You are on page 1of 10

The n e w e ng l a n d j o u r na l of m e dic i n e

Brief Report

Acute Myeloid Leukemia Case after Gene


Therapy for Sickle Cell Disease
Sunita Goyal, M.D., John Tisdale, M.D., Manfred Schmidt, Ph.D.,
Julie Kanter, M.D., Jennifer Jaroscak, M.D., Dustin Whitney, Ph.D.,
Hans Bitter, Ph.D., Philip D. Gregory, Ph.D., Geoffrey Parsons, Ph.D.,
Marianna Foos, M.S., Ashish Yeri, Ph.D., Maple Gioia, A.L.M.,
Sarah B. Voytek, Ph.D., Alex Miller, B.S., Jessie Lynch, M.S.,
Richard A. Colvin, M.D., Ph.D., and Melissa Bonner, Ph.D.​​

Sum m a r y

From Bluebird Bio, Cambridge, MA (S.G., Gene therapy with LentiGlobin for sickle cell disease (bb1111, lovotibeglogene
D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., autotemcel) consists of autologous transplantation of a patient’s hematopoietic stem
S.B.V., A.M., J.L., R.A.C., M.B.); the Cel-
lular and Molecular Therapeutics Branch, cells transduced with the BB305 lentiviral vector that encodes the βA-T87Q-globin
National Heart, Lung, and Blood Insti- gene. Acute myeloid leukemia developed in a woman approximately 5.5 years after
tute–National Institute of Diabetes and she had received LentiGlobin for sickle cell disease as part of the initial cohort
Digestive and Kidney Diseases, National
Institutes of Health, Bethesda, MD (J.T.); (Group A) of the HGB-206 study. An analysis of peripheral-blood samples revealed
GeneWerk, Heidelberg, Germany (M.S.); that blast cells contained a BB305 lentiviral vector insertion site. The results of an
the University of Alabama at Birming- investigation of causality indicated that the leukemia was unlikely to be related to
ham, Birmingham (J.K.); and the Division
of Pediatric Hematology–Oncology, Med- vector insertion, given the location of the insertion site, the very low transgene
ical University of South Carolina, Charles- expression in blast cells, and the lack of an effect on expression of surrounding
ton (J.J.). Dr. Bonner can be contacted at genes. Several somatic mutations predisposing to acute myeloid leukemia were
­mbonner@​­bluebirdbio​.­com or at Bluebird
Bio, 60 Binney St., Cambridge, MA 02142. present after diagnosis, which suggests that patients with sickle cell disease are at
increased risk for hematologic malignant conditions after transplantation, most
This article was published on December
12, 2021, at NEJM.org. likely because of a combination of risks associated with underlying sickle cell disease,
transplantation procedure, and inadequate disease control after treatment. (Funded
N Engl J Med 2022;386:138-47.
DOI: 10.1056/NEJMoa2109167 by Bluebird Bio.)
Copyright © 2021 Massachusetts Medical Society.

S
ickle cell disease is a progressive, genetic disease caused by a
point mutation in the β-globin gene and is associated with substantial mor-
bidity and early mortality.1,2 Sickle hemoglobin production and subsequent
polymerization leads to red-cell sickling, clinically manifesting as chronic hemo-
lytic anemia, vasculopathy, and vaso-occlusion.1,2 Current management of sickle
cell disease relies on lifelong use of palliative short-term and long-term thera-
pies.1,2 Allogeneic hematopoietic stem-cell transplantation is the only accepted,
potentially curative treatment option for patients with sickle cell disease; however,
its use is limited by a paucity of matched donors, immunologic complications, and
the inherent risks of transplantation-related morbidity and mortality.1,3,4
Gene therapy with LentiGlobin for sickle cell disease (bb1111, lovotibeglogene
autotemcel) consists of autologous transplantation of the patients’ own hemato-
poietic stem cells (HSCs) transduced with the BB305 lentiviral vector encoding the
βA-T87Q-globin gene, which is designed to produce antisickling hemoglobin (HbAT87Q).5-7
Through the use of autologous HSCs, gene therapy with LentiGlobin circumvents
the need for a donor and may avoid the immunologic complications associated with
allogeneic HSC transplantation. The phase 1–2 HGB-206 study is evaluating the ef-
ficacy and safety of LentiGlobin for sickle cell disease, and an unprespecified in-

138 n engl j med 386;2 nejm.org January 13, 2022

The New England Journal of Medicine


Downloaded from nejm.org at UNIVERSIDAD INTERNACIONAL DEL ECUADOR UIDE on September 18, 2023. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
Brief Report

terim analysis of the results is now reported in vaso-occlusive crisis in December 2020, the patient
the Journal.8 Here, we report a case of acute my- was noted to have 2% blast cells in peripheral
eloid leukemia that developed in a female patient blood, which was thought to be related to recov-
approximately 5.5 years after she had received ery after infection; the presence of blast cells
gene therapy with LentiGlobin as part of the ini- was not reported in subsequent blood analyses.
tial cohort (Group A) of the HGB-206 study, as In January 2021, neutropenia (defined as an ab-
well as the results of investigations to deter- solute neutrophil count of <1000 per microliter)
mine whether the development of acute myeloid developed, and in February 2021, acute myeloid
leukemia was vector-mediated. leukemia (subtype M0 according to the French–
American–British classification) was diagnosed
C a se R ep or t after the detection of 29% blast cells in peripheral
blood and 22 to 50% blast cells in bone marrow
Before receiving gene therapy with LentiGlobin, (Table 1). Given the potential of insertional on-
the patient had sickle cell disease (βS/βS geno- cogenesis by the lentiviral vector and the expo-
type) with recurrent vaso-occlusive crises that had sure to myelotoxic chemotherapy with busulfan,
led to frequent hospitalizations despite the use of the acute myeloid leukemia was studied to deter-
long-term hydroxyurea therapy. She was 31 years mine causality and relation to gene therapy. Chro-
of age when she received a diagnosis of acute mosomal microarray analysis of the bone marrow
myeloid leukemia in February 2021 (Table 1). samples revealed monosomy 7 in 70% of cells
In August 2015, as part of the Group A cohort and partial loss of 11p involving WT1 in 50% of
in the HGB-206 study, the patient received Lenti­ cells. A next-generation sequencing myeloid mu-
Globin produced from HSCs harvested from bone tation panel revealed a RUNX1 frameshift muta-
marrow through an earlier version of the drug- tion and a PTPN11 missense mutation (Table 1).
product manufacturing process as part of the Retrospective next-generation sequencing and
initial study protocol. The manufacturing process single-nucleotide-polymorphism microarray anal-
was later modified for subsequent patients en- yses of retained pretreatment peripheral-blood
rolled in Groups B and C (Table S1 in the Sup- leukocytes did not reveal any genetic abnormali-
plementary Appendix, available with the full text ties, nor were any abnormalities observed in any
of this article at NEJM.org). The drug product retained post-treatment sample up to month 48
administered in the patient had a total CD34+ (the most recent sample obtained before the di-
cell dose of 2.6 × 106 cells per kilogram of body agnosis of acute myeloid leukemia). Further anal-
weight (of which 1.5 × 106 cells per kilogram were ysis of the peripheral-blood samples revealed that
CD34hi long-term repopulating hematopoietic stem the blast cells were positive for BB305 lentiviral
and progenitor cells [HSPCs]) and a vector copy vector. Specifically, CD34+ blast-enriched cell pop-
number of 0.57 copies per diploid genome (Ta- ulations in samples of both peripheral-blood and
ble 1). At day 16 after LentiGlobin treatment, the bone marrow contained an integrated lentiviral
patient received granulocyte colony-stimulating vector, with vector copy numbers of 1.12 copies
factor because of fever, and subsequent neutro- per diploid genome and 1.17 copies per diploid
phil and platelet engraftments occurred at 19 genome, respectively. The percentage of lentivi-
and 31 days after treatment, respectively. At 6 ral vector–positive cells was 92.1% in the CD34+
months after treatment, the vector copy number peripheral-blood sample and 86.7% in the CD34+
in peripheral blood was 0.05 copies per diploid bone marrow sample. The investigator assessed
genome and the level of gene therapy–derived the acute myeloid leukemia as possibly related to
hemoglobin (HbAT87Q) was 4.33% of total non- her gene therapy.
transfused hemoglobin, findings that suggest low After acute myeloid leukemia was diagnosed,
engraftment of gene-modified cells and low trans- the patient underwent three cycles of induction
gene expression with minimal clinical benefit, chemotherapy, which led to morphologic remis-
because the pretransplantation sickle cell disease sion, but she remained positive for minimal resid-
phenotype persisted and led to the need for ual disease with a blast-cell percentage of 0.2%.
hydroxyurea therapy and packed red-cell trans- She underwent a human leukocyte antigen–hap-
fusions (Table 1 and Table S2). loidentical peripheral-blood stem-cell transplan-
During an emergency department visit for a tation in June 2021 and remained positive for

n engl j med 386;2 nejm.org January 13, 2022 139


The New England Journal of Medicine
Downloaded from nejm.org at UNIVERSIDAD INTERNACIONAL DEL ECUADOR UIDE on September 18, 2023. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Table 1. Baseline and Treatment Characteristics and Clinical Outcomes in a Patient with AML after LentiGlobin Treatment for Sickle Cell Disease.*

Variable Patient Information


Demographic characteristic
Age at treatment (date of treatment) 25 yr (Aug. 2015)
Sex Female
Genotype βS/βS
Race Black
Family history of AML or myelodysplastic syndrome No
Smoking history Never smoked
Clinical feature of sickle cell disease in the 2 yr preceding
consent
Hydroxyurea use Yes (approximately 6 yr of long-term use; treatment was discontinued
approximately 6 mo before LentiGlobin treatment)
Regular transfusions of packed red cells No
Stroke No
Previous complications of sickle cell disease Recurrent vaso-occlusive crisis, osteonecrosis, avascular necrosis of right
hip, chronic pain due to sickle cell disease, deep-vein thrombosis
Transplantation characteristic
Mean estimated area under the plasma 4084 μmol × min
busulfan concentration–time curve
Neutrophil engraftment† 19 days after treatment
Platelet engraftment‡ 31 days after treatment
Drug-product characteristic
Source of hematopoietic stem cells in Group A Manufactured from bone marrow harvest§
Mean VCN in the drug product 0.57 copies/diploid genome
Total CD34+ cell dose 2.6 × 106/kg
Long-term hematopoietic stem and progenitor cells (CD34 ) hi
1.5 × 106/kg
Post-treatment clinical course
VCN in peripheral blood at 6 mo 0.05 copies/diploid genome
Gene therapy–derived hemoglobin (HbAT87Q) level at 6 mo 4.33% of nontransfused total hemoglobin
Clinical signs and complications No improvement in hemolysis, continued vaso-occlusive crisis, chronic pain
Sickle cell disease–related concomitant medications and Granulocyte colony-stimulating factor (days 16 and 17 after treatment),
transfusions regular transfusions of packed red cells, hydroxyurea (days 186–229,
282–520, and 1018–1754 after treatment)
AML diagnosis
Interval between treatment and diagnosis (date of diagnosis) Approximately 5.5 yr (Feb. 2021)
Clinical course leading to AML diagnosis
Dec. 28, 2020 2% Blast cells in peripheral blood (resolved, thought to be due to infection
recovery); white-cell count, 7030 per microliter
Feb. 2, 2021 9% Blast cells in peripheral blood; white-cell count, 6450 per microliter
Feb. 8, 2021 29% Blast cells in peripheral blood; white-cell count, 10,900 per microliter
Feb. 9, 2021 22–50% Blast cells in bone marrow with confirmed diagnosis of AML (sub-
type M0 according to the French–American–British classification); white-
cell count, 12,800 per microliter
Feb. 9, 2021 Analysis of bone marrow identified RUNX1 exon 5 stop-gained p.A149*fs
(VAF, 26%); PTPN11 exon 3 missense p.A72V (VAF, 30%); monosomy 7
(70% of cells); partial loss of 11p (50% of cells)

140 n engl j med 386;2 nejm.org January 13, 2022

The New England Journal of Medicine


Downloaded from nejm.org at UNIVERSIDAD INTERNACIONAL DEL ECUADOR UIDE on September 18, 2023. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
Brief Report

Table 1. (Continued.)

Variable Patient Information


Vector and blast-cell analysis
CD34+ blast cells determined to be lentiviral vector–positive
VCN in peripheral blood 1.12 copies/diploid genome (92.1% LVV+ cells)
VCN in bone marrow 1.17 copies/diploid genome (86.7% LVV+ cells)
Integration-site analysis Vector in blast cells with a single insertion site in VAMP4

* AML denotes acute myeloid leukemia, LVV+ lentiviral vector–positive, VAF variant allele frequency, and VCN vector copy number.
† Neutrophil engraftment was defined as the first day of absolute neutrophil count of at least 500 per microliter for three consecutive mea-
surements on different days after the initial post-transplant nadir by day 43, without receiving backup cells at any time during the neutrope-
nic phase.
‡ Platelet engraftment was defined as the first of 3 consecutive platelet counts of at least 50,000 per microliter on different days after the ini-
tial post-treatment nadir, without platelet transfusions for 7 days immediately preceding and during the evaluation period.
§ The drug-product manufacturing process was later modified for subsequent patients enrolled in Groups B and C in the HGB-206 study.

minimal residual disease. The patient had a re- ed specific quantitative polymerase-chain-reac-
lapse with the reappearance of circulating blast tion analyses designed to estimate the fractional
cells 90 days after transplantation and underwent contribution of the clone carrying the VAMP4
additional chemotherapy; however, she later died insertion site in peripheral blood. Until January
from complications of progressive acute myeloid 2020 (54 months after treatment), the VAMP4
leukemia. insertion site was present in approximately 1%
of peripheral-blood cells in the patient, and by
August 2020, this increased to 7% of peripheral-
Me thods
blood cells (Fig. 1B). This increase coincided
The phase 1–2 HGB-206 study evaluated the use with an overall increase in the vector copy num-
of LentiGlobin in patients with sickle cell disease. ber in peripheral blood during this period,
The drug product was manufactured by trans- which increased from approximately 0.05 copies
duction of autologous enriched CD34+ stem per diploid genome to 0.13 copies per diploid
cells with BB305 lentiviral vector encoding the genome. Concurrent with this rise, decreases in
antisickling βA-T87Q-globin gene. The structure of white-cell and absolute neutrophil counts were
the BB305 lentiviral vector has been previously also observed; these findings suggest a potential
described.5 To assess whether the acute myeloid correlation of these measures (Fig. 1C).
leukemia in the patient was due to insertional A single insertion was identified in VAMP4 in
oncogenesis, we performed integration site analy- the intronic region between exons 4 and 5 that
sis and RNA sequencing, as described previous- did not disrupt a putative enhancer (approxi-
ly9-13 and detailed in the Supplementary Appendix. mately 5 kilobases upstream) or the promoter
(approximately 30 kilobases upstream). An in-
sertion site in the VAMP4 gene body was present
R e sult s
in most patients (25 of 35 [71%]) in the HGB-206
Integration Site Analysis study who had received LentiGlobin and had
We performed an integration site analysis to available data from integration site analysis. A
evaluate the role of vector insertion in the devel- total of 60 unique insertions in the VAMP4 gene
opment of acute myeloid leukemia. Many unique locus were mapped, of which 14 were localized
insertion sites (a total of 2997) were detected in to the intronic region between exons 4 and 5,
the patient; however, the insertion site in the including the VAMP4 insertion site in the patient
vesicle-associated membrane protein 4 gene with acute myeloid leukemia (Fig. 1D). As com-
(VAMP4) increased over time, reaching a relative pared with other protein-coding genes, VAMP4
frequency of more than 60% in August 2020 was in the 68th percentile for insertion sites when
(Fig. 1A) and 98.6% in February 2021. The in- normalized for gene length. In all other patients
crease in VAMP4 insertion-site frequency prompt- in the HGB-206 study, VAMP4 insertion sites were

n engl j med 386;2 nejm.org January 13, 2022 141


The New England Journal of Medicine
Downloaded from nejm.org at UNIVERSIDAD INTERNACIONAL DEL ECUADOR UIDE on September 18, 2023. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A B
100 VAMP4 1.0

PB VCN (copies/diploid genome)


90 MSH5
80 RHOT1
Sequence Count (%) 70 LOC100294145
60 WNK1
STOM
50 0.5
FBXW7
40 POLR2G
30 Insertion
NOSIP site–specific
20 VAV1 VCN
10 All other insertion sites Overall VCN

0 0.0
August 2020 0 12 24 36 48 60 72
Months since LentiGlobin Treatment

C
20,000

White-cell count
Cell Count (per microliter)

15,000

10,000

5,000
Absolute neutrophil count

0
0.0 0.5 1.0 12 24 36 48 60 64 65 66 67
Months since LentiGlobin Treatment

D
BB305 VAMP4 insertion sites BB305 VAMP4 insertion sites BB305 VAMP4 insertion site
in all patients with insertion sites in all patients with insertion sites in patient with AML
outside the intronic region between inside the intronic region between
exons 4 and 5 exons 4 and 5

BB305
Insertion Site

VAMP4
Exon 8 Exon 7 Exon 5 Exon 4 Exon 3 Exon 2 Exon 1
Exon 6
17,167× 104 17,168× 104 17,169× 104 17,170× 104 17,171× 104 17,172× 104 17,173× 104
Location on Chromosome 1 (no. of base pairs)

not associated with sequelae, including clonal was similar to that in mobilized peripheral-blood
expansion. CD34+ cells from a healthy donor (Fig. 2A and
Table S3). Next, we measured HBB expression lev-
Gene Expression Analysis els, including lentiviral vector–derived βA-T87Q and
We performed RNA-sequencing analysis to de- endogenous βS transcripts, in different cell frac-
termine the transcriptional effect of the VAMP4 tions. The highest expression levels of HBA1 and
insertion site. Given the location of the insertion HBB were in unsorted and CD34− bone marrow
site, we first determined whether VAMP4 expres- cells, which is expected in cell populations en-
sion had changed. An analysis of the CD34+ cells riched for the presence of erythroblasts and
from the patient showed VAMP4 expression that erythroid progenitors. In contrast, in CD34+ pe-

142 n engl j med 386;2 nejm.org January 13, 2022

The New England Journal of Medicine


Downloaded from nejm.org at UNIVERSIDAD INTERNACIONAL DEL ECUADOR UIDE on September 18, 2023. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
Brief Report

indicated very low expression levels in blast cells.


Figure 1 (facing page). Lentiviral Vector Integration-Site
Among the CD34+ peripheral-blood HBB tran-
Analysis and Cell Counts in a Patient with AML after
LentiGlobin Treatment for Sickle Cell Disease. scripts, the majority (60%) were endogenous βS
transcripts — 40% derived from the therapeutic
Acute myeloid leukemia (AML) developed in a patient
who had been treated in the initial cohort (Group A) of
transgene βA-T87Q. In a finding consistent with
the HGB-206 study of LentiGlobin for sickle cell disease
the site and orientation of vector insertion, we
(bb1111, lovotibeglogene autotemcel).8 Panel A shows
also detected very low levels of HBB–VAMP4 and
the distribution of gene-marked cell clones at the un-
VAMP4–HBB fusion transcripts (<1 normalized
scheduled visit by the patient in August 2020, as deter-
fusion fragment transcript per million for CD34+
mined by integration-site analysis at GeneWerk. Each
of the top 10 most frequently detected unique insertion
and CD34− peripheral-blood samples). These re-
sites are indicated by a different color, with red show-
sults showed that the BB305 lentiviral vector had
ing VAMP4 (64.9%) and gray showing the cumulative
minimal transcriptional activity in blast cells
proportion of all other unique insertion sites (723 sites
and no noticeable effect on VAMP4 expression.
[27.6%]). Other than VAMP4, none of the insertion sites
We investigated whether the VAMP4 insertion
showed a relative frequency of more than 5% of the to-
tal retrieved insertion sites. Panel B shows the overall
site affected gene expression across the 10-mega-
vector copy number (VCN) in peripheral blood (PB),
base region flanking VAMP4. An analysis of the
as determined by central laboratory analysis, and the
CD34+ cells from the patient, as compared with
VAMP4 insertion site–specific PB VCN, as determined
CD34+ cells from a healthy donor, showed gene-
by analysis at GeneWerk. Insertion site–specific VCN
expression ratios centered around 1 (Fig. 2B).
can be used to estimate the fractional contribution of
the VAMP4-containing clone within the analyzed popu-
These population-level data indicated that ex-
lation. Samples were analyzed from scheduled visits
pression levels of genes proximal to the VAMP4
(≤60 months after LentiGlobin treatment) and unsched-
locus in CD34+ bone marrow cells from the
uled visits (>60 months after treatment, August 2020–
patient were similar to those in the CD34+ mo-
February 2021). Panel C shows the white-cell count and
bilized peripheral-blood cells from the healthy
absolute neutrophil count, as determined by central-
laboratory analysis of data from scheduled visits; these
donor. An assessment of genes within 100 kilo-
data are available from the clinical databases of the
bases of the VAMP4 locus revealed no or minimal
HGB-206 study and its long-term follow-up study LTF-
changes in expression level in analyzed genes in
307 (ClinicalTrials.org number, NCT04628585). Analysis
all patient samples, particularly in genes with a
of samples from unscheduled visits (>60 months; De-
transcripts-per-million value of 1.0 or greater
cember 2020–February 2021) were determined by local
laboratory analysis; these data were available from the
(Fig. 2B and Table S3). An analysis of all chro-
safety database of the LTF-307 study. Panel D shows
mosomes showed a ratio of protein-coding gene
the frequency and location of VAMP4 insertions in the
transcripts centered around 1, indicating that
patients treated with LentiGlobin in the HGB-206 study;
the only chromosome-wide differences in gene
integration-site analysis data were derived from an ex-
expression between the patient and the healthy
ploratory research database (first integration-site analy-
sis data at 6 months after treatment). Open black trian-
donor occurred in chromosome 7 (Fig. S2). The
gles represent unique insertion sites, and open red
shift in the gene-expression ratio for chromo-
triangles represent insertion sites inside the intronic
some 7 is consistent with monosomy 7. Finally,
­region between exons 4 and 5, including the VAMP4
targeted whole-genome sequencing analysis of
­insertion site of interest in the patient with AML repre-
the 10-megabase flanking region of the VAMP4
sented by a solid red triangle. The triangles are offset
from each other to enhance visibility. Below the inser-
insertion site showed no chromosomal abnor-
tion sites, the 8 exons of VAMP4 are represented by
malities or copy-number alterations, which sug-
purple bars, and the intronic regions are in gray. VAMP4
gests that BB305 lentiviral vector insertion did not
exon boundaries, derived from the principal isoform
result in unexpected chromosomal damage in this
from the APPRIS (annotating principal splice isoforms)
region (data not shown). These results indicated
database, are extracted from GENCODE, version 36,
and overlaid onto the GRCh37 (hg19) human reference
that gene expression within 10 megabases of the
genome.
VAMP4 insertion site was not noticeably affected,
and no chromosomal abnormalities were ob-
served. The overall transcriptional profile in the
ripheral-blood cells enriched for leukemic blast blast cells was consistent with monosomy 7, a
cells, HBB expression was 0.6% of that detected common genetic lesion in alkylating agent-induced
in the sample of CD34− bone marrow cells, which acute myeloid leukemia.15

n engl j med 386;2 nejm.org January 13, 2022 143


The New England Journal of Medicine
Downloaded from nejm.org at UNIVERSIDAD INTERNACIONAL DEL ECUADOR UIDE on September 18, 2023. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
144
A
VAMP4 HBA1 HBB
Blast- Blast- Blast- Blast- Blast- Blast-
enriched depleted enriched depleted enriched depleted
80 25,000 120,000
60 20,000 90,000
15,000
40 60,000
10,000
20 5,000 30,000
0 0 0
M B P B PB 1 2 PB M B M PB PB 1 2 PB PB B PB 1 2 PB

Transcripts per Million


BM ,B ,P BM −, m ,# ,# BM ,B ,P ,B −, m ,# ,# m BM , BM BM − , P m ,# ,# m
d + + −, , M M ,m d + + − , M M , d + 4 +, −, 4 , M M ,
rte 34 34 34 34 3+ B B + rte 34 34 34 34 3+ ,B ,B 4+ tr e 3 4 3 3 4 3 + B B +
so CD CD CD 3 −, −, 34 so CD CD CD 3 − − 3 so CD CD CD 33 −, −, 34
CD CD 34 34 CD CD CD 34 34 CD CD CD 34 34 CD
Un D D Un Un D D
C C CD CD C C

Patient with AML Healthy Donors Patient with AML Healthy Donors Patient with AML Healthy Donors

B
Chromosome 1 10-Megabase flanking VAMP4
400 25
The

300 20
15
200
10
100

No. of Genes
5
0 0
0 2 4 0 2 4

n engl j med 386;2


Expression Ratio Expression Ratio
DNM3 METTL13 MYOC
Blast- Blast- Blast- Blast- Blast- Blast-

nejm.org
enriched depleted enriched depleted enriched depleted
8 30 25
6 25 20
n e w e ng l a n d j o u r na l

20 15

The New England Journal of Medicine


4 15
of

10 10
2 5 5
0 0 0
B B PB 1 2 PB
BM , BM ,P ,# ,#

January 13, 2022


BM − , P
PFN1P1 RPL4P3 d + 4+ 4 −, 4 M ,m
rte 3 4 3 3 3 +, m BM B +
Blast- Blast- Blast- Blast- so CD CD CD 33 −, −, 34

Copyright © 2022 Massachusetts Medical Society. All rights reserved.


CD CD 34 34 CD
m e dic i n e

enriched depleted enriched depleted Un


0.8 0.4 CD CD
0.6 0.3

Transcripts per Million


Patient with AML Healthy Donors
0.4 0.2
0.2 0.1
0.0 0.0
M PB P B PB 1 2 PB M PB M PB PB 1 2 PB
BM ,B BM −, m,# ,# BM ,B ,B −, m ,# ,# m
d + +, −, , M M ,m d + +, − , M M ,
rte 34 34 34 34 3+ B B + rte 34 34 34 34 3+ ,B ,B 4+
so CD CD CD 3 −, −, 34 so CD CD CD 3 − − 3
CD CD 34 34 CD CD CD 34 34 CD
Un Un
CD CD CD CD

Patient with AML Healthy Donors Patient with AML Healthy Donors

Downloaded from nejm.org at UNIVERSIDAD INTERNACIONAL DEL ECUADOR UIDE on September 18, 2023. For personal use only. No other uses without permission.
Brief Report

Figure 2 (facing page). Gene Expression Analysis


leukemic blast cells, which suggests that blast cells
in AML Blast Cells and within 10 Megabases originated from a transduced HSC and not from
of the VAMP4 Locus. residual unablated host cells exposed to busulfan.
Panel A shows the normalized expression levels for This report provides several lines of evidence
VAMP4, HBA1, and HBB, as determined by bulk RNA- showing that the development of this case of
sequencing analysis. The increased expression of VAMP4 acute myeloid leukemia most likely occurred in-
in blast-depleted CD34− bone marrow (BM) cells is
probably driven by CD34+ myeloblasts differentiating
dependently of insertional oncogenesis. First,
into CD34− erythroid progenitor cells.14 This increased the insertion site was in VAMP4, a gene that is
expression of VAMP4 is supported by the concurrent known to play a role in Golgi structure and
increase in HBA1 and HBB expression in this erythro- function16,17 but has no known role in cellular
blast-containing population, the increase in antisickling proliferation or oncogenesis on the basis of an
hemoglobin HbAT87Q that coincides with the emergence
of this clone (Fig. S1), and the increased detection of
analysis of the published literature and gene
fusion transcripts between VAMP4 and the transgenic databases (COSMIC [Catalogue of Somatic Muta-
βA-T87Q in this CD34− BM cell population, the number tions in Cancer], cBioPortal, DepMap [Dependency
of which was more than 10 times as high as that in Map], and CCGD [Candidate Cancer Gene Data-
CD34− PB cells. The y axis scale was adjusted for each base]), which included sequenced cancer tissue
gene. Panel B shows histograms of gene-expression
ratios on chromosome 1 (upper left) and within 10
samples from more than 2000 patients with acute
megabases of VAMP4 (upper right) comparing CD34+ myeloid leukemia and 800 human-derived can-
BM cells from the patient with AML with CD34+ mobi- cer cell lines (see the Supplementary Appendix).
lized PB (mPB) cells from a healthy donor. The labels Second, the insertion site mapped to a noncoding
of the upper-left and upper-right histograms indicate region of VAMP4 in a location with no annotated
the compared loci; the x axes represent the expression
ratio between CD34+ BM cells from the patient with
genomic features, a finding that is in line with
AML (numerator) and CD34+ BM cells from a healthy the frequent integration of lentiviral vector into
donor (denominator); and the y axes denote the num- introns18 because of their larger size relative to
ber of genes that populate each binned range. The red exons in a gene. Third, a VAMP4 insertion site was
dotted lines indicate an expression ratio of one. The present in most evaluated patients with sickle
chromosome for which the distribution peak is on the
dotted line indicates no overall change in protein-coding
cell disease (71%) who received treatment in the
gene expression for the region of interest. The bottom HGB-206 study and had no sequelae. Our findings
set of graphs shows the normalized expression levels are in keeping with those reported for other
for the five genes closest to VAMP4 (DNM3, METTL13, lentiviral vectors, which frequently integrate into
MYOC, PFN1P1, and RPL4P3), as determined by bulk VAMP4.19 Finally, the BB305 lentiviral vector was
RNA-sequencing analysis; the y axis scales were ad-
justed for each gene. In Panels A and B, the purity of
minimally transcriptionally active in blast cells and
sorted fractions ranged from 90.69 to 97.90% (CD34+ did not noticeably alter the expression of proxi-
PB, 90.69%; CD34− PB, 95.57%; CD34+ BM, 95.38%; mal genes, and no chromosome-wide changes in
and CD34− BM, 97.90%). CD34+ and CD33+ mPB cells gene expression were observed, except in chromo-
were derived from a single healthy donor, and each some 7, which is consistent with monosomy 7.
CD34– BM sample (#1 and #2) was derived from a
different healthy donor.
Because the development of acute myeloid
leukemia was most likely independent of vector
insertion, other contributing factors in both pa-
tients should be considered. Genetic testing iden-
Discussion
tified several known driver alterations of acute
Among the seven patients who received Lenti- myeloid leukemia in the blast cells from the cur-
Globin treatment in the initial cohort (Group A) rent patient (RUNX1, PTPN11, monosomy 7, and
of the HGB-206 study, this patient is the second partial loss of 11p), which suggests a molecular
to present with acute myeloid leukemia. Under- basis for the findings observed in this case. In
standing potential risk factors for the develop- the previous case diagnosed in 2018, a similar
ment of hematologic malignant conditions is mutation profile (RUNX1, PTPN11, and monoso-
important for communities affected by sickle cell my 7) was observed.15 Both patients could have
disease and persons who are considering gene accumulated several mutations predisposing them
therapy. In contrast to the case of acute myeloid to the development of acute myeloid leukemia
leukemia that was diagnosed in 2018,15 the case over the course of their sickle cell disease and
in the current patient showed vector present in their treatment, resulting in a clone that led to

n engl j med 386;2 nejm.org January 13, 2022 145


The New England Journal of Medicine
Downloaded from nejm.org at UNIVERSIDAD INTERNACIONAL DEL ECUADOR UIDE on September 18, 2023. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

acute myeloid leukemia. The mechanism behind expression and an inadequate therapeutic response,
the accumulation of these somatic mutations is with ongoing hemolysis and persistent anemia
likely to be multifactorial, with contributions from — outcomes that subjected transplanted and
the transplantation procedure and its associated endogenous bone marrow cells to continued he-
risks, the underlying sickle cell disease, and the matopoietic stress after treatment, providing fur-
continued hematopoietic stress resulting from ther opportunity for accumulation of mutations.
the minimal clinical benefit derived from Lenti- Before the development of malignant disease
Globin, which was observed in both cases.15 in these two patients, alterations to the HGB-206
Among patients with sickle cell disease, the study protocol were necessary to improve clinical
risk of hematologic malignant conditions is 2 to benefit; thus, the initial protocol and manufac-
11 times as high as that in the general popula- turing process used in Group A underwent sev-
tion.20,21 Factors that probably contribute to this eral changes to improve HSC collection and trans-
increased baseline risk include the underlying duction efficiency. The protocol used in Group C
pathophysiological mechanism of chronic hypoxia, includes a minimum of 60 days of packed red-cell
the generation of reactive oxygen species with as- transfusions before HSC collection, HSC collec-
sociated oxidative stress, the chronic inflamma- tion by mobilization with plerixafor followed by
tion, the endothelial and vascular damage and apheresis, and an increased target value for the
constant erythropoietic stress, the high cell turn- area under the plasma busulfan concentration–
over, and the damage to the HSC compartment time curve during conditioning. Some of these
in bone marrow.1,2,20,21 Use of hydroxyurea for modifications have positively affected the quality
sickle cell disease has been suggested as a pos- (CD34hi) and number of HSCs available for ex vivo
sible risk factor, but analyses of data accumu- manipulation (Table S1). In addition, the improve-
lated over decades from patients with sickle cell ment in transduction efficiency resulting from
disease and chronic myeloid leukemia have not the refining of the manufacturing process has
established a link.22 The transplantation proce- resulted in improved levels of antisickling hemo-
dure, including the use of myeloablative condi- globin.27 Together, these protocol improvements
tioning agents such as busulfan, carries inherent are anticipated to reduce proliferative and hema-
risks of malignant disease.23-25 Furthermore, af- topoietic stresses and therefore may contribute
ter myeloablation, the bone marrow niche under- to reducing the risk of acute myeloid leukemia
goes extensive proliferation of HSCs and HSPCs, after transplantation in patients in Group C. Al-
generating proliferative stress that may lead to though follow-up was limited (median duration,
the rise and accumulation of mutations as part 20.9 months [range, 8.5 to 43.1] as of July 26,
of the normal engraftment process.26 If a lower 2021), no cases of malignant disease have been
cell dose is administered to a patient after mye- reported in Group C patients to date.
loablation, fewer HSCs and HSPCs are available In summary, acute myeloid leukemia devel-
to repopulate the bone marrow, which leads to oped in a patient who had received LentiGlobin
the need for more replication cycles and hence for sickle cell disease as part of the initial cohort
additional proliferative stress and a higher prob- (Group A) of the HGB-206 study; the develop-
ability of acquiring a mutation that could lead to ment of this malignant condition was most
acute myeloid leukemia. likely independent of insertional oncogenesis.
The two patients in Group A of the HGB-206 We have suggested possible contributing factors
study in whom acute myeloid leukemia developed to the development of acute myeloid leukemia;
had received LentiGlobin produced from bone however, a higher proportion of transgene-express-
marrow–harvested HSCs through an earlier ver- ing cells may eliminate many of them. Longer
sion of the drug-product manufacturing process. follow-up is required to assess the effect of trans-
Cell collections from the bone marrow with the plant-mediated gene therapy for patients with
use of this process resulted in low cell doses, sickle cell disease.
with fewer CD34hi long-term HSPCs, leading to Supported by Bluebird Bio. Medical writing support was
increased proliferative stress (Table S1). The low provided by Luke K. Burke, Ph.D. (Synergy Medical Communica-
tions, UK) and was funded by Bluebird Bio.
transduction efficiency of the drug product in Disclosure forms by the authors are available with the full
these patients resulted in minimal transgene text of this article at NEJM.org.

146 n engl j med 386;2 nejm.org January 13, 2022

The New England Journal of Medicine


Downloaded from nejm.org at UNIVERSIDAD INTERNACIONAL DEL ECUADOR UIDE on September 18, 2023. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
Brief Report

We thank the staff at HGB-206 clinical site for their important Smith, Joyce O’Connell (former employee), Perrine Geoffroy,
contributions to the care of the patient described in this article; and Katherine Lewis for their contributions; Donald B. Kohn
members of the Bluebird Bio team: Alberto De Iaco (former em- (Broad Stem Cell Research Center, University of California, Los
ployee), Brandon Gentile, Brandon V. Nguyen, Chetanya Pandya Angeles), Andreas E. Kulozik (Department of Pediatric Oncol-
(former employee), Christopher Mucci, Cynthia Rogers, James ogy, Hematology, and Immunology, University of Heidelberg),
Rottman (former employee), Katie Groglio (former employee), Franco Locatelli (IRCCS Ospedale Pediatrico Bambino Gesù),
Lauryn M. Christiansen, Madhumita Mahajan (former employ- Ryotaro Nakamura (Division of Hematology and HCT, City of
ee), Maggie Chen (former employee), Mauris Nnamani (former Hope National Medical Center), Eli Pasackow (Generation Bio),
employee), Megan D. Hoban, Nick Fenger, Nick Rouillard (for- Christof von Kalle (National Center for Tumor Diseases, Heidel-
mer employee), Noel Namai (former employee), Omar Elbeta- berg and German Cancer Research Center), David A. Williams
nony (former employee), Russell W. Goetze (former employee), (Boston Children’s Hospital and Dana–Farber Cancer Institute,
Ryan Leenay, Shelby LaBarre (former employee), Stephanie Harvard Medical School and Harvard Stem Cell Institute), and
Hintzen-Matoian, Swathi Krishnan (former employee), Tiffany John Wagner (Division of Blood and Marrow Transplantation,
Hu (former employee), Xinyan Zhang, Suzanne Plezier Jonkheer, Department of Pediatrics, University of Minnesota) for helpful
Dennis Kim (former employee), Briana Deary, Natalia Mackow- discussions; and Pamela Y. Chan of Bluebird Bio for writing and
iak, Leanne Ianniello, Heidi Elliot, Laura Demopoulos, Frances editorial assistance.

References
1. Kato GJ, Piel FB, Reid CD, et al. Sickle 11. Aiuti A, Biasco L, Scaramuzza S, et al. 20. Brunson A, Keegan THM, Bang H,
cell disease. Nat Rev Dis Primers 2018;​4:​ Lentiviral hematopoietic stem cell gene Mahajan A, Paulukonis S, Wun T. In-
18010. therapy in patients with Wiskott-Aldrich creased risk of leukemia among sickle cell
2. Sundd P, Gladwin MT, Novelli EM. syndrome. Science 2013;​341:​1233151. disease patients in California. Blood 2017;​
Pathophysiology of sickle cell disease. 12. Paruzynski A, Arens A, Gabriel R, et al. 130:​1597-9.
Annu Rev Pathol 2019;​14:​263-92. Genome-wide high-throughput integrome 21. Seminog OO, Ogunlaja OI, Yeates D,
3. Sheth S, Licursi M, Bhatia M. Sickle analyses by nrLAM-PCR and next-genera- Goldacre MJ. Risk of individual malig-
cell disease: time for a closer look at treat- tion sequencing. Nat Protoc 2010;​5:​1379- nant neoplasms in patients with sickle
ment options? Br J Haematol 2013;​ 162:​ 95. cell disease: English national record link-
455-64. 13. Schmidt M, Schwarzwaelder K, Bar- age study. J R Soc Med 2016;​109:​303-9.
4. Walters MC, De Castro LM, Sullivan tholomae C, et al. High-resolution inser- 22. Cuthbert D, Stein BL. Therapy-associ-
KM, et al. Indications and results of HLA- tion-site analysis by linear amplification- ated leukemic transformation in myelo-
identical sibling hematopoietic cell trans- mediated PCR (LAM-PCR). Nat Methods proliferative neoplasms — what do we
plantation for sickle cell disease. Biol 2007;​4:​1051-7. know? Best Pract Res Clin Haematol 2019;​
Blood Marrow Transplant 2016;​22:​207-11. 14. Agarwal A, Bolosky WJ, Wilson DB, 32:​65-73.
5. Negre O, Bartholomae C, Beuzard Y, et al. Differentiation of leukemic blasts is 23. Pagano L, Pulsoni A, Tosti ME, et al.
et al. Preclinical evaluation of efficacy not completely blocked in acute myeloid Acute lymphoblastic leukaemia occurring
and safety of an improved lentiviral vector leukemia. Proc Natl Acad Sci U S A 2019;​ as second malignancy: report of the
for the treatment of β-thalassemia and 116:​24593-9. GIMEMA archive of adult acute leukae-
sickle cell disease. Curr Gene Ther 2015;​ 15. Hsieh MM, Bonner M, Pierciey FJ, et al. mia. Br J Haematol 1999;​106:​1037-40.
15:​64-81. Myelodysplastic syndrome unrelated to 24. Adès L, Guardiola P, Sociè G. Second
6. Ribeil JA, Hacein-Bey-Abina S, Payen lentiviral vector in a patient treated with malignancies after allogeneic hematopoi-
E, et al. Gene therapy in a patient with gene therapy for sickle cell disease. Blood etic stem cell transplantation: new insight
sickle cell disease. N Engl J Med 2017;​376:​ Adv 2020;​4:​2058-63. and current problems. Blood Rev 2002;​16:​
848-55. 16. Shitara A, Shibui T, Okayama M, et al. 135-46.
7. Thompson AA, Walters MC, Kwiat- VAMP4 is required to maintain the ribbon 25. Leone G, Mele L, Pulsoni A, Equitani
kowski J, et al. Gene therapy in patients structure of the Golgi apparatus. Mol Cell F, Pagano L. The incidence of secondary
with transfusion-dependent β-thalassemia. Biochem 2013;​380:​11-21. leukemias. Haematologica 1999;​84:​937-
N Engl J Med 2018;​378:​1479-93. 17. Tran THT, Zeng Q, Hong W. VAMP4 45.
8. Kanter J, Walters MC, Krishnamurti cycles from the cell surface to the trans- 26. Nawas MT, Schetelig J, Damm F, et al.
L, et al. Biologic and clinical efficacy of Golgi network via sorting and recycling The clinical implications of clonal hema-
LentiGlobin for sickle cell disease. N Engl endosomes. J Cell Sci 2007;​120:​1028-41. topoiesis in hematopoietic cell transplan-
J Med. DOI:​10.1056/NEJMoa2117175. 18. Heffner GC, Bonner M, Christiansen tation. Blood Rev 2021;​46:​100744.
9. Six E, Guilloux A, Denis A, et al. Clon- L, et al. Prostaglandin E2 increases lentivi- 27. Tisdale JF, Pierciey FJ Jr, Bonner M, et al.
al tracking in gene therapy patients re- ral vector transduction efficiency of adult Safety and feasibility of hematopoietic
veals a diversity of human hematopoietic human hematopoietic stem and progeni- progenitor stem cell collection by mobili-
differentiation programs. Blood 2020;​ tor cells. Mol Ther 2018;​26:​320-8. zation with plerixafor followed by apher-
135:​1219-31. 19. Shao W, Shan J, Kearney MF, et al. esis vs bone marrow harvest in patients
10. Oshlack A, Robinson MD, Young MD. Retrovirus Integration Database (RID): with sickle cell disease in the multi-center
From RNA-seq reads to differential ex- a public database for retroviral insertion HGB-206 trial. Am J Hematol 2020;​95(9):​
pression results. Genome Biol 2010;​11:​ sites into host genomes. Retrovirology E239-E242.
220. 2016;​13:​47. Copyright © 2021 Massachusetts Medical Society.

n engl j med 386;2 nejm.org January 13, 2022 147


The New England Journal of Medicine
Downloaded from nejm.org at UNIVERSIDAD INTERNACIONAL DEL ECUADOR UIDE on September 18, 2023. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.

You might also like