1
|
Wistinghausen B, Toner K, Barkauskas DA, Jerkins LP, Kinoshita H, Chansky P, Pezzella G, Saguilig L, Hayashi RJ, Abhyankar H, Scull B, Karri V, Tanna J, Hanley P, Hermiston ML, Allen CE, Bollard CM. Durable immunity to EBV after rituximab and third-party LMP-specific T cells: a Children's Oncology Group study. Blood Adv 2024; 8:1116-1127. [PMID: 38163318 PMCID: PMC10909726 DOI: 10.1182/bloodadvances.2023010832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/07/2023] [Accepted: 11/12/2023] [Indexed: 01/03/2024] Open
Abstract
ABSTRACT Posttransplant lymphoproliferative disease (PTLD) in pediatric solid organ transplant (SOT) recipients is characterized by uncontrolled proliferation of Epstein-Barr virus-infected (EBV+) B cells due to decreased immune function. This study evaluated the feasibility, safety, clinical and immunobiological outcomes in pediatric SOT recipients with PTLD treated with rituximab and third-party latent membrane protein-specific T cells (LMP-TCs). Newly diagnosed (ND) patients without complete response to rituximab and all patients with relapsed/refractory (R/R) disease received LMP-TCs. Suitable LMP-TC products were available for all eligible subjects. Thirteen of 15 patients who received LMP-TCs were treated within the prescribed 14-day time frame. LMP-TC therapy was generally well tolerated. Notable adverse events included 3 episodes of rejection in cardiac transplant recipients during LMP-TC therapy attributed to subtherapeutic immunosuppression and 1 episode of grade 3 cytokine release syndrome. Clinical outcomes were associated with disease severity. Overall response rate (ORR) after LMP-TC cycle 1 was 70% (7/10) for the ND cohort and 20% (1/5) for the R/R cohort. For all cohorts combined, the best ORR for LMP-TC cycles 1 and 2 was 53% and the 2-year overall survival was 70.7%. vβT-cell receptor sequencing showed persistence of adoptively transferred third-party LMP-TCs for up to 8 months in the ND cohort. This study establishes the feasibility of administering novel T-cell therapies in a cooperative group clinical trial and demonstrates the potential for positive outcomes without chemotherapy for ND patients with PTLD. This trial was registered at www.clinicaltrials.gov as #NCT02900976 and at the Children's Oncology Group as ANHL1522.
Collapse
Affiliation(s)
- Birte Wistinghausen
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
- The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Keri Toner
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
- The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Donald A. Barkauskas
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- Children’s Oncology Group Statistics and Data Center, Monrovia, CA
| | - Lauren P Jerkins
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Hannah Kinoshita
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
- The George Washington University School of Medicine and Health Sciences, Washington, DC
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Pamela Chansky
- The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Gloria Pezzella
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
| | - Lauren Saguilig
- Children’s Oncology Group Statistics and Data Center, Monrovia, CA
| | - Robert J. Hayashi
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis Children’s Hospital, St. Louis, MO
| | - Harshal Abhyankar
- Baylor College of Medicine, Texas Children’s Hospital Cancer Center, Houston, TX
| | - Brooks Scull
- Baylor College of Medicine, Texas Children’s Hospital Cancer Center, Houston, TX
| | | | - Jay Tanna
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
| | - Patrick Hanley
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
- The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Michelle L. Hermiston
- Department of Pediatrics, Benioff Children’s Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | - Carl E. Allen
- Baylor College of Medicine, Texas Children’s Hospital Cancer Center, Houston, TX
| | - Catherine M. Bollard
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
- The George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
2
|
Kuchar E, Karlikowska-Skwarnik M, Wawrzuta D. Anti-Inflammatory Therapy of Infections. ENCYCLOPEDIA OF INFECTION AND IMMUNITY 2022. [PMCID: PMC8496905 DOI: 10.1016/b978-0-12-818731-9.00181-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Anti-inflammatory treatment of infections is challenging due to the heterogeneity of etiologic agents and complex immune interactions. Nevertheless, anti-inflammatory medications are commonly used in infections to reduce unpleasant symptoms and to modify host response. They may play a fundamental role in managing infection with over-inflammation by decreasing inflammatory organ damage, e.g., COVID-19. However, by its inherent inhibition of immune functions, they might also contribute to the development of serious bacterial infections. Moreover, reducing a patient's symptoms and signs may provide a false sense of security and delay diagnosing threatening infections.
Collapse
|
3
|
Di Napoli ML, Rohan V, Nadig SN, Lin A. Small Bowel Perforation as the Initial Manifestation of Post-Transplant Lymphoproliferative Disorder in a Kidney and Pancreas Transplant Recipient: A Case Report. Transplant Proc 2020; 52:2827-2830. [PMID: 32900475 DOI: 10.1016/j.transproceed.2020.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/02/2020] [Indexed: 11/16/2022]
Abstract
Post-transplant lymphoproliferative disorder (PTLD) comprises a broad spectrum of diseases and is a rare but serious complication of solid organ transplantation. We report the case of a 45-year-old simultaneous pancreas and kidney (SPK) transplant recipient with diffuse, early-onset PTLD, manifesting as jejunal perforation at 6 months after transplantation. The patient underwent urgent small bowel resection of the affected portion of jejunum. The surgical pathology report was significant for diffuse large B-cell lymphoma. Subsequently, the patient underwent a full workup, including upper and lower endoscopy and whole-body positron emission tomography that revealed involvement of the axial skeleton and multiple abdominal organs with sparing of the grafts. He was treated with rituximab and intrathecal methotrexate for central nervous system prophylaxis. The patient experienced complete resolution of disease by positron emission tomography 8 months after initial presentation. We found no previous report in the literature of intestinal perforation as the initial presentation of PTLD in SPK transplant recipients.
Collapse
Affiliation(s)
- Marissa L Di Napoli
- Division of Transplant Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Vinayak Rohan
- Division of Transplant Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Satish N Nadig
- Division of Transplant Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Angello Lin
- Division of Transplant Surgery, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
4
|
Metzger ML, Mauz-Körholz C. Epidemiology, outcome, targeted agents and immunotherapy in adolescent and young adult non-Hodgkin and Hodgkin lymphoma. Br J Haematol 2019; 185:1142-1157. [PMID: 30729493 DOI: 10.1111/bjh.15789] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The epidemiology, outcome and targeted immunotherapy in adolescent and young adult non-Hodgkin and Hodgkin lymphoma were discussed during the 6th International Symposium on Childhood, Adolescent and Young Adult Non-Hodgkin Lymphoma September 26th-29th 2018 in Rotterdam, the Netherlands. This review summarizes some of those presentations, as well as other current and novel antibody therapy, immune check-point inhibitors, chimeric antigen receptor T cells, cancer vaccines and cytotoxic T lymphocyte therapy.
Collapse
Affiliation(s)
- Monika L Metzger
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Christine Mauz-Körholz
- Pädiatrische Hämatologie und Onkologie, Justus-Liebig-Universität Gießen and Medical Faculty of the Martin-Luther University of Halle, Germany
| |
Collapse
|
5
|
Gan R, Xie X, He J, Liu X, Hong L, Tang Y, Liu F, Xie H. Gene Analysis of Epstein-Barr Virus-Associated Lymphomas in Hu-PBL/SCID Chimeras. TUMORI JOURNAL 2018; 96:465-72. [DOI: 10.1177/030089161009600315] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Aims and background The mechanisms of Epstein-Barr virus (EBV)-associated tumor development are incompletely understood. The aim of this study was to investigate the gene expression of EBV-associated lymphomas in hu-PBL/SCID mice. Methods Human peripheral blood lymphocytes (hu-PBL) from EBV-seropositive donors were transplanted into severe combined immunodeficiency (SCID) mice. In situ hybridization was used to detect EBV-encoded small RNA-1 (EBER1) in tumor tissues. Mutation of TP53 exons 5–8 in EBV-induced lymphomas was analyzed by PCR-SSCP. Immunohistochemical staining was used to examine EBV gene products and cellular oncoproteins. Results Twenty-one of 29 mice developed tumors. EBER1 was positive in the nuclei of almost all tumor cells. Immunohistochemistry showed positive staining of LMP1, EBNA2 and ZEBRA in a small number of tumor cells. Immunohistochemically detectable p53 protein expression was common (85.7%), but TP53 gene mutations were identified in only four cases (19.1%) of EBV-associated lymphomas. Positivity rates of C-myc, Bcl-2 and Bax expression were 100%, 95.2%, and 90.5%, respectively, in the 21 cases of EBV-associated lymphomas. Conclusions Our preliminary findings suggest that EBV-associated lymphomas in hu-PBL/SCID chimeras show EBV infection, expression of oncogenic viral genes, and overexpression of cellular oncogenes. TP53 gene mutations are rare but p53 protein is commonly expressed in EBV-associated lymphomas.
Collapse
Affiliation(s)
- Runliang Gan
- Cancer Research Institute, University of South China, Hengyang City, Hunan 421001, China
| | - Xiaoli Xie
- Cancer Research Institute, University of South China, Hengyang City, Hunan 421001, China
| | - Jie He
- Cancer Research Institute, University of South China, Hengyang City, Hunan 421001, China
| | - Xiaomin Liu
- Cancer Research Institute, University of South China, Hengyang City, Hunan 421001, China
| | - Li Hong
- Cancer Research Institute, University of South China, Hengyang City, Hunan 421001, China
| | - Yunlian Tang
- Cancer Research Institute, University of South China, Hengyang City, Hunan 421001, China
| | - Fang Liu
- Cancer Research Institute, University of South China, Hengyang City, Hunan 421001, China
| | - Hailong Xie
- Cancer Research Institute, University of South China, Hengyang City, Hunan 421001, China
| |
Collapse
|
6
|
Bollard CM, Cruz CR, Barrett AJ. Directed T-cell therapies for leukemia and lymphoma after hematopoietic stem cell transplant: beyond chimeric antigen receptors. Int J Hematol Oncol 2015. [DOI: 10.2217/ijh.15.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This review focuses on the recent advances utilizing adoptive T-cell immunotherapies for patients after hematopoietic stem cell transplant using T cells after autologous transplant to treat the highest risk patients. The particular emphasis is the use of T cells to treat leukemias and lymphomas with gene transfer and nongene transfer approaches to direct specificity to tumor associated antigens. In this review, we will highlight how these novel therapeutics can be successfully used to prevent or treat high-risk patients who relapse after hematopoietic stem cell transplant.
Collapse
Affiliation(s)
- Catherine M Bollard
- Children’s National Health System & The George Washington University, Washington, DC, USA
| | - C Russell Cruz
- Children’s National Health System & The George Washington University, Washington, DC, USA
| | - A John Barrett
- National Heart Lung & Blood Institute, National Institutes for Health, Bethesda, MD, USA
| |
Collapse
|
7
|
Bollard CM, Barrett AJ. Cytotoxic T lymphocytes for leukemia and lymphoma. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2014; 2014:565-569. [PMID: 25696912 DOI: 10.1182/asheducation-2014.1.565] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This chapter focuses on the recent advances in adoptive T-cell immunotherapies, not only for patients after hematopoietic stem cell transplantation, but also in the autologous setting using T cells early in the disease process for the treatment of the highest-risk patients with leukemias and lymphomas. The particular emphasis is to highlight the role of T-cell therapies for hematologic malignancies using a non-gene-transfer approach to direct specificity, including the clinical use of T-cell therapies for EBV-associated lymphomas and strategies for targeting nonviral lymphoma- and leukemia-associated antigens.
Collapse
Affiliation(s)
- Catherine M Bollard
- Children's National Health System and The George Washington University, Washington, DC; and
| | - A John Barrett
- National Heart, Lung, and Blood Institute, National Institutes for Health, Bethesda, MD
| |
Collapse
|
8
|
Immunotherapy: opportunities, risks and future perspectives. Cytotherapy 2014; 16:S120-9. [PMID: 24629797 DOI: 10.1016/j.jcyt.2014.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/31/2014] [Accepted: 02/04/2014] [Indexed: 01/06/2023]
Abstract
This review is intended to reflect upon the current status and perspectives of cell-based immunotherapy at a time when the promise of extensive pre-clinical research has been translated into encouraging clinical responses. However, some of these have also been complicated by significant adverse reactions. As the field moves towards definitive late stage trials, with a growing interest from pharmaceutical companies, we realize that novel cell therapy strategies pose questions that are familiar to traditional drug development, along with new considerations due to the potential of T cells to persist long term and to expand after adoptive transfer. These questions address the safety of the product, the efficacy, the mode of action, and the anticipation of risks. From different perspectives, we intend to address exciting opportunities and safety concerns in current concepts of cellular immunotherapy.
Collapse
|
9
|
Ameratunga R, Woon ST, Neas K, Love DR. The clinical utility of molecular diagnostic testing for primary immune deficiency disorders: a case based review. Allergy Asthma Clin Immunol 2010; 6:12. [PMID: 20529312 PMCID: PMC2903612 DOI: 10.1186/1710-1492-6-12] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 06/08/2010] [Indexed: 12/18/2022] Open
Abstract
Primary immune deficiency disorders (PIDs) are a group of diseases associated with a genetic predisposition to recurrent infections, malignancy, autoimmunity and allergy. The molecular basis of many of these disorders has been identified in the last two decades. Most are inherited as single gene defects. Identifying the underlying genetic defect plays a critical role in patient management including diagnosis, family studies, prognostic information, prenatal diagnosis and is useful in defining new diseases. In this review we outline the clinical utility of molecular testing for these disorders using clinical cases referred to Auckland Hospital. It is written from the perspective of a laboratory offering a wide range of tests for a small developed country.
Collapse
Affiliation(s)
- Rohan Ameratunga
- Department of Clinical Immunology Auckland City Hospital, Park Rd, Grafton, Auckland New Zealand.
| | | | | | | |
Collapse
|
10
|
Chang CM, Yu KJ, Mbulaiteye SM, Hildesheim A, Bhatia K. The extent of genetic diversity of Epstein-Barr virus and its geographic and disease patterns: a need for reappraisal. Virus Res 2009; 143:209-21. [PMID: 19596032 PMCID: PMC2731007 DOI: 10.1016/j.virusres.2009.07.005] [Citation(s) in RCA: 165] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 06/30/2009] [Accepted: 07/02/2009] [Indexed: 12/12/2022]
Abstract
Epstein-Barr virus (EBV) is a ubiquitous, gamma-1 lymphotrophic virus etiologically linked to nasopharyngeal carcinoma (NPC), endemic to Southern China, and Burkitt lymphoma (BL), endemic to equatorial Africa, both of which are rare elsewhere in the world. Why EBV is associated with different malignancies in different geographic regions remains puzzling and may be related to EBV genotypic variability through specific disease and geographic associations. We review the literature on sequence variation in EBV genes, focusing on LMP-1, EBNA-1, and BZLF-1 and their distribution by geography and disease. Given the limitations of current studies, definitive conclusions regarding the link between EBV genotypes, disease and geography are not possible. We suggest that the true extent of EBV diversity is likely to be greater than is currently recognized. Additional studies conducted in carefully selected populations, that are sufficiently powered to provide robust estimates, and that utilize testing approaches that permit full characterization of viral diversity are needed to further our understanding of patterns of EBV genetic variation and their association with malignancies in different regions.
Collapse
Affiliation(s)
- Cindy M Chang
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, 6120 Executive Blvd., EPS 7074, Rockville, MD 20892, USA.
| | | | | | | | | |
Collapse
|
11
|
Salama ME, Parham DM, Perkins SL, Bahler DW, Ellison DA. Fatal non-transplant-related epstein-barr virus-associated atypical lymphoid proliferations in infants and children: a clinicopathologic study. Pediatr Dev Pathol 2008; 11:443-9. [PMID: 18473605 DOI: 10.2350/07-12-0386.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2007] [Accepted: 04/27/2008] [Indexed: 11/20/2022]
Abstract
Most Epstein-Barr virus (EBV)-related infections in infants and children are asymptomatic or self-limited mild viral illnesses, but rare cases of a rapidly fatal disorder have been described. Failure of the cellular response to control EBV-related lymphoid proliferation leads to severe disease with multiple complications, including a fatal outcome or development of an EBV-driven, clonal lymphoid neoplasm. In this report we characterize 3 cases of fatal, nontransplant, or immunodeficiency-related EBV infection in very young children with immunophenotypic and molecular evidence of B/natural killer (NK)-T cell clonal expansion. An immunohistochemical staining panel included testing for B-cell antigen (CD20), and T/NK cell antigens including CD2, CD3, CD4, CD8, CD56, CD57, and TIA-1. T-cell and B-cell PCR clonality testing was performed on paraffin tissue specimens to identify clonal populations. The ages of these 3 patients ranged from 22 months to 4 years. Initial clinical presentations included pneumonia, abnormal liver function tests and fever, and lymphadenopathy. The 3 patients died within 17 to 72 days of presentation, and autopsy was performed on 1 patient. All cases demonstrated prominent atypical lymphoid or lymphohistiocytic infiltrates, and necrosis was present in 2 of the 3 cases. The atypical lymphocytes were positive for CD3 (cytoplasmic), CD2, CD8, TIA-1, and CD57 and negative for CD4. We molecularly identified B-cell clones in the 2 tested patients, who also showed evidence of hemophagocytosis. Fatal EBV infection is characterized by a morphologic spectrum with atypical lymphoid infiltrates and variable necrosis. Our molecular studies of these patients suggest a clonally-derived expansive process, most likely driven by EBV infection. Our results also suggest that development of clonality is associated with an aggressive clinical course and may be useful in predicting greater risk for fatal outcome. A high index of suspicion, coupled with appropriate serologic and molecular testing, aids in early recognition and diagnosis of these lymphoproliferative processes.
Collapse
Affiliation(s)
- Mohamed E Salama
- Department of Pathology, University of Utah and ARUP Lab, 500 Chipeta Way, Salt Lake City, Utah 84108, USA.
| | | | | | | | | |
Collapse
|
12
|
Carbone A, Gloghini A, Dotti G. EBV-associated lymphoproliferative disorders: classification and treatment. Oncologist 2008; 13:577-85. [PMID: 18515742 DOI: 10.1634/theoncologist.2008-0036] [Citation(s) in RCA: 176] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Since its discovery as the first human tumor virus, Epstein-Barr virus (EBV) has been implicated in the development of a wide range of B-cell lymphoproliferative disorders, including Burkitt's lymphoma, classic Hodgkin's lymphoma, and lymphomas arising in immunocompromised individuals (post-transplant and HIV-associated lymphoproliferative disorders). T-cell lymphoproliferative disorders that have been reported to be EBV associated include a subset of peripheral T-cell lymphomas, angioimmunoblastic T-cell lymphoma, extranodal nasal type natural killer/T-cell lymphoma, and other rare histotypes. EBV encodes a series of products interacting with or exhibiting homology to a wide variety of antiapoptotic molecules, cytokines, and signal transducers, hence promoting EBV infection, immortalization, and transformation. However, the exact mechanism by which EBV promotes oncogenesis is an area of active debate. The focus of this review is on the pathology, diagnosis, classification, and pathogenesis of EBV-associated lymphomas. Recent advances in EBV cell-based immunotherapy, which is beginning to show promise in the treatment of EBV-related disorders, are discussed.
Collapse
MESH Headings
- Adult
- Disease Susceptibility
- Epstein-Barr Virus Infections/classification
- Epstein-Barr Virus Infections/immunology
- Epstein-Barr Virus Infections/virology
- Herpesvirus 4, Human/classification
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/immunology
- Humans
- Immunocompromised Host/immunology
- Immunotherapy
- Lymphoma, B-Cell/classification
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Lymphoma, B-Cell/virology
- Lymphoma, T-Cell/classification
- Lymphoma, T-Cell/immunology
- Lymphoma, T-Cell/pathology
- Lymphoma, T-Cell/virology
Collapse
Affiliation(s)
- Antonino Carbone
- Department of Pathology, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, Milano I-20133, Italy.
| | | | | |
Collapse
|
13
|
Zakrzewski JL, Suh D, Markley JC, Smith OM, King C, Goldberg GL, Jenq R, Holland AM, Grubin J, Cabrera-Perez J, Brentjens RJ, Lu SX, Rizzuto G, Sant'Angelo DB, Riviere I, Sadelain M, Heller G, Zúñiga-Pflücker JC, Lu C, van den Brink MRM. Tumor immunotherapy across MHC barriers using allogeneic T-cell precursors. Nat Biotechnol 2008; 26:453-61. [PMID: 18376399 PMCID: PMC2731996 DOI: 10.1038/nbt1395] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2008] [Accepted: 03/03/2008] [Indexed: 01/02/2023]
Abstract
We present a strategy for adoptive immunotherapy using T-lineage committed lymphoid precursor cells generated by Notch1-based culture. We found that allogeneic T-cell precursors can be transferred to irradiated individuals irrespective of major histocompatibility complex (MHC) disparities and give rise to host-MHC restricted and host-tolerant functional allogeneic T cells, improving survival in irradiated recipients as well as enhancing anti-tumor responses. T-cell precursors transduced to express a chimeric receptor targeting hCD19 resulted in significant additional anti-tumor activity, demonstrating the feasibility of genetic engineering of these cells. We conclude that ex vivo generated MHC-disparate T-cell precursors from any donor can be used universally for 'off-the-shelf' immunotherapy, and can be further enhanced by genetic engineering for targeted immunotherapy.
Collapse
Affiliation(s)
- Johannes L Zakrzewski
- Department Immunology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Meerbach A, Wutzler P, Häfer R, Zintl F, Gruhn B. Monitoring of Epstein-Barr virus load after hematopoietic stem cell transplantation for early intervention in post-transplant lymphoproliferative disease. J Med Virol 2008; 80:441-54. [PMID: 18205222 DOI: 10.1002/jmv.21096] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Epstein-Barr virus (EBV)-associated post-transplant lymphoproliferative disease is a life-threatening complication following hematopoietic stem cell transplantation. A quantitative polymerase chain reaction to evaluate EBV-genome copy numbers based on a nested polymerase chain reaction and an end-point dilution was used. Applying this assay EBV load was prospectively screened weekly in 123 patients after transplantation. The results demonstrate that EBV reactivations with more than 1,000 EBV-genome copies measured in 10(5) peripheral blood mononuclear cells were observed in 31 patients (25.2%). Three patients developed lymphoproliferative disease with extremely high EBV-genome copies in peripheral blood mononuclear cells (>100,000 copies/10(5) cells) and plasma. After combined antiviral and immune therapy two of three patients showed a dramatic decrease of EBV load and survived, while the third patient died of lymphoma. A subclinical EBV reactivation was observed in 24 cases (19.5%) with EBV-genome copies in 10(5) peripheral blood mononuclear cells ranging between 2,500 and mostly 10,000. After reduction of immunosuppression the EBV levels normalized. In four patients, the high copy number of > or =80,000 copies/10(5) peripheral blood mononuclear cells and plasma positivity prompted us to start pre-emptive therapy with rituximab and cidofovir for prevention of lymphoproliferative disease. After drug administration the high EBV load was reduced remarkably. Ninety-two patients (74.8%) who had < or =1,000 copies/10(5) peripheral blood mononuclear cells did not develop EBV-associated lymphoproliferative disease. In conclusion, monitoring of EBV load is a sensitive and useful parameter in the surveillance of EBV reactivation for early intervention in EBV-associated lymphoproliferative disease as well as for follow-up of the efficacy of therapy.
Collapse
Affiliation(s)
- Astrid Meerbach
- Institute of Virology and Antiviral Therapy, Medical Center, Friedrich-Schiller University Jena, Jena, Germany.
| | | | | | | | | |
Collapse
|
15
|
Suwiwat S, Pradutkanchana J, Ishida T, Mitarnun W. Quantitative analysis of cell-free Epstein-Barr virus DNA in the plasma of patients with peripheral T-cell and NK-cell lymphomas and peripheral T-cell proliferative diseases. J Clin Virol 2007; 40:277-83. [PMID: 17996493 DOI: 10.1016/j.jcv.2007.08.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 08/21/2007] [Accepted: 08/21/2007] [Indexed: 11/18/2022]
Abstract
BACKGROUND The level of circulating EBV DNA is a prognostic marker in patients with some EBV-associated malignant diseases. OBJECTIVES To investigate the presence and nature of Epstein-Barr virus (EBV) DNA in the plasma and to evaluate the correlation of plasma concentrations of EBV DNA with the EBV genomic status in peripheral blood T-cells and neoplastic cells and with the clinical outcome of patients with peripheral T-cell and NK-cell lymphomas (PTCL) and peripheral T-cell proliferative diseases (PTPD). STUDY DESIGN EBV DNA in the plasma of 45 patients and 45 controls was measured using real-time PCR. The presence of the EBV genome in the isolated peripheral blood lymphocytes (CD3+ and CD3- cells) was analysed by PCR. Detection of EBV-encoded early RNA (EBER) in corresponding tumor tissues was carried out using in situ hybridization. DNase I digestion was applied to plasma samples to detect naked EBV DNA. RESULTS Cell-free EBV DNA was detected in 32/38 (84%) of PTCL patients and 5/7 (71%) of PTPD patients, but not in the controls. Patients with EBV genome in peripheral blood CD3+ cells and EBV genome (EBER) in the tumor cells, compared to those without these findings, had significantly higher plasma EBV DNA levels. The majority of circulating EBV DNA molecules was naked form. The plasma EBV DNA levels were not related to survival. CONCLUSIONS The concentration of EBV DNA in the plasma was not a prognostic marker in PTCL and PTPD patients.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- CD3 Complex/immunology
- Case-Control Studies
- Child
- Child, Preschool
- DNA, Viral/blood
- Female
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/isolation & purification
- Humans
- In Situ Hybridization/methods
- Killer Cells, Natural/pathology
- Lymphocyte Activation
- Lymphoma, Extranodal NK-T-Cell/blood
- Lymphoma, Extranodal NK-T-Cell/immunology
- Lymphoma, Extranodal NK-T-Cell/pathology
- Lymphoma, Extranodal NK-T-Cell/virology
- Lymphoma, T-Cell, Peripheral/blood
- Lymphoma, T-Cell, Peripheral/immunology
- Lymphoma, T-Cell, Peripheral/pathology
- Lymphoma, T-Cell, Peripheral/virology
- Male
- Middle Aged
- Polymerase Chain Reaction/methods
- T-Lymphocytes/pathology
Collapse
Affiliation(s)
- Supaporn Suwiwat
- Department of Pathology, Faculty of Medicine, Prince of Songkla University, Hat-Yai, Songkhla 90110, Thailand
| | | | | | | |
Collapse
|
16
|
Maecker B, Jack T, Zimmermann M, Abdul-Khaliq H, Burdelski M, Fuchs A, Hoyer P, Koepf S, Kraemer U, Laube GF, Müller-Wiefel DE, Netz H, Pohl M, Toenshoff B, Wagner HJ, Wallot M, Welte K, Melter M, Offner G, Klein C. CNS or bone marrow involvement as risk factors for poor survival in post-transplantation lymphoproliferative disorders in children after solid organ transplantation. J Clin Oncol 2007; 25:4902-8. [PMID: 17971586 DOI: 10.1200/jco.2006.10.2392] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To identify prognostic factors of survival in pediatric post-transplantation lymphoproliferative disorder (PTLD) after solid organ transplantation. PATIENTS AND METHODS A multicenter, retrospective case analysis of 55 pediatric solid organ graft recipients (kidney, liver, heart/lung) developing PTLD were reported to the German Pediatric-PTLD registry. Patient charts were analyzed for tumor characteristics (histology, immunophenotypes, cytogenetics, Epstein-Barr virus [EBV] detection), stage, treatment, and outcome. Probability of overall and event-free survival was analyzed in defined subgroups using univariate and Cox regression analyses. RESULTS PTLD was diagnosed at a median time of 29 months after organ transplantation, with a significantly shorter lag time in liver (0.83 years) versus heart or renal graft recipients (3.33 and 3.10 years, respectively; P = .001). The 5-year overall and event-free survival was 68% and 59%, respectively, with 59% of patients surviving 10 years. Stage IV disease with bone marrow and/or CNS involvement was associated independently with poor survival (P = .0005). No differences in outcome were observed between early- and late-onset PTLD, monomorphic or polymorphic PTLD, and EBV-positive or EBV-negative PTLD, respectively. Patients with Burkitt or Burkitt-like PTLD and c-myc translocations had short survival (< 1 year). CONCLUSION Stage IV disease is an independent risk factor for poor survival in pediatric PTLD patients. Prospective multicenter trials are needed to delineate additional risk factors and to assess treatment approaches for pediatric PTLD.
Collapse
Affiliation(s)
- Britta Maecker
- Department of Pediatric Hematology/Oncology, Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Maecker B, Klein C. Lymphoproliferative Erkrankungen nach solider Organtransplantation. Monatsschr Kinderheilkd 2007. [DOI: 10.1007/s00112-007-1629-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
18
|
Kennedy-Nasser AA, Bollard CM. T cell therapies following hematopoietic stem cell transplantation: surely there must be a better way than DLI? Bone Marrow Transplant 2007; 40:93-104. [PMID: 17502898 DOI: 10.1038/sj.bmt.1705667] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Advances in the past few years have significantly improved adoptive immunotherapy strategies available following autologous and allogeneic hematopoietic stem cell transplantation (HSCT). Minimal residual disease, relapsed disease and viral infections remain a significant cause of mortality in patients undergoing HSCT. Novel therapies are critically needed to overcome these management dilemmas, while sparing the graft-versus-tumor effect and avoiding graft-versus-host disease. This review focuses on the T-cell strategies currently available to allay disease while minimizing toxicities in patients who have undergone HSCT.
Collapse
Affiliation(s)
- A A Kennedy-Nasser
- Center for Cell and Gene Therapy, Baylor College of Medicine, 6621 Fannin Street, Houston, TX 77030, USA
| | | |
Collapse
|
19
|
Auwaerter PG. Recent advances in the understanding of infectious mononucleosis: are prospects improved for treatment or control? Expert Rev Anti Infect Ther 2006; 4:1039-1049. [PMID: 17181419 DOI: 10.1586/14787210.4.6.1039] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Symptomatic primary Epstein-Barr virus infection is known more commonly as infectious mononucleosis, an illness known for afflicting adolescents and younger adults as a febrile illness accompanied by pharyngitis and lymphadenopathy. Historically believed to be generally benign, infectious mononucleosis has been linked more recently to increased risks of developing Hodgkin's lymphoma and multiple sclerosis. Advances in the understanding of host immune responses to Epstein-Barr virus have begun to elucidate the reasons why younger children typically experience subclinical infection whereas older individuals develop infectious mononucleosis. This review will highlight recent advances in the understanding of primary Epstein-Barr virus infection, and whether prospective treatments or vaccine strategies may affect native infection and its sequelae.
Collapse
Affiliation(s)
- Paul G Auwaerter
- The Johns Hopkins University School of Medicine, Division of Infectious Diseases, 1830 East Monument Street, #449, Baltimore, MD 21205, USA.
| |
Collapse
|
20
|
Tattevin P, Le Tulzo Y, Minjolle S, Person A, Chapplain JM, Arvieux C, Thomas R, Michelet C. Increasing incidence of severe Epstein-Barr virus-related infectious mononucleosis: surveillance study. J Clin Microbiol 2006; 44:1873-4. [PMID: 16672427 PMCID: PMC1479173 DOI: 10.1128/jcm.44.5.1873-1874.2006] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Older patients are more susceptible to severe Epstein-Barr virus (EBV)-related infectious mononucleosis (IM). This condition may increase in industrialized countries where primary EBV infection occurs later in life. Between 1990 and 2004, 38 patients were admitted to our department with EBV-related IM. Two patients died. The annual incidence increased significantly (r = 0.623; P = 0.013).
Collapse
Affiliation(s)
- Pierre Tattevin
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, Rennes, France.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Capello D, Rossi D, Gaidano G. Post-transplant lymphoproliferative disorders: molecular basis of disease histogenesis and pathogenesis. Hematol Oncol 2006; 23:61-7. [PMID: 16216037 DOI: 10.1002/hon.751] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Post-transplant lymphoproliferative disorders (PTLD) represent a serious complication of solid organ and allogeneic bone marrow transplantation. PTLD generally display B-cell lineage derivation, involvement of extranodal sites, aggressive histology and clinical behaviour, and frequent association with EBV infection. The occurrence of IgV mutations in the overwhelming majority of PTLD documents that malignant transformation targets germinal centre (GC) B-cells and their descendants both in EBV-positive and EBV-negative cases. Analysis of phenotypic markers of B-cell histogenesis, namely BCL6, MUM-1 and CD138, allows further distinction of PTLD histogenetic categories. PTLD expressing the BCL6(+)/MUM1(+/-)/CD138(-) profile reflect B-cells actively experiencing the GC reaction and comprise diffuse large B-cell lymphoma (DLBCL) centroblastic and Burkitt lymphoma. PTLD expressing the BCL6(-)/MUM1(+)/CD138(-) phenotype putatively derive from B-cells that have concluded the GC reaction and comprise the majority of polymorphic PTLD and a fraction of DLBCL. A third group of PTLD is reminiscent of post-GC and pre- terminally differentiated B-cells that show the BCL6(-)/MUM1(+)/CD138(+) phenotype and are morphologically represented by either polymorphic PTLD or DLBCL immunoblastic. The molecular pathogenesis of PTLD involves infection by oncogenic viruses, namely Epstein-Barr virus, as well as genetic or epigenetic alterations of several cellular genes. At variance with lymphoma arising in immunocompetent hosts, whose genome is relatively stable, a fraction of PTLD are characterized by microsatellite instability as a consequence of defects in the DNA mismatch repair mechanism. Apart from microsatellite instability, molecular alterations of cellular genes recognized in PTLD include alterations of c-MYC, BCL-6, p53, DNA hypermethylation, and aberrant somatic hypermutation of proto-oncogenes.
Collapse
Affiliation(s)
- Daniela Capello
- Division of Hematology, Department of Medical Sciences and IRCAD, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | | | | |
Collapse
|
22
|
Kremer M, Quintanilla-Martínez L, Nährig J, von Schilling C, Fend F. Immunohistochemistry in bone marrow pathology: a useful adjunct for morphologic diagnosis. Virchows Arch 2005; 447:920-37. [PMID: 16231177 DOI: 10.1007/s00428-005-0070-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2005] [Accepted: 08/23/2005] [Indexed: 12/11/2022]
Abstract
Pathomorphological examination of trephine biopsies of the bone marrow (BM) represents a standard method for the diagnosis and staging of hematologic neoplasms and other disorders involving the BM. The increasing knowledge about the genetic basis and biology of hematologic neoplasms, as well as the recently proposed WHO classification system, provide the framework for an accurate diagnosis. Although conventional morphology remains the gold standard for paraffin-embedded BM trephines, immunohistochemical stainings have become an integral part of the diagnostic workup. Antibodies suitable for paraffin sections are generally applicable to BM trephines, but modifications of staining protocols may be necessary due to the alternative fixatives and decalcification procedures used for BM biopsies. The indications for immunostainings range from confirmation and classification of lymphoma involvement, subclassification of acute leukemias, and estimating blast counts in myelodysplastic and myeloproliferative syndromes to characterization of BM involvement in nonhematologic neoplasms. Although subtyping of NHL in the BM is more difficult from the point of morphology, classification of the entities that frequently involve the BM, especially the small B-cell lymphomas, can easily be achieved with the help of immunohistochemistry. In this review, we try to summarize the current state of the art in BM immunohistochemistry for the diagnosis of hematologic disorders. Moreover, diagnostic algorithms and useful antibody panels are proposed for a rational and cost-effective approach.
Collapse
Affiliation(s)
- Marcus Kremer
- Institute of Pathology, Technical University Munich, Ismaningerstrasse 22, 81675 Munich, Germany
| | | | | | | | | |
Collapse
|
23
|
Ferrà C, Castellví J. [Cervical adenopathy in a patient with acute leukemia and stem cell transplantation]. Med Clin (Barc) 2005; 125:270-7. [PMID: 16137489 DOI: 10.1157/13078102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
MESH Headings
- Acute Disease
- Adult
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Diagnosis, Differential
- Epstein-Barr Virus Infections/diagnosis
- Hepatitis, Viral, Human/diagnosis
- Hepatitis, Viral, Human/etiology
- Hepatitis, Viral, Human/pathology
- Hepatitis, Viral, Human/virology
- Herpes Simplex/diagnosis
- Herpesvirus 1, Human/isolation & purification
- Herpesvirus 3, Human/isolation & purification
- Herpesvirus 4, Human/isolation & purification
- Humans
- Leukemia, Myeloid/therapy
- Lymphatic Metastasis/diagnosis
- Lymphoproliferative Disorders/diagnosis
- Lymphoproliferative Disorders/etiology
- Lymphoproliferative Disorders/pathology
- Lymphoproliferative Disorders/virology
- Male
- Neck
- Necrosis
- Remission Induction
- Stem Cell Transplantation/adverse effects
Collapse
Affiliation(s)
- Christelle Ferrà
- Servicio de Hematología Clínica, Hospital Germans Trias i Pujol, Institut Català d'Oncologia, Badalona, Barcelona, Spain
| | | |
Collapse
|
24
|
Yang CR, Hsieh SL, Ho FM, Lin WW. Decoy receptor 3 increases monocyte adhesion to endothelial cells via NF-kappa B-dependent up-regulation of intercellular adhesion molecule-1, VCAM-1, and IL-8 expression. THE JOURNAL OF IMMUNOLOGY 2005; 174:1647-56. [PMID: 15661928 DOI: 10.4049/jimmunol.174.3.1647] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Decoy receptor 3 (DcR3), a soluble receptor for FasL, LIGHT and TL1A, is highly expressed in cancer cells. We show that pretreatment of HUVECs with DcR3 enhances the adhesion of THP-1 and U937 cells and primary monocytes. A similar stimulatory effect of DcR3 on THP-1 adhesion was also observed in human microvascular endothelial cells (HMVECs). Flow cytometry and ELISA showed that DcR3-treated HUVECs exhibited significant increases in ICAM-1 and VCAM-1 expression. We also demonstrate the ability of DcR3 to stimulate the secretion of IL-8 by HUVECs. RT-PCR and reporter assays revealed that the expression of adhesion molecules and IL-8 are regulated at the level of gene transcription. Experiments with pyrrolidine dithiocarbamate indicated the involvement of an NF-kappaB signaling pathway. DcR3 was found to induce IkappaB kinase activation, IkappaB degradation, p65 nuclear translocation, and NF-kappaB DNA-binding activity. The enhancement by DcR3 of cell adhesion to HUVECs was not mimicked by the TL1A-Ab, which has been shown in our previous work to be a neutralizing Ab against TL1A, thereby inducing HUVECs angiogenesis. Moreover, DcR3-induced cell adhesion could be detected in human aortic endothelial cells (ECs) in which TL1A expression is lacking. Together, our data demonstrate that DcR3 increases monocyte adhesion to ECs via NF-kappaB activation, leading to the transcriptional up-regulation of adhesion molecules and IL-8 in ECs. This novel action appears not to be due to TL1A neutralization, but occurs through an as yet undefined target(s). This study implicates DcR3 in the relationship between inflammation and cancer development.
Collapse
Affiliation(s)
- Chia-Ron Yang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | |
Collapse
|
25
|
Loughrey M, Trivett M, Lade S, Murray W, Turner H, Waring P. Diagnostic application of Epstein-Barr virus-encoded RNA in situ hybridisation. Pathology 2005; 36:301-8. [PMID: 15370127 DOI: 10.1080/0031302042000224584] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIMS Epstein-Barr virus (EBV) has been implicated in the pathogenesis of nasopharyngeal carcinoma and a range of proliferative lymphoid conditions. In situ hybridisation (ISH) looking for virus-encoded RNA (EBER) transcripts is performed simply using a commercially available probe. We aimed to examine the application of this test in a routine diagnostic setting. METHODS In total, 26 cases in which EBV ISH was requested for diagnostic purposes were examined. We looked at the indication for testing, the result and its implication for the final diagnosis. RESULTS Cases were classified into three categories: possible nasopharyngeal carcinoma; possible EBV-related lymphoma; and possible immunodeficiency-associated lymphoproliferative disorder. Six of nine cases of possible nasopharyngeal carcinoma were EBV ISH positive (3/3 primary and 3/6 secondary), confirming the diagnosis. Three of 14 possible lymphoma cases were EBV ISH positive which, along with appropriate ancillary tests, assisted in making the diagnoses of Burkitt's lymphoma, lymphomatoid granulomatosis and extranodal NK/T-cell lymphoma of nasal type. All of three immunodeficiency-associated cases were EBV ISH positive. Two of these were post-transplant lymphoproliferative disorders, monomorphic type. The third case was classified as HIV-related polymorphic lymphoproliferative disorder. CONCLUSIONS In our experience, EBV ISH is a straightforward and rapid procedure to perform, giving unequivocal results. Used in the appropriate clinicopathological setting it can be a highly useful ancillary diagnostic aid.
Collapse
Affiliation(s)
- Maurice Loughrey
- Department of Pathology, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Victoria 3002, Australia.
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
Abstract
Epstein-Barr virus (EBV) is associated with several different types of aggressive non-Hodgkin lymphoma (NHL). Individuals with primary or secondary immunodeficiency are susceptible to developing B cell lymphoproliferation due to outgrowth of EBV-infected B cells that express type III latency characterized by expression of all nine latent-cycle EBV antigens. These cells would normally be susceptible to control by EBV-specific T cells, and strategies to restore EBV-specific immune responses may be effective therapeutically. EBV-associated lymphomas occurring in individuals who do not have a known immunodeficiency include NK and T malignancies with cytotoxic phenotypes, sporadic cases of B-NHL and lymphomatoid granulomatosis. These malignancies respond poorly to standard chemoradiotherapy, and immunotherapeutic or pharmacologic strategies targeting EBV are being explored.
Collapse
Affiliation(s)
- Helen E Heslop
- Baylor College of Medicine, Center for Cell and Gene Therapy, Houston, TX 77030-2303, USA.
| |
Collapse
|