PBL Trigger For Groups 13 To 15

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 2

Editorial

Gene therapy for SCID, now up to 3!


ne
Alain Fischer, MD, PhD,a,b,c and Be dicte Neven, MD, PhDa,d Paris, France

Key words: Gene therapy pathways used to fix DNA lesions. Patients with ART-SCID
are known to experience extra hematopoietic abnormalities years
Since 2000, gene therapy has become an option to treat after HSCT, including dental, growth, kidney, and endocrinal ab-
severe combined immunodeficiency (SCID), a rare but lethal normalities8 that are likely enhanced by use of alkylating agents
condition characterized by fully defective T lymphocyte differ- in the conditioning regimen. Cowan et al therefore decided to
entiation caused by pathogenic variants in least 18 genes. The reduce the use of alkylating agents in the conditioning for gene
current treatment by allogeneic hematopoietic stem cell trans- therapy to 25% of the classical dosage. They designed a lentiviral
plantation (HSCT) is effective but carries the risk of graft- vector in which the Artemis (DCLREIC) cDNA is placed under
versus-host disease. This treatment is based on ex vivo gene the control of 1-kb endogenous promoter to reduce the potential
transfer by using retroviral vector (initially gammaretrovirus, risk associated with overexpression. In all, 10 patients have been
now lentivirus) to transduce the patient bone marrow CD34 treated, mostly at a young age thanks to early diagnosis by
cells.1,2 Stable reconstitution of T-cell counts and function newborn screening. The results are encouraging overall. No
with a clear-cut clinical benefit has been achieved in the treat- adverse events linked to the procedure have been observed. Im-
ment of X-linked SCID (gc) deficiency and adenosine deami- mune reconstitution of T cells occurred in 9 of the 10 patients
nase (ADA)-SCID (Fig 1).1,2 Initial genotoxicity observed (and in all 10 following a second attempt), and some patients
with first-generation gammaretroviral vectors leading to onco- (n 5 4) are no longer receiving immunoglobulin replacement
gene transactivation appears to now be prevented by using therapy, with the median follow-up extending to 31 months. It
self-inactivating vectors in which an internal promoter is instead will be necessary to compare these results with those of HSCT,
used to drive transgene expression.1,2 Use of lentiviral vectors notably in the long term. There is an obvious clinical benefit pro-
as compared with gammaretrovirus-based vectors and some vided by this gene therapy approach. Transduced T and B lym-
conditioning regimen3,4 has led to safe, more efficient transduc- phocytes carry a mean vector copy number (;3-5) higher than
tion and more efficient engraftment of hematopoietic cells, giv- that of transduced CD341 cells (ie, 2.5). This suggests that there
ing rise to differentiation of all subsets of transduced has been a selection of such cells, implying a relatively low effi-
lymphocytes (T, B, and natural killer) and long-lasting immune cacy of the endogenous promoter used, nevertheless enabling
reconstitution. In 2016, these advances led to the authorization adequate regulation of Artemis protein expression. A slight
of marketing of gene therapy for ADA-SCID.2 Thus, today, warning sign comes from the occurrence of autoimmune hemo-
gene therapy for X-linked SCID and ADA-SCID appears to lytic anemia in 4 of the patients, an event that has occasionally
be a safe and efficient therapeutic option. Nevertheless, it is been observed after gene therapy or after HSCT in patients
fair to say that meanwhile, advances in HSCT have occurred, with SCID and is usually related to slow immune reconstitution.
leading to significant improvements in success (ie, in survival The good news is that in all 4 cases the autoimmune hemolytic
and quality of immune reconstitution).5,6 anemia was transient. Nevertheless, it raises the question of the
Cowan et al have now reported on the success of gene therapy balance between reducing conditioning to lower the risk of devel-
for a third SCID condition, namely, Artemis deficiency.7 This opment of ART-SCID–related long-term complications and
was an awaited step because Artemis SCID (ART-SCID) raises increasing the fraction of engrafted transduced stem cells. In
specific issues. The Artemis protein is part of the nonhomologous the future, besides potential improvement in transduction proto-
end-joining repair machinery required for the V-D-J recombina- col, strategies aimed at selectively killing or displacing nontrans-
tion process of antigen receptor genes in T and B lymphocytes.7 duced stem cells in vivo are of potential interest to optimize
Nonhomologous end-joining is also one of the important repair engraftment of transduced progenitors and avoid toxicity. It is
exciting to see a third SCID disease for which 2 therapeutic op-
tions can now be considered (Fig 1). An alternative, as far as the
T-cell deficiency is concerned, might be in vitro generation and
From athe Pediatric Hematology-Immunology and Rheumatology Department, H^opital gene correction of pro-T cells. It is likely that the same technol-
Necker-Enfants Malades, AP-HP Centre Universite de Paris; bthe Institut Imagine, IN- ogy will soon deliver new advances in gene therapy for SCID, at
SERM UMR 1163, Paris; cthe College de France, Paris; and dthe Universite Paris-Cite,
Imagine Institute, Laboratory of Immunogenetics of Pediatric 2, Autoimmunity,
least for RAG-1 SCID, which is a second most frequent form of
INSERM UMR 1163, Paris. SCID.1,2 In addition, the prospect of genome editing9 based on
Disclosure of potential conflict of interest: The authors declare that they have no relevant the CRISPR-Cas9 technology, or likely even better, on base or
conflicts of interest. prime genome editing10 of mutated SCID genes could offer addi-
Received for publication January 24, 2023; revised February 9, 2023; accepted for
tional advances in the treatment of SCID. There is even the pros-
publication February 20, 2023.
Available online February 23, 2023. pect that the later technologies might be used without ex vivo cell
Corresponding author: Alain Fischer, MD, PhD, Institut Imagine, 24 Boulevard Montpar- handling. Looking back on the 55 years that have now passed
nasse, 75015 Paris. E-mail: alain.fischer@aphp.fr. since the first success with HSCT in SCID, several bottleneck
J Allergy Clin Immunol 2023;151:1255-6. steps have been overcome, making it possible to offer HSCT to
0091-6749/$36.00
Ó 2023 American Academy of Allergy, Asthma & Immunology
virtually all patients and to now consider gene therapy for at least
https://doi.org/10.1016/j.jaci.2023.02.015 3 SCID conditions.

1255
1256 FISCHER AND NEVEN J ALLERGY CLIN IMMUNOL
MAY 2023

γc deficiency
IL-7/IL-15 driven expansion
2000 : γRV-GT
2009 : SIN γRV-GT
2019 : SIN LV-GT

NK NK
precursor

lymphoid
progenitors
T T
precursor

B B
precursor

HSC
ADA Artemis
deficiency deficiency
deoxynucleotides supply VDJ recombination
2002 : γRV-GT 2022 : SIN LV-GT
myeloid 2021 : SIN LV-GT
progenitors

FIG 1. Lymphopoiesis and SCID diseases corrected by gene therapy. The mechanism impaired in given
SCID diseases is shown (gray shaded boxes). gRV, Gammaretrovirus; GT, gene therapy; HSC, hematopoi-
etic stem cell; LV, lentivirus; SIN, self-inactivating vector.

REFERENCES 6. Lankester AC, Neven B, Mahlaoui N, von Asmuth EGJ, Courteille V, Alligon M,
1. Fischer A. Gene therapy for inborn errors of immunity: past, present and future. et al. Hematopoietic cell transplantation in severe combined immunodeficiency:
Nat Rev Immunol 2023;23:51-62. The SCETIDE 2006-2014 European cohort. J Allergy Clin Immunol 2022;149:
2. Tucci F, Scaramuzza S, Aiuti A, Mortellaro A. Update on clinical ex vivo hematopoietic 1744-54.e8.
stem cell gene therapy for inherited monogenic diseases. Mol Ther 2021;29:489-504. 7. Cowan MJ, Yu J, Facchino J, Fraser-Browne C, Sanford U, Kawahara M, et al.
3. Mamcarz E, Zhou S, Lockey T, Abdelsamed H, Cross SJ, Kang G, et al. Lentiviral Lentiviral gene therapy for artemis-deficient SCID. N Engl J Med 2022;387:
gene therapy combined with low-dose busulfan in infants with SCID-X1. N Engl J 2344-55.
Med 2019;380:1525-34. 8. Schuetz C, Neven B, Dvorak CC, Leroy S, Ege MJ, Pannicke U, et al. SCID pa-
4. Kohn DB, Booth C, Shaw KL, Xu-Bayford J, Garabedian E, Trevisan V, et al. tients with ARTEMIS vs RAG deficiencies following HCT: increased risk of late
Autologous ex vivo lentiviral gene therapy for adenosine deaminase deficiency. toxicity in ARTEMIS-deficient SCID. Blood 2014;123:281-9.
N Engl J Med 2021;384:2002-13. 9. Porteus MH. A New class of medicines through DNA editing. N Engl J Med 2019;
5. Cuvelier GDE, Logan BR, Prockop SE, Buckley RH, Kuo CY, Griffith LM, et al. 380:947-59.
Outcomes following treatment for ADA-deficient severe combined immunodefi- 10. Newby GA, Liu DR. In vivo somatic cell base editing and prime editing. Mol Ther
ciency: a report from the PIDTC. Blood 2022;140:685-705. 2021;29:3107-24.

You might also like