Mycobacteria-Specific T Cells May Be Expanded From Healthy Donors and Are Near Absent in Primary Immunodeficiency Disorders

Mycobacterial Infections can be severe in patients with T-cell deficiency or phagocyte disorders, and treatment is frequently complicated by antimicrobial resistance. Restoration of T-cell immunity via stem cell transplantation facilitates control of mycobacterial infections, but presence of active...

Full description

Bibliographic Details
Main Authors: Shabnum Patel, Haili Lang, Gelina Sani, Alexandra F. Freeman, Jennifer Leiding, Patrick J. Hanley, Conrad Russell Cruz, Melanie Grant, Yunfei Wang, Benjamin Oshrine, Cindy Palmer, Steven M. Holland, Catherine M. Bollard, Michael D. Keller
Format: Article
Language:English
Published: Frontiers Media S.A. 2019-03-01
Series:Frontiers in Immunology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fimmu.2019.00621/full
id doaj-537ffc6a02874c9180d3ffe55bd51802
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Shabnum Patel
Shabnum Patel
Haili Lang
Gelina Sani
Alexandra F. Freeman
Jennifer Leiding
Jennifer Leiding
Jennifer Leiding
Patrick J. Hanley
Patrick J. Hanley
Conrad Russell Cruz
Conrad Russell Cruz
Melanie Grant
Yunfei Wang
Benjamin Oshrine
Cindy Palmer
Steven M. Holland
Catherine M. Bollard
Catherine M. Bollard
Catherine M. Bollard
Michael D. Keller
Michael D. Keller
spellingShingle Shabnum Patel
Shabnum Patel
Haili Lang
Gelina Sani
Alexandra F. Freeman
Jennifer Leiding
Jennifer Leiding
Jennifer Leiding
Patrick J. Hanley
Patrick J. Hanley
Conrad Russell Cruz
Conrad Russell Cruz
Melanie Grant
Yunfei Wang
Benjamin Oshrine
Cindy Palmer
Steven M. Holland
Catherine M. Bollard
Catherine M. Bollard
Catherine M. Bollard
Michael D. Keller
Michael D. Keller
Mycobacteria-Specific T Cells May Be Expanded From Healthy Donors and Are Near Absent in Primary Immunodeficiency Disorders
Frontiers in Immunology
immunotherapy
T cells
mycobacteria
primary immunodeficiency
mendelian suspectibility to mycobacteria
hematopoietic stem cell transplantation
author_facet Shabnum Patel
Shabnum Patel
Haili Lang
Gelina Sani
Alexandra F. Freeman
Jennifer Leiding
Jennifer Leiding
Jennifer Leiding
Patrick J. Hanley
Patrick J. Hanley
Conrad Russell Cruz
Conrad Russell Cruz
Melanie Grant
Yunfei Wang
Benjamin Oshrine
Cindy Palmer
Steven M. Holland
Catherine M. Bollard
Catherine M. Bollard
Catherine M. Bollard
Michael D. Keller
Michael D. Keller
author_sort Shabnum Patel
title Mycobacteria-Specific T Cells May Be Expanded From Healthy Donors and Are Near Absent in Primary Immunodeficiency Disorders
title_short Mycobacteria-Specific T Cells May Be Expanded From Healthy Donors and Are Near Absent in Primary Immunodeficiency Disorders
title_full Mycobacteria-Specific T Cells May Be Expanded From Healthy Donors and Are Near Absent in Primary Immunodeficiency Disorders
title_fullStr Mycobacteria-Specific T Cells May Be Expanded From Healthy Donors and Are Near Absent in Primary Immunodeficiency Disorders
title_full_unstemmed Mycobacteria-Specific T Cells May Be Expanded From Healthy Donors and Are Near Absent in Primary Immunodeficiency Disorders
title_sort mycobacteria-specific t cells may be expanded from healthy donors and are near absent in primary immunodeficiency disorders
publisher Frontiers Media S.A.
series Frontiers in Immunology
issn 1664-3224
publishDate 2019-03-01
description Mycobacterial Infections can be severe in patients with T-cell deficiency or phagocyte disorders, and treatment is frequently complicated by antimicrobial resistance. Restoration of T-cell immunity via stem cell transplantation facilitates control of mycobacterial infections, but presence of active infections during transplantation is associated with a higher risk of mortality. Adoptive T cell immunotherapy has been successful in targeting viruses, but has not been attempted to treat mycobacterial infections. We sought to expand and characterize mycobacterial-specific T-cells derived from healthy donors in order to determine suitability for adoptive immunotherapy. Mycobacteria-specific T-cells (MSTs) were generated from 10 healthy donors using a rapid ex vivo expansion protocol targeting five known mycobacterial target proteins (AG85B, PPE68, ESXA, ESXB, and ADK). MSTs were compared to T-cells expanded from the same donors using lysate from M. tuberculosis or purified protein derivative from M. avium (sensitin). MST expansion from seven patients with primary immunodeficiency disorders (PID) and two patients with IFN-γ autoantibodies and invasive M. avium infections. MSTs expanded from healthy donors recognized a median of 3 of 5 antigens, with production of IFN-γ, TNF, and GM-CSF in CD4+ T cells. Comparison of donors who received BCG vaccine (n = 6) to those who did not (n = 4) showed differential responses to PPE68 (p = 0.028) and ADK (p = 0.015) by IFN-γ ELISpot. MSTs expanded from lysate or sensitin also recognized multiple mycobacterial antigens, with a statistically significant differences noted only in the response to PPE68 (p = 0.016). MSTs expanded from patients with primary immunodeficiency (PID) and invasive mycobacterial infections showed activity against mycobacterial antigens in only two of seven subjects, whereas both patients with IFN-γ autoantibodies recognized mycobacterial antigens. Thus, MSTs can be generated from donors using a rapid expansion protocol regardless of history of BCG immunization. Most tested PID patients had no detectable T-cell immunity to mycobacteria despite history of infection. MSTs may have clinical utility for adoptive immunotherapy in T-cell deficient patients with invasive mycobacterial infections.
topic immunotherapy
T cells
mycobacteria
primary immunodeficiency
mendelian suspectibility to mycobacteria
hematopoietic stem cell transplantation
url https://www.frontiersin.org/article/10.3389/fimmu.2019.00621/full
work_keys_str_mv AT shabnumpatel mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT shabnumpatel mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT haililang mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT gelinasani mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT alexandraffreeman mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT jenniferleiding mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT jenniferleiding mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT jenniferleiding mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT patrickjhanley mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT patrickjhanley mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT conradrussellcruz mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT conradrussellcruz mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT melaniegrant mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT yunfeiwang mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT benjaminoshrine mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT cindypalmer mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT stevenmholland mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT catherinembollard mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT catherinembollard mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT catherinembollard mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT michaeldkeller mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
AT michaeldkeller mycobacteriaspecifictcellsmaybeexpandedfromhealthydonorsandarenearabsentinprimaryimmunodeficiencydisorders
_version_ 1725238819554328576
spelling doaj-537ffc6a02874c9180d3ffe55bd518022020-11-25T00:53:12ZengFrontiers Media S.A.Frontiers in Immunology1664-32242019-03-011010.3389/fimmu.2019.00621442314Mycobacteria-Specific T Cells May Be Expanded From Healthy Donors and Are Near Absent in Primary Immunodeficiency DisordersShabnum Patel0Shabnum Patel1Haili Lang2Gelina Sani3Alexandra F. Freeman4Jennifer Leiding5Jennifer Leiding6Jennifer Leiding7Patrick J. Hanley8Patrick J. Hanley9Conrad Russell Cruz10Conrad Russell Cruz11Melanie Grant12Yunfei Wang13Benjamin Oshrine14Cindy Palmer15Steven M. Holland16Catherine M. Bollard17Catherine M. Bollard18Catherine M. Bollard19Michael D. Keller20Michael D. Keller21Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United StatesGW Cancer Center, George Washington University, Washington, DC, United StatesCenter for Cancer and Immunology Research, Children's National Health System, Washington, DC, United StatesCenter for Cancer and Immunology Research, Children's National Health System, Washington, DC, United StatesLaboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, MD, United StatesDivision of Allergy & Immunology, University of South Florida, St. Petersburg, FL, United StatesDepartment of Pediatrics, University of South Florida, St. Petersburg, FL, United StatesCancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St Petersburg, FL, United StatesCenter for Cancer and Immunology Research, Children's National Health System, Washington, DC, United StatesDivision of Blood and Marrow Transplantation, Children's National Health System, Washington, DC, United StatesCenter for Cancer and Immunology Research, Children's National Health System, Washington, DC, United StatesGW Cancer Center, George Washington University, Washington, DC, United StatesCenter for Cancer and Immunology Research, Children's National Health System, Washington, DC, United StatesClinical and Translational Science Institute, Children's National Health System, Washington, DC, United StatesCancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St Petersburg, FL, United StatesLaboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, MD, United StatesLaboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, MD, United StatesCenter for Cancer and Immunology Research, Children's National Health System, Washington, DC, United StatesGW Cancer Center, George Washington University, Washington, DC, United StatesDivision of Blood and Marrow Transplantation, Children's National Health System, Washington, DC, United StatesCenter for Cancer and Immunology Research, Children's National Health System, Washington, DC, United StatesDivision of Allergy & Immunology, Children's National Health System, Washington, DC, United StatesMycobacterial Infections can be severe in patients with T-cell deficiency or phagocyte disorders, and treatment is frequently complicated by antimicrobial resistance. Restoration of T-cell immunity via stem cell transplantation facilitates control of mycobacterial infections, but presence of active infections during transplantation is associated with a higher risk of mortality. Adoptive T cell immunotherapy has been successful in targeting viruses, but has not been attempted to treat mycobacterial infections. We sought to expand and characterize mycobacterial-specific T-cells derived from healthy donors in order to determine suitability for adoptive immunotherapy. Mycobacteria-specific T-cells (MSTs) were generated from 10 healthy donors using a rapid ex vivo expansion protocol targeting five known mycobacterial target proteins (AG85B, PPE68, ESXA, ESXB, and ADK). MSTs were compared to T-cells expanded from the same donors using lysate from M. tuberculosis or purified protein derivative from M. avium (sensitin). MST expansion from seven patients with primary immunodeficiency disorders (PID) and two patients with IFN-γ autoantibodies and invasive M. avium infections. MSTs expanded from healthy donors recognized a median of 3 of 5 antigens, with production of IFN-γ, TNF, and GM-CSF in CD4+ T cells. Comparison of donors who received BCG vaccine (n = 6) to those who did not (n = 4) showed differential responses to PPE68 (p = 0.028) and ADK (p = 0.015) by IFN-γ ELISpot. MSTs expanded from lysate or sensitin also recognized multiple mycobacterial antigens, with a statistically significant differences noted only in the response to PPE68 (p = 0.016). MSTs expanded from patients with primary immunodeficiency (PID) and invasive mycobacterial infections showed activity against mycobacterial antigens in only two of seven subjects, whereas both patients with IFN-γ autoantibodies recognized mycobacterial antigens. Thus, MSTs can be generated from donors using a rapid expansion protocol regardless of history of BCG immunization. Most tested PID patients had no detectable T-cell immunity to mycobacteria despite history of infection. MSTs may have clinical utility for adoptive immunotherapy in T-cell deficient patients with invasive mycobacterial infections.https://www.frontiersin.org/article/10.3389/fimmu.2019.00621/fullimmunotherapyT cellsmycobacteriaprimary immunodeficiencymendelian suspectibility to mycobacteriahematopoietic stem cell transplantation