1
|
Babutzka S, Gehrke M, Papadopoulou A, Diedrichs-Möhring M, Giannaki M, Hennis L, Föhr B, Kooyman C, Osterman A, Yannaki E, Wildner G, Ammer H, Michalakis S. A novel platform for engineered AAV-based vaccines. Mol Ther Methods Clin Dev 2025; 33:101418. [PMID: 40008090 PMCID: PMC11850754 DOI: 10.1016/j.omtm.2025.101418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 01/20/2025] [Indexed: 02/27/2025]
Abstract
Engineering of adeno-associated virus (AAV) capsids allowed for the development of gene therapy vectors with improved tropism and enhanced transduction efficiency. Capsid engineering can also be used to adapt the AAV technology for applications outside gene therapy. Here, we investigated modified AAV capsids as scaffolds for the presentation of large immunogenic antigens to elicit a strong and specific immune response against pathogens. Using SARS-CoV-2 as a model pathogen, we introduced ∼200 amino acids of the SARS-CoV-2 receptor-binding domain (RBD) into a surface-exposed variable loop region of AAV2 and AAV9, resulting in AAV2.RBD and AAV9.RBD capsids (AAV.RBDs). This engineering endowed AAV.RBDs with SARS-CoV-2-like properties, such as angiotensin-converting enzyme 2 receptor affinity. In line with this, AAV.RBDs were neutralized by sera from human donors vaccinated against SARS-CoV-2. When administered subcutaneously to rabbits, AAV.RBDs elicited a strong humoral response against SARS-CoV-2 RBD. Moreover, the AAV.RBDs were able to trigger RBD-specific cellular immune responses in peripheral human lymphocytes. In conclusion, this novel AAV-based next-generation vaccine platform allows for the presentation of large antigenic sequences to elicit strong and specific immune responses. This versatile vaccine technology could be explored in the context of diseases where conventional immunization approaches have been unsuccessful.
Collapse
Affiliation(s)
- Sabrina Babutzka
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Miranda Gehrke
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Anastasia Papadopoulou
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, 570 10 Thessaloniki, Greece
| | | | - Maria Giannaki
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, 570 10 Thessaloniki, Greece
| | - Lena Hennis
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Bastian Föhr
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Cale Kooyman
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Andreas Osterman
- Max Von Pettenkofer Institute and Gene Center, Virology, LMU Munich, 80336 Munich, Germany
| | - Evangelia Yannaki
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, 570 10 Thessaloniki, Greece
- Department of Medicine, University of Washington, Seattle, WA 91895, USA
| | - Gerhild Wildner
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Hermann Ammer
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, 80539 Munich, Germany
| | - Stylianos Michalakis
- Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| |
Collapse
|
2
|
Im KI, Kim N, Lee J, Oh UH, Lee HW, Lee DG, Min GJ, Lee R, Lee J, Kim S, Cho SG. SARS-CoV-2-Specific T-Cell as a Potent Therapeutic Strategy against Immune Evasion of Emerging COVID-19 Variants. Int J Mol Sci 2024; 25:10512. [PMID: 39408840 PMCID: PMC11477143 DOI: 10.3390/ijms251910512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Despite advances in vaccination and therapies for coronavirus disease, challenges remain due to reduced antibody longevity and the emergence of virulent variants like Omicron (BA.1) and its subvariants (BA.1.1, BA.2, BA.3, and BA.5). This study explored the potential of adoptive immunotherapy and harnessing the protective abilities using virus-specific T cells (VSTs). Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) VSTs were generated by stimulating donor-derived peripheral blood mononuclear cells with spike, nucleocapsid, and membrane protein peptide mixtures. Phenotypic characterization, including T-cell receptor (TCR) vβ and pentamer analyses, was performed on the ex vivo-expanded cells. We infected human leukocyte antigen (HLA)-partially matched human Calu-3 cells with various authentic SARS-CoV-2 strains in a Biosafety Level 3 facility and co-cultured them with VSTs. VSTs exhibited a diverse TCR vβ repertoire, confirming their ability to target a broad range of SARS-CoV-2 antigens from both the ancestral and mutant strains, including Omicron BA.1 and BA.5. These ex vivo-expanded cells exhibited robust cytotoxicity and low alloreactivity against HLA-partially matched SARS-CoV-2-infected cells. Their cytotoxic effects were consistent across variants, targeting conserved spike and nucleocapsid epitopes. Our findings suggest that third-party partial HLA-matching VSTs could counter immune-escape mechanisms posed by emerging variants of concern.
Collapse
Affiliation(s)
- Keon-Il Im
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Nayoun Kim
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Junseok Lee
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
| | - Ui-Hyeon Oh
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Hye-Won Lee
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Dong-Gun Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.-G.L.); (R.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Gi-June Min
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Department of Hematology, Seoul St. Mary’s Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Raeseok Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.-G.L.); (R.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jinah Lee
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea; (J.L.); (S.K.)
| | - Seungtaek Kim
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea; (J.L.); (S.K.)
| | - Seok-Goo Cho
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
- Department of Hematology, Seoul St. Mary’s Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
3
|
Koukoulias K, Papayanni PG, Jones J, Kuvalekar M, Watanabe A, Velazquez Y, Gilmore S, Papadopoulou A, Leen AM, Vasileiou S. Assessment of the cytolytic potential of a multivirus-targeted T cell therapy using a vital dye-based, flow cytometric assay. Front Immunol 2023; 14:1299512. [PMID: 38187380 PMCID: PMC10766817 DOI: 10.3389/fimmu.2023.1299512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024] Open
Abstract
Reliable and sensitive characterization assays are important determinants of the successful clinical translation of immunotherapies. For the assessment of cytolytic potential, the chromium 51 (51Cr) release assay has long been considered the gold standard for testing effector cells. However, attaining the approvals to access and use radioactive isotopes is becoming increasingly complex, while technical aspects [i.e. sensitivity, short (4-6 hours) assay duration] may lead to suboptimal performance. This has been the case with our ex vivo expanded, polyclonal (CD4+ and CD8+) multivirus-specific T cell (multiVST) lines, which recognize 5 difficult-to-treat viruses [Adenovirus (AdV), BK virus (BKV), cytomegalovirus (CMV), Epstein Barr virus (EBV), and human herpes virus 6 (HHV6)] and when administered to allogeneic hematopoietic stem cell (HCT) or solid organ transplant (SOT) recipients have been associated with clinical benefit. However, despite mediating potent antiviral effects in vivo, capturing in vitro cytotoxic potential has proven difficult in a traditional 51Cr release assay. Now, in addition to cytotoxicity surrogates, including CD107a and Granzyme B, we report on an alternative, vital dye -based, flow cytometric platform in which superior sensitivity and prolonged effector:target co-culture duration enabled the reliable detection of both CD4- and CD8-mediated in vitro cytolytic activity against viral targets without non-specific effects.
Collapse
Affiliation(s)
- Kiriakos Koukoulias
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Penelope G. Papayanni
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Julia Jones
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Manik Kuvalekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Ayumi Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Yovana Velazquez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | | | - Anastasia Papadopoulou
- Hematology Department- Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, Thessaloniki, Greece
| | - Ann M. Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Spyridoula Vasileiou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
4
|
Piñana JL, Pérez A, Chorão P, Guerreiro M, García-Cadenas I, Solano C, Martino R, Navarro D. Respiratory virus infections after allogeneic stem cell transplantation: Current understanding, knowledge gaps, and recent advances. Transpl Infect Dis 2023; 25 Suppl 1:e14117. [PMID: 37585370 DOI: 10.1111/tid.14117] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023]
Abstract
Before the COVID-19 pandemic, common community-acquired seasonal respiratory viruses (CARVs) were a significant threat to the health and well-being of allogeneic hematopoietic cell transplant (allo-HCT) recipients, often resulting in severe illness and even death. The pandemic has further highlighted the significant risk that immunosuppressed patients, including allo-HCT recipients, face when infected with SARS-CoV-2. As preventive transmission measures are relaxed and CARVs circulate again among the community, including in allo-HSCT recipients, it is crucial to understand the current state of knowledge, gaps, and recent advances regarding CARV infection in allo-HCT recipients. Urgent research is needed to identify seasonal respiratory viruses as potential drivers for future pandemics.
Collapse
Affiliation(s)
- Jose L Piñana
- Hematology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Fundación INCLIVA, Instituto de Investigación Sanitaria Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Ariadna Pérez
- Hematology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Fundación INCLIVA, Instituto de Investigación Sanitaria Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Pedro Chorão
- Hematology Division, Hospital universitario y politécnico La Fe, Valencia, Spain
- Instituto de Investigación La Fe, Hospital Universitário y Politécncio La Fe, Valencia, Spain
| | - Manuel Guerreiro
- Hematology Division, Hospital universitario y politécnico La Fe, Valencia, Spain
- Instituto de Investigación La Fe, Hospital Universitário y Politécncio La Fe, Valencia, Spain
| | | | - Carlos Solano
- Hematology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Fundación INCLIVA, Instituto de Investigación Sanitaria Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Rodrigo Martino
- Hematology Division, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - David Navarro
- Microbiology department, Hospital Clinico Universitario de Valencia, Spain
- Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
5
|
Fylaktou A, Stai S, Kasimatis E, Xochelli A, Nikolaidou V, Papadopoulou A, Myserlis G, Lioulios G, Asouchidou D, Giannaki M, Yannaki E, Tsoulfas G, Papagianni A, Stangou M. Humoral and Cellular Immunity Are Significantly Affected in Renal Transplant Recipients, following Vaccination with BNT162b2. Vaccines (Basel) 2023; 11:1670. [PMID: 38006002 PMCID: PMC10674678 DOI: 10.3390/vaccines11111670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Renal transplant recipients (RTRs) tend to mount weaker immune responses to vaccinations, including vaccines against the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). METHODS Humoral immunity was assessed using anti-receptor binding domain (RBD) and neutralizing antibodies (NAb) serum levels measured by ELISA, and cellular immunity was assessed using T-, B-, NK, natural killer-like T (NKT)-cell subpopulations, and monocytes measured by flow cytometry, and also specific T-cell immunity, at predefined time points after BNT162b2 vaccination, in 57 adult RTRs. RESULTS Administration of three booster doses was necessary to achieve anti-RBD and NAb protective levels in almost all patients (92.98%). Ab production, at several time points, was positively correlated with the corresponding renal function and inversely correlated with hemodialysis vintage (HDV) and treatment with mycophenolic acid (MPA). A gradual rise in several cell subpopulations, including total lymphocytes (p = 0.026), memory B cells (p = 0.028), activated CD4 (p = 0.005), and CD8 cells (p = 0.001), was observed even after the third vaccination dose, while a significant reduction in CD3+PD1+ (p = 0.002), NKT (p = 0.011), and activated NKT cells (p = 0.034) was noted during the same time interval. Moreover, SARS-CoV-2-specific T-cells were present in 41% of the patients who were unable to develop Nabs, and their positivity rates four months after the second dose were in inverse correlation with monocytes (p = 0.045) and NKT cells (p = 0.01). CONCLUSIONS SARS-CoV-2-specific T-cell responses preceded the humoral ones, while two booster doses were needed for this group of immunocompromised patients to mount a protective immune response.
Collapse
Affiliation(s)
- Asimina Fylaktou
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece; (A.F.); (A.X.); (V.N.); (D.A.)
| | - Stamatia Stai
- Department of Nephrology, Hippokration Hospital, 54642 Thessaloniki, Greece; (S.S.); (E.K.); (G.L.); (A.P.)
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Efstratios Kasimatis
- Department of Nephrology, Hippokration Hospital, 54642 Thessaloniki, Greece; (S.S.); (E.K.); (G.L.); (A.P.)
| | - Aliki Xochelli
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece; (A.F.); (A.X.); (V.N.); (D.A.)
| | - Vasiliki Nikolaidou
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece; (A.F.); (A.X.); (V.N.); (D.A.)
| | - Anastasia Papadopoulou
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, 57010 Thessaloniki, Greece; (A.P.); (M.G.); (E.Y.)
| | - Grigorios Myserlis
- Department of Transplant Surgery, Hippokration Hospital, 54642 Thessaloniki, Greece;
| | - Georgios Lioulios
- Department of Nephrology, Hippokration Hospital, 54642 Thessaloniki, Greece; (S.S.); (E.K.); (G.L.); (A.P.)
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Despoina Asouchidou
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece; (A.F.); (A.X.); (V.N.); (D.A.)
| | - Maria Giannaki
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, 57010 Thessaloniki, Greece; (A.P.); (M.G.); (E.Y.)
| | - Evangelia Yannaki
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, 57010 Thessaloniki, Greece; (A.P.); (M.G.); (E.Y.)
| | - Georgios Tsoulfas
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Department of Transplant Surgery, Hippokration Hospital, 54642 Thessaloniki, Greece;
| | - Aikaterini Papagianni
- Department of Nephrology, Hippokration Hospital, 54642 Thessaloniki, Greece; (S.S.); (E.K.); (G.L.); (A.P.)
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Maria Stangou
- Department of Nephrology, Hippokration Hospital, 54642 Thessaloniki, Greece; (S.S.); (E.K.); (G.L.); (A.P.)
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| |
Collapse
|
6
|
Papadopoulou A, Karavalakis G, Papadopoulou E, Xochelli A, Bousiou Z, Vogiatzoglou A, Papayanni PG, Georgakopoulou A, Giannaki M, Stavridou F, Vallianou I, Kammenou M, Varsamoudi E, Papadimitriou V, Giannaki C, Sileli M, Stergiouda Z, Stefanou G, Kourlaba G, Gounelas G, Triantafyllidou M, Siotou E, Karaglani A, Zotou E, Chatzika G, Boukla A, Papalexandri A, Koutra MG, Apostolou D, Pitsiou G, Morfesis P, Doumas M, Karampatakis T, Kapravelos N, Bitzani M, Theodorakopoulou M, Serasli E, Georgolopoulos G, Sakellari I, Fylaktou A, Tryfon S, Anagnostopoulos A, Yannaki E. SARS-CoV-2-specific T cell therapy for severe COVID-19: a randomized phase 1/2 trial. Nat Med 2023; 29:2019-2029. [PMID: 37460756 DOI: 10.1038/s41591-023-02480-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 06/28/2023] [Indexed: 07/22/2023]
Abstract
Despite advances, few therapeutics have shown efficacy in severe coronavirus disease 2019 (COVID-19). In a different context, virus-specific T cells have proven safe and effective. We conducted a randomized (2:1), open-label, phase 1/2 trial to evaluate the safety and efficacy of off-the-shelf, partially human leukocyte antigen (HLA)-matched, convalescent donor-derived severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cells (CoV-2-STs) in combination with standard of care (SoC) in patients with severe COVID-19 compared to SoC during Delta variant predominance. After a dose-escalated phase 1 safety study, 90 participants were randomized to receive CoV-2-ST+SoC (n = 60) or SoC only (n = 30). The co-primary objectives of the study were the composite of time to recovery and 30-d recovery rate and the in vivo expansion of CoV-2-STs in patients receiving CoV-2-ST+SoC over SoC. The key secondary objective was survival on day 60. CoV-2-ST+SoC treatment was safe and well tolerated. The study met the primary composite endpoint (CoV-2-ST+SoC versus SoC: recovery rate 65% versus 38%, P = 0.017; median recovery time 11 d versus not reached, P = 0.052, respectively; rate ratio for recovery 1.71 (95% confidence interval 1.03-2.83, P = 0.036)) and the co-primary objective of significant CoV-2-ST expansion compared to SοC (CoV-2-ST+SoC versus SoC, P = 0.047). Overall, in hospitalized patients with severe COVID-19, adoptive immunotherapy with CoV-2-STs was feasible and safe. Larger trials are needed to strengthen the preliminary evidence of clinical benefit in severe COVID-19. EudraCT identifier: 2021-001022-22 .
Collapse
Affiliation(s)
- Anastasia Papadopoulou
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - George Karavalakis
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Efthymia Papadopoulou
- Department of Respiratory Medicine, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Aliki Xochelli
- Department of Immunology, National Peripheral Histocompatibility Center, Hippokration General Hospital, Thessaloniki, Greece
| | - Zoi Bousiou
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | | | - Penelope-Georgia Papayanni
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aphrodite Georgakopoulou
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Giannaki
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Fani Stavridou
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Ioanna Vallianou
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Maria Kammenou
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Evangelia Varsamoudi
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Vasiliki Papadimitriou
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Chrysavgi Giannaki
- 'A' Intensive Care Unit, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Maria Sileli
- 'B' Intensive Care Unit, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Zoi Stergiouda
- Department of Anesthesiology, George Papanikolaou Hospital, Thessaloniki, Greece
| | | | - Georgia Kourlaba
- Department of Nursing, University of Peloponnese, Tripolis, Greece
| | | | - Maria Triantafyllidou
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Eleni Siotou
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | | | - Eleni Zotou
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgia Chatzika
- Department of Immunology, National Peripheral Histocompatibility Center, Hippokration General Hospital, Thessaloniki, Greece
| | - Anna Boukla
- Department of Immunology, National Peripheral Histocompatibility Center, Hippokration General Hospital, Thessaloniki, Greece
| | - Apostolia Papalexandri
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Maria-Georgia Koutra
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Dimitra Apostolou
- Department of Respiratory Failure, George Papanikolaou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgia Pitsiou
- Department of Respiratory Failure, George Papanikolaou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Petros Morfesis
- 1st Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Michalis Doumas
- 2nd Propedeutic Department of Internal Medicine, Hippokrateio Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | - Militsa Bitzani
- 'A' Intensive Care Unit, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Maria Theodorakopoulou
- National and Kapodistrian University of Athens, Evaggelismos General Hospital, Athens, Greece
| | - Eva Serasli
- Department of Respiratory Medicine, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Grigorios Georgolopoulos
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Ioanna Sakellari
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Asimina Fylaktou
- Department of Immunology, National Peripheral Histocompatibility Center, Hippokration General Hospital, Thessaloniki, Greece
| | - Stavros Tryfon
- Department of Respiratory Medicine, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Achilles Anagnostopoulos
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Evangelia Yannaki
- Hematopoietic Cell Transplantation Unit, Department of Hematology Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece.
- Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
7
|
Vasileiou S, Hill L, Kuvalekar M, Workineh AG, Watanabe A, Velazquez Y, Lulla S, Mooney K, Lapteva N, Grilley BJ, Heslop HE, Rooney CM, Brenner MK, Eagar TN, Carrum G, Grimes KA, Leen AM, Lulla P. Allogeneic, off-the-shelf, SARS-CoV-2-specific T cells (ALVR109) for the treatment of COVID-19 in high-risk patients. Haematologica 2023; 108:1840-1850. [PMID: 36373249 PMCID: PMC10316279 DOI: 10.3324/haematol.2022.281946] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/31/2022] [Indexed: 07/22/2023] Open
Abstract
Defects in T-cell immunity to SARS-CoV-2 have been linked to an increased risk of severe COVID-19 (even after vaccination), persistent viral shedding and the emergence of more virulent viral variants. To address this T-cell deficit, we sought to prepare and cryopreserve banks of virus-specific T cells, which would be available as a partially HLA-matched, off-the-shelf product for immediate therapeutic use. By interrogating the peripheral blood of healthy convalescent donors, we identified immunodominant and protective T-cell target antigens, and generated and characterized polyclonal virus-specific T-cell lines with activity against multiple clinically important SARS-CoV-2 variants (including 'delta' and 'omicron'). The feasibility of making and safely utilizing such virus-specific T cells clinically was assessed by administering partially HLA-matched, third-party, cryopreserved SARS-CoV-2-specific T cells (ALVR109) in combination with other antiviral agents to four individuals who were hospitalized with COVID-19. This study establishes the feasibility of preparing and delivering off-the-shelf, SARS-CoV-2-directed, virus-specific T cells to patients with COVID-19 and supports the clinical use of these products outside of the profoundly immune compromised setting (ClinicalTrials.gov number, NCT04401410).
Collapse
Affiliation(s)
- Spyridoula Vasileiou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX.
| | - LaQuisa Hill
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Manik Kuvalekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Aster G Workineh
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Ayumi Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Yovana Velazquez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Suhasini Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Kimberly Mooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Natalia Lapteva
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Bambi J Grilley
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Todd N Eagar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - George Carrum
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Kevin A Grimes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Ann M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| | - Premal Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX
| |
Collapse
|
8
|
Fernández-Moreno R, Valle-Arroyo J, Páez-Vega A, Salinas A, Cano A, Pérez AB, Torre-Cisneros J, Cantisán S. Memory SARS-CoV-2 T-cell response in convalescent COVID-19 patients with undetectable specific IgG antibodies: a comparative study. Front Immunol 2023; 14:1142918. [PMID: 37180143 PMCID: PMC10169638 DOI: 10.3389/fimmu.2023.1142918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/07/2023] [Indexed: 05/15/2023] Open
Abstract
Background During the COVID-19 pandemic, a variable percentage of patients with SARS-CoV-2 infection failed to elicit humoral response. This study investigates whether patients with undetectable SARS-CoV-2 IgG are able to generate SARS-CoV-2 memory T cells with proliferative capacity upon stimulation. Methods This cross-sectional study was conducted with convalescent COVID-19 patients, diagnosed with a positive real-time PCR (RT-PCR) from nasal and pharyngeal swab specimens. COVID-19 patients were enrolled ≥3 months after the last PCR positive. Proliferative T-cell response after whole blood stimulation was assessed using the FASCIA assay. Results A total of 119 participants (86 PCR-confirmed COVID-19 patients and 33 healthy controls) were randomly filtered from an initial cohort. Of these 86 patients, 59 had detectable (seropositive) and 27 had undetectable (seronegative) SARS-CoV-2 IgG. Seropositive patients were subclassified as asymptomatic/mild or severe according to the oxygen supplementation requirement. SARS-CoV-2 CD3+ and CD4+ T cells showed significantly lower proliferative response in seronegative than in seropositive patients. The ROC curve analysis indicated that ≥ 5 CD4+ blasts/μL of blood defined a "positive SARS-CoV-2 T cell response". According to this cut-off, 93.2% of seropositive patients had a positive T-cell response compared to 50% of seronegative patients and 20% of negative controls (chi-square; p < 0.001). Conclusions This proliferative assay is useful not only to discriminate convalescent patients from negative controls, but also to distinguish seropositive patients from those with undetectable SARS-CoV-2 IgG antibodies. Memory T cells in seronegative patients are able to respond to SARSCoV-2 peptides, although at a lower magnitude than seropositive patients.
Collapse
Affiliation(s)
- Raquel Fernández-Moreno
- Spanish Network for Research in Infectious Diseases (REIPI), Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
| | - Jorge Valle-Arroyo
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
| | - Aurora Páez-Vega
- Spanish Network for Research in Infectious Diseases (REIPI), Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
| | - Ana Salinas
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
| | - Angela Cano
- Spanish Network for Research in Infectious Diseases (REIPI), Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
- Infectious Diseases Unit, Reina Sofía University Hospital, Cordoba, Spain
| | - Ana B Pérez
- Spanish Network for Research in Infectious Diseases (REIPI), Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
- Microbiology Unit, Reina Sofía University Hospital, Cordoba, Spain
| | - Julián Torre-Cisneros
- Spanish Network for Research in Infectious Diseases (REIPI), Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
- Infectious Diseases Unit, Reina Sofía University Hospital, Cordoba, Spain
| | - Sara Cantisán
- Spanish Network for Research in Infectious Diseases (REIPI), Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
- Infectious Diseases Unit, Reina Sofía University Hospital, Cordoba, Spain
| |
Collapse
|
9
|
Al-Akioui-Sanz K, Pascual-Miguel B, Díaz-Almirón M, Mestre-Durán C, Navarro-Zapata A, Clares-Villa L, Martín-Cortázar C, Vicario JL, Moreno MÁ, Balas A, De Paz R, Minguillón J, Pérez-Martínez A, Ferreras C. Donor selection for adoptive cell therapy with CD45RA - memory T cells for patients with coronavirus disease 2019, and dexamethasone and interleukin-15 effects on the phenotype, proliferation and interferon gamma release. Cytotherapy 2023; 25:330-340. [PMID: 36585293 PMCID: PMC9742221 DOI: 10.1016/j.jcyt.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND AIMS We have previously demonstrated the safety and feasibility of adoptive cell therapy with CD45RA- memory T cells containing severe acute respiratory syndrome coronavirus 2-specific T cells for patients with coronavirus disease 2019 from an unvaccinated donor who was chosen based on human leukocyte antigen compatibility and cellular response. In this study, we examined the durability of cellular and humoral immunity within CD45RA- memory T cells and the effect of dexamethasone, the current standard of care treatment, and interleukin-15, a cytokine critically involved in T-cell maintenance and survival. METHODS We performed a longitudinal analysis from previously severe acute respiratory syndrome coronavirus 2-infected and infection-naïve individuals covering 21 months from infection and 10 months after full vaccination with the BNT162b2 Pfizer/BioNTech vaccine. RESULTS We observed that cellular responses are maintained over time. Humoral responses increased after vaccination but were gradually lost. In addition, dexamethasone did not alter cell functionality or proliferation of CD45RA- T cells, and interleukin-15 increased the memory T-cell activation state, regulatory T cell expression, and interferon gamma release. CONCLUSIONS Our results suggest that the best donors for adoptive cell therapy would be recovered individuals and 2 months after vaccination, although further studies with larger cohorts would be needed to confirm this finding. Dexamethasone did not affect the characteristics of the memory T cells at a concentration used in the clinical practice and IL-15 showed a positive effect on SARS-CoV-2-specific CD45RA- T cells.
Collapse
Affiliation(s)
- Karima Al-Akioui-Sanz
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Bárbara Pascual-Miguel
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | | | - Carmen Mestre-Durán
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Alfonso Navarro-Zapata
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Laura Clares-Villa
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Carla Martín-Cortázar
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - José Luis Vicario
- Histocompatibility Unit, Transfusion Center of Madrid, Madrid, Spain
| | | | - Antonio Balas
- Histocompatibility Unit, Transfusion Center of Madrid, Madrid, Spain
| | - Raquel De Paz
- Cell Therapy Unit, Hematology Department, La Paz University Hospital, Madrid, Spain
| | - Jordi Minguillón
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Antonio Pérez-Martínez
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain; Pediatric Hemato-oncology Department, La Paz University Hospital, Madrid, Spain; Faculty of Medicine Autonomous, University of Madrid, Madrid, Spain
| | - Cristina Ferreras
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain.
| |
Collapse
|
10
|
Quach DH, Lulla P, Rooney CM. Banking on virus-specific T cells to fulfill the need for off-the-shelf cell therapies. Blood 2023; 141:877-885. [PMID: 36574622 PMCID: PMC10023738 DOI: 10.1182/blood.2022016202] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/28/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
Adoptively transferred virus-specific T cells (VSTs) have shown remarkable safety and efficacy for the treatment of virus-associated diseases and malignancies in hematopoietic stem cell transplant (HSCT) recipients, for whom VSTs are derived from the HSCT donor. Autologous VSTs have also shown promise for the treatment of virus-driven malignancies outside the HSCT setting. In both cases, VSTs are manufactured as patient-specific products, and the time required for procurement, manufacture, and release testing precludes their use in acutely ill patients. Further, Good Manufacturing Practices-compliant products are expensive, and failures are common in virus-naive HSCT donors and patient-derived VSTs that are rendered anergic by immunosuppressive tumors. Hence, highly characterized, banked VSTs (B-VSTs) that can be used for multiple unrelated recipients are highly desirable. The major challenges facing B-VSTs result from the inevitable mismatches in the highly polymorphic and immunogenic human leukocyte antigens (HLA) that present internally processed antigens to the T-cell receptor, leading to the requirement for partial HLA matching between the B-VST and recipient. HLA mismatches lead to rapid rejection of allogeneic T-cell products and graft-versus-host disease induced by alloreactive T cells in the infusion product. Here, we summarize the clinical outcomes to date of trials of B-VSTs used for the treatment of viral infections and malignancies and their potential as a platform for chimeric antigen receptors targeting nonviral tumors. We will highlight the properties of VSTs that make them attractive off-the-shelf cell therapies, as well as the challenges that must be overcome before they can become mainstream.
Collapse
Affiliation(s)
- David H. Quach
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Premal Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Cliona M. Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX
- Department of Molecular Virology and Immunology, Baylor College of Medicine, Houston, TX
| |
Collapse
|
11
|
Pathogen-specific T Cells: Targeting Old Enemies and New Invaders in Transplantation and Beyond. Hemasphere 2023; 7:e809. [PMID: 36698615 PMCID: PMC9831191 DOI: 10.1097/hs9.0000000000000809] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/07/2022] [Indexed: 01/27/2023] Open
Abstract
Adoptive immunotherapy with virus-specific cytotoxic T cells (VSTs) has evolved over the last three decades as a strategy to rapidly restore virus-specific immunity to prevent or treat viral diseases after solid organ or allogeneic hematopoietic cell-transplantation (allo-HCT). Since the early proof-of-principle studies demonstrating that seropositive donor-derived T cells, specific for the commonest pathogens post transplantation, namely cytomegalovirus or Epstein-Barr virus (EBV) and generated by time- and labor-intensive protocols, could effectively control viral infections, major breakthroughs have then streamlined the manufacturing process of pathogen-specific T cells (pSTs), broadened the breadth of target recognition to even include novel emerging pathogens and enabled off-the-shelf administration or pathogen-naive donor pST production. We herein review the journey of evolution of adoptive immunotherapy with nonengineered, natural pSTs against infections and virus-associated malignancies in the transplant setting and briefly touch upon recent achievements using pSTs outside this context.
Collapse
|
12
|
El Chaer F, Auletta JJ, Chemaly RF. How I treat and prevent COVID-19 in patients with hematologic malignancies and recipients of cellular therapies. Blood 2022; 140:673-684. [PMID: 35776899 PMCID: PMC9249429 DOI: 10.1182/blood.2022016089] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/16/2022] [Indexed: 12/15/2022] Open
Abstract
Patients with hematologic malignancies and recipients of hematopoietic cell transplantation (HCT) are more likely to experience severe coronavirus disease 2019 (COVID-19) and have a higher risk of morbidity and mortality after infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Compared with the general population, these patients have suboptimal humoral responses to COVID-19 vaccines and subsequently increased risk for breakthrough infections, underscoring the need for additional therapies, including pre- and postexposure prophylaxis, to attenuate clinical progression to severe COVID-19. Therapies for COVID-19 are mostly available for adults and in the inpatient and outpatient settings. Selection and administration of the best treatment options are based on host factors; virus factors, including circulating SARS-CoV-2 variants; and therapeutic considerations, including the clinical efficacy, availability, and practicality of treatment and its associated side effects, including drug-drug interactions. In this paper, we discuss how we approach managing COVID-19 in patients with hematologic malignancies and recipients of HCT and cell therapy.
Collapse
Affiliation(s)
- Firas El Chaer
- Division of Hematology and Oncology, Department of Medicine, University of Virginia, Charlottesville, VA
| | - Jeffery J Auletta
- National Marrow Donor Program/Be The Match, Minneapolis, MN
- Divisions of Hematology, Oncology, Blood and Marrow Transplant & Infectious Diseases, Nationwide Children's Hospital, Columbus, OH; and
| | - Roy F Chemaly
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
13
|
Papadopoulou A, Stavridou F, Giannaki M, Paschoudi K, Chatzopoulou F, Gavriilaki E, Georgolopoulos G, Anagnostopoulos A, Yannaki E. Robust SARS-COV-2-specific T-cell immune memory persists long-term in immunocompetent individuals post BNT162b2 double shot. Heliyon 2022; 8:e09863. [PMID: 35815135 PMCID: PMC9250414 DOI: 10.1016/j.heliyon.2022.e09863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/23/2022] [Accepted: 06/29/2022] [Indexed: 11/08/2022] Open
Abstract
Background A robust efficiency of mRNA vaccines against coronavirus disease-2019 has been demonstrated, however, the intended long-term protection against SARS-CoV-2 has been challenged by the waning humoral and cellular immunity over time, leading to a third vaccination dose recommendation for immunocompetent individuals, six months after completion of primary mRNA vaccination. Methods We here measured humoral responses via an immunoassay measuring SARS-CoV-2 neutralizing antibodies and T-cell responses using Elispot for interferon-γ 1- and 8- months post full BNT162b2 vaccination, in 10 health-care professionals. To explore whether the declining abundance of coronavirus-specific T-cells (CoV-2-STs) truly reflects decreased capacity for viral control, rather than the attenuating viral stimulus over time, we modeled ex vivo the T-cellular response upon viral challenge in fully vaccinated immunocompetent individuals, 1- and 8-months post BNT162b2. Findings. Notwithstanding the declining CoV-2-neutralizing antibodies and CoV-2-STs, re-challenged CoV-2-STs, 1- and 8-months post vaccination, presented similar functional characteristics including high cytotoxicity against both the unmutated virus and the delta variant. Interpretation. These findings suggest robust and sustained cellular immune response upon SARS-CοV-2 antigen exposure, 8 months post mRNA vaccination, despite declining CοV-2-STs over time in the presence of an attenuating viral stimulus.
Collapse
|
14
|
Gavriilaki E, Papadopoulou A, Touloumenidou T, Stavridou F, Koravou EE, Giannaki M, Papalexandri A, Karavalakis G, Batsis I, Kourelis A, Chatzopoulou F, Chatzidimitriou D, Sotiropoulos D, Yannaki E, Sakellari I, Anagnostopoulos A. Neutralizing antibody and T cell responses to SARS-CoV-2 vaccination in hematopoietic cell transplant recipients. Bone Marrow Transplant 2022; 57:1183-1186. [PMID: 35449455 PMCID: PMC9022618 DOI: 10.1038/s41409-022-01675-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 12/30/2022]
Affiliation(s)
- Eleni Gavriilaki
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Anastasia Papadopoulou
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Tasoula Touloumenidou
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Fani Stavridou
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Evaggelia-Evdoxia Koravou
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Maria Giannaki
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Apostolia Papalexandri
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Georgios Karavalakis
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Ioannis Batsis
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Andreas Kourelis
- Microbiology Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Fani Chatzopoulou
- Microbiology Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Damianos Sotiropoulos
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Evangelia Yannaki
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece.
| | - Ioanna Sakellari
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Achilles Anagnostopoulos
- Hematology Department - Hematopoietic Cell Transplant (HCT) Unit - Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| |
Collapse
|
15
|
Panikkar A, Lineburg KE, Raju J, Chew KY, Ambalathingal GR, Rehan S, Swaminathan S, Crooks P, Le Texier L, Beagley L, Best S, Solomon M, Matthews KK, Srihari S, Neller MA, Short KR, Khanna R, Smith C. SARS-CoV-2-specific T cells generated for adoptive immunotherapy are capable of recognizing multiple SARS-CoV-2 variants. PLoS Pathog 2022; 18:e1010339. [PMID: 35157735 PMCID: PMC8880869 DOI: 10.1371/journal.ppat.1010339] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/25/2022] [Accepted: 02/04/2022] [Indexed: 12/13/2022] Open
Abstract
Adoptive T-cell immunotherapy has provided promising results in the treatment of viral complications in humans, particularly in the context of immunocompromised patients who have exhausted all other clinical options. The capacity to expand T cells from healthy immune individuals is providing a new approach to anti-viral immunotherapy, offering rapid off-the-shelf treatment with tailor-made human leukocyte antigen (HLA)-matched T cells. While most of this research has focused on the treatment of latent viral infections, emerging evidence that SARS-CoV-2-specific T cells play an important role in protection against COVID-19 suggests that the transfer of HLA-matched allogeneic off-the-shelf virus-specific T cells could provide a treatment option for patients with active COVID-19 or at risk of developing COVID-19. We initially screened 60 convalescent individuals and based on HLA typing and T-cell response profile, 12 individuals were selected for the development of a SARS-CoV-2-specific T-cell bank. We demonstrate that these T cells are specific for up to four SARS-CoV-2 antigens presented by a broad range of both HLA class I and class II alleles. These T cells show consistent functional and phenotypic properties, display cytotoxic potential against HLA-matched targets and can recognize HLA-matched cells infected with different SARS-CoV-2 variants. These observations demonstrate a robust approach for the production of SARS-CoV-2-specific T cells and provide the impetus for the development of a T-cell repository for clinical assessment. Since the emergence of SARS-CoV-2 variants that reduce the effectiveness of vaccines, it is evident that other interventional strategies will be needed to treat COVID-19, particularly in patients with a compromised immune system who are at an increased risk of developing severe COVID-19. Off-the-shelf T-cell immunotherapy is proving to be a powerful tool to treat viral disease in patients with a compromised immune system. Here, we report here that a small number of SARS-CoV-2 exposed individuals can be used generate a bank of specific T cells that provide broad population coverage. Importantly, we demonstrate that most of the epitopes recognized by these T cells remain unchanged in different variants and that the T cells can recognize cells infected with three different variants of SARS-CoV-2. We believe these observations provide critical proof-of-concept that T-cell based immunotherapy may offer an option for the future treatment of immunocompromised patients who remain susceptible to the severe complications associated with COVID-19.
Collapse
Affiliation(s)
- Archana Panikkar
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Katie E. Lineburg
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Jyothy Raju
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Keng Yih Chew
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia Queensland, Australia
| | - George R. Ambalathingal
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Sweera Rehan
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Srividhya Swaminathan
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Pauline Crooks
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Laetitia Le Texier
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Leone Beagley
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Shannon Best
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Matthew Solomon
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Katherine K. Matthews
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Sriganesh Srihari
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Michelle A. Neller
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Kirsty R. Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia Queensland, Australia
| | - Rajiv Khanna
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
- * E-mail:
| |
Collapse
|
16
|
Kim N, Lee JM, Oh EJ, Jekarl DW, Lee DG, Im KI, Cho SG. Off-the-Shelf Partial HLA Matching SARS-CoV-2 Antigen Specific T Cell Therapy: A New Possibility for COVID-19 Treatment. Front Immunol 2022; 12:751869. [PMID: 35003063 PMCID: PMC8733616 DOI: 10.3389/fimmu.2021.751869] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022] Open
Abstract
Background Immunological characteristics of COVID-19 show pathological hyperinflammation associated with lymphopenia and dysfunctional T cell responses. These features provide a rationale for restoring functional T cell immunity in COVID-19 patients by adoptive transfer of SARS-CoV-2 specific T cells. Methods To generate SARS-CoV-2 specific T cells, we isolated peripheral blood mononuclear cells from 7 COVID-19 recovered and 13 unexposed donors. Consequently, we stimulated cells with SARS-CoV-2 peptide mixtures covering spike, membrane and nucleocapsid proteins. Then, we culture expanded cells with IL-2 for 21 days. We assessed immunophenotypes, cytokine profiles, antigen specificity of the final cell products. Results Our results show that SARS-CoV-2 specific T cells could be expanded in both COVID-19 recovered and unexposed groups. Immunophenotypes were similar in both groups showing CD4+ T cell dominance, but CD8+ and CD3+CD56+ T cells were also present. Antigen specificity was determined by ELISPOT, intracellular cytokine assay, and cytotoxicity assays. One out of 14 individuals who were previously unexposed to SARS-CoV-2 failed to show antigen specificity. Moreover, ex-vivo expanded SARS-CoV-2 specific T cells mainly consisted of central and effector memory subsets with reduced alloreactivity against HLA-unmatched cells suggesting the possibility for the development of third-party partial HLA-matching products. Discussion In conclusion, our findings show that SARS-CoV-2 specific T cell can be readily expanded from both COVID-19 and unexposed individuals and can therefore be manufactured as a biopharmaceutical product to treat severe COVID-19 patients. One Sentence Summary Ex-vivo expanded SARS-CoV-2 antigen specific T cells developed as third-party partial HLA-matching products may be a promising approach for treating severe COVID-19 patients that do not respond to previous treatment options.
Collapse
Affiliation(s)
- Nayoun Kim
- Product Development Division, LucasBio Co., Ltd., Seoul, South Korea
| | - Jong-Min Lee
- Division of Respiratory, Allergy and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Eun-Jee Oh
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Dong Wook Jekarl
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Dong-Gun Lee
- Division of Infectious Diseases, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Keon-Il Im
- Product Development Division, LucasBio Co., Ltd., Seoul, South Korea.,Institute for Translational Research and Molecular Imaging, The Catholic University of Korea, Seoul, South Korea
| | - Seok-Goo Cho
- Product Development Division, LucasBio Co., Ltd., Seoul, South Korea.,Institute for Translational Research and Molecular Imaging, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
17
|
Khanolkar A. Elucidating T Cell and B Cell Responses to SARS-CoV-2 in Humans: Gaining Insights into Protective Immunity and Immunopathology. Cells 2021; 11:cells11010067. [PMID: 35011627 PMCID: PMC8750814 DOI: 10.3390/cells11010067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
The SARS-CoV-2 pandemic is an unprecedented epochal event on at least two fronts. Firstly, in terms of the rapid spread and the magnitude of the outbreak, and secondly, on account of the equally swift response of the scientific community that has galvanized itself into action and has successfully developed, tested and deployed highly effective and novel vaccines in record time to combat the virus. The sophistication and diversification of the scientific toolbox we now have at our disposal has enabled us to interrogate both the breadth and the depth of the immune response to a degree that is unparalleled in recent memory. In terms of our understanding of what is critical to contain the virus and mitigate the effects the pandemic, neutralizing antibodies to SARS-CoV-2 garner most of the attention, however, it is essential to recognize that it is the quality and the fitness of the virus-specific T cell and B cell response that lays the foundation and the backdrop for an effective neutralizing antibody response. In this report, we will review some of the key findings that have helped define and delineate some of the essential attributes of T and B cell responses in the setting of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Aaruni Khanolkar
- Department of Pathology, Ann and Robert H. Lurie Children’s Hospital of Chicago, 225 East Chicago Avenue, Box 82, Chicago, IL 60611, USA; ; Tel.: +1-312-227-8073
- Department of Pathology, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|