1
|
Nath A, Kolson DL. Reemerging Infectious Diseases and Neuroimmunologic Complications. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200356. [PMID: 39693583 PMCID: PMC11658811 DOI: 10.1212/nxi.0000000000200356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 11/07/2024] [Indexed: 12/20/2024]
Abstract
During the past decade (and beyond), neurologists have become aware of the emergence, persistence, and consequences of some familiar and new infections affecting the nervous system. Even among the familiar CNS infections, such as herpes virus, polyoma virus/JC, influenza, arbovirus, and hepatitis, challenges remain in developing effective antiviral treatments and treatments of postinfection sequelae. With the changing environment and increased global travel, arthropod vectors that mediate zoonotic disease transmission have spread unfamiliar viruses such as West Nile virus, dengue, chikungunya, equine encephalitis, and Zika, among others. Although the global health impact of these diseases has not risen to that of COVID-19 and HIV, it is likely to dramatically increase with continued spread of transmission vectors and the emergence of new zoonotic animal-to-human diseases mediated by those transmission vectors. Furthermore, specific virus-targeting treatments or effective vaccines for arboviral infections are not yet available, and this represents a major challenge in limiting the morbidity of these infections. By contrast, HIV-1, a disease that originated by direct transmission from nonhuman primates to humans (as early as the 1930s), after many years of intense study, is now targeted by highly specific and effective antiviral drugs that can limit the spread of infection and extend human life and health in all populations. Even with these dramatic therapeutic effects of suppressing HIV replication, neurologic dysfunction (primarily cognitive impairment) affects significant numbers of persons living with HIV. This emphasizes not only the importance of treating the underlying infection but also developing treatments for legacy effects of the initial infection even after antiviral therapy. Notably, the rapid emergence of SARS-CoV-2 infection was met with rapid implementation of highly effective and specific antiviral therapies. This resulted in early and dramatic lowering of the morbidity and mortality of SARS-CoV-2 infection. Nonetheless, the postinfectious complications of SARS-CoV-2 infection (long COVID) are now among the more costly consequences of emerging zoonotic infections worldwide. Developing new antiviral therapies that can penetrate the CNS, vaccines, and therapies that target host immune responses and metabolic dysfunction will be necessary for management of infectious and postinfectious complications of established and emerging infections.
Collapse
Affiliation(s)
- Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD; and
| | - Dennis L Kolson
- Department of Neurology, University of Pennsylvania, Philadelphia
| |
Collapse
|
2
|
Alkan B, Tuncer MA, İnkaya AÇ. Advances in virus-specific T-cell therapy for polyomavirus infections: A comprehensive review. Int J Antimicrob Agents 2024; 64:107333. [PMID: 39245328 DOI: 10.1016/j.ijantimicag.2024.107333] [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: 12/21/2023] [Revised: 08/14/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Polyomaviruses are a group of small, non-enveloped, double-stranded DNA viruses that can infect various hosts, including humans. BKPyV causes conditions such as human polyomavirus-associated nephropathy (HPyVAN), human polyomavirus-associated haemorrhagic cystitis (HPyVHC), and human polyomavirus-associated urothelial cancer (HPyVUC). JC polyomavirus (JCPyV), on the other hand, is the causative agent of progressive multifocal leukoencephalopathy (PML), a severe demyelinating disease of the central nervous system. PML primarily affects immunocompromised individuals, including those with HIV, recipients of certain immunosuppressive therapies, and transplant patients. The treatment options for HPyV infections have been limited, but recent developments in virus-specific T cell (VST) therapy have shown promise. Although VST therapy has shown potential in treating both BKPyV and JCPyV infections, several challenges remain. These include the time-consuming and costly preparation of VSTs, the need for sophisticated production facilities, and uncertainties regarding the optimal cell type and infusion frequency. To the best of our knowledge, 85 patients with haemorrhagic cystitis, 27 patients with BKPyV viremia, 2 patients with BKPyV nephritis, 14 patients with haemorrhagic cystitis and BKPyV viremia, and 32 patients with PML have been treated with VST in the literature. The overall response results were 82 complete response, 33 partial response, 35 no response, and 10 no-outcome-reported. This review underscores the importance of VST therapy as a promising treatment approach for polyomavirus infections, emphasising the need for continued research and clinical trials to refine and expand this innovative immunotherapeutic strategy.
Collapse
Affiliation(s)
- Baran Alkan
- Hacettepe University, Faculty of Medicine, Ankara
| | - M Asli Tuncer
- Hacettepe University, Faculty of Medicine, Department of Neurology, Ankara
| | - A Çağkan İnkaya
- Hacettepe University, Faculty of Medicine, Department of Infectious Diseases, Ankara.
| |
Collapse
|
3
|
Lara-de-León AG, Mora-Buch R, Cantó E, Peña-Gómez C, Rudilla F. Identification of Candidate Immunodominant Epitopes and Their HLA-Binding Prediction on BK Polyomavirus Proteins in Healthy Donors. HLA 2024; 104:e15722. [PMID: 39435889 DOI: 10.1111/tan.15722] [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: 03/25/2024] [Revised: 09/12/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024]
Abstract
BK polyomavirus infection is an important cause of graft loss in transplant patients, however, currently available therapies lack effectiveness against this pathogen. Identification of immunological targets for potential treatments is therefore necessary. The aim of this study was to predict candidates of immunodominant epitopes within four BK virus proteins (VP1, VP2, VP3 and LTA) using PBMCs from 44 healthy donors. We used the ELISpot epitope mapping method to evaluate the T-cell response, and HLA-peptide binding was predicted using the NetMHCpan algorithm. A total of 11 potential peptides were selected for VP1, 3 for VP2/VP3 and 13 for LTA. Greater reactivity was observed for VP1 and LTA proteins compared with VP2/VP3. Most of the peptides selected as potential immunodominant candidates were restricted towards several HLA class I and II alleles, with predominant HLA class I binding by computational predictions. Based on these findings, the sequences of the selected immunodominant epitopes candidates and their corresponding HLA restrictions could contribute to the optimisation of functional assays and aid in the design and improvement of immunotherapy strategies against BK virus infections.
Collapse
Affiliation(s)
- Ana Gabriela Lara-de-León
- Advanced & Cell Therapy Services, Banc de Sang i Teixits (Blood and Tissue Bank, BST), Barcelona, Spain
| | - Rut Mora-Buch
- Advanced & Cell Therapy Services, Banc de Sang i Teixits (Blood and Tissue Bank, BST), Barcelona, Spain
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Ester Cantó
- Advanced & Cell Therapy Services, Banc de Sang i Teixits (Blood and Tissue Bank, BST), Barcelona, Spain
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Cleofé Peña-Gómez
- Mental Health and Neurosciences, Mixt Unit, Parc Taulí Research and Innovation Institute (I3PT), Barcelona, Spain
| | - Francesc Rudilla
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
- Immunogenetics and Histocompatibility Laboratory, Banc de Sang i Teixits (Blood and Tissue Bank, BST), Barcelona, Spain
| |
Collapse
|
4
|
Schweitzer L, Muranski P. Virus-specific T cell therapy to treat refractory viral infections in solid organ transplant recipients. Am J Transplant 2024; 24:1558-1566. [PMID: 38857784 DOI: 10.1016/j.ajt.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024]
Abstract
Solid organ transplant recipients require ongoing immunosuppression to prevent acute rejection, which puts them at risk of opportunistic infections. Viral infections are particularly challenging to prevent and treat as many establish latency and thus cannot be eliminated, whereas targets for small molecule antiviral medications are limited. Resistance to antivirals and unacceptable toxicity also complicate treatment. Virus-specific T cell therapies aim to restore host-specific immunity to opportunistic viruses that is lacking due to ongoing immunosuppressive therapy. This minireview will provide a state-of-the-art update of the current virus-specific T cell pipeline and translational research that is likely to lead to further treatment options for viral infections in solid organ transplant recipients.
Collapse
Affiliation(s)
- Lorne Schweitzer
- Department of Medicine, Division of Infectious Diseases, Columbia University Irving Medical Center, New York, New York, USA; Columbia Center for Translational Immunology, New York, New York, USA
| | - Pawel Muranski
- Department of Medicine, Division of Hematology, Columbia University Irving Medical Center, New York, New York, USA; Columbia Center for Translational Immunology, New York, New York, USA.
| |
Collapse
|
5
|
Sempere A, Castillo N, Rudilla F, Querol S, Enrich E, Prat-Vidal C, Codinach M, Cofan F, Torregrossa V, Dieckmann F, Bodro M. Successful BK virus-specific T cell therapy in a kidney transplant recipient with progressive multifocal leukoencephalopathy. Am J Transplant 2024; 24:1698-1702. [PMID: 38734417 DOI: 10.1016/j.ajt.2024.05.003] [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: 03/06/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
The strategy for progressive multifocal leukoencephalopathy (PML) in solid organ transplant recipients primarily focuses on reducing immunosuppressive therapy. However, this approach offers limited efficacy and carries a high risk of graft loss. Here, we present the case of a 64-year-old male kidney transplant recipient with a high degree of immunosuppression who developed PML in October 2022. Despite the standard reduction of immunosuppressive therapy, the patient's condition continued to deteriorate, as evidenced by worsening neurological symptoms and increasing JC virus (JCV) DNA levels in cerebrospinal fluid. This prompted the innovative use of BKPyV-virus-specific T cell (BKPyV-VST) therapy, given the genetic similarities between BK and JCVs. Infusion of third-party donor BKPyV-VST resulted in clinical stabilization, a significant reduction in JCV-DNA levels, and the emergence of a JCV-specific T cell response, as observed in enzyme-linked immunospot assays and TCRβ sequencing. This represents the first case report of successful third-party BKPyV-VST therapy in a kidney recipient presenting PML, without graft-versus-host disease or graft dysfunction.
Collapse
Affiliation(s)
- Abiu Sempere
- Infectious Diseases Department. Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona. Spain
| | - Nerea Castillo
- Advanced Cell Therapy Service, Banc de Sang i Teixits (Blood and Tissue Bank, BST), Barcelona, Spain; Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Francesc Rudilla
- Immunogenetics and Histocompatibility Laboratory, Banc de Sang i Teixits (Blood and Tissue Bank, BST), Barcelona, Spain; Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Sergi Querol
- Advanced Cell Therapy Service, Banc de Sang i Teixits (Blood and Tissue Bank, BST), Barcelona, Spain; Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Emma Enrich
- Immunogenetics and Histocompatibility Laboratory, Banc de Sang i Teixits (Blood and Tissue Bank, BST), Barcelona, Spain; Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Cristina Prat-Vidal
- Advanced Cell Therapy Service, Banc de Sang i Teixits (Blood and Tissue Bank, BST), Barcelona, Spain; Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Margarita Codinach
- Cell Laboratory, Banc de Sang i Teixits (Blood and Tissue Bank, BST), Barcelona, Spain; Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Frederic Cofan
- Department of Nephrology and Renal Transplantation, IDIBAPS, University of Barcelona, Barcelona. Spain
| | - Vicens Torregrossa
- Department of Nephrology and Renal Transplantation, IDIBAPS, University of Barcelona, Barcelona. Spain
| | - Fritz Dieckmann
- Department of Nephrology and Renal Transplantation, IDIBAPS, University of Barcelona, Barcelona. Spain
| | - Marta Bodro
- Infectious Diseases Department. Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona. Spain.
| |
Collapse
|
6
|
Deffner M, Schneider-Hohendorf T, Schulte-Mecklenbeck A, Falk S, Lu IN, Ostkamp P, Müller-Miny L, Schumann EM, Goelz S, Cahir-McFarland E, Thakur KT, De Jager PL, Klotz L, Meyer Zu Hörste G, Gross CC, Wiendl H, Grauer OM, Schwab N. Chemokine-mediated cell migration into the central nervous system in progressive multifocal leukoencephalopathy. Cell Rep Med 2024; 5:101622. [PMID: 38917802 PMCID: PMC11293326 DOI: 10.1016/j.xcrm.2024.101622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/10/2024] [Accepted: 06/02/2024] [Indexed: 06/27/2024]
Abstract
Progressive multifocal leukoencephalopathy (PML) has been associated with different forms of immune compromise. This study analyzes the chemokine signals and attracted immune cells in cerebrospinal fluid (CSF) during PML to define immune cell subpopulations relevant for the PML immune response. In addition to chemokines that indicate a general state of inflammation, like CCL5 and CXCL10, the CSF of PML patients specifically contains CCL2 and CCL4. Single-cell transcriptomics of CSF cells suggests an enrichment of distinct CD4+ and CD8+ T cells expressing chemokine receptors CCR2, CCR5, and CXCR3, in addition to ITGA4 and the genetic PML risk genes STXBP2 and LY9. This suggests that specific immune cell subpopulations migrate into the central nervous system to mitigate PML, and their absence might coincide with PML development. Monitoring them might hold clues for PML risk, and boosting their recruitment or function before therapeutic immune reconstitution might improve its risk-benefit ratio.
Collapse
Affiliation(s)
- Marie Deffner
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Tilman Schneider-Hohendorf
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Andreas Schulte-Mecklenbeck
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Simon Falk
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - I-Na Lu
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Patrick Ostkamp
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Louisa Müller-Miny
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Eva Maria Schumann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Susan Goelz
- Oregon Health & Science University, Portland, OR, USA; Biogen, Cambridge, MA, USA
| | | | - Kiran T Thakur
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Gerd Meyer Zu Hörste
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Catharina C Gross
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Oliver M Grauer
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Nicholas Schwab
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.
| |
Collapse
|
7
|
Nay S, Möhn N, Grote-Levi L, Bonifacius A, Saßmann ML, Karacondi K, Tischer-Zimmermann S, Pöter H, Mahmoudi N, Wattjes MP, Maecker-Kolhoff B, Höglinger G, Eiz-Vesper B, Skripuletz T. Combined treatment with allogeneic Epstein-Barr- and human polyomavirus 1 specific T-cells in progressive multifocal leukoencephalopathy and EBV infection: a case report. Ther Adv Neurol Disord 2024; 17:17562864241253917. [PMID: 38813521 PMCID: PMC11135084 DOI: 10.1177/17562864241253917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/23/2024] [Indexed: 05/31/2024] Open
Abstract
Opportunistic viral infections in individuals with severe immunodeficiency can lead to fatal conditions such as progressive multifocal leukoencephalopathy (PML), for which treatment options are limited. These infections pose significant risks, especially when co-infections with other viruses occur. We describe a combined therapy approach using directly isolated allogeneic Human Polyomavirus 1 (also known as BKV) and Epstein-Barr virus (EBV) specific cytotoxic T-cells for the treatment of PML in conjunction with identified EBV in the cerebrospinal fluid (CSF) of a male patient infected with human immunodeficiency virus (HIV). A 53-year-old HIV-positive male, recently diagnosed with PML, presented with rapidly worsening symptoms, including ataxia, tetraparesis, dysarthria, and dysphagia, leading to respiratory failure. The patient developed PML even after commencing highly active antiretroviral therapy (HAART) 3 months prior. Brain magnetic resonance imaging (MRI) revealed multifocal demyelination lesions involving the posterior fossa and right thalamus suggestive of PML. In addition to the detection of human polyomavirus 2 (also known as JCV), analysis of CSF showed positive results for EBV deoxyribonucleic acid (DNA). His neurological condition markedly deteriorated over the following 2 months. Based on MRI, there was no evidence of Immune Reconstitution Inflammatory Syndrome contributing to this decline. The patient did not have endogenous virus-specific T-cells. We initiated an allogeneic, partially human leukocyte antigen-matched transfer of EBV and utilizing the cross-reactivity between BKV and JCV-BKV specific T-cells. This intervention led to notable neurological improvement and partial resolution of the MRI lesions within 6 weeks. Our case of a patient with acquired immune deficiency syndrome demonstrates that PML and concurrent EBV co-infection can still occur despite undergoing HAART treatment. This innovative experimental therapy, involving a combination of virus-specific T-cells, was demonstrated to be an effective treatment option in this patient.
Collapse
Affiliation(s)
- Sandra Nay
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Nora Möhn
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Lea Grote-Levi
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Mieke L. Saßmann
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Kevin Karacondi
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | | - Henning Pöter
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Nima Mahmoudi
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
- Department of Neuroradiology, Charité Berlin, Corporate Member of Freie Universität zu Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mike P. Wattjes
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
- Department of Neuroradiology, Charité Berlin, Corporate Member of Freie Universität zu Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Britta Maecker-Kolhoff
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Hannover, Germany
| | - Günter Höglinger
- Department of Neurology, LMU University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Hannover, Germany
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, Hannover 30625, Germany
- Centre for Individualised Infection Medicine, Hannover, Germany
| |
Collapse
|
8
|
Rudilla F, Carrasco-Benso MP, Pasamar H, López-Montañés M, Andrés-Rozas M, Tomás-Marín M, Company D, Moya C, Larrea L, Guerreiro M, Barba P, Arbona C, Querol S. Development and characterization of a cell donor registry for virus-specific T cell manufacture in a blood bank. HLA 2024; 103:e15419. [PMID: 38450972 DOI: 10.1111/tan.15419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 01/19/2024] [Accepted: 02/16/2024] [Indexed: 03/08/2024]
Abstract
Adoptive cell therapy using virus-specific T cells (VST) is a strategy for treating common opportunistic viral infections after transplantation, particularly when these infections do not resolve through antiviral drug therapy. The availability of third-party healthy donors allows for the immediate use of cells for allogeneic therapy in cases where patients lack an appropriate donor. Here, we present the creation of a cell donor registry of human leukocyte antigen (HLA)-typed blood donors, REDOCEL, a strategic initiative to ensure the availability of compatible cells for donation when needed. Currently, the registry consists of 597 healthy donors with a median age of 29 years, 54% of whom are women. The most represented blood groups were A positive and O positive, with 36.52% and 34.51%, respectively. Also, donors were screened for cytomegalovirus (CMV) and Epstein-Barr virus (EBV). Almost 65% of donors were CMV-seropositive, while less than 5% were EBV-seronegative. Of the CMV-seropositive donors, 98% were also EBV-seropositive. High-resolution HLA-A, -B, -C, -DRB1 and -DQB1 allele and haplotype frequencies were determined in the registry. Prevalent HLA alleles and haplotypes were well represented to ensure donor-recipient HLA-matching, including alleles reported to present viral immunodominant epitopes. Since the functional establishment of REDOCEL, in May 2019, 87 effective donations have been collected, and the effective availability of donors with the first call has been greater than 75%. Thus, almost 89% of patients receiving an effective donation had available at least 5/10 HLA-matched cell donors (HLA-A, -B, -C, -DRB1, and -DQB1). To summarize, based on our experience, a cell donor registry from previously HLA-typed blood donors is a useful tool for facilitating access to VST therapy.
Collapse
Affiliation(s)
- Francesc Rudilla
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma of Barcelona (VHIR-UAB), Barcelona, Spain
- Immunogenetics and Histocompatibility Laboratory, Blood and Tissue Bank, Barcelona, Spain
| | - María Paz Carrasco-Benso
- Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (Fisabio), Valencia, Spain
| | - Helena Pasamar
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma of Barcelona (VHIR-UAB), Barcelona, Spain
- Advanced & Cell Therapy Services, Blood and Tissue Bank, Barcelona, Spain
| | - María López-Montañés
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma of Barcelona (VHIR-UAB), Barcelona, Spain
- Advanced & Cell Therapy Services, Blood and Tissue Bank, Barcelona, Spain
| | - María Andrés-Rozas
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma of Barcelona (VHIR-UAB), Barcelona, Spain
- Advanced & Cell Therapy Services, Blood and Tissue Bank, Barcelona, Spain
| | - Maria Tomás-Marín
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma of Barcelona (VHIR-UAB), Barcelona, Spain
- Advanced & Cell Therapy Services, Blood and Tissue Bank, Barcelona, Spain
| | - Desirée Company
- Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (Fisabio), Valencia, Spain
| | - Cristina Moya
- Blood Donors Management Department, Blood and Tissue Bank, Barcelona, Spain
| | - Luis Larrea
- Centro de Transfusión de la Comunitat Valenciana, Valencia, Spain
| | - Manuel Guerreiro
- Department of Hematology, La Fe Polytechnic and University Hospital, Valencia, Spain
| | - Pere Barba
- Hospital Vall d'Hebron, Barcelona, Spain
| | - Cristina Arbona
- Centro de Transfusión de la Comunitat Valenciana, Valencia, Spain
| | - Sergio Querol
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma of Barcelona (VHIR-UAB), Barcelona, Spain
- Advanced & Cell Therapy Services, Blood and Tissue Bank, Barcelona, Spain
| |
Collapse
|
9
|
Schreiber B, Tripathi S, Nikiforow S, Chandraker A. Adoptive Immune Effector Cell Therapies in Cancer and Solid Organ Transplantation: A Review. Semin Nephrol 2024; 44:151498. [PMID: 38555223 DOI: 10.1016/j.semnephrol.2024.151498] [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] [Indexed: 04/02/2024]
Abstract
Cancer is one of the most devastating complications of kidney transplantation and constitutes one of the leading causes of morbidity and mortality among solid organ transplantation (SOT) recipients. Immunosuppression, although effective in preventing allograft rejection, inherently inhibits immune surveillance against oncogenic viral infections and malignancy. Adoptive cell therapy, particularly immune effector cell therapy, has long been a modality of interest in both cancer and transplantation, though has only recently stepped into the spotlight with the development of virus-specific T-cell therapy and chimeric antigen receptor T-cell therapy. Although these modalities are best described in hematopoietic cell transplantation and hematologic malignancies, their potential application in the SOT setting may hold tremendous promise for those with limited therapeutic options. In this review, we provide a brief overview of the development of adoptive cell therapies with a focus on virus-specific T-cell therapy and chimeric antigen receptor T-cell therapy. We also describe the current experience of these therapies in the SOT setting as well as the challenges in their application and future directions in their development.
Collapse
Affiliation(s)
- Brittany Schreiber
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sudipta Tripathi
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sarah Nikiforow
- Division of Medical Oncology, Department of Medicine, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Anil Chandraker
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Renal Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA.
| |
Collapse
|
10
|
Ardakani R, Jia L, Matthews E, Thakur KT. Therapeutic advances in neuroinfectious diseases. Ther Adv Infect Dis 2024; 11:20499361241274246. [PMID: 39314743 PMCID: PMC11418331 DOI: 10.1177/20499361241274246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 07/05/2024] [Indexed: 09/25/2024] Open
Abstract
There have been several major advances in therapeutic options for the treatment of neurological infections over the past two decades. These advances encompass both the development of new antimicrobial therapies and the repurposing of existing agents for new indications. In addition, advances in our understanding of the host immune response have allowed for the development of new immunomodulatory strategies in the treatment of neurological infections. This review focuses on the key advances in the treatment of neurological infections, including viral, bacterial, fungal, and prion diseases, with a particular focus on immunomodulatory treatment options. This review also highlights the process by which clinicians can request access to therapeutic agents on a compassionate or emergency basis when they may not be commercially available. While many therapeutic advances have been achieved in the past several years, there remains a pressing need for the continued development of additional therapeutic agents in the treatment of neurological infections.
Collapse
Affiliation(s)
- Rumyar Ardakani
- Neuro-Infectious Diseases Group, Department of Neurology and Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lucy Jia
- Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Elizabeth Matthews
- Neuro-Infectious Diseases Group, Department of Neurology and Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kiran T. Thakur
- Department of Neurology, Columbia University Irving Medical Center, 177 Fort Washington Avenue, Milstein Hospital, 8GS-300, New York, NY 10032, USA
- Program in Neuroinfectious Diseases, Department of Neurology, Columbia University Irving Medical Center-New York Presbyterian Hospital
| |
Collapse
|
11
|
Soni N, Ora M, Mangla R, Singh R, Ellika S, Agarwal A, Meyers SP, Bathla G. Radiological abnormalities in progressive multifocal leukoencephalopathy: Identifying typical and atypical imaging patterns for early diagnosis and differential considerations. Mult Scler Relat Disord 2023; 77:104830. [PMID: 37418930 DOI: 10.1016/j.msard.2023.104830] [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: 03/29/2023] [Revised: 05/22/2023] [Accepted: 06/11/2023] [Indexed: 07/09/2023]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a rare viral central nervous system (CNS) demyelinating disease primarily associated with a compromised immune system. PML is seen mainly in individuals with human immunodeficiency virus, lymphoproliferative disease, and multiple sclerosis. Patients on immunomodulators, chemotherapy, and solid organ or bone marrow transplants are predisposed to PML. Recognition of various PML-associated typical and atypical imaging abnormalities is critical for early diagnosis and differentiating it from other conditions, especially in high-risk populations. Early PML recognition should expedite efforts at immune-system restoration, allowing for a favorable outcome. This review aims to provide a practical overview of radiological abnormalities in PML patients and address differential considerations.
Collapse
Affiliation(s)
- Neetu Soni
- Radiodiagnosis (Neuroradiology and Nuclear Medicine), University of Rochester Medical Center, Rochester, NY 14618, USA.
| | - Manish Ora
- Department of Nuclear Medicine, SGPGIMS, Lucknow, Uttar Pradesh, India
| | | | - Rohit Singh
- Division of Hematology-Oncology at the University of Vermont Medical Center, Burlington, VT, USA
| | - Shehanaz Ellika
- Radiodiagnosis (Neuroradiology and Nuclear Medicine), University of Rochester Medical Center, Rochester, NY 14618, USA
| | - Amit Agarwal
- Radiology, Mayo Clinic in Florida, San Pablo Dr, Jacksonville, FL 32224-1865, USA
| | - Steven P Meyers
- Radiodiagnosis (Neuroradiology and Nuclear Medicine), University of Rochester Medical Center, Rochester, NY 14618, USA
| | | |
Collapse
|
12
|
Grote-Levi L, Möhn N, Bonifacius A, Tischer-Zimmermann S, Schweitzer F, Mahmoudi N, Silling S, Warnke C, Maecker-Kolhoff B, Wattjes MP, Eiz-Vesper B, Höglinger GU, Skripuletz T. Adoptive Allogeneic T-Cell Therapy Improves the Clinical Outcome of JC Virus Granule Cell Neuronopathy: A Case Report. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200138. [PMID: 37385737 PMCID: PMC10474852 DOI: 10.1212/nxi.0000000000200138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/08/2023] [Indexed: 07/01/2023]
Abstract
OBJECTIVES JC virus granule cell neuronopathy is a potentially fatal otherwise highly disabling disease without an approved therapeutic option. This case report presents the positive record to T-cell therapy in JC virus granule cell neuronopathy. METHODS The patient represented with subacute cerebellar symptoms. Diagnosis of JC virus granule cell neuronopathy was made because of infratentorially accentuated brain volume atrophy shown by brain MRI and the detection of JC virus DNA in the CSF. RESULTS Six doses of virus-specific T cells were administered. Within 12 months after therapy initiation, the patient showed clear clinical benefit with improvement of symptoms, and JC viral DNA load significantly declined. DISCUSSION We present the case report of a positive response to T-cell therapy in JC virus granule cell neuronopathy, leading to an improvement of symptoms.
Collapse
Affiliation(s)
- Lea Grote-Levi
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.)
| | - Nora Möhn
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.)
| | - Agnes Bonifacius
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.)
| | - Sabine Tischer-Zimmermann
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.)
| | - Finja Schweitzer
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.)
| | - Nima Mahmoudi
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.)
| | - Steffi Silling
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.)
| | - Clemens Warnke
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.)
| | - Britta Maecker-Kolhoff
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.)
| | - Mike P Wattjes
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.)
| | - Britta Eiz-Vesper
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.)
| | - Günter U Höglinger
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.)
| | - Thomas Skripuletz
- From the Department of Neurology (L.G.-L., N. Möhn, G.U.H., T.S.); Institute of Transfusion Medicine and Transplant Engineering (A.B., S.T.-Z., B.E.-V.), Hannover Medical School; Department of Neurology (F.S., C.W.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Department of Diagnostic and Interventional Neuroradiology (N. Mahmoudi, M.P.W.), Hannover Medical School; National Reference Center for Papilloma- and Polyomaviruses (S.S.), Institute of Virology, University of Cologne; Department of Pediatric Hematology and Oncology (B.M.-K.), Hannover Medical School, Germany; and German Center for Infection Research (DZIF) (B.M.-K., B.E.-V.).
| |
Collapse
|
13
|
Lambert N, El Moussaoui M, Baron F, Maquet P, Darcis G. Virus-Specific T-Cell Therapy for Viral Infections of the Central Nervous System: A Review. Viruses 2023; 15:1510. [PMID: 37515196 PMCID: PMC10383098 DOI: 10.3390/v15071510] [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: 06/08/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Opportunistic viral infections of the central nervous system represent a significant cause of morbidity and mortality among an increasing number of immunocompromised patients. Since antiviral treatments are usually poorly effective, the prognosis generally relies on the ability to achieve timely immune reconstitution. Hence, strategies aimed at reinvigorating antiviral immune activity have recently emerged. Among these, virus-specific T-cells are increasingly perceived as a principled and valuable tool to treat opportunistic viral infections. Here we briefly discuss how to develop and select virus-specific T-cells, then review their main indications in central nervous system infections, including progressive multifocal leukoencephalopathy, CMV infection, and adenovirus infection. We also discuss their potential interest in the treatment of progressive multiple sclerosis, or EBV-associated central nervous system inflammatory disease. We finish with the key future milestones of this promising treatment strategy.
Collapse
Affiliation(s)
- Nicolas Lambert
- Department of Neurology, University Hospital of Liège, 4000 Liège, Belgium
| | - Majdouline El Moussaoui
- Department of General Internal Medicine and Infectious Diseases, University Hospital of Liège, 4000 Liège, Belgium
| | - Frédéric Baron
- Department of Hematology, University Hospital of Liège, 4000 Liège, Belgium
| | - Pierre Maquet
- Department of Neurology, University Hospital of Liège, 4000 Liège, Belgium
| | - Gilles Darcis
- Department of General Internal Medicine and Infectious Diseases, University Hospital of Liège, 4000 Liège, Belgium
| |
Collapse
|
14
|
McEntire CRS, Fletcher A, Toledano M, Epstein S, White E, Tan CS, Mao-Draayer Y, Banks SA, Aksamit AJ, Gelfand JM, Thakur KT, Anand P, Cortese I, Bhattacharyya S. Characteristics of Progressive Multifocal Leukoencephalopathy Associated With Sarcoidosis Without Therapeutic Immune Suppression. JAMA Neurol 2023; 80:624-633. [PMID: 37093609 PMCID: PMC10126944 DOI: 10.1001/jamaneurol.2023.0841] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/17/2023] [Indexed: 04/25/2023]
Abstract
Importance Progressive multifocal leukoencephalopathy can occur in the context of systemic sarcoidosis (S-PML) in the absence of therapeutic immune suppression and can initially be mistaken for neurosarcoidosis or other complications of sarcoidosis. Earlier recognition of S-PML could lead to more effective treatment of the disease. Objective To describe characteristics of patients with S-PML. Design, Setting, and Participants For this case series, records from 8 academic medical centers in the United States were reviewed from 2004 to 2022. A systematic review of literature from 1955 to 2022 yielded data for additional patients. Included were patients with S-PML who were not receiving therapeutic immune suppression. The median follow-up time for patients who survived the acute range of illness was 19 months (range, 2-99). Data were analyzed in February 2023. Exposures Sarcoidosis without active therapeutic immune suppression. Main Outcomes and Measures Clinical, laboratory, and radiographic features of patients with S-PML. Results Twenty-one patients with S-PML not receiving therapeutic immune suppression were included in this study, and data for 37 patients were collected from literature review. The median age of the 21 study patients was 56 years (range, 33-72), 4 patients (19%) were female, and 17 (81%) were male. The median age of the literature review patients was 49 years (range, 21-74); 12 of 34 patients (33%) with reported sex were female, and 22 (67%) were male. Nine of 21 study patients (43%) and 18 of 31 literature review patients (58%) had simultaneous presentation of systemic sarcoidosis and PML. Six of 14 study patients (43%) and 11 of 19 literature review patients (58%) had a CD4+ T-cell count greater than 200/μL. In 2 study patients, a systemic flare of sarcoidosis closely preceded S-PML development. Ten of 17 study patients (59%) and 21 of 35 literature review patients (60%) died during the acute phase of illness. No meaningful predictive differences were found between patients who survived S-PML and those who did not. Conclusions and Relevance In this case series, patients with sarcoidosis developed PML in the absence of therapeutic immune suppression, and peripheral blood proxies of immune function were often only mildly abnormal. Systemic sarcoidosis flares may rarely herald the onset of S-PML. Clinicians should consider PML in any patient with sarcoidosis and new white matter lesions on brain magnetic resonance imaging.
Collapse
Affiliation(s)
| | - Anita Fletcher
- Neuroimmunology Clinic, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Michel Toledano
- Department of Neurology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Samantha Epstein
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut
| | - Emily White
- Department of Neurology, Boston Medical Center, Boston, Massachusetts
| | - C. Sabrina Tan
- Division of Infectious Diseases, Center for Virology and Vaccines Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Division of Infectious Diseases, Department of Medicine, University of Iowa, Iowa City
| | | | - Samantha A. Banks
- Department of Neurology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Allen J. Aksamit
- Department of Neurology, Mayo Clinic Rochester, Rochester, Minnesota
| | | | - Kiran T. Thakur
- Department of Neurology, Columbia University Irving Medical Center–New York Presbyterian Hospital, New York
| | - Pria Anand
- Department of Neurology, Boston Medical Center, Boston, Massachusetts
| | - Irene Cortese
- Experimental Immunotherapeutics Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | | |
Collapse
|
15
|
Pathogen-specific T Cells: Targeting Old Enemies and New Invaders in Transplantation and Beyond. Hemasphere 2023; 7:e809. [PMID: 36698615 PMCID: PMC9831191 DOI: 10.1097/hs9.0000000000000809] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/07/2022] [Indexed: 01/27/2023] Open
Abstract
Adoptive immunotherapy with virus-specific cytotoxic T cells (VSTs) has evolved over the last three decades as a strategy to rapidly restore virus-specific immunity to prevent or treat viral diseases after solid organ or allogeneic hematopoietic cell-transplantation (allo-HCT). Since the early proof-of-principle studies demonstrating that seropositive donor-derived T cells, specific for the commonest pathogens post transplantation, namely cytomegalovirus or Epstein-Barr virus (EBV) and generated by time- and labor-intensive protocols, could effectively control viral infections, major breakthroughs have then streamlined the manufacturing process of pathogen-specific T cells (pSTs), broadened the breadth of target recognition to even include novel emerging pathogens and enabled off-the-shelf administration or pathogen-naive donor pST production. We herein review the journey of evolution of adoptive immunotherapy with nonengineered, natural pSTs against infections and virus-associated malignancies in the transplant setting and briefly touch upon recent achievements using pSTs outside this context.
Collapse
|
16
|
Schmidt K, Skusa R, Großmann A. [Fatal neurological side effect of anti-CD20 antibody treatment]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2023; 64:193-196. [PMID: 36422661 PMCID: PMC9894990 DOI: 10.1007/s00108-022-01431-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/13/2022] [Indexed: 11/27/2022]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a rare, often fatal infection of the central nervous system caused by reactivation of John Cunningham virus (JCV). The case of a 59-year-old woman presenting with neurological disorders after treatment of her relapsed lymphoma with rituximab, among others, is reported. Magnetic resonance imaging showed fast-growing white matter lesions of both hemisphere and cerebellar that were neither space-consuming nor enhancing contrast media. Clinical and radiological suspicion of PML was confirmed by detection of JCV-DNA in cerebrospinal fluid. The patient died from devastating neurological decline only 11 days after the diagnosis was made. Due to the wider indication of monoclonal antibodies in almost every medical specialty we must always consider iatrogenic PML in addition to classic PML associated with acquired immunodeficiency syndrome (AIDS).
Collapse
Affiliation(s)
- Kathie Schmidt
- Zentrum für Innere Medizin, Medizinische Klinik III, Klinik für Hämatologie, Onkologie und Palliativmedizin, Universitätsmedizin Rostock, Ernst-Heydemann-Str. 6, 18057 Rostock, Deutschland
| | - Romy Skusa
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsmedizin Rostock, Rostock, Deutschland
| | - Annette Großmann
- Institut für Diagnostische und Interventionelle Radiologie, Kinder- und Neuroradiologie, Abteilung Neuroradiologie, Campus Gehlsdorf, Universitätsmedizin Rostock, Rostock, Deutschland
| |
Collapse
|
17
|
Wiercinska E, Bönig H. Zelltherapie in den Zeiten von SARS-CoV-2. TRANSFUSIONSMEDIZIN 2022. [DOI: 10.1055/a-1720-7975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
ZusammenfassungEin breites Spektrum von Disruptionen, aber auch blitzschnelle Innovationen, hat
die SARS-CoV-2 Pandemie gebracht. Dieser Übersichtsartikel betrachtet
die Pandemie aus der Warte der Zelltherapie; konkret werden vier Aspekte
untersucht: Wie unterscheiden sich die Risiken von Zelltherapie-Patienten mit
SARS-CoV-2 Infektion und COVID von denen der Allgemeinbevölkerung? Sind
Empfänger von Zelltherapien, hier speziell autologe und allogene
Stammzelltransplantationsempfänger sowie Empfänger von
CAR-T-Zell-Präparaten, klinisch relevant durch SARS-CoV-2 Vakzine
immunisierbar? Welche Auswirkungen hat die Pandemie mit Spenderausfallrisiko und
Zusammenbruch von Supply Chains auf die Versorgung mit Zelltherapeutika? Gibt es
Zelltherapeutika, die bei schwerem COVID therapeutisch nutzbringend eingesetzt
werden können? In aller Kürze, das erwartete massiv
erhöhte Risiko von Zelltherapie-Patienten, im Infektionsfall einen
schweren Verlauf zu erleiden oder zu sterben, wurde bestätigt. Die
Vakzine induziert jedoch bei vielen dieser Patienten humorale und
zelluläre Immunität, wenn auch weniger zuverlässig als
bei Gesunden. Dank kreativer Lösungen gelang es, die Versorgung mit
Zelltherapeutika im Wesentlichen uneingeschränkt aufrecht zu erhalten.
SARS-CoV-2-spezifische T-Zell-Präparate für den adoptiven
Immuntransfer wurden entwickelt, eine therapeutische Konstellation diese
anzuwenden ergab sich jedoch nicht. Therapiestudien mit mesenchymalen
Stromazellen beim schweren COVID laufen weltweit; die Frage der Wirksamkeit
bleibt zurzeit offen, bei jedoch substanziellem Optimismus in der Szene. Einige
der Erkenntnisse und Innovationen aus der SARS-CoV-2-Pandemie können
möglicherweise verallgemeinert werden und so auf die Zeit nach ihrem
Ende langfristig nachwirken.
Collapse
Affiliation(s)
- Eliza Wiercinska
- DRK-Blutspendedienst Baden-Württemberg-Hessen, Institut
Frankfurt, Frankfurt a.M
| | - Halvard Bönig
- DRK-Blutspendedienst Baden-Württemberg-Hessen, Institut
Frankfurt, Frankfurt a.M
- Goethe Universität, Institut für Transfusionsmedizin
und Immunhämatologie, Frankfurt a.M
- University of Washington, Seattle, WA
| |
Collapse
|
18
|
Bonifacius A, Tischer-Zimmermann S, Santamorena MM, Mausberg P, Schenk J, Koch S, Barnstorf-Brandes J, Gödecke N, Martens J, Goudeva L, Verboom M, Wittig J, Maecker-Kolhoff B, Baurmann H, Clark C, Brauns O, Simon M, Lang P, Cornely OA, Hallek M, Blasczyk R, Seiferling D, Köhler P, Eiz-Vesper B. Rapid Manufacturing of Highly Cytotoxic Clinical-Grade SARS-CoV-2-specific T Cell Products Covering SARS-CoV-2 and Its Variants for Adoptive T Cell Therapy. Front Bioeng Biotechnol 2022; 10:867042. [PMID: 35480981 PMCID: PMC9036989 DOI: 10.3389/fbioe.2022.867042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Objectives: Evaluation of the feasibility of SARS-CoV-2-specific T cell manufacturing for adoptive T cell transfer in COVID-19 patients at risk to develop severe disease. Methods: Antiviral SARS-CoV-2-specific T cells were detected in blood of convalescent COVID-19 patients following stimulation with PepTivator SARS-CoV-2 Select using Interferon-gamma Enzyme-Linked Immunospot (IFN-γ ELISpot), SARS-CoV-2 T Cell Analysis Kit (Whole Blood) and Cytokine Secretion Assay (CSA) and were characterized with respect to memory phenotype, activation state and cytotoxic potential by multicolor flow cytometry, quantitative real-time PCR and multiplex analyses. Clinical-grade SARS-CoV-2-specific T cell products were generated by stimulation with MACS GMP PepTivator SARS-CoV-2 Select using CliniMACS Prodigy and CliniMACS Cytokine Capture System (IFN-gamma) (CCS). Functionality of enriched T cells was investigated in cytotoxicity assays and by multiplex analysis of secreted cytotoxic molecules upon target recognition. Results: Donor screening via IFN-γ ELISpot allows for pre-selection of potential donors for generation of SARS-CoV-2-specific T cells. Antiviral T cells reactive against PepTivator SARS-CoV-2 Select could be magnetically enriched from peripheral blood of convalescent COVID-19 patients by small-scale CSA resembling the clinical-grade CCS manufacturing process and showed an activated and cytotoxic T cell phenotype. Four clinical-grade SARS-CoV-2-specific T cell products were successfully generated with sufficient cell numbers and purities comparable to those observed in donor pretesting via CSA. The T cells in the generated products were shown to be capable to replicate, specifically recognize and kill target cells in vitro and secrete cytotoxic molecules upon target recognition. Cell viability, total CD3+ cell number, proliferative capacity and cytotoxic potential remained stable throughout storage of up to 72 h after end of leukapheresis. Conclusion: Clinical-grade SARS-CoV-2-specific T cells are functional, have proliferative capacity and target-specific cytotoxic potential. Their function and phenotype remain stable for several days after enrichment. The adoptive transfer of partially matched, viable human SARS-CoV-2-specific T lymphocytes collected from convalescent individuals may provide the opportunity to support the immune system of COVID-19 patients at risk for severe disease.
Collapse
Affiliation(s)
- Agnes Bonifacius
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Sabine Tischer-Zimmermann
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Maria Michela Santamorena
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Philip Mausberg
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Josephine Schenk
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Stephanie Koch
- Deutsche Gesellschaft für Gewebetransplantation, Hannover, Germany
| | - Johanna Barnstorf-Brandes
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Nina Gödecke
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Jörg Martens
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Lilia Goudeva
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Murielle Verboom
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | - Jana Wittig
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Britta Maecker-Kolhoff
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | | | - Caren Clark
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Olaf Brauns
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Martina Simon
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Peter Lang
- Department of Pediatric Hematology and Oncology, University Children's Hospital, University of Tuebingen, Tuebingen, Germany
| | - Oliver A Cornely
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, Cologne, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Rainer Blasczyk
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| | | | - Philipp Köhler
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Britta Eiz-Vesper
- Hannover Medical School, Institute of Transfusion Medicine and Transplant Engineering, Hannover, Germany
| |
Collapse
|
19
|
Innovative therapeutic concepts of progressive multifocal leukoencephalopathy. J Neurol 2022; 269:2403-2413. [PMID: 34994851 PMCID: PMC8739669 DOI: 10.1007/s00415-021-10952-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 02/04/2023]
Abstract
Progressive multifocal leukoencephalopathy (PML) is an opportunistic viral disease of the brain-caused by human polyomavirus 2. It affects patients whose immune system is compromised by a corresponding underlying disease or by drugs. Patients with an underlying lymphoproliferative disease have the worst prognosis with a mortality rate of up to 90%. Several therapeutic strategies have been proposed but failed to show any benefit so far. Therefore, the primary therapeutic strategy aims to reconstitute the impaired immune system to generate an effective endogenous antiviral response. Recently, anti-PD-1 antibodies and application of allogeneic virus-specific T cells demonstrated promising effects on the outcome in individual PML patients. This article aims to provide a detailed overview of the literature with a focus on these two treatment approaches.
Collapse
|
20
|
Schulze Lammers FC, Bonifacius A, Tischer-Zimmermann S, Goudeva L, Martens J, Lepenies B, von Karpowitz M, Einecke G, Beutel G, Skripuletz T, Blasczyk R, Beier R, Maecker-Kolhoff B, Eiz-Vesper B. Antiviral T-Cell Frequencies in a Healthy Population: Reference Values for Evaluating Antiviral Immune Cell Profiles in Immunocompromised Patients. J Clin Immunol 2022; 42:546-558. [PMID: 34989946 PMCID: PMC9015970 DOI: 10.1007/s10875-021-01205-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/23/2021] [Indexed: 11/30/2022]
Abstract
Viral infections and reactivations are major causes of morbidity and mortality after hematopoietic stem cell (HSCT) and solid organ transplantation (SOT) as well as in patients with immunodeficiencies. Latent herpesviruses (e.g., cytomegalovirus, Epstein-Barr virus, and human herpesvirus 6), lytic viruses (e.g., adenovirus), and polyomaviruses (e.g., BK virus, JC virus) can cause severe complications. Antiviral drugs form the mainstay of treatment for viral infections and reactivations after transplantation, but they have side effects and cannot achieve complete viral clearance without prior reconstitution of functional antiviral T-cell immunity. The aim of this study was to establish normal ranges for virus-specific T-cell (VST) frequencies in healthy donors. Such data are needed for better interpretation of VST frequencies observed in immunocompromised patients. Therefore, we measured the frequencies of VSTs against 23 viral protein-derived peptide pools from 11 clinically relevant human viruses in blood from healthy donors (n = 151). Specifically, we determined the VST frequencies by interferon-gamma enzyme-linked immunospot assay and classified their distribution according to age and gender to allow for a more specific evaluation and prediction of antiviral immune responses. The reference values established here provide an invaluable tool for immune response evaluation, intensity of therapeutic drugs and treatment decision-making in immunosuppressed patients. This data should make an important contribution to improving the assessment of immune responses in immunocompromised patients.
Collapse
Affiliation(s)
- Friederike C Schulze Lammers
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, DE, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, DE, Germany
| | - Sabine Tischer-Zimmermann
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, DE, Germany
| | - Lilia Goudeva
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, DE, Germany
| | - Jörg Martens
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, DE, Germany
| | - Bernd Lepenies
- Institute for Immunology & Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, DE, Germany
| | | | - Gunilla Einecke
- Department of Nephrology, Hannover Medical School, Hannover, DE, Germany
| | - Gernot Beutel
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, DE, Germany
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, Hannover, DE, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, DE, Germany
| | - Rita Beier
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, DE, Germany
| | - Britta Maecker-Kolhoff
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, DE, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, DE, Germany.
| |
Collapse
|
21
|
Beyond antivirals: virus-specific T-cell immunotherapy for BK virus haemorrhagic cystitis and JC virus progressive multifocal leukoencephalopathy. Curr Opin Infect Dis 2021; 34:627-634. [PMID: 34751182 DOI: 10.1097/qco.0000000000000794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW The clinical manifestations of the polyomaviruses BK and JC in immunocompromised patients include BK virus (BKV) induced haemorrhagic cystitis and nephropathy, and JC virus (JCV) associated progressive multifocal leukoencephalopathy (PML) and are typically a consequence of impaired adaptive immunity in the host. To date, little clinical success has been achieved with antiviral agents or other drug therapies to treat these conditions. Here we review the methods and outcomes of the most recent clinical studies utilising adoptive immunotherapy with BK and/or JC virus-specific T-cells (VST) as either prophylaxis or treatment alternatives. RECENT FINDINGS In the last 12-18 months, several clinical trials have been published in the post-haemopoietic stem cell transplant (HSCT) setting showing good clinical success with the use of VST for treatment of BK viremia ± haemorrhagic cystitis. Between 82 and 100% clinical response has been observed in haemorrhagic cystitis using either third-party or donor-derived VST. The therapy was well tolerated with few cases of graft versus host disease in HSCT recipients, but immune mediated renal allograft loss was observed in one renal transplant recipient. Studies using BKV/JCV VST to treat PML are hindered by few patients who are sufficiently stable to receive VST. In a condition that otherwise carries such poor prognosis, VST were associated with clearance of JC virus, clinical and radiological improvement in some patients. Immune reconstitution inflammatory syndrome was a noted adverse event. SUMMARY Restoration of BK and JC virus immunity using VST immunotherapy has shown good clinical outcomes in BKV associated infections. Further evaluation with the administration of VST earlier in the course of disease is warranted for the treatment of BKV associated nephropathy in renal allograft and in JCV PML. In both indications, larger cohorts and standardisation of dosing and outcome measures would be of benefit.
Collapse
|
22
|
Baldassari LE, Wattjes MP, Cortese ICM, Gass A, Metz I, Yousry T, Reich DS, Richert N. The neuroradiology of progressive multifocal leukoencephalopathy: a clinical trial perspective. Brain 2021; 145:426-440. [PMID: 34791056 DOI: 10.1093/brain/awab419] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/29/2021] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is an opportunistic infection of the central nervous system caused by the JC virus, which infects white and grey matter cells and leads to irreversible demyelination and neuroaxonal damage. Brain magnetic resonance imaging (MRI), in addition to the clinical presentation and demonstration of JC virus DNA either in the CSF or by histopathology, is an important tool in the detection of PML. In clinical practice, standard MRI pulse sequences are utilized for screening, diagnosis, and monitoring of PML, but validated imaging-based outcome measures for use in prospective, interventional clinical trials for PML have yet to be established. We review the existing literature regarding the use of MRI and positron emission tomography imaging in PML and discuss the implications of PML histopathology for neuroradiology. MRI not only demonstrates the localization and extent of PML lesions, but also mirrors the tissue destruction, ongoing viral spread, and resulting inflammation. Finally, we explore the potential for imaging measures to serve as an outcome in PML clinical trials and provide recommendations for current and future imaging outcome measure development in this area.
Collapse
Affiliation(s)
- Laura E Baldassari
- Division of Neurology 2, Office of Neuroscience, Office of New Drugs, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Mike P Wattjes
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, 30625 Hannover, Germany
| | - Irene C M Cortese
- Neuroimmunology Clinic, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Achim Gass
- Department of Neurology/Neuroimaging, Mannheim Center of Translational Neuroscience, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Imke Metz
- Institute of Neuropathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Tarek Yousry
- Neuroradiological Academic Unit, UCL IoN; Lysholm Department of Neuroradiology, UCLH National Hospital for Neurology and Neurosurgery, London, UK
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | | |
Collapse
|
23
|
PML: BK-Virus-spezifische T-Zell-Therapie erfolgreich eingesetzt. TRANSFUSIONSMEDIZIN 2021. [DOI: 10.1055/a-1580-4044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
24
|
Bernard-Valnet R, Koralnik IJ, Du Pasquier R. Advances in Treatment of Progressive Multifocal Leukoencephalopathy. Ann Neurol 2021; 90:865-873. [PMID: 34405435 PMCID: PMC9291129 DOI: 10.1002/ana.26198] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 12/19/2022]
Abstract
Progressive multifocal encephalopathy (PML) is a severe demyelinating disease of the central nervous system (CNS) caused by JC virus (JCV), which occurs in immunocompromised individuals. Management of PML relies on restoration of immunity within the CNS. However, when this restoration cannot be readily achieved, PML has a grim prognosis. Innovative strategies have shown promise in promoting anti‐JCV immune responses, and include T‐cell adoptive transfer or immune checkpoint inhibitor therapies. Conversely, management of immune reconstitution inflammatory syndrome, particularly in iatrogenic PML, remains a major challenge. In this paper, we review recent development in the treatment of PML. ANN NEUROL 2021;90:865–873
Collapse
Affiliation(s)
- Raphaël Bernard-Valnet
- Service of Neurology, Department of Clinical Neurosciences, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Igor J Koralnik
- Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Renaud Du Pasquier
- Service of Neurology, Department of Clinical Neurosciences, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|