ArticlePDF Available

Autopsy Study Defines Composition and Dynamics of the HIV-1 Reservoir after Allogeneic Hematopoietic Stem Cell Transplantation with CCR5Δ32/Δ32 Donor Cells

MDPI
Viruses
Authors:

Abstract and Figures

Allo-HSCT with CCR5Δ32/Δ32 donor cells is the only curative HIV-1 intervention. We investigated the impact of allo-HSCT on the viral reservoir in PBMCs and post-mortem tissue in two patients. IciS-05 and IciS-11 both received a CCR5Δ32/Δ32 allo-HSCT. Before allo-HSCT, ultrasensitive HIV-1 RNA quantification; HIV-1-DNA quantification; co-receptor tropism analysis; deep-sequencing and viral characterization in PBMCs and bone marrow; and post-allo-HSCT, ultrasensitive RNA and HIV-1-DNA quantification were performed. Proviral quantification, deep sequencing, and viral characterization were done in post-mortem tissue samples. Both patients harbored subtype B CCR5-tropic HIV-1 as determined genotypically and functionally by virus culture. Pre-allo-HSCT, HIV-1-DNA could be detected in both patients in bone marrow, PBMCs, and T-cell subsets. Chimerism correlated with detectable HIV-1-DNA LTR copies in cells and tissues. Post-mortem analysis of IciS-05 revealed proviral DNA in all tissue biopsies, but not in PBMCs. In patient IciS-11, who was transplanted twice, no HIV-1-DNA could be detected in PBMCs at the time of death, whereas HIV-1-DNA was detectable in the lymph node. In conclusion, shortly after CCR5Δ32/Δ32, allo-HSCT HIV-1-DNA became undetectable in PBMCs. However, HIV-1-DNA variants identical to those present before transplantation persisted in post-mortem-obtained tissues, indicating that these tissues play an important role as viral reservoirs.
Content may be subject to copyright.
Citation: Huyveneers, L.E.P.; Bruns,
A.; Stam, A.; Ellerbroek, P.; de Jong,
D.; Nagy, N.A.; Gumbs, S.B.H.;
Tesselaar, K.; Bosman, K.; Salgado,
M.; et al. Autopsy Study Defines
Composition and Dynamics of the
HIV-1 Reservoir after Allogeneic
Hematopoietic Stem Cell
Transplantation with CCR532/32
Donor Cells. Viruses 2022,14, 2069.
https://doi.org/10.3390/v14092069
Academic Editor: Sonia Moretti
Received: 30 August 2022
Accepted: 14 September 2022
Published: 17 September 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
viruses
Article
Autopsy Study Defines Composition and Dynamics of the
HIV-1 Reservoir after Allogeneic Hematopoietic Stem Cell
Transplantation with CCR532/32 Donor Cells
Laura E. P. Huyveneers 1, Anke Bruns 2,3, Arjen Stam 1, Pauline Ellerbroek 2, Dorien de Jong 1, Noémi A. Nagy 1,
Stephanie B. H. Gumbs 1, Kiki Tesselaar 4, Kobus Bosman 1, Maria Salgado 5,6 , Gero Hütter 7, Lodewijk A.
A. Brosens 8, Mi Kwon 9, Jose Diez Martin 9, Jan T. M. van der Meer 10 , Theun M. de Kort 11, Asier Sáez-Cirión12 ,
Julian Schulze zur Wiesch 13 , Jaap Jan Boelens 14,15 , Javier Martinez-Picado 5,6,16,17 , Jürgen H. E. Kuball 3,4 ,
Annemarie M. J. Wensing 1,*, and Monique Nijhuis 1,† ,‡ on behalf of the IciStem Consortium
1Department of Medical Microbiology, University Medical Center Utrecht (UMCU),
3584 CX Utrecht, The Netherlands
2Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU),
3584 CX Utrecht, The Netherlands
3
Department of Hematology, University Medical Center Utrecht (UMCU), 3584 CX Utrecht, The Netherlands
4Central Laboratory of Translational Immunology, University Medical Center Utrecht (UMCU),
3584 CX Utrecht, The Netherlands
5IrsiCaixa AIDS Research Institute and Institute for Health Science Research Germans Trias i Pujol (IGTP),
08916 Badalona, Spain
6
Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto
de Salud Carlos III, 28029 Madrid, Spain
7DKMS CC, 72072 Tübingen, Germany
8Department of Pathology, University Medical Center Utrecht (UMCU), 3584 CX Utrecht, The Netherlands
9Department of Hematology, Hospital General Universitario Gregorio Maranon, 28007 Madrid, Spain
10 Department of Internal Medicine, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
11 Department of Anatomy and Embryology, Leiden University Medical Center,
2333 ZA Leiden, The Netherlands
12 HIV Inflammation and Persistence, Institut Pasteur, UniversitéParis Cité, 75015 Paris, France
13 Department of Internal Medicine, UMC Hamburg-Eppendorf, 20251 Hamburg, Germany
14 Department of Hematology and Oncology, Princess Maxima Center, 3584 CS Utrecht, The Netherlands
15 Stem Cell Transplant and Cellular Therapies, Pediatrics, Memorial Sloan Kettering Cancer Center,
New York, NY 10065, USA
16 Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
17 University of Vic—Central University of Catalonia (UVic—UCC), 08500 Vic, Spain
*Correspondence: a.m.j.wensing@umcutrecht.nl
These authors contributed equally to this work.
Study Group team members are listed in the Acknowledgments.
Abstract:
Allo-HSCT with CCR5
32/
32 donor cells is the only curative HIV-1 intervention. We
investigated the impact of allo-HSCT on the viral reservoir in PBMCs and post-mortem tissue in
two patients. IciS-05 and IciS-11 both received a CCR5
32/
32 allo-HSCT. Before allo-HSCT, ul-
trasensitive HIV-1 RNA quantification; HIV-1-DNA quantification; co-receptor tropism analysis;
deep-sequencing and viral characterization in PBMCs and bone marrow; and post-allo-HSCT, ul-
trasensitive RNA and HIV-1-DNA quantification were performed. Proviral quantification, deep
sequencing, and viral characterization were done in post-mortem tissue samples. Both patients har-
bored subtype B CCR5-tropic HIV-1 as determined genotypically and functionally by virus culture.
Pre-allo-HSCT, HIV-1-DNA could be detected in both patients in bone marrow, PBMCs, and T-cell
subsets. Chimerism correlated with detectable HIV-1-DNA LTR copies in cells and tissues. Post-
mortem analysis of IciS-05 revealed proviral DNA in all tissue biopsies, but not in PBMCs. In patient
IciS-11, who was transplanted twice, no HIV-1-DNA could be detected in PBMCs at the time of death,
whereas HIV-1-DNA was detectable in the lymph node. In conclusion, shortly after CCR5
32/
32,
allo-HSCT HIV-1-DNA became undetectable in PBMCs. However, HIV-1-DNA variants identical to
those present before transplantation persisted in post-mortem-obtained tissues, indicating that these
tissues play an important role as viral reservoirs.
Viruses 2022,14, 2069. https://doi.org/10.3390/v14092069 https://www.mdpi.com/journal/viruses
Viruses 2022,14, 2069 2 of 12
Keywords: HIV-1; HIV persistence; reservoir; cure; tissue; CCR532; allo-HSCT
1. Introduction
Although antiretroviral therapy (ART) successfully averts HIV-1-related disease pro-
gression and saves lives of millions of HIV-infected individuals, the ongoing inflammation
and costs urgently call for curative strategies [
1
]. Current antiretroviral compounds target
several steps in the viral life cycle but do not target the integrated provirus nor suppress
HIV-1 transcription and production from the cellular reservoir. This integrated provirus
forms a stable viral reservoir and is the major barrier to HIV-1 eradication [1].
Several approaches are being explored to eliminate the viral reservoir and ultimately
convert HIV-1 infection into a curable disease. Successful viral remission is known in four
cases of treatment interruption after allogeneic hematopoietic stem cell transplantation
(allo-HSCT) with homozygous CCR5
32 donor cells: “The Berlin patient”, two partici-
pants in the IciStem cohort; IciS-36, IciS-19—also named “The London patient” and “The
Düsseldorf patient”; and recently in a woman of mixed race in New York. The cells used
for transplantation lack expression of surface CCR5, rendering them resistant to infection
by CCR5-tropic HIV-1 [
2
7
]. Though no viral rebound was observed in these patients
after treatment interruption (ATI), traces of proviral HIV-1-DNA could still be found in
some samples [
3
,
5
,
7
,
8
]. Also, in patients transplanted with CCR5-expressing donor cells
(CCR5WT), a profound reduction in the viral reservoir was observed. However, in these
patients, therapy interruption resulted in a delayed rebound of viruses comparable to
the pre-allo-HSCT PBMC population in “the Boston patients”, but not in “the Minnesota
patient” [912].
These data highlight the knowledge gap on how allo-HSCT impacts the viral reservoir
and which cellular or anatomical compartments fuel HIV-1 rebound. IciStem is an interna-
tional collaboration to guide and investigate the potential for HIV-1 cure after allo-HSCT
and has compiled the largest cohort of HIV-1-infected individuals receiving allo-HSCT
(www.icistem.org, accessed on 15 August 2022). Two IciStem CCR5
32 patients (IciS-05
and IciS-11) died after allo-HSCT. Post-mortem biopsies of these patients provided a unique
opportunity to broadly investigate the viral reservoir after allo-HSCT in multiple tissues,
which would never be feasible in living patients.
1.1. Cases
1.1.1. Patient IciS-05
A 38-year-old male presented in 1998 with a CD4
+
T-cell count of 46 cells/
µ
L whole
blood and an HIV-1 viral load (VL) >500,000 RNA copies/mL plasma. Despite several
changes in his therapy regimen, full suppression was not reached until 2008 (Table 1). Ex-
tensive drug resistance was selected and genotypic analyses of the viral envelope predicted
infection with CCR5-tropic virus. In 2011, poor-risk myelodysplastic syndrome (MDS)
was diagnosed, necessitating allo-HSCT in May 2012. Because no adult HLA-matched
unrelated CCR5
32 donor was found, a cord-blood transplant of a homozygous CCR5
32
donor (HLA-match 4/6) combined with an infusion of CD34+-cells of an HLA-unmatched
haploidentical family donor (CCR5wt) was performed, to aim for HIV-1 cure but also
to ensure faster engraftment (Table 1). Using general diagnostic assays, viral load was
20 copies/mL on the day of transplant and remained suppressed (<50 copies/mL) after
transplantation. Cord-blood donor chimerism in peripheral blood lymphocytes (PBLs) was
57% on day +16 and 100% on day +36. On day +54, chimerism reduced to 95%, and further
down to 85% at day +65. Unfortunately, the patient died of severe sepsis on day +68.
Viruses 2022,14, 2069 3 of 12
Table 1. Baseline clinical characteristics of patients.
IciS-05 IciS-11
Hematological Data
Hematological diagnosis MDS AML
Donor type/graft source Cord blood + CD34+-cells third party donor HLA-matched unrelated donor
Donor CCR5 Homozygous CCR532 First donor: homozygous CCR532, second donor:
heterozygous CCR532/WT
Recipient HLA
HLA-A*03:01;24:02;
HLA-B*07:02;35:01;
HLA-Cw*04:01;07:02; HLA-DRB1*01:01;04:04;
HLA-DQB1*03:02;05:01
HLA-A*01:01;02:01;
HLA-B*07:02;-;
HLA-Cw*07:02;-;
HLA-DRB1*15:01;-;
HLA-DQB1*06:02; -
Donor-recipient HLA match
4/6 cord blood donor (A*03:01; -; B*07:02; 35:02;
C*07:02; 04;01;
DRB1*01:01; 11:04);
50% haploidentical family member
(A*24:02;26:01;B*07:02; 14:01;C*07:02;08:02;
DRB1*04:04; 14:54; DQB1*03:02; 05:03)
10/10; 10/10
Pre-transplant chemotherapy None 2 induction cycles with cytarabine and idarubicin
Conditioning ATG, fludarabine with busulvex
ATG, fludarabine, and low-dose TBI before initial
transplant; ATG fludarabine and treosulfan before
second transplant
GvHD prophylaxis Cyclosporine, prednisone Cyclosporine, prednisone, and mycophenolic acid
mofetil
GvHD severity Acute skin GvHD grade 1 No GvHD
Virological data
Time from HIV-1 diagnosis to allo-HSCT 14 years 22 years
Time from start cART to allo-HSCT 14 years 19 years
Host CCR5 CCR5WT/WT CCR5WT/WT
Predicted HIV-1 co-receptor tropism CCR5-tropic virus (FPR 68.0–96.2%) CCR5-tropic virus (FPR 9.7–77.3%)
Phenotypic HIV-1 co-receptor tropism CCR5-tropic virus CCR5-tropic virus
HIV-1 subtype HIV-1 subtype B HIV-1 subtype B
cART History
1998: AZT/3TC, NFV. 2005: TNF, EFV, ATV/r 2006:
TNF, LPV/r, NVP 2006: TNF, SQV/r, NVP
2008: AZT/3TC, TNF, SQV/r
1996: AZT, DDI.
1996: D4T, 3TC, SQV
1997: D4T, 3TC, SQV/r
1999: D4T, 3TC, NVP
2003: TNF, 3TC, NVP
cART during allo-HSCT procedure
Day -152: TNF/FTC, DRV/r, RAL
Day -34 until +68: TNF/FTC, RAL, MVC, ENF
Day -5 until +37: ETR added
Day -19: TNF, FTC, DTG
Day +20 until +107: ABC/3TC, DTG
Plasma HIV RNA load at allo-HSCT 20 copies/ml <50/TND copies/ml
HCV Anti-HCV negative Anti-HCV negative
HBV HbsAg negative, anti-HBc positive, anti-Hbs
negative
HbsAg negative, anti-HBc positive, anti-Hbs
positive
CMV status pre-SCT Positive Positive
Donor CMV status Positive Positive
3TC, lamivudine. ABC, abacavir. Allo-HSCT, allogeneic hematopoietic stem cell transplantation. AML, acute
myeloid leukemia. Anti-HBc, antibody against hepatitis B core antigen. Anti-HBs, antibody against hepatitis B
surface antigen. ATG, anti-thymocyte globulin. ATV/r, ritonavir boosted atazanavir. AZT, zidovudine. cART,
combined antiretroviral therapy. CCR5, C-C chemokine receptor type 5. CCR5
32, C-C chemokine receptor
type 5 delta 32 mutation. CMV, cytomegalovirus. DDI, didanosine. DRV/r, ritonavir boosted darunavir. DTG,
dolutegravir. D4T, stavudine. EBV, Epstein-Barr virus. EBV Vca, EBV viral capsid antigen antibody. EBV EBNA,
EBV nuclear antigen antibody. EFV, efavirenz. ENF, enfuvirtide. ETR, etravirine. FTC, emtricitabine. FPR, False
Positive Ratio. GvHD, Graft-versus-host disease. HBV, hepatitis B virus. HbsAG, hepatitis B surface antigen. HCV,
hepatitis C virus. HLA, human leukocyte antigen. LPV/r, ritonavir boosted lopinavir. MDS, myelodysplastic
syndrome. MVC, maraviroc. NFV, nelfivnavir. NVP, nevirapine. RAL, raltegravir. RS, respiratory syncytial virus.
RTV or r, ritonavir. SQV/r, ritonavir boosted saquinavir. TBI, total body irradiation. TDF, tenofovir disoproxil
fumarate. TND, target not detected. TPHA, Treponema pallidum haemagluttination test. WT, wild type.
1.1.2. Patient IciS-11
A 38-year-old male was diagnosed with HIV-1 in 1993 with a CD4+ T-cell count of
440 cells
/. In March 1996, when CD4+ T-cell count fell below 200 cells/
µ
L whole blood, the
patient started dual therapy, quickly followed by triple therapy (Table 1). In December 2014,
Viruses 2022,14, 2069 4 of 12
when VL was suppressed and CD4+ T-cells were around 700 cells/
µ
L, he was diagnosed
with poor-risk AML. Genotypic analyses of the viral envelope predicted infection with
CCR5-tropic virus.
In April 2015, the patient received allo-HSCT with cells of a 10/10 HLA-matched
unrelated donor homozygous for the CCR5
32 mutation (Table 1). Before allo-HSCT, CD4
count was 283 cells/
µ
L and plasma VL remained suppressed. Donor chimerism decreased
from 60% to 0% between day +27 and +55. On day +41, donor chimerism in bone marrow
(BM) was only 18%, further indicating graft failure. On day +71, the patient received a
second transplant of a heterozygous CCR5
32/WT unrelated donor (HLA-match 10/10);
the first donor was not available. A month after the second allo-HSCT, on day +100 after the
first allo-HSCT, full donor chimerism was reached. On day +108 after the first allo-HSCT
and day +37 after the second allo-HSCT, the patient died due to respiratory failure.
2. Materials and Methods
2.1. Patients and Patient Material
This study included two IciStem patients. IciS-05 was included after signing a general
waiver for the use of body material for future research, according to the regulations of
the UMC Utrecht HSCT Biobank (HSCT study number 3440). IciS-11 was included after
written informed consent was acquired. Ethical approval by the Institutional Medical
Ethics Committee of the UMCU was obtained under protocol number NL53114.041.15.
Before allo-HSCT, plasma, serum, PBMCs, and BM were obtained and a leukaphere-
sis was performed. From the leukapheresis, 2–3
×
10
8
PBMCs were sorted into T
N
(CD3
+
CD4
+
CD45RO
-
CCR7
+
CD27
+
Fas
-
), T
SCM
(CD3
+
CD4
+
CD45RO
-
CCR7
+
CD27
+
Fas
+
),
T
CM
(CD3
+
CD4
+
CD45RO
+
CCR7
+
CD27
+
), T
TM
(CD3
+
CD4
+
CD45RO
+
CCR7
-
CD27
+
Fas
+
)
and T
EM
(CD3
+
CD4
+
CD45RO
+
CCR7
-
CD27
-
Fas
+
). CD4
+
T-cells were obtained from base-
line PBMCs and (CD4
+
-cell Isolation Kit, negative selection) (Miltenyi Biotec). At different
time points after, allo-HSCT plasma and PBMCs were obtained. Post-mortem biopsies were
obtained the day after death from terminal ileum, lung, liver, and spleen. From patient
IciS-11, also brain biopsies were obtained and CD4
+
-cells were isolated from fresh ileum
and a mediastinal lymph node.
2.2. DNA Isolation
Total DNA was isolated from frozen tissue biopsies using two different methods
because of size diffence of the biopsy size and the maximum input in the the DNeasy
Blood & Tissue kit (Qiagen, Hilden, Germany). Biopsy 1 was sliced into small pieces and
cells were lysed using the MagNALyser (Roche, Basel, Switzerland) and purified using
QIAquick (Qiagen, Hilden, Germany). Biopsy 2 was lysed and purified using the DNeasy
Blood & Tissue kit (Qiagen, Hilden, Germany). Total DNA from PBMCs or CD4
+
-cells was
isolated using the DNeasy Blood & Tissue kit.
2.3. Patient and Donor CCR5 Genotype Determination
To determine the CCR5 genotype, 30 ng total cellular DNA was amplified using
CCR5forward2 5
0
-GATAGGTACCTGGCTGTCGTCCAT-3
0
, and CCR5d 5
0
-CCTGTGCCTCT
TCTTCTCATTTCG-3
0
. As a control, plasmids containing the wild type CCR5 sequence or
the CCR5
32 sequence were also amplified and lengths of all amplified fragments were
compared by gel-electrophoresis and the Bioanalyzer (Agilent, Santa Clara, United States).
Deletions in CCR5 were confirmed by sequence analysis.
2.4. Ultra-Sensitive Viral Reservoir Quantification
Ultra-sensitive HIV-1 proviral DNA quantification was performed using primers in the
conserved HIV-LTR region and pol region and total cell DNA was quantified using an RPP30
(Ribonuclease P/MRP Subunit P30) primer and probe set as described in Bosman et al. [
13
].
HIV-1 DNA was quantified using de droplet digital PCR (ddPCR, Bio-Rad). The threshold
was set manually at a conservative fluorescence level. Water was used as a no-template
Viruses 2022,14, 2069 5 of 12
control and DNA isolated from PBMCs of HIV-negative donors was used as a DNA template
control. U1 cells were tested as a positive control. In 3/100 assays, a single positive droplet
was observed. Thus, measurement of a single droplet is interpreted as a trace.
2.5. Micro-Chimerism Assessment
In addition to routine clinical chimerism testing, ultra-sensitive chimerism PCR was
performed on peripheral blood (day +55, +106), post-mortem lymph node, and CD4
+
-
cells from the lymph node of patient IciS-11 using the Mentype DIPscreen and Mentype
DIPquant assays (Biotype), with a sensitivity of 0.01–0.001%, depending on quality and
quantity of the purified DNA.
2.6. Ultra-Sensitive Plasma HIV-1 RNA Determination
Ultra-sensitive VL was measured using Cobas Ampliprep/Cobas TaqMan HIV-1 Test
v2.0 (Roche Molecular Systems, Inc. Pleasanton, CA, USA) after ultracentrifugation of
4–9 mL of plasma [14].
2.7. HIV-1 Co-Receptor Prediction and Determination
Genotypic HIV-1 co-receptor tropism was predicted by deep sequence analysis of the
V3 loop of the viral envelope (gp160-V3). Total cellular DNA was amplified in triplicate in
a nested approach using primers envF1.1 5
0
-GGATATAATCAGYYTATGGGA-3
0
, envF1.2
5
0
-GAGGATATAATCAGTTTATGG
0
, envR1.1 5
0
-GGTGGGTGCTAYTCCYAITG-3
0
, envR1.2
5
0
-GGTGGGTGCTATTCCTAATGG-3
0
for the first amplification and primers envF2.1 5
0
-
GATCAAAGCCTAAARCCATGT-3
0
, envF2.2 5
0
-GATCAAAGCCTAAAGCCATG-3
0
, envR2.1
5
0
-CTCCAATTGTCCYTCATHTYTCC-3
0
, envR2.2 5
0
-ACTTCTCCAATTGTCCCTCATAT-3
0
in the nested amplification. The DNA concentration of the purified amplicons was deter-
mined using Quant-iT PicoGreen dsDNA Assay (Thermo Fisher Scientific, Waltham, MA,
USA) and a sequence library was formed using the Nextera DNA Library Preparation Kit
(Illumina, San Diego, CA, United States). Finally, the DNA concentration of the library was
measured (Quant-iT PicoGreen dsDNA Assay Kit, Thermo Fisher Scientific, MA, USA),
diluted to 2 ng/mm
3
denatured, and sequenced using the MiSeq Reagent Kit v2 (500-cycles)
(Illumina, San Diego, USA). Viral tropism was predicted in silico using geno2pheno (454)
with a 3.5% cut-off indicating the probability of classifying an R5-tropic virus falsely as
CXCR4-tropic virus (FPR; false-positive-rate) [15].
Phenotypic HIV-1 co-receptor was determined using cells and virus culture. Cells:
U373-MAGI-CCR5E, U373-MAGICXCR4CEM [
16
], MT2 cell lines [
17
] (NIH AIDS Reagent
Program), and SupT1R5X4 (personal gift from J. Hoxie) were maintained as recommended.
PBMCs from five healthy donors (homozygous for CCR5WT) were prepared by Ficoll–
Paque density gradient centrifugation of heparinized blood. The mix was stimulated
for
3 days
with phytohaemagglutinin (2 mg/L) in culture medium [RPMI1640 with L-
glutamine (BioWhittaker, Lonza, Basel, Switzerland), 10% fetal bovine serum (FBS; Biochrom
AG, Berlin, Germany) and 10 mg/L gentamicin (Gibco)]. Virus culture: PBMCs from
healthy donors were co-cultured with purified CD4
+
-cells from patients IciS-05 and IciS-11
for 2 h at 37
C, after which cells were washed twice. Subsequently, cells were cultured with
25 U/mL IL-2 in 10 mL CM. Virus was cultured for 3 weeks, and twice-weekly half of the
culture was replaced with fresh culture medium with 25 U/mL recombinant human IL-2
(Thermo Fisher Scientific, Massachusetts, United States) and once weekly freshly stimulated
PBMCs from healthy donors were added. Twice weekly HIV-1 replication was monitored
by CA-p24 ELISA and culture supernatant was stored for analyses of viral co-receptor
tropism in MT2 cells (expressing CD4 and CXCR4) and as a control SupT1R5
×
4 cells
(expressing CD4, CCR5 and CXCR4).
Analysis of HIV-1 co-receptor usage in U373-MAGI cells: Infection of these cells was
done using 2 ng of CA-p24 of the patient-derived viral isolate or the control viruses [6].
Phylogenetic sequence analysis; viral evolution and compartmentalization: HIV-1
subtype was determined using REGA HIV Subtyping Tool [
18
]. The evolutionary history
Viruses 2022,14, 2069 6 of 12
was inferred using Maximum Likelihood (MEGA 7) (500 bootstraps) [
19
]. The tree with
the highest log likelihood is shown. Initial tree (s) for the heuristic search were obtained
automatically by applying Neighbor-Join and BioNJ algorithms to a matrix of pairwise
distances estimated using the Maximum Composite Likelihood (MCL) approach, and then
selecting the topology with superior log likelihood value. The tree is drawn to scale, with
branch lengths measured in the number of substitutions per site. All positions containing
gaps and missing data were eliminated.
3. Results
3.1. Patient IciS-05
Twenty days before allo-HSCT, only an unquantifiably low amount of HIV-1 RNA could
be detected with routine diagnostics. Ultra-sensitive plasma viral load was
15 copies/mL
(Figure 1a). The viral reservoir was quantified in BM, PBMCs, naïve T-cells, and all memory
T-cell subsets with HIV-1-DNA being most abundant in the more differentiated T-cell subsets
(1135-6924 HIV-1-LTR copies/106cells) (Table 2).
Table 2. Results of HIV-1-DNA quantification and characterization in patient IciS-05.
Time
Point
(Days)
Chimerism
(%)
Ultrasensitive RNA
Quantification
(Copies/mL)
Patient Material
ddPCR
(Copies/106Cells)
Gp120V3-Sequence (Dominant
FPRs; Range, %)
HIV-1 LTR HIV-1
Pol
Pre allo-HSCT
20 15
PBMCs 1967 432 87.2, 89.7 (68.8–96.2)
Tn 1284 167 87.2, 89.7 (87.2–89.7)
Tcm 3074 609 87.2, 89.7 (87.2–91.0)
Ttm 5600 1622 87.2, 89.7 (68.0–89.7)
Tem 6924 1886 87.2, 89.7 (68.8–89.7)
19 BM 1135 167 87.2, 89.7(73.7–89.7)
Post allo-HSCT
+5 3 PBMCs 949 ND
+16 57 PBMCs 278 <21
+36 100 0 PBMCs <5 ND
+54 95 8 PBMCs 19 ND
+61 PBMCs <20 <20
+65 85
+68
(died) 8 PBMCs <7 <22
Post-mortem biopsies (one biopsy from same site is separated in two parts; 1,2)
+69
Liver, biopsy 1; 2 54; trace ND; <8 No amplification
Lung left, biopsy 1; 2 36; 49 ND; trace 89.7, 87.2 (87.2–89.7)
Lung right, biopsy 1; 2 90; 32 ND; trace 89.7, 87.2 (71.4–92.2)
Spleen, biopsy 1; 2 43; 67 ND; 34 No amplification
Terminal Ileum,
biopsy 1; 2 549; 81 ND; 89 89.7, 87.2 (64.4–89.7)
Allo-HSCT, allogeneic hematopoietic stem cell transplantation. BM, bone marrow. ddPCR, droplet digital PCR.
FPR, false-positive-rate. LTR, long terminal repeat DNA sequence. ND, not done. NTC, no template control.
PBMCs, peripheral blood mononuclear cells. pol, DNA polymerase DNA sequence. Tcm, central memory T-cell.
Tem, effector memory T-cell. Tn, naive T-cell. Ttm, transitional memory T-cell.
Viruses 2022,14, 2069 7 of 12
Viruses 2022, 14, 2069 7 of 13
Twenty days before allo-HSCT, only an unquantifiably low amount of HIV-1 RNA
could be detected with routine diagnostics. Ultra-sensitive plasma viral load was 15
copies/mL (Figure 1a). The viral reservoir was quantified in BM, PBMCs, naïve T-cells,
and all memory T-cell subsets with HIV-1-DNA being most abundant in the more
differentiated T-cell subsets (1135-6924 HIV-1-LTR copies/106 cells) (Table 2).
Figure 1. Clinical and virological data of IciS-05 (a) and IciS-11 (b) pre- and post-allo-HSCT. HIV-1
proviral DNA expressed as LTR DNA copies/million PBMC, and HIV-1 RNA as copies/mL for
plasma and chimerism. Open symbols represent values under the level of quantification.
Abbreviations: allo-HSCT, allogeneic hematopoietic stem cell transplantation. PBMCs, peripheral
blood mononuclear cells.
Table 2. Results of HIV-1-DNA quantification and characterization in patient IciS-05.
Time
Point
(Days)
Chimerism
(%)
Ultrasensitive
RNA
Quantification
(Copies/mL)
Patient
Material
ddPCR
(Copies/106 Cells)
Gp120V3-Sequence
(Dominant FPRs;
Range, %)
HIV-1 LTR HIV-1 pol
Pre allo-HSCT
20 15
PBMCs 1967 432 87.2, 89.7 (68.8–96.2)
Tn 1284 167 87.2, 89.7 (87.2–89.7)
Tcm 3074 609 87.2, 89.7 (87.2–91.0)
Ttm 5600 1622 87.2, 89.7 (68.0–89.7)
Tem 6924 1886 87.2, 89.7 (68.8–89.7)
19 BM 1135 167 87.2, 89.7(73.7–89.7)
Post allo-HSCT
+5 3 PBMCs 949 ND
+16 57 PBMCs 278 <21
+36 100 0 PBMCs <5 ND
+54 95 8 PBMCs 19 ND
+61 PBMCs <20 <20
+65 85
+68 (died) 8 PBMCs <7 <22
Post-mortem biopsies (one biopsy from same site is separated in two parts; 1,2)
+69
Liver,
biopsy 1; 2 54; trace ND; <8 No amplification
Lung left,
biopsy 1; 2 36; 49 ND; trace 89.7, 87.2 (87.2–89.7)
Lung right,
biopsy 1; 2 90; 32 ND; trace 89.7, 87.2 (71.4–92.2)
Spleen,
biopsy 1; 2 43; 67 ND; 34 No amplification
Figure 1.
Clinical and virological data of IciS-05 (
a
) and IciS-11 (
b
) pre- and post-allo-HSCT. HIV-1
proviral DNA expressed as LTR DNA copies/million PBMC, and HIV-1 RNA as copies/mL for
plasma and chimerism. Open symbols represent values under the level of quantification. Abbre-
viations: allo-HSCT, allogeneic hematopoietic stem cell transplantation. PBMCs, peripheral blood
mononuclear cells.
In-depth genotypic analyses of the viral reservoir showed that the population was
rather homogeneous with two viral variants dominating all cell types (Supplementary data
Figure S1a). Genotypic analyses predicted CCR5 co-receptor dependency for both variants
(FPR 87.2% and 89.7%) (Table 2). Viruses cultured from PBMCs also showed CCR5-tropism
and were unable to infect MAGI-X4 cells (Supplementary data Figure S1b) and MT2 cells.
After allo-HSCT with CCR5
32 donor cells, a decrease in proviral DNA was observed
in PBMCs (Figure 1a, Table 2). On day +36, when full chimerism was observed in PBLs,
proviral DNA in PBMCs and plasma VL were both below the limit of quantification.
Subsequently, a decrease in chimerism was observed in peripheral T-cells, and HIV-1-DNA
in PBMCs and plasma VL both became detectable on day +54. On day +68, when the
patient deceased, plasma VL was detectable (8 copies/mL) but proviral DNA in PBMCs
was still undetectable. Interestingly, at the same time, proviral DNA could be detected in
all post-mortem biopsies (Table 2). Viral envelope could be amplified and sequenced from
lung tissue and terminal ileum and revealed the dominance of the same CCR5-tropic viral
variants as observed in peripheral blood and BM prior to allo-HSCT (Supplementary data
Figure S1a).
3.2. Patient IciS-11
Shortly before allo-HSCT, low-level plasma HIV-1 RNA was detectable by ultrasen-
sitive viral load assay (2 copies/mL) (Figure 1b). HIV-1-DNA could be detected in BM,
PBMCs, naïve T-cells, and all memory T-cell subsets with most HIV-1-LTR DNA in the
differentiated subsets (73-4629 HIV-1-LTR copies/106 cells) (Table 3).
While proviral DNA levels were lower in comparison to IciS-05, in-depth genotypic
analyses of the viral envelope sequence demonstrated more inter- and intra-sample vari-
ation (Table 3, Supplementary data Figure S1a). All variants were predicted for the use
of the CCR5 co-receptor for viral entry (FPR 9.7–77.3%) (Supplementary data Figure S1a).
HIV-1 variants cultured from BM and PBMCs were also CCR5-tropic and unable to use the
alternative CXCR4 co-receptor as present in MAGI-X4 cells (Supplementary data Figure
S1b) and MT2 cells.
Shortly after allo-HSCT, plasma HIV-1 RNA could no longer be detected and also
PBMC proviral DNA rapidly declined (Table 3, Figure 1b). Over time, with decreasing
chimerism, proviral DNA increased to pre-transplantation levels and plasma ultrasensitive
VL became detectable (3 copies/mL). On day +55 when the graft was rejected, proviral
DNA in PBMCs was twice the pre-transplant level. Considering the relatively low cell
counts after allo-HSCT, a relatively big increase in proviral DNA was observed. In-depth
sequence analyses revealed the same viral sequences as seen before transplantation, and
thus no signs of viral evolution or escape of CCR5 co-receptor usage were observed.
Viruses 2022,14, 2069 8 of 12
Table 3. Results of HIV-1-DNA quantification and characterization in patient IciS-11.
Time
Point
(Days)
Chimerism
(%)
Ultrasensitive RNA
Quantification
(Copies/mL)
Patient Material ddPCR (Copies/106Cells) Gp120V3-Sequence
(Dominant FPR; Range, %)
HIV-1 LTR HIV-1 Pol
Pre allo-HSCT
22 PBMCs 279 21 18.0 (18.0–70.5)
20 BM 73 <14 70.5 (39.4–70.5)
14 2 Tn 571 69 31.8 (18.0–70.5)
Tscm 490 ND 9.7 (9.7–50.3)
Tcm 2222 544 42.4 (22.1–77.3)
Ttm 2780 838 42.4 (18.0–70.5)
Tem 4629 882 21.8 (18.0–39.4)
Post allo-HSCT
+5 0 PBMCs trace <5
+27 60 3 PBMCs 378 ND
+55 0 2 PBMCs 534 72 31.8 (31.8–70.5)
Post 2nd allo-HSCT (days post 1st allo-HSCT)
+11
(+82) 0 PBMCs 8 ND
+27
(+98) 0 PBMCs <2 <4
+35
(+106) 100 0 PBMCs <2 ND
Post-mortem biopsies (one biopsy from same site is separated in two parts; 1,2)
+37
(+108) Liver, biopsy 1; 2 60; <7 ND; trace No amplification
Lung left, biopsy 1; 2 28; <4 ND; <4 No amplification
Lung right, biopsy 1; 2 trace; <3 ND; <3 No amplification
Spleen, biopsy 1; 2 60; <6 ND; <6 No amplification
Brain, biopsy 1; 2 <4; <14 <4; <4 No amplification
38 LN CD4+cells 10 <4 No amplification
Allo-HSCT, allogeneic hematopoietic stem cell transplantation. BM, bone marrow. ddPCR, droplet digital PCR.
FPR, false-positive-rate. LN, lymph node. LTR, long terminal repeat DNA sequence ND, not done. NTC, no
template control. PBMCs, peripheral blood mononuclear cells. pol, DNA polymerase DNA sequence. Tcm,
central memory T-cell. Tem, effector memory T-cell. Tn, naive T-cell. Tscm, stem memory T-cell. Ttm, transitional
memory T-cell.
On day +71, IciS-11 received a second allo-HSCT with heterozygous CCR5
32/WT
donor cells. In the first week after transplantation, a rapid decrease of proviral DNA was
observed. On day +35, when chimerism was 100%, PBMC proviral DNA and
plasma VL
were undetectable. Two days later the patient died of respiratory failure. Multiple post-
mortem biopsies were obtained from liver, lung, spleen, brain, and lymph node. In contrast
to patient IciS-05, detection of HIV-1-DNA in these tissues varied between the different
tissues and biopsies, with lung and brain being negative and other tissues being positive in
only one of the biopsies. The viral envelope could not be successfully amplified from any
of the biopsies (Table 3).
4. Discussion
To date, allo-HSCT is the only strategy to profoundly reduce the HIV-1 reservoir
irrespective of whether CCR5 wild-type stem cells or stem cells lacking expression of the
CCR5 co-receptor for HIV-1 entry are used [25,79,11,2022].
Autopsy of patients IciS-05 and IciS-11 provided a unique opportunity to investigate
the impact of allo-HSCT on the dynamics and composition of the viral reservoir in PBMCs
as well as in different tissue compartments. In patient IciS-05 who was transplanted in a
non-myeloablative procedure with CCR5
32/
32 donor cells, no HIV-1-DNA could be
detected by ddPCR in PBMCs but HIV-1-DNA could readily be detected in all biopsies
taken from lung, liver, spleen, and ileum. A study describing post-mortem HIV-1 reservoir
Viruses 2022,14, 2069 9 of 12
analysis in a perinatally infected child who also underwent a myeloablative procedure
using cord blood CCR5
32 donor cells could not detect HIV-1-DNA in blood but could
detect viral DNA in some of the tissues analyzed using sensitive in-situ hybridization
techniques [
21
]. The sensitivity of the ddPCR as employed in the study of Rothenberger
et al. was likely affected by the smaller number of cells that could be assessed from the
tissue biopsies as compared to our study.
In patient IciS-11, who was also transplanted with CCR5
32 donor cells, an initial
decrease of proviral DNA in PBMCs was followed by an increase in proviral DNA perPBMC.
Eberhard et al. showed an HIV-1-specific T-cell activation in IciStem patients after allo-
HSCT and the risk of viral escape from the HIV-1 reservoir shortly after transplantation [
23
].
In-depth sequence analyses of the viral envelope gene revealed identical viral sequences as
seen before transplantation of patient IciS-11 and as such showed no signs of viral evolution
or escape of CCR5 co-receptor usage. As patient IciS-11 experienced cytomegalovirus
(CMV) reactivation after allo-HSCT during incomplete chimerism, we hypothesize that the
increase of proviral DNA could be due to the expansion of particular T-cell populations
after allo-HSCT. This observation is in line with the described expansion of HIV-1-infected
CD4+ T-cells in response to CMV or Epstein–Barr viral antigens, contributing to sustain the
HIV-1-DNA reservoir following chemotherapy [12].
Patient IciS-11 did not suffer from GvHD and, like the Berlin patient, was transplanted
twice. HIV-1-DNA copy number in the tissues was at the detection level of our ddPCR
assays with discrepancies between different tissue biopsies and target regions. These
discrepancies show the difficulties analyzing proviral DNA in high-volume post-mortem
tissues and emphasize the challenges that we face in the accurate detection and quantifica-
tion of proviral DNA in low-volume biopsies of patients undergoing curative strategies.
Though with a successful transplantation full chimerism in the peripheral blood is reached
in the weeks after allo-HSCT, little is known about the turnover and donor chimerism in
tissues. In the Berlin patient, CCR5-positive macrophages were still detected in rectal tissue
more than 5 months after allo-HSCT [
4
]. In patient IciS-11, who died 1 month after his
second transplant, chimerism in PBLs was 100% and no HIV-1-DNA was detected in blood.
Simultaneously, in CD4
+
-cells obtained from the lymph node, where donor chimerism only
reached 38%, HIV-1-DNA was detected, most likely in the recipient cells.
In patient IciS-05, proviral DNA from the different tissue samples was genotypically
characterized and phylogenetic analysis indicated that all tissue-derived sequences in-
termingled with pre-transplantation viral populations as present in the different T-cell
subsets and BM. These data show no evident compartmentalization between viruses ob-
tained from peripheral blood and BM before the transplant compared to viruses obtained
from lung and ileum afterwards. Unlike in the Berlin patient, sequences obtained from
the different post-mortem tissues after allo-HSCT showed no evidence of persistence of
macrophage-tropic viral variants. In general, our data are in agreement with post-mortem
studies demonstrating the lack of compartmentalization of HIV-1 envelope sequences
observed in lung, lymph nodes, and colon [
24
,
25
]. Previous ATI studies performed after
allo-HSCT suggested that the observed viral rebound was caused by pre-existing viral
variants present in the pre-transplant PBLs [
12
,
26
,
27
]. However, these patients’ tissues
were not characterized. Since no HIV-1-DNA-positive cells could be found in blood, the
rebounding viruses post allo-SCT most likely originated from tissue compartments.
A limitation of this study that should be addressed is the short follow-up of the
patients after allo-HSCT. Both patients died shortly after allo-HSCT, hampering the follow-
up of the dynamics of the donor cells in different tissues. Also because sampling occurred
shortly after allo-HSCT, only limited cells were available due to the allo-HSCT procedure.
Analyses of the proviral DNA could only be performed on PBMCs and not on a more
concentrated CD4+ T-cell subset, which could have underestimated the true size of the
peripheral reservoir after allo-HSCT.
In conclusion: This is the first report that performed in-depth post-mortem tissue
quantification and genotypic characterization after allo-HSCT with CCR5
32/
32 donor
Viruses 2022,14, 2069 10 of 12
cells of HIV-1-infected individuals. We demonstrate that the detection of proviral DNA
in tissues was prolonged compared to PBMCs. Tissues clearly play an essential role as
a long-standing viral reservoir and routine sampling in living HIV-1-individuals will be
insufficient to represent the extent of this reservoir. Before conducting ATI, the role of the
tissue reservoir has to be taken into consideration.
Supplementary Materials:
The following supporting information can be downloaded at: https:
//www.mdpi.com/article/10.3390/v14092069/s1, Figure S1: Phylogenetic analysis and relative
coreceptor preference.
Author Contributions:
Conceptualization, A.M.J.W., M.N., J.M.-P.; A.M.J.W., and A.S. were involved
in the ethical approval. A.B., P.E., J.H.E.K. and J.T.M.v.d.M. were involved in the clinical process
and provided clinical knowledge. L.A.A.B. performed the autopsy. D.d.J., N.A.N., S.B.H.G., K.T.,
K.B., M.S. and T.M.d.K. generated and analyzed the data. L.E.P.H., A.M.J.W., M.N. and N.A.N. were
involved in draft preparation. L.E.P.H. editing the manuscript. M.S., G.H., M.K., J.D.M., J.T.M.v.d.M.,
A.S.-C., J.J.B., J.S.z.W., J.M.-P., J.H.E.K., A.M.J.W. and M.N. provided scientific guidance and domain
knowledge. All authors have read and agreed to the published version of the manuscript.
Funding:
This work was supported by amfAR, the Foundation for AIDS Research through the
ARCHE program [108930-56-RGRL, 109293-59-RGRL, 109552-61-RGRL], and by the Dutch Aidsfonds
[P-2013034, P-60802, P-13204].
Institutional Review Board Statement:
The study was conducted in accordance with the Decla-
ration of Helsinki, and approved by the Institutional Medical Ethics Committee of the UMCU
(NL35448.041.11 on 27 November 2012 for IciS-05 and NL53114.041.15. on 14 April 2015 for IciS-11).
Informed Consent Statement:
IciS-05 was included after signing a general waiver for the use of
body material for future research, according to the regulations of the UMC Utrecht HSCT Biobank
(HSCT study number 3440). IciS-11 was included after written informed consent was acquired.
Data Availability Statement:
The authors confirm that the data supporting the findings of this study
are available within the article. Further inquiries can be directed to the corresponding author.
Acknowledgments:
The authors thank the following members of the IciStem Consortium who were
not named as authors: Vanderson Rocha (Churchill Hospital, Oxford, United Kingdom); JuliàBlanco
and Jorge Carillo (IrsiCaixa AIDS Research Institute and Institute for Health Science Research Ger-
mans Trias i Pujol (IGTP), Badalona, Spain); Johanna Eberhard and Maximilian Christopeit (University
Medical Center Hamburg-Eppendorf, Hamburg, Germany); Rebeca Bailén and Pascual Balsalobre
(Hospital Gregorio Marañón, Madrid, Spain); Linos Vandekerckhove Marie-Angélique de Scheerder,
and Eva Steel (University of Ghent, Ghent, Belgium); Lisa Barrett, Sharon Oldford, Jill Moore, and
Clarissa Brisseau (NSHA/Dalhousie University, Halifax, Canada); Dieter Häussinger, Guido Kobbe,
and Björn Jensen (University Hospital Düsseldorf, Düsseldorf, Germany); Rolf Kaise and, Elena
Knops (University of Cologne, Cologne, Germany); Marek Widera (University Hospital Essen, Essen,
Germany); Alessandra Bandera, Antonio Muscatello, and Alessandro Soria (San Gerardo Hospital,
Monza, Italy); Gabriella Scarlatti and Simona Piemontese (Istituto Scientifico San Raffaele, Milan,
Italy); Jon Badiola and Manuel Jurado Chacón (Complejo Hospitalario Universitario de Granada,
Granda, Spain); Raquel Saldana (Hospital General de Jerez de la Frontera, Jerez de la Frontera,
Spain); Luz Martín Carbonero (La Paz Hospital, Madrid, Spain); Ildefonso Espigado (University
Hospital Virgen del Rocío, Seville, Spain); Piotr Nowak and Anders Sonnerborg (Karolinska Institutet,
Solna, Sweden); Mitja Nabergoj and Alexandra Calmy (Hôpitaux Universitaires de Genève, Genève,
Switzerland); Kavita Raj, Fabio Cruciani, Varun Mehra, and Carmel Rice (Kings College Hospital,
London, United Kingdom); Angela Bailey (Imperial College Healthcare NHS Trust, London, United
Kingdom); Waseem Qasim (Institute of Child Health & Great Ormond Street Hospital, London,
United Kingdom); Ravindra Gupta (University College London Hospital, London, United Kingdom);
and Koen van Besien (New York Presbyterian Hospital, New York, United States). The authors also
thank Agueda Hernández Rodríguez, from the Microbiology Department of the Hospital Universitari
Germans Trias i Pujol.
Conflicts of Interest:
All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts
of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been
disclosed. AJS resports a grant form Dutch Government (ZonMw) and is a committee member for
Viruses 2022,14, 2069 11 of 12
revision Dutch guideline on Sexually Transmitted Diseases in primary care. JJB reports consultancy
fees for the following companies: Avrobio, Omeros, Advanced Clinical, Bluebird Bio, Equillium, Sobi,
and SmartImmune. MS reports grants or contracts from Spanisch Ministry of research and private TV
fundraising funding (MaratóTV3) and payments or honoraria for lectures, presentations, speakers
bureaus, manuscript writing, or educational events from Autonomous University of Barcelona and
GESIDA Spain. ASC report grants or contracts from ANRS, NIH, and MSDAVENIR; payments from
MSD, ViiV healthcare, and Gilead, and a patent for a method for reprogramming CD8+ T-cells to
enhance their therapeutic potential and applications thereof; U.S. Patent application # 63/301, 532
and chair of the Sidaction Scientific and Medical Committee. JHEK reports grants or contracts from
Novartis, Miltenyi, and Gadeta. MN reports grants from Gilead: Rosetta (Global INI resistance);
Dutch Government (ZonMw/NWO): VIMP2, Bridge project; COVIDex-vivo models, Health Holland:
Clear COVID organoids2cureHIV, amfAR: Epigenetic engineering of HIV, NSF: SARS-CoV-2 mutation
frequency and evolution and honoraria for lectures from Virology Education. AW reports grants
from Gilead, Aidfsonds, Dutch Government (ZonMw/NOW), and Health Holland/NLF4 cure, as
well as consulting fees from Gilead, Janssen, Viiv/GSK, and honoraria for lectures from Virology
Education and Southern African HIV Clinicians Society.
References
1.
Deeks, S.G.; Archin, N.; Cannon, P.; Collins, S.; Jones, R.B.; de Jong, M.A.W.P.; Lambotte, O.; Lamplough, R.; Ndung’u, T.;
Sugarman, J.; et al. Research priorities for an HIV cure: International AIDS Society Global Scientific Strategy 2021. Nat. Med.
2021
,
12, 2085–2098. [CrossRef] [PubMed]
2.
Gupta, R.K.; Abdul-Jawad, S.; McCoy, L.E.; Mok, H.P.; Peppa, D.; Salgado, M.; Martinez-Picado, J.; Nijhuis, M.; Wensing, A.M.J.;
Lee, H.; et al. HIV-1 remission following CCR5
32/
32 haematopoietic stem-cell transplantation. Nature
2019
,7751, 244–248.
[CrossRef] [PubMed]
3.
Gupta, R.K.; Peppa, D.; Hill, A.L.; Gálvez, C.; Salgado, M.; Pace, M.; McCoy, L.E.; Griffith, S.A.; Thornhill, J.; Alrubayyi, A.;
et al. Evidence for HIV-1 cure after CCR5
32/
32 allogeneic haemopoietic stem-cell transplantation 30 months post analytical
treatment interruption: A case report. Lancet HIV 2020,7, e340–e347. [CrossRef]
4.
Hütter, G.; Nowak, D.; Mossner, M.; Ganepola, S.; Müssig, A.; Allers, K.; Schneider, T.; Hofmann, J.; Kücherer, C.; Blau, O.; et al.
Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N. Engl. J. Med.
2009
,360, 692–698. [CrossRef]
[PubMed]
5.
Jensen, B.; Haussinger, D.; Knops, E. CCR5
32 SCT-induced HIV remission: Traces of HIV-DNA but fading immune reactivity
[abstract 348]. Presented at The Annual Conference on Retroviruses and Opportunistic Infections 2020, Boston, MA, USA, 8–11
March 2020.
6.
Symons, J.; Vandekerckhove, L.; Hütter, G.; Wensing, A.M.; van Ham, P.M.; Deeks, S.G.; Nijhuis, M. Dependence on the CCR5
coreceptor for viral replication explains the lack of rebound of CXCR4-predicted HIV variants in the Berlin patient. Clin. Infect.
Dis. 2014,59, 596–600. [CrossRef] [PubMed]
7.
Hsu, J.; Van Besien, K.; Glesby, M.J. HIV-1 remission with CCR5
32
32 haplo-cord transplant in a US woman: IMPAACT P1107
[abstract 65]. In Proceedings of the Conference on Retroviruses and Opportunistic Infections (CROI), Virtual, 12–24 February
2022.
8.
Yukl, S.A.; Boritz, E.; Busch, M.; Bentsen, C.; Chun, T.W.; Douek, D.; Eisele, E.; Haase, A.; Ho, Y.C.; Hütter, G.; et al. Challenges
in Detecting HIV Persistence during Potentially Curative Interventions: A Study of the Berlin Patient. PLoS Pathog.
2013
,9,
e1003347. [CrossRef] [PubMed]
9.
Avettand-Fenoel, V.; Mahlaoui, N.; Chaix, M.L.; Milliancourt, C.; Burgard, M.; Cavazzana-Calvo, M.; Rouzioux, C. Failure of bone
marrow transplantation to eradicate HIV reservoir despite efficient HAART. AIDS 2007,6, 776–777. [CrossRef] [PubMed]
10.
Capoferri, A.A.; Redd, A.D.; Gocke, C.D.; Clark, L.R.; Quinn, T.C.; Ambinder, R.F.; Durand, C.M. Rebound HIV viremia with
meningoencephalitis following antiretroviral therapy interruption after allogeneic bone marrow transplant. J. Acquir. Immune
Defic. Syndr. 2021,89, 297–302. [CrossRef]
11.
Cummins, N.W.; Rizza, S.; Litzow, M.R.; Hua, S.; Lee, G.Q.; Einkauf, K.; Chun, T.W.; Rhame, F.; Baker, J.V.; Busch, M.P.; et al.
Extensive virologic and immunologic characterization in an HIV-infected individual following allogeneic stem cell transplant
and analytic cessation of antiretroviral therapy: A case study. PLoS Med. 2017,14, e1002461. [CrossRef]
12.
Henrich, T.J.; Hanhauser, E.; Marty, F.M.; Sirignano, M.N.; Keating, S.; Lee, T.H.; Robles, Y.P.; Davis, B.T.; Li, J.Z.; Heisey, A.; et al.
Antiretroviral-free HIV-1 remission and viral rebound after allogeneic stem cell transplantation: Report of 2 cases. Ann. Intern
Med. 2014,161, 319–327. [CrossRef]
13.
Bosman, K.J.; Wensing, A.M.; Pijning, A.E.; van Snippenberg, W.J.; van Ham, P.M.; de Jong, D.M.; Hoepelman, A.I.; Nijhuis, M.
Development of sensitive ddPCR assays to reliably quantify the proviral DNA reservoir in all common circulating HIV subtypes
and recombinant forms. J. Int. AIDS Soc. 2018,21, e25185. [CrossRef] [PubMed]
14.
Salgado, M.; Kwon, M.; Gálvez, C.; Badiola, J.; Nijhuis, M.; Bandera, A.; Balsalobre, P.; Miralles, P.; Buño, I.; Martinez-Laperche,
C.; et al. Mechanisms That Contribute to a Profound Reduction of the HIV-1 Reservoir After Allogeneic Stem Cell Transplant.
Ann. Intern Med. 2018,169, 674–683. [CrossRef] [PubMed]
Viruses 2022,14, 2069 12 of 12
15.
Lengauer, T.; Sander, O.; Sierra, S.; Thielen, A.; Kaiser, R. Bioinformatics prediction of HIV coreceptor usage. Nat. Biotechnol.
2007
,
12, 1407–1410. [CrossRef]
16.
Pineda-Peña, A.C.P.; Faria, N.R.; Imbrechts, S.; Libin, P.; Abecasis, A.B.; Deforche, K.; Gómez-López, A.; Camacho, R.J.; de Oliveira,
T.; Vandamme, A.M. Automated subtyping of HIV-1 genetic sequences for clinical and surveillance purposes: Performance
evaluation of the new REGA version 3 and seven other tools. Infect Genet. Evol. 2013,19, 337–348. [CrossRef] [PubMed]
17.
Kumar, S.; Stecher, G.; Tamura, K. MEGA7: Molecular Evolutionary Genetics Analysis Version 7.0 for Bigger Datasets. Mol. Biol.
Evol. 2016,7, 1870–1874. [CrossRef] [PubMed]
18.
Vodicka, M.A.; Goh, W.C.; Wu, L.I.; Rogel, M.E.; Bartz, S.R.; Schweickart, V.L.; Raport, C.J.; Emerman, M. Indicator cell lines for
detection of primary strains of human and simian immunodeficiency viruses. Virology
1997
,233, 193–198. [CrossRef] [PubMed]
19.
Harada, S.; Koyanag, Y.; Yamamoto, N. Infection of HTLV-III/LAV in HTLV-I-carrying cells MT-2 and MT-4 and application in a
plaque assay. Science 1985,713, 563–566. [CrossRef]
20.
Duarte, R.F.; Salgado, M.; Sánchez-Ortega, I.; Arnan, M.; Canals, C.; Domingo-Domenech, E.; Fernández-de-Sevilla, A.; González-
Barca, E.; Morón-López, S.; Nogues, N.; et al. CCR5
32 homozygous cord blood allogeneic transplantation in a patient with hiv:
A case report. Lancet HIV 2015,2, e236–e242. [CrossRef]
21.
Rothenberger, M.K.; Wagner, J.E.; Haase, A.; Richman, D.; Grzywacz, B.; Strain, M.; Lada, S.; Estes, J.; Fletcher, C.V.; Podany, A.T.;
et al. Transplantation of CCR5
32 Homozygous Umbilical Cord Blood in a Child With Acute Lymphoblastic Leukemia and
Perinatally Acquired HIV Infection. Open Forum Infect. Dis. 2018,5, ofy090. [CrossRef]
22.
Koelsch, K.K.; Rasmussen, T.A.; Hey-Nguyen, W.J.; Pearson, C.; Xu, Y.; Bailey, M.; Marks, K.H.; Sasson, S.C.; Taylor, M.S.; Tantau,
R.; et al. Impact of Allogeneic Hematopoietic Stem Cell Transplantation on the HIV Reservoir and Immune Response in 3
HIV-Infected Individuals. J. Acquir. Immune Defic. Syndr. 2017,75, 328–337. [CrossRef]
23.
Eberhard, J.M.; Angin, M.; Passaes, C.; Salgado, M.; Monceaux, V.; Knops, E.; Kobbe, G.; Jensen, B.; Christopeit, M.; Kröger, N.;
et al. Vulnerability to reservoir reseeding due to high immune activation after allogeneic hematopoietic stem cell transplantation
in individuals with HIV-1. Sci. Transl. Med. 2020,12, eaay9355. [CrossRef] [PubMed]
24.
Brese, R.L.; Gonzalez-Perez, M.P.; Koch, M.; O’Connell, O.; Luzuriaga, K.; Somasundaran, M.; Clapham, P.R.; Dollar, J.J.; Nolan,
D.J.; Rose, R.; et al. Ultradeep single-molecule real-time sequencing of HIV envelope reveals complete compartmentalization of
highly macrophage-tropic R5 proviral variants in brain and CXCR4-using variants in immune and peripheral tissues. J. Neurovirol.
2018,24, 439–453. [CrossRef]
25.
Chaillon, A.; Gianella, S.; Dellicour, S.; Rawlings, S.A.; Schlub, T.E.; De Oliveira, M.F.; Ignacio, C.; Porrachia, M.; Vrancken, B.;
Smith, D.M. HIV persists throughout deep tissues with repopulation from multiple anatomical sources. J. Clin. Investig.
2020
,130,
1699–1712. [CrossRef] [PubMed]
26.
Kordelas, L.; Verheyen, J.; Beelen, D.W.; Horn, P.A.; Heinold, A.; Kaiser, R.; Trenschel, R.; Schadendorf, D.; Dittmer, U.; Esser,
S.; et al. Shift of HIV tropism in stem-cell transplantation with CCR5 Delta32 mutation. N. Engl. J. Med.
2014
,371, 880–882.
[CrossRef] [PubMed]
27.
Verheyen, J.; Thielen, A.; Lübke, N.; Dirks, M.; Widera, M.; Dittmer, U.; Kordelas, L.; Däumer, M.; de Jong, D.C.M.; Wensing, A.M.J.;
et al. Rapid Rebound of a Preexisting CXCR4-tropic Human Immunodeficiency Virus Variant After Allogeneic Transplantation
With CCR5 32 Homozygous Stem Cells. Clin. Infect. Dis. 2018,68, 684–687. [CrossRef] [PubMed]
... But in July 2014, HIV was detected in her body again [40]. Thus, an observation period of at least 30 months after stopping ART is a convincing time point for determining a functional cure; research indicates that after allo-HSCT, the virus is cleared in a tissue-specific and step-by-step manner [73,74]. HIV and the infected cells are first cleared from the peripheral blood, then from the peripheral lymph nodes, and finally from the mesenteric lymph nodes that drain the gastrointestinal tract. ...
Article
Full-text available
Antiretroviral therapy (ART) can effectively suppress the replication of human immunodeficiency virus (HIV), but it cannot completely eradicate the virus. The persistent existence of the HIV reservoir is a major obstacle in the quest for a cure. To date, there have been a total of seven cured cases of HIV worldwide. These patients all cleared HIV while undergoing allogeneic stem cell transplantation (allo-HSCT) for hematological malignancies. However, in these cases, the specific mechanism by which allo-HSCT leads to the eradication of HIV remains unclear, so it is necessary to conduct an in-depth analysis. Due to the difficulty in obtaining donors and the risks associated with transplantation, this treatment method is not applicable to all HIV patients. There is still a need to explore new treatment strategies. In recent years, emerging therapies such as neutralizing antibody immunotherapy, chimeric antigen receptor T cell (CAR-T) therapy, gene editing, and antiviral therapies targeting the reservoir have attracted wide attention due to their ability to effectively inhibit HIV replication. This article first elaborates on the nature of the HIV reservoir, then deeply explores the treatment modalities and potential success factors of HIV cured cases, and finally discusses the current novel treatment methods, hoping to provide comprehensive and feasible strategies for achieving the cure of HIV.
... The intact proviral DNA assay (IPDA) 23 detected potentially intact proviruses in two samples that had been obtained during ART-suppressed viremia 17 and 32 months before allo-HSCT in the context of his participation in the Swiss HIV cohort study (Fig. 2c). By contrast, potentially intact proviruses were never detected following people with HIV who did not achieve full donor chimerism 12,17 or in tissue sanctuaries analyzed in necropsy studies 18 . Moreover, during the weeks following allo-HSCT, a window of vulnerability occurs when highly activated CD4 + T cells from both donor and recipient coexist 19 , thereby increasing the risk of reservoir reseeding if infection of donor cells is not prevented by pharmacological or genetic and host barriers. ...
Article
Full-text available
HIV cure has been reported for five individuals who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT) with cells from CCR5Δ32 homozygous donors. By contrast, viral rebound has occurred in other people living with HIV who interrupted antiretroviral treatment after undergoing allo-HSCT, with cells mostly from wild-type CCR5 donors. Here we report the case of a male individual who has achieved durable HIV remission following allo-HSCT with cells from an unrelated HLA-matched (9 of 10 matching for HLA-A, HLA-B, HLA-C, HLA-DRB1 and HLA-DQB1 alleles) wild-type CCR5 donor to treat an extramedullary myeloid tumor. To date, plasma viral load has remained undetectable for 32 months after the interruption of antiretroviral treatment. Treatment with ruxolitinib has been maintained during this period to treat chronic graft-versus-host disease. Low levels of proviral DNA were detected sporadically after allo-HSCT, including defective but not intact HIV DNA. No virus could be amplified in cultures of CD4⁺ T cells obtained after antiretroviral treatment interruption, while CD4⁺ T cells remained susceptible to HIV-1 infection in vitro. Declines in HIV antibodies and undetectable HIV-specific T cell responses further corroborate the absence of viral rebound after antiretroviral treatment interruption. These results suggest that HIV remission could be achieved in the context of allo-HSCT with wild-type CCR5.
Article
Purpose of review Long-lasting HIV remission has been reported in a small group of people with HIV (PWH) following allogenic hematopoietic stem cell transplants (HSCT) for the treatment of hematologic malignancies. While the mechanisms of HIV remission following release from antiretroviral therapy (ART) were not initially known, subsequent findings from clinical cases and preclinical nonhuman primate studies have implicated mechanisms of clearance. Here, we review the six currently published human cases of long-term ART-free HIV remission. Recent findings Since the first report of ART-free HIV remission following HSCT, five subsequent cases of HSCT-induced sustained HIV remission have been published. While the pre- and posttransplant treatment conditions vary greatly, all but one received cells from donors homozygous for a 32 bp deletion in the gene that encodes CCR5 ( ccr5Δ32 ), the major HIV coreceptor. Studies in nonhuman primates and the newest published individual suggest that while CCR5 deficiency can protect donor cells from infection early posttransplant, it is not required for long term remission, as ablation of the viral reservoir is likely due to allogeneic immunity mediating a graft-versus-reservoir response. Summary Studies of HSCT in PLWH and simian immunodeficiency virus (SIV)-infected monkeys show that those with durable remission are likely cured, demonstrated by complete ablation of the replication-competent HIV reservoir, gradual loss of anti-HIV immunity, and greater than 5 years of aviremia.
Article
Full-text available
Erratum to: Progress Note 2024: Curing HIV; Not in My Lifetime or Just Around the Corner? doi: 10.20411/pai.v8i2.665 In the original publication, the comments provided by Santiago Ávila-Ríos were mistakenly omitted. In this version, his comments are included in the “Comments by Leaders” section, and his name has been included in the list of authors. ---Once a death sentence, HIV is now considered a manageable chronic disease due to the development of antiretroviral therapy (ART) regimens with minimal toxicity and a high barrier for genetic resistance. While highly effective in arresting AIDS progression and rendering the virus untransmissible in people living with HIV (PLWH) with undetectable viremia (U=U) [1, 2]), ART alone is incapable of eradicating the “reservoir” of resting, latently infected CD4+ T cells from which virus recrudesces upon treatment cessation. As of 2022 estimates, there are 39 million PLWH, of whom 86% are aware of their status and 76% are receiving ART [3]. As of 2017, ART-treated PLWH exhibit near normalized life expectancies without adjustment for socioeconomic differences [4]. Furthermore, there is a global deceleration in the rate of new infections [3] driven by expanded access to pre-exposure prophylaxis (PrEP), HIV testing in vulnerable populations, and by ART treatment [5]. Therefore, despite outstanding issues pertaining to cost and access in developing countries, there is strong enthusiasm that aggressive testing, treatment, and effective viral suppression may be able to halt the ongoing HIV epidemic (ie, UNAIDS’ 95-95-95 targets) [6–8]; especially as evidenced by recent encouraging observations in Sydney [9]. Despite these promising efforts to limit further viral transmission, for PLWH, a “cure” remains elusive; whether it be to completely eradicate the viral reservoir (ie, cure) or to induce long-term viral remission in the absence of ART (ie, control; Figure 1). In a previous salon hosted by Pathogens and Immunity in 2016 [10], some researchers were optimistic that a cure was a feasible, scalable goal, albeit with no clear consensus on the best route. So, how are these cure strategies panning out? In this commentary, 8 years later, we will provide a brief overview on recent advances and failures towards identifying determinants of viral persistence and developing a scalable cure for HIV. Based on these observations, and as in the earlier salon, we have asked several prominent HIV cure researchers for their perspectives.
Article
Full-text available
Once a death sentence, HIV is now considered a manageable chronic disease due to the development of antiretroviral therapy (ART) regimens with minimal toxicity and a high barrier for genetic resistance. While highly effective in arresting AIDS progression and rendering the virus untransmissible in people living with HIV (PLWH) with undetectable viremia (U=U) [1, 2]), ART alone is incapable of eradicating the “reservoir” of resting, latently infected CD4+ T cells from which virus recrudesces upon treatment cessation. As of 2022 estimates, there are 39 million PLWH, of whom 86% are aware of their status and 76% are receiving ART [3]. As of 2017, ART-treated PLWH exhibit near normalized life expectancies without adjustment for socioeconomic differences [4]. Furthermore, there is a global deceleration in the rate of new infections [3] driven by expanded access to pre-exposure prophylaxis (PrEP), HIV testing in vulnerable populations, and by ART treatment [5]. Therefore, despite outstanding issues pertaining to cost and access in developing countries, there is strong enthusiasm that aggressive testing, treatment, and effective viral suppression may be able to halt the ongoing HIV epidemic (ie, UNAIDS’ 95-95-95 targets) [6–8]; especially as evidenced by recent encouraging observations in Sydney [9]. Despite these promising efforts to limit further viral transmission, for PLWH, a “cure” remains elusive; whether it be to completely eradicate the viral reservoir (ie, cure) or to induce long-term viral remission in the absence of ART (ie, control; Figure 1). In a previous salon hosted by Pathogens and Immunity in 2016 [10], some researchers were optimistic that a cure was a feasible, scalable goal, albeit with no clear consensus on the best route. So, how are these cure strategies panning out? In this commentary, 8 years later, we will provide a brief overview on recent advances and failures towards identifying determinants of viral persistence and developing a scalable cure for HIV. Based on these observations, and as in the earlier salon, we have asked several prominent HIV cure researchers for their perspectives.
Article
Allogeneic hematopoietic stem cell transplantation (alloHSCT) from donors lacking C-C chemokine receptor 5 (CCR5Δ32/Δ32) can cure HIV, yet mechanisms remain speculative. To define how alloHSCT mediates HIV cure, we performed MHC-matched alloHSCT in SIV+, anti-retroviral therapy (ART)-suppressed Mauritian cynomolgus macaques (MCMs) and demonstrated that allogeneic immunity was the major driver of reservoir clearance, occurring first in peripheral blood, then peripheral lymph nodes, and finally in mesenteric lymph nodes draining the gastrointestinal tract. While allogeneic immunity could extirpate the latent viral reservoir and did so in two alloHSCT-recipient MCMs that remained aviremic >2.5 years after stopping ART, in other cases, it was insufficient without protection of engrafting cells afforded by CCR5-deficiency, as CCR5-tropic virus spread to donor CD4+ T cells despite full ART suppression. These data demonstrate the individual contributions of allogeneic immunity and CCR5 deficiency to HIV cure and support defining targets of alloimmunity for curative strategies independent of HSCT.
Article
Acquired immunodeficiency syndrome (AIDS), caused by the human immunodeficiency virus (HIV), has become a heavy burden of disease and an important public health problem in the world. Although current antiretroviral therapy (ART) is effective at suppressing the virus in the blood, HIV still remains in two different types of reservoirs-the latently infected cells (represented by CD4+ T cells) and the tissues containing those cells, which may block access to ART, HIV-neutralizing antibodies and latency-reversing agents. The latter is the focus of our review, as blood viral load drops below detectable levels after ART, a deeper and more systematic understanding of the HIV tissue reservoirs is imperative. In this review, we take the lymphoid system (including lymph nodes, gut-associated lymphoid tissue, spleen and bone marrow), nervous system, respiratory system, reproductive system (divided into male and female), urinary system as the order, focusing on the particularity and importance of each tissue in HIV infection, the infection target cell types of each tissue, the specific infection situation of each tissue quantified by HIV DNA or HIV RNA and the evidence of compartmentalization and pharmacokinetics. In summary, we found that the present state of HIV in different tissues has both similarities and differences. In the future, the therapeutic principle we need to follow is to respect the discrepancy on the basis of grasping the commonality. The measures taken to completely eliminate the virus in the whole body cannot be generalized. It is necessary to formulate personalized treatment strategies according to the different characteristics of the HIV in the various tissues, so as to realize the prospect of curing AIDS as soon as possible.
Article
Full-text available
Recent development of human three-dimensional organoid cultures has opened new doors and opportunities ranging from modelling human development in vitro to personalised cancer therapies. These new in vitro systems are opening new horizons to the classic understanding of human development and disease. However, the complexity and heterogeneity of these models requires cutting-edge techniques to capture and trace global changes in gene expression to enable identification of key players and uncover the underlying molecular mechanisms. Rapid development of sequencing approaches made possible global transcriptome analyses and epigenetic profiling. Despite challenges in organoid culture and handling, these techniques are now being adapted to embrace organoids derived from a wide range of human tissues. Here, we review current state-of-the-art multi-omics technologies, such as single-cell transcriptomics and chromatin accessibility assays, employed to study organoids as a model for development and a platform for precision medicine.
Article
Previously, two men were cured of HIV-1 through CCR5Δ32 homozygous (CCR5Δ32/Δ32) allogeneic adult stem cell transplant. We report the first remission and possible HIV-1 cure in a mixed-race woman who received a CCR5Δ32/Δ32 haplo-cord transplant (cord blood cells combined with haploidentical stem cells from an adult) to treat acute myeloid leukemia (AML). Peripheral blood chimerism was 100% CCR5Δ32/Δ32 cord blood by week 14 post-transplant and persisted through 4.8 years of follow-up. Immune reconstitution was associated with (1) loss of detectable replication-competent HIV-1 reservoirs, (2) loss of HIV-1-specific immune responses, (3) in vitro resistance to X4 and R5 laboratory variants, including pre-transplant autologous latent reservoir isolates, and (4) 18 months of HIV-1 control with aviremia, off antiretroviral therapy, starting at 37 months post-transplant. CCR5Δ32/Δ32 haplo-cord transplant achieved remission and a possible HIV-1 cure for a person of diverse ancestry, living with HIV-1, who required a stem cell transplant for acute leukemia.
Article
Full-text available
We present the latest version of the Molecular Evolutionary Genetics Analysis (MEGA) software, which contains many sophisticated methods and tools for phylogenomics and phylomedicine. In this major upgrade, MEGA has been optimized for use on 64-bit computing systems for analyzing bigger datasets. Researchers can now explore and analyze tens of thousands of sequences in MEGA. The new version also provides an advanced wizard for building timetrees and includes a new functionality to automatically predict gene duplication events in gene family trees. The 64-bit MEGA is made available in two interfaces: graphical and command line. The graphical user interface (GUI) is a native Microsoft Windows application that can also be used on Mac OSX. The command line MEGA is available as native applications for Windows, Linux, and Mac OSX. They are intended for use in high-throughput and scripted analysis. Both versions are available from www.megasoftware.net free of charge.
Article
Full-text available
Background The London patient (participant 36 in the IciStem cohort) underwent allogeneic stem-cell transplantation with cells that did not express CCR5 (CCR5Δ32/Δ32); remission was reported at 18 months after analytical treatment interruption (ATI). Here, we present longer term data for this patient (up to 30 months after ATI), including sampling from diverse HIV-1 reservoir sites. Methods We used ultrasensitive viral load assays of plasma, semen, and cerebrospinal fluid (CSF) samples to detect HIV-1 RNA. In gut biopsy samples and lymph-node tissue, cell-copy number and total HIV-1 DNA levels were quantified in multiple replicates, using droplet digital PCR (ddPCR) and quantitative real-time PCR. We also analysed the presence of intact proviral DNA using multiplex ddPCR targeting the packaging signal (ψ) and envelope (env). We did intracellular cytokine staining to measure HIV-1-specific T-cell responses. We used low-sensitive and low-avidity antibody assays to measure the humoral response to HIV-1. We predicted the probability of rebound using a mathematical model and inference approach. Findings HIV-1 viral load in plasma remained undetectable in the London patient up to 30 months (last tested on March 4, 2020), using an assay with a detection limit of 1 copy per mL. The patient's CD4 count was 430 cells per μL (23·5% of total T cells) at 28 months. A very low-level positive signal for HIV-1 DNA was recorded in peripheral CD4 memory cells at 28 months. The viral load in semen was undetectable in both plasma (lower limit of detection [LLD] <12 copies per mL) and cells (LLD 10 copies per 10⁶ cells) at 21 months. CSF was within normal parameters at 25 months, with HIV-1 RNA below the detection limit (LLD 1 copy per mL). HIV-1 DNA by ddPCR was negative in rectum, caecum, and sigmoid colon and terminal ileum tissue samples at 22 months. Lymph-node tissue from axilla was positive for the long-terminal repeat (33 copies per 10⁶ cells) and env (26·1 copies per 10⁶ cells), negative for ψ and integrase, and negative by the intact proviral DNA assay, at 27 months. HIV-1-specific CD4 and CD8 T-cell responses have remained absent at 27 months. Low-avidity Env antibodies have continued to decline. Mathematical modelling suggests that the probability of remission for life (cure) is 98% in the context of 80% donor chimerism in total HIV target cells and greater than 99% probability of remission for life with 90% donor chimerism. Interpretation The London patient has been in HIV-1 remission for 30 months with no detectable replication-competent virus in blood, CSF, intestinal tissue, or lymphoid tissue. Donor chimerism has been maintained at 99% in peripheral T cells. We propose that these findings represent HIV-1 cure. Funding Wellcome Trust and amfAR (American Foundation for AIDS Research).
Article
Full-text available
Allogeneic stem cell transplantation (alloSCT) of homozy-gous CCR5 Δ32 stem cells once resulted in the cure of human immunodeficiency virus (HIV) infection. We have recently reported a viral breakthrough in a similar setting. Here, we demonstrate that the rapid rebound after alloSCT was related to a highly replicative CXCR4-tropic HIV variant, which could already be detected before alloSCT.
Article
Full-text available
A cure for HIV-1 remains unattainable as only one case has been reported, a decade ago1,2. The individual—who is known as the ‘Berlin patient’—underwent two allogeneic haematopoietic stem-cell transplantation (HSCT) procedures using a donor with a homozygous mutation in the HIV coreceptor CCR5 (CCR5Δ32/Δ32) to treat his acute myeloid leukaemia. Total body irradiation was given with each HSCT. Notably, it is unclear which treatment or patient parameters contributed to this case of long-term HIV remission. Here we show that HIV-1 remission may be possible with a less aggressive and toxic approach. An adult infected with HIV-1 underwent allogeneic HSCT for Hodgkin’s lymphoma using cells from a CCR5Δ32/Δ32 donor. He experienced mild gut graft-versus-host disease. Antiretroviral therapy was interrupted 16 months after transplantation. HIV-1 remission has been maintained over a further 18 months. Plasma HIV-1 RNA has been undetectable at less than one copy per millilitre along with undetectable HIV-1 DNA in peripheral CD4 T lymphocytes. Quantitative viral outgrowth assays from peripheral CD4 T lymphocytes show no reactivatable virus using a total of 24 million resting CD4 T cells. CCR5-tropic, but not CXCR4-tropic, viruses were identified in HIV-1 DNA from CD4 T cells of the patient before the transplant. CD4 T cells isolated from peripheral blood after transplantation did not express CCR5 and were susceptible only to CXCR4-tropic virus ex vivo. HIV-1 Gag-specific CD4 and CD8 T cell responses were lost after transplantation, whereas cytomegalovirus-specific responses were detectable. Similarly, HIV-1-specific antibodies and avidities fell to levels comparable to those in the Berlin patient following transplantation. Although at 18 months after the interruption of treatment it is premature to conclude that this patient has been cured, these data suggest that a single allogeneic HSCT with homozygous CCR5Δ32 donor cells may be sufficient to achieve HIV-1 remission with reduced intensity conditioning and no irradiation, and the findings provide further support for the development of HIV-1 remission strategies based on preventing CCR5 expression.
Article
Full-text available
Introduction The latent reservoir is the main barrier on the road to HIV cure, and clinical approaches towards eradication are often evaluated by their effect on proviral DNA. To ensure inclusiveness and representativeness in HIV cure studies, proviral DNA quantification assays that are able to detect all common circulating HIV clades are urgently needed. Here, three HIV DNA assays targeting three different genomic regions were evaluated for their sensitivity and subtype‐tolerance using digital PCR. Methods A subtype‐B‐specific assay targeting gag (GAG) and two assays targeting conserved sequences in ltr and pol (LTR and JO) were assessed for their sensitivity and subtype‐tolerance in digital PCR (Bio‐Rad QX200), using a panel of serially diluted subtype reference plasmids as well as a panel of clinical isolates. Both panels represent subtypes A, B, C, D, F, G and circulating recombinant forms (CRFs) AE and AG, which together are responsible for 94% of HIV infections worldwide. Results HIV subtype was observed to greatly affect HIV DNA quantification results. Robust regression analysis of the serially diluted plasmid panel showed that the GAG assay was only able to linearly quantify subtype B, D and G isolates (4/13 reference plasmids, average R² = 0.99), whereas LTR and JO were able to quantify all tested isolates (13/13 reference plasmids, respective average R² = 0.99 and 0.98). In the clinical isolates panel, isolates were considered detectable if all replicates produced a positive result. The GAG assay could detect HIV DNA in four out of five subtype B and one out of two subtype D isolates, whereas the LTR and JO assays detected HIV DNA in all twenty‐nine tested isolates. LTR and JO results were found to be equally precise but more precise than GAG. Conclusions The results demonstrate the need for a careful validation of proviral reservoir quantification assays prior to investigations into non‐B subtype reservoirs. The LTR and JO assays can sensitively and reliably quantify HIV DNA in a panel that represents the worldwide most prevalent subtypes and CRFs (A, B, C, D, AE, F, G and AG), justifying their application in future trials aimed at global HIV cure.
Article
Despite the success of antiretroviral therapy (ART) for people living with HIV, lifelong treatment is required and there is no cure. HIV can integrate in the host genome and persist for the life span of the infected cell. These latently infected cells are not recognized as foreign because they are largely transcriptionally silent, but contain replication-competent virus that drives resurgence of the infection once ART is stopped. With a combination of immune activators, neutralizing antibodies, and therapeutic vaccines, some nonhuman primate models have been cured, providing optimism for these approaches now being evaluated in human clinical trials. In vivo delivery of gene-editing tools to either target the virus, boost immunity or protect cells from infection, also holds promise for future HIV cure strategies. In this Review, we discuss advances related to HIV cure in the last 5 years, highlight remaining knowledge gaps and identify priority areas for research for the next 5 years.
Article
Background: Allogeneic bone marrow transplant (alloBMT) in people living with HIV (PLWH) can lead to the undetectable levels of HIV reservoirs in blood, even using highly sensitive assays. However, with antiretroviral therapy (ART) interruption, rebound of HIV viremia occurs. The source of this rebound viremia is of interest in HIV cure strategies. Methods: Within a trial of alloBMT in individuals with hematologic malignancies and HIV (ClinicalTrials.gov, NCT01836068), one recipient self-interrupted ART after achieving >99.5% host cell replacement in peripheral blood by day 147 and developed severe acute retroviral syndrome with meningoencephalitis at 156 days post-alloBMT. We isolated replication-competent HIV using a quantitative viral outgrowth assay at -100 and -25 days pre-alloBMT and from the same time points pre-alloBMT for HIV DNA and cell-associated RNA from peripheral blood mononuclear cells and resting memory CD4+ T-cells. We isolated HIV RNA in plasma and cerebrospinal fluid (CSF) at viral rebound. We sequenced the RT-region of pol and performed neighbor-joining phylogenetic reconstruction. Results: Phylogenetic analysis revealed an identical viral sequence at both pre-alloBMT time-points accounting for 9/34 sequences (26%) of the sampled HIV reservoir. This sequence population grouped with viral rebound sequences from plasma and CSF with high sequence homology. Discussion: Despite >99.5% replacement of host cells in peripheral blood, ART interruption led to HIV viral rebound in plasma and CSF. Further, the rebound virus matched replication competent virus from resting memory CD4+ T-cells pre-alloBMT. This case underscores that HIV-infected recipient cells can persist after alloBMT, and that latent replication-competent virus can re-establish infection.
Article
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only medical intervention that has led to an HIV cure. Whereas the HIV reservoir sharply decreases after allo-HSCT, the dynamics of the T cell reconstitution has not been comprehensively described. We analyzed the activation and differentiation of CD4 ⁺ and CD8 ⁺ T cells, and the breadth and quality of HIV- and CMV-specific CD8 ⁺ T cell responses in 16 patients with HIV who underwent allo-HSCT (including five individuals who received cells from CCR5Δ32/Δ32 donors) to treat their underlying hematological malignancy and who remained on antiretroviral therapy (ART). We found that reconstitution of the T cell compartment after allo-HSCT was slow and heterogeneous with an initial expansion of activated CD4 ⁺ T cells that preceded the expansion of CD8 ⁺ T cells. Although HIV-specific CD8 ⁺ T cells disappeared immediately after allo-HSCT, weak HIV-specific CD8 ⁺ T cell responses were detectable several weeks after transplant and could still be detected at the time of full T cell chimerism, indicating that de novo priming, and hence antigen exposure, occurred during the time of T cell expansion. These HIV-specific T cells had limited functionality compared with CMV-specific CD8 ⁺ T cells and persisted years after allo-HSCT. In conclusion, immune reconstitution was slow, heterogeneous, and incomplete and coincided with de novo detection of weak HIV-specific T cell responses. The initial short phase of high T cell activation, in which HIV antigens were present, may constitute a window of vulnerability for the reseeding of viral reservoirs, emphasizing the importance of maintaining ART directly after allo-HSCT.
Article
Background: Understanding HIV dynamics across the human body is important for cure efforts. This goal has been hampered by technical difficulties and the challenge to obtain fresh tissues. Methods: This observational study evaluated 6 persons with HIV (4 virally suppressed with antiretroviral therapy and 2 with rebound viremia after stopping therapy) who provided blood serially before death and their bodies for rapid autopsy. HIV reservoirs were characterized by digital droplet PCR and single genome amplification and sequencing of full-length (FL) envelope HIV. Phylogeographic methods reconstructed HIV spread and generalized linear models tested for viral factors associated with dispersal. Results: Across participants, HIV DNA levels varied from ~0 to 659 copies/106 cells (IQR:22.9-126.5). A total of 605 intact FL env sequences were recovered in antemortem blood cells and across 28 tissues (IQR:5-9). Sequence analysis showed: 1) emergence of large, identical, intact HIV RNA populations in blood after stopping therapy, which repopulated tissues throughout the body, 2) multiple sites acted as hubs for HIV dissemination but blood and lymphoid tissues were the main source, and 3) viral exchanges occurred within brain areas and across the blood brain barrier, and 4) migration was associated with low HIV divergence between sites and higher diversity at the recipient site. Conclusion: HIV reservoirs persist in all deep tissues, and blood is the main source of dispersal. This may explain why eliminating HIV susceptibility in circulating T cells via bone marrow transplants allowed some people with HIV to have therapy free remission, even though deeper tissue reservoirs were not targeted. Trial registration: Not applicable. Funding: National Institute of Health Grants (P01 AI31385, P30 AI036214, AI131971-01, AI120009AI036214,HD094646, AI027763, AI134295, AI68636).
Article
Background: The multifactorial mechanisms associated with radical reductions in HIV-1 reservoirs after allogeneic hematopoietic stem cell transplant (allo-HSCT), including a case of HIV cure, are not fully understood. Objective: To investigate the mechanism of HIV-1 eradication associated with allo-HSCT. Design: Nested case series within the IciStem observational cohort. Setting: Multicenter European study. Participants: 6 HIV-infected, antiretroviral-treated participants who survived more than 2 years after allo-HSCT with CCR5 wild-type donor cells. Measurements: HIV DNA analysis, HIV RNA analysis, and quantitative viral outgrowth assay were performed in blood, and HIV DNA was also measured in lymph nodes, ilea, bone marrow, and cerebrospinal fluid. A humanized mouse model was used for in vivo detection of the replication-competent blood cell reservoir. HIV-specific antibodies were measured in plasma. Results: Analysis of the viral reservoir showed that 5 of 6 participants had full donor chimera in T cells within the first year after transplant, undetectable proviral HIV DNA in blood and tissue, and undetectable replication-competent virus (<0.006 infectious unit per million cells). The only participant with detectable virus received cord blood stem cells with an antithymocyte globulin-containing conditioning regimen, did not develop graft-versus-host disease, and had delayed complete standard chimerism in T cells (18 months) with mixed ultrasensitive chimera. Adoptive transfer of peripheral CD4+ T cells to immunosuppressed mice resulted in no viral rebound. HIV antibody levels decreased over time, with 1 case of seroreversion. Limitation: Few participants. Conclusion: Allo-HSCT resulted in a profound long-term reduction in the HIV reservoir. Such factors as stem cell source, conditioning, and a possible "graft-versus-HIV-reservoir" effect may have contributed. Understanding the mechanisms involved in HIV eradication after allo-HSCT can enable design of new curative strategies. Primary funding source: The Foundation for AIDS Research (amFAR).