1
|
Schweitzer L, Muranski P. Virus-specific T cell therapy to treat refractory viral infections in solid organ transplant recipients. Am J Transplant 2024; 24:1558-1566. [PMID: 38857784 DOI: 10.1016/j.ajt.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024]
Abstract
Solid organ transplant recipients require ongoing immunosuppression to prevent acute rejection, which puts them at risk of opportunistic infections. Viral infections are particularly challenging to prevent and treat as many establish latency and thus cannot be eliminated, whereas targets for small molecule antiviral medications are limited. Resistance to antivirals and unacceptable toxicity also complicate treatment. Virus-specific T cell therapies aim to restore host-specific immunity to opportunistic viruses that is lacking due to ongoing immunosuppressive therapy. This minireview will provide a state-of-the-art update of the current virus-specific T cell pipeline and translational research that is likely to lead to further treatment options for viral infections in solid organ transplant recipients.
Collapse
Affiliation(s)
- Lorne Schweitzer
- Department of Medicine, Division of Infectious Diseases, Columbia University Irving Medical Center, New York, New York, USA; Columbia Center for Translational Immunology, New York, New York, USA
| | - Pawel Muranski
- Department of Medicine, Division of Hematology, Columbia University Irving Medical Center, New York, New York, USA; Columbia Center for Translational Immunology, New York, New York, USA.
| |
Collapse
|
2
|
Kakavandi S, Hajikhani B, Azizi P, Aziziyan F, Nabi-Afjadi M, Farani MR, Zalpoor H, Azarian M, Saadi MI, Gharesi-Fard B, Terpos E, Zare I, Motamedifar M. COVID-19 in patients with anemia and haematological malignancies: risk factors, clinical guidelines, and emerging therapeutic approaches. Cell Commun Signal 2024; 22:126. [PMID: 38360719 PMCID: PMC10868124 DOI: 10.1186/s12964-023-01316-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/13/2023] [Indexed: 02/17/2024] Open
Abstract
Extensive research in countries with high sociodemographic indices (SDIs) to date has shown that coronavirus disease 2019 (COVID-19) may be directly associated with more severe outcomes among patients living with haematological disorders and malignancies (HDMs). Because individuals with moderate to severe immunodeficiency are likely to undergo persistent infections, shed virus particles for prolonged periods, and lack an inflammatory or abortive phase, this represents an overall risk of morbidity and mortality from COVID-19. In cases suffering from HDMs, further investigation is needed to achieve a better understanding of triviruses and a group of related variants in patients with anemia and HDMs, as well as their treatment through vaccines, drugs, and other methods. Against this background, the present study aimed to delineate the relationship between HDMs and the novel COVID-19, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Besides, effective treatment options for HDM cases were further explored to address this epidemic and its variants. Therefore, learning about how COVID-19 manifests in these patients, along with exploiting the most appropriate treatments, may lead to the development of treatment and care strategies by clinicians and researchers to help patients recover faster. Video Abstract.
Collapse
Affiliation(s)
- Sareh Kakavandi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahareh Hajikhani
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Paniz Azizi
- Psychological and Brain Science Departments, Program in Neuroscience, Indiana University, Bloomington, IN, USA
| | - Fatemeh Aziziyan
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Marzieh Ramezani Farani
- Department of Biological Sciences and Bioengineering, Nano Bio High-Tech Materials Research Center, Inha University, Incheon, 22212, Republic of Korea
| | - Hamidreza Zalpoor
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Maryam Azarian
- Department of Radiology, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
| | | | | | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co., Ltd., Shiraz, 7178795844, Iran.
| | - Mohammad Motamedifar
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Shiraz HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Gopcsa L, Réti M, Andrikovics H, Bobek I, Bekő G, Bogyó J, Ceglédi A, Dobos K, Giba-Kiss L, Jankovics I, Kis O, Lakatos B, Mathiász D, Meggyesi N, Miskolczi G, Németh N, Paksi M, Riczu A, Sinkó J, Szabó B, Szilvási A, Szlávik J, Tasnády S, Reményi P, Vályi-Nagy I. Effective virus-specific T-cell therapy for high-risk SARS-CoV-2 infections in hematopoietic stem cell transplant recipients: initial case studies and literature review. GeroScience 2024; 46:1083-1106. [PMID: 37414968 PMCID: PMC10828167 DOI: 10.1007/s11357-023-00858-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023] Open
Abstract
The COVID-19 pandemic has exacerbated mortality rates among immunocompromised patients, accentuating the need for novel, targeted therapies. Transplant recipients, with their inherent immune vulnerabilities, represent a subgroup at significantly heightened risk. Current conventional therapies often demonstrate limited effectiveness in these patients, calling for innovative treatment approaches. In immunocompromised transplant recipients, several viral infections have been successfully treated by adoptive transfer of virus-specific T-cells (VST). This paper details the successful application of SARS-CoV-2-specific memory T-cell therapy, produced by an interferon-γ cytokine capture system (CliniMACS® Prodigy device), in three stem cell transplant recipients diagnosed with COVID-19 (case 1: alpha variant, cases 2 and 3: delta variants). These patients exhibited persistent SARS-CoV-2 PCR positivity accompanied by bilateral pulmonary infiltrates and demonstrated only partial response to standard treatments. Remarkably, all three patients recovered and achieved viral clearance within 3 to 9 weeks post-VST treatment. Laboratory follow-up investigations identified an increase in SARS-CoV-2-specific T-cells in two of the cases. A robust anti-SARS-CoV-2 S (S1/S2) IgG serological response was also recorded, albeit with varying titers. The induction of memory T-cells within the CD4 + compartment was confirmed, and previously elevated interleukin-6 (IL-6) and IL-8 levels normalized post-VST therapy. The treatment was well tolerated with no observed adverse effects. While the need for specialized equipment and costs associated with VST therapy present potential challenges, the limited treatment options currently available for COVID-19 within the allogeneic stem cell transplant population, combined with the risk posed by emerging SARS-CoV-2 mutations, underscore the potential of VST therapy in future clinical practice. This therapeutic approach may be particularly beneficial for elderly patients with multiple comorbidities and weakened immune systems.
Collapse
Affiliation(s)
- László Gopcsa
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, 1 Nagyvárad Square, P.B. 1097, Budapest, Hungary.
| | - Marienn Réti
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, 1 Nagyvárad Square, P.B. 1097, Budapest, Hungary
| | - Hajnalka Andrikovics
- Laboratory of Molecular Genetics, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Ilona Bobek
- Department of Intensive Care Unit, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Gabriella Bekő
- Department of Central Laboratory, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Judit Bogyó
- Hungarian National Blood Transfusion Service, Karolina Út 19-21, 1113, Budapest, Hungary
| | - Andrea Ceglédi
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, 1 Nagyvárad Square, P.B. 1097, Budapest, Hungary
| | - Katalin Dobos
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, 1 Nagyvárad Square, P.B. 1097, Budapest, Hungary
| | - Laura Giba-Kiss
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, 1 Nagyvárad Square, P.B. 1097, Budapest, Hungary
| | - István Jankovics
- National Public Health and Medical Officer Service, Albert Florian Út 2-6, 1097, Budapest, Hungary
| | - Orsolya Kis
- Department of Intensive Care Unit, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Botond Lakatos
- Department of Infectious Diseases, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Dóra Mathiász
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, 1 Nagyvárad Square, P.B. 1097, Budapest, Hungary
| | - Nóra Meggyesi
- Laboratory of Molecular Genetics, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Gottfried Miskolczi
- Department of Central Laboratory, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Noémi Németh
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, 1 Nagyvárad Square, P.B. 1097, Budapest, Hungary
| | - Melinda Paksi
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, 1 Nagyvárad Square, P.B. 1097, Budapest, Hungary
| | - Alexandra Riczu
- Department of Infectious Diseases, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - János Sinkó
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, 1 Nagyvárad Square, P.B. 1097, Budapest, Hungary
| | - Bálint Szabó
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, 1 Nagyvárad Square, P.B. 1097, Budapest, Hungary
| | - Anikó Szilvási
- Hungarian National Blood Transfusion Service, Karolina Út 19-21, 1113, Budapest, Hungary
| | - János Szlávik
- Department of Infectious Diseases, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Szabolcs Tasnády
- Department of Central Laboratory, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Péter Reményi
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, 1 Nagyvárad Square, P.B. 1097, Budapest, Hungary
| | - István Vályi-Nagy
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, 1 Nagyvárad Square, P.B. 1097, Budapest, Hungary
| |
Collapse
|
4
|
Herrera S, Aguado JM, Candel FJ, Cordero E, Domínguez-Gil B, Fernández-Ruiz M, Los Arcos I, Len Ò, Marcos MÁ, Muñez E, Muñoz P, Rodríguez-Goncer I, Sánchez-Céspedes J, Valerio M, Bodro M. Executive summary of the consensus statement of the group for the study of infection in transplantation and other immunocompromised host (GESITRA-IC) of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC) on the treatment of SARS-CoV-2 infection in solid organ transplant recipients. Transplant Rev (Orlando) 2023; 37:100788. [PMID: 37591117 DOI: 10.1016/j.trre.2023.100788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023]
Affiliation(s)
- Sabina Herrera
- Department of Infectious Diseases, Hospital Clínic, IDIBAPS (Institut D'Investigacions Biomèdiques Agust Pi I Sunyer), Universitat de Barcelona, Barcelona, Spain
| | - Jose M Aguado
- Infectious Diseases Unit, Hospital Universitario 12 de Octubre (Madrid), Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Francisco Javier Candel
- Clinical Microbiology & Infectious Diseases, Transplant Coordination, Hospital Clínico Universitario San Carlos, Madrid 28040, Spain; Department of Clinical Microbiology and Infectious Diseases, Hospital Clínico San Carlos, Madrid, Spain
| | - Elisa Cordero
- Infectious Diseases Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina Sevilla, Sevilla, Spain
| | | | - Mario Fernández-Ruiz
- Infectious Diseases Unit, Hospital Universitario 12 de Octubre (Madrid), Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Ibai Los Arcos
- Infectious Diseases Department, Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | - Òscar Len
- Infectious Diseases Department, Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | | | - Elena Muñez
- Infectious Diseases Unit, Internal Medicine Department, University Hospital Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Patricia Muñoz
- Department of Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 9 Madrid, Spain
| | - Isabel Rodríguez-Goncer
- Infectious Diseases Unit, Hospital Universitario 12 de Octubre (Madrid), Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Javier Sánchez-Céspedes
- Infectious Diseases Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina Sevilla, Sevilla, Spain
| | - Maricela Valerio
- Department of Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 9 Madrid, Spain
| | - Marta Bodro
- Department of Infectious Diseases, Hospital Clínic, IDIBAPS (Institut D'Investigacions Biomèdiques Agust Pi I Sunyer), Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
5
|
Chen Q, Chia A, Hang SK, Lim A, Koh WK, Peng Y, Gao F, Chen J, Ho Z, Wai LE, Kunasegaran K, Tan AT, Le Bert N, Loh CY, Goh YS, Renia L, Dong T, Vathsala A, Bertoletti A. Engineering immunosuppressive drug-resistant armored (IDRA) SARS-CoV-2 T cells for cell therapy. Cell Mol Immunol 2023; 20:1300-1312. [PMID: 37666955 PMCID: PMC10616128 DOI: 10.1038/s41423-023-01080-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023] Open
Abstract
Solid organ transplant (SOT) recipients receive immunosuppressive drugs (ISDs) and are susceptible to developing severe COVID-19. Here, we analyze the Spike-specific T-cell response after 3 doses of mRNA vaccine in a group of SOT patients (n = 136) treated with different ISDs. We demonstrate that a combination of a calcineurin inhibitor (CNI), mycophenolate mofetil (MMF), and prednisone (Pred) treatment regimen strongly suppressed the mRNA vaccine-induced Spike-specific cellular response. Such defects have clinical consequences because the magnitude of vaccine-induced Spike-specific T cells was directly proportional to the ability of SOT patients to rapidly clear SARS-CoV-2 after breakthrough infection. To then compensate for the T-cell defects induced by immunosuppressive treatment and to develop an alternative therapeutic strategy for SOT patients, we describe production of 6 distinct SARS-CoV-2 epitope-specific ISD-resistant T-cell receptor (TCR)-T cells engineered using the mRNA electroporation method with reactivity minimally affected by mutations occurring in Beta, Delta, Gamma, and Omicron variants. This strategy with transient expression characteristics marks an improvement in the immunotherapeutic field and provides an attractive and novel therapeutic possibility for immunosuppressed COVID-19 patients.
Collapse
Affiliation(s)
- Qi Chen
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore
| | - Adeline Chia
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore
| | - Shou Kit Hang
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore
| | - Amy Lim
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
| | - Wee Kun Koh
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
| | - Yanchun Peng
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Fei Gao
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Jili Chen
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Zack Ho
- Lion TCR Pte Ltd, Singapore, Singapore
| | - Lu-En Wai
- Lion TCR Pte Ltd, Singapore, Singapore
| | - Kamini Kunasegaran
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore
| | - Anthony Tanoto Tan
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore
| | - Nina Le Bert
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore
| | - Chiew Yee Loh
- A*STAR ID labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Yun Shan Goh
- A*STAR ID labs, Agency for Science, Technology and Research, Singapore, Singapore
| | - Laurent Renia
- A*STAR ID labs, Agency for Science, Technology and Research, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Tao Dong
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Anantharaman Vathsala
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Antonio Bertoletti
- Emerging Infectious Disease Program, Duke-NUS Medical School, Singapore, Singapore.
- Singapore Immunology Network, A*STAR, Singapore, Singapore.
| |
Collapse
|
6
|
Kampouri E, Hill JA, Dioverti V. COVID-19 after hematopoietic cell transplantation and chimeric antigen receptor (CAR)-T-cell therapy. Transpl Infect Dis 2023; 25 Suppl 1:e14144. [PMID: 37767643 DOI: 10.1111/tid.14144] [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: 06/15/2023] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
More than 3 years have passed since Coronavirus disease 2019 (COVID-19) was declared a global pandemic, yet COVID-19 still severely impacts immunocompromised individuals including those treated with hematopoietic cell transplantation (HCT) and chimeric antigen receptor-T-cell therapies who remain at high risk for severe COVID-19 and mortality. Despite vaccination efforts, these patients have inadequate responses due to immunosuppression, which underscores the need for additional preventive approaches. The optimal timing, schedule of vaccination, and immunological correlates for protective immunity remain unknown. Antiviral therapies used early during disease can reduce mortality and severity due to COVID-19. The combination or sequential use of antivirals could be beneficial to control replication and prevent the development of treatment-related mutations in protracted COVID-19. Despite conflicting data, COVID-19 convalescent plasma remains an option in immunocompromised patients with mild-to-moderate disease to prevent progression. Protracted COVID-19 has been increasingly recognized among these patients and has been implicated in intra-host emergence of SARS-CoV-2 variants. Finally, novel SARS-CoV2-specific T-cells and natural killer cell-boosting (or -containing) products may be active against multiple variants and are promising therapies in immunocompromised patients.
Collapse
Affiliation(s)
- Eleftheria Kampouri
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Veronica Dioverti
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Haidar G, Jacobs JL, Kramer KH, Naqvi A, Heaps A, Parikh U, McCormick KD, Sobolewski MD, Agha M, Bogdanovich T, Bushunow V, Farah R, Hensley M, Hsu YMS, Johnson B, Klamar-Blain C, Kozar J, Lendermon E, Macatangay BJC, Marino CC, Raptis A, Salese E, Silveira FP, Leen AM, Marshall WL, Miller M, Patel B, Atillasoy E, Mellors JW. Therapy With Allogeneic Severe Acute Respiratory Syndrome Coronavirus-2-Specific T Cells for Persistent Coronavirus Disease 2019 in Immunocompromised Patients. Clin Infect Dis 2023; 77:696-702. [PMID: 37078720 PMCID: PMC10495124 DOI: 10.1093/cid/ciad233] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 04/21/2023] Open
Abstract
We administered severe acute respiratory syndrome coronavirus-2 viral-specific T cells (VSTs) under emergency investigational new drug applications to 6 immunocompromised patients with persistent coronavirus disease 2019 (COVID-19) and characterized clinical and virologic responses. Three patients had partial responses after failing other therapies but then died. Two patients completely recovered, but the role of VSTs in recovery was unclear due to concomitant use of other antivirals. One patient had not responded to 2 courses of remdesivir and experienced sustained recovery after VST administration. The use of VSTs in immunocompromised patients with persistent COVID-19 requires further study.
Collapse
Affiliation(s)
- Ghady Haidar
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jana L Jacobs
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kailey Hughes Kramer
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Asma Naqvi
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Amy Heaps
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Urvi Parikh
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kevin D McCormick
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Michele D Sobolewski
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mounzer Agha
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Tatiana Bogdanovich
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Vasilii Bushunow
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Rafic Farah
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Matthew Hensley
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Yen-Michael S Hsu
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Bruce Johnson
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Cynthia Klamar-Blain
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jennifer Kozar
- Department of Pharmacy, Investigational Drug Services, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Elizabeth Lendermon
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Bernard J C Macatangay
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Christopher C Marino
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Anastasios Raptis
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Erin Salese
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Fernanda P Silveira
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ann M Leen
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | - John W Mellors
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
8
|
Reimann H, Moosmann C, Schober K, Lang V, Verhagen J, Zeun J, Mackensen A, Kremer AN, Völkl S, Aigner M. Identification and characterization of T-cell receptors with therapeutic potential showing conserved specificity against all SARS-CoV 2 strains. Immunobiology 2023; 228:152720. [PMID: 37541134 DOI: 10.1016/j.imbio.2023.152720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/06/2023]
Abstract
INTRODUCTION Treatment of severe COVID-19 disease can be challenging in immunocompromized patients due to newly emerging virus variants of concern (VOC) escaping the humoral response. Thus, T cells recognizing to date unmutated epitopes are not only relevant for patients' immune responses against VOC, but might also serve as a therapeutic option for patients with severe COVID-19 disease in the future, e.g. following allogenic stem cell transplantation. METHODS To this purpose, the activation, cytokine profile and specificity of T-cell clones against unmutated and omicron Spike (S)-protein was analyzed, HLA restriction was determined and most promising T-cell receptor (TCR) was introduced into allogeneic T cells via CRISPR/Cas9-mediated orthotopic TCR replacement. Finally, T-cell responses of engineered T cells was determined and durability of the TCR replacement measured. PERSPECTIVE SARS-CoV-2 specific engineered T cells recognizing a genomically stable region of the S-protein of all SARS-CoV 2 variants were successfully generated. Such transgenic T cells exhibit favorable effector functions and provide a treatment option of immunocompromised COVID-19 patients.
Collapse
Affiliation(s)
- Hannah Reimann
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany.
| | - Carolin Moosmann
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Kilian Schober
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Vanessa Lang
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Johan Verhagen
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Department of Internal Medicine 3, Rheumatology and Immunology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Zeun
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Anita N Kremer
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Michael Aigner
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| |
Collapse
|
9
|
Vasileiou S, Kuvalekar M, Velazquez Y, Watanabe A, Narula M, Workineh AG, French-Kim M, Chavez AT, Gilmore S, Rooney CM, Leen AM. Longitudinal analysis of the evolution of cellular immunity to SARS-CoV-2 induced by infection and vaccination. Haematologica 2023; 108:1934-1939. [PMID: 36373251 PMCID: PMC10316232 DOI: 10.3324/haematol.2022.281947] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/31/2022] [Indexed: 06/26/2024] Open
Affiliation(s)
- Spyridoula Vasileiou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston.
| | - Manik Kuvalekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston
| | - Yovana Velazquez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston
| | - Ayumi Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston
| | - Mansi Narula
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston
| | - Aster G Workineh
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston
| | - Matthew French-Kim
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston
| | - Alejandro Torres Chavez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston
| | | | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston
| | - Ann M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston
| |
Collapse
|
10
|
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
|
11
|
Kegler A, Drewitz L, Arndt C, Daglar C, Rodrigues Loureiro L, Mitwasi N, Neuber C, González Soto KE, Bartsch T, Baraban L, Ziehr H, Heine M, Nieter A, Moreira-Soto A, Kühne A, Drexler JF, Seliger B, Laube M, Máthé D, Pályi B, Hajdrik P, Forgách L, Kis Z, Szigeti K, Bergmann R, Feldmann A, Bachmann M. A novel ACE2 decoy for both neutralization of SARS-CoV-2 variants and killing of infected cells. Front Immunol 2023; 14:1204543. [PMID: 37383226 PMCID: PMC10293748 DOI: 10.3389/fimmu.2023.1204543] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/17/2023] [Indexed: 06/30/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) led to millions of infections and deaths worldwide. As this virus evolves rapidly, there is a high need for treatment options that can win the race against new emerging variants of concern. Here, we describe a novel immunotherapeutic drug based on the SARS-CoV-2 entry receptor ACE2 and provide experimental evidence that it cannot only be used for (i) neutralization of SARS-CoV-2 in vitro and in SARS-CoV-2-infected animal models but also for (ii) clearance of virus-infected cells. For the latter purpose, we equipped the ACE2 decoy with an epitope tag. Thereby, we converted it to an adapter molecule, which we successfully applied in the modular platforms UniMAB and UniCAR for retargeting of either unmodified or universal chimeric antigen receptor-modified immune effector cells. Our results pave the way for a clinical application of this novel ACE2 decoy, which will clearly improve COVID-19 treatment.
Collapse
Affiliation(s)
- Alexandra Kegler
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Laura Drewitz
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Claudia Arndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Cansu Daglar
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Liliana Rodrigues Loureiro
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Nicola Mitwasi
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Christin Neuber
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Karla Elizabeth González Soto
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Tabea Bartsch
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Larysa Baraban
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Holger Ziehr
- Department of Pharmaceutical Biotechnology, Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Braunschweig, Germany
| | - Markus Heine
- Department of Pharmaceutical Biotechnology, Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Braunschweig, Germany
| | - Annabel Nieter
- Department of Pharmaceutical Biotechnology, Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Braunschweig, Germany
| | - Andres Moreira-Soto
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Arne Kühne
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jan Felix Drexler
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Barbara Seliger
- Medical Faculty, Martin-Luther-University Halle-Wittenberg, Halle, Germany
- Institute of Translational Immunology, Medical High School, Brandenburg an der Havel, Germany
| | - Markus Laube
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Hungarian Centre of Excellence for Molecular Medicine, In Vivo Imaging Advanced Core Facility, Szeged, Hungary
- CROmed Translational Research Ltd., Budapest, Hungary
| | - Bernadett Pályi
- National Biosafety Laboratory, Division of Microbiological Reference Laboratories, National Public Health Center, Budapest, Hungary
| | - Polett Hajdrik
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - László Forgách
- Semmelweis University School of Pharmacy, Semmelweis University, Budapest, Hungary
| | - Zoltán Kis
- National Biosafety Laboratory, Division of Microbiological Reference Laboratories, National Public Health Center, Budapest, Hungary
| | - Krisztián Szigeti
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Ralf Bergmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Anja Feldmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT), German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Bachmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT), German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Kodali L, Budhiraja P, Gea-Banacloche J. COVID-19 in kidney transplantation-implications for immunosuppression and vaccination. Front Med (Lausanne) 2022; 9:1060265. [PMID: 36507509 PMCID: PMC9727141 DOI: 10.3389/fmed.2022.1060265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
COVID-19 pandemic continues to challenge the transplant community, given increased morbidity and mortality associated with the disease and poor response to prevention measures such as vaccination. Transplant recipients have a diminished response to both mRNA and vector-based vaccines compared to dialysis and the general population. The currently available assays to measure response to vaccination includes commercially available antibody assays for anti-Spike Ab, or anti- Receptor Binding Domain Ab. Positive antibody testing on the assays does not always correlate with neutralizing antibodies unless the antibody levels are high. Vaccinations help with boosting polyfunctional CD4+ T cell response, which continues to improve with subsequent booster doses. Ongoing efforts to improve vaccine response by using additional booster doses and heterologous vaccine combinations are underway. There is improved antibody response in moderate responders; however, the ones with poor response to initial vaccination doses, continue to have a poor response to sequential boosters. Factors associated with poor vaccine response include diabetes, older age, specific immunosuppressants such as belatacept, and high dose mycophenolate. In poor responders, a decrease in immunosuppression can increase response to vaccination. COVID infection or vaccination has not been associated with an increased risk of rejection. Pre- and Post-exposure monoclonal antibodies are available to provide further protection against COVID infection, especially in poor vaccine responders. However, the efficacy is challenged by the emergence of new viral strains. A recently approved bivalent vaccine offers better protection against the Omicron variant.
Collapse
Affiliation(s)
- Lavanya Kodali
- Department of Internal Medicine, Mayo Clinic, Phoenix, AZ, United States
- Division of Nephrology, Transplant Center, Mayo Clinic, Phoenix, AZ, United States
| | - Pooja Budhiraja
- Department of Internal Medicine, Mayo Clinic, Phoenix, AZ, United States
- Division of Nephrology, Transplant Center, Mayo Clinic, Phoenix, AZ, United States
| | - Juan Gea-Banacloche
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| |
Collapse
|
15
|
Dioverti V, Salto-Alejandre S, Haidar G. Immunocompromised Patients with Protracted COVID-19: a Review of "Long Persisters". CURRENT TRANSPLANTATION REPORTS 2022; 9:209-218. [PMID: 36407883 PMCID: PMC9660019 DOI: 10.1007/s40472-022-00385-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2022] [Indexed: 11/13/2022]
Abstract
Purpose of Review Certain immunocompromised individuals are at risk for protracted COVID-19, in which SARS-CoV-2 leads to a chronic viral infection. However, the pathogenesis, diagnosis, and management of this phenomenon remain ill-defined. Recent Findings Herein, we review key aspects of protracted SARS-CoV-2 infection in immunocompromised individuals, or the so-called long persisters, and describe the clinical presentation, risk factors, diagnosis, and treatment modalities of this condition, as well as intra-host viral evolution. Based on the available data, we also propose a framework of criteria with which to approach this syndrome. Summary Protracted COVID-19 is an uncharacterized syndrome affecting patients with B-cell depletion; our proposed diagnostic approach and definitions will inform much needed future research.
Collapse
Affiliation(s)
- Veronica Dioverti
- Division of Infectious Diseases, Johns Hopkins University, 600 N Wolfe St, Baltimore, MD 21287 USA
| | - Sonsoles Salto-Alejandre
- Division of Infectious Diseases, Johns Hopkins University, 600 N Wolfe St, Baltimore, MD 21287 USA
- Clinical Unit of Infectious Diseases and Microbiology, Institute of Biomedicine of Seville (IBiS)/Virgen del Rocío University Hospital/University of Seville/CSIC, Avda. Manuel Siurot s/n 41013, Seville, Spain
| | - Ghady Haidar
- Division of Infectious Diseases, University of Pittsburgh and UPMC, 3601 Fifth Ave, Falk Medical Building, Suite 5B, Pittsburgh, PA 15213 USA
| |
Collapse
|
16
|
Avery RK. Update on COVID-19 Therapeutics for Solid Organ Transplant Recipients, Including the Omicron Surge. Transplantation 2022; 106:1528-1537. [PMID: 35700481 PMCID: PMC9311293 DOI: 10.1097/tp.0000000000004200] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 12/15/2022]
Abstract
Major changes have occurred in therapeutics for coronavirus-19 (COVID-19) infection over the past 12-18 mo, most notably in early outpatient therapy. In most cases, solid organ transplant recipients were not included in the original clinical trials of these agents, so studies of real-world outcomes have been important in building our understanding of their utility. This review examines what is known about clinical outcomes in solid organ transplant recipients with newer therapies. SARS-CoV-2 monoclonal antibodies for early treatment or prophylaxis have likely prevented many hospitalizations and deaths. In addition, convalescent plasma, the oral drugs nirmatrelvir/ritonavir and molnupiravir, remdesivir for early outpatient treatment, anti-inflammatory therapy, and investigational virus-specific T-cell therapy will be discussed. Finally, the later consequences of COVID-19, such as secondary infections, long COVID symptoms, and persistent active infection, are identified as areas for future research.
Collapse
Affiliation(s)
- Robin Kimiko Avery
- Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
17
|
Reddy R, Mintz J, Golan R, Firdaus F, Ponce R, Van Booven D, Manoharan A, Issa I, Blomberg BB, Arora H. Antibody Diversity in Cancer: Translational Implications and Beyond. Vaccines (Basel) 2022; 10:vaccines10081165. [PMID: 35893814 PMCID: PMC9331493 DOI: 10.3390/vaccines10081165] [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: 05/04/2022] [Revised: 06/14/2022] [Accepted: 06/22/2022] [Indexed: 12/10/2022] Open
Abstract
Patients with cancer tend to develop antibodies to autologous proteins. This phenomenon has been observed across multiple cancer types, including bladder, lung, colon, prostate, and melanoma. These antibodies potentially arise due to induced inflammation or an increase in self-antigens. Studies focusing on antibody diversity are particularly attractive for their diagnostic value considering antibodies are present at an early diseased stage, serum samples are relatively easy to obtain, and the prevalence of antibodies is high even when the target antigen is minimally expressed. Conversely, the surveillance of serum proteins in cancer patients is relatively challenging because they often show variability in expression and are less abundant. Moreover, an antibody’s presence is also useful as it suggests the relative immunogenicity of a given antigen. For these reasons, profiling antibodies’ responses is actively considered to detect the spread of antigens following immunotherapy. The current review focuses on expanding the knowledge of antibodies and their diversity, and the impact of antibody diversity on cancer regression and progression.
Collapse
Affiliation(s)
- Raghuram Reddy
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (R.R.); (F.F.); (A.M.)
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Joel Mintz
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, FL 33328, USA;
| | - Roei Golan
- College of Medicine, Florida State University, Tallahassee FL 32304, USA;
| | - Fakiha Firdaus
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (R.R.); (F.F.); (A.M.)
| | - Roxana Ponce
- Department of Biology, Florida International University, Miami, FL 33199, USA;
| | - Derek Van Booven
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33143, USA; (D.V.B.); (I.I.)
| | - Aysswarya Manoharan
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (R.R.); (F.F.); (A.M.)
| | - Isabelle Issa
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33143, USA; (D.V.B.); (I.I.)
| | - Bonnie B. Blomberg
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Himanshu Arora
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (R.R.); (F.F.); (A.M.)
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33143, USA; (D.V.B.); (I.I.)
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- The Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Correspondence:
| |
Collapse
|
18
|
Durkee-Shock J, Lazarski CA, Jensen-Wachspress MA, Zhang A, Son A, Kankate VV, Field NE, Webber K, Lang H, Conway SR, Hanley PJ, Bollard CM, Keller MD, Schwartz DM. Transcriptomic analysis reveals optimal cytokine combinations for SARS-CoV-2-specific T cell therapy products. Mol Ther Methods Clin Dev 2022; 25:439-447. [PMID: 35506060 PMCID: PMC9050197 DOI: 10.1016/j.omtm.2022.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 04/25/2022] [Indexed: 10/29/2022]
Abstract
Adoptive T cell immunotherapy has been used to restore immunity against multiple viral targets in immunocompromised patients after bone-marrow transplantation and has been proposed as a strategy for preventing coronavirus 2019 (COVID-19) in this population. Ideally, expanded severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-virus-specific T cells (CSTs) should demonstrate marked cell expansion, T cell specificity, and CD8+ T cell skewing prior to adoptive transfer. However, current methodologies using IL-4 + IL-7 result in suboptimal specificity, especially in CD8+ cells. Using a microexpansion platform, we screened various cytokine cocktails (IL-4 + IL-7, IL-15, IL-15 + IL-4, IL-15 + IL-6, and IL-15 + IL-7) for the most favorable culture conditions. IL-15 + IL-7 optimally balanced T cell expansion, polyfunctionality, and CD8+ T cell skewing of a final therapeutic T cell product. Additionally, the transcriptomes of CD4+ and CD8+ T cells cultured with IL-15 + IL-7 displayed the strongest induction of antiviral type I interferon (IFN) response genes. Subsequently, microexpansion results were successfully translated to a Good Manufacturing Practice (GMP)-applicable format where IL-15 + IL-7 outperformed IL-4 + IL-7 in specificity and expansion, especially in the desirable CD8+ T cell compartment. These results demonstrate the functional implications of IL-15-, IL-4-, and IL-7-containing cocktails for therapeutic T cell expansion, which could have broad implication for cellular therapy, and pioneer the use of RNA sequencing (RNA-seq) to guide viral-specific T cell (VST) product manufacturing.
Collapse
Affiliation(s)
- Jessica Durkee-Shock
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA.,National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher A Lazarski
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
| | | | - Anqing Zhang
- GW Cancer Center, George Washington University, Washington, DC, USA
| | - Aran Son
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Vaishnavi V Kankate
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Naomi E Field
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Kathleen Webber
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Haili Lang
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Susan R Conway
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA.,GW Cancer Center, George Washington University, Washington, DC, USA.,Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA.,GW Cancer Center, George Washington University, Washington, DC, USA.,Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA
| | - Michael D Keller
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA.,GW Cancer Center, George Washington University, Washington, DC, USA.,Division of Allergy and Immunology, Children's National Hospital, Washington, DC, USA
| | - Daniella M Schwartz
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
19
|
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
|
20
|
Mycophenolate-mofetil/prednisone/tacrolimus. REACTIONS WEEKLY 2022. [PMCID: PMC9051753 DOI: 10.1007/s40278-022-14300-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
21
|
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
|