Long-term follow-up of IPEX syndrome patients after different therapeutic strategies: An international multicenter retrospective study

Federica Barzaghi, Laura Cristina Amaya Hernandez, Benedicte Neven, Silvia Ricci, Zeynep Yesim Kucuk, Jack J Bleesing, Zohreh Nademi, Mary Anne Slatter, Erlinda Rose Ulloa, Anna Shcherbina, Anna Roppelt, Austen Worth, Juliana Silva, Alessandro Aiuti, Luis Murguia-Favela, Carsten Speckmann, Magda Carneiro-Sampaio, Juliana Folloni Fernandes, Safa Baris, Ahmet Ozen, Elif Karakoc-Aydiner, Ayca Kiykim, Ansgar Schulz, Sandra Steinmann, Lucia Dora Notarangelo, Eleonora Gambineri, Paolo Lionetti, William Thomas Shearer, Lisa R Forbes, Caridad Martinez, Despina Moshous, Stephane Blanche, Alain Fisher, Frank M Ruemmele, Come Tissandier, Marie Ouachee-Chardin, Frédéric Rieux-Laucat, Marina Cavazzana, Waseem Qasim, Barbarella Lucarelli, Michael H Albert, Ichiro Kobayashi, Laura Alonso, Cristina Diaz De Heredia, Hirokazu Kanegane, Anita Lawitschka, Jong Jin Seo, Marta Gonzalez-Vicent, Miguel Angel Diaz, Rakesh Kumar Goyal, Martin G Sauer, Akif Yesilipek, Minsoo Kim, Yesim Yilmaz-Demirdag, Monica Bhatia, Julie Khlevner, Erick J Richmond Padilla, Silvana Martino, Davide Montin, Olaf Neth, Agueda Molinos-Quintana, Justo Valverde-Fernandez, Arnon Broides, Vered Pinsk, Antje Ballauf, Filomeen Haerynck, Victoria Bordon, Catharina Dhooge, Maria Laura Garcia-Lloret, Robbert G Bredius, Krzysztof Kałwak, Elie Haddad, Markus Gerhard Seidel, Gregor Duckers, Sung-Yun Pai, Christopher C Dvorak, Stephan Ehl, Franco Locatelli, Frederick Goldman, Andrew Richard Gennery, Mort J Cowan, Maria-Grazia Roncarolo, Rosa Bacchetta, Primary Immune Deficiency Treatment Consortium (PIDTC) and the Inborn Errors Working Party (IEWP) of the European Society for Blood and Marrow Transplantation (EBMT), Federica Barzaghi, Laura Cristina Amaya Hernandez, Benedicte Neven, Silvia Ricci, Zeynep Yesim Kucuk, Jack J Bleesing, Zohreh Nademi, Mary Anne Slatter, Erlinda Rose Ulloa, Anna Shcherbina, Anna Roppelt, Austen Worth, Juliana Silva, Alessandro Aiuti, Luis Murguia-Favela, Carsten Speckmann, Magda Carneiro-Sampaio, Juliana Folloni Fernandes, Safa Baris, Ahmet Ozen, Elif Karakoc-Aydiner, Ayca Kiykim, Ansgar Schulz, Sandra Steinmann, Lucia Dora Notarangelo, Eleonora Gambineri, Paolo Lionetti, William Thomas Shearer, Lisa R Forbes, Caridad Martinez, Despina Moshous, Stephane Blanche, Alain Fisher, Frank M Ruemmele, Come Tissandier, Marie Ouachee-Chardin, Frédéric Rieux-Laucat, Marina Cavazzana, Waseem Qasim, Barbarella Lucarelli, Michael H Albert, Ichiro Kobayashi, Laura Alonso, Cristina Diaz De Heredia, Hirokazu Kanegane, Anita Lawitschka, Jong Jin Seo, Marta Gonzalez-Vicent, Miguel Angel Diaz, Rakesh Kumar Goyal, Martin G Sauer, Akif Yesilipek, Minsoo Kim, Yesim Yilmaz-Demirdag, Monica Bhatia, Julie Khlevner, Erick J Richmond Padilla, Silvana Martino, Davide Montin, Olaf Neth, Agueda Molinos-Quintana, Justo Valverde-Fernandez, Arnon Broides, Vered Pinsk, Antje Ballauf, Filomeen Haerynck, Victoria Bordon, Catharina Dhooge, Maria Laura Garcia-Lloret, Robbert G Bredius, Krzysztof Kałwak, Elie Haddad, Markus Gerhard Seidel, Gregor Duckers, Sung-Yun Pai, Christopher C Dvorak, Stephan Ehl, Franco Locatelli, Frederick Goldman, Andrew Richard Gennery, Mort J Cowan, Maria-Grazia Roncarolo, Rosa Bacchetta, Primary Immune Deficiency Treatment Consortium (PIDTC) and the Inborn Errors Working Party (IEWP) of the European Society for Blood and Marrow Transplantation (EBMT)

Abstract

Background: Immunodysregulation polyendocrinopathy enteropathy x-linked (IPEX) syndrome is a monogenic autoimmune disease caused by FOXP3 mutations. Because it is a rare disease, the natural history and response to treatments, including allogeneic hematopoietic stem cell transplantation (HSCT) and immunosuppression (IS), have not been thoroughly examined.

Objective: This analysis sought to evaluate disease onset, progression, and long-term outcome of the 2 main treatments in long-term IPEX survivors.

Methods: Clinical histories of 96 patients with a genetically proven IPEX syndrome were collected from 38 institutions worldwide and retrospectively analyzed. To investigate possible factors suitable to predict the outcome, an organ involvement (OI) scoring system was developed.

Results: We confirm neonatal onset with enteropathy, type 1 diabetes, and eczema. In addition, we found less common manifestations in delayed onset patients or during disease evolution. There is no correlation between the site of mutation and the disease course or outcome, and the same genotype can present with variable phenotypes. HSCT patients (n = 58) had a median follow-up of 2.7 years (range, 1 week-15 years). Patients receiving chronic IS (n = 34) had a median follow-up of 4 years (range, 2 months-25 years). The overall survival after HSCT was 73.2% (95% CI, 59.4-83.0) and after IS was 65.1% (95% CI, 62.8-95.8). The pretreatment OI score was the only significant predictor of overall survival after transplant (P = .035) but not under IS.

Conclusions: Patients receiving chronic IS were hampered by disease recurrence or complications, impacting long-term disease-free survival. When performed in patients with a low OI score, HSCT resulted in disease resolution with better quality of life, independent of age, donor source, or conditioning regimen.

Keywords: FOXP3; IPEX; Treg cells; enteropathy; genetic autoimmunity; hematopoietic stem cell transplantation; immunosuppression; neonatal diabetes; primary immune deficiency; rapamycin.

Conflict of interest statement

Disclosure of potential conflict of interest:

The rest of the authors declare that they have no relevant conflicts of interest.

Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

FIG 1
FIG 1
Patients’ demographics and disease-related characteristics. A, Diagnostic delay scatter plot displaying correlation between age at diagnosis and age at onset (n = 96). Spearman’s rank correlation ρ = 0.456, P < .001. B, Histogram distribution of number of patients diagnosed between 2001 and 2015. Overlaid density distribution showed in blue. C, Heat map of number of symptoms at onset grouped by age at onset. Symptoms present in each age group (indicated by rows) were scaled (z-score or standardized score) and then converted to colors from yellow (low = less frequent) to red (high = highly frequent). Data were not available (NA) for 3 patients; 1 patient has the mutation but has not yet experienced the onset; and for 2 patients, the first symptom is unknown. D, Bar graph comparing frequency of symptoms at onset and later during disease evolution. Every bar indicates number of patients presenting each symptom. However, each patient can exhibit >1 symptom at once. E, Uncommon manifestations. Every bar indicates number of patients presenting each symptom. F, Scatter plot of FOXP3 gene mutations grouped by domain, indicating age at onset (circles) and number of symptoms at onset (asterisks), with median (n = 87; for 10 patients, cDNA FOXP3 mutations were not specified). Gene structure: N-terminal proline-rich (PRR) domain (orange), zinc-finger (ZF) domain (green), leucine-zipper (LZ) domain (blue), LZ-FKH loop (yellow), and FKH domain (red). Mutations were grouped as follows: <c.1 to c.570> 5 E1-5 N-terminal domain, <c.591 to c.666> = E5-6 ZF domain, <c.717 to c.780> = E6-7 LZ domain, <c.781 to c.1010> = E7-9 LZ-FKH loop domain, <c.1011 to c.1251> = E9-11 FKH domain. AIHA, Autoimmune hemolytic anemia; AIN, autoimmune neutropenia; AT, autoimmune thyroiditis; FTT, failure to thrive; ITP, idiopathic thrombocytopenic purpura; Neph, nephropathy; LN, lymphadenopathy; PRR, proline-rich region; UTR, untranslated region.
FIG 2
FIG 2
Timeline of natural history and disease evolution. Patients undergoing HSCT (A) and IS (B). Each line represents a patient identified by his FOXP3 mutation in order of localization on the gene. The end on the line represents the last day of follow-up, and different symbols represent age at onset (circles) and age at HSCT (triangles) or the beginning of IS. An X at the end of the line indicates the age of death. The color of the line indicates the disease status after treatment, whether the patient went into remission (blue), was still diabetic (gray) or not cured (red). NA, Not available.
FIG 3
FIG 3
Immunosuppressive therapy. A, Bar graph indicating numbers of patients exhibiting response, partial benefit, or no response according to a specific immunosuppressive drug administered (n = 34; however, each patient received >1 drug). B, Bar graph indicating each patient’s outcome after treatment with a combination of drugs, as last treatment. Each bar represents the number of patients receiving the treatment, distinguishing those in remission from those with additional or persistent autoimmunity (n = 34). C, Pre- and post-IS patients’ conditions, each bar represents the number of patients presenting each condition. On the right side, the relative percentage is reported (n = 34; however, each patient could present with >1 condition at once). D, Percentage of survival for patients undergoing IS (n = 34) according to score post-IS (P = .0444). AZA, Azathioprine; Ca Inhib, calcineurin inhibitors; CTLA, cytotoxic T lymphocyte–associated antigen; MTX, methotrexate; MMF, mycophenolate mofetil; “others”, any different IS (eg, 6-mercaptopurina, mesalazine); RAPA, rapamycin.
FIG 4
FIG 4
Survival analysis of patients undergoing HSCT. Percentage of survival of patients undergoing HSCT (n = 58) according to conditioning (log-rank test, P = .234) (A), donor type (P = .886) (B), age at HSCT (P = .359) (C), score pre-HSCT (P = .003) (D), score and conditioning (P = .010) (E), and score and age at HSCT (P = .019) (F). A survival probability table accompanies those plots that show significant differences (time points: 6 months, and 1, 3, 5, and 10 years). Full C, Full conditioning regimen; MMRD, mismatched related donor; MMUCB, mismatched unrelated cord blood; MMUD, mismatched unrelated donor; MSD, matched sibling donor; MUD, matched unrelated donor.
FIG 5
FIG 5
Probability of survival and disease status after treatment. A, Survival analysis of IPEX patients undergoing HSCT or IS (n = 92, P = .055). B, Disease-free survival analysis of IPEX patients undergoing IS or HSCT censored for deaths (n = 81, P = .419).

References

    1. Powell BR, Buist NR, Stenzel P. An x-linked syndrome of diarrhea, polyendocrinopathy, and fatal infection in infancy. J Pediatr. 1982;100:731–7.
    1. Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, et al. The immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27:20–1.
    1. Wildin RS, Ramsdell F, Peake J, Peake J, Faravelli F, Casanova JL, Buist N, et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet. 2001;27:18–20.
    1. Barzaghi F, Passerini L, Bacchetta R. Immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome: a paradigm of immunodeficiency with autoimmunity. Front Immunol. 2012;3:211.
    1. Bacchetta R, Barzaghi F, Roncarolo Mg. From IPEX syndrome to FOXP3 mutation: a lesson on immune dysregulation. Ann N Y Acad Sci. 2016 Feb 25; [E-pub ahead of print].
    1. Bacchetta R, Passerini L, Gambineri E, Gambineri E, Dai M, Allan SE, Perroni L, et al. Defective regulatory and effector T cell functions in patients with FOXP3 mutations. J Clin Invest. 2006;116:1713–22.
    1. Kinnunen T, Chamberlain N, Morbach H, Morbach H, Choi J, Kim S, Craft J, et al. Accumulation of peripheral autoreactive B cells in the absence of functional human regulatory T cells. Blood. 2013;121:1595–603.
    1. Passerini L, Olek S, Di Nunzio S, Barzaghi F, Hambleton S, Abinun M, et al. Forkhead box protein 3 (FOXP3) mutations lead to increased th17 cell numbers and regulatory T-cell instability. J Allergy Clin Immunol. 2011;128:1376–9.e1.
    1. Bin Dhuban K, Piccirillo CA. The immunological and genetic basis of immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome. Curr Opin Allergy Clin Immunol. 2015;15:525–32.
    1. Sheikine Y, Woda CB, Lee PY, Chatila TA, Keles S, Charbonnier LM, et al. Renal involvement in the immunodysregulation, polyendocrinopathy, enteropathy, x-linked (IPEX) disorder. Pediatr Nephrol. 2015;30:1197–202.
    1. Zama D, Cocchi I, Masetti R, Specchia F, Alvisi P, Gambineri E, et al. Late-onset of immunodysregulation, polyendocrinopathy, enteropathy, x-linked syndrome (IPEX) with intractable diarrhea. Ital J Pediatr. 2014;40:68.
    1. De Benedetti F, Insalaco A, Diamanti A, Cortis E, Muratori F, Lamioni A, et al. Mechanistic associations of a mild phenotype of immunodysregulation, polyendocrinopathy, enteropathy, x-linked syndrome. Clin Gastroenterol Hepatol. 2006;4:653–9.
    1. Mazzolari E, Forino C, Fontana M, D’Ippolito C, Lanfranchi A, Gambineri E, et al. A new case of IPEX receiving bone marrow transplantation. Bone Marrow Transplant. 2005;35:1033–4.
    1. Seidel Mg, Fritsch G, Lion T, Jurgens B, Heitger A, Bacchetta R, et al. Selective engraftment of donor CD4125high FOXP3-positive T cells in IPEX syndrome after nonmyeloablative hematopoietic stem cell transplantation. Blood. 2009;113:5689–91.
    1. Seidel Mg, Boztug K, Haas Oa. Immune dysregulation syndromes (IPEX, CD27 deficiency, and others): always doomed from the start? J Clin Immunol. 2016;36:6–7.
    1. Veys P. Reduced intensity transplantation for primary immunodeficiency disorders. Pediatr Rep. 2011;3(suppl 2):E11.
    1. Apperley J, Carreras D, Gluckman E, Masszi T, editors. The EBMT handbook on haematopoietic stem cell transplantation. 6th. Leiden, The Netherlands: EBMT; 2012.
    1. Shin CR, Kim MO, Li D, Bleesing JJ, Harris R, Mehta P, et al. Outcomes following hematopoietic cell transplantation for Wiskott-Aldrich syndrome. Bone Marrow Transplant. 2012;47:1428–35.
    1. Bindl L, Torgerson T, Perroni L, Youssef N, Ochs HD, Goulet O, et al. Successful use of the new immune-suppressor sirolimus in IPEX (immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome) J Pediatr. 2005;147:256–9.
    1. Yong PL, Russo P, Sullivan KE. Use of sirolimus in IPEX and IPEX-like children. J Clin Immunol. 2008;28:581–7.
    1. Baud O, Goulet O, Canioni D, Le Deist F, Radford I, Rieu D, et al. Treatment of the immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome (IPEX) by allogeneic bone marrow transplantation. N Engl J Med. 2001;344:1758–62.
    1. Wildin RS, Smyk-Pearson S, Filipovich AH. Clinical and molecular features of the immunodysregulation, polyendocrinopathy, enteropathy, x linked (IPEX) syndrome. J Med Genet. 2002;39:537–45.
    1. Taddio A, Faleschini E, Valencic E, Granzotto M, Tommasini A, Lepore L, et al. Medium-term survival without haematopoietic stem cell transplantation in a case of IPEX: insights into nutritional and immunosuppressive therapy. Eur J Pediatr. 2007;166:1195–7.
    1. Gambineri E, Perroni L, Passerini L, Bianchi L, Doglioni C, Meschi F, et al. Clinical and molecular profile of a new series of patients with immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome: inconsistent correlation between forkhead box protein 3 expression and disease severity. J Allergy Clin Immunol. 2008;122:1105–12.e1.
    1. Kucuk ZY, Bleesing JJ, Marsh R, Zhang K, Davies S, Filipovich AH. A challenging undertaking: stem cell transplantation for immune dysregulation, polyendocrinopathy, enteropathy, x-linked (IPEX) syndrome. J Allergy Clinical Immunol. 2016;137:953–5.e4.
    1. Nademi Z, Slatter M, Gambineri E, Mannurita SC, Barge D, Hodges S, et al. Single centre experience of haematopoietic SCT for patients with immunodysregulation, polyendocrinopathy, enteropathy, x-linked syndrome. Bone Marrow Transplant. 2014;49:310–2.
    1. Lucas Kg, Ungar D, Comito M, Bayerl M, Groh B. Submyeloablative cord blood transplantation corrects clinical defects seen in IPEX syndrome. Bone Marrow Transplant. 2007;39:55–6.
    1. Rao A, Kamani N, Filipovich A, Lee SM, Davies SM, Dalal J, et al. Successful bone marrow transplantation for IPEX syndrome after reduced-intensity conditioning. Blood. 2007;109:383–5.
    1. Burroughs LM, Torgerson TR, Storb R, Carpenter PA, Rawlings DJ, Sanders J, et al. Stable hematopoietic cell engraftment after low-intensity nonmyeloablative conditioning in patients with immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome. J Allergy Clin Immunol. 2010;126:1000–5.
    1. Kasow KA, Morales-Tirado VM, Wichlan D, Shurtleff SA, Abraham A, Persons DA, et al. Therapeutic in vivo selection of thymic-derived natural t regulatory cells following non-myeloablative hematopoietic stem cell transplant for IPEX. Clin Immunol. 2011;141:169–76.
    1. Slatter MA, Boztug H, Potschger U, Sykora KW, Lankester A, Yaniv I, et al. Treosulfan-based conditioning regimens for allogeneic haematopoietic stem cell transplantation in children with non-malignant diseases. Bone Marrow Transplant. 2015;50:1536–41.
    1. d’Hennezel E, Bin Dhuban K, Torgerson T, Piccirillo CA. The immunogenetics of immune dysregulation, polyendocrinopathy, enteropathy, x linked (IPEX) syndrome. J Med Genet. 2012;49:291–302.
    1. Barzaghi F, Passerini L, Gambineri E, Ciullini Mannurita S, Cornu T, Kang ES, et al. Demethylation analysis of the FOXP3 locus shows quantitative defects of regulatory T cells in IPEX-like syndrome. J Autoimmun. 2012;38:49–58.
    1. Iizuka-Koga M, Nakatsukasa H, Ito M, Akanuma T, Lu Q, Yoshimura A. Induction and maintenance of regulatory T cells by transcription factors and epigenetic modifications. J Autoimmun. 2017;83:113–21.

Source: PubMed

3
Abonnieren