Skip to main content

MINI REVIEW article

Front. Pediatr., 30 October 2019
Sec. Pediatric Immunology
This article is part of the Research Topic HSCT for Primary Immunodeficiencies and Rare Metabolic Diseases View all 16 articles

Hematopoietic Stem Cell Transplantation for Primary Immunodeficiencies

\nAndrew R. Gennery,
Andrew R. Gennery1,2*Michael H. AlbertMichael H. Albert3Mary A. Slatter,Mary A. Slatter1,2Arjan LankesterArjan Lankester4 on behalf of Inborn Errors Working Party (IEWP) of the European Society for Blood and Marrow Transplantation (EBMT)
  • 1Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
  • 2Pediatric Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
  • 3Pediatric SCT Program, Dr. von Hauner University Children's Hospital, Ludwig-Maximilians Universität, Munich, Germany
  • 4Department of Pediatrics, Stem Cell Transplantation Program, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands

The field of primary immunodeficiencies has pioneered many of the advances in haematopoietic stem cell transplantation and cellular therapies over the last 50 years. The first patients to demonstrate sustained benefit and prolonged cure from the primary genetic defect following allogeneic haematopoietic stem cell transplantation were patients with primary immunodeficiencies. Although primary immunodeficiency patients began the modern era of haematopoietic stem cell transplantation, the history is nevertheless short-in answer to the question “what is the long term outcome of patients transplanted for primary immunodeficiencies?” we often have to say that we do not know. We believe that most patients who undergo haematopoietic stem cell transplantation for primary immunodeficiencies should live a normal lifespan with a fully corrected immune system. We are now beginning to understanding long term outcomes, the relationship to the underlying genetic defect, age, and pre-morbid condition of the patient at time of transplantation, stem cell source and donor, and effect of pre-transplant cytoreductive chemotherapy conditioning. The long term consequences of post-transplant complications such as graft vs. host disease, veno-occlusive disease, or immune dysregulation are also being recognized. Additionally, some genetic defects have a systemic distribution, and we are learning the natural history of these defects once the immunodeficiency has been removed.

The field of primary immunodeficiencies has pioneered the way in many of the advances in hematopoietic stem cell transplantation and cellular therapies over the last 50 years. In 1968, three patients with primary immunodeficiencies—one with Wiskott Aldrich syndrome and two with X-linked severe combined immunodeficiencies—were the first patients to demonstrate sustained benefit and prolonged cure from the primary genetic defect following allogeneic hematopoietic stem cell transplantation (13). The story of our specialty, whilst at the inception of hematopoietic stem cell transplantation, is thus short—in answer to the question “what is the long term outcome of patients transplanted for primary immunodeficiencies?,” we often have to say that we do not really know. We believe, in many cases, that patients who undergo hematopoietic stem cell transplantation for primary immunodeficiencies will live a normal lifespan with a fully corrected immune system. However, it is only now that we are beginning to dissect long term outcomes and the relationship to the underlying genetic defect, age and pre-morbid condition of the patient at time of transplantation, stem cell source and donor, and effect of pre-transplant cytoreductive chemotherapy conditioning (48). The long term consequences of post-transplant complications such as graft vs. host disease, veno-occlusive disease or immune dysregulation are also being recognized. Additionally, some genetic defects have a systemic distribution, and we are learning the natural history of these defects once the immunodeficiency has been removed.

We are hindered by dealing with small numbers of patients, rare diseases, changing protocols and transplant techniques, as well as suboptimal methods of measuring immune function and repertoire, and incomplete follow up information. Furthermore, the information we gather in our retrospective studies often pertains to historic rather than current practice (9). Importantly, we are also, for many diseases, beginning to understand the natural history of the disease without intervention with transplantation, so that, for some of the more common diseases, we are able to compare data between transplanted and non-transplanted cohorts (1014).

Nevertheless, we are entering an era where we are beginning to understand the implications and consequences of previous treatment practice. Data that we are now gathering are important to aid our understanding of the impact of transplantation on patient survival, immune function, long term organ dysfunction/toxicity including fertility, and quality of life (refer to chapter on Long Term Outcome and Immune Function After Hematopoietic Stem Cell Transplantation for Primary Immunodeficiency). It is now clear that early transplant before the onset of significant infection or organ dysfunction results in better outcomes for all immunodeficiencies (14, 15). In recent years, this knowledge and has heralded the introduction of newborn screening to identify patients with severe combined immunodeficiency before symptom onset (16) (refer to chapter on Newborn Screening for SCID). Furthermore, data are emerging to suggest that best early and longer term outcomes of immune function require some degree of myeloid engraftment which reflects hematopoietic stem cell progenitor engraftment, and by implication, some form of pre-transplant conditioning (4, 6, 17, 18).

Our understanding of the underlying genetic defects has led us to realize that the degree of donor chimerism required for optimal outcome differs depending on the primary disease—a small percentage of donor myeloid chimerism in patients with RAG-deficient severe combined immunodeficiency is sufficient to restore complete T- and B-lymphocyte repertoire and function, whereas incomplete donor chimerism in patients with Wiskott-Aldrich syndrome is associated with an increased risk of autoimmunity (7). Patients with gain-of-function diseases such as STAT-1 or APDS appear to be more likely to experience recurrence of disease manifestations when complete donor chimerism is not achieved (19, 20) (refer to chapter on Long Term Outcome and Immune Function After Hematopoietic Stem Cell Transplantation for Primary Immunodeficiency). Detailed information from larger cohorts of these patients, and those treated with small molecules or specific pathway inhibitors will help us to appropriately select patients for transplantation in the future (21).

An understanding of the toxicities resulting from our treatment approaches has driven the search for safer approaches to therapy, and resulted in less toxic chemotherapy conditioning regimens (22, 23), and is leading to approaches in antibody-based conditioning regimens (24, 25) (refer to chapter on Conditioning Perspectives for Primary Immunodeficiency Stem cell Transplants) and safer approaches to curative therapy including genetic correction of autologous cells by gene addition (26) or gene editing (refer to chapter Autologous stem cell-based gene therapy for inherited disorders: state-of-the-art and future prospects). Our understanding of disease phenotype is being complicated as the genetic revolution, powered by new generation sequencing techniques and analysis of big data sets, reveals patients with new, less severe phenotypes who harbor mutations in genes previously understood to confer a severe phenotype. At the same time as transplantation becomes safer, it is also becoming clear that not everyone with a specific primary immunodeficiency will require transplantation (27). Nowhere is this dilemma sharper than in the expanding field of adolescent and young adult transplantation for primary immunodeficiency (28, 29) (refer to chapter on HSCT in Adolescents and Young Adults with Primary Immunodeficiencies). It should be acknowledged that most of our data pertain to treatment of infants or children, and cannot be automatically extrapolated to older patients. Recognizing who in this group of patients requires transplantation, or can be adequately managed with newly emerging immune-specific therapies is probably one of the most difficult current challenges. The need for collaboration between specialists, particularly through organizations like the Inborn Errors Working Party of the European Society for Blood and Marrow Transplantation (IEWP-EBMT), the Primary Immune Deficiency Treatment Consortium (PIDTC) of North America, the European Society for Immunodeficiencies (ESID), and Stem CEll Transplant for primary Immune Deficiencies in Europe (SCETIDE) and the harvesting of good quality data into specialist registries has never been greater.

As we celebrate 50 years of transplantation for primary immunodeficiencies, we can acknowledge the tremendous progress that we have achieved and look to a bright future for our patients fuelled by enthusiastic international professional collaborations between clinical specialists, basic scientists, patient organizations and industry. These collaborations should now focus on specific questions, identifying current knowledge (30), and formulating practical research questions for the future (31). Given the rarity of our patients, and relatively small cohorts of patients, it is only by close collaborative efforts, ideally between relevant societies such as those listed above, and careful use of combined registries data, with carefully directed questions that we will begin to gather the answers. Retrospective data harvesting has formed the foundation of our knowledge base to date, piecing together knowledge from different studies, often measuring diverse parameters. Perhaps now, building on the knowledge we have now accumulated, is the time to design prospective clinical trials for our patients, in order to accurately answer outstanding questions.

Author Contributions

AG, MA, MS, and AL wrote and edited the manuscript.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Bach FH, Albertini RJ, Joo P, Anderson JL, Bortin MM. Bone-marrow transplantation in a patient with the Wiskott-Aldrich syndrome. Lancet. (1968) 2:1364–6. doi: 10.1016/S0140-6736(68)92672-X

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Gatti RA, Meuwissen HJ, Allen HD, Hong R, Good RA. Immunological reconstitution of sex- linked lymphopenic immunological deficiency. Lancet. (1968) 2:1366–9. doi: 10.1016/S0140-6736(68)92673-1

PubMed Abstract | CrossRef Full Text | Google Scholar

3. De Koning J, Van Bekkum DW, Dicke KA, Dooren LJ, Rádl J, Van Rood JJ. Transplantation of bone-marrow cells and fetal thymus in an infant with lymphopenic immunological deficiency. Lancet. (1969) 1:1223–7. doi: 10.1016/S0140-6736(69)92112-6

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Abd Hamid IJ, Slatter MA, McKendrick F, Pearce MS, Gennery AR. Long-term outcome IL2RG/JAK3 SCID post-hematopoietic stem cell transplantation: a cohort report. Blood. (2017) 129:2198–201. doi: 10.1182/blood-2016-11-748616

CrossRef Full Text | Google Scholar

5. Abd Hamid IJ, Slatter MA, McKendrick F, Pearce MS, Gennery AR. Long term health outcome and quality of life post-HSCT for IL7Rα-, Artemis-, RAG1- and RAG2-deficient Severe Combined Immunodeficiency: a single centre report. J Clin Immunol. (2018) 38:727–32. doi: 10.1007/s10875-018-0540-9

CrossRef Full Text | Google Scholar

6. Cavazzana-Calvo M, Carlier F, Le Deist F, Morillon E, Taupin P, Gautier D, et al. Long-term T-cell reconstitution after hematopoietic stem-cell transplantation in primary T-cell-immunodeficient patients is associated with myeloid chimerism and possibly the primary disease phenotype. Blood. (2007) 109:4575–81. doi: 10.1182/blood-2006-07-029090

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Moratto D, Giliani S, Bonfim C, Mazzolari E, Fischer A, Ochs HD, et al. Long-term outcome and lineage-specific chimerism in 194 patients with Wiskott-Aldrich syndrome treated by hematopoietic cell transplantation in the period 1980-2009: an international collaborative study. Blood. (2011) 118:1675–84. doi: 10.1182/blood-2010-11-319376

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Schuetz C, Neven B, Dvorak CC, Leroy S, Ege MJ, Pannicke U, et al. SCID patients with ARTEMIS vs RAG deficiencies following HCT: increased risk of late toxicity in ARTEMIS-deficient SCID. Blood. (2014) 123:281–9. doi: 10.1182/blood-2013-01-476432

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Gennery AR, Slatter MA, Grandin L, Taupin P, Cant AJ, Veys P, et al. Transplantation of hematopoietic stem cells and long-term survival for primary immunodeficiencies in Europe: entering a new century, do we do better? J Allergy Clin Immunol. (2010) 126:602–10. doi: 10.1016/j.jaci.2010.06.015

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Cole T, Pearce MS, Cant AJ, Cale CM, Goldblatt D, Gennery AR. Clinical outcome in children with chronic granulomatous disease managed conservatively or with hematopoietic stem cell transplantation. J Allergy Clin Immunol. (2013) 132:1150–5. doi: 10.1016/j.jaci.2013.05.031

CrossRef Full Text | Google Scholar

11. de la Morena MT, Leonard D, Torgerson TR, Cabral-Marques O, Slatter M, Aghamohammadi A, et al. Long-term out- comes of 176 patients with X-linked hyper-IgM syndrome treated with or without hematopoietic cell transplantation. J Allergy Clin Immunol. (2017) 139:1282–92. doi: 10.1016/j.jaci.2016.07.039

CrossRef Full Text | Google Scholar

12. Barzaghi F, Amaya Hernandez LC, Neven B, Ricci S, Kucuk ZY, Bleesing JJ, et al. IPEX syndrome survivors following different therapeutic strategies: an international multicenter retrospective study. J Allergy Clin Immunol. (2018) 141:1036–49.e5. doi: 10.1016/j.jaci.2017.10.041

CrossRef Full Text | Google Scholar

13. Booth C, Gilmour KC, Veys P, Gennery AR, Slatter MA, Chapel H, et al. X-linked Lymphoproliferative Disease due to SAP/SH2D1A deficiency: a multicentre study on the manifestations, management and outcome of the disease. Blood. (2011) 117:53–62. doi: 10.1182/blood-2010-06-284935

CrossRef Full Text | Google Scholar

14. Pai SY, Logan BR, Griffith LM, Buckley RH, Parrott RE, Dvorak CC, et al. Transplantation outcomes for severe combined immunodeficiency, 2000–2009. N Engl J Med. (2014) 371:434–46. doi: 10.1056/NEJMoa1401177

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Ferrua F, Galimberti S, Courteille V, Slatter MA, Booth C, Moshous D, et al. Hematopoietic stem cell transplantation for CD40 ligand deficiency: results from an EBMT/ESID-IEWP-SCETIDE-PIDTC Study. J Allergy Clin Immunol. (2019) 143:2238–53. doi: 10.1016/j.jaci.2018.12.1010

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Kwan A, Abraham RS, Currier R, Brower A, Andruszewski K, Abbott JK, et al. Newborn screening for severe combined immunodeficiency in 11 screening programs in the United States. JAMA. (2014) 312:729–38. doi: 10.1001/jama.2014.9132

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Haddad E, Logan BR, Griffith LM, Buckley RH, Parrott RE, Prockop SE, et al. SCID genotype and 6-month posttransplant CD4 count predict survival and immune recovery: a PIDTC retrospective study. Blood. (2018) 132:1737–49. doi: 10.1182/blood-2018-03-840702

CrossRef Full Text | Google Scholar

18. Borghans JA, Bredius RG, Hazenberg MD, Roelofs H, Jol-van der Zijde EC, Heidt J, et al. Early determinants of long-term T-cell reconstitution after hematopoietic stem cell transplantation for severe combined immunodeficiency. Blood. (2006) 108:763–9. doi: 10.1182/blood-2006-01-009241

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Leiding JW, Okada S, Hagin D, Abinun M, Shcherbina A, Balashov DN, et al. Hematopoietic stem cell transplantation in patients with Gain of Function STAT1 Mutation. J Allergy Clin Immunol. (2018) 141:704–17.e5. doi: 10.1016/j.jaci.2017.03.049

CrossRef Full Text | Google Scholar

20. Nademi Z, Slatter MA, Dvorak CC, Neven B, Fischer A, Suarez F, et al. Haematopoietic stem cell transplant in patients with activated PI3K delta syndrome. J Allergy Clin Immunol. (2017) 139:1046–9. doi: 10.1016/j.jaci.2016.09.040

CrossRef Full Text | Google Scholar

21. Forbes LR, Vogel TP, Cooper MA, Castro-Wagner J, Schussler E, Weinacht KG, et al. Jakinibs for the Treatment of Immunodysregulation in Patients with Gain of Function STAT1 or STAT3 Mutations. J Allergy Clin Immunol. (2018) 142:1665–9. doi: 10.1016/j.jaci.2018.07.020

CrossRef Full Text | Google Scholar

22. Güngör T, Teira P, Slatter M, Stussi G, Stepensky P, Moshous D, et al. Reduced-intensity conditioning and HLA-matched haemopoietic stem-cell transplantation in patients with chronic granulomatous disease: a prospective multicentre study. Lancet. (2014) 838:436–48. doi: 10.1016/S0140-6736(13)62069-3

CrossRef Full Text | Google Scholar

23. Morillo-Gutierrez B, Beier R, Rao K, Burroughs L, Schulz A, Ewins AM, et al. Treosulfan-based conditioning for allogeneic HSCT in children with chronic granulomatous disease: a multicenter experience. Blood. (2016) 128:440–8. doi: 10.1182/blood-2016-03-704015

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Schulz AS, Glatting G, Hoenig M, Schuetz C, Gatz SA, Grewendorf S, et al. Radioimmunotherapy-based conditioning for hematopoietic cell transplantation in children with malignant and nonmalignant diseases. Blood. (2011) 117:4642–50. doi: 10.1182/blood-2010-06-284349

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Straathof KC, Rao K, Eyrich M, Hale G, Bird P, Berrie E, et al. Haemopoietic stem-cell transplantation with antibody-based minimal-intensity conditioning: a phase 1/2 study. Lancet. (2009) 374:912–20. doi: 10.1016/S0140-6736(09)60945-4

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Cicalese MP, Ferrua F, Castagnaro L, Pajno R, Barzaghi F, Giannelli S, et al. Update on the safety and efficacy of retroviral gene therapy for immunodeficiency due to adenosine deaminase deficiency. Blood. (2016) 128:45–54. doi: 10.1182/blood-2016-01-688226

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Speckmann C, Doerken S, Aiuti A, Albert MH, Al-Herz W, Allende LM, et al. A prospective study on the natural history of patients with profound combined immunodeficiency (P-CID): a “first fifty” analysis. J Allergy Clin Immunol. (2017) 139:1302–10.e4. doi: 10.1016/j.jaci.2016.07.040

CrossRef Full Text | Google Scholar

28. Fox TA, Chakraverty R, Burns S, Carpenter B, Thomson K, Lowe D, et al. Successful outcome following allogeneic hematopoietic stem cell transplantation in adults with primary immunodeficiency. Blood. (2018) 131:917–31. doi: 10.1182/blood-2017-09-807487

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Albert MH, Hauck F, Wiebking V, Aydin S, Notheis G, Koletzko S, et al. Allogeneic stem cell transplantation in adolescents and young adults with primary immunodeficiencies. J Allergy Clin Immunol Pract. (2018) 6:298–301.e2. doi: 10.1016/j.jaip.2017.07.045

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Heimall J, Puck J, Buckley R, Fleisher TA, Gennery AR, Neven B, et al. Current knowledge and priorities for future research in late effects after hematopoietic stem cell transplantation (HCT) for severe combined immunodeficiency patients: a consensus statement from the second pediatric blood and marrow transplant consortium international conference on late effects after pediatric HCT. Biol Blood Marrow Transplant. (2017) 23:379–87. doi: 10.1016/j.bbmt.2016.12.619

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Heimall J, Buckley RH, Puck J, Fleisher TA, Gennery AR, Haddad E, et al. Recommendations for screening and management of late effects in patients with severe combined immunodeficiency after allogenic hematopoietic cell transplantation: a consensus statement from the second pediatric blood and marrow transplant consortium international conference on late effects after pediatric HCT. Biol Blood Marrow Transplant. (2017) 23:1229–40. doi: 10.1016/j.bbmt.2017.04.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: primary immunodeficiency, severe combined immnunodeficiency, Wiskott Aldrich syndrome, chronic granulomatous disease, conditioning

Citation: Gennery AR, Albert MH, Slatter MA and Lankester A (2019) Hematopoietic Stem Cell Transplantation for Primary Immunodeficiencies. Front. Pediatr. 7:445. doi: 10.3389/fped.2019.00445

Received: 31 July 2019; Accepted: 14 October 2019;
Published: 30 October 2019.

Edited by:

Rosa Bacchetta, Stanford University, United States

Reviewed by:

Ricardo U. Sorensen, Louisiana State University, United States
Alice Bertaina, Stanford University, United States

Copyright © 2019 Gennery, Albert, Slatter and Lankester. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Andrew R. Gennery, a.r.gennery@ncl.ac.uk

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.