You are currently viewing a new version of our website. To view the old version click .
International Journal of Molecular Sciences
  • Review
  • Open Access

6 September 2020

In Vitro-Transcribed mRNA Chimeric Antigen Receptor T Cell (IVT mRNA CAR T) Therapy in Hematologic and Solid Tumor Management: A Preclinical Update

,
,
and
1
Department of Biotechnology, School of Life Sciences, Karpagam Academy of Higher Education, Coimbatore 641021, India
2
Karpagam Cancer Research Centre, Karpagam Academy of Higher Education, Coimbatore 641021, India
3
IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Palermo, Contrada Casazza–S.S.113, 98124 Messina, Italy
*
Author to whom correspondence should be addressed.
This article belongs to the Special Issue Cytokines/Chemokines in Cancer Metastasis

Abstract

Adoptive T cell immunotherapy has received considerable interest in the treatment of cancer. In recent years, chimeric antigen receptor T cell (CAR T) therapy has emerged as a promising therapy in cancer treatment. In CAR T therapy, T cells from the patients are collected, reprogrammed genetically against tumor antigens, and reintroduced into the patients to trigger an immense immune response against cancer cells. CAR T therapy is successful in hematologic malignancies; however, in solid tumors, CAR T therapy faces multiple challenges, including the on-target off-tumor phenomenon, as most of the tumor-associated antigens are expressed in normal cells as well. Consequently, a transient in vitro-transcribed anti-mRNA-based CAR T cell (IVT mRNA CAR T) approach has been investigated to produce controlled cytotoxicity for a limited duration to avoid any undesirable effects in patients. In vitro and in vivo studies demonstrated the therapeutic ability of mRNA-engineered T cells in solid tumors, including melanoma, neuroblastoma and ovarian cancer; however, very few clinical trials are registered. In the present review, we discuss the effect of IVT mRNA CAR T therapy in preclinical studies related to hematologic malignancies and solid tumor management. In addition, we discuss the clinical trial studies based on IVT mRNA CAR T therapy in cancer.

1. Introduction

In recent years, adoptive T cell immunotherapy has emerged as a promising therapy for cancer patients. It is based on two methods: (i) to isolate the tumor-infiltrating lymphocytes from the primary tumor tissues of patients [1] and (ii) to construct T cells with defined specificity against tumor antigens using gene modification approaches [2]. Two gene modification approaches have been used to manufacture the monoclonal T cells with predetermined antigen specificity, namely T cell receptor (TCR) gene transfer and chimeric antigen receptor (CAR) gene transfer [2]. CAR T (CAR T) cells have received significant attention as the most promising adoptive immunotherapy for cancer. CAR T cells are genetically reprogrammed to express an antigen-specific, non-MHC-restricted receptor. This receptor is composed of the extracellular antigen recognition domain, which is most commonly derived from the single-chain variable fragment (scFv) of a monoclonal antibody fused to a hinge, a transmembrane domain, an intracellular signaling domain and/or co-stimulatory molecules [3,4]. Transformed CAR T cells are constructed using a plasmid or viral vector.
CAR T therapy is successful in hematologic malignancies, for example, B cell malignancies, which include acute lymphoblastic leukemia, chronic lymphoblastic leukemia and non-Hodgkin lymphoma [5]. However, in solid tumors, CAR T therapy faces multiple challenges, with limited success. For example, unlike hematologic malignancies, finding an ideal single target antigen is more difficult in solid tumors. On the other hand, it is more common to detect a tumor-associated antigen(s) (TAA) in a solid tumor. TAAs are overexpressed in tumors but also expressed at the physiological level in normal non-tumor tissues. Proteins such as epidermal growth factor receptor (EGFR), carcinoembryonic antigen (CEA), epidermal growth factor receptor 2 (ERBB2), prostate-specific membrane antigen (PSMA) and mesothelin are examples of frequently targeted TAAs present in solid tumors [6]. Indeed, a lack of tumor antigen specificity of CAR T cells enhances the risk of substantial on-target off-tumor toxicity in normal tissues, which occurs when the indefinite period of CAR expression in T cells attacks non-tumor cells that display the intended antigens. This is one of the clinical challenges in the conventional CAR T therapy in cancer treatment. Other challenges include a lack of knowledge of appropriate tumor specific antigen (s) (TSAs)/TAAs, heterogeneity of tumor antigens, difficulties of CAR T cells to enter into tumor sites and the negative effect of the tumor microenvironment on CAR T cells [7]. In order to circumvent on-target off-tumor toxicity, in vitro transcribed mRNA CAR T (IVT mRNA CAR T) cells are emerging as a safe therapeutic approach, where T cells are transiently reprogrammed with mRNA that encodes chimeric membrane antigen receptor protein against a TSA or TAA. Due to the labile nature of mRNA, IVT- mRNA CAR T reduces the side effects associated with on-target off-tumor toxicity [8]. However, there are limitations associated with the IVT mRNA approach, which include a lack of sufficient longevity of mRNA-redirected T cells, which results in the expression of encoded protein for a few days, poor tumor infiltration, manufacturing challenges when a limited quantity of T cells are available and the risk of side effects when repeated doses of CAR T cells are injected. The scheme of IVT mRNA CAR T therapy in cancer patients is shown in Figure 1.
Figure 1. The scheme of IVT mRNA CAR T therapy in cancer patients. IVT mRNA CAR T: in vitro transcribed mRNA chimeric antigen receptor T cells; TSA: Tumor-specific antigen; TAA: Tumor-associated antigen.
In the present review, we discuss the preclinical reports on the effect of IVT mRNA CAR T in hematologic and solid malignancies. Furthermore, we discuss the clinical trial studies based on IVT mRNA CAR T therapy in cancer. In order to collect research articles related to IVT mRNA CAR T therapy, we performed PubMed and Google Scholar searches using the following key words: “In vitro transcribed mRNA chimeric antigen receptor T cells and cancer”, “IVT- mRNA CAR T and cancer”, “In vitro mRNA CAR T and cancer”, “In vitro mRNA CAR T and hematologic tumors”, “In vitro mRNA CAR T and leukemia”, “In vitro mRNA CAR T and lymphoma”, “In vitro mRNA CAR T and solid tumors”, and “In vitro mRNA CAR T and cancer and case reports”.

4. IVT mRNA-Based Clinical Trials in Hematologic and Solid Tumors

Clinical trials using IVT mRNA CAR T for hematologic and solid tumors have been initiated worldwide. Trials with anti-mesothelin CARs and anti-cMet CARs against malignant pleural mesothelioma (NCT01355965), metastatic pancreatic ductal adenocarcinoma (NCT01897415) and metastatic triple-negative breast cancer (NCT01837602) have been completed. A few of the ongoing clinical trials are studying the therapeutic effect of anti-cMet CARs, anti-CD19 CARs and anti-CD20 CARs against malignant melanoma breast cancer (NCT03060356), Hodgkin’s lymphoma (NCT02624258), B cell leukemia B cell lymphoma (NCT03166878) and non-Hodgkin’s lymphoma B cell chronic lymphocyticeukemia (NCT02315118). In a pilot clinical trial, CAR T cells reprogrammed with mRNA that targeted CD123 were tested in relapsed/refractory acute myeloid leukemia patients [46]. Although the method was safe, no antitumor effect was elicited by CAR T cells. Moreover, the study reported a poor quality of T cells from the patients and a lack of persistence of administered CAR T cells. In 2018, Svoboda et al. reported the transient responses of CD19-targeted mRNA engineered T cells (CD19 CAR T) in patients with relapsed or refractory classical Hodgkin’s lymphoma (cHL). This lymphoma is characterized by scant CD19Hodgkin and Reed-Sternberg (HRS) cells within an immunosuppressive tumor microenvironment which causes limitations for cellular therapies directly targeting antigens expressed on HRS cells [47]. In this trial, the CD19 antigen was targeted against CD19+ B cells present in the tumor microenvironment and putative circulating CD19+HRS cells, which may result in the disruption of the tumor microenvironment necessary for the survival of HRS cells. Four patients were administered CD19 CART cells and obtained transient responses; one patient achieved a complete response and one patient achieved a partial response. Two patients showed no responses and this resulted in progressive disease and stable disease outcomes. All of them were shown to have no severe toxicity owing to the transient expression of the inserted CAR mRNA of the T cells.
Phase I clinical trials have been initiated to investigate the efficacy of these IVT CAR T cells redirected for mesothelin (CARTmeso cells) in malignant pleural mesothelioma patients and other tumors that overexpress mesothelin [48]. During the trial, severe anaphylaxis was observed in one patient, which was attributed to the sudden rise in immunoglobulin G (IgG) after two initial injections. Anaphylaxis may also result from an IgE-driven immune response to foreign CAR moieties that degranulate mast cells [49] and that multiple doses of CAR T cells may contribute to anaphylaxis reactions [48]. Consequently, the dosage schema was modified, followed by the absence of anaphylaxis. In a follow-up study, two of these patients showed partial antitumor responses as evidenced by the presence of novel circulating antitumor antibodies and tumor regression with a lack of apparent on-target off-tumor side effects in normal cells [50]. In another phase I clinical trial with six pancreatic ductal adenocarcinoma patients, CARTmeso cell treatment resulted in an elevated level of circulating antibodies produced against protumor-related proteins such as B cell maturation antigen, Ras-related protein Rab-11B, signal-transducing adaptor protein 1, programmed cell death protein 1 (PD1), programmed death ligand 1 (PDL1), and transducin-like enhancer protein 3 in one patient [51]. Tchou et al. reported that no side effect was noticed in metastatic breast cancer patients who intratumorally received c-Met IVT mRNA CAR T cells in a phase 0 clinical trial. Interestingly, immunohistochemical analysis from excised tumors displayed a high degree of necrosis, suggesting the anti-cancer effect of c-Met IVT mRNA CAR T cells in breast cancer patients [33]. A few more clinical trials are ongoing with breast cancer and colorectal cancer patients; however, no clinical updates have been provided.
An overview of the results obtained in clinical trials related to solid and hematologic malignancies is shown in Table 3.
Table 3. Clinical trials regarding the effects of IVT mRNA CAR T cells in hematologic and solid malignancies (i.v., intravenous).

5. Future Directions

A considerable number of preclinical studies have reported the promising and safer therapeutic ability of IVT mRNA CAR T cells in hematologic and solid tumors. Given the promising preclinical results from IVT mRNA CAR T cells, more clinical trials are warranted to investigate the therapeutic efficacy of mRNA-redirected CAR T cells in different cancers. To achieve this translational part, additional preclinical studies are required to study the cytotoxic and tumor-reducing efficacies of new mRNA-engineered CAR constructs manufactured against novel protumor antigens and to elicit the antitumor response via intravenous injections. In addition, future studies are warranted to circumvent the lack of sustainable function and potency associated with IVT mRNA. Foster et al. purified mRNA using recent RNA technology by inserting a modified 1-methylpseudouridin nucleoside into the mRNA to avoid immune stimulation and purified it of possible double-stranded RNA contaminants, which may halt the translation, with the help of RNase III [23]. Human CAR T cells engineered with purified CD19 mRNA exerted a twofold increase in cytotoxicity towards the Nalm-6 cell line and a 100-fold inhibition of leukemia burden in humanized ALL mice with enhanced persistence. More animal studies designed to investigate the purity level of mRNA required before transfection and to study the potential of re-constructed mRNA in T cells against tumor-specific antigens together with cytokine stimulatory signals are warranted, which might increase the number of clinical trials on IVT mRNA CAR T therapy in hematologic and solid tumors in the future. Last, but not least, more studies should focus on improving the homing of mRNA CAR T cells into the tumor microenvironment by neutralizing the localized immunosuppressive cues.

6. Conclusions

IVT mRNA CAR T cell therapy, a form of adoptive T cell therapy, has received significant attention due to its ability to drive away the tumor immune response from the on-target off-tumor phenomenon in hematologic and solid tumors. Preclinical studies have demonstrated the efficacy of these transiently redirected T cells in different hematologic and solid tumor models. Selective antigen-specific proinflammatory cytokine secretion, cell death induction, tumor growth reduction and prolonged survival are the key results from in vitro and in vivo studies. Limited clinical trials have reported positive responses for IVT mRNA CAR T cell therapy in the management of hematologic and solid tumors. Additional clinical trials with large sets of patients are warranted to test the therapeutic efficacy of IVT mRNA CAR T cells in hematologic and solid malignancies.

Author Contributions

Conceptualization, T.S.R., P.B. and E.M.; investigation, T.S.R. and A.G.; writing—original draft preparation, T.S.R. All authors have read and agreed to the published version of the manuscript.

Funding

The present article was supported by DST-FIST to the Department of Biotechnology, Karpagam Academy of Higher Education (SR/FST/LS-1/2018/187; dated 26 December 2018) and current research funds 2020, Ministry of Health, of IRCCS-Centro Neurolesi “Bonino-Pulejo”, Messina, Italy.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Abbreviations

CAR TChimeric antigen receptor T cell
IVT mRNA CAR Tin vitro transcribed anti-mRNA-based CAR T cells
TCRT cell receptor
CARChimeric antigen receptor
scFvSingle-chain variable fragment
TAATumor-associated antigen
EGFREpidermal growth factor receptor
CEACarcinoembryonic antigen
ERBB2Epidermal growth factor receptor 2
PSMAProstate-specific membrane antigen
B-CLLB cell chronic lymphocytic leukemia
B-NHLB cell non-Hodgkin’s lymphoma
FcγRFragment c gamma receptor
BiTEsBispecific T cell engager
AMLAcute myeloid leukemia
NKG2DNatural killer group 2D
ESFTEwing’s sarcoma family of tumors
TGFβRIImutTransforming growth factor β receptor II frameshift mutation
GM-CSFGranulocyte–macrophage colony Stimulating factor
FRαFolate receptor alpha
EpCAMEpithelial cell adhesion molecule
GBMGlioblastoma multiforme
gp100Glycoprotein 100
TRP-1Tyrosinase-related protein 1
TRP-2Tyrosinase-related protein 2
GD2Disialoganglioside 2
L1-CAML1 cell adhesion molecule
MCSPMelanoma-associated chondroitin sulfate proteoglycan
TETARsT cells expressing two additional receptors
CSPG4Chondroitin sulfate proteoglycan 4
NKTNatural killer T
VEGFR2Vascular endothelial growth factor receptor 2
CARTmeso cellsCAR T cells redirected for mesothelin
IgGImmunoglobulin G
PD1Programmed cell death protein 1
PDL1Programmed death ligand 1

References

  1. Cheadle, E.J.; Gornall, H.; Baldan, V.; Hanson, V.; Hawkins, R.E.; Gilham, D.E. CAR T cells: Driving the road from the laboratory to the clinic. Immunol. Rev. 2014, 257, 91–106. [Google Scholar] [CrossRef] [PubMed]
  2. Sharpe, M.; Mount, N. Genetically modified T cells in cancer therapy: Opportunities and challenges. Dis. Models Mech. 2015, 8, 337–350. [Google Scholar] [CrossRef] [PubMed]
  3. Newick, K.; O’Brien, S.; Moon, E.; Albelda, S.M. CAR T Cell Therapy for Solid Tumors. Annu. Rev. Med. 2017, 68, 139–152. [Google Scholar] [CrossRef] [PubMed]
  4. Garber, K. Driving T-cell immunotherapy to solid tumors. Nat. Biotechnol. 2018, 36, 215–219. [Google Scholar] [CrossRef] [PubMed]
  5. Kenderian, S.S.; Porter, D.L.; Gill, S. Chimeric Antigen Receptor T Cells and Hematopoietic Cell Transplantation: How Not to Put the CART Before the Horse. Biol. Blood Marrow Transplant. 2017, 23, 235–246. [Google Scholar] [CrossRef]
  6. Martinez, M.; Moon, E.K. CAR T Cells for Solid Tumors: New Strategies for Finding, Infiltrating, and Surviving in the Tumor Microenvironment. Front. Immunol. 2019, 10, 128. [Google Scholar] [CrossRef]
  7. Zhang, Q.; Ping, J.; Huang, Z.; Zhang, X.; Zhou, J.; Wang, G.; Liu, S.; Ma, J. CAR-T Cell Therapy in Cancer: Tribulations and Road Ahead. J. Immunol. Res. 2020, 2020, 1924379. [Google Scholar] [CrossRef]
  8. Miliotou, A.N.; Papadopoulou, L.C. In Vitro-Transcribed (IVT)-mRNA CAR Therapy Development. Methods Mol. Biol. 2020, 2086, 87–117. [Google Scholar] [CrossRef]
  9. Braun, D.A.; Wu, C.J. Antigen Discovery and Therapeutic Targeting in Hematologic Malignancies. Cancer J. 2017, 23, 115–124. [Google Scholar] [CrossRef]
  10. Rabinovich, P.M.; Komarovskaya, M.E.; Ye, Z.J.; Imai, C.; Campana, D.; Bahceci, E.; Weissman, S.M. Synthetic messenger RNA as a tool for gene therapy. Hum. Gene Ther. 2006, 17, 1027–1035. [Google Scholar] [CrossRef]
  11. Rabinovich, P.M.; Komarovskaya, M.E.; Wrzesinski, S.H.; Alderman, J.L.; Budak-Alpdogan, T.; Karpikov, A.; Guo, H.F.; Flavell, R.A.; Cheung, N.K.; Weissman, S.M.; et al. Chimeric Receptor mRNA Transfection as a Tool to Generate Antineoplastic Lymphocytes. Hum. Gene Ther. 2009, 20, 51–61. [Google Scholar] [CrossRef] [PubMed]
  12. Almasbak, H.; Rian, E.; Hoel, H.J.; Pule, M.; Walchli, S.; Kvalheim, G.; Gaudernack, G.; Rasmussen, A.M. Transiently redirected T cells for adoptive transfer. Cytotherapy 2011, 13, 629–640. [Google Scholar] [CrossRef] [PubMed]
  13. Koksal, H.; Dillard, P.; Josefsson, S.E.; Maggadottir, S.M.; Pollmann, S.; Fane, A.; Blaker, Y.N.; Beiske, K.; Huse, K.; Kolstad, A.; et al. Preclinical development of CD37CAR T-cell therapy for treatment of B-cell lymphoma. Blood Adv. 2019, 3, 1230–1243. [Google Scholar] [CrossRef] [PubMed]
  14. Panjwani, M.K.; Smith, J.B.; Schutsky, K.; Gnanandarajah, J.; O’Connor, C.M.; Powell, D.J., Jr.; Mason, N.J. Feasibility and Safety of RNA-transfected CD20-specific Chimeric Antigen Receptor T Cells in Dogs with Spontaneous B Cell Lymphoma. Mol. Ther. J. Am. Soc. Gene Ther. 2016, 24, 1602–1614. [Google Scholar] [CrossRef] [PubMed]
  15. Barrett, D.M.; Zhao, Y.; Liu, X.; Jiang, S.; Carpenito, C.; Kalos, M.; Carroll, R.G.; June, C.H.; Grupp, S.A. Treatment of advanced leukemia in mice with mRNA engineered T cells. Hum. Gene Ther. 2011, 22, 1575–1586. [Google Scholar] [CrossRef]
  16. Barrett, D.M.; Liu, X.; Jiang, S.; June, C.H.; Grupp, S.A.; Zhao, Y. Regimen-specific effects of RNA-modified chimeric antigen receptor T cells in mice with advanced leukemia. Hum. Gene Ther. 2013, 24, 717–727. [Google Scholar] [CrossRef]
  17. Almasbak, H.; Walseng, E.; Kristian, A.; Myhre, M.R.; Suso, E.M.; Munthe, L.A.; Andersen, J.T.; Wang, M.Y.; Kvalheim, G.; Gaudernack, G.; et al. Inclusion of an IgG1-Fc spacer abrogates efficacy of CD19 CAR T cells in a xenograft mouse model. Gene Ther. 2015, 22, 391–403. [Google Scholar] [CrossRef]
  18. Hudecek, M.; Sommermeyer, D.; Kosasih, P.L.; Silva-Benedict, A.; Liu, L.; Rader, C.; Jensen, M.C.; Riddell, S.R. The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol. Res. 2015, 3, 125–135. [Google Scholar] [CrossRef]
  19. Till, B.G.; Jensen, M.C.; Wang, J.; Qian, X.; Gopal, A.K.; Maloney, D.G.; Lindgren, C.G.; Lin, Y.; Pagel, J.M.; Budde, L.E.; et al. CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: Pilot clinical trial results. Blood 2012, 119, 3940–3950. [Google Scholar] [CrossRef]
  20. Al-Hussaini, M.; Rettig, M.P.; Ritchey, J.K.; Karpova, D.; Uy, G.L.; Eissenberg, L.G.; Gao, F.; Eades, W.C.; Bonvini, E.; Chichili, G.R.; et al. Targeting CD123 in acute myeloid leukemia using a T-cell-directed dual-affinity retargeting platform. Blood 2016, 127, 122–131. [Google Scholar] [CrossRef]
  21. Kenderian, S.S.; Ruella, M.; Shestova, O.; Klichinsky, M.; Aikawa, V.; Morrissette, J.J.D.; Scholler, J.; Song, D.; Porter, D.L.; Carroll, M.; et al. CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity against human acute myeloid leukemia. Leukemia 2015, 29, 1637–1647. [Google Scholar] [CrossRef] [PubMed]
  22. Tasian, S.K.; Kenderian, S.S.; Shen, F.; Ruella, M.; Shestova, O.; Kozlowski, M.; Li, Y.; Schrank-Hacker, A.; Morrissette, J.J.D.; Carroll, M.; et al. Optimized depletion of chimeric antigen receptor T cells in murine xenograft models of human acute myeloid leukemia. Blood 2017, 129, 2395–2407. [Google Scholar] [CrossRef] [PubMed]
  23. Foster, J.B.; Choudhari, N.; Perazzelli, J.; Storm, J.; Hofmann, T.J.; Jain, P.; Storm, P.B.; Pardi, N.; Weissman, D.; Waanders, A.J.; et al. Purification of mRNA Encoding Chimeric Antigen Receptor Is Critical for Generation of a Robust T-Cell Response. Hum. Gene Ther. 2019, 30, 168–178. [Google Scholar] [CrossRef] [PubMed]
  24. Yu, S.; Li, A.; Liu, Q.; Li, T.; Yuan, X.; Han, X.; Wu, K. Chimeric antigen receptor T cells: A novel therapy for solid tumors. J. Hematol. Oncol. 2017, 10, 78. [Google Scholar] [CrossRef]
  25. Wagner, S.; Mullins, C.S.; Linnebacher, M. Colorectal cancer vaccines: Tumor-associated antigens vs. neoantigens. World J. Gastroenterol. 2018, 24, 5418–5432. [Google Scholar] [CrossRef]
  26. Zhao, Y.B.; Moon, E.; Carpenito, C.; Paulos, C.M.; Liu, X.J.; Brennan, A.L.; Chew, A.; Carroll, R.G.; Scholler, J.; Levine, B.L.; et al. Multiple Injections of Electroporated Autologous T Cells Expressing a Chimeric Antigen Receptor Mediate Regression of Human Disseminated Tumor. Cancer Res. 2010, 70, 9053–9061. [Google Scholar] [CrossRef]
  27. Lehner, M.; Gotz, G.; Proff, J.; Schaft, N.; Dorrie, J.; Full, F.; Ensser, A.; Muller, Y.A.; Cerwenka, A.; Abken, H.; et al. Redirecting T cells to Ewing’s sarcoma family of tumors by a chimeric NKG2D receptor expressed by lentiviral transduction or mRNA transfection. PLoS ONE 2012, 7, e31210. [Google Scholar] [CrossRef]
  28. Mirzaei, H.R.; Rodriguez, A.; Shepphird, J.; Brown, C.E.; Badie, B. Chimeric Antigen Receptors T Cell Therapy in Solid Tumor: Challenges and Clinical Applications. Front. Immunol. 2017, 8, 1850. [Google Scholar] [CrossRef]
  29. Yoon, S.H.; Lee, J.M.; Cho, H.I.; Kim, E.K.; Kim, H.S.; Park, M.Y.; Kim, T.G. Adoptive immunotherapy using human peripheral blood lymphocytes transferred with RNA encoding Her-2/neu-specific chimeric immune receptor in ovarian cancer xenograft model. Cancer Gene Ther. 2009, 16, 489–497. [Google Scholar] [CrossRef]
  30. Schutsky, K.; Song, D.G.; Lynn, R.; Smith, J.B.; Poussin, M.; Figini, M.; Zhao, Y.B.; Powell, D.J. Rigorous optimization and validation of potent RNA CAR T cell therapy for the treatment of common epithelial cancers expressing folate receptor. Oncotarget 2015, 6, 28911–28928. [Google Scholar] [CrossRef]
  31. Hung, C.F.; Xu, X.Q.; Li, L.H.; Ma, Y.; Jin, Q.; Viley, A.; Allen, C.; Natarajan, P.; Shivakumar, R.; Peshwa, M.V.; et al. Development of Anti-Human Mesothelin-Targeted Chimeric Antigen Receptor Messenger RNA-transfected Peripheral Blood Lymphocytes for Ovarian Cancer Therapy. Hum. Gene Ther. 2018, 29, 614–625. [Google Scholar] [CrossRef] [PubMed]
  32. Ang, W.X.; Li, Z.D.; Chi, Z.X.; Du, S.H.; Chen, C.; Tay, J.C.K.; Toh, H.C.; Connolly, J.E.; Xu, X.H.; Wang, S. Intraperitoneal immunotherapy with T cells stably and transiently expressing anti-EpCAM CAR in xenograft models of peritoneal carcinomatosis. Oncotarget 2017, 8, 13545–13559. [Google Scholar] [CrossRef] [PubMed]
  33. Tchou, J.; Zhao, Y.B.; Levine, B.L.; Zhang, P.J.; Davis, M.M.; Melenhorst, J.J.; Kulikovskaya, I.; Brennan, A.L.; Liu, X.J.; Lacey, S.F.; et al. Safety and Efficacy of Intratumoral Injections of Chimeric Antigen Receptor (CAR) T Cells in Metastatic Breast Cancer. Cancer Immunol. Res. 2017, 5, 1152–1161. [Google Scholar] [CrossRef] [PubMed]
  34. Tang, X.; Zhao, S.; Zhang, Y.; Wang, Y.; Zhang, Z.; Yang, M.; Zhu, Y.; Zhang, G.; Guo, G.; Tong, A.; et al. B7-H3 as a Novel CAR-T Therapeutic Target for Glioblastoma. Mol. Ther. Oncolytics 2019, 14, 279–287. [Google Scholar] [CrossRef]
  35. Singh, N.; Liu, X.; Hulitt, J.; Jiang, S.; June, C.H.; Grupp, S.A.; Barrett, D.M.; Zhao, Y. Nature of tumor control by permanently and transiently modified GD2 chimeric antigen receptor T cells in xenograft models of neuroblastoma. Cancer Immunol. Res. 2014, 2, 1059–1070. [Google Scholar] [CrossRef]
  36. Caruso, H.G.; Torikai, H.; Zhang, L.; Matti, S.; Dai, J.L.; Do, K.A.; Singh, H.; Huls, H.; Lee, D.A.; Champlin, R.E.; et al. Redirecting T-Cell Specificity to EGFR Using mRNA to Self-limit Expression of Chimeric Antigen Receptor. J.Immunother. 2016, 39, 205–217. [Google Scholar] [CrossRef]
  37. Ilieva, K.M.; Cheung, A.; Mele, S.; Chiaruttini, G.; Crescioli, S.; Griffin, M.; Nakamura, M.; Spicer, J.F.; Tsoka, S.; Lacy, K.E.; et al. Chondroitin Sulfate Proteoglycan 4 and Its Potential As an Antibody Immunotherapy Target across Different Tumor Types. Front. Immunol. 2017, 8, 1911. [Google Scholar] [CrossRef]
  38. Krug, C.; Birkholz, K.; Paulus, A.; Schwenkert, M.; Schmidt, P.; Hoffmann, N.; Hombach, A.; Fey, G.; Abken, H.; Schuler, G.; et al. Stability and activity of MCSP-specific chimeric antigen receptors (CARs) depend on the scFv antigen-binding domain and the protein backbone. Cancer Immunol. Immun. 2015, 64, 1623–1635. [Google Scholar] [CrossRef]
  39. Harrer, D.C.; Simon, B.; Fujii, S.I.; Shimizu, K.; Uslu, U.; Schuler, G.; Gerer, K.F.; Hoyer, S.; Dorrie, J.; Schaft, N. RNA-transfection of gamma/delta T cells with a chimeric antigen receptor or an alpha/beta T-cell receptor: A safer alternative to genetically engineered alpha/beta T cells for the immunotherapy of melanoma. BMC Cancer 2017, 17, 551. [Google Scholar] [CrossRef]
  40. Uslu, U.; Schuler, G.; Dorrie, J.; Schaft, N. Combining a chimeric antigen receptor and a conventional T-cell receptor to generate T cells expressing two additional receptors (TETARs) for a multi-hit immunotherapy of melanoma. Exp. Dermatol. 2016, 25, 872–879. [Google Scholar] [CrossRef]
  41. Simon, B.; Harrer, D.C.; Schuler-Thurner, B.; Schuler, G.; Uslu, U. Arming T Cells with a gp100-Specific TCR and a CSPG4-Specific CAR Using Combined DNA- and RNA-Based Receptor Transfer. Cancers 2019, 11, 696. [Google Scholar] [CrossRef] [PubMed]
  42. Wiesinger, M.; Marz, J.; Kummer, M.; Schuler, G.; Dorrie, J.; Schuler-Thurner, B.; Schaft, N. Clinical-Scale Production of CAR-T Cells for the Treatment of Melanoma Patients by mRNA Transfection of a CSPG4-Specific CAR under Full GMP Compliance. Cancers 2019, 11, 1198. [Google Scholar] [CrossRef] [PubMed]
  43. Simon, B.; Wiesinger, M.; Marz, J.; Wistuba-Hamprecht, K.; Weide, B.; Schuler-Thurner, B.; Schuler, G.; Dorrie, J.; Uslu, U. The Generation of CAR-Transfected Natural Killer T Cells for the Immunotherapy of Melanoma. Int. J. Mol. Sci. 2018, 19, 2365. [Google Scholar] [CrossRef] [PubMed]
  44. Walseng, E.; Koksal, H.; Sektioglu, I.M.; Fane, A.; Skorstad, G.; Kvalheim, G.; Gaudernack, G.; Inderberg, E.M.; Walchli, S. A TCR-based Chimeric Antigen Receptor. Sci. Rep. 2017, 7, 10713. [Google Scholar] [CrossRef] [PubMed]
  45. Inoo, K.; Inagaki, R.; Fujiwara, K.; Sasawatari, S.; Kamigaki, T.; Nakagawa, S.; Okada, N. Immunological quality and performance of tumor vessel-targeting CAR-T cells prepared by mRNA-EP for clinical research. Mol. Ther. Oncolytics 2016, 3. [Google Scholar] [CrossRef] [PubMed]
  46. Cummins, K.D.; Frey, N.; Nelson, A.M.; Schmidt, A.; Luger, S.; Isaacs, R.E.; Lacey, S.F.; Hexner, E.; Melenhorst, J.J.; June, C.H.; et al. Treating Relapsed/Refractory (RR) AML with Biodegradable Anti-CD123 CAR Modified T Cells. Blood 2017, 130, 1359. [Google Scholar]
  47. Svoboda, J.; Rheingold, S.R.; Gill, S.I.; Grupp, S.A.; Lacey, S.F.; Kulikovskaya, I.; Suhoski, M.M.; Melenhorst, J.J.; Loudon, B.; Mato, A.R.; et al. Nonviral RNA chimeric antigen receptor-modified T cells in patients with Hodgkin lymphoma. Blood 2018, 132, 1022–1026. [Google Scholar] [CrossRef]
  48. Maus, M.V.; Haas, A.R.; Beatty, G.L.; Albelda, S.M.; Levine, B.L.; Liu, X.J.; Zhao, Y.B.; Kalos, M.; June, C.H. T Cells Expressing Chimeric Antigen Receptors Can Cause Anaphylaxis in Humans. Cancer Immunol. Res. 2013, 1, 26–31. [Google Scholar] [CrossRef]
  49. Caruso, H.G.; Heimberger, A.B.; Cooper, L.J.N. Steering CAR T cells to distinguish friend from foe. Oncoimmunology 2019, 8, e1271857. [Google Scholar] [CrossRef]
  50. Beatty, G.L.; Haas, A.R.; Maus, M.V.; Torigian, D.A.; Soulen, M.C.; Plesa, G.; Chew, A.; Zhao, Y.; Levine, B.L.; Albelda, S.M.; et al. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol. Res. 2014, 2, 112–120. [Google Scholar] [CrossRef]
  51. Beatty, G.L.; O’Hara, M.H.; Lacey, S.F.; Torigian, D.A.; Nazimuddin, F.; Chen, F.; Kulikovskaya, I.M.; Soulen, M.C.; McGarvey, M.; Nelson, A.M.; et al. Activity of Mesothelin-Specific Chimeric Antigen Receptor T Cells Against Pancreatic Carcinoma Metastases in a Phase 1 Trial. Gastroenterology 2018, 155, 29–32. [Google Scholar] [CrossRef] [PubMed]

Article Metrics

Citations

Article Access Statistics

Multiple requests from the same IP address are counted as one view.