T-Cell Therapeutics Targeting Human Parainfluenza Virus 3 Are Broadly Epitope Specific and Are Cross Reactive With Human Parainfluenza Virus 1

Katherine M Harris, Sarah E Horn, Melanie L Grant, Haili Lang, Gelina Sani, Mariah A Jensen-Wachspress, Vaishnavi V Kankate, Anushree Datar, Christopher A Lazarski, Catherine M Bollard, Michael D Keller, Katherine M Harris, Sarah E Horn, Melanie L Grant, Haili Lang, Gelina Sani, Mariah A Jensen-Wachspress, Vaishnavi V Kankate, Anushree Datar, Christopher A Lazarski, Catherine M Bollard, Michael D Keller

Abstract

Human Parainfluenza Virus-3 (HPIV3) causes severe respiratory illness in immunocompromised patients and lacks approved anti-viral therapies. A phase I study of adoptively transferred virus-specific T-cells (VSTs) targeting HPIV3 following bone marrow transplantation is underway (NCT03180216). We sought to identify immunodominant epitopes within HPIV3 Matrix protein and their cross-reactivity against related viral proteins. VSTs were generated from peripheral blood of healthy donors by ex-vivo expansion after stimulation with a 15-mer peptide library encompassing HPIV3 matrix protein. Epitope mapping was performed using IFN-γ ELIspot with combinatorial peptide pools. Flow cytometry was used to characterize products with intracellular cytokine staining. In 10 VST products tested, we discovered 12 novel immunodominant epitopes. All products recognized an epitope at the C-terminus. On IFN-γ ELISpot, individual peptides eliciting activity demonstrated mean IFN-γ spot forming units per well (SFU)/1x105 cells of 115.5 (range 24.5-247.5). VST products were polyfunctional, releasing IFN-γ and TNF-α in response to identified epitopes, which were primarily HLA Class II restricted. Peptides from Human Parainfluenza Virus-1 corresponding to the HPIV3 epitopes showed cross-reactivity for HPIV1 in 11 of 12 tested epitopes (mean cross reactivity index: 1.19). Characterization of HPIV3 epitopes may enable development of third-party VSTs to treat immune suppressed patients with HPIV infection.

Keywords: T-cell; anti-viral; bone marrow transplant; immunotherapy; parainfluenza.

Copyright © 2020 Harris, Horn, Grant, Lang, Sani, Jensen-Wachspress, Kankate, Datar, Lazarski, Bollard and Keller.

Figures

Figure 1
Figure 1
(A) Virus-specific T-cells (VST) Product Phenotyping. Phenotyping data of the hexaviral-specific T cell products specific for cytomegalovirus (CMV), Epstein Barr virus (EBV), AdV, HHV6, BKV, and HPIV3 (product #s 1-4, 6, 8-10) and monoviral-specific T cell products specific only for HPIV3 (product #s 5, 7) was obtained using flow cytometry. Phenotypes, including memory differentiation status panel, are reported as % of total CD3+ cells. Product #5 was unavailable for memory/differentiation phenotyping. Definitions of phenotypes are as follows: Naïve: CD45RA+CD45RO-CCR7+CD62L+CD95- Stem Cell Memory: CD45RA+CD45RO-CCR7+CD62L+CD95+ Central Memory: CD45RA-CD45RO+CCR7+CD62L+ Effector Memory: CD45RA-CD45RO+CCR7-CD62L- Terminal Effector Memory: CD45RA-CD45RO-CCR7-CD62L- (B) Overall HPIV3 Matrix specificity by product. All 10 viral-specific T cell products were stimulated with HPIV3 pepmix. Response was measured as spot forming units per well (SFU/1x105 cells) by anti-IFN-γ ELISpot assay. Unstimulated T-cells (CTL only) and stimulation with actin pepmix (irrelevant antigen) were used as negative controls.
Figure 2
Figure 2
Peptide Pool specificity by product. Virus-specific T-cells (VSTs) were stimulated with combinatorial HPIV3 peptide pools. Response was measured as spots per well (SFU/1x105 cells) by anti-IFN-γ ELISpot assay. Unstimulated T-cells (CTL only) and stimulation with actin (irrelevant antigen) were used as negative controls.
Figure 3
Figure 3
(A) Cross reactivity between HPIV3 and HPIV1. Virus-specific T-cells (VSTs) were stimulated with individual HPIV3 peptides and corresponding HPIV1 peptides. Response was measured as spots per well (SFU/1x105 cells) by anti-IFN-γ ELISpot assay, and mean responses were calculated among responding products. Black bars represent HPIV3 peptides and gray bars represent HPIV1 peptides. (B) CD4+ T Cell Responses against HPIV3 and HPIV1 peptides. Intracellular flow cytometry was performed on VSTs following stimulation with Actin, SEB, HPIV3 or HPIV1 peptides and anti-CD28/CD49. After 2 h stimulation with peptides, Brefeldin A was added for an additional 4 h following which cells were labeled with dead cell exclusion dye, Fc Receptor block and antibodies against CD3, CD4, CD8, TNF-α, IFN-γ, and CD95. Following fixation and permeabilization cells were labeled intracellularly with antibodies targeting TNF-α, IFN-γ, Percentage of CD4+ and CD8+ T-cells producing both IFNγ+ and/or TNFα+ were measured.
Figure 4
Figure 4
(A) HLA Restriction Mapping. Two phytohemagglutinin (PHA) blast lines (each partially matched to a Virus-specific T-cell (VST) product at HLA-DR*15:01 and HLA-DQ*03:01, respectively) were pulsed with HPIV3 peptides 82, 83, and 84 for 60 min, triple washed, and then presented to VSTs. T cell activity was detected using anti-IFN-γ ELISpot. (B) HPIV3 Matrix diagram with epitopes and C-terminal domain. HPIV3 Matrix protein contains 353 amino acids. Diamonds represent the novel epitopes identified during mapping. The oligomerization domain at the C-terminus contains 7 out of 12 epitopes, including the three most immunodominant peptides, 82–84. The black arrow denotes the L302 residue, which is a target of ubiquitination during virion production.

References

    1. Boeckh M, Boeckh M, Boeckh M, Boeckh M, Boeckh M, Boeckh M. The Challenge of Respiratory Virus Infections in Hematopoietic Cell Transplant Recipients. Br J Haematol (2008) 143(4):455–67. 10.1111/j.1365-2141.2008.07295.x
    1. Falsey A. Current Management of parainfluenza pneumonitis in immunocompromised patients: a review. Infect Drug Resist (2012) 5:121–7. 10.2147/IDR.S25874
    1. Srinivasan A, Wang C, Yang J, Shenep J, Leung W, Hayden R, et al. Symptomatic Parainfluenza Virus Infections in Children Undergoing Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant (2011) 17:1520–7. 10.1016/j.bbmt.2011.03.001
    1. Nichols WG, Corey L, Gooley T, Davis C, Boeckh M. Parainfluenza virus infections after hematopoietic stem cell transplantation: risk factors, response to antiviral therapy, and effect on transplant outcome. Blood (2001) 98(3):573–8. 10.1182/blood.V98.3.573
    1. Walter EA, Greenberg PD, Gilbert MJ, Finch RJ, Watanabe KS, Thomas ED, Riddell SR, et al. Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T cell clones from the donor. N Engl J Med (1995) 333(16):1038–44. 10.1056/NEJM199510193331603
    1. Peggs KS, Verfuerth A, Pizzey A, Khan N, Guiver N, Moss PS, et al. Adoptive cellular therapy for early cytomegalovirus infection after allogeneic stem-cell transplantation with virus-specific T cell lines. Lancet (2003) 362(9393):1375–7. 10.1016/S0140-6736(03)14634-X
    1. Leen AM, Christin A, Myers GD, Liu H, Cruz CR, Hanley PJ, et al. Cytotoxic T lymphocyte therapy with donor T cells prevents and treats adenovirus and Epstein-Barr virus infections after haploidentical and matched unrelated stem cell transplantation. Blood (2009) 114(19):4283–92. 10.1182/blood-2009-07-232454
    1. Leen AM, Myers GD, Sili U, Hulu MH, Weiss H, Leung KS, et al. Monoculture-derived T lymphocytes specific for multiple viruses expand and produce clinically relevant effects in immunocompromised individuals. Nat Med (2006) 12(10):1160–6. 10.1038/nm1475
    1. Papadopoulou A, Gerdemann U, Katari U, Tzannou I, Liu H, Martinez C, et al. Activity of broad-spectrum T cells as treatment for AdV, EBV, CMV, BKV and HHV6 infections after HSCT. Sci Transl Med (2014) 6(242):242–83. 10.1126/scitranslmed.3008825
    1. McLaughlin L, Lang H, Williams E, Wright KE, Powell A, Cruz CR, et al. Human Parainfluenza Virus-3 can be Targeted by Rapidly ex vivo Expanded T-Lymphocytes. Cytotherapy (2016) 18(12):1515–24. 10.1016/j.jcyt.2016.08.010
    1. Leen AM, Bollard CM, Mendizabal AM, Shpall EJ, Szabolcs P, Antin JH, et al. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood (2013) 121:5113–23. 10.1182/blood-2013-02-486324
    1. Tzannou I, Papadopoulou A, Naik S, Leung K, Martinez CA, Ramos CA, et al. Off-the-shelf virus-specific T cells to treat BK virus, human herpesvirus 6, cytomegalovirus, Epstein-Barr virus, and adenovirus infections after allogeneic hematopoietic stem-cell transplantation. J Clin Oncol (2017) 35:3547–57. 10.1200/JCO.2017.73.0655
    1. Gerdemann, Keirnan JM, Katari UL, Yanagisawa R, Christin AS, Huye LE, et al. Rapidly generated multivirus-specific cytotoxic T l ymphocytes for the prophylaxis and treatment of viral infections. Mol Ther (2012) 20(8):1622–32. 10.1038/mt.2012.130
    1. Stokes A, Tierney EL, Murphy BR, Hall SL. The complete nucleotide sequence of the JS strain of human parainfluenza virus type 3: comparison with the Wash/47885/57 prototype strain. Virus Res (1992) 25(1-2):91–103. 10.1016/0168-1702(92)90102-F
    1. Withers E, Blyth E, Clancy LE, Yong A, Fraser C, Burgess J, et al. Long-term control of recurrent or refractory viral infections after allogeneic HSCT with third-party virus-specific T cells. Blood Adv (2017) 1(24):2193–205. 10.1182/cloodadvances.2017010223
    1. Tzannou I, Watanabe A, Naik S, Daum R, Kuvalekar M, Leung KS, et al. Mini" bank of only 8 donors supplies CMV-directed T cells to diverse recipients. Blood Adv (2019) 3(17):2571–80. 10.1182/bloodadvances.2019000371
    1. Ding B, Zhang L, Li Z, Zhong Y, Tang Q, Quin Y, et al. The Matrix Protein of Human Parainfluenza Virus Type 3 Induces Mitophagy that Suppresses Interferon Responses. Cell Host Microbe (2017) 21:538–47. 10.1016/j.chom.2017.03.004
    1. Henrickson K. Parainfluenza viruses. Clin Microbiol Rev (2003) 16(2):242–64. 10.1128/CMR.16.2.242-264.2003
    1. Englund JA, Karron RA, Cunningham CK, LaRussa P, Melvin A, Yogev R, et al. Safety and infectivity of two doses of live attenuated recombinant cold-passaged human parainfluenza type 3 virus vaccine rHPIV3cp45 in HPIV3-seronegative young children. Vaccine (2013) 31(48):5706–12. 10.1016/j.vaccine.2013.09.046
    1. Leen A, Christin A, Khalil M, Weiss H, Gee AP, Brenner MK, et al. Identification of Hexon-Specific CD4 and CD8 T-Cell Epitopes for Vaccine and Immunotherapy. J Virol (2008) 82(1):546–54. 10.1128/JVI.01689-07
    1. Feuchtinger T, Richard C, Joachim S, Scheible MH, Schumm M, Hamprecht K, et al. Clinical grade generation of hexon-specific T cells for adoptive T-cell transfer as a treatment of adenovirus infection after allogeneic stem cell transplantation. J Immunother (2008) 31(2):199–206. 10.1097/CJI.0b013e31815ef862
    1. Olive M, Eisenlohr L, Flomenberg N, Hsu H, Flomenberg P, et al. The adenovirus capsid protein hexon contains a highly conserved human CD4+ T-cell epitope. Hum Gene Ther (2002) 13(10):1167–78. 10.1089/104303402320138952
    1. Prockop S, Doubrovina E, Suser S, Heller G, Barker J, Dahi P. Off-the-shelf EBV-specific T cell immunotherapy for rituximab-refractory EBV-associated lymphoma following transplantation. J Clin Invest (2020) 130(2):733–47. 10.1172/JCI121127
    1. Dave, Allan JE, Sloblod KS, Smith FS, Ryan KW, Takimoto T, et al. Viral Cross-Reactivity and Antigenic Determinants Recognized by Human Parainfluenza Virus Type 1-Specific Cytotoxic T-Cells. Virology (1994) 199(2):376–83. 10.1006/viro.1994.1135
    1. Ziola B, Smith RH. T Cell Cross-Reactivity Among Viruses of the Paramyxoviridae. Viral Immunol (1987) 1(2):111. 10.1089/vim.1987.1.111

Source: PubMed

3
Se inscrever