1
|
Vasileiou S, Kuvalekar M, Velazquez Y, Watanabe A, Leen AM, Gilmore SA. Phenotypic and functional characterization of posoleucel, a multivirus-specific T cell therapy for the treatment and prevention of viral infections in immunocompromised patients. Cytotherapy 2024; 26:869-877. [PMID: 38597860 DOI: 10.1016/j.jcyt.2024.03.012] [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: 10/23/2023] [Revised: 02/09/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Deficits in T cell immunity translate into increased risk of severe viral infection in recipients of solid organ and hematopoietic cell transplants. Thus, therapeutic strategies that employ the adoptive transfer of virus-specific T cells are being clinically investigated to treat and prevent viral diseases in these highly immunocompromised patients. Posoleucel is an off-the-shelf multivirus-specific T cell investigational product for the treatment and prevention of infections due to adenovirus, BK virus, cytomegalovirus, Epstein-Barr virus, human herpesvirus 6 or JC virus. METHODS Herein we perform extensive characterization of the phenotype and functional profile of posoleucel to illustrate the cellular properties that may contribute to its in vivo activity. RESULTS AND CONCLUSIONS Our results demonstrate that posoleucel is enriched for central and effector memory CD4+ and CD8+ T cells with specificity for posoleucel target viruses and expressing a broad repertoire of T cell receptors. Antigen-driven upregulation of cell-surface molecules and production of cytokine and effector molecules indicative of proliferation, co-stimulation, and cytolytic potential demonstrate the specificity of posoleucel and its potential to mount a broad, polyfunctional, and effective Th1-polarized antiviral response upon viral exposure. We also show the low risk for off-target and nonspecific effects as evidenced by the enrichment of posoleucel in memory T cells, low frequency of naive T cells, and lack of demonstrated alloreactivity in vitro. The efficacy of posoleucel is being explored in four placebo-controlled clinical trials in transplant recipients to treat and prevent viral infections (NCT05179057, NCT05305040, NCT04390113, NCT04605484).
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; AlloVir, Inc., Waltham, MA
| | - Manik Kuvalekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX; AlloVir, Inc., Waltham, MA
| | - Yovana Velazquez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX; AlloVir, Inc., Waltham, MA
| | - Ayumi Watanabe
- 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; AlloVir, Inc., Waltham, MA
| | | |
Collapse
|
2
|
Wang J, Xu Z, Lai Y, Zhang Y, Zhang P, Mu Q, Yang S, Sun Y, Sheng L, Ouyang G. Silencing of PD-1 combined with EBV-specific killer T cells for the treatment of EBV-associated B lymphoma. Transl Oncol 2024; 40:101831. [PMID: 38039947 PMCID: PMC10716022 DOI: 10.1016/j.tranon.2023.101831] [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: 09/04/2023] [Revised: 10/31/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023] Open
Abstract
Epstein-Barr Virus (EBV) infection is closely associated with the development of lymphoma, as it plays a significant role in the malignant transformation of lymphocytes. The expression of programmed death-1 (PD-1), which binds to PD-L1 in tumor cells, can lead to immune evasion by lymphoma cells and promote tumor progression. In this study, immortalized B lymphoblastoid cell lines (B-LCLs) positive for EBV-specific proteins were established from human peripheral mononuclear cells (PBMCs) using EBV induction along with CpG-ODN 2006 and cyclosporin A. EBV-specific T cells (EBVST) were generated by multiple immunizations of CD3+ T lymphocytes using irradiated B-LCLs. Flow cytometry analysis confirmed the activation of EBVST through the detection of CD3+, CD4+, and CD8+ markers. Co-incubation of EBVST with EBV-positive B lymphocyte cell lines resulted in the secretion of perforin by EBVST, leading to granzyme B-mediated cell death and an increase in LDH levels. Silencing PD-1 in EBVST cells enhanced perforin production, increased granzyme B release, and upregulated cell death in co-incubated B lymphocytes. In a nude mice tumor transplantation model, silencing PD-1 in combination with EBV-specific killer T cells exhibited the maximum inhibition of B-lymphoblastoma. This treatment upregulated the expression of proteins associated with apoptosis and immune response, while inhibiting anti-apoptotic protein expression in tumor tissues. Silencing PD-1 also increased the infiltration of EBV-specific killer T cells in the tumor tissues. Overall, PD-1 silencing enhanced the tumor targeting effect of EBV-specific killer T cells on EBV-infected B lymphocytes and attenuated the immune escape effect mediated by the PD-1 pathway.
Collapse
Affiliation(s)
- Jiaping Wang
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Zhijuan Xu
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Yanli Lai
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Yanli Zhang
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Ping Zhang
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Qitian Mu
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Shujun Yang
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Yongcheng Sun
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Lixia Sheng
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China.
| | - Guifang Ouyang
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China.
| |
Collapse
|
3
|
O’Reilly RJ, Prockop S, Oved JH. Virus-specific T-cells from third party or transplant donors for treatment of EBV lymphoproliferative diseases arising post hematopoietic cell or solid organ transplantation. Front Immunol 2024; 14:1290059. [PMID: 38274824 PMCID: PMC10808771 DOI: 10.3389/fimmu.2023.1290059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
EBV+ lymphomas constitute a significant cause of morbidity and mortality in recipients of allogeneic hematopoietic cell (HCT) and solid organ transplants (SOT). Phase I and II trials have shown that in HCT recipients, adoptive transfer of EBV-specific T-cells from the HCT donor can safely induce durable remissions of EBV+ lymphomas including 70->90% of patients who have failed to respond to treatment with Rituximab. More recently, EBV-specific T-cells generated from allogeneic 3rd party donors have also been shown to induce durable remission of EBV+ lymphomas in Rituximab refractory HCT and SOT recipients. In this review, we compare results of phase I and II trials of 3rd party and donor derived EBV-specific T-cells. We focus on the attributes and limitations of each product in terms of access, safety, responses achieved and durability. The limited data available regarding donor and host factors contributing to T cell persistence is also described. We examine factors contributing to treatment failures and approaches to prevent or salvage relapse. Lastly, we summarize strategies to further improve results for virus-specific immunotherapies for post-transplant EBV lymphomas.
Collapse
Affiliation(s)
- Richard J. O’Reilly
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Susan Prockop
- Pediatric Stem Cell Transplantation, Boston Children’s Hospital/Dana-Farber Cancer Institute, Boston, MA, United States
| | - Joseph H. Oved
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
4
|
Jalili A, Hajifathali A, Mohammadian M, Sankanian G, Sayahinouri M, Dehghani Ghorbi M, Roshandel E, Aghdami N. Virus-Specific T Cells: Promising Adoptive T Cell Therapy Against Infectious Diseases Following Hematopoietic Stem Cell Transplantation. Adv Pharm Bull 2023; 13:469-482. [PMID: 37646062 PMCID: PMC10460803 DOI: 10.34172/apb.2023.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 09/25/2022] [Accepted: 11/02/2022] [Indexed: 09/01/2023] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a life-saving therapy for various hematologic disorders. Due to the bone marrow suppression and its long recovery period, secondary infections, like cytomegalovirus (CMV), Epstein-Bar virus (EBV), and adenovirus (AdV), are the leading causes of morbidity and mortality in HSCT cases. Drug resistance to the antiviral pharmacotherapies makes researchers develop adoptive T cell therapies like virus-specific T cell therapy. These studies have faced major challenges such as finding the most effective T cell expansion methods, isolating the expected subtype, defining the functionality of the end-cell population, product quality control, and clinical complications after the injection. This review discusses the viral infections after HSCT, T cells characteristics during chronic viral infection, application of virus-specific T cells (VSTs) for refractory infections, standard methods for producing VSTs and their limitation, clinical experiences on VSTs, focusing on outcomes and side effects that can be helpful in decision-making for patients and further researches.
Collapse
Affiliation(s)
- Arsalan Jalili
- Department of Applied Cell Sciences, Faculty of Basic Sciences and Advanced Medical Technologies, Royan Institute, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mozhdeh Mohammadian
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ghazaleh Sankanian
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Sayahinouri
- Department of Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Parvaz Research Ideas Supporter institute, Tehran, Iran
| | - Mahmoud Dehghani Ghorbi
- Department of Internal Medicine, Imam Hossein Hospital, School of Medicine Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
5
|
Epstein-Barr Virus-Positive Lymphomas Exploit Ectonucleotidase Activity To Limit Immune Responses and Prevent Cell Death. mBio 2023; 14:e0345922. [PMID: 36786572 PMCID: PMC10127690 DOI: 10.1128/mbio.03459-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
Epstein-Barr virus (EBV) is a cancer-associated virus that infects more than 90% of adults. Unfortunately, many EBV-driven malignancies, including numerous B cell lymphomas, are highly aggressive and lack acceptable therapeutic outcomes. The concentrations of extracellular purines, namely, ATP and adenosine, are highly dysregulated in the tumor microenvironment and significantly impact the degree of immune responses to the tumor. Additionally, many tumor cells adapt to this dysregulation by overexpressing one or more ectonucleotidases, enzymes that degrade extracellular nucleotides to nucleosides. The degradation of immunostimulatory extracellular ATP to immunosuppressive adenosine through ectonucleotidase activity is one example of tumor cell exploitation of the purinergic signaling pathway. As such, preclinical studies targeting the purinergic signaling pathway have found it to be a promising immunotherapeutic target for the treatment of solid tumors; however, the extent to which purinergic signaling impacts the development and survival of EBV+ B cell lymphoma remains unstudied. Here, we demonstrate robust ectonucleotidase expression on multiple types of EBV-positive B cell non-Hodgkin lymphoma (NHL). Furthermore, the presence of high concentrations of extracellular ATP resulted in the expression of lytic viral proteins and exhibited cytotoxicity toward EBV+ B cell lines, particularly when CD39 was inhibited. Inhibition of CD39 also significantly prolonged survival in an aggressive cord blood humanized mouse model of EBV-driven lymphomagenesis and was correlated with an enhanced inflammatory immune response and reduced tumor burden. Taken together, these data suggest that EBV+ B cell lymphomas exploit ectonucleotidase activity to circumvent ATP-mediated inflammation and cell death. IMPORTANCE EBV is a ubiquitous pathogen responsible for significant global lymphoma burden, including Hodgkin lymphoma, numerous non-Hodgkin B, T, and NK cell lymphomas, and lymphoproliferative disorders. EBV is also associated with epithelial cancers and autoimmune diseases, such as multiple sclerosis. Many of these diseases are highly aggressive and exhibit poor outcomes. As such, new treatments for EBV-driven cancers have the potential to benefit a large number of patients. We use in vitro and in vivo models to demonstrate the therapeutic potential of targeting the purinergic signaling pathway in the context of EBV-driven B cell lymphoma. These findings lend credence to the manipulation of purinergic signaling as a viable therapeutic approach to EBV+ malignancies and support the feasibility of immunotherapeutic treatments for viral lymphoma.
Collapse
|
6
|
Motta CM, Keller MD, Bollard CM. Applications of Virus specific T cell Therapies Post BMT. Semin Hematol 2022; 60:10-19. [PMID: 37080705 DOI: 10.1053/j.seminhematol.2022.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) has been used as a curative standard of care for moderate to severe primary immunodeficiency disorders as well as relapsed hematologic malignancies for over 50 years [1,2]. However, chronic and refractory viral infections remain a leading cause of morbidity and mortality in the immune deficient period following HSCT, where use of available antiviral pharmacotherapies is limited by toxicity and emerging resistance [3]. Adoptive immunotherapy using virus-specific T cells (VSTs) has been explored for over 2 decades [4,5] in patients post-HSCT and has been shown prior phase I-II studies to be safe and effective for treatment or preventions of viral infections including cytomegalovirus, Epstein-Barr virus, BK virus, and adenovirus with minimal toxicity and low risk of graft vs host disease [6-9]. This review summarizes methodologies to generate VSTs the clinical results utilizing VST therapeutics and the challenges and future directions for the field.
Collapse
|
7
|
Khatamzas E, Antwerpen MH, Rehn A, Graf A, Hellmuth JC, Hollaus A, Mohr AW, Gaitzsch E, Weiglein T, Georgi E, Scherer C, Stecher SS, Gruetzner S, Blum H, Krebs S, Reischer A, Leutbecher A, Subklewe M, Dick A, Zange S, Girl P, Müller K, Weigert O, Hopfner KP, Stemmler HJ, von Bergwelt-Baildon M, Keppler OT, Wölfel R, Muenchhoff M, Moosmann A. Accumulation of mutations in antibody and CD8 T cell epitopes in a B cell depleted lymphoma patient with chronic SARS-CoV-2 infection. Nat Commun 2022; 13:5586. [PMID: 36151076 PMCID: PMC9508331 DOI: 10.1038/s41467-022-32772-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 08/15/2022] [Indexed: 11/29/2022] Open
Abstract
Antibodies against the spike protein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) can drive adaptive evolution in immunocompromised patients with chronic infection. Here we longitudinally analyze SARS-CoV-2 sequences in a B cell-depleted, lymphoma patient with chronic, ultimately fatal infection, and identify three mutations in the spike protein that dampen convalescent plasma-mediated neutralization of SARS-CoV-2. Additionally, four mutations emerge in non-spike regions encoding three CD8 T cell epitopes, including one nucleoprotein epitope affected by two mutations. Recognition of each mutant peptide by CD8 T cells from convalescent donors is reduced compared to its ancestral peptide, with additive effects resulting from double mutations. Querying public SARS-CoV-2 sequences shows that these mutations have independently emerged as homoplasies in circulating lineages. Our data thus suggest that potential impacts of CD8 T cells on SARS-CoV-2 mutations, at least in those with humoral immunodeficiency, warrant further investigation to inform on vaccine design.
Collapse
Affiliation(s)
- Elham Khatamzas
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.
- Division of Infectious Diseases and Tropical Medicine, Center for Infectious Diseases, Heidelberg Hospital, Heidelberg, Germany.
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.
| | - Markus H Antwerpen
- Bundeswehr, Institute of Microbiology Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Alexandra Rehn
- Bundeswehr, Institute of Microbiology Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Alexander Graf
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Johannes Christian Hellmuth
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Alexandra Hollaus
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Anne-Wiebe Mohr
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Erik Gaitzsch
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Tobias Weiglein
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Enrico Georgi
- Bundeswehr, Institute of Microbiology Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Clemens Scherer
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Stephanie-Susanne Stecher
- Department of Medicine II, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Stefanie Gruetzner
- Institute for Transfusion Medicine and Haemostasis, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Anna Reischer
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Alexandra Leutbecher
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Andrea Dick
- Laboratory for Immunogenetics, University of Munich, LMU, Munich, Germany
| | - Sabine Zange
- Bundeswehr, Institute of Microbiology Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Philipp Girl
- Bundeswehr, Institute of Microbiology Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Katharina Müller
- Bundeswehr, Institute of Microbiology Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Oliver Weigert
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich, Germany
| | - Karl-Peter Hopfner
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hans-Joachim Stemmler
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich, Germany
| | - Oliver T Keppler
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
- Max von Pettenkofer Institute & Gene Center, Virology, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany
| | - Roman Wölfel
- Bundeswehr, Institute of Microbiology Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Maximilian Muenchhoff
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
- Max von Pettenkofer Institute & Gene Center, Virology, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany
| | - Andreas Moosmann
- Department of Medicine III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| |
Collapse
|
8
|
Cirac A, Poirey R, Dieckmeyer M, Witter K, Delecluse HJ, Behrends U, Mautner J. Immunoinformatic Analysis Reveals Antigenic Heterogeneity of Epstein-Barr Virus Is Immune-Driven. Front Immunol 2021; 12:796379. [PMID: 34975903 PMCID: PMC8716887 DOI: 10.3389/fimmu.2021.796379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 11/30/2021] [Indexed: 12/05/2022] Open
Abstract
Whole genome sequencing of Epstein-Barr virus (EBV) isolates from around the world has uncovered pervasive strain heterogeneity, but the forces driving strain diversification and the impact on immune recognition remained largely unknown. Using a data mining approach, we analyzed more than 300 T-cell epitopes in 168 published EBV strains. Polymorphisms were detected in approximately 65% of all CD8+ and 80% of all CD4+ T-cell epitopes and these numbers further increased when epitope flanking regions were included. Polymorphisms in CD8+ T-cell epitopes often involved MHC anchor residues and resulted in changes of the amino acid subgroup, suggesting that only a limited number of conserved T-cell epitopes may represent generic target antigens against different viral strains. Although considered the prototypic EBV strain, the rather low degree of overlap with most other viral strains implied that B95.8 may not represent the ideal reference strain for T-cell epitopes. Instead, a combinatorial library of consensus epitopes may provide better targets for diagnostic and therapeutic purposes when the infecting strain is unknown. Polymorphisms were significantly enriched in epitope versus non-epitope protein sequences, implicating immune selection in driving strain diversification. Remarkably, CD4+ T-cell epitopes in EBNA2, EBNA-LP, and the EBNA3 family appeared to be under negative selection pressure, hinting towards a beneficial role of immune responses against these latency type III antigens in virus biology. These findings validate this immunoinformatics approach for providing novel insight into immune targets and the intricate relationship of host defense and virus evolution that may also pertain to other pathogens.
Collapse
Affiliation(s)
- Ana Cirac
- Children’s Hospital, School of Medicine, Technische Universität München, Munich, Germany
- German Centre for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Remy Poirey
- German Cancer Research Center (DKFZ) Unit F100 and Institut National de la Santé et de la Recherche Médicale Unit U1074, Heidelberg, Germany
| | - Michael Dieckmeyer
- Department of Diagnostic and Interventional Neuroradiology, Technische Universität München, Munich, Germany
| | - Klaus Witter
- Laboratory of Immunogenetics, Ludwig-Maximilians-Universität, München, Germany
| | - Henri-Jacques Delecluse
- German Cancer Research Center (DKFZ) Unit F100 and Institut National de la Santé et de la Recherche Médicale Unit U1074, Heidelberg, Germany
| | - Uta Behrends
- Children’s Hospital, School of Medicine, Technische Universität München, Munich, Germany
- German Centre for Infection Research (DZIF), partner site Munich, Munich, Germany
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
| | - Josef Mautner
- Children’s Hospital, School of Medicine, Technische Universität München, Munich, Germany
- German Centre for Infection Research (DZIF), partner site Munich, Munich, Germany
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- *Correspondence: Josef Mautner,
| |
Collapse
|
9
|
Epstein Barr virus-positive B-cell lymphoma is highly vulnerable to MDM2 inhibitors in vivo. Blood Adv 2021; 6:891-901. [PMID: 34861697 PMCID: PMC8945299 DOI: 10.1182/bloodadvances.2021006156] [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: 09/14/2021] [Accepted: 11/25/2021] [Indexed: 11/20/2022] Open
Abstract
MDM2 inhibitors have potent in vivo activity against and could be a novel therapy for EBV-positive B-cell lymphoma. EBV positivity or loss of BCL6 expression can be a potential predictive biomarker for response to MDM2 inhibitors in patients with lymphoma
Epstein-Barr virus–positive (EBV-positive) B-cell lymphomas are common in immunocompromised patients and remain an unmet medical need. Here we report that MDM2 inhibitors (MDM2is) navtemadlin and idasanutlin have potent in vivo activity in EBV-positive B-cell lymphoma established in immunocompromised mice. Tumor regression was observed in all 5 EBV-positive xenograft–associated B-cell lymphomas treated with navtemadlin or idasanutlin. Molecular characterization showed that treatment with MDM2is resulted in activation of p53 pathways and downregulation of cell cycle effectors in human lymphoma cell lines that were either EBV-positive or had undetectable expression of BCL6, a transcriptional inhibitor of the TP53 gene. Moreover, treatment with navtemadlin resulted in tumor regression and prevented systemic dissemination of EBV-positive lymphoma derived from 2 juvenile patients with posttransplant lymphoproliferative diseases, including 1 whose tumor was resistant to virus-specific T-cell therapy. These results provide proof-of-concept for targeted therapy of EBV-positive lymphoma with MDM2is and the feasibility of using EBV infection or loss of BCL6 expression to identify responders to MDM2is.
Collapse
|
10
|
Agrawal V, Gbolahan OB, Stahl M, Zeidan AM, Zaid MA, Farag SS, Konig H. Vaccine and Cell-based Therapeutic Approaches in Acute Myeloid Leukemia. Curr Cancer Drug Targets 2021; 20:473-489. [PMID: 32357813 DOI: 10.2174/1568009620666200502011059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 03/05/2020] [Accepted: 03/29/2020] [Indexed: 12/13/2022]
Abstract
Over the past decade, our increased understanding of the interactions between the immune system and cancer cells has led to paradigm shifts in the clinical management of solid and hematologic malignancies. The incorporation of immune-targeted strategies into the treatment landscape of acute myeloid leukemia (AML), however, has been challenging. While this is in part due to the inability of the immune system to mount an effective tumor-specific immunogenic response against the heterogeneous nature of AML, the decreased immunogenicity of AML cells also represents a major obstacle in the effort to design effective immunotherapeutic strategies. In fact, AML cells have been shown to employ sophisticated escape mechanisms to evade elimination, such as direct immunosuppression of natural killer cells and decreased surface receptor expression leading to impaired recognition by the immune system. Yet, cellular and humoral immune reactions against tumor-associated antigens (TAA) of acute leukemia cells have been reported and the success of allogeneic stem cell transplantation and monoclonal antibodies in the treatment of AML clearly provides proof that an immunotherapeutic approach is feasible in the management of this disease. This review discusses the recent progress and persisting challenges in cellular immunotherapy for patients with AML.
Collapse
Affiliation(s)
- Vaibhav Agrawal
- Department of Medicine, Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Olumide B Gbolahan
- Division of Hematology and Oncology, University of Alabama School of Medicine, Birmingham, AL 35294, United States
| | - Maximilian Stahl
- Department of Medicine, Division of Hematology and Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT 06510, United States
| | - Mohammad Abu Zaid
- Department of Medicine, Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Sherif S Farag
- Department of Medicine, Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Heiko Konig
- Department of Medicine, Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| |
Collapse
|
11
|
Shahid S, Prockop SE. Epstein-Barr virus-associated post-transplant lymphoproliferative disorders: beyond chemotherapy treatment. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:646-664. [PMID: 34485854 PMCID: PMC8415721 DOI: 10.20517/cdr.2021.34] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/10/2021] [Accepted: 05/19/2021] [Indexed: 12/30/2022]
Abstract
Post-transplant lymphoproliferative disorder (PTLD) is a rare but life-threatening complication of both allogeneic solid organ (SOT) and hematopoietic cell transplantation (HCT). The histology of PTLD ranges from benign polyclonal lymphoproliferation to a lesion indistinguishable from classic monoclonal lymphoma. Most commonly, PTLDs are Epstein-Barr virus (EBV) positive and result from loss of immune surveillance over EBV. Treatment for PTLD differs from the treatment for typical non-Hodgkin lymphoma because prognostic factors are different, resistance to treatment is unique, and there are specific concerns for organ toxicity. While recipients of HCT have a limited time during which they are at risk for this complication, recipients of SOT have a lifelong requirement for immunosuppression, so approaches that limit compromising or help restore immune surveillance are of high interest. Furthermore, while EBV-positive and EBV-negative PTLDs are not intrinsically resistant to chemotherapy, the poor tolerance of chemotherapy in the post-transplant setting makes it essential to minimize potential treatment-related toxicities and explore alternative treatment algorithms. Therefore, reduced-toxicity approaches such as single-agent CD20 monoclonal antibodies or bortezomib, reduced dosing of standard chemotherapeutic agents, and non-chemotherapy-based approaches such as cytotoxic T cells have all been explored. Here, we review the chemotherapy and non-chemotherapy treatment landscape for PTLD.
Collapse
Affiliation(s)
| | - Susan E. Prockop
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
12
|
Karavalakis G, Yannaki E, Papadopoulou A. Reinforcing the Immunocompromised Host Defense against Fungi: Progress beyond the Current State of the Art. J Fungi (Basel) 2021; 7:jof7060451. [PMID: 34204025 PMCID: PMC8228486 DOI: 10.3390/jof7060451] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Despite the availability of a variety of antifungal drugs, opportunistic fungal infections still remain life-threatening for immunocompromised patients, such as those undergoing allogeneic hematopoietic cell transplantation or solid organ transplantation. Suboptimal efficacy, toxicity, development of resistant variants and recurrent episodes are limitations associated with current antifungal drug therapy. Adjunctive immunotherapies reinforcing the host defense against fungi and aiding in clearance of opportunistic pathogens are continuously gaining ground in this battle. Here, we review alternative approaches for the management of fungal infections going beyond the state of the art and placing an emphasis on fungus-specific T cell immunotherapy. Harnessing the power of T cells in the form of adoptive immunotherapy represents the strenuous protagonist of the current immunotherapeutic approaches towards combating invasive fungal infections. The progress that has been made over the last years in this field and remaining challenges as well, will be discussed.
Collapse
Affiliation(s)
- Georgios Karavalakis
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, 57010 Thessaloniki, Greece; (G.K.); (E.Y.)
| | - Evangelia Yannaki
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, 57010 Thessaloniki, Greece; (G.K.); (E.Y.)
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Anastasia Papadopoulou
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, 57010 Thessaloniki, Greece; (G.K.); (E.Y.)
- Correspondence: ; Tel.: +30-2313-307-693; Fax: +30-2313-307-521
| |
Collapse
|
13
|
Heslop HE, Sharma S, Rooney CM. Adoptive T-Cell Therapy for Epstein-Barr Virus-Related Lymphomas. J Clin Oncol 2021; 39:514-524. [PMID: 33434061 DOI: 10.1200/jco.20.01709] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX
| | - Sandhya Sharma
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, 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
| |
Collapse
|
14
|
Prockop S, Doubrovina E, Suser S, Heller G, Barker J, Dahi P, Perales MA, Papadopoulos E, Sauter C, Castro-Malaspina H, Boulad F, Curran KJ, Giralt S, Gyurkocza B, Hsu KC, Jakubowski A, Hanash AM, Kernan NA, Kobos R, Koehne G, Landau H, Ponce D, Spitzer B, Young JW, Behr G, Dunphy M, Haque S, Teruya-Feldstein J, Arcila M, Moung C, Hsu S, Hasan A, O'Reilly RJ. Off-the-shelf EBV-specific T cell immunotherapy for rituximab-refractory EBV-associated lymphoma following transplantation. J Clin Invest 2020; 130:733-747. [PMID: 31689242 DOI: 10.1172/jci121127] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 10/22/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUNDAdoptive transfer of donor-derived EBV-specific cytotoxic T-lymphocytes (EBV-CTLs) can eradicate EBV-associated lymphomas (EBV-PTLD) after transplantation of hematopoietic cell (HCT) or solid organ (SOT) but is unavailable for most patients.METHODSWe developed a third-party, allogeneic, off-the-shelf bank of 330 GMP-grade EBV-CTL lines from specifically consented healthy HCT donors. We treated 46 recipients of HCT (n = 33) or SOT (n = 13) with established EBV-PTLD, who had failed rituximab therapy, with third-party EBV-CTLs. Treatment cycles consisted of 3 weekly infusions of EBV-CTLs and 3 weeks of observation.RESULTSEBV-CTLs did not induce significant toxicities. One patient developed grade I skin graft-versus-host disease. Complete remission (CR) or sustained partial remission (PR) was achieved in 68% of HCT recipients and 54% of SOT recipients. For patients who achieved CR/PR or stable disease after cycle 1, one year overall survival was 88.9% and 81.8%, respectively. In addition, 3 of 5 recipients with POD after a first cycle who received EBV-CTLs from a different donor achieved CR or durable PR (60%) and survived longer than 1 year. Maximal responses were achieved after a median of 2 cycles.CONCLUSIONThird-party EBV-CTLs of defined HLA restriction provide safe, immediately accessible treatment for EBV-PTLD. Secondary treatment with EBV-CTLs restricted by a different HLA allele (switch therapy) can also induce remissions if initial EBV-CTLs are ineffective. These results suggest a promising potential therapy for patients with rituximab-refractory EBV-associated lymphoma after transplantation.TRIAL REGISTRATIONPhase II protocols (NCT01498484 and NCT00002663) were approved by the Institutional Review Board at Memorial Sloan Kettering Cancer Center, the FDA, and the National Marrow Donor Program.FUNDINGThis work was supported by NIH grants CA23766 and R21CA162002, the Aubrey Fund, the Claire Tow Foundation, the Major Family Foundation, the Max Cure Foundation, the Richard "Rick" J. Eisemann Pediatric Research Fund, the Banbury Foundation, the Edith Robertson Foundation, and the Larry Smead Foundation. Atara Biotherapeutics licensed the bank of third-party EBV-CTLs from Memorial Sloan Kettering Cancer Center in June 2015.
Collapse
Affiliation(s)
- Susan Prockop
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pediatric Hematology/Oncology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, New York, USA
| | - Ekaterina Doubrovina
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Center for Immune Cellular Therapy
| | - Stephanie Suser
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Juliet Barker
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Parastoo Dahi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Miguel A Perales
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Esperanza Papadopoulos
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Craig Sauter
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Hugo Castro-Malaspina
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Farid Boulad
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pediatric Hematology/Oncology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, New York, USA
| | - Kevin J Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pediatric Hematology/Oncology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, New York, USA
| | - Sergio Giralt
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Boglarka Gyurkocza
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Katharine C Hsu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Ann Jakubowski
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Alan M Hanash
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Nancy A Kernan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pediatric Hematology/Oncology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, New York, USA
| | - Rachel Kobos
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Jansen Pharmaceuticals, Raritan, New Jersey, USA
| | - Guenther Koehne
- Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Heather Landau
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Doris Ponce
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Barbara Spitzer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pediatric Hematology/Oncology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, New York, USA
| | - James W Young
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Gerald Behr
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mark Dunphy
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sofia Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Julie Teruya-Feldstein
- Department of Pathology, Icahn School of Medicine, Mount Sinai Health System, New York, New York, USA
| | - Maria Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Christine Moung
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Susan Hsu
- American Red Cross, Philadelphia, Pennsylvania, USA
| | - Aisha Hasan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,GlaxoSmithKline, Oncology, Collegeville, Pennsylvania, USA
| | - Richard J O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pediatric Hematology/Oncology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
15
|
Münz C. Probing Reconstituted Human Immune Systems in Mice With Oncogenic γ-Herpesvirus Infections. Front Immunol 2020; 11:581419. [PMID: 33013936 PMCID: PMC7509489 DOI: 10.3389/fimmu.2020.581419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022] Open
Abstract
Mice with reconstituted human immune systems can mount cell-mediated immune responses against the human tumor viruses Epstein Barr virus (EBV) and Kaposi sarcoma associated herpesvirus (KSHV). Primarily cytotoxic lymphocytes protect the vast majority of persistently infected carriers of these tumor viruses from the respective malignancies for life. Thus, EBV and KSHV infection can teach us how this potent immune control is induced, what phenotype and functions characterize the protective lymphocyte compartments and if similar immune responses could be induced by vaccination. This review will summarize similarities and differences between EBV and KSHV associated pathologies and their immune control in patients and mice with reconstituted human immune systems. Furthermore, it will high-light which aspects of the near perfect immune control can be modeled in the latter preclinical animal models and discuss their relevance for cancer immunology in general.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, Zurich, Switzerland
| |
Collapse
|
16
|
O'Reilly RJ, Prockop S, Hasan A, Doubrovina E. Therapeutic advantages provided by banked virus-specific T-cells of defined HLA-restriction. Bone Marrow Transplant 2020; 54:759-764. [PMID: 31431697 DOI: 10.1038/s41409-019-0614-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We have developed banks of EBV and CMV-specific T-cell lines generated from healthy seropositive third party donors and characterized them as to their HLA type, virus specificity, lack of alloreactivity, and HLA restriction. We here summarize results of studies employing these immediately accessible, broadly-applicable third party virus-specific T-cells for adoptive therapy of EBV lymphomas and CMV infections in allo-HCT recipients. We describe the characteristics contributing to their safety. We also discuss several distinctive advantages of banked third party virus-specific T-cells selected on the basis of their HLA restriction, particularly in the treatment of Rituximab-non-responsive EBV+ lymphomas and drug refractory CMV infections complicating HLA non-identical transplants.
Collapse
Affiliation(s)
- Richard J O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Susan Prockop
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aisha Hasan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ekaterina Doubrovina
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Viral and fungal infections cause significant morbidity and mortality following hematopoietic stem-cell transplantation (HSCT), primarily due to the prolonged and complex immunodeficient state that results from conditioning chemo-radiotherapy and subsequent prophylaxis of graft vs. host disease. Although currently available antimicrobial pharmacotherapies have demonstrated short-term efficacy, their toxicities often preclude long-term use, and cessation if frequently associated with recurrent infection. Adoptive cell therapy (ACT) offers the potential to more rapidly reconstitute antimicrobial immune responses in the posttransplant setting. RECENT FINDINGS Traditional approaches to manufacture of adoptive T-cell therapies are time consuming and limited to single pathogen specificity. Recent advances in the understanding of immunogenic epitopes, improved methods for pathogen-specific T-cell isolation and cultureware technologies is allowing for rapid generation of ACTs for clinical use. SUMMARY The current review summarizes the potential infectious targets and manufacturing methodologies for ACTs and contrasts their clinical efficacy and safety to currently available pharmacotherapies for patients recovering after HSCT.
Collapse
|
18
|
Caduff N, McHugh D, Murer A, Rämer P, Raykova A, Landtwing V, Rieble L, Keller CW, Prummer M, Hoffmann L, Lam JKP, Chiang AKS, Raulf F, Azzi T, Berger C, Rubic-Schneider T, Traggiai E, Lünemann JD, Kammüller M, Münz C. Immunosuppressive FK506 treatment leads to more frequent EBV-associated lymphoproliferative disease in humanized mice. PLoS Pathog 2020; 16:e1008477. [PMID: 32251475 PMCID: PMC7162544 DOI: 10.1371/journal.ppat.1008477] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 04/16/2020] [Accepted: 03/15/2020] [Indexed: 12/13/2022] Open
Abstract
Post-transplant lymphoproliferative disorder (PTLD) is a potentially fatal complication after organ transplantation frequently associated with the Epstein-Barr virus (EBV). Immunosuppressive treatment is thought to allow the expansion of EBV-infected B cells, which often express all eight oncogenic EBV latent proteins. Here, we assessed whether HLA-A2 transgenic humanized NSG mice treated with the immunosuppressant FK506 could be used to model EBV-PTLD. We found that FK506 treatment of EBV-infected mice led to an elevated viral burden, more frequent tumor formation and diminished EBV-induced T cell responses, indicative of reduced EBV-specific immune control. EBV latency III and lymphoproliferation-associated cellular transcripts were up-regulated in B cells from immunosuppressed animals, akin to the viral and host gene expression pattern found in EBV-PTLD. Utilizing an unbiased gene expression profiling approach, we identified genes differentially expressed in B cells of EBV-infected animals with and without FK506 treatment. Upon investigating the most promising candidates, we validated sCD30 as a marker of uncontrolled EBV proliferation in both humanized mice and in pediatric patients with EBV-PTLD. High levels of sCD30 have been previously associated with EBV-PTLD in patients. As such, we believe that humanized mice can indeed model aspects of EBV-PTLD development and may prove useful for the safety assessment of immunomodulatory therapies.
Collapse
Affiliation(s)
- Nicole Caduff
- University of Zurich, Viral Immunobiology, Institute of Experimental Immunology, Zurich, Switzerland
| | - Donal McHugh
- University of Zurich, Viral Immunobiology, Institute of Experimental Immunology, Zurich, Switzerland
| | - Anita Murer
- University of Zurich, Viral Immunobiology, Institute of Experimental Immunology, Zurich, Switzerland
| | - Patrick Rämer
- University of Zurich, Viral Immunobiology, Institute of Experimental Immunology, Zurich, Switzerland
| | - Ana Raykova
- University of Zurich, Viral Immunobiology, Institute of Experimental Immunology, Zurich, Switzerland
| | - Vanessa Landtwing
- University of Zurich, Viral Immunobiology, Institute of Experimental Immunology, Zurich, Switzerland
| | - Lisa Rieble
- University of Zurich, Viral Immunobiology, Institute of Experimental Immunology, Zurich, Switzerland
| | - Christian W Keller
- University Hospital of Münster, Department of Neurology with Institute of Translational Neurology, Münster, Germany
| | - Michael Prummer
- Nexus Personalized Health Technologies, ETH Zurich, Zurich Switzerland, and Swiss Institute for Bioinformatics (SIB), Zurich, Switzerland
| | | | - Janice K P Lam
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Alan K S Chiang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Friedrich Raulf
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Tarik Azzi
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Christoph Berger
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland
| | | | | | - Jan D Lünemann
- University Hospital of Münster, Department of Neurology with Institute of Translational Neurology, Münster, Germany
| | | | - Christian Münz
- University of Zurich, Viral Immunobiology, Institute of Experimental Immunology, Zurich, Switzerland
| |
Collapse
|
19
|
Maloney EM, Busque VA, Hui ST, Toh J, Fernandez-Vina M, Krams SM, Esquivel CO, Martinez OM. Genomic variations in EBNA3C of EBV associate with posttransplant lymphoproliferative disorder. JCI Insight 2020; 5:131644. [PMID: 32213705 DOI: 10.1172/jci.insight.131644] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 02/26/2020] [Indexed: 01/22/2023] Open
Abstract
Epstein-Barr Virus (EBV) is a ubiquitous virus linked to a variety of lymphoid and epithelial malignancies. In solid organ and hematopoietic stem cell transplant recipients, EBV is causally associated with posttransplant lymphoproliferative disorder (PTLD), a group of heterogeneous lymphoid diseases. EBV+ B cell lymphomas that develop in the context of PTLD are generally attributed to the immunosuppression required to promote graft survival, but little is known regarding the role of EBV genome diversity in the development of malignancy. We deep-sequenced the EBV genome from the peripheral blood of 18 solid organ transplant recipients, including 6 PTLD patients. Sequences from 6 EBV+ spontaneous lymphoblastoid B cell lines (SLCL) were similarly analyzed. The EBV genome from PTLD patients had a significantly greater number of variations than EBV from transplant recipients without PTLD. Importantly, there were 15 nonsynonymous variations, including 8 in the latent cycle gene EBNA3C that were associated with the development of PTLD. One of the nonsynonymous variations in EBNA3C is located within a previously defined T cell epitope. These findings suggest that variations in the EBV genome can contribute to the pathogenesis of PTLD.
Collapse
Affiliation(s)
| | - Vincent A Busque
- Division of Abdominal Transplantation, Department of Surgery, and
| | - Sin Ting Hui
- Division of Abdominal Transplantation, Department of Surgery, and
| | | | - Marcelo Fernandez-Vina
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Sheri M Krams
- Stanford Immunology.,Division of Abdominal Transplantation, Department of Surgery, and
| | | | - Olivia M Martinez
- Stanford Immunology.,Division of Abdominal Transplantation, Department of Surgery, and
| |
Collapse
|
20
|
McHugh D, Caduff N, Murer A, Engelmann C, Deng Y, Zdimerova H, Zens K, Chijioke O, Münz C. Infection and immune control of human oncogenic γ-herpesviruses in humanized mice. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180296. [PMID: 30955487 DOI: 10.1098/rstb.2018.0296] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epstein-Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus (KSHV) comprise the oncogenic human γ-herpesvirus family and are responsible for 2-3% of all tumours in man. With their prominent growth-transforming abilities and high prevalence in the human population, these pathogens have probably shaped the human immune system throughout evolution for near perfect immune control of the respective chronic infections in the vast majority of healthy pathogen carriers. The exclusive tropism of EBV and KSHV for humans has, however, made it difficult in the past to study their infection, tumourigenesis and immune control in vivo. Mice with reconstituted human immune system components (humanized mice) support replication of both viruses with both persisting latent and productive lytic infection. Moreover, B-cell lymphomas can be induced by EBV alone and KSHV co-infection with gene expression hallmarks of human malignancies that are associated with both viruses. Furthermore, cell-mediated immune control by primarily cytotoxic lymphocytes is induced upon infection and can be probed for its functional characteristics as well as putative requirements for its priming. Insights that have been gained from this model and remaining questions will be discussed in this review. This article is part of the theme issue 'Silent cancer agents: multi-disciplinary modelling of human DNA oncoviruses'.
Collapse
Affiliation(s)
- Donal McHugh
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Nicole Caduff
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Anita Murer
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Christine Engelmann
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Yun Deng
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Hana Zdimerova
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Kyra Zens
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Obinna Chijioke
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| |
Collapse
|
21
|
Keller MD, Bollard CM. Virus-specific T-cell therapies for patients with primary immune deficiency. Blood 2020; 135:620-628. [PMID: 31942610 PMCID: PMC7046606 DOI: 10.1182/blood.2019000924] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 09/23/2019] [Indexed: 02/07/2023] Open
Abstract
Viral infections are common and are potentially life-threatening in patients with moderate to severe primary immunodeficiency disorders. Because T-cell immunity contributes to the control of many viral pathogens, adoptive immunotherapy with virus-specific T cells (VSTs) has been a logical and effective way of combating severe viral disease in immunocompromised patients in multiple phase 1 and 2 clinical trials. Common viral targets include cytomegalovirus, Epstein-Barr virus, and adenovirus, though recent published studies have successfully targeted additional pathogens, including HHV6, BK virus, and JC virus. Though most studies have used VSTs derived from allogenic stem cell donors, the use of banked VSTs derived from partially HLA-matched donors has shown efficacy in multicenter settings. Hence, this approach could shorten the time for patients to receive VST therapy thus improving accessibility. In this review, we discuss the usage of VSTs for patients with primary immunodeficiency disorders in clinical trials, as well as future potential targets and methods to broaden the applicability of virus-directed T-cell immunotherapy for this vulnerable patient population.
Collapse
Affiliation(s)
- Michael D Keller
- Center for Cancer and Immunology Research and
- Division of Allergy and Immunology, Children's National Health System, Washington, DC
- GW Cancer Center, George Washington University, Washington, DC; and
| | - Catherine M Bollard
- Center for Cancer and Immunology Research and
- GW Cancer Center, George Washington University, Washington, DC; and
- Division of Blood and Marrow Transplantation, Children's National Health System, Washington, DC
| |
Collapse
|
22
|
Immune Control and Vaccination against the Epstein-Barr Virus in Humanized Mice. Vaccines (Basel) 2019; 7:vaccines7040217. [PMID: 31861045 PMCID: PMC6963577 DOI: 10.3390/vaccines7040217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/12/2019] [Accepted: 12/14/2019] [Indexed: 12/12/2022] Open
Abstract
Mice with reconstituted human immune system components (humanized mice) offer the unique opportunity to test vaccines preclinically in the context of vaccine adjuvant sensing by human antigen presenting cells and priming of human cytotoxic lymphocyte populations. These features are particularly attractive for immune control of the Epstein–Barr virus (EBV), which represents the most potent growth-transforming pathogen in man and exclusively relies on cytotoxic lymphocytes for its asymptomatic persistence in the vast majority of healthy virus carriers. This immune control is particularly impressive because EBV infects more than 95% of the human adult population and persists without pathology for more than 50 years in most of them. This review will discuss the pathologies that EBV elicits in humanized mice, which immune responses control it in this model, as well as which passive and active vaccination schemes with adoptive T cell transfer and with virus-like particles or individual antigens, respectively, have been explored in this model so far. EBV-specific CD8+ T cell priming in humanized mice could provide crucial insights into how cytotoxic lymphocytes against other viruses and tumors might be elicited by vaccination in humans.
Collapse
|
23
|
Ando M, Ando J, Yamazaki S, Ishii M, Sakiyama Y, Harada S, Honda T, Yamaguchi T, Nojima M, Ohshima K, Nakauchi H, Komatsu N. Long-term eradication of extranodal natural killer/T-cell lymphoma, nasal type, by induced pluripotent stem cell-derived Epstein-Barr virus-specific rejuvenated T cells in vivo. Haematologica 2019; 105:796-807. [PMID: 31296577 PMCID: PMC7049350 DOI: 10.3324/haematol.2019.223511] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/10/2019] [Indexed: 12/31/2022] Open
Abstract
Functionally rejuvenated induced pluripotent stem cell (iPSC)-derived antigen-specific cytotoxic T lymphocytes (CTL) are expected to be a potent immunotherapy for tumors. When L-asparaginase-containing standard chemotherapy fails in extranodal natural killer/T-cell lymphoma, nasal type (ENKL), no effective salvage therapy exists. The clinical course then is miserable. We demonstrate prolonged and robust eradication of ENKL in vivo by Epstein-Barr virus-specific iPSC-derived antigen-specific CTL, with iPSC-derived antigen-specific CTL persisting as central memory T cells in the mouse spleen for at least six months. The anti-tumor response is so strong that any concomitant effect of the programmed cell death 1 (PD-1) blockade is unclear. These results suggest that long-term persistent Epstein-Barr virus-specific iPSC-derived antigen-specific CTL contribute to a continuous anti-tumor effect and offer an effective salvage therapy for relapsed and refractory ENKL.
Collapse
Affiliation(s)
- Miki Ando
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan .,Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jun Ando
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | - Satoshi Yamazaki
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Midori Ishii
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yumi Sakiyama
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sakiko Harada
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | - Tadahiro Honda
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | - Tomoyuki Yamaguchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masanori Nojima
- Center for Translational Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koichi Ohshima
- Department of Pathology, School of Medicine, Kurume University, Fukuoka, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Norio Komatsu
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Pump WC, Schulz R, Huyton T, Kunze-Schumacher H, Martens J, Hò GGT, Blasczyk R, Bade-Doeding C. Releasing the concept of HLA-allele specific peptide anchors in viral infections: A non-canonical naturally presented human cytomegalovirus-derived HLA-A*24:02 restricted peptide drives exquisite immunogenicity. HLA 2019; 94:25-38. [PMID: 30912293 PMCID: PMC6593758 DOI: 10.1111/tan.13537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 03/18/2019] [Accepted: 03/23/2019] [Indexed: 11/30/2022]
Abstract
T‐cell receptors possess the unique ability to survey and respond to their permanently modified ligands, self HLA‐I molecules bound to non‐self peptides of various origin. This highly specific immune function is impaired following hematopoietic stem cell transplantation (HSCT) for a timespan of several months needed for the maturation of T‐cells. Especially, the progression of HCMV disease in immunocompromised patients induces life‐threatening situations. Therefore, the need for a new immune system that delivers vital and potent CD8+ T‐cells carrying TCRs that recognize even one human cytomegalovirus (HCMV) peptide/HLA molecule and clear the viral infection long term becomes obvious. The transcription and translation of HCMV proteins in the lytic cycle is a precisely regulated cascade of processes, therefore, it is a highly sensitive challenge to adjust the exact time point of HCMV‐peptide recruitment over self‐peptides. We utilized soluble HLA technology in HCMV‐infected fibroblasts and sequenced naturally sHLA‐A*24:02 presented HCMV‐derived peptides. One peptide of 14 AAs length derived from the IE2 antigen induced the strongest T‐cell responses; this peptide can be detected with a low ranking score in general peptide prediction databanks. These results highlight the need for elaborate and HLA‐allele specific peptide selection.
Collapse
Affiliation(s)
- Wiebke C Pump
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Rebecca Schulz
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Trevor Huyton
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | | | - Jörg Martens
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Gia-Gia T Hò
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Rainer Blasczyk
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
25
|
Adoptive T Cell Therapy Strategies for Viral Infections in Patients Receiving Haematopoietic Stem Cell Transplantation. Cells 2019; 8:cells8010047. [PMID: 30646564 PMCID: PMC6356262 DOI: 10.3390/cells8010047] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/08/2019] [Accepted: 01/11/2019] [Indexed: 01/18/2023] Open
Abstract
Adverse outcomes following virus-associated disease in patients receiving allogeneic haematopoietic stem cell transplantation (HSCT) have encouraged strategies to control viral reactivation in immunosuppressed patients. However, despite timely treatment with antiviral medication, some viral infections remain refractory to treatment, which hampers outcomes after HSCT, and are responsible for a high proportion of transplant-related morbidity and mortality. Adoptive transfer of donor-derived lymphocytes aims to improve cellular immunity and to prevent or treat viral diseases after HSCT. Early reports described the feasibility of transferring nonspecific lymphocytes from donors, which led to the development of cell therapy approaches based on virus-specific T cells, allowing a targeted treatment of infections, while limiting adverse events such as graft versus host disease (GvHD). Both expansion and direct selection techniques have yielded comparable results in terms of efficacy (around 70–80%), but efficacy is difficult to predict for individual cases. Generating bespoke products for each donor–recipient pair can be expensive, and there remains the major obstacle of generating products from seronegative or poorly responsive donors. More recent studies have focused on the feasibility of collecting and infusing partially matched third-party virus-specific T cells, reporting response rates of 60–70%. Future development of this approach will involve the broadening of applicability to multiple viruses, the optimization and cost-control of manufacturing, larger multicentred efficacy trials, and finally the creation of cell banks that can provide prompt access to virus-specific cellular product. The aim of this review is to summarise present knowledge on adoptive T cell manufacturing, efficacy and potential future developments.
Collapse
|
26
|
Harris KM, Davila BJ, Bollard CM, Keller MD. Virus-Specific T Cells: Current and Future Use in Primary Immunodeficiency Disorders. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2018; 7:809-818. [PMID: 30581131 DOI: 10.1016/j.jaip.2018.10.049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/31/2018] [Accepted: 10/31/2018] [Indexed: 12/28/2022]
Abstract
Viral infections are common and can be potentially fatal in patients with primary immunodeficiency disorders (PIDDs). Because viral susceptibility stems from poor to absent T-cell function in most patients with moderate to severe forms of PIDD, adoptive immunotherapy with virus-specific T cells (VSTs) has been used to combat viral infections in the setting of hematopoietic stem cell transplantation in multiple clinical trials. Most trials to date have targeted cytomegalovirus, EBV, and adenovirus either alone or in combination, although newer trials have expanded the number of targeted pathogens. Use of banked VSTs produced from third-party donors has also been studied as a method of expanding access to this therapy. Here we review the clinical experience with VST therapy for patients with PIDDs as well as future potential targets and approaches for the use of VSTs to improve clinical outcomes for this specific patient population.
Collapse
Affiliation(s)
- Katherine M Harris
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC
| | - Blachy J Davila
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC; Division of Blood and Marrow Transplantation, Children's National Health System, Washington, DC
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC; Division of Blood and Marrow Transplantation, Children's National Health System, Washington, DC
| | - Michael D Keller
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC; Division of Allergy and Immunology, Children's National Health System, Washington, DC.
| |
Collapse
|
27
|
Toptan T, Abere B, Nalesnik MA, Swerdlow SH, Ranganathan S, Lee N, Shair KH, Moore PS, Chang Y. Circular DNA tumor viruses make circular RNAs. Proc Natl Acad Sci U S A 2018; 115:E8737-E8745. [PMID: 30150410 PMCID: PMC6140489 DOI: 10.1073/pnas.1811728115] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Epstein-Barr virus (EBV) and Kaposi's sarcoma herpesvirus (KSHV) cause ∼2% of all human cancers. RNase R-resistant RNA sequencing revealed that both gammaherpesviruses encode multiple, uniquely stable, circular RNAs (circRNA). EBV abundantly expressed both exon-only and exon-intron circRNAs from the BamHI A rightward transcript (BART) locus (circBARTs) formed from a spliced BART transcript and excluding the EBV miRNA region. The circBARTs were expressed in all verified EBV latency types, including EBV-positive posttransplant lymphoproliferative disease, Burkitt lymphoma, nasopharyngeal carcinoma, and AIDS-associated lymphoma tissues and cell lines. Only cells infected with the B95-8 EBV strain, with a 12-kb BART locus deletion, were negative for EBV circBARTs. Less abundant levels of EBV circRNAs originating from LMP2- and BHLF1-encoding genes were also identified. The circRNA sequencing of KSHV-infected primary effusion lymphoma cells revealed a KSHV-encoded circRNA from the vIRF4 locus (circvIRF4) that was constitutively expressed. In addition, KSHV polyadenylated nuclear (PAN) RNA locus generated a swarm (>100) of multiply backspliced, low-abundance RNase R-resistant circRNAs originating in both sense and antisense directions consistent with a novel hyperbacksplicing mechanism. In EBV and KSHV coinfected cells, exon-only EBV circBARTs were located more in the cytoplasm, whereas the intron-retaining circBARTs were found in the nuclear fraction. KSHV circvIRF4 and circPANs were detected in both nuclear and cytoplasmic fractions. Among viral circRNAs tested, none were found in polysome fractions from KSHV-EBV coinfected BC1 cells, although low-abundance protein translation from viral circRNAs could not be excluded. The circRNAs are a new class of viral transcripts expressed in gammaherpesvirus-related tumors that might contribute to viral oncogenesis.
Collapse
Affiliation(s)
- Tuna Toptan
- Hillman Cancer Center, Cancer Virology Program, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213
| | - Bizunesh Abere
- Hillman Cancer Center, Cancer Virology Program, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213
| | - Michael A Nalesnik
- Division of Transplantation and Hepatic Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Steven H Swerdlow
- Division of Hematopathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213
| | | | - Nara Lee
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213
| | - Kathy H Shair
- Hillman Cancer Center, Cancer Virology Program, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213
| | - Patrick S Moore
- Hillman Cancer Center, Cancer Virology Program, University of Pittsburgh, Pittsburgh, PA 15213;
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213
| | - Yuan Chang
- Hillman Cancer Center, Cancer Virology Program, University of Pittsburgh, Pittsburgh, PA 15213;
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213
| |
Collapse
|
28
|
Abels WC, Manandhar T, Kunze-Schumacher H, Blasczyk R, Bade-Döding C. The polymorphism at residue 156 determines the HLA-B*35 restricted peptide repertoire during HCMV infection. Immunogenetics 2018; 70:639-646. [PMID: 30128813 PMCID: PMC6182399 DOI: 10.1007/s00251-018-1077-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/16/2018] [Indexed: 12/27/2022]
Abstract
Peptide selection in infected cells is not fully understood yet, but several indications point to the fact that there are differences to uninfected cells, especially in productive HCMV infection, since HCMV evolved various strategies to disable the hosts immune system, including presentation of peptide-HLA complexes to immune effector cells. Therefore, peptide predictions for specific HLA alleles are limited in these cases and the naturally presented peptide repertoire of HCMV-infected cells is of major interest to optimize adoptive T cell therapies. The allotypes HLA-B*35:01 and B*35:08 differ at a single amino acid at position 156 and have been described to differ in their peptide features and in their association with the peptide loading complex. Virus specific T cells recognizing the allelic pHLA-B*35 complexes could be detected, indicating a significant role of this HLA subtypes in viral immunity. However, naturally selected and presented viral peptides have not been described so far. In this study, we analyzed the peptide binding repertoire for HLA-B*35:01 and HLA-B*35:08 in HCMV-infected cells. The isolated peptides from both allelic subtypes were of extraordinary length, however differed in their features, origin, and sequence. For these HCMV-originated peptides, no overlap in the peptide repertoire could be observed between the two allelic subtypes. These findings reveal the discrepancies between predicted and naturally presented immunogenic epitopes and support the need of comprehensive peptide recruitment data for personalized and effective cellular therapies.
Collapse
Affiliation(s)
- Wiebke C Abels
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Trishna Manandhar
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | | | - Rainer Blasczyk
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
29
|
Fernandes Q, Merhi M, Raza A, Inchakalody VP, Abdelouahab N, Zar Gul AR, Uddin S, Dermime S. Role of Epstein-Barr Virus in the Pathogenesis of Head and Neck Cancers and Its Potential as an Immunotherapeutic Target. Front Oncol 2018; 8:257. [PMID: 30035101 PMCID: PMC6043647 DOI: 10.3389/fonc.2018.00257] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 06/22/2018] [Indexed: 12/18/2022] Open
Abstract
The role of Epstein-Barr virus (EBV) infection in the development and progression of tumor cells has been described in various cancers. Etiologically, EBV is a causative agent in certain variants of head and neck cancers such as nasopharyngeal cancer. Proteins expressed by the EVB genome are involved in invoking and perpetuating the oncogenic properties of the virus. However, these protein products were also identified as important targets for therapeutic research in the past decades, particularly within the context of immunotherapy. The adoptive transfer of EBV-targeted T-cells as well as the development of EBV vaccines has opened newer lines of research to conceptualize novel therapeutic approaches toward the disease. This review addresses the most important aspects of the association of EBV with head and neck cancers from an immunological perspective. It also aims to highlight the current and future prospects of enhanced EBV-targeted immunotherapies.
Collapse
Affiliation(s)
- Queenie Fernandes
- Translational Cancer Research Facility, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Afsheen Raza
- Translational Cancer Research Facility, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Varghese Philipose Inchakalody
- Translational Cancer Research Facility, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Nassima Abdelouahab
- Translational Cancer Research Facility, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Abdul Rehman Zar Gul
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Interim Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
30
|
Li Y, Sun H, Yan Y, Sun T, Wang S, Ma H. Long-Term Survival Rates of Patients with Stage III-IV Hodgkin Lymphoma According to Age, Sex, Race, and Socioeconomic Status, 1984-2013. Oncologist 2018; 23:1328-1336. [PMID: 29739895 DOI: 10.1634/theoncologist.2017-0541] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 03/23/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Long-term survival rates for patients with stage III-IV Hodgkin lymphoma, or advanced Hodgkin lymphoma (aHL), have increased substantially since the 1960s. Because large-scale research of aHL is rare, we aimed to demonstrate the differences in incidence and survival of aHL according to four patient variables in recent decades, with a focus on the outcomes of treatment of aHL and the advancement of public health care. MATERIALS AND METHODS Data on aHL cases diagnosed during 1984-2013 were extracted from the Surveillance, Epidemiology, and End Results Program database. Relative survival, Kaplan-Meier, and Cox proportional hazards regression analyses were performed to identify prognosis indicators for aHL. RESULTS The incidence rates for aHL were 1.1, 0.8, and 1.0 per 100,000 in the first, second, and third decades, respectively, during 1984-2013. The 120-month relative survival rate improved continuously in each decade from 58.5% to 64.6% to 72.1%. In addition, disparities in the 120-month relative survival rate between male and female patients and among patients of different races narrowed over time. The difference in long-term survival rate between the poor (medium and high poverty) and rich (low poverty) groups narrowed across the 3 decades. CONCLUSION The long-term survival rate for patients with aHL increased in each decade, whereas survival rate disparities according to sex, race, and socioeconomic status narrowed, except for older patients aged >60 years and the high-poverty group. IMPLICATIONS FOR PRACTICE Long-term survival rates of patients with advanced Hodgkin lymphoma were elaborated in this article. The disparities according to sex, race, and socioeconomic status of survival condition were analyzed and showed the development of the public health care system and modern medicine technology.
Collapse
Affiliation(s)
- Yushi Li
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, People's Republic of China
| | - Huanhuan Sun
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, People's Republic of China
| | - Yan Yan
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, People's Republic of China
| | - Tiantian Sun
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Shuncong Wang
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, People's Republic of China
| | - Haiqing Ma
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, People's Republic of China
| |
Collapse
|
31
|
McLaughlin LP, Bollard CM, Keller MD. Adoptive T Cell Therapy for Epstein-Barr Virus Complications in Patients With Primary Immunodeficiency Disorders. Front Immunol 2018; 9:556. [PMID: 29616044 PMCID: PMC5867312 DOI: 10.3389/fimmu.2018.00556] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/05/2018] [Indexed: 12/28/2022] Open
Abstract
Patients with primary immunodeficiency disorders (PID) have an increased risk from acute and chronic Epstein–Barr Virus (EBV) viral infections and EBV-associated malignancies. Hematopoietic stem cell transplantation (HSCT) is a curative strategy for many patients with PID, but EBV-related complications are common in the immediate post-transplant period due to delayed reconstitution of T cell immunity. Adoptive T cell therapy with EBV-specific T cells is a promising therapeutic strategy for patients with PID both before and after HSCT. Here we review the methods used to manufacture EBV-specific T cells, the clinical outcomes, and the ongoing challenges for future development of the strategy.
Collapse
Affiliation(s)
- Lauren P McLaughlin
- Center for Cancer and Immunology Research, Children's National Health System, The George Washington University, Washington, DC, United States.,Division of Oncology, Children's National Health System, Washington, DC, United States
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Health System, The George Washington University, Washington, DC, United States.,Division of Allergy and Immunology, Children's National Health System, Washington, DC, United States.,Division of Blood and Marrow Transplantation, Children's National Health System, Washington, DC, United States
| | - Michael D Keller
- Center for Cancer and Immunology Research, Children's National Health System, The George Washington University, Washington, DC, United States.,Division of Allergy and Immunology, Children's National Health System, Washington, DC, United States
| |
Collapse
|
32
|
Ono T, Fujita Y, Matano T, Takahashi S, Morio T, Kawana-Tachikawa A. Characterization of In Vitro Expanded Virus-Specific T cells for Adoptive Immunotherapy against Virus Infection. Jpn J Infect Dis 2018; 71:122-128. [PMID: 29491233 DOI: 10.7883/yoken.jjid.2017.500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Adoptive transfer of virus-specific T cells has emerged as a promising therapeutic approach for treatment of virus infections in immunocompromised hosts. Characterization of virus-specific T cells provides essential information about the curative mechanism of the treatment. In this study, we developed a T cell epitope mapping system for 718 overlapping peptides spanning 6 proteins of 3 viruses (pp65 and IE1 from cytomegalovirus; LMP1, EBNA1, and BZLF1 from Epstein-Barr virus; Penton from adenovirus). Peripheral blood mononuclear cells (PBMCs) from 33 healthy Japanese donors were stimulated with these peptides and virus-specific CD4+ and CD8+ T cells were expanded in vitro in the presence of interleukin (IL) 4 and IL7. A median of 13 (minimum-maximum, 2-46) peptides was recognized in the cohort. Both fresh and cryopreserved PBMCs were used for in vitro expansion. The expansion and breadth of T cell responses were not significantly different between the 2 PBMC sets. We assessed viral regions frequently recognized by T cells in a Japanese cohort that could become pivotal T cell targets for immunotherapy in Japan. We tested epitope prediction for CD8+ T cell responses against a common target region using a freely available online tool. Some epitopes were considered to be predictive.
Collapse
Affiliation(s)
- Toshiaki Ono
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University
| | - Yuriko Fujita
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo.,Yokohama Municipal Citizen's Hospital
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases.,Department of AIDS Vaccine Development, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo
| | - Satoshi Takahashi
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University
| | - Ai Kawana-Tachikawa
- AIDS Research Center, National Institute of Infectious Diseases.,Department of AIDS Vaccine Development, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo
| |
Collapse
|
33
|
Münz C. Human γ-Herpesvirus Infection, Tumorigenesis, and Immune Control in Mice with Reconstituted Human Immune System Components. Front Immunol 2018; 9:238. [PMID: 29483919 PMCID: PMC5816265 DOI: 10.3389/fimmu.2018.00238] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/29/2018] [Indexed: 12/12/2022] Open
Abstract
The human γ-herpesviruses Epstein–Barr virus (EBV or HHV4) and Kaposi sarcoma-associated herpesvirus (KSHV or HHV8) are each associated with around 2% of all tumors in humans worldwide. However, investigations into their infection, oncogenesis, and immune responses that protect from the associated tumors have been hampered by the exclusive tropism of these pathogens for humans. Mice with reconstituted human immune system components (HIS mice) provide the unique opportunity to study persistent infection, virus associated lymphoma formation, and cell-mediated immune control of EBV and KSHV. Moreover, since these pathogens are unique stimuli for cytotoxic human lymphocyte responses, they also allow us to characterize long-lasting cell-mediated immune control and the requirements for its initiation, which would also be desirable to achieve during antitumor vaccination in general. Thus, human γ-herpesvirus infection of HIS mice provides unique insights into the biology of these important human pathogens and human cell-mediated immune responses that are considered to be the main protective entity against tumors.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
34
|
Virus-Specific T Cells for Hematopoietic Stem Cell Transplantation. CURRENT STEM CELL REPORTS 2017. [DOI: 10.1007/s40778-017-0107-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
35
|
Ba Abdullah MM, Palermo RD, Palser AL, Grayson NE, Kellam P, Correia S, Szymula A, White RE. Heterogeneity of the Epstein-Barr Virus (EBV) Major Internal Repeat Reveals Evolutionary Mechanisms of EBV and a Functional Defect in the Prototype EBV Strain B95-8. J Virol 2017; 91:e00920-17. [PMID: 28904201 PMCID: PMC5686732 DOI: 10.1128/jvi.00920-17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/02/2017] [Indexed: 12/28/2022] Open
Abstract
Epstein-Barr virus (EBV) is a ubiquitous pathogen of humans that can cause several types of lymphoma and carcinoma. Like other herpesviruses, EBV has diversified through both coevolution with its host and genetic exchange between virus strains. Sequence analysis of the EBV genome is unusually challenging because of the large number and lengths of repeat regions within the virus. Here we describe the sequence assembly and analysis of the large internal repeat 1 of EBV (IR1; also known as the BamW repeats) for more than 70 strains. The diversity of the latency protein EBV nuclear antigen leader protein (EBNA-LP) resides predominantly within the exons downstream of IR1. The integrity of the putative BWRF1 open reading frame (ORF) is retained in over 80% of strains, and deletions truncating IR1 always spare BWRF1. Conserved regions include the IR1 latency promoter (Wp) and one zone upstream of and two within BWRF1. IR1 is heterogeneous in 70% of strains, and this heterogeneity arises from sequence exchange between strains as well as from spontaneous mutation, with interstrain recombination being more common in tumor-derived viruses. This genetic exchange often incorporates regions of <1 kb, and allelic gene conversion changes the frequency of small regions within the repeat but not close to the flanks. These observations suggest that IR1-and, by extension, EBV-diversifies through both recombination and breakpoint repair, while concerted evolution of IR1 is driven by gene conversion of small regions. Finally, the prototype EBV strain B95-8 contains four nonconsensus variants within a single IR1 repeat unit, including a stop codon in the EBNA-LP gene. Repairing IR1 improves EBNA-LP levels and the quality of transformation by the B95-8 bacterial artificial chromosome (BAC).IMPORTANCE Epstein-Barr virus (EBV) infects the majority of the world population but causes illness in only a small minority of people. Nevertheless, over 1% of cancers worldwide are attributable to EBV. Recent sequencing projects investigating virus diversity to see if different strains have different disease impacts have excluded regions of repeating sequence, as they are more technically challenging. Here we analyze the sequence of the largest repeat in EBV (IR1). We first characterized the variations in protein sequences encoded across IR1. In studying variations within the repeat of each strain, we identified a mutation in the main laboratory strain of EBV that impairs virus function, and we suggest that tumor-associated viruses may be more likely to contain DNA mixed from two strains. The patterns of this mixing suggest that sequences can spread between strains (and also within the repeat) by copying sequence from another strain (or repeat unit) to repair DNA damage.
Collapse
Affiliation(s)
- Mohammed M Ba Abdullah
- Section of Virology, Imperial College Faculty of Medicine, St. Mary's Hospital, Norfolk Place, London, United Kingdom
| | - Richard D Palermo
- Section of Virology, Imperial College Faculty of Medicine, St. Mary's Hospital, Norfolk Place, London, United Kingdom
| | - Anne L Palser
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | | | - Paul Kellam
- Section of Virology, Imperial College Faculty of Medicine, St. Mary's Hospital, Norfolk Place, London, United Kingdom
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
- Kymab, Babraham Research Campus, Cambridge, United Kingdom
| | - Samantha Correia
- Section of Virology, Imperial College Faculty of Medicine, St. Mary's Hospital, Norfolk Place, London, United Kingdom
| | - Agnieszka Szymula
- Section of Virology, Imperial College Faculty of Medicine, St. Mary's Hospital, Norfolk Place, London, United Kingdom
| | - Robert E White
- Section of Virology, Imperial College Faculty of Medicine, St. Mary's Hospital, Norfolk Place, London, United Kingdom
| |
Collapse
|
36
|
Cellular therapy for multiple pathogen infections after hematopoietic stem cell transplant. Cytotherapy 2017; 19:1284-1301. [DOI: 10.1016/j.jcyt.2017.07.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/18/2017] [Accepted: 07/27/2017] [Indexed: 11/22/2022]
|
37
|
Tashiro H, Sauer T, Shum T, Parikh K, Mamonkin M, Omer B, Rouce RH, Lulla P, Rooney CM, Gottschalk S, Brenner MK. Treatment of Acute Myeloid Leukemia with T Cells Expressing Chimeric Antigen Receptors Directed to C-type Lectin-like Molecule 1. Mol Ther 2017; 25:2202-2213. [PMID: 28676343 DOI: 10.1016/j.ymthe.2017.05.024] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/28/2017] [Accepted: 05/31/2017] [Indexed: 01/05/2023] Open
Abstract
The successful immunotherapy of acute myeloid leukemia (AML) has been hampered because most potential antigenic targets are shared with normal hematopoietic stem cells (HSCs), increasing the risk of sustained and severe hematopoietic toxicity following treatment. C-type lectin-like molecule 1 (CLL-1) is a membrane glycoprotein expressed by >80% of AML but is absent on normal HSCs. Here we describe the development and evaluation of CLL-1-specific chimeric antigen receptor T cells (CLL-1.CAR-Ts) and we demonstrate their specific activity against CLL-1+ AML cell lines as well as primary AML patient samples in vitro. CLL-1.CAR-Ts selectively reduced leukemic colony formation in primary AML patient peripheral blood mononuclear cells compared to control T cells. In a human xenograft mouse model, CLL-1.CAR-Ts mediated anti-leukemic activity against disseminated AML and significantly extended survival. By contrast, the colony formation of normal progenitor cells remained intact following CLL-1.CAR-T treatment. Although CLL-1.CAR-Ts are cytotoxic to mature normal myeloid cells, the selective sparing of normal hematopoietic progenitor cells should allow full myeloid recovery once CLL-1.CAR-T activity terminates. To enable elective ablation of the CAR-T, we therefore introduced the inducible caspase-9 suicide gene system and we show that exposure to the activating drug rapidly induced a controlled decrease of unwanted CLL-1.CAR-T activity against mature normal myeloid cells.
Collapse
Affiliation(s)
- Haruko Tashiro
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, Houston, TX 77030, USA.
| | - Tim Sauer
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, Houston, TX 77030, USA; Department of Internal Medicine A, Hematology and Oncology, University of Muenster, 48149 Muenster, Germany
| | - Thomas Shum
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, Houston, TX 77030, USA; Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kathan Parikh
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, Houston, TX 77030, USA
| | - Maksim Mamonkin
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bilal Omer
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rayne H Rouce
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, TX 77030, USA
| | - Premal Lulla
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, Houston, TX 77030, USA; Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular Virology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephen Gottschalk
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
38
|
Keane C, Gould C, Jones K, Hamm D, Talaulikar D, Ellis J, Vari F, Birch S, Han E, Wood P, Le-Cao KA, Green MR, Crooks P, Jain S, Tobin J, Steptoe RJ, Gandhi MK. The T-cell Receptor Repertoire Influences the Tumor Microenvironment and Is Associated with Survival in Aggressive B-cell Lymphoma. Clin Cancer Res 2017; 23:1820-1828. [PMID: 27649554 DOI: 10.1158/1078-0432.ccr-16-1576] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/31/2016] [Accepted: 09/02/2016] [Indexed: 11/16/2022]
Abstract
Purpose: To investigate the relationship between the intra-tumoral T-cell receptor (TCR) repertoire and the tumor microenvironment (TME) in de novo diffuse large B-cell lymphoma (DLBCL) and the impact of TCR on survival.Experimental Design: We performed high-throughput unbiased TCRβ sequencing on a population-based cohort of 92 patients with DLBCL treated with conventional (i.e., non-checkpoint blockade) frontline "R-CHOP" therapy. Key immune checkpoint genes within the TME were digitally quantified by nanoString. The primary endpoints were 4-year overall survival (OS) and progression-free survival (PFS).Results: The TCR repertoire within DLBCL nodes was abnormally narrow relative to non-diseased nodal tissues (P < 0.0001). In DLBCL, a highly dominant single T-cell clone was associated with inferior 4-year OS rate of 60.0% [95% confidence interval (CI), 31.7%-79.6%], compared with 79.8% in patients with a low dominant clone (95% CI, 66.7%-88.5%; P = 0.005). A highly dominant clone also predicted inferior 4-year PFS rate of 46.6% (95% CI, 22.5%-76.6%) versus 72.6% (95% CI, 58.8%-82.4%, P = 0.008) for a low dominant clone. In keeping, clonal expansions were most pronounced in the EBV+ DLBCL subtype that is known to express immunogenic viral antigens and is associated with particularly poor outcome. Increased T-cell diversity was associated with significantly elevated PD-1, PD-L1, and PD-L2 immune checkpoint molecules.Conclusions: Put together, these findings suggest that the TCR repertoire is a key determinant of the TME. Highly dominant T-cell clonal expansions within the TME are associated with poor outcome in DLBCL treated with conventional frontline therapy. Clin Cancer Res; 23(7); 1820-8. ©2016 AACR.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal, Murine-Derived/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Cyclophosphamide/administration & dosage
- Disease-Free Survival
- Doxorubicin/administration & dosage
- Female
- High-Throughput Nucleotide Sequencing
- Humans
- Kaplan-Meier Estimate
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Male
- Middle Aged
- Neoplasm Staging
- Prednisone/administration & dosage
- Prognosis
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Rituximab/administration & dosage
- Tumor Microenvironment/genetics
- Vincristine/administration & dosage
Collapse
Affiliation(s)
- Colm Keane
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Australia.
- Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Clare Gould
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Australia
- Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Kimberley Jones
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Australia
| | - David Hamm
- Adaptive Biotechnologies, Seattle, Washington
| | - Dipti Talaulikar
- Canberra Hospital, Canberra, Australian Capital Territory, Australia
- Australian National University Medical School, Australian Capital Territory, Australia
| | - Jonathan Ellis
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Australia
| | - Frank Vari
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Australia
| | - Simone Birch
- Princess Alexandra Hospital, Brisbane, Queensland, Australia
- Pathology Queensland, Brisbane, Queensland, Australia
| | - Erica Han
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Australia
| | - Peter Wood
- Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Kim-Anh Le-Cao
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Australia
| | - Michael R Green
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska
| | - Pauline Crooks
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Australia
| | - Sanjiv Jain
- Canberra Hospital, Canberra, Australian Capital Territory, Australia
| | - Josh Tobin
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Australia
- Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Raymond J Steptoe
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Australia
| | - Maher K Gandhi
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Australia.
- Princess Alexandra Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
39
|
Epstein-Barr virus microRNAs reduce immune surveillance by virus-specific CD8+ T cells. Proc Natl Acad Sci U S A 2016; 113:E6467-E6475. [PMID: 27698133 DOI: 10.1073/pnas.1605884113] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Infection with Epstein-Barr virus (EBV) affects most humans worldwide and persists life-long in the presence of robust virus-specific T-cell responses. In both immunocompromised and some immunocompetent people, EBV causes several cancers and lymphoproliferative diseases. EBV transforms B cells in vitro and encodes at least 44 microRNAs (miRNAs), most of which are expressed in EBV-transformed B cells, but their functions are largely unknown. Recently, we showed that EBV miRNAs inhibit CD4+ T-cell responses to infected B cells by targeting IL-12, MHC class II, and lysosomal proteases. Here we investigated whether EBV miRNAs also counteract surveillance by CD8+ T cells. We have found that EBV miRNAs strongly inhibit recognition and killing of infected B cells by EBV-specific CD8+ T cells through multiple mechanisms. EBV miRNAs directly target the peptide transporter subunit TAP2 and reduce levels of the TAP1 subunit, MHC class I molecules, and EBNA1, a protein expressed in most forms of EBV latency and a target of EBV-specific CD8+ T cells. Moreover, miRNA-mediated down-regulation of the cytokine IL-12 decreases the recognition of infected cells by EBV-specific CD8+ T cells. Thus, EBV miRNAs use multiple, distinct pathways, allowing the virus to evade surveillance not only by CD4+ but also by antiviral CD8+ T cells.
Collapse
|
40
|
T cells for viral infections after allogeneic hematopoietic stem cell transplant. Blood 2016; 127:3331-40. [PMID: 27207801 DOI: 10.1182/blood-2016-01-628982] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/11/2016] [Indexed: 12/21/2022] Open
Abstract
Despite recent advances in the field of allogeneic hematopoietic stem cell transplantation (HSCT), viral infections are still a major complication during the period of immune suppression that follows the procedure. Adoptive transfer of donor-derived virus-specific cytotoxic T cells (VSTs) is a strategy to rapidly restore virus-specific immunity to prevent or treat viral diseases after HSCT. Early proof of principle studies demonstrated that the administration of donor-derived T cells specific for cytomegalovirus or Epstein-Barr virus (EBV) could effectively restore virus-specific immunity and control viral infections. Subsequent studies using different expansion or direct selection techniques have shown that donor-derived VSTs confer protection in vivo after adoptive transfer in 70% to 90% of recipients. Because a major cause of failure is lack of immunity to the infecting virus in a naïve donor, more recent studies have infused closely matched third-party VSTs and reported response rates of 60% to 70%. Current efforts have focused on broadening the applicability of this approach by: (1) extending the number of viral antigens being targeted, (2) simplifying manufacture, (3) exploring strategies for recipients of virus-naïve donor grafts, and (4) developing and optimizing "off the shelf" approaches.
Collapse
|
41
|
Barth MJ, Chu Y, Hanley PJ, Cairo MS. Immunotherapeutic approaches for the treatment of childhood, adolescent and young adult non-Hodgkin lymphoma. Br J Haematol 2016; 173:597-616. [PMID: 27062282 DOI: 10.1111/bjh.14078] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
With the introduction of the anti-CD20 monoclonal antibody rituximab, B-cell non-Hodgkin lymphoma was the first malignancy successfully treated with an immunotherapeutic agent. Since then, numerous advances have expanded the repertoire of immunotherapeutic agents available for the treatment of a variety of malignancies, including many lymphoma subtypes. These include the introduction of monoclonal antibodies targeting a variety of cell surface proteins, including the successful targeting of immunoregulatory checkpoint receptors present on T-cells or tumour cells. Additionally, cellular immunotherapeutic approaches utilize T- or Natural Killer-cells generated with chimeric antigen receptors against cell surface proteins or Epstein-Barr virus-associated latent membrane proteins. The following review describes the current state of immunotherapy for non-Hodgkin lymphoma including a summary of currently available data and promising agents currently in clinical development with future promise in the treatment of childhood, adolescent and young adult non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Matthew J Barth
- Department of Pediatrics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Division of Pediatric Hematology/Oncology, University at Buffalo, Buffalo, NY, USA
| | - Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - Patrick J Hanley
- Program for Cell Enhancement and Technologies for Immunotherapy, Division of Blood and Marrow Transplantation, Sheikhz Zayed Institute for Pediatric Surgical Innovation, Washington, D.C., USA.,Center for Cancer and Immunology Research, Children's National Health System, The George Washington University, Washington, D.C., USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.,Department of Medicine, New York Medical College, Valhalla, NY, USA.,Department of Pathology, New York Medical College, Valhalla, NY, USA.,Department of Microbiology & Immunology, New York Medical College, Valhalla, NY, USA.,Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
42
|
Virus-specific T-cell banks for 'off the shelf' adoptive therapy of refractory infections. Bone Marrow Transplant 2016; 51:1163-72. [PMID: 27042851 DOI: 10.1038/bmt.2016.17] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 01/13/2016] [Indexed: 02/07/2023]
Abstract
Adoptive immunotherapy with transplant donor-derived virus-specific T cells has emerged as a potentially curative approach for the treatment of drug-refractory EBV+lymphomas as well as CMV and adenovirus infections complicating allogeneic hematopoietic cell transplants. Adoptive transfer of HLA partially matched virus-specific T cells from healthy third party donors has also shown promise in the treatment of these conditions, with disease response rates of 50-76% and strikingly low incidences of toxicity or GVHD recorded in initial trials. In this review, we examine the reported experience with transplant donor and third party donor-derived virus-specific T cells, identifying characteristics of the viral pathogen, the T cells administered and the diseased host that contribute to treatment response or failure. We also describe the characteristics of virus-specific T-cell lines in our center's bank and the frequency with which in vitro culture promotes expansion of immunodominant T cells specific for epitopes that are presented by a limited array of prevalent HLA alleles, which facilitates their broad applicability for treatment.
Collapse
|
43
|
Nowakowska J, Stuehler C, Egli A, Battegay M, Rauser G, Bantug GR, Brander C, Hess C, Khanna N. T cells specific for different latent and lytic viral proteins efficiently control Epstein-Barr virus-transformed B cells. Cytotherapy 2016; 17:1280-91. [PMID: 26276009 DOI: 10.1016/j.jcyt.2015.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 05/19/2015] [Accepted: 06/07/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND AIMS Epstein-Barr virus (EBV)-associated post-transplant lymphoproliferative disorders (PTLD) belong to the most dreaded complications of immunosuppression. The efficacy of EBV-specific T-cell transfer for PTLD has been previously shown, yet the optimal choice of EBV-derived antigens inducing polyclonal CD4(+) and CD8(+) T cells that cover a wide range of human leukocyte antigen types and efficiently control PTLD remains unclear. METHODS A pool of 125 T-cell epitopes from seven latent and nine lytic EBV-derived proteins (EBVmix) and peptide pools of EBNA1, EBNA3c, LMP2a and BZLF1 were used to determine T-cell frequencies and to isolate T cells through the use of the interferon (IFN)-γ cytokine capture system. We further evaluated the phenotype and functionality of the generated T-cell lines in vitro. RESULTS EBVmix induced significantly higher T-cell frequencies and allowed selecting more CD4(+)IFN-γ(+) and CD8(+)IFN-γ(+) cells than single peptide pools. T cells of all specificities expanded similarly in vitro, recognized cognate antigen, and, to a lower extent, EBV-infected cells, exerted moderate cytotoxicity and showed reduced alloreactivity. However, EBVmix-specific cells most efficiently controlled EBV-infected lymphoblastoid cell lines (LCLs). This control was mainly mediated by EBV-specific CD8(+) cells with an oligoclonal epitope signature covering both latent and lytic viral proteins. Notably, EBV-specific CD4(+) cells unable to control LCLs produced significantly less perforin and granzyme B, probably because of limited LCL epitope presentation. CONCLUSIONS EBVmix induces a broader T-cell response, probably because of its coverage of latent and lytic EBV-derived proteins that may be important to control EBV-transformed B cells and might offer an improvement of T-cell therapies.
Collapse
Affiliation(s)
- Justyna Nowakowska
- Infection Biology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Switzerland
| | - Claudia Stuehler
- Infection Biology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Switzerland
| | - Adrian Egli
- Infection Biology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Switzerland; Clinical Microbiology, University Hospital of Basel, Switzerland
| | - Manuel Battegay
- Division of Infectious Diseases and Hospital Epidemiology, Department of Biomedicine and Clinical Research University Hospital of Basel, Switzerland
| | - Georg Rauser
- Research and Development, Miltenyi Biotec, Bergisch-Gladbach, Germany
| | - Glenn Robert Bantug
- Immunobiology, Department of Biomedicine, University and University Hospital of Basel, Switzerland
| | - Christian Brander
- AIDS Research Institute-IrsiCaixa and AIDS Unit, Hospital Germans Trias i Pujol, Autonomous University of Barcelona, Badalona, Spain, Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain; University of Vic and Central Catalonia, Vic, Spain
| | - Christoph Hess
- Immunobiology, Department of Biomedicine, University and University Hospital of Basel, Switzerland
| | - Nina Khanna
- Infection Biology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, Department of Biomedicine and Clinical Research University Hospital of Basel, Switzerland.
| |
Collapse
|
44
|
Abstract
Post-transplant lymphoproliferative disorders (PTLDs) are a group of conditions that involve uncontrolled proliferation of lymphoid cells as a consequence of extrinsic immunosuppression after organ or haematopoietic stem cell transplant. PTLDs show some similarities to classic lymphomas in the non-immunosuppressed general population. The oncogenic Epstein-Barr virus (EBV) is a key pathogenic driver in many early-onset cases, through multiple mechanisms. The incidence of PTLD varies with the type of transplant; a clear distinction should therefore be made between the conditions after solid organ transplant and after haematopoietic stem cell transplant. Recipient EBV seronegativity and the intensity of immunosuppression are among key risk factors. Symptoms and signs depend on the localization of the lymphoid masses. Diagnosis requires histopathology, although imaging techniques can provide additional supportive evidence. Pre-emptive intervention based on monitoring EBV levels in blood has emerged as the preferred strategy for PTLD prevention. Treatment of established disease includes reduction of immunosuppression and/or administration of rituximab (a B cell-specific antibody against CD20), chemotherapy and EBV-specific cytotoxic T cells. Despite these strategies, the mortality and morbidity remains considerable. Patient outcome is influenced by the severity of presentation, treatment-related complications and risk of allograft loss. New innovative treatment options hold promise for changing the outlook in the future.
Collapse
|
45
|
Abstract
Epstein-Barr virus (EBV) is arguably one of the most successful pathogens of humans, persistently infecting over ninety percent of the world's population. Despite this high frequency of carriage, the virus causes apparently few adverse effects in the vast majority of infected individuals. Nevertheless, the potent growth transforming ability of EBV means the virus has the potential to cause malignancies in infected individuals. Indeed, EBV is thought to cause 1% of human malignancies, equating to 200,000 malignancies each year. A clear factor as to why virus-induced disease is relatively infrequent in healthy infected individuals is the presence of a potent immune response to EBV, in particular, that mediated by T cells. Thus, patient groups with immunodeficiencies or whose cellular immune response is suppressed have much higher frequencies of EBV-induced disease and, in at least some cases, these diseases can be controlled by restoration of the T-cell compartment. In this chapter, we will primarily review the role the αβ subset of T cells in the control of EBV in healthy and diseased individuals.
Collapse
Affiliation(s)
- Andrew D Hislop
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Graham S Taylor
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
46
|
Mittal P, St Rose MC, Wang X, Ryan JM, Wasser JS, Vella AT, Adler AJ. Tumor-Unrelated CD4 T Cell Help Augments CD134 plus CD137 Dual Costimulation Tumor Therapy. THE JOURNAL OF IMMUNOLOGY 2015; 195:5816-26. [PMID: 26561553 DOI: 10.4049/jimmunol.1502032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/15/2015] [Indexed: 11/19/2022]
Abstract
The ability of immune-based cancer therapies to elicit beneficial CD8(+) CTLs is limited by tolerance pathways that inactivate tumor-specific CD4 Th cells. A strategy to bypass this problem is to engage tumor-unrelated CD4 Th cells. Thus, CD4 T cells, regardless of their specificity per se, can boost CD8(+) CTL priming as long as the cognate epitopes are linked via presentation on the same dendritic cell. In this study, we assessed the therapeutic impact of engaging tumor-unrelated CD4 T cells during dual costimulation with CD134 plus CD137 that provide help via the above-mentioned classical linked pathway, as well as provide nonlinked help that facilitates CTL function in T cells not directly responding to cognate Ag. We found that engagement of tumor-unrelated CD4 Th cells dramatically boosted the ability of dual costimulation to control the growth of established B16 melanomas. Surprisingly, this effect depended upon a CD134-dependent component that was extrinsic to the tumor-unrelated CD4 T cells, suggesting that the dual costimulated helper cells are themselves helped by a CD134(+) cell(s). Nevertheless, the delivery of therapeutic help tracked with an increased frequency of tumor-infiltrating granzyme B(+) effector CD8 T cells and a reciprocal decrease in Foxp3(+)CD4(+) cell frequency. Notably, the tumor-unrelated CD4 Th cells also infiltrated the tumors, and their deletion several days following initial T cell priming negated their therapeutic impact. Taken together, dual costimulation programs tumor-unrelated CD4 T cells to deliver therapeutic help during both the priming and effector stages of the antitumor response.
Collapse
Affiliation(s)
- Payal Mittal
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030; and
| | - Marie-Clare St Rose
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030; and
| | - Xi Wang
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030; and
| | - Joseph M Ryan
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030; and
| | - Jeffrey S Wasser
- The Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Anthony T Vella
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030; and
| | - Adam J Adler
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030; and
| |
Collapse
|
47
|
Characterization and Functional Analysis of scFv-based Chimeric Antigen Receptors to Redirect T Cells to IL13Rα2-positive Glioma. Mol Ther 2015; 24:354-363. [PMID: 26514825 DOI: 10.1038/mt.2015.199] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/23/2015] [Indexed: 12/30/2022] Open
Abstract
Immunotherapy with T cells expressing chimeric antigen receptors (CARs) is an attractive approach to improve outcomes for patients with glioblastoma (GBM). IL13Rα2 is expressed at a high frequency in GBM but not in normal brain, making it a promising CAR T-cell therapy target. IL13Rα2-specific CARs generated up to date contain mutated forms of IL13 as an antigen-binding domain. While these CARs target IL13Rα2, they also recognize IL13Rα1, which is broadly expressed. To overcome this limitation, we constructed a panel of IL13Rα2-specific CARs that contain the IL13Rα2-specific single-chain variable fragment (scFv) 47 as an antigen binding domain, short or long spacer regions, a transmembrane domain, and endodomains derived from costimulatory molecules and CD3.ζ (IL13Rα2-CARs). IL13Rα2-CAR T cells recognized IL13Rα2-positive target cells in coculture and cytotoxicity assays with no cross-reactivity to IL13Rα1. However, only IL13Rα2-CAR T cells with a short spacer region produced IL2 in an antigen-dependent fashion. In vivo, T cells expressing IL13Rα2-CARs with short spacer regions and CD28.ζ, 41BB.ζ, and CD28.OX40.ζ endodomains had potent anti-glioma activity conferring a significant survival advantage in comparison to mice that received control T cells. Thus, IL13Rα2-CAR T cells hold the promise to improve current IL13Rα2-targeted immunotherapy approaches for GBM and other IL13Rα2-positive malignancies.
Collapse
|
48
|
Koehne G, Hasan A, Doubrovina E, Prockop S, Tyler E, Wasilewski G, O'Reilly RJ. Immunotherapy with Donor T Cells Sensitized with Overlapping Pentadecapeptides for Treatment of Persistent Cytomegalovirus Infection or Viremia. Biol Blood Marrow Transplant 2015; 21:1663-78. [PMID: 26028505 PMCID: PMC4537838 DOI: 10.1016/j.bbmt.2015.05.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/18/2015] [Indexed: 11/22/2022]
Abstract
We conducted a phase I trial of allogeneic T cells sensitized in vitro against a pool of pentadecapeptides (15-mer peptides) spanning the sequence of CMVpp65 for adoptive therapy of 17 allogeneic hematopoietic cell transplant recipients with cytomegalovirus (CMV) viremia or clinical infection persisting despite prolonged treatment with antiviral drugs. All but 3 of the patients had received T cell-depleted transplants without graft-versus-host disease (GVHD) prophylaxis with immunosuppressive drugs after transplantation. The CMVpp65-specific T cells (CMVpp65CTLs) generated were oligoclonal and specific for only 1 to 3 epitopes, presented by a limited set of HLA class I or II alleles. T cell infusions were well tolerated without toxicity or GVHD. Of 17 patients treated with transplant donor (n = 16) or third-party (n = 1) CMVpp65CTLs, 15 cleared viremia, including 3 of 5 with overt disease. In responding patients, the CMVpp65CTLs infused consistently proliferated and could be detected by T cell receptor Vβ usage in CMVpp65/HLA tetramer + populations for period of 120 days to up to 2 years after infusion. Thus, CMVpp65CTLs generated in response to synthetic 15-mer peptides of CMVpp65 are safe and can clear persistent CMV infections in the post-transplantation period.
Collapse
Affiliation(s)
- Guenther Koehne
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York; Transplantation Biology Laboratory, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York.
| | - Aisha Hasan
- Transplantation Biology Laboratory, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York; Bone Marrow Transplantation Service, Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Ekaterina Doubrovina
- Transplantation Biology Laboratory, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York; Bone Marrow Transplantation Service, Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Susan Prockop
- Transplantation Biology Laboratory, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York; Bone Marrow Transplantation Service, Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Eleanor Tyler
- Transplantation Biology Laboratory, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Gloria Wasilewski
- Bone Marrow Transplantation Service, Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Richard J O'Reilly
- Transplantation Biology Laboratory, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York; Bone Marrow Transplantation Service, Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York
| |
Collapse
|
49
|
Abstract
Lymphomas arise from clonal expansions of B, T, or NK cells at different stages of differentiation. Because they occur in the immunocyte-rich lymphoid tissues, they are easily accessible to antibodies and cell-based immunotherapy. Expressing chimeric antigen receptors (CARs) on T cells is a means of combining the antigen-binding site of a monoclonal antibody with the activating machinery of a T cell, enabling antigen recognition independent of major histocompatibility complex restriction, while retaining the desirable antitumor properties of a T cell. Here, we discuss the basic design of CARs and their potential advantages and disadvantages over other immune therapies for lymphomas. We review current clinical trials in the field and consider strategies to improve the in vivo function and safety of immune cells expressing CARs. The ultimate driver of CAR development and implementation for lymphoma will be the demonstration of their ability to safely and cost-effectively cure these malignancies.
Collapse
Affiliation(s)
- Carlos A Ramos
- Center for Cell and Gene Therapy, Houston Methodist Hospital, Texas Children's Hospital, and Baylor College of Medicine, Houston, Texas 77030.,Dan L. Duncan Cancer Center.,Department of Medicine, and
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Houston Methodist Hospital, Texas Children's Hospital, and Baylor College of Medicine, Houston, Texas 77030.,Dan L. Duncan Cancer Center.,Department of Medicine, and.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030; , ,
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Houston Methodist Hospital, Texas Children's Hospital, and Baylor College of Medicine, Houston, Texas 77030.,Dan L. Duncan Cancer Center.,Department of Medicine, and.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030; , ,
| |
Collapse
|
50
|
Tzannou I, Leen AM. Preventing stem cell transplantation-associated viral infections using T-cell therapy. Immunotherapy 2015; 7:793-810. [PMID: 26250410 DOI: 10.2217/imt.15.43] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hematopoietic stem cell transplantation is the treatment of choice for many hematologic malignancies and genetic diseases. However, viral infections continue to account for substantial post-transplant morbidity and mortality. While antiviral drugs are available against some viruses, they are associated with significant side effects and are frequently ineffective. This review focuses on the immunotherapeutic strategies that have been used to prevent and treat infections over the past 20 years and outlines different refinements that have been introduced with the goal of moving this therapy beyond specialized academic centers.
Collapse
Affiliation(s)
- Ifigeneia Tzannou
- Center for Cell & Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital & Texas Children's Hospital, 1102 Bates Street, Suite 1770, Houston, TX 77030, USA
| | - Ann M Leen
- Center for Cell & Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital & Texas Children's Hospital, 1102 Bates Street, Suite 1770, Houston, TX 77030, USA
| |
Collapse
|