. 2020, 348, 104041.
  48. Yang, Y.; Xu, W.; Peng, D.; Wang, H.; Zhang, X.; Wang, H.; Xiao, F.; Zhu, Y.; Ji, Y.; Gulukota, K.; et al. An Oncolytic Adenovirus Targeting Transforming Growth Factor β Inhibits Protumorigenic Signals and Produces Immune Activation: A Novel Approach to Enhance Anti-PD-1 and Anti-CTLA-4 Therapy. Hum. Gene Ther. 2019, 30, 1117–1132.
  49. Hassan, R.; Bera, T.; Pastan, I. Mesothelin. Clin. Cancer Res. 2004, 10, 3937–3942.
  50. Watanabe, K.; Luo, Y.; Da, T.; Guedan, S.; Ruella, M.; Scholler, J.; Keith, B.; Young, R.M.; Engels, B.; Sorsa, S.; et al. Pancreatic cancer therapy with combined mesothelin-redirected chimeric antigen receptor T cells and cytokine-armed oncolytic adenoviruses. JCI Insight. 2018, 3, e99573.
  51. Marchini, A.; Scott, E.; Rommelaere, J. Overcoming Barriers in Oncolytic Virotherapy with HDAC Inhibitors and Immune Checkpoint Blockade. Viruses. 2016, 8, 9.
  52. Nishikawa, T.; Tung, L.Y.; Kaneda, Y. Systemic Administration of Platelets Incorporating Inactivated Sendai Virus Eradicates Melanoma in Mice. Mol. Ther. 2014, 22, 2046–2055.
  53. Denton, N.; Chen, C.Y.; Scott, T.; Cripe, T. Tumor-Associated Macrophages in Oncolytic Virotherapy: Friend or Foe? Biomedicines 2016, 4, 13.
  54. 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.
More
Information
Subjects: Virology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 290
Revisions: 2 times (View History)
Update Date: 28 Jun 2022
1000/1000