Tailored therapies for primary immunodeficiencies

Main Article Content

Bianca Cinicola
Federica Pulvirenti
Giulia Brindisi
Gian Luigi Marseglia
Riccardo Castagnoli
Thomas Foiadelli
Carlo Caffarelli
Amelia Licari
Michele Miraglia Del Giudice
Anna Maria Zicari
Marzia Duse
Fabio Cardinale

Keywords

Primary immunodeficiencies, autoimmunity, immune dysregulation, target, tailored treatment, immunoglobulin, gene therapy

Abstract

Primary immunodeficiency disorders (PIDs) are rare inherited monogenic disorders of the immune system, characterized by an increased risk of infection, immune dysregulation and malignancies. To date, more than 420 PIDs have been identified. The recent introduction of high throughput sequencing technologies has led to identifying the molecular basis of the underlying aberrant immune pathway, and candidate targets to develop precision treatment, aimed at modifying the clinical course of the disease. In PID, targeted therapies are especially effective to manage immune dysregulation and autoimmunity, also reducing the incidence of side effects compared to conventional treatments, sparing the use of steroids and immunosuppressive drugs. Moreover, in the last years, the approach of conventional treatments such as immunoglobulin replacement therapies has evolved and the indication has expanded to new diseases, leading to individualized strategies to both improve infection control and quality of life.  Similarly, the new advent of gene therapy in selected PIDs has introduced the benefit to correct the immunological defect, reducing at the same time the complications related to the hematopoietic stem cell transplantation. Here, we illustrate the most recent findings on tailored treatments for PIDs.

Abstract 814 | PDF Downloads 417

References

1. Tangye SG, Al-Herz W, Bousfiha A, et al. Human inborn errors of immunity: 2019 update on the classification from the International Union of Immunological Societies Expert Committee. J Clin Immunol 2020;40:24-64.
2. Castagnoli R, Lougaris V, Giardino G, et al. Inborn errors of immunity with atopic phenotypes: a practical guide for allergists. World Allergy Organ J 2021;14:100513.
3. Heimall J. Genetic testing to diagnose primary immunodeficiency disorders and to identify targeted therapy. Immunol Allergy Clin North Am 2019;39:129-40.
4. Kohn LA, Kohn DB. Gene therapies for primary immune deficiencies. Front Immunol 12:648951.
5. Wasserman RL. Personalized therapy: immunoglobulin replacement for antibody deficiency. Immunol Allergy Clin North Am 2019;39:95-111.
6. Chikuma S. CTLA-4, an essential immune-checkpoint for T-cell activation. Curr Top Microbiol Immunol 2017;410:99-126.
7. Mitsuiki N, Schwab C, Grimbacher B. What did we learn from CTLA-4 insufficiency on the human immune system? Immunol Rev 2019;287:33-49.
8. Schubert D, Bode C, Kenefeck R, et al. Autosomal dominant immune dysregulation
syndrome in humans with CTLA4 mutations. Nat Med 2014;20:1410–6.
9. Kuehn HS, Ouyang W, Lo B, et al. Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science 2014;345:1623-7.
10. Schwab C, Gabrysch A, Olbrich P, et al. Phenotype, penetrance, and treatment of 133 cytotoxic T-lymphocyte antigen 4-insufficient subjects. J Allergy Clin Immunol 2018;142:1932-46.
11. Pombo-Suarez M, Gomez-Reino JJ. Abatacept for the treatment of rheumatoid arthritis. Expert Rev Clin Immunol 2019;15:319-26.
12. Lee S, Moon JS, Lee CR, et al. Abatacept alleviates severe autoimmune symptoms in a patient carrying a de novo variant in CTLA-4. J Allergy Clin Immunol 2016;137:327-30.
13. Egg D, Rump IC, Mitsuiki N, et al. Therapeutic options for CTLA-4 insufficiency. J Allergy Clin Immunol 2021;S0091-6749(21)00891-5, Epub ahead of print.
14. van Leeuwen EM, Cuadrado E, Gerrits AM, Witteveen E, de Bree GJ. Treatment of intracerebral lesions with abatacept in a CTLA4-haploinsufficient patient. J Clin Immunol 2018;38:464-7.
15. Navarini AA, Hruz P, Berger CT, et al. Vedolizumab as a successful treatment of CTLA-4-associated autoimmune enterocolitis. J Allergy Clin Immunol 2017;139:1043-6.
16. Slatter MA, Engelhardt KR, Burroughs LM, et al. Hematopoietic stem cell transplantation for CTLA4 deficiency. J Allergy Clin Immunol 2016;138:615-9.
17. Hou TZ, Verma N, Wanders J, et al. Identifying functional defects in patients with immune dysregulation due to LRBA and CTLA-4 mutations. Blood 2017;129:1458-68.
18. Lopez-Herrera G, Tampella G, Pan-Hammarstrom Q, et al. Deleterious mutations in LRBA are associated with a syndrome of immune deficiency and autoimmunity. Am J Hum Genet 2012;90:986-1001.
19. Alkhairy OK, Abolhassani H, Rezaei N, et al. Spectrum of phenotypes associated
with mutations in LRBA. J Clin Immunol 2016;36:33–45.
20. Habibi S, Zaki-Dizaji M, Rafiemanesh H, et al. Clinical, immunologic, and molecular spectrum of patients with LPS-responsive beige-like anchor protein deficiency: a systematic review. J Allergy Clin Immunol Pract 2019;7:2379-86.
21. Gamez-Diaz L, August D, Stepensky P, et al. The extended phenotype of LPS-responsive beige-like anchor protein (LRBA) deficiency. J Allergy Clin Immunol 2016;137:223–30.
22. Charbonnier LM, Janssen E, Chou J, et al. Regulatory T-cell deficiency and immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like disorder caused by loss-of-function mutations in LRBA. J Allergy Clin Immunol 2015;135:217–27.
23. Levy E, Stolzenberg MC, Bruneau J, et al. LRBA deficiency with autoimmunity
and early onset chronic erosive polyarthritis. Clin Immunol 2016;168:88–93.
24. Bratanic N, Kovac J, Pohar K, et al. Multifocal gastric adenocarcinoma in a patient
with LRBA deficiency. Orphanet J Rare Dis 2017;12:131.
25. Azizi G, Mirshafiey A, Abolhassani H, et al. The imbalance of circulating T helper subsets and regulatory T cells in patients with LRBA deficiency: correlation with disease severity. J Cell Physiol 2018;233:8767-77.
26. Revel-Vilk S, Fischer U, Keller B, et al. Autoimmune lymphoproliferative syndrome-like disease in patients with LRBA mutation. Clin Immunol 2015;159:84-92.
27. Alroqi FJ, Charbonnier LM, Baris S, et al. Exaggerated follicular helper T-cell responses in patients with LRBA deficiency caused by failure of CTLA4- mediated regulation. J Allergy Clin Immunol 2018;141:1050-9.
28. Jamee M, Hosseinzadeh S, Sharifinejad N, et al. Comprehensive comparison between 222 CTLA-4 haploinsufficiency and 212 LRBA deficiency patients: a systematic review. Clin Exp Immunol 2021;205:28-43.
29. Lo B, Zhang K, Lu W, et al. Autoimmune disease. Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy. Science 2015;349:436–40.
30. Kiykim A, Ogulur I, Dursun E, et al. Abatacept as a long-term targeted therapy for
LRBA deficiency. J Allergy Clin Immunol Pract 2019;7:2790–800.
31. Tesch VK, Abolhassani H, Shadur B, et al. Inborn errors, clinical, and registry working parties of the European Society for Blood and Marrow Transplantation and the European Society for Immunodeficiencies. Long-term outcome of LRBA deficiency in 76 patients after various treatment modalities as evaluated by the Immune Deficiency and Dysregulation Activity (IDDA) score. J Allergy Clin Immunol 2020;145:1452-63.
32. Cantley LC. The phosphoinositide 3-kinase pathway. Science. 2002;296:1655-7.
33. Angulo I, Vadas O, Garcon F, et al. Phosphoinositide 3-kinase delta gene mutation predisposes to respiratory infection and airway damage. Science 2013;342:866–71.
34. Lucas CL, Kuehn HS, Zhao F, et al. Dominant-activating germline mutations in the gene encoding the PI(3)K catalytic subunit p110delta result in T cell senescence and human immunodeficiency. Nat Immunol 2014;15:88–97.
35. Brodsky NN, Lucas CL. Infections in activated PI3K delta syndrome (APDS). Curr Opin Immunol 2021;72:146–57.
36. Elgizouli M, Lowe DM, Speckmann C, et al. Activating PI3Kδ mutations in a cohort of 669 patients with primary immunodeficiency. Clin Exp Immunol 2016;183:221-9.
37. Elkaim E, Neven B, Bruneau J, et al. Clinical and immunologic phenotype associated with activated phosphoinositide 3-kinase δ syndrome 2: a cohort study. J Allergy Clin Immunol 2016;138:210-8.
38. Coulter TI, Chandra A, Bacon CM, et al. Clinical spectrum and features of activated phosphoinositide 3-kinase δ syndrome: A large patient cohort study. J Allergy Clin Immunol 2017;139:597-606.
39. Durandy A, Kracker S. Increased activation of PI3 kinase-δ predisposes to B-cell lymphoma. Blood 2020;135:638–43.
40. Edwards ESJ, Bier J, Cole TS, et al. Activating PIK3CD mutations impair human cytotoxic lymphocyte differentiation and function and EBV immunity. J Allergy Clin Immunol 2019; 143:276-91.
41. Carpier JM, Lucas CL. Epstein-Barr virus susceptibility in Activated PI3Kδ Syndrome (APDS) immunodeficiency. Front Immunol 2018;8:2005.
42. Jamee M, Moniri S, Zaki-Dizaji M, et al. Clinical, immunological, and genetic features in patients with activated PI3Kδ syndrome (APDS): a systematic review. Clin Rev Allergy Immunol 2020;59:323–33.
43. Coulter TI, Cant AJ. The treatment of activated PI3Kdelta syndrome. Front Immunol 2018;9:2043.
44. Nademi Z, Slatter MA, Dvorak CC, et al. Hematopoietic stem cell transplant in patients with activated PI3K delta syndrome. J Allergy Clin Immunol 2017;139:1046-9.
45. Okano T, Imai K, Tsujita Y, et al. Hematopoietic stem cell transplantation for progressive combined immunodeficiency and lymphoproliferation in patients with activated phosphatidylinositol- 3-OH kinase δ syndrome type 1. J Allergy Clin Immunol 2019; 143: 266–75.
46. Dimitrova D, Nademi Z, Maccari ME, et al. International retrospective study of allogeneic hematopoietic cell transplantation for activated PI3K-delta syndrome. J Allergy Clin Immunol 2021;S0091-6749(21)00810-1. Epub ahead of print.
47. Maccari ME, Abolhassani H, Aghamohammadi A, et al. Disease evolution and response to rapamycin in activated phosphoinositide 3-Kinase δ syndrome: the European Society for Immunodeficiencies-activated phosphoinositide 3-Kinase δ syndrome registry. Front Immunol 2018;9:1–8.
48. Rao VK, Webster S, Dalm V, et al. Effective “activated PI3Kdelta syndrome”-targeted therapy with the PI3Kdelta inhibitor leniolisib. Blood 2017;130:2307-16.
49. Diaz N, Juarez M, Cancrini C, et al. Seletalisib for activated PI3Kδ syndromes: open-label phase 1b and extension studies. J Immunol 2020;205:2979-87.
50. Cahn A, Hamblin JN, Begg M, et al. Safety, pharmacokinetics and dose-response characteristics of GSK2269557, an inhaled PI3Kdelta inhibitor under development for the treatment of COPD. Pulm Pharmacol Ther 2017;46:69–77.
51. Harrison DA. The Jak/STAT pathway. Cold Spring Harb Perspect Biol 2012;4:a011205.
52. Zhang Y, Ma CA, Lawrence MG, et al. PD-L1 up-regulation restrains Th17 cell differentiation in STAT3 loss- and STAT1 gain-of-function patients. J Exp Med 2017;214:2523-33.
53. Zhang W, Chen X, Gao G, et al. Clinical relevance of gain- and loss-of-function germline mutations in STAT1: a systematic review. Front Immunol 2021;12:654406.
54. Sampaio EP, Hsu AP, Pechacek J, et al. Signal transducer and activator of transcription 1 (STAT1) gain-of-function mutations and disseminated coccidioidomycosis and histoplasmosis. J Allergy Clin Immunol 2013;131:1624-34.
55. Mizoguchi Y, Okada S. Inborn errors of STAT1 immunity. Curr Opin Immunol 2021;72:59-64.
56. Toubiana J, Okada S, Hiller J, et al. International STAT1 gain-of-function study group. Heterozygous STAT1 gain-of-function mutations underlie an unexpectedly broad clinical phenotype. Blood 2016;127:3154-64.
57. Uzel G, Sampaio EP, Lawrence MG, et al. Dominant gain-of-function STAT1 mutations in FOXP3 wild-type immune dysregulation polyendocrinopathy enteropathy-X-linked-like syndrome. J Allergy Clin Immunol 2013;131:1611–23.
58. Baris S, Alroqi F, Kiykim A, et al. Severe early-onset combined immunodeficiency due to heterozygous gain-of function mutations in STAT1. J Clin Immunol 2016;36:641-8.
59. Ma CS, Wong N, Rao G, et al. Monogenic mutations differentially affect the quantity and quality of T follicular helper cells in patients with human primary immunodeficiencies. J Allergy Clin Immunol 2015;136:993-1006.
60. Kiykim A, Charbonnier LM, Akcay A, et al. Hematopoietic stem cell transplantation in patients with heterozygous STAT1 gain-of-function mutation. J Clin Immunol 2019;39:37-44.
61. Aldave JC, Cachay E, Núñez L, et al. A 1-year-old girl with a gain-of-function STAT1 mutation treated with hematopoietic stem cell transplantation. J Clin Immunol 2013;33:1273-5.
62. Gadina M, Chisolm DA, Philips RL, McInness IB, Changelian PS, O'Shea JJ. Translating JAKs to jakinibs. J Immunol 2020;204:2011-20.
63. Forbes LR, Vogel TP, Cooper MA, et al. Jakinibs for the treatment of immune dysregulation in patients with gain-of-function signal transducer and activator of transcription 1 (STAT1) or STAT3 mutations. J Allergy Clin Immunol 2018;142:1665-9.
64. Meesilpavikkai K, Dik WA, Schrijver B, et al. Baricitinib treatment in a patient with a gain-of-function mutation in signal transducer and activator of transcription 1 (STAT1). J Allergy Clin Immunol 2018;142:328–30.
65. Vargas-Hernandez A, Mace EM, Zimmerman O, et al. Ruxolitinib partially reverses functional NK cell deficiency in patients with STAT1 gain-of-function mutations. J Allergy Clin Immunol 2018;141:2142-55.
66. Weinacht KG, Charbonnier LM, Alroqi F, et al. Ruxolitinib reverses dysregulated T helper cell responses and controls autoimmunity caused by a novel signal transducer and activator of transcription 1 (STAT1) gain-of-function mutation. J Allergy Clin Immunol 2017; 139:1629–40.
67. Vogel TP, Milner JD, Cooper MA. The ying and yang of STAT3 in human disease. J Clin Immunol 2015;35:615-23.
68. Sharma S, Saikia B, Goel S, et al. TH17 cells in STAT3 related hyper-IgE syndrome. Indian J Pediatr 2016;83:1104-8.
69. Chandrasekaran P, Zimmerman O, Paulson M, et al. Distinct mutations at the same positions of STAT3 cause either loss or gain of function. J Allergy Clin Immunol 2016;138:1222-4.
70. Flanagan SE, Haapaniemi E, Russell MA, et al. Activating germline mutations in STAT3 cause early-onset multi-organ autoimmune disease. Nat Genet 2014;46:812-4.
71. Fabre A, Marchal S, Barlogis V, et al. Clinical aspects of STAT3 gain-of-function germline mutations: a systematic review. J Allergy Clin Immunol Pract 2019;7:1958-69.
72. Milner JD, Vogel TP, Forbes L, et al. Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations. Blood 2015; 125: 591-9.
73. Gutiérrez M. Activating mutations of STAT3: impact on human growth. Mol Cell Endocrinol 2020;518:110979.
74. Haapaniemi EM, Kaustio M, Rajala HL, et al. Autoimmunity, hypogammaglobulinemia, lymphoproliferation, and mycobacterial disease in patients with activating mutations in STAT3. Blood 2015; 125: 639–48.
75. Wang W, Liu L, Hui X, et al. Efficacy of tocilizumab therapy in a patient with severe pancytopenia associated with a STAT3 gain-of-function mutation. BMC Immunol 2021;22:19.
76. Ammann AJ, Ashman RF, Buckley RH, et al. Use of intravenous gamma-globulin in antibody immunodeficiency: results of a multicenter controlled trial. Clin Immunol Immunopathol 1982;22:60-7.
77. European Medicine Agency. Core summary of product characteristics for human normal immunoglobulin for intravenous administration (IVIg). Available at https://www.ema.europa.eu/en/core-summary-product-characteristics-human-normal-immunoglobulin-intravenous-administration-ivig. Accessed on 28 July 2021.
78. Lederman HM, Winkelstein JA. X-linked agammaglobulinemia: an analysis of 96 patients. Medicine (Baltimore) 1985;64:145-56.
79. Lougaris V, Soresina A, Baronio M, et al. Long-term follow-up of 168 patients with X-linked agammaglobulinemia reveals increased morbidity and mortality. J Allergy Clin Immunol 2020;146:429-37.
80. Quinti I, Soresina A, Guerra A, et al. Effectiveness of immunoglobulin replacement therapy on clinical outcome in patients with primary antibody deficiencies: results from a multicenter prospective cohort study. J Clin Immunol 2011;3:315-22.
81. Hurst JR, Workman S, Garcha DS, Seneviratne SL, Haddock JA, Grimbacher B. Activity, severity and impact of respiratory disease in primary antibody deficiency syndromes. J Clin Immunol 2014;34:68-75.
82. Cinetto F, Scarpa R, Pulvirenti F, Quinti I, Agostini C, Milito C. Appropriate lung management in patients with primary antibody deficiencies. Expert Rev Respir Med 2019;13:823-38.
83. Buckley RH. B-cell function in severe combined immunodeficiency after stem cell or gene therapy: a review. J Allergy Clin Immunol 2010;125:790-7.
84. Serana F, Sottini A, Chiarini M, et al. The different extent of B and T cell immune reconstitution after hematopoietic stem cell transplantation and enzyme replacement therapies in SCID patients with adenosine deaminase deficiency. J Immunol 2010;185:7713-22.
85. Perez EE, Orange JS, Bonilla F, et al. Update on the use of immunoglobulin in human disease: A review of evidence. J Allergy Clin Immunol 2017;139(3S):S1-S46.
86. Buckley RH, Schiff RI. The use of intravenous immune globulin in immunodeficiency diseases. N Engl J Med 1991;325:110-7.
87. Gelfand EW, Ochs HD, Shearer WT. Controversies in IgG replacement therapy in patients with antibody deficiency diseases. J Allergy Clin Immunol 2013;131:1001-5.
88. Farrugia A, Visentini M, Quinti I. Editorial: Immunoglobulin therapy in the 21st century - the dark side of the moon. Front Immunol 2015;6:436.
89. Gathmann B, Mahlaoui N, Gérard L, et al. European Society for Immunodeficiencies registry working party. Clinical picture and treatment of 2212 patients with common variable immunodeficiency. J Allergy Clin Immunol 2014;134:116–26.
90. Quartier P, Debre M, De Blic J, et al. Early and prolonged intravenous immunoglobulin replacement therapy in childhood agammaglobulinemia: a retrospective survey of 31 patients. J Pediatr 1999;134:589-96.
91. Bonagura VR, Marchlewski R, Cox A, Rosenthal DW. Biologic IgG level in primary immunodeficiency disease: the IgG level that protects against recurrent infection. J Allergy Clin Immunol 2008;122:210‐2.
92. Lechanska-Helman J, Sobocinska A, Jerzynska J, Stelmach I. The influence of hospital-based intravenous immunoglobulin and home-based self-administrated subcutaneous immunoglobulin therapy in young children with primary immunodeficiency diseases on their parents' / caregivers' satisfaction. Pediatr Int 2020;62:316-8.
93. Anterasian C, Duong R, Gruenemeier P, Ernst C, Kitsen J, Geng B. Quality of life differences for primary immunodeficiency patients on home SCIG versus IVIG. J Clin Immunol 2019;39:814-22.
94. Ameratunga R, Ahn Y, Steele R, Woon ST. The natural history of untreated primary hypogammaglobulinemia in adults: implications for the diagnosis and treatment of Common Variable Immunodeficiency Disorders (CVID). Front Immunol 2019;10:1541.
95. Berger M. Choices in IgG replacement therapy for primary immune deficiency diseases: subcutaneous IgG vs. intravenous IgG and selecting an optimal dose. Curr Opin Allergy Clin Immunol 2011;11:532‐8.
96. Pulvirenti F, Cinetto F, Pecoraro A, et al. Health-related quality of life in patients with CVID under different schedules of immunoglobulin administration: prospective multicenter study. J Clin Immunol 2019;39:159-70.
97. Matucci A, Maggi E, Vultaggio A. Mechanisms of action of Ig preparations: immunomodulatory and anti-inflammatory effects. Front Immunol 2015;5:690.
98. Gennery AR, Albert MH, Slatter MA, Lankester A. Hematopoietic stem cell transplantation for primary immunodeficiencies. Front Pediatr 2019;7:445.
99. Ferrari G, Thrasher AJ, Aiuti A. Gene therapy using haematopoietic stem and progenitor cells. Nat Rev Genet 2021;22:216-34.
100. Dunbar CE, High KA, Joung JK, Kohn DB, Ozawa K, Sadelain M. Gene therapy comes of age. Science 2018;359:eaan4672.
101. Houghton BC, Booth C. Gene therapy for primary immunodeficiency. Hemasphere 2020;5:e509.
102. Kohn DB, Booth C, Shaw KL, et al. Autologous ex vivo lentiviral gene therapy for adenosine deaminase deficiency. N Engl J Med 2021;384:2002-13.
103. European Medicine Agency. Strimvelis. Avalaible at https://www.ema.europa.eu/en/medicines/human/EPAR/strimvelis. Accessed on 31 July 2021.
104. Fox TA, Booth C. Gene therapy for primary immunodeficiencies. Br J Haematol 2021;193:1044-59.