Abstract
The evolution of drug-resistant viral strains and anatomical and cellular reservoirs of HIV pose significant clinical challenges to antiretroviral therapy. CCR5 is a coreceptor critical for HIV host cell fusion, and a homozygous 32-bp gene deletion (Δ32) leads to its loss of function. Interestingly, an allogeneic HSCT from an HIV-negative Δ32 donor to an HIV-1-infected recipient demonstrated a curative approach by rendering the recipient’s blood cells resistant to viral entry. Ex vivo gene editing tools, such as CRISPR/Cas9, hold tremendous promise in generating allogeneic HSC grafts that can potentially replace allogeneic Δ32 HSCTs. Here, we review antiretroviral therapeutic challenges, clinical successes, and failures of allogeneic and allogeneic Δ32 HSCTs, and newer exciting developments within CCR5 editing using CRISPR/Cas9 in the search to cure HIV.
Keywords: CCR5, Δ32, HIV, CRISPR/Cas9, antiretroviral therapy
1. Introduction
Human immunodeficiency virus (HIV) infection remains a significant global public health threat responsible for tens of millions of deaths [1]. Currently, the treatment of HIV includes oral and extended-release injectable drugs [2]. Since its inception, antiretroviral therapy (ART) has significantly reduced morbidity, mortality, and the global burden of the virus [1]. ART, started within the first six months after infection, reduces viral load to undetectable levels, improves CD4 T cell counts, inhibits HIV transmission, and significantly improves a patient’s quality of life [3].
Forty-six FDA-approved antiretroviral (ARV) drugs administered in efficacious combinations target viral proteins and are effective [2]. ARVs are not very accessible in resource-limited areas and, unfortunately, have inadequate reach to nations needing them most [4]. ARVs are generally not well-tolerated and are at constant risk of obsolescence due to the emergence of drug-resistant strains [5]. Most ARVs have a limited capacity to penetrate the blood-brain barrier (BBB); thus, their access to the CNS remains a significant ongoing challenge to treating HIV sequestered in several immune and drug-privileged sites [6]. Not surprisingly, a complete or even functional HIV cure via ART is currently impossible.
HIV infects different compartments of the host immune system. In the circulation, for example, where the virus is most infectious, HIV replicates fastest in proliferating CD4+ T cells [7]. In contrast, HIV usually remains latent in resting CD4+ T and phagocytic cells in dispersed tissues and organ systems [8]. Consequently, latently infected CD4+ cells act as sanctuaries for the integrated provirus to hide undetected for years or even decades before emergence [9].
HIV infection begins with the interaction of the viral surface protein, gp120, with a CD4 molecule expressed on several types of immune cells [10]. This interaction critically requires C-C Chemokine Receptor Type 5 (CCR5) coreceptor engagement, allowing membrane fusion of the HIV particle to the CD4+ host cell [11]. Intriguingly, a genetic mutation identified in a small set of individuals of European descent leads to a functional loss of CCR5 activity, resulting in significant HIV-1 resistance [12]. Individuals with the Δ32/Δ32 homozygous mutation (Δ32) of their CCR5 genes are resistant to HIV-1 [13], and stem cell transplantation (SCT) from Δ32 healthy donors to HIV patients leads to presumed cures. To date, allogeneic hematopoietic stem cell transplantation (HSCT) from Δ32 donors has provided the only curative therapy for HIV [14–18].
Here, we will discuss HIV infection and treatment challenges, the CCR5 gene and its Δ32 mutation, allogeneic Δ32 stem cell transplants, clinical successes, and potential clustered regularly interspaced short palindromic repeats (CRISPR)-based methods to reproduce this mutation in individuals living with HIV.
2. Infection
HIV is an infectious retrovirus that contains two copies of positive-sense single-stranded (ss) RNA. Encoded and equipped with many critical essential and regulatory enzymes, HIV exploits the host’s cellular machinery to replicate itself, leaving infected CD4+ cells metabolically exhausted, after which infected cells scavenge their membranes to bud new virions and die [19].
HIV can be transmitted from infected individuals through sexual contact, during pregnancy or breastfeeding, or through exposure to infected materials, including blood products and contaminated needles [20]. Once HIV enters the individual, the virus infects CD4+ cells. This primary infection is associated with a fever and other common flu-like symptoms, which quickly resolve, and can be difficult to distinguish from a superficial influenza infection [21]. After this, the virus starts infecting other CD4+ cells, and subsequent symptoms appear, including skin rashes, fatigue, lymphadenopathy, sore throat, repeated episodes of fever, and infection with opportunistic pathogens [21]. If left untreated, HIV leads to a collapse in CD4+ T cell numbers, with the host immune system deteriorating to a point where multiple infections or HIV-specific cancers arise, overwhelming the host, terminating in the virus’s terminal end-stage, acquired immunodeficiency syndrome (AIDS) [21].
3. Treatment
ARVs are anti-HIV drugs administered in a combination that inhibits structural or functional targets critical for viral pathogenesis. ART is the only cost-effective treatment for HIV and is the current standard of care [22]. Approved ARVs have distinct targets, including viral reverse transcriptase (Non- Nucleoside Reverse Transcriptase Inhibitors and Nucleoside Reverse Transcriptase Inhibitors) [23], HIV protease (Protease Inhibitors) [24], HIV fusion (Fusion inhibitors) [25], CCR5 (CCR5 antagonists) [26], HIV integrase (Integrase Strand Transfer Inhibitor and Integrase inhibitors) [27], viral gp120 protein (Attachment inhibitors) [28] and host CD4 receptor (post-attachment inhibitors) [29] (Table 1). Other pharmacokinetic enhancers, such as ritonavir and cobicistat, can increase the effectiveness of some ARVs in an anti-HIV regimen [30].
Table 1.
FDA-approved antiretroviral drugs for HIV.
Drug Class | Mode of action | Drugs |
---|---|---|
Nucleoside Reverse Transcriptase Inhibitors | Reverse transcriptase blocker | Abacavir* |
Emtricitabine* | ||
Lamivudine* | ||
Tenofovir disoproxil fumarate* Tenofovir alafenamide* | ||
Zidovudine* | ||
Non-Nucleoside Reverse Transcriptase Inhibitors | Reverse transcriptase blocker | Doravirine |
Efavirenz* | ||
Etravirine | ||
Nevirapine* | ||
Rilpivirine | ||
Protease Inhibitors | HIV protease blocker | Atazanavir* |
Darunavir* | ||
Fosamprenavir | ||
Ritonavir* | ||
Lopinavir* | ||
Tipranavir | ||
HIV capsid inhibitor | Interferes with capsid assembly | Lenacapavir |
Fusion Inhibitor | Binds CD4 receptors, blocking fusion | Enfuvirtide |
CCR5 Antagonist | Binds CCR5, preventing viral entry | Maraviroc |
Integrase Strand Transfer Inhibitors | Binds the integrase-viral DNA complex | Cabotegravir |
Dolutegravir* | ||
Raltegravir* | ||
Bictegravir | ||
Attachment Inhibitor | Binds gp120, preventing viral entry | Fostemsavir |
Post-Attachment Inhibitor | Binds gp120-CD4 complex, preventing coreceptor engagement | Ibalizumab |
Pharmacokinetic Enhancer | Inhibits key liver enzymes that decrease bioavailability of protease inhibitors | Cobicistat |
Combination | Multiple targets | Bictegravir Emtricitabine Tenofovir alafenamide |
WHO recommended first and second line regimen drugs.
HIV treatment relies on combinations of ARVs given for life. After testing positive for the virus, the treatment protocol is to start first-line ART immediately, which consists of two nucleoside reverse transcriptase inhibitors combined with a third ARV from one of three other classes, such as a protease inhibitor, an integrase strand transfer inhibitor, or a non-nucleoside reverse transcriptase inhibitor [22]. If the patient is non-compliant and/or the initial drugs are no longer effective, then second-line ART is considered [22]. Alternative ART commences if viral load shows no further improvement with second-line drugs, usually due to cross-drug resistance. ART administration is not benign and can lead to chronic adverse effects and toxicities with associated comorbidities [31]. ART administration can also induce lactic acidosis associated with fatigue, pulmonary complications, altered liver function, circulation complications, heart palpitations, and weight loss [32]. Patients on ART commonly complain of chronic nausea, vomiting, and abdominal discomfort.
3.1. Drug resistance
ARVs were initially designed to target HIV structures or viral enzymes, hoping that effective agents would be curative. However, viral drug resistance and other escape mechanisms challenged the continuing efficacy of ARVs. Retroviruses like HIV use reverse transcriptase enzymes and the infected host cell transcriptional machinery to synthesize cDNA based on their genomic (g) RNA template. HIV reverse transcriptase has limited proofreading activity and a reported error rate of 1/1,700 bp’s [33], an exceptionally high error rate, even for a retrovirus. Thus, each time the virus synthesizes new DNA strands during replication, mistakes go uncorrected and are incorporated into its nucleic acid, producing new gRNA variant copies and, consequently, new virion variants [34]. The error-prone process usually leads to replicative incompetent virions or, less often and more importantly, allows the virus to escape the effect of the original ARVs. Conventional ARVs target native forms of HIV enzymes, but newly emerging virion variants produce nonnative enzymes, and often ARVs may no longer be effective [35].
For this reason, individuals on long-term ART (~10 years) may suddenly show high viral loads despite being ART compliant and may need to switch to second or alternative-line drugs to manage the disease. Not surprisingly, ART non-compliance, for whatever reason, hastens this process by giving the virus time to replicate, mutate, and develop resistance [36]. Nevertheless, ART effectively suppresses viral load, improves CD4+ T cell numbers, dramatically reduces viral transmission, improves patient quality of life, and delays symptomatic disease, but it is not curative.
3.2. Inaccessible anatomical sites
Inaccessible HIV reservoirs in immune- and drug-privileged sites can act as sanctuaries for the provirus and pose ongoing clinical challenges to ART. For example, HIV enters the CNS early after infection, and in almost half of ART-treated patients, progresses to HIV-1-associated neurologic disorders (HAND) [37]. HAND is associated with dementia, cognitive motor disorders, and neurocognitive disorders, which can become severe and debilitating over time. For a cure, the principal HIV-infected cell subsets in the CNS that harbor the provirus must be eliminated, including CD4+ macrophages, CD4+ microglia, CD4+ T cells, and perhaps CD4− astrocytes [38]. HIV can infect resident CNS cells by cell-free (cis) mechanisms and direct cell-to-cell (trans) transmission [39–41]. HIV-infected cells, such as monocytes and CD4+ T cells, can transit from the bloodstream across the BBB and into the CNS, directly infecting microglia [41]. Astrocytes are presumed infected by a CD4-independent cell-to-cell contact mechanism in which immature HIV virions bud off lymphocytes, binding directly to CXCR4 and fusing with the astrocyte membrane [42].
Most ARVs cannot penetrate the BBB. However, some ARVs, such as non-nucleoside reverse transcriptase inhibitors, including efavirenz, etravirine, rilpivirine, and nevirapine, can disrupt BBB integrity and enter the CNS, leading to potentially deleterious consequences and increasing the likelihood and severity of a life-threatening stroke [43]. Newer methods to facilitate the bioavailability of other ARVs to the CNS can be envisioned, such as nasal delivery directly to the brain; however, these strategies need careful and thorough assessment [44].
The testes, fetus, and placenta are other immune-privileged sites most ARVs cannot reach [45]. Fortunately, mothers can dramatically lessen the risk of fetal HIV transmission when on ART due to very low or undetectable viral loads [46].
3.3. Reservoir cells
HIV preferentially binds CD4 receptors but can bind to other receptors, including DC-SIGN, Syndecan-3, and Siglec-1, to acquire cell membrane access [47–49]. Once within the cell, HIV integrase, a critical pathogenic enzyme, inserts the newly generated HIV dsDNA into the host genome [50]. The virus resides in resting and productive target cells, resulting in latent and active HIV-infected cells. Viral antigens, such as gp160, are not expressed on latently infected cells, which are immunologically inert and quiescent, with the provirus remaining transcriptionally silent for years or decades [8]. However, virus replication can resume once the cognate antigen activates the host T cell.
It is still unclear how HIV infects so many different types of cells, but CCR5, expressed by many immune and epithelial cells, appears as a central receptor facilitating host cell-viral fusion [51]. In addition, cell-to-cell viral transmission by mechanisms including efferocytosis [52], tunneling nanotubes [53], virus-containing compartments (VCC) [54], cell-cell fusion [55], and virological synapses [56] are also responsible for expanding the pool of target cells. Monocytes and macrophages also display CD4 receptors and likely play an essential role in generating additional proviral latent reservoirs. Once in local tissues, infected peripheral monocytes become infected resident macrophages (monocyte-derived macrophages), which act as a long-term reservoir of HIV [57]. Translation induces intracellular immunologically inert VCC formation when these monocytes transcribe viral genes such as gag, which encodes the viral core’s structural components. Each VCC contains numerous copies of HIV virions bound to the tetherin/BST-2 protein sheltered in monocytes [58]. During immunological synapse formation, viral transmission occurs by coreceptor and adhesion molecule engagement on the surface of infected antigen-presenting cells, infected CD4+ T cells, productively infected cells, or uninfected target cells [56]. Finally, CD14+CD11c+ monocyte-derived dendritic cells and langerin-expressing conventional dendritic cells can present HIV particles to CD4+ cells following vaginal or anal sex, leading to transmission [59].
HIV transmission primarily occurs by exchanging infected blood or bodily fluids harboring latently infected CD4+ T cells or infectious virus [8]. The virus then spreads to different compartments, including the gastrointestinal tract-associated lymphoid tissues, CNS, spleen, liver, lungs, and kidneys. HIV particles at these sites can be genetically distinct from those in the peripheral circulation. Surprisingly, local CD4+ T cell subsets are frequently unlike those observed in the peripheral circulation concerning phenotype and function. The stability of the CD4+ T cell reservoir is unique to each infected cell that resides in a unique environment of regulatory cytokines and chemokines [60]. In a constant back-and-forth battle, immune cells mount proinflammatory responses against HIV by cytokine and chemokine secretion. However, HIV hijacks many of these cytokines to establish and maintain latent reservoirs [61]. Many of these cytokines regulate HIV replication and innate and adaptive responses. For example, TNF-α, IL-2, and IL-6 break latency in resting CD4+ cells, promoting proviral emergence, and IL-18 boosts viral replication.
As the disease progresses from acute to chronic, a decline in healthy immune cells occurs, and the immune response shifts from attack to defense. In this phase, immunosuppressive cytokines, including IL-10 and TGF-β, are secreted, reducing T helper (Th)1 type responses, MHC II expression, and antigen presentation by APCs [62]. IL-10 also upregulates programmed cell death protein (PD)-1, and TGF-β promotes the expression of cytotoxic T lymphocyte antigen (CTLA)-4, leading to T-cell exhaustion and reduced activation [63]. By downregulating T cells, HIV facilitates its latency and promotes reservoir persistence. Additionally, chemokines such as CXCL9, CXCL10, CCL19, CCL20, and CCL21[64], and cytokines such as IL-2, IL-4, IL-7, and IL-15, enable resting CD4+ T cells to become infected [65]. Interestingly, IL-7 activates the AKT/FOXO3a pathway, reducing the expression of proapoptotic FasL and Bim, promoting memory T cell survival, and stabilizing the latent reservoir [66]. Thus, the latent reservoir of HIV-infected cells remains constant by the homeostatic and cognate antigen-stimulated proliferation of infected cells.
4. CCR5 – gene, protein, and the Δ32 mutation
At position 21 on the short arm of chromosome 3, the human CCR5 gene has two or three exons that encode its three transcript variants. The sense strand has two upstream promoters, offering potential binding sites for several interferon-stimulated response elements, κB factors, and cAMP-response elements, providing a binding platform for interferon regulatory elements, NF-κB, and the common activator of transcription CREB-1, respectively [67]. The CCR5 antisense strand (CCR5AS) encodes long non-coding RNA sequences, further regulating CCR5 expression. Single nucleotide polymorphisms associated with one CCR5AS allelomorph increase CCR5 expression. These polymorphisms increase viral load, promote latency, and decrease CD4 T cell counts, accelerating disease progression, and can play a negative role in disease outcomes [68,69].
The CCR5 gene encodes C-C chemokine receptor 5, a beta chemokine receptor structurally similar to G protein-coupled receptors (GPCRs) [70]. This protein is expressed in many cells, including T cells, monocytes, macrophages, natural killer cells, microglia, astrocytes, and dendritic cells. It is a critical coreceptor for macrophage-tropic HIV strains [71]. Like other GPCRs, CCR5 has one extracellular N-terminal domain (NTD), three extracellular (ECL1–3) loop segments, seven transmembrane hydrophobic α-helical domains, one cytoplasmic C-terminal domain (CTD), and three intracellular (ICL1–3) loop segments [72] (Fig. 2A). The ligand recognition site comprises a tyrosine-rich motif in the NTD and various conserved amino acids in ECL1 and ECL2 [73]. CCR5 ligands are inflammatory chemokines, including RANTES, MIP-1a, MCP-2, and MIP-1b [74]. Site-directed mutagenesis and molecular docking studies demonstrated that core domains of ligands initially interact with ECL1, ECL2, and the NTD, then bind later to the transmembrane helical bundle of CCR5 via the N-terminus of the ligand [75]. Once activated in its two-step binding process, a conformational change in CCR5 desensitizes PKC/GRK-dependent phosphorylation of the CTD and ICL3 and induces CCR5 internalization. This results in the recruitment of cytotoxic lymphocytes and activation of antigen-specific T cells via secondary signaling pathways, including PI3K/Akt and MAPK/ERK, by G-protein subunit release and engagement of other effector molecules [76]. Once the ligand undocks, CCR5 is recycled back to the surface. HIV gp120 hijacks the exact mechanism of ligand binding and activation of CCR5+ cells. The NTD of CCR5 contains four tyrosine residues; sulfation of two residues appears essential for high-affinity binding to gp120. Gp120 has two sites able to interact with sulfated tyrosine residues and two others able to interact with non-sulfated tyrosine residues [77].
Fig. 2.
CCR5 structure (A); and Stages of CCR5-mediated HIV entry (B).
CD4bs, CD4 binding site.
In classical molecular dynamics and site-directed mutagenesis models, four gp120s stabilize CCR5 [78]. Simulated structure studies revealed that gp120 molecules stabilize CCR5 in different conformational states, reorienting ECL2 and ECL3 and allowing access to the transmembrane binding cavity. Gp120 also reshapes this cavity giving rise to distinct positions of transmembrane helices 5, 6, and 7 and ICL3 cytoplasmic ends that may influence G protein binding regions [78]. To facilitate binding and access to the transmembrane cavity, HIV gp120 also changes conformation after its first interaction with the CD4 receptor. This exposes the hypervariable V3 loop, which has a high affinity for ECL2 and NTD of CCR5 (Fig. 2B).
The Δ32 CCR5 mutation is a 32-bp deletion resulting in a frameshift mutation with a premature stop codon leading to a truncated form of the protein [79]. The mutant protein remains cytosolic, unable to translocate to the cell surface. Moreover, the specific amino acids 298–329 in the hypervariable V3 loop of HIV gp120 that determine cellular tropism bind with the 2D7 epitope in the ECL2 of CCR5 [80]. This truncated form of CCR5 structurally lacks the 2D7 loop since the frameshift occurs before 2D7 in the open reading frame, making the cytosol bound truncated CCR5 protein inaccessible to the free virus.
Predominantly found in Europe and Western Asia, the Δ32 genotypes resulted from positive natural selection of the Δ32/Δ32 alleles [81,82]. The Δ32 mutation evolved approximately 700 years ago and, amid the smallpox endemics of the eighteenth century, which exerted selective pressure on European and Western Asian populations, increasing the penetrance of the allele [83]. Roughly 10% of Europeans retain paired missense mutations C101X and FS299 or C20S and C178R, responsible for the Δ32 mutation [84]. Interestingly, the Δ32 CCR5 genotype appears protective against several other viruses, including smallpox [85] and SARS-CoV-2 [86], and CCR5 blockade inhibited Dengue and Zika virus infection [87,88]. Similarly, genetic polymorphisms of CCR5 ligands are involved in protection against Trypanosoma cruzi infection [89]. Even more surprisingly, individuals with Δ32 mutations show improved neuronal plasticity and faster neuronal recovery following a stroke or traumatic brain injury, indicating a significant role for the immune response in these presumed immune-independent conditions [90]. The Δ32 mutation can also have negative consequences, including increased susceptibility to the West Nile virus [91], increased risk of death from the influenza virus [92], and detrimental bone development due to defective osteoclastogenesis and osteoclast communication [93].
5. Δ32 CCR5 as a functional HIV cure?
Allogeneic HSCTs carrying the Δ32 mutation have cured five patients to date. Initially, two male HIV+ Caucasians, the ‘Berlin’ and ‘London’ patients, were cured of HIV-1 following allogeneic HSCT from Δ32 CCR5 donors (Fig. 1) [14,15]. Both patients had been on ART for many years and developed HIV-associated hematologic malignancies. The HSCT replaced recipient immune cells damaged during chemotherapy, HIV-1 infection, and ART with healthy donor immune cells containing the Δ32 homozygous mutations in CCR5, allowing the recipients to reconstitute their immune cells with CCR5-deficient cells. Homozygous Δ32 donors were explicitly selected so that following immune reconstitution, the new immune cells would be highly resistant to HIV-1 infection. Thus, patients could stop ART following immune reconstitution. Findings from distinct HIV-1 reservoir sites in these patients, such as plasma, semen, and cerebrospinal fluid, revealed undetectable levels of HIV-1 RNA, intact proviral DNA, viral packaging signals ψ, or envelope [14,15]. The treatment, however, was rather intense, requiring chemo- or radiotherapy to ablate existing bone marrow cells and immunosuppressive drug administration to control graft versus host disease (GVHD). Both patients were declared free of HIV-1.
Fig. 1.
Clinical developments in CCR5-based cell replacement therapies.
AML, Acute myeloid leukemia; ALL, Acute lymphocytic leukemia
The ‘City of Hope’ patient was the third to be cured. This Caucasian male had lived the longest with the virus before the transplant [16]. He was an ART-compliant patient who presented with acute myeloid leukemia (AML) and received an allogeneic HSCT from a Δ32 CCR5 donor treating both AML and HIV-1. This patient subsequently stopped ART, and clinical findings at reservoir sites were negative. He has remained in dual HIV-1 and AML remission since 2019.
A 53-year-old Caucasian male, also known as the ‘Düsseldorf patient,’ is the fourth patient to be cured of HIV-1 following allogeneic HSCT from a Δ32 CCR5 donor [17]. Like the City of Hope patient, this individual also developed AML and underwent HSCT to treat both conditions. However, his AML relapsed after four months, forcing the patient to continue ART. Five years later, in 2018, the patient stopped ART and was assessed for viral load. Residual, non-replicative HIV-RNA and HIV-DNA were detected, yet the patient remains HIV-1-negative.
A fifth case in New York has generated high hopes of developing an HIV cure using a haplo-cord transplant method [18]. This female patient from a mixed genetic background marked a significant step toward an HIV cure.
Haplo-cord transplants involve cord blood SC transfusions followed by adult bone marrow SC transfusions [94]. Cord blood transplants generally contain few SCs and often require serial transplants to achieve a reconstituting dose [95]. Double cord transplants are rarely obtained from individual donors; unfortunately, single cord blood transplants face significant graft failure and a high incidence of infection [95, 96]. Therefore, in haplo-cord transplants, cord blood SCs are accompanied by mismatched (haploidentical) healthy adult marrow SCs, supporting the recipient against diseases [94]. For a patient of African or mixed ancestry like this New York patient, finding a Δ32-matched donor is challenging because, to date, the Δ32 screening registries have only reported Caucasians to be Δ32 homozygous; however, most HIV-infected individuals are of African ancestry. Δ32 pre-screened cord blood banks hold promise for providing hematopoietic Δ32 SCs. In the New York patient’s case, Δ32 cord blood SCs and wildtype adult SCs from a close relative with a partial HLA mismatch were administered. Thus, Δ32 SCs from the cord blood donor could adapt more readily to the new host with the aid of adult SCs. This approach could hold significant promise for allogeneic Δ32 transplants in non-Caucasian populations.
6. R5 and X4 viral tropism
Synthetic CCR5 based cell or gene therapy cannot offer a complete cure for HIV-1, as this therapy does not treat infections caused by CXC chemokine receptor 4 (CXCR4) - tropic (X4) HIV-1 virus. Host cell surface expression of the HIV entry receptor complex composed of CD4 plus CXC chemokine receptor 4 (CXCR4) or CCR5 determines viral tropism [97]. Other chemokine receptors on non-CD4+ target cells, including CCR3 and CCR8, can also aid in viral fusion and expand the pool of infectable cells [98]. R5-tropic viruses bind CCR5 for host cell fusion and are usually present in the early stages of HIV infection. In contrast, X4-tropic strains, which utilize CXCR4 to promote host cell fusion, begin to dominate during later infection stages [99]. Intriguingly, the normal progression of HIV infection begins with R5 tropism and then switches to an X4 tropic virus. Strains that can bind both CCR5 and CXCR4 for host cell fusion characterize intermediate pathogenic stages, in which HIV appears to prefer CXCR4 receptors on naïve T cells and CCR5 receptors on memory T cells [100]. Surprisingly, this R5 to X4 phenotypic switch has not yet occurred in the five clinical Δ32 CCR5 HSCT cases cured of HIV-1 discussed above, suggesting that CXCR4 tropic viruses were absent or, if present, could not infect donor CCR5-deficient cells [14–18].
It is important to understand the mechanism of phenotypic switching during disease progression. In normal individuals, memory T cells divide nearly ten times as frequently as naïve cells [101]. Therefore, preferential R5 tropism for memory T cells in early infection can augment viral expansion [102]. As the disease progresses, the frequency of naïve and memory T cell division equalizes, allowing viral strains that can exploit both chemokine receptors to dominate. In the latter stages of HIV infection, viral load increases and CD4+ counts drop; consequently, naïve T cell division frequency exceeds that of the remaining memory T cells, and X4 tropic viruses predominate [103].
Notably, genetic ablation of CCR5 is a naturally validated approach to cure HIV-1. However, ablation of CXCR4 did not support a viable treatment option for X4 tropic HIV-1 in a preclinical setting 104]. Nevertheless, X4- (NL4–3) and R5- (YU-2) tropic strains failed to infect CCR5 and CXCR4 ablated CD4+ T cells in vitro, and this dual ablation was suggested as a prophylactic strategy in developing nascent cell-based therapeutics for curing HIV-1[105].
7. CCR5 gene editing using CRISPR/Cas9
CRISPR-Cas is an adaptive immune mechanism targeting foreign genetic elements in bacteria and archaea [106]. This intra- or extra-chromosomal genetic system deploys nucleases, such as Cas, followed by a leader sequence and a CRISPR array composed of short, direct repeats stacked with foreign DNA sequences called spacers. CRISPR-Cas systems are highly diverse and divided into many subtypes. The most frequently used gene editing CRISPR system is adapted from Streptococcus pyogenes. This type II system uses the Cas9 endonuclease and crRNA (guide), and tracrRNA (scaffold) to perform dsDNA nicks in an RNA-guided manner [107]. Cas9 comprises six domains: Rec I, Rec II, Bridge Helix, HNH, RuvC, and protospacer adjacent nucleotide motif (PAM) interacting. Synthetically designed single guide-RNA (sg-RNA) binds to the Rec I domain, activating Cas9 to interrogate the host DNA for a specific PAM signature. The PAM sequence binds the Cas9-sgRNA complex allowing the sg-RNA to form a heteroduplex with the target DNA. This heteroduplex then activates the endonuclease domains HNH and RuvC, breaking the phosphodiester bonds between the third and fourth nucleotides upstream of the PAM. CRISPR-based targeting exploits two major active DNA repair pathways to facilitate gene editing: HDR (homology-directed repair) for applications such as abnormal gene replacement or wild-type gene insertions and NHEJ (Non-homologous end joining) for therapeutic deletions [108,109]. However, other DNA repair pathways, such as HITI (homology-independent targeted integration) [110], BER (base excision repair), and PE (prime editing), were artificially developed for more precise CRISPR editing applications (Fig. 3) [111,112].
Fig. 3.
The CCR5 gene is situated at 3p.21.31, and a 32-bp deletion leads to a truncated, non-functional protein. Various CRISPR approaches can artificially reproduce this deletion, including knock-out, knock-in, base-editing, and prime editing.
A. CRISPR-Cas9 editing. Cas9-induced double-stranded DNA breaks (DSBs) initiate intrinsic DNA repair mechanisms, resulting in an edited CCR5 gene. Primary DSB repair mechanisms include homology-directed repair (HDR) and non-homologous end-joining (NHEJ). HDR, which is accurate and most active in dividing cells, employs an homologous template to mend DSBs. In contrast, NHEJ directly ligates nicked ends in a homology-independent manner, often introducing small insertions and deletions at the joined ends. In non-dividing cells, homology-independent targeted integration (HITI) serves as an assisted endogenous NHEJ method for knock-in applications. The HITI insert cassette contains short target flanking sequences, enabling both the target and insert to share the same sgRNA-Cas9 site. Targeting these sequences produces identical ends (S1 and S2) in genomic DNA and insert cassette. Correct insert orientation eliminates the sgRNA target site, while incorrect orientation recreates the sgRNA target.
B. Base editing. Single-strand DNA breaks (SSBs) yield better results and demonstrate an enhanced insertion and deletion (indel) frequency compared to DSBs. Base editing utilizes single-strand nicking Cas9. The cytosine base editor (CBE) consists of a D10A Cas9 nickase (RuvC mutant) bound to cytidine deaminase (Apobec1) and a uracil glycosylase inhibitor (UGI). Apobec1 converts C to U, while UGI prevents the conversion of U to G. Mismatch repair replaces U with T; subsequently, on the complementary strand, it replaces G with A. Cytidine base editing can transform C-T into T-A and introduce premature stop codons at desired positions within the CCR5 coding region. The adenine base editor (ABE) converts A to G and comprises D10A bound to adenine deaminase (TadA-8e). TadA-8e converts A to G, and mismatch repair replaces T on the complementary strand with a C. ABE can eliminate the transcription start site by changing the start codon ATG to GTG or ACG.
C. Prime editing. The prime editor (PE) consists of H840A Cas9 nickase (HNH mutant) bound to M-MLV reverse transcriptase (M-MLV RT) and enables rewriting of the target sequence up to 80-bp. PE employs a modified guide RNA called prime editing guide RNA (pegRNA). PegRNA comprises four components: i) guide sequence, ii) scaffold sequence, iii) RT template, and iv) primer binding sequence. H840A nicks the target, creating an open 3’ end for M-MLV RT to initiate DNA synthesis. The newly synthesized sequence replaces the original sequence, resulting in a 5’ flap. Cellular endonucleases remove the 5’ flap, and mismatch repair mechanisms replace the desired sequence on the complementary strand.
CRISPR/Cas9 has demonstrated superior outcomes in the last two decades compared to other gene editing approaches, including zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), small interfering RNAs (shRNA/siRNA), peptide-nucleic acids, and ribozymes, in reproducing the naturally occurring Δ32 CCR5 mutation [113]. CCR5 gene editing using CRISPR-Cas9 shows 4.8 times more efficiency than TALENs [114] and is associated with increased biallelic dsDNA breaks at the CCR5 locus [115]. Indeed, multiple reports show higher efficiency, specificity, and lower cytotoxicity via CRISPR/Cas9-mediated CCR5 gene editing [114,116].
In vivo, off-target Cas9 activity raises important questions about the utility of applying this approach without further technique modifications [117]. Indeed, wild-type SpCas9-generates dsDNA breaks, leading to unwanted risks by increasing the frequency of insertions and deletions (indels). Consequently, Cas modifications are underway to enhance cleavage, specificity, and PAM recognition [118,119]. AsCpf1, a Cas12a endonuclease recognizing ‘TTTN’ PAMs, demonstrated enhanced specificity and superior efficiency compared to Cas9, with no off-target events [120]. A high-fidelity Cas9 shows promise for delivering ribonucleoprotein complexes (RNPs) with no off-target events in editing CCR5 [121]. Engineering gRNAs can increase target specificity and Cas9 activity [122,123]. Assays such as ZeoR and qEva-CRISPR can effectively test for on and off-target activity of Cas9s [124,125]. Nevertheless, preclinical ex vivo data suggest that cell editing by CRISPR circumvents many of the recognized cell-associated risks of previous techniques, off-target events, immune-reactivation, and delivery-associated issues [126].
Biological delivery of CRISPR/Cas9 components relies on several methods, including lentiviral, adenoviral, adeno-associated viral (AAV), non-viral vectors (plasmids, transposons), all-RNA CRISPR, and RNPs [127]. However, host exposure to these viruses can lead to immunoreactivity, and integrating vectors such as lentivirus allow permanent expression of Cas9, exposing cells to recurrent dsDNA breaks and increasing vulnerability to neoplastic development [128]. Moreover, since lentivirus integrates, it is also possible that these ‘random’ viral DNA insertions could occur in oncogenes, albeit this risk is minimal [129]. Adenovirus is non-integrating, but transcription of its components may lead to platelet activation and coagulopathic effects [130]. AAVs are less immunogenic; transfection can be complex, and outcomes could be more efficient. Synthetic vectors, including lipids, polymeric, and inorganic particles, are considerably easier to scale from a pharmaceutical perspective. Lipofection, electroporation, and microinjections effectively deliver CRISPR/Cas9 components to target cells; however, these methods must be thoroughly assessed before clinical use. Although RNP delivery to target cells is routinely accomplished by electroporation, newer approaches using transmembrane internalization assisted by membrane filtration (TRIAMF) [131] and induced transduction by osmocytosis and propane betaine (iTOP) [132] offer promising solutions for ex vivo manipulation of target cells, particularly HSCs. TRIAMF is a technique that delivers RNPs into HSPCs by passing RNPs and cells through a filter membrane. The TRIAMF technique retains the multilineage colony-forming capacity and engraftment potential of HSPCs. iTOP exploits micropinocytosis to uptake RNPs using non-detergent sulfobetaines, achieving efficient CRISPR editing in HSCs. iTOP may also be a viable method to scale transfections for clinical use.
Ex vivo CRISPR studies have failed to achieve comparable CCR5 disruption as CCR5-ablated lymphocytes (Table 2). In clinical studies, Xu and colleagues successfully created and engrafted CRISPR-mediated HSPCs carrying the Δ32 CCR5 mutation [133]. Although the cells were well-tolerated in patients, infusing 1 billion HSCs did not produce sufficient CD4+ cells with the CCR5 mutation—only ~5% [134,135]. Post-infusion HSPC activation is presumed as a significant clinical challenge in ex vivo cell therapy. Administration of CXCR4 antagonists in mice, monkeys, and humans [136,137] helps release HSPCs from bone-marrow stroma via blocking stromal cell-derived factors, such as SDF-1α, that interact with CXCR4 [82]. Others have reported that endogenous CD26 protease activity is involved in HSC bone marrow homing, the loss of which resulted in HSC peripheral blood mobilization [82]. Finally, cytokines such as type II IFNs promote differentiation of myeloid-derived HSCs [138], and mesenchymal stem cells can secrete angiopoietin-like 5, supporting HSC expansion in NSG mice [139].
Table 2.
CRISPR-based methods to edit CCR5.
Gene editing tool | Approach | En gra ftm ent type | Target cells | Study outcome |
---|---|---|---|---|
CRISPR-Cas9 knockout | Reproduced Δ32 mutation | in vitro | Human CD4+T cells | R5-tropic resistance was conferred to primary cells [146]. |
CRISPR-Cas9 TALENs | CRISPR and TALENs used to reproduce Δ32 mutation | in vitro | iPSCs | CRISPR showed better targeting efficiency than TALENs [115]. |
CRISPR-Cas9 knockout | Reproduced Δ32 mutation | in vitro | iPSCs, human CD4+T cells | Multiple sgRNA pairs generated CCR5 knockouts and resistance to R5-tropic HIV 116,147]. |
CRISPR-Cas9 knockout | Reproduced Δ32 mutation | in vitro | TZM-bl cells | Cas9-sgRNA-edited cells showed few off-target events, and lentiviral delivery was safe [148]. |
CRISPR-Cas9 RNP technology | Reproduced Δ32 mutation | in vitro | Human CD4+T cells | RNP-mediated delivery was safe, disrupted CCR5, and provided R5-tropic resistance [149]. |
CRISPR-Cas9 knockout | Reproduced Δ32 mutation | in vitro | A549, HeLa, primary human skeletal myoblasts | Δ32 mutation induced resistance to HIV [150]. |
CRISPR-Cas9 HDR | Reproduced Δ32 mutation | humice | HSPCs | Δ32 HSPCs conferred HIV resistance [134]. |
CRISPR-Cas9 knockout | Flanking regions of the Δ32 locus were targeted to minimize off-target events | in vitro | Human CD4+T cells, Jurkat Cells | Knockout conferred resistance to R5-tropic virus without off-target effects [151]. |
CRISPR-Cas9-tracrRNA structure modification | sgRNA structure was investigated to improve indel outcomes for safer CCR5 ablation. | in vitro | Human CD4+T cells | Modified tracrRNA enhanced CCR5 cleavage and improved indel frequency [123]. |
CRISPR-SaCas9 | S. aureus(Sa) and S. pyogenes(Sp) Cas9s were compared for CCR5 ablating efficiency | humice | CD4+T cells, HSPCs | SaCas9 showed superior CCR5 cleavage efficiency than SpCas9 [152]. |
CRISPR-Cas9 NHEJ | Series of sgRNAs were assessed for CCR5 ablating efficiency | in vitro | HSCs | CCR5 ablation conferred HIV resistance [153]. |
CRISPR-Cas9 HDR | Reproduced Δ32 mutation | NH P | HSCs | Autologous CCR5-deficient HSCs provided short-term anti-SIV protection [154]. |
TALEN and CRISPR-Cas9 | PiggyBac-mediated CRISPR to reproduce the Δ32 mutation | in vitro | iPSCs | Δ32 macrophages showed resistance to HIV [155]. |
CRISPR-Cas9 knockout | Ablated the CCR5 gene and placed X4 infected cells under ganciclovir-augmented suicidal control | in vitro | TZM cells, H7 cells | CCR5 knock-out and Tat-activated conditional X4 inhibition conferred complete resistance to HIV [156]. |
CRISPR-Cas9 knockout | Deletion of a 198-bp CCR5 fragment from exon 2 | in vitro | iPSCs from primate T cells and fibroblasts | Knockout cells prevented replication of the CCR5 tropic- SIVmac239 and -SIVmac316 [157]. |
CRISPR-Cas9 knock-in | Induced a frameshift insertion in the CCR5 gene | in vitro | HSPCs, SCs, CD4+ T cells | Knock-ins disrupted CCR5 and conferred HIV resistance [158]. |
CRISPR-Cas9 knockout | Induced CCR5 and CXCR4 single and dual knockouts | humice | Human CD4+T cells and PBMCs | Single knockouts resulted in HIV resistance, and dual knockouts inhibited CD4+T cells engraftment in BM [159]. |
Base editing | evoCDA-BE4max converted CAA, CAG, CGA, and TGG to stop codons. ABE, using different PAM-specific nCas9s, showed broader coverage in mutating start codons | in vitro | HSPCs, Human CD4+ T cells | CBE and ABE complexes inhibited R5-tropic strains. CBE also inhibited X4 tropic strains [112]. |
Prime editing | PE3max and uPEn converted C-T, disrupting CCR5 | in vitro | HEK293T cells | uPEn significantly reduced indel frequencies and ablated CCR5 [120]. |
TALENs, Transcription activator-like effector nucleases; HDR, Homology-directed repair; hu, humanized; NHP, non-human primate; NHEJ, Nonhomologous end joining; CBE, Cytosine base editor; ABE, Adenine base editor
Nevertheless, autologous transplantation of CCR5 edited cells shows limited capacity to clear the HIV latent reservoir, providing strong evidence that an allogeneic immune response is also likely required to clear viral reservoirs in ART-suppressed animals [140]. Indeed, following HSCT, latent virus was cleared which was associated with an increase in donor chimerism leading to a complete remission within 2.5 years [140]. Based on these data, it appears that CCR5-editing in autologous HSCs may not alone be sufficient to eradicate HIV-1, strongly suggesting that allogeneic immunity in the context of CCR5-based cure approaches need further evaluation. Moreover, development of additional preclinical models to further understand the mechanisms of reservoir clearance is highly warranted [141].
A major limiting factor in treating HIV with HSCT is associated toxicity with conditioning protocols [142]. Conventional conditioning procedures employ high doses of radiation and/or chemotherapy resulting in nonspecific tissue injury, potentially inducing or aggravating GVHD. However, antibody-based conditioning methods, including native antibody, radiolabeled antibody conjugates, and antibody drug conjugates result in tissue specific injury [142], suggesting that milder preparative regimens could be critical in developing better CCR5-based HSCT methods.
8. Conclusion
The introduction of the first single-tablet ARV (azidothymidine) to treat HIV infection almost four decades ago, followed by the development of subsequent ARVs, dramatically reduced HIV-associated morbidity and mortality. Today, ART allows restoration of CD4+ T cell counts with the virus becoming undetectable and non-transmissible, yet treatment interruption results in rapid viral rebound. Drug resistance, long-term ART-associated comorbid conditions, and a stable HIV latent reservoir necessitate further exploration for new curative approaches.
CCR5, in tandem with the CD4 molecule, interacts with gp120 of HIV, promoting host cell-to-virus fusion. Defective Δ32 alleles of CCR5, evolutionarily selected during the smallpox pandemic of the eighteenth century, confer significant resistance to HIV-1. Thus, allogeneic HSCTs from Δ32 donors cured several recipients and showed proof-of-concept for a Δ32-based replacement approach; yet, the Berlin patient died from malignant disease, the London patient suffered graft versus host disease complications, and the other three patients are still under observation. Allogeneic transplants were never a real option for generalized HIV treatment. However, an ex vivo intervention based on HSCTs carrying Δ32 designs may still be viable.
Several gene editing approaches, including ZFNs and TALENs, were referenced when designing ex vivo modified HIV-1-resistant SC infusions. However, the poor generation of HIV-1-resistant lymphocytes derived from CCR5-modified SCs remains challenging. The New York patient was cured of HIV-1 by donor cord blood SCs carrying Δ32 and wildtype donor adult SCs, findings which hold great promise in the pursuit of a cure. Editing CCR5 with CRISPR/Cas9 shows improved nuclease efficiency compared to previous methods; however, off-target Cas9 activity, in vivo SC activation, and required allogeneic immune responses are impeding progress into the clinic. Targeted conditioning and development of animal models to further understand the mechanisms of HIV reservoir elimination are required to develop a better HSC transplantation program.
CCR5 gene-editing using CRISPR/Cas9 to reproduce the Δ32 mutations certainly requires optimization, thorough in vitro testing, and preclinical validation. Simple and incremental advances optimizing CRISPR should allow future clinical studies with ex vivo generated allogeneic HSCTs.
Highlights.
Allogeneic hematopoietic stem cell transplant (HSCT) from Δ32 donors has been established as a cure for HIV; however, the procedure involves life-threatening risks limiting its widespread use.
CRISPR-Cas9 targeting the CCR5 gene generates autogeneic Δ32 hematopoietic stem cell grafts, potentially replacing allogeneic Δ32 HSCTs.
This review describes difficulties with antiretroviral therapy, the use of CRISPR/Cas9 to replicate the natural Δ32 mutation, and challenges with ex vivo edited hematopoietic stem cells in curing HIV.
Acknowledgments
This is manuscript number 1,071 from The Scintillon Institute. The authors would like to thank Drs. Joel Buxbaum and Jiwu Wang for their critical reading of the manuscript.
Funding
This work was supported by the California Institute of Regenerative Medicine (CIRM) [Grant No. DISC2-13510] and the National Institutes of Health (NIH) [Grant No. 7R01AI150381-02].
Footnotes
Declaration of Competing Interest
None.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Number of people dying from HIV-related causes, (n.d.). https://www.who.int/data/gho/data/indicators/indicator-details/GHO/number-of-deaths-due-to-hiv-aids (accessed February 20, 2023).
- [2].FDA-Approved HIV Medicines | NIH, (n.d.). https://hivinfo.nih.gov/understanding-hiv/fact-sheets/fda-approved-hiv-medicines (accessed February 20, 2023). [Google Scholar]
- [3].Cohen MS, Chen YQ, McCauley M, Gamble T, Hosseinipour MC, Kumarasamy N, Hakim JG, Kumwenda J, Grinsztejn B, Pilotto JHS, Godbole SV, Chariyalertsak S, Santos BR, Mayer KH, Hoffman IF, Eshleman SH, Piwowar-Manning E, Cottle L, Zhang XC, Makhema J, Mills LA, Panchia R, Faesen S, Eron J, Gallant J, Havlir D, Swindells S, Elharrar V, Burns D, Taha TE, Nielsen-Saines K, Celentano DD, Essex M, Hudelson SE, Redd AD, Fleming TR, Antiretroviral Therapy for the Prevention of HIV-1 Transmission, N. Engl. J. Med. 375 (2016) 830–839. 10.1056/NEJMoa1600693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Estimated antiretroviral therapy coverage among people living with HIV (%), (n.d.). https://www.who.int/data/gho/data/indicators/indicator-details/GHO/estimated-antiretroviral-therapy-coverage-among-people-living-with-hiv-(−) (accessed February 21, 2023).
- [5].Hosseinipour MC, Gupta RK, Van Zyl G, Eron JJ, Nachega JB, Emergence of HIV Drug Resistance During First- and Second-Line Antiretroviral Therapy in Resource-Limited Settings, J. Infect. Dis. 207 (2013) S49–S56. 10.1093/infdis/jit107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Osborne O, Peyravian N, Nair M, Daunert S, Toborek M, The Paradox of HIV Blood–Brain Barrier Penetrance and Antiretroviral Drug Delivery Deficiencies, Trends Neurosci. 43 (2020) 695–708. 10.1016/j.tins.2020.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Warren JA, Zhou S, Xu Y, Moeser MJ, MacMillan DR, Council O, Kirchherr J, Sung JM, Roan NR, Adimora AA, Joseph S, Kuruc JD, Gay CL, Margolis DM, Archin N, Brumme ZL, Swanstrom R, Goonetilleke N, The HIV-1 latent reservoir is largely sensitive to circulating T cells, ELife. 9 (2020) e57246. 10.7554/eLife.57246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, Quinn TC, Chadwick K, Margolick J, Brookmeyer R, Gallant J, Markowitz M, Ho DD, Richman DD, Siliciano RF, Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy, Science. 278 (1997) 1295–1300. 10.1126/SCIENCE.278.5341.1295. [DOI] [PubMed] [Google Scholar]
- [9].Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K, Pierson T, Smith K, Lisziewicz J, Lori F, Flexner C, Quinn TC, Chaisson RE, Rosenberg E, Walker B, Gange S, Gallant J, Siliciano RF, Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy, Nat. Med. 5 (1999) 512–517. 10.1038/8394. [DOI] [PubMed] [Google Scholar]
- [10].Kwong PD, Wyatt R, Robinson J, Sweet RW, Sodroski J, Hendrickson WA, Structure of an HIV gp120 envelope glycoprotein in complex with the CD4 receptor and a neutralizing human antibody, Nature. 393 (1998) 648–659. 10.1038/31405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, Marzio PD, Marmon S, Sutton RE, Hill CM, Davis CB, Peiper SC, Schall TJ, Littman DR, Landau NR, Identification of a major co-receptor for primary isolates of HIV-1, Nature. 381 (1996) 661–666. 10.1038/381661a0. [DOI] [PubMed] [Google Scholar]
- [12].Samson M, Libert F, Doranz BJ, Rucker J, Liesnard C, Farber CM, Saragosti S, Lapoumeroulie C, Cognaux J, Forceille C, Muyldermans G, Verhofstede C, Burtonboy G, Georges M, Imai T, Rana S, Yi Y, Smyth RJ, Collman RG, Doms RW, Vassart G, Parmentier M, Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene, Nature. 382 (1996) 722–725. 10.1038/382722a0. [DOI] [PubMed] [Google Scholar]
- [13].Ni J, Wang D, Wang S, The CCR5-Delta32 Genetic Polymorphism and HIV-1 Infection Susceptibility: a Meta-analysis, Open Med. 13 (2018) 467–474. 10.1515/med-2018-0062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Hütter G, Nowak D, Mossner M, Ganepola S, Müssig A, Allers K, Schneider T, Hofmann J, Kücherer C, Blau O, Blau IW, Hofmann WK, Thiel E, Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation, N. Engl. J. Med. 360 (2009) 692–698. 10.1056/NEJMoa0802905. [DOI] [PubMed] [Google Scholar]
- [15].Gupta RK, Abdul-Jawad S, McCoy LE, Mok HP, Peppa D, Salgado M, Martinez-Picado J, Nijhuis M, Wensing AMJ, Lee H, Grant P, Nastouli E, Lambert J, Pace M, Salasc F, Monit C, Innes AJ, Muir L, Waters L, Frater J, Lever AML, Edwards SG, Gabriel IH, Olavarria E, HIV-1 remission following CCR5Δ32/Δ32 haematopoietic stem-cell transplantation, Nature. 568 (2019) 244–248. 10.1038/s41586-019-1027-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].(n.d.). https://programme.aids2022.org/Abstract/Abstract/?abstractid=12508 (accessed May 13, 2023).
- [17].Jensen B-EO, Knops E, Cords L, Lübke N, Salgado M, Busman-Sahay K, Estes JD, Huyveneers LEP, Perdomo-Celis F, Wittner M, Gálvez C, Mummert C, Passaes C, Eberhard JM, Münk C, Hauber I, Hauber J, Heger E, De Clercq J, Vandekerckhove L, Bergmann S, Dunay GA, Klein F, Häussinger D, Fischer JC, Nachtkamp K, Timm J, Kaiser R, Harrer T, Luedde T, Nijhuis M, Sáez-Cirión A, Schulze zur Wiesch J, Wensing AMJ, Martinez-Picado J, Kobbe G, In-depth virological and immunological characterization of HIV-1 cure after CCR5Δ32/Δ32 allogeneic hematopoietic stem cell transplantation, Nat. Med. (2023) 1–5. 10.1038/s41591-023-02213-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Hsu J, Van Besien K, Glesby MJ, Pahwa S, Coletti A, Warshaw MG, Petz L, Moore TB, Chen YH, Pallikkuth S, Dhummakupt A, Cortado R, Golner A, Bone F, Baldo M, Riches M, Mellors JW, Tobin NH, Browning R, Persaud D, Bryson Y, International Maternal Pediatric Adolescent AIDS Clinical Trials Network (IMPAACT) P1107 Team, HIV-1 remission and possible cure in a woman after haplo-cord blood transplant, Cell. 186 (2023) 1115–1126.e8. 10.1016/j.cell.2023.02.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Martin GE, Sen DR, Pace M, Robinson N, Meyerowitz J, Adland E, Thornhill JP, Jones M, Ogbe A, Parolini L, Olejniczak N, Zacharopoulou P, Brown H, Willberg CB, Nwokolo N, Fox J, Fidler S, Haining WN, Frater J, Epigenetic Features of HIV-Induced T-Cell Exhaustion Persist Despite Early Antiretroviral Therapy, Front. Immunol. 12 (2021). 10.3389/fimmu.2021.647688 (accessed February 21, 2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Ways HIV Can Be Transmitted | HIV Transmission | HIV Basics | HIV/AIDS | CDC, (2022). https://www.cdc.gov/hiv/basics/hiv-transmission/ways-people-get-hiv.html (accessed February 20, 2023). [Google Scholar]
- [21].WHO clinical staging of HIV disease in adults, adolescents and children, World Health Organization, 2016. https://www.ncbi.nlm.nih.gov/books/NBK374293/ (accessed February 20, 2023). [Google Scholar]
- [22].Antiretroviral Drugs for Treatment and Prevention of HIV Infection in Adults: 2022 Recommendations of the International Antiviral Society–USA Panel | JAMA | JAMA Network, (n.d.). https://jamanetwork.com/journals/jama/fullarticle/2799240 (accessed February 20, 2023). [DOI] [PubMed] [Google Scholar]
- [23].Li G, Wang Y, De Clercq E, Approved HIV reverse transcriptase inhibitors in the past decade, Acta Pharm. Sin. B. 12 (2022) 1567–1590. 10.1016/j.apsb.2021.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Patick AK, Potts KE, Protease inhibitors as antiviral agents, Clin. Microbiol. Rev. 11 (1998) 614–627. 10.1128/CMR.11.4.614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Singh IP, Chauthe SK, Small molecule HIV entry inhibitors: Part II. Attachment and fusion inhibitors: 2004–2010, Expert Opin. Ther. Pat. 21 (2011) 399–416. 10.1517/13543776.2011.550876. [DOI] [PubMed] [Google Scholar]
- [26].Shaheen F, Collman RG, Co-receptor antagonists as HIV-1 entry inhibitors, Curr. Opin. Infect. Dis. 17 (2004) 7–16. 10.1097/00001432-200402000-00003. [DOI] [PubMed] [Google Scholar]
- [27].Zhao AV, Crutchley RD, Guduru RC, Ton K, Lam T, Min AC, A clinical review of HIV integrase strand transfer inhibitors (INSTIs) for the prevention and treatment of HIV-1 infection, Retrovirology. 19 (2022) 22. 10.1186/s12977-022-00608-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Governa P, Manetti F, Recent research results have converted gp120 binders to a therapeutic option for the treatment of HIV-1 infection. A medicinal chemistry point of view, Eur. J. Med. Chem. 229 (2022) 114078. 10.1016/j.ejmech.2021.114078. [DOI] [PubMed] [Google Scholar]
- [29].Chandra I, Prabhu SV, Nayak C, Singh SK, E-pharmacophore based screening to identify potential HIV-1 gp120 and CD4 interaction blockers for wild and mutant types, SAR QSAR Environ. Res. 32 (2021) 353–377. 10.1080/1062936X.2021.1901310. [DOI] [PubMed] [Google Scholar]
- [30].De Castro N, Brun A, Sellier P, Hamet G, Mechaï F, Garrait V, Chabrol A, Bouldouyre M-A, Froguel E, Troisvallets D, Caraux-Paz P, Delaugerre C, Rozenbaum W, Molina J-M, Safety and efficacy of switching to elvitegravir, cobicistat, emtricitabine, tenofovir disoproxil fumarate in treatment-experienced people with HIV: a multicenter cohort study, AIDS Res. Ther. 20 (2023) 1. 10.1186/s12981-022-00499-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Paudel M, Prajapati G, Buysman EK, Goswami S, Mao J, McNiff K, Kumar P, Comorbidity and comedication burden among people living with HIV in the United States, Curr. Med. Res. Opin. 38 (2022) 1443–1450. 10.1080/03007995.2022.2088714. [DOI] [PubMed] [Google Scholar]
- [32].Montessori V, Press N, Harris M, Akagi L, Montaner JSG, Adverse effects of antiretroviral therapy for HIV infection, CMAJ Can. Med. Assoc. J. 170 (2004) 229–238. [PMC free article] [PubMed] [Google Scholar]
- [33].Yeo JY, Goh G-R, Su CT-T, Gan SK-E, The Determination of HIV-1 RT Mutation Rate, Its Possible Allosteric Effects, and Its Implications on Drug Resistance, Viruses. 12 (2020) 297. 10.3390/v12030297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Leitner T, Phylogenetics in HIV transmission: taking within-host diversity into account, Curr. Opin. HIV AIDS. 14 (2019) 181. 10.1097/COH.0000000000000536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].de Azevedo SSD, Delatorre E, Gaido CM, Silva-de-Jesus C, Guimarães ML, Couto-Fernandez JC, Morgado MG, HIV-1 Diversity and Drug Resistance in Treatment-Naïve Children and Adolescents from Rio de Janeiro, Brazil, Viruses. 14 (2022) 1761. 10.3390/v14081761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Cadosch D, Bonhoeffer S, Kouyos R, Assessing the impact of adherence to anti-retroviral therapy on treatment failure and resistance evolution in HIV, J. R. Soc. Interface. 9 (2012) 2309–2320. 10.1098/rsif.2012.0127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Wang Y, Liu M, Lu Q, Farrell M, Lappin JM, Shi J, Lu L, Bao Y, Global prevalence and burden of HIV-associated neurocognitive disorder: A meta-analysis, Neurology. 95 (2020) e2610–e2621. 10.1212/WNL.0000000000010752. [DOI] [PubMed] [Google Scholar]
- [38].León-Rivera R, Veenstra M, Donoso M, Tell E, Eugenin EA, Morgello S, Berman JW, Central Nervous System (CNS) Viral Seeding by Mature Monocytes and Potential Therapies To Reduce CNS Viral Reservoirs in the cART Era, MBio. 12 (2021) e03633–20. 10.1128/mBio.03633-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Kandel SR, Luo X, He JJ, Nef inhibits HIV transcription and gene expression in astrocytes and HIV transmission from astrocytes to CD4+ T cells, J. Neurovirol. 28 (2022) 552–565. 10.1007/s13365-022-01091-2. [DOI] [PubMed] [Google Scholar]
- [40].Veenstra M, León-Rivera R, Li M, Gama L, Clements JE, Berman JW, Mechanisms of CNS Viral Seeding by HIV+ CD14+ CD16+ Monocytes: Establishment and Reseeding of Viral Reservoirs Contributing to HIV-Associated Neurocognitive Disorders, MBio. 8 (2017) e01280–17. 10.1128/mBio.01280-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Gumbs SBH, Berdenis van Berlekom A, Kübler R, Schipper PJ, Gharu L, Boks MP, Ormel PR, Wensing AMJ, de Witte LD, Nijhuis M, Characterization of HIV-1 Infection in Microglia-Containing Human Cerebral Organoids, Viruses. 14 (2022) 829. 10.3390/v14040829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Li G-H, Anderson C, Jaeger L, Do T, Major EO, Nath A, Cell-to-cell contact facilitates HIV transmission from lymphocytes to astrocytes via CXCR4, AIDS Lond. Engl. 29 (2015) 755–766. 10.1097/QAD.0000000000000605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Bertrand L, Dygert L, Toborek M, Antiretroviral Treatment with Efavirenz Disrupts the Blood-Brain Barrier Integrity and Increases Stroke Severity, Sci. Rep. 6 (2016) 39738. 10.1038/srep39738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Pokharkar V, Patil-Gadhe A, Palla P, Efavirenz loaded nanostructured lipid carrier engineered for brain targeting through intranasal route: In-vivo pharmacokinetic and toxicity study, Biomed. Pharmacother. Biomedecine Pharmacother. 94 (2017) 150–164. 10.1016/j.biopha.2017.07.067. [DOI] [PubMed] [Google Scholar]
- [45].Rimawi BH, Johnson E, Rajakumar A, Tao S, Jiang Y, Gillespie S, Schinazi RF, Mirochnick M, Badell ML, Chakraborty R, Pharmacokinetics and Placental Transfer of Elvitegravir, Dolutegravir, and Other Antiretrovirals during Pregnancy, Antimicrob. Agents Chemother. 61 (2017) e02213–16. 10.1128/AAC.02213-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Agabu A, Baughman AL, Fischer-Walker C, de Klerk M, Mutenda N, Rusberg F, Diergaardt D, Pentikainen N, Sawadogo S, Agolory S, Dinh T-H, National-level effectiveness of ART to prevent early mother to child transmission of HIV in Namibia, PLoS ONE. 15 (2020) e0233341. 10.1371/journal.pone.0233341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Geijtenbeek TB, Kwon DS, Torensma R, van Vliet SJ, van Duijnhoven GC, Middel J, Cornelissen IL, Nottet HS, KewalRamani VN, Littman DR, Figdor CG, van Kooyk Y, DC-SIGN, a dendritic cell-specific HIV-1-binding protein that enhances trans-infection of T cells, Cell. 100 (2000) 587–597. 10.1016/s0092-8674(00)80694-7. [DOI] [PubMed] [Google Scholar]
- [48].de Witte L, Bobardt M, Chatterji U, Degeest G, David G, Geijtenbeek TBH, Gallay P, Syndecan-3 is a dendritic cell-specific attachment receptor for HIV-1, Proc. Natl. Acad. Sci. U. S. A. 104 (2007) 19464–19469. 10.1073/pnas.0703747104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Izquierdo-Useros N, Lorizate M, Puertas MC, Rodriguez-Plata MT, Zangger N, Erikson E, Pino M, Erkizia I, Glass B, Clotet B, Keppler OT, Telenti A, Kräusslich H-G, Martinez-Picado J, Siglec-1 is a novel dendritic cell receptor that mediates HIV-1 trans-infection through recognition of viral membrane gangliosides, PLoS Biol. 10 (2012) e1001448. 10.1371/journal.pbio.1001448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Bushman FD, Fujiwara T, Craigie R, Retroviral DNA integration directed by HIV integration protein in vitro, Science. 249 (1990) 1555–1558. 10.1126/science.2171144. [DOI] [PubMed] [Google Scholar]
- [51].Rottman JB, Ganley KP, Williams K, Wu L, Mackay CR, Ringler DJ, Cellular localization of the chemokine receptor CCR5. Correlation to cellular targets of HIV-1 infection., Am. J. Pathol. 151 (1997) 1341–1351. [PMC free article] [PubMed] [Google Scholar]
- [52].Chua BA, Ngo JA, Situ K, Ramirez CM, Nakano H, Morizono K, Protein S and Gas6 induce efferocytosis of HIV-1-infected cells, Virology. 515 (2018) 176–190. 10.1016/j.virol.2017.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Okafo G, Valdebenito S, Donoso M, Luu R, Ajasin D, Prideaux B, Gorantha S, Eugenin EA, Role of tunneling nanotube (TNT)-like structures during the early events of HIV-infection: novel features of tissue compartmentalization and mechanism of HIV spread., J. Immunol. Baltim. Md 1950. 205 (2020) 2726–2741. 10.4049/jimmunol.2000803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Hammonds JE, Beeman N, Ding L, Takushi S, Francis AC, Wang J-J, Melikyan GB, Spearman P, Siglec-1 initiates formation of the virus-containing compartment and enhances macrophage-to-T cell transmission of HIV-1, PLoS Pathog. 13 (2017) e1006181. 10.1371/journal.ppat.1006181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Wexler-Cohen Y, Ashkenazi A, Viard M, Blumenthal R, Shai Y, Virus-cell and cell-cell fusion mediated by the HIV-1 envelope glycoprotein is inhibited by short gp41 N-terminal membrane-anchored peptides lacking the critical pocket domain, FASEB J. 24 (2010) 4196–4202. 10.1096/fj.09-151704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Wang L, Izadmehr S, Kamau E, Kong X-P, Chen BK, Sequential trafficking of Env and Gag to HIV-1 T cell virological synapses revealed by live imaging, Retrovirology. 16 (2019) 2. 10.1186/s12977-019-0464-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Graziano F, Aimola G, Forlani G, Turrini F, Accolla R, Vicenzi E, Poli G, D-105 Reversible HIV-1 Latency Induced in Primary Human Monocyte-Derived Macrophages by Repeated M1 Polarization, JAIDS J. Acquir. Immune Defic. Syndr. 77 (2018) 40. 10.1097/01.qai.0000532597.67178.f2. [DOI] [Google Scholar]
- [58].Chu H, Wang J-J, Qi M, Yoon J-J, Chen X, Wen X, Hammonds J, Ding L, Spearman P, Tetherin/BST-2 is essential for the formation of the intracellular virus-containing compartment in HIV-infected macrophages, Cell Host Microbe. 12 (2012) 360–372. 10.1016/j.chom.2012.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Rhodes JW, Botting RA, Bertram KM, Vine EE, Rana H, Baharlou H, Vegh P, O’Neil TR, Ashhurst AS, Fletcher J, Parnell GP, Graham JD, Nasr N, Lim JJK, Barnouti L, Haertsch P, Gosselink MP, Di Re A, Reza F, Ctercteko G, Jenkins GJ, Brooks AJ, Patrick E, Byrne SN, Hunter E, Haniffa MA, Cunningham AL, Harman AN, Human anogenital monocyte-derived dendritic cells and langerin+cDC2 are major HIV target cells, Nat. Commun. 12 (2021) 2147. 10.1038/s41467-021-22375-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, Boucher G, Boulassel M-R, Ghattas G, Brenchley JM, Schacker TW, Hill BJ, Douek DC, Routy J-P, Haddad EK, Sékaly R-P, HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation, Nat. Med. 15 (2009) 893–900. 10.1038/nm.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Chun T-W, Engel D, Mizell SB, Ehler LA, Fauci AS, Induction of HIV-1 Replication in Latently Infected CD4+ T Cells Using a Combination of Cytokines, J. Exp. Med. 188 (1998) 83–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Elrefaei M, Burke CM, Baker CAR, Jones NG, Bousheri S, Bangsberg DR, Cao H, TGF-β and IL-10 Production by HIV-Specific CD8+ T Cells Is Regulated by CTLA-4 Signaling on CD4+ T Cells, PLOS ONE. 4 (2009) e8194. 10.1371/journal.pone.0008194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Said EA, Dupuy FP, Trautmann L, Zhang Y, Shi Y, El-Far M, Hill BJ, Noto A, Ancuta P, Peretz Y, Fonseca SG, Van Grevenynghe J, Boulassel MR, Bruneau J, Shoukry NH, Routy J-P, Douek DC, Haddad EK, Sekaly R-P, Programmed death-1-induced interleukin-10 production by monocytes impairs CD4+ T cell activation during HIV infection, Nat. Med. 16 (2010) 452–459. 10.1038/nm.2106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Cameron PU, Saleh S, Sallmann G, Solomon A, Wightman F, Evans VA, Boucher G, Haddad EK, Sekaly R-P, Harman AN, Anderson JL, Jones KL, Mak J, Cunningham AL, Jaworowski A, Lewin SR, Establishment of HIV-1 latency in resting CD4+ T cells depends on chemokine-induced changes in the actin cytoskeleton, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) 16934–16939. 10.1073/pnas.1002894107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Unutmaz D, KewalRamani VN, Marmon S, Littman DR, Cytokine Signals Are Sufficient for HIV-1 Infection of Resting Human T Lymphocytes, J. Exp. Med. 189 (1999) 1735–1746. 10.1084/jem.189.11.1735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Riou C, Yassine-Diab B, Van grevenynghe J, Somogyi R, Greller LD, Gagnon D, Gimmig S, Wilkinson P, Shi Y, Cameron MJ, Campos-Gonzalez R, Balderas RS, Kelvin D, Sekaly R-P, Haddad EK, Convergence of TCR and cytokine signaling leads to FOXO3a phosphorylation and drives the survival of CD4+ central memory T cells, J. Exp. Med. 204 (2007) 79–91. 10.1084/jem.20061681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Mummidi S, Ahuja SS, McDaniel BL, Ahuja SK, The human CC chemokine receptor 5 (CCR5) gene. Multiple transcripts with 5’-end heterogeneity, dual promoter usage, and evidence for polymorphisms within the regulatory regions and noncoding exons, J. Biol. Chem. 272 (1997) 30662–30671. 10.1074/jbc.272.49.30662. [DOI] [PubMed] [Google Scholar]
- [68].Kulkarni S, Lied A, Kulkarni V, Rucevic M, Martin MP, Walker-Sperling V, Anderson SK, Ewy R, Singh S, Nguyen H, McLaren PJ, Viard M, Naranbhai V, Zou C, Lin Z, Gatanaga H, Oka S, Takiguchi M, Thio CL, Margolick J, Kirk GD, Goedert JJ, Hoots WK, Deeks SG, Haas DW, Michael N, Walker B, Le Gall S, Chowdhury FZ, Yu XG, Carrington M, CCR5AS lncRNA variation differentially regulates CCR5, influencing HIV disease outcome, Nat. Immunol. 20 (2019) 824–834. 10.1038/s41590-019-0406-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Mehlotra RK, New Knowledge About CCR5, HIV Infection, and Disease Progression: Is “Old” Still Valuable?, AIDS Res. Hum. Retroviruses. 36 (2020) 795–799. 10.1089/AID.2020.0060. [DOI] [PubMed] [Google Scholar]
- [70].Scurci I, Martins E, Hartley O, CCR5: Established paradigms and new frontiers for a “celebrity” chemokine receptor, Cytokine. 109 (2018) 81–93. 10.1016/j.cyto.2018.02.018. [DOI] [PubMed] [Google Scholar]
- [71].Edinger AL, Amedee A, Miller K, Doranz BJ, Endres M, Sharron M, Samson M, Lu Z, Clements JE, Murphey-Corb M, Peiper SC, Parmentier M, Broder CC, Doms RW, Differential utilization of CCR5 by macrophage and T cell tropic simian immunodeficiency virus strains, Proc. Natl. Acad. Sci. 94 (1997) 4005–4010. 10.1073/pnas.94.8.4005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Yang J, Zhang Y-W, Huang J-F, Zhang Y-P, Liu C-Q, Structure analysis of CCR5 from human and primates, J. Mol. Struct. THEOCHEM. 505 (2000) 199–210. 10.1016/S0166-1280(99)00393-0. [DOI] [Google Scholar]
- [73].Duma L, Häussinger D, Rogowski M, Lusso P, Grzesiek S, Recognition of RANTES by extracellular parts of the CCR5 receptor, J. Mol. Biol. 365 (2007) 1063–1075. 10.1016/j.jmb.2006.10.040. [DOI] [PubMed] [Google Scholar]
- [74].Howard OMZ, Shirakawa A-K, Turpin JA, Maynard A, Tobin GJ, Carrington M, Oppenheim JJ, Dean M, Naturally Occurring CCR5 Extracellular and Transmembrane Domain Variants Affect HIV-1 Co-receptor and Ligand Binding Function*, J. Biol. Chem. 274 (1999) 16228–16234. 10.1074/jbc.274.23.16228. [DOI] [PubMed] [Google Scholar]
- [75].Tan Q, Zhu Y, Li J, Chen Z, Han GW, Kufareva I, Li T, Ma L, Fenalti G, Li J, Zhang W, Xie X, Yang H, Jiang H, Cherezov V, Liu H, Stevens RC, Zhao Q, Wu B, Structure of the CCR5 Chemokine Receptor–HIV Entry Inhibitor Maraviroc Complex, Science. 341 (2013) 1387–1390. 10.1126/science.1241475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Signoret N, Pelchen-Matthews A, Mack M, Proudfoot AE, Marsh M, Endocytosis and recycling of the HIV coreceptor CCR5, J. Cell Biol. 151 (2000) 1281–1294. 10.1083/jcb.151.6.1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Moseri A, Akabayov SR, Cohen LS, Naider F, Anglister J, Multiple binding modes of an N-terminal CCR5-peptide in complex with HIV-1 gp120, FEBS J. 289 (2022) 3132–3147. 10.1111/febs.16328. [DOI] [PubMed] [Google Scholar]
- [78].Jacquemard C, Koensgen F, Colin P, Lagane B, Kellenberger E, Modeling of CCR5 Recognition by HIV-1 gp120: How the Viral Protein Exploits the Conformational Plasticity of the Coreceptor, Viruses. 13 (2021) 1395. 10.3390/v13071395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Benkirane M, Jin D-Y, Chun RF, Koup RA, Jeang K-T, Mechanism of Transdominant Inhibition of CCR5-mediated HIV-1 Infection by ccr5Δ32*, J. Biol. Chem. 272 (1997) 30603–30606. 10.1074/jbc.272.49.30603. [DOI] [PubMed] [Google Scholar]
- [80].Siciliano SJ, Kuhmann SE, Weng Y, Madani N, Springer MS, Lineberger JE, Danzeisen R, Miller MD, Kavanaugh MP, DeMartino JA, Kabat D, A Critical Site in the Core of the CCR5 Chemokine Receptor Required for Binding and Infectivity of Human Immunodeficiency Virus Type 1*, J. Biol. Chem. 274 (1999) 1905–1913. 10.1074/jbc.274.4.1905. [DOI] [PubMed] [Google Scholar]
- [81].Novembre J, Galvani AP, Slatkin M, The Geographic Spread of the CCR5 Δ32 HIV-Resistance Allele, PLoS Biol. 3 (2005) e339. 10.1371/journal.pbio.0030339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Stumpf MPH, Wilkinson-Herbots HM, Allelic histories: positive selection on a HIV-resistance allele, Trends Ecol. Evol. 19 (2004) 166–168. 10.1016/j.tree.2004.02.001. [DOI] [PubMed] [Google Scholar]
- [83].Lucotte G, Frequencies of 32 base pair deletion of the (Delta 32) allele of the CCR5 HIV-1 co-receptor gene in Caucasians: a comparative analysis, Infect. Genet. Evol. J. Mol. Epidemiol. Evol. Genet. Infect. Dis. 1 (2002) 201–205. 10.1016/s1567-1348(02)00027-8. [DOI] [PubMed] [Google Scholar]
- [84].Parmentier M, CCR5 and HIV Infection, a View from Brussels, Front. Immunol. 6 (2015). 10.3389/fimmu.2015.00295 (accessed February 23, 2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Galvani AP, Slatkin M, Evaluating plague and smallpox as historical selective pressures for the CCR5-Δ32 HIV-resistance allele, Proc. Natl. Acad. Sci. 100 (2003) 15276–15279. 10.1073/pnas.2435085100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Hubacek JA, Dusek L, Majek O, Adamek V, Cervinkova T, Dlouha D, Pavel J, Adamkova V, CCR5Delta32 deletion as a protective factor in Czech first-wave COVID-19 subjects, Physiol. Res. 70 (2021) 111–115. 10.33549/physiolres.934647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Mladinich MC, Conde JN, Schutt WR, Sohn S-Y, Mackow ER, Blockade of Autocrine CCL5 Responses Inhibits Zika Virus Persistence and Spread in Human Brain Microvascular Endothelial Cells, MBio. 12 (2021) e01962–21. 10.1128/mBio.01962-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Marques RE, Guabiraba R, Del Sarto JL, Rocha RF, Queiroz AL, Cisalpino D, Marques PE, Pacca CC, Fagundes CT, Menezes GB, Nogueira ML, Souza DG, Teixeira MM, Dengue virus requires the CC-chemokine receptor CCR5 for replication and infection development, Immunology. 145 (2015) 583–596. 10.1111/imm.12476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Calzada JE, Nieto A, Beraún Y, Martín J, Chemokine receptor CCR5 polymorphisms and Chagas’ disease cardiomyopathy, Tissue Antigens. 58 (2001) 154–158. 10.1034/j.1399-0039.2001.580302.x. [DOI] [PubMed] [Google Scholar]
- [90].Joy MT, Assayag EB, Shabashov-Stone D, Liraz-Zaltsman S, Mazzitelli J, Arenas M, Abduljawad N, Kliper E, Korczyn AD, Thareja NS, Kesner EL, Zhou M, Huang S, Silva TK, Katz N, Bornstein NM, Silva AJ, Shohami E, Carmichael ST, CCR5 Is a Therapeutic Target for Recovery after Stroke and Traumatic Brain Injury, Cell. 176 (2019) 1143–1157.e13. 10.1016/j.cell.2019.01.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Glass WG, McDermott DH, Lim JK, Lekhong S, Yu SF, Frank WA, Pape J, Cheshier RC, Murphy PM, CCR5 deficiency increases risk of symptomatic West Nile virus infection, J. Exp. Med. 203 (2006) 35–40. 10.1084/jem.20051970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Falcon A, Cuevas MT, Rodriguez-Frandsen A, Reyes N, Pozo F, Moreno S, Ledesma J, Martínez-Alarcón J, Nieto A, Casas I, CCR5 deficiency predisposes to fatal outcome in influenza virus infection, J. Gen. Virol. 96 (2015) 2074–2078. 10.1099/vir.0.000165. [DOI] [PubMed] [Google Scholar]
- [93].Lee J-W, Hoshino A, Inoue K, Saitou T, Uehara S, Kobayashi Y, Ueha S, Matsushima K, Yamaguchi A, Imai Y, Iimura T, The HIV co-receptor CCR5 regulates osteoclast function, Nat. Commun. 8 (2017) 2226. 10.1038/s41467-017-02368-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Nagler A, Mohty M, In 2022, which is preferred: haploidentical or cord transplant?, Hematol. Am. Soc. Hematol. Educ. Program. 2022 (2022) 64–73. 10.1182/hematology.2022000327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Konuma T, Kato S, Oiwa-Monna M, Tanoue S, Ogawa M, Isobe M, Tojo A, Takahashi S, Cryopreserved CD34+ Cell Dose, but Not Total Nucleated Cell Dose, Influences Hematopoietic Recovery and Extensive Chronic Graft-versus-Host Disease after Single-Unit Cord Blood Transplantation in Adult Patients, Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 23 (2017) 1142–1150. 10.1016/j.bbmt.2017.03.036. [DOI] [PubMed] [Google Scholar]
- [96].Gutman JA, Riddell SR, McGoldrick S, Delaney C, Double unit cord blood transplantation: Who wins-and why do we care?, Chimerism. 1 (2010) 21–22. 10.4161/chim.1.1.12141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Connell BJ, Hermans LE, Wensing AMJ, Schellens I, Schipper PJ, van Ham PM, de Jong DTCM, Otto S, Mathe T, Moraba R, Borghans JAM, Papathanasopoulos MA, Kruize Z, Venter FWD, Kootstra NA, Tempelman H, Tesselaar K, Nijhuis M, Immune activation correlates with and predicts CXCR4 co-receptor tropism switch in HIV-1 infection, Sci. Rep. 10 (2020) 15866. 10.1038/s41598-020-71699-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Singh A, Besson G, Mobasher A, Collman RG, Patterns of Chemokine Receptor Fusion Cofactor Utilization by Human Immunodeficiency Virus Type 1 Variants from the Lungs and Blood, J. Virol. 73 (1999) 6680–6690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Maeda Y, Takemura T, Chikata T, Kuwata T, Terasawa H, Fujimoto R, Kuse N, Akahoshi T, Murakoshi H, Tran GV, Zhang Y, Pham CH, Pham AHQ, Monde K, Sawa T, Matsushita S, Nguyen TV, Nguyen KV, Hasebe F, Yamashiro T, Takiguchi M, Existence of Replication-Competent Minor Variants with Different Coreceptor Usage in Plasma from HIV-1-Infected Individuals, J. Virol. 94 (2020) e00193–20. 10.1128/JVI.00193-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Fernàndez G, Llano A, Esgleas M, Clotet B, Esté JA, Martínez MA, Purifying selection of CCR5-tropic human immunodeficiency virus type 1 variants in AIDS subjects that have developed syncytium-inducing, CXCR4-tropic viruses, J. Gen. Virol. 87 (2006) 1285–1294. 10.1099/vir.0.81722-0. [DOI] [PubMed] [Google Scholar]
- [101].Akondy RS, Fitch M, Edupuganti S, Yang S, Kissick HT, Li KW, Youngblood BA, Abdelsamed HA, McGuire DJ, Cohen KW, Alexe G, Nagar S, McCausland MM, Gupta S, Tata P, Haining WN, McElrath MJ, Zhang D, Hu B, Greenleaf WJ, Goronzy JJ, Mulligan MJ, Hellerstein M, Ahmed R, Origin and differentiation of human memory CD8 T cells after vaccination, Nature. 552 (2017) 362–367. 10.1038/nature24633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Yang X, Jiao Y, Wang R, Ji Y, Zhang H, Zhang Y, Chen D, Zhang T, Wu H, High CCR5 Density on Central Memory CD4+ T Cells in Acute HIV-1 Infection Is Mostly Associated with Rapid Disease Progression, PLOS ONE. 7 (2012) e49526. 10.1371/journal.pone.0049526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Brumme ZL, Goodrich J, Mayer HB, Brumme CJ, Henrick BM, Wynhoven B, Asselin JJ, Cheung PK, Hogg RS, Montaner JSG, Harrigan PR, Molecular and clinical epidemiology of CXCR4-using HIV-1 in a large population of antiretroviral-naive individuals, J. Infect. Dis. 192 (2005) 466–474. 10.1086/431519. [DOI] [PubMed] [Google Scholar]
- [104].Xu L, Wang J, Liu Y, Xie L, Su B, Mou D, Wang L, Liu T, Wang X, Zhang B, Zhao L, Hu L, Ning H, Zhang Y, Deng K, Liu L, Lu X, Zhang T, Xu J, Li C, Wu H, Deng H, Chen H, CRISPR-Edited Stem Cells in a Patient with HIV and Acute Lymphocytic Leukemia., N. Engl. J. Med. 381 (2019) 1240–1247. 10.1056/NEJMoa1817426. [DOI] [PubMed] [Google Scholar]
- [105].Liu X, Zhang Y, Cheng C, Cheng AW, Zhang X, Li N, Xia C, Wei X, Liu X, Wang H, CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells, Cell Res. 27 (2017) 154–157. 10.1038/cr.2016.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E, A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity, Science. 337 (2012) 816–821. 10.1126/science.1225829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Sternberg SH, Redding S, Jinek M, Greene EC, Doudna JA, DNA interrogation by the CRISPR RNA-guided endonuclease Cas9, Nature. 507 (2014) 62–67. 10.1038/nature13011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Artegiani B, Hendriks D, Beumer J, Kok R, Zheng X, Joore I, Chuva S de Sousa Lopes, J. van Zon, S. Tans, H. Clevers, Fast and efficient generation of knock-in human organoids using homology-independent CRISPR-Cas9 precision genome editing, Nat. Cell Biol. 22 (2020) 321–331. 10.1038/s41556-020-0472-5. [DOI] [PubMed] [Google Scholar]
- [109].Li X, Sun B, Qian H, Ma J, Paolino M, Zhang Z, A high-efficiency and versatile CRISPR/Cas9-mediated HDR-based biallelic editing system, J. Zhejiang Univ. Sci. B. 23 (2022) 141–152. 10.1631/jzus.B2100196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Suzuki K, Tsunekawa Y, Hernandez-Benitez R, Wu J, Zhu J, Kim EJ, Hatanaka F, Yamamoto M, Araoka T, Li Z, Kurita M, Hishida T, Li M, Aizawa E, Guo S, Chen S, Goebl A, Soligalla RD, Qu J, Jiang T, Fu X, Jafari M, Esteban CR, Berggren WT, Lajara J, Nuñez-Delicado E, Guillen P, Campistol JM, Matsuzaki F, Liu G-H, Magistretti P, Zhang K, Callaway EM, Zhang K, Belmonte JCI, In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration, Nature. 540 (2016) 144–149. 10.1038/nature20565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Li X, Zhang G, Huang S, Liu Y, Tang J, Zhong M, Wang X, Sun W, Yao Y, Ji Q, Wang X, Liu J, Zhu S, Huang X, Development of a versatile nuclease prime editor with upgraded precision, Nat. Commun. 14 (2023) 305. 10.1038/s41467-023-35870-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Knipping F, Newby GA, Eide CR, McElroy AN, Nielsen SC, Smith K, Fang Y, Cornu TI, Costa C, Gutierrez-Guerrero A, Bingea SP, Feser CJ, Steinbeck B, Hippen KL, Blazar BR, McCaffrey A, Mussolino C, Verhoeyen E, Tolar J, Liu DR, Osborn MJ, Disruption of HIV-1 co-receptors CCR5 and CXCR4 in primary human T cells and hematopoietic stem and progenitor cells using base editing, Mol. Ther. 30 (2022) 130–144. 10.1016/j.ymthe.2021.10.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Schmidt JK, Strelchenko N, Park MA, Kim YH, Mean KD, Schotzko ML, Kang HJ, Golos TG, Slukvin II, Genome editing of CCR5 by CRISPR-Cas9 in Mauritian cynomolgus macaque embryos, Sci. Rep. 10 (2020) 18457. 10.1038/s41598-020-75295-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Nerys-Junior A, Braga-Dias LP, Pezzuto P, Cotta-de-Almeida V, Tanuri A, Comparison of the editing patterns and editing efficiencies of TALEN and CRISPR-Cas9 when targeting the human CCR5 gene., Genet. Mol. Biol. 41 (2018) 167–179. 10.1590/1678-4685-GMB-2017-0065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Ye L, Wang J, Beyer AI, Teque F, Cradick TJ, Qi Z, Chang JC, Bao G, Muench MO, Yu J, Levy JA, Kan YW, Seamless modification of wild-type induced pluripotent stem cells to the natural CCR5Δ32 mutation confers resistance to HIV infection, Proc. Natl. Acad. Sci. U. S. A. 111 (2014) 9591–9596. 10.1073/pnas.1407473111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Kang H, Minder P, Park MA, Mesquitta W-T, Torbett BE, Slukvin II, CCR5 Disruption in Induced Pluripotent Stem Cells Using CRISPR/Cas9 Provides Selective Resistance of Immune Cells to CCR5-tropic HIV-1 Virus, Mol. Ther. - Nucleic Acids. 4 (2015). 10.1038/mtna.2015.42. [DOI] [PubMed] [Google Scholar]
- [117].Pacesa M, Lin C-H, Cléry A, Saha A, Arantes PR, Bargsten K, Irby MJ, Allain FH-T, Palermo G, Cameron P, Donohoue PD, Jinek M, Structural basis for Cas9 off-target activity, Cell. 185 (2022) 4067–4081.e21. 10.1016/j.cell.2022.09.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Josipović G, Zoldoš V, Vojta A, Active fusions of Cas9 orthologs, J. Biotechnol. 301 (2019) 18–23. 10.1016/j.jbiotec.2019.05.306. [DOI] [PubMed] [Google Scholar]
- [119].Finnigan GC, Thorner J, mCAL: A New Approach for Versatile Multiplex Action of Cas9 Using One sgRNA and Loci Flanked by a Programmed Target Sequence, G3 Bethesda Md. 6 (2016) 2147–2156. 10.1534/g3.116.029801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Liu Z, Liang J, Chen S, Wang K, Liu X, Liu B, Xia Y, Guo M, Zhang X, Sun G, Tian G, Genome editing of CCR5 by AsCpf1 renders CD4+T cells resistance to HIV-1 infection, Cell Biosci. 10 (2020) 85. 10.1186/s13578-020-00444-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Vakulskas CA, Dever DP, Rettig GR, Turk R, Jacobi AM, Collingwood MA, Bode NM, McNeill MS, Yan S, Camarena J, Lee CM, Park SH, Wiebking V, Bak RO, Gomez-Ospina N, Pavel-Dinu M, Sun W, Bao G, Porteus MH, Behlke MA, A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human hematopoietic stem and progenitor cells, Nat. Med. 24 (2018) 1216–1224. 10.1038/s41591-018-0137-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Yang H, Eremeeva E, Abramov M, Jacquemyn M, Groaz E, Daelemans D, Herdewijn P, CRISPR-Cas9 recognition of enzymatically synthesized base-modified nucleic acids, Nucleic Acids Res. 51 (2023) 1501–1511. 10.1093/nar/gkac1147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Scott T, Urak R, Soemardy C, Morris KV, Improved Cas9 activity by specific modifications of the tracrRNA, Sci. Rep. 9 (2019) 16104. 10.1038/s41598-019-52616-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Dabrowska M, Czubak K, Juzwa W, Krzyzosiak WJ, Olejniczak M, Kozlowski P, qEva-CRISPR: a method for quantitative evaluation of CRISPR/Cas-mediated genome editing in target and off-target sites, Nucleic Acids Res. 46 (2018) e101. 10.1093/nar/gky505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Xu H, Luo D, Gao X, Liu X, Wang Q, Sun A, Shen J, He R, Lu G, Li K, Zhang J, Xiao L, ZeoR: A Candidate for Assays Testing Enzymatic Activities and Off-Target Effects of Gene-Editing Enzymes, J. Biomed. Nanotechnol. 15 (2019) 662–673. 10.1166/jbn.2019.2720. [DOI] [PubMed] [Google Scholar]
- [126].Li Y, Glass Z, Huang M, Chen Z-Y, Xu Q, Ex vivo cell-based CRISPR/Cas9 genome editing for therapeutic applications, Biomaterials. 234 (2020) 119711. 10.1016/j.biomaterials.2019.119711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Asmamaw Mengstie M, Viral Vectors for the in Vivo Delivery of CRISPR Components: Advances and Challenges, Front. Bioeng. Biotechnol. 10 (2022) 895713. 10.3389/fbioe.2022.895713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Manjón AG, Linder S, Teunissen H, Friskes A, Zwart W, de Wit E, Medema RH, Unexpected gene activation following CRISPR-Cas9-mediated genome editing, EMBO Rep. 23 (2022) e53902. 10.15252/embr.202153902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Ferrari S, Jacob A, Cesana D, Laugel M, Beretta S, Varesi A, Unali G, Conti A, Canarutto D, Albano L, Calabria A, Vavassori V, Cipriani C, Castiello MC, Esposito S, Brombin C, Cugnata F, Adjali O, Ayuso E, Merelli I, Villa A, Di Micco R, Kajaste-Rudnitski A, Montini E, Penaud-Budloo M, Naldini L, Choice of template delivery mitigates the genotoxic risk and adverse impact of editing in human hematopoietic stem cells, Cell Stem Cell. 29 (2022) 1428–1444.e9. 10.1016/j.stem.2022.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Jacobson BF, Schapkaitz E, Takalani A, Rowji PF, Louw V, Opie J, Bekker LG, Garrett N, Goga A, Reddy T, Zuma NY, Sanne I, Seocharan I, Peter J, Robinson M, Collie S, Khan A, Takuva S, Gray G, Vascular thrombosis after single dose Ad26.COV2.S vaccine in healthcare workers in South Africa: open label, single arm, phase 3B study (Sisonke study), BMJ Med. 2 (2023) e000302. 10.1136/bmjmed-2022-000302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Yen J, Fiorino M, Liu Y, Paula S, Clarkson S, Quinn L, Tschantz WR, Klock H, Guo N, Russ C, Yu VWC, Mickanin C, Stevenson SC, Lee C, Yang Y, TRIAMF: A New Method for Delivery of Cas9 Ribonucleoprotein Complex to Human Hematopoietic Stem Cells, Sci. Rep. 8 (2018) 16304. 10.1038/s41598-018-34601-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Kholosy WM, Visscher M, Ogink K, Buttstedt H, Griffin K, Beier A, Gerlach JP, Molenaar JJ, Geijsen N, de Boer M, Chatsisvili A, Simple, fast and efficient iTOP-mediated delivery of CRISPR/Cas9 RNP in difficult-to-transduce human cells including primary T cells, J. Biotechnol. 338 (2021) 71–80. 10.1016/j.jbiotec.2021.07.006. [DOI] [PubMed] [Google Scholar]
- [133].Xu L, Wang J, Liu Y, Xie L, Su B, Mou D, Wang L, Liu T, Wang X, Zhang B, Zhao L, Hu L, Ning H, Zhang Y, Deng K, Liu L, Lu X, Zhang T, Xu J, Li C, Wu H, Deng H, Chen H, CRISPR-Edited Stem Cells in a Patient with HIV and Acute Lymphocytic Leukemia., N. Engl. J. Med. 381 (2019) 1240–1247. 10.1056/NEJMoa1817426. [DOI] [PubMed] [Google Scholar]
- [134].Xu L, Yang H, Gao Y, Chen Z, Xie L, Liu Y, Liu Y, Wang X, Li H, Lai W, He Y, Yao A, Ma L, Shao Y, Zhang B, Wang C, Chen H, Deng H, CRISPR/Cas9-Mediated CCR5 Ablation in Human Hematopoietic Stem/Progenitor Cells Confers HIV-1 Resistance In Vivo, Mol. Ther. 25 (2017) 1782–1789. 10.1016/j.ymthe.2017.04.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Xu M, CCR5-Δ32 biology, gene editing, and warnings for the future of CRISPR-Cas9 as a human and humane gene editing tool., Cell Biosci. 10 (2020) 48. 10.1186/s13578-020-00410-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Cashen AF, Nervi B, DiPersio J, AMD3100: CXCR4 antagonist and rapid stem cell-mobilizing agent, Future Oncol. Lond. Engl. 3 (2007) 19–27. 10.2217/14796694.3.1.19. [DOI] [PubMed] [Google Scholar]
- [137].Luo C, Wu G, Huang X, Zhang Y, Ma Y, Huang Y, Huang Z, Li H, Hou Y, Chen J, Li X, Xu S, Efficacy of hematopoietic stem cell mobilization regimens in patients with hematological malignancies: a systematic review and network meta-analysis of randomized controlled trials, Stem Cell Res. Ther. 13 (2022) 123. 10.1186/s13287-022-02802-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Matatall KA, Shen C-C, Challen GA, King KY, Type II interferon promotes differentiation of myeloid-biased hematopoietic stem cells, Stem Cells Dayt. Ohio. 32 (2014) 3023–3030. 10.1002/stem.1799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Khoury M, Drake A, Chen Q, Dong D, Leskov I, Fragoso MF, Li Y, Iliopoulou BP, Hwang W, Lodish HF, Chen J, Mesenchymal Stem Cells Secreting Angiopoietin-Like-5 Support Efficient Expansion of Human Hematopoietic Stem Cells Without Compromising Their Repopulating Potential, Stem Cells Dev. 20 (2011) 1371–1381. 10.1089/scd.2010.0456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Wu HL, Busman-Sahay K, Weber WC, Waytashek CM, Boyle CD, Bateman KB, Reed JS, Hwang JM, Shriver-Munsch C, Swanson T, Northrup M, Armantrout K, Price H, Robertson-LeVay M, Uttke S, Kumar MR, Fray EJ, Taylor-Brill S, Bondoc S, Agnor R, Junell SL, Legasse AW, Moats C, Bochart RM, Sciurba J, Bimber BN, Sullivan MN, Dozier B, MacAllister RP, Hobbs TR, Martin LD, Panoskaltsis-Mortari A, Colgin LMA, Siliciano RF, Siliciano JD, Estes JD, Smedley JV, Axthelm MK, Meyers G, Maziarz RT, Burwitz BJ, Stanton JJ, Sacha JB, Allogeneic immunity clears latent virus following allogeneic stem cell transplantation in SIV-infected ART-suppressed macaques, Immunity. 56 (2023) 1649–1663.e5. 10.1016/j.immuni.2023.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Schmidt JK, Reynolds MR, Golos TG, Slukvin II, CRISPR/Cas9 genome editing to create nonhuman primate models for studying stem cell therapies for HIV infection, Retrovirology. 19 (2022) 17. 10.1186/s12977-022-00604-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Saha A, Blazar BR, Antibody based conditioning for allogeneic hematopoietic stem cell transplantation, Front. Immunol. 13 (2022) 1031334. 10.3389/fimmu.2022.1031334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].DiGiusto DL, Krishnan A, Li L, Li H, Li S, Rao A, Mi S, Yam P, Stinson S, Kalos M, Alvarnas J, Lacey SF, Yee J-K, Li M, Couture L, Hsu D, Forman SJ, Rossi JJ, Zaia JA, RNA-based gene therapy for HIV with lentiviral vector-modified CD34(+) cells in patients undergoing transplantation for AIDS-related lymphoma, Sci. Transl. Med. 2 (2010) 36ra43. 10.1126/scitranslmed.3000931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, Spratt SK, Surosky RT, Giedlin MA, Nichol G, Holmes MC, Gregory PD, Ando DG, Kalos M, Collman RG, Binder-Scholl G, Plesa G, Hwang W-T, Levine BL, June CH, Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV, N. Engl. J. Med. 370 (2014) 901–910. 10.1056/NEJMoa1300662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Tebas P, Jadlowsky JK, Shaw PA, Tian L, Esparza E, Brennan AL, Kim S, Naing SY, Richardson MW, Vogel AN, Maldini CR, Kong H, Liu X, Lacey SF, Bauer AM, Mampe F, Richman LP, Lee G, Ando D, Levine BL, Porter DL, Zhao Y, Siegel DL, Bar KJ, June CH, Riley JL, CCR5-edited CD4+ T cells augment HIV-specific immunity to enable post-rebound control of HIV replication, J. Clin. Invest. 131 (2021) e144486, 144486. 10.1172/JCI144486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Wang W, Ye C, Liu J, Zhang D, Kimata JT, Zhou P, CCR5 Gene Disruption via Lentiviral Vectors Expressing Cas9 and Single Guided RNA Renders Cells Resistant to HIV-1 Infection, PLOS ONE. 9 (2014) e115987. 10.1371/journal.pone.0115987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].Li C, Guan X, Du T, Jin W, Wu B, Liu Y, Wang P, Hu B, Griffin GE, Shattock RJ, Hu Q, Inhibition of HIV-1 infection of primary CD4+ T-cells by gene editing of CCR5 using adenovirus-delivered CRISPR/Cas9, J. Gen. Virol. 96 (2015) 2381–2393. 10.1099/vir.0.000139. [DOI] [PubMed] [Google Scholar]
- [148].Choi JG, Dang Y, Abraham S, Ma H, Zhang J, Guo H, Cai Y, Mikkelsen JG, Wu H, Shankar P, Manjunath N, Lentivirus pre-packed with Cas9 protein for safer gene editing, Gene Ther. 23 (2016) 627–633. 10.1038/gt.2016.27. [DOI] [PubMed] [Google Scholar]
- [149].Hultquist JF, Schumann K, Woo JM, Manganaro L, McGregor MJ, Doudna J, Simon V, Krogan NJ, Marson A, A Cas9 Ribonucleoprotein Platform for Functional Genetic Studies of HIV-Host Interactions in Primary Human T Cells, Cell Rep. 17 (2016) 1438–1452. 10.1016/j.celrep.2016.09.080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Ehrke-Schulz E, Schiwon M, Leitner T, Dávid S, Bergmann T, Liu J, Ehrhardt A, CRISPR/Cas9 delivery with one single adenoviral vector devoid of all viral genes, Sci. Rep. 7 (2017) 17113. 10.1038/s41598-017-17180-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Qi C, Li D, Jiang X, Jia X, Lu L, Wang Y, Sun J, Shao Y, Wei M, Inducing CCR5Δ32/Δ32 Homozygotes in the Human Jurkat CD4+ Cell Line and Primary CD4+ Cells by CRISPR-Cas9 Genome-Editing Technology, Mol. Ther. Nucleic Acids. 12 (2018) 267–274. 10.1016/j.omtn.2018.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Xiao Q, Chen S, Wang Q, Liu Z, Liu S, Deng H, Hou W, Wu D, Xiong Y, Li J, Guo D, CCR5 editing by Staphylococcus aureus Cas9 in human primary CD4+ T cells and hematopoietic stem/progenitor cells promotes HIV-1 resistance and CD4+ T cell enrichment in humanized mice, Retrovirology. 16 (2019) 15. 10.1186/s12977-019-0477-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Lin D, Scheller SH, Robinson MM, Izadpanah R, Alt EU, Braun SE, Increased Efficiency for Biallelic Mutations of the CCR5 Gene by CRISPR-Cas9 Using Multiple Guide RNAs As a Novel Therapeutic Option for Human Immunodeficiency Virus, CRISPR J. 4 (2021) 92–103. 10.1089/crispr.2020.0019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Yu S, Ou Y, Xiao H, Li J, Adah D, Liu S, Zhao S, Qin L, Yao Y, Chen X, Experimental Treatment of SIV-Infected Macaques via Autograft of CCR5-Disrupted Hematopoietic Stem and Progenitor Cells, Mol. Ther. - Methods Clin. Dev. 17 (2020) 520–531. 10.1016/j.omtm.2020.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Teque F, Ye L, Xie F, Wang J, Morvan MG, Kan YW, Levy JA, Genetically-edited induced pluripotent stem cells derived from HIV-1-infected patients on therapy can give rise to immune cells resistant to HIV-1 infection, AIDS Lond. Engl. 34 (2020) 1141–1149. 10.1097/QAD.0000000000002539. [DOI] [PubMed] [Google Scholar]
- [156].Mehmetoglu-Gurbuz T, Yeh R, Garg H, Joshi A, Combination gene therapy for HIV using a conditional suicidal gene with CCR5 knockout, Virol. J. 18 (2021) 31. 10.1186/s12985-021-01501-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].D’Souza SS, Kumar A, Weinfurter J, Park MA, Maufort J, Tao L, Kang H, Dettle ST, Golos T, Thomson JA, Reynolds MR, Slukvin I, Generation of SIV-resistant T cells and macrophages from nonhuman primate induced pluripotent stem cells with edited CCR5 locus, Stem Cell Rep. 17 (2022) 953–963. 10.1016/j.stemcr.2022.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [158].Scheller SH, Rashad Y, Saleh FM, Willingham KA, Reilich A, Lin D, Izadpanah R, Alt EU, Braun SE, Biallelic, Selectable, Knock-in Targeting of CCR5 via CRISPR-Cas9 Mediated Homology Directed Repair Inhibits HIV-1 Replication, Front. Immunol. 13 (2022) 821190. 10.3389/fimmu.2022.821190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Li S, Holguin L, Burnett JC, CRISPR-Cas9-mediated gene disruption of HIV-1 co-receptors confers broad resistance to infection in human T cells and humanized mice., Mol. Ther. Methods Clin. Dev. 24 (2022) 321–331. 10.1016/j.omtm.2022.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]