Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jul 1.
Published in final edited form as: Curr Opin HIV AIDS. 2023 May 15;18(4):191–208. doi: 10.1097/COH.0000000000000803

Delivery Platforms for Broadly Neutralizing Antibodies

Lok R Joshi a, Nicolás MS Gálvez a, Sukanya Ghosh b, David B Weiner b, Alejandro B Balazs a,*
PMCID: PMC10247185  NIHMSID: NIHMS1896271  PMID: 37265268

Abstract

Purpose of review

Passive administration of broadly neutralizing antibodies (bNAbs) is being evaluated as a therapeutic approach to prevent or treat HIV infections. However, a number of challenges face the widespread implementation of passive transfer for HIV. To reduce the need of recurrent administrations of bNAbs, gene-based delivery approaches have been developed which overcome the limitations of passive transfer.

Recent findings

The use of DNA and mRNA for the delivery of bNAbs has made significant progress. DNA-encoded monoclonal antibodies (DMAbs) have shown great promise in animal models of disease and the underlying DNA-based technology is now being tested in vaccine trials for a variety of indications. The COVID-19 pandemic greatly accelerated the development of mRNA-based technology to induce protective immunity. These advances are now being successfully applied to the delivery of monoclonal antibodies using mRNA in animal models. Delivery of bNAbs using viral vectors, primarily adeno-associated virus (AAV), has shown great promise in preclinical animal models and more recently in human studies. Most recently, advances in genome editing techniques have led to engineering of monoclonal antibody expression from B cells. These efforts aim to turn B cells into a source of evolving antibodies that can improve through repeated exposure to the respective antigen.

Summary

The use of these different platforms for antibody delivery has been demonstrated across a wide range of animal models and disease indications, including HIV. While each approach has unique strengths and weaknesses, additional advances in efficiency of gene delivery and reduced immunogenicity will be necessary to drive widespread implementation of these technologies. Considering the mounting clinical evidence of the potential of bNAbs for HIV treatment and prevention, overcoming the remaining technical challenges for gene-based bNAb delivery represents a relatively straightforward path towards practical interventions against HIV infection.

Keywords: HIV, Broadly Neutralizing Antibody, Vectored ImmunoProphylaxis, mRNA, DNA Delivery, Broadly neutralizing antibodies, DNA-based therapies, mRNA-based therapies, Adeno-Associated Virus, B cell editing

INTRODUCTION

Since their initial discovery, broadly neutralizing antibodies (bNAbs) targeting diverse HIV-1 isolates have fundamentally altered the landscape of HIV prevention and treatment research [1]. Given their promise, significant effort has been focused on the development of vaccine approaches capable of eliciting bNAbs [2]. Yet passive transfer of bNAb proteins has shown significant effects in studies of either prevention [3-5] or treatment [6-9] of HIV infection. The relatively short half-life of passively transferred monoclonal antibodies necessitates regular infusions to maintain functional circulating titers, hindering the utility of bNAb passive transfer for both treatment and prevention of HIV [5]. This shortcoming has stimulated multiple lines of investigation into alternative approaches for the delivery of bNAbs in more convenient or longer-lived formats. These approaches include genetically encoding bNabs for delivery as plasmid DNA, modified mRNA, or adeno associated virus vectors, with the most recent efforts aimed at genetically engineering host B cells. In this article, we review prior studies describing the use of each technology as a means of delivering monoclonal antibodies, with specific emphasis on approaches used for HIV bNAb delivery.

DNA for Delivery of bNAbs

The in vivo expression of recombinant proteins by exogenous nucleic acid injected into skeletal muscle was reported for the first time in the early 90s [10,11]. In this approach, protein-coding expression transgenes are encoded in plasmids or other DNA forms for expression in vivo. Originally, DNA was advanced as a delivery platform for diverse vaccine and immunization strategies [12,13]. Numerous clinical trials have been performed and several are currently in progress evaluating DNA immunization against infectious diseases and cancers (NCT04090528, NCT03110770, NCT04131413, NCT04251117). During the recent SARS-CoV-2 pandemic, India granted emergency authorization to license ZyCoV-D, a plasmid DNA vaccine delivered by Jet injection for use in adults and children of 12 years and older [14] for prevention of SARS-CoV-2 infection. However, based on improved platform performance, they are being studied for in vivo person-specific protein production. Conceptually, DNA biologics exhibit important features relevant for globally distributable products, including temperature stability, an excellent safety profile and lack of vector-induced immune responses allowing for repeat delivery. A single DNA vector backbone can be re-administered repeatedly for the delivery of either the same or different genes without the induction of vector-specific immunity. The approach has yielded months-long expression from a single inoculation, with a focus on intramuscular (IM) or intradermal (ID) delivery to simplify use in the field. A major goal for improvement of DNA platforms has been to increase their expression levels and immunogenicity when delivering vaccine antigens. Approaches such as codon and RNA optimization, improved leader sequences and improved delivery formulations and delivery methods are under investigation [14,15]. These approaches include needle-based injections of naked DNA or ballistic DNA, forms of jet delivery, multiple electroporation approaches, sonoporation, and photoporation among others, which can improve in vivo expression from gene-encoded DNA cassettes [12,14].

Studies over the past decade have shown continued improvement in the expression levels achieved with DNA-encoded monoclonal antibodies (DMAbs) in animal studies [16-20,15,21-24,12,25,26,13,27-29]. DMAbs hold promise to accelerate the deployment of new therapeutic interventions and provide preclinical tools for rapid evaluation of biological products. DMAbs have been tested for IgG production targeting prevention and treatment of diverse infectious diseases [15,20,22,23,25,28-40], and cancer [41-48] (Table 1. Current approaches for DNA delivery of antibodies). For example, DV87.1, a dengue-specific neutralizing antibody, was encoded as a DMAb for delivery of multi-serotype neutralizing antibodies produced in vivo upon IM injection that was sufficient for protection in animal challenges [15]. Studies have reported the delivery of combinations of DMAbs resulting in protection against viral or bacterial disease models. For example, the concomitant use of two broadly neutralizing DMAbs for H1 or H3 influenza viruses resulted in a total of 3μg/mL of IgG circulating antibodies, which were able to protect against a lethal dose challenge with either influenza strain, independently [23]. Studies also focused on the engineering of V regions (among others) as an important approach for increasing serum concentrations from DMAb delivery. This was first described using a panel of anti-Ebola MAbs, which were redeveloped for enhanced in vivo expression using a pDNA delivery format [24]. After enhancement, Patel et al. demonstrated that a single injection with DMAbs resulted in months of expression, achieving 48 μg/ml peak serum concentration and providing single-dose protection against Ebola challenge in animals. As a response to Zika infection, a potent anti-Zika MAb, ZK-190, was studied first in mice and then in non-human primates (NHPs) and was shown to protect against viral challenge [26]. Through a collaborative academic/industry partnership, two potent SARS-CoV2 mAbs were redesigned for DNA delivery and advanced rapidly into clinical studies as DMAbs. Administration of these DMAbs to BALB/c mice induced peak expression of 5-50 μg/mL of circulating human IgG within 21 days and recapitulated the immune phenotype of the parental mAbs [40]. Expression from a single IM administration was confirmed for over 200 days, showing a prolonged slope of decay. These animals were protected from SARS-CoV-2 challenge, to a similar degree as animals given passive transfer [40]. Clinical testing of this dual DMAb approach has moved into a human clinical trial in the last few months (NCT05293249). This study could provide important information on the safety, deliverability, and expression levels of the DMAb approaches for dual mAb delivery in humans.

Table 1.

Current approaches for DNA delivery of antibodies.

Type of
disease
Disease Target Antibodies expressed Organism
tested
Comments References
Proof of concept N/A Human thyroglobulin Tg10 Mice · IM electroporation of naked DNA allows both constitutive and regulatable expression Perez et al., 2004
Mouse CD25 and Human CTLA-4 PC61 5.3 and A3.6B10 NA · An expression vector system developed for expression of fully functional and antigen-specific human antibodies with correct isotype specificities
· RNA and Codon optimized leader sequence demonstrated increased secretion correct isotype specificities
Morrow et al., 2009
Infectious Pathogen Influenza Hemaglutanin pHA mAb Mice · Multiple delivery of single DMAb at different sites enhances expression and neutralization
· Protection against IFV lethal challenge
Yamazaki et al., 2011
C179, S139/1, 9H10 Mice · First report- enhanced expression (32 weeks)
· Multiple mAb delivery in same animal.
· Oligoclonal mAb protection
· Heterosubtypic immunity
Andrews et al., 2017
2-12C Mice, Pig and Sheep · Reduction in viral load and lung pathology after pandemic H1N1 influenza challenge at a prophylactic dose
· Reduction in only lung pathology at lower dose
· MAbs based on bovine Abs demonstrate durable circulating serum concentrations for ~75 days in mice
· Peak levels of 7-12 μg/ml for 7-14 days in sheep, with no ADA responses
McNee et al., 2020
FluA, FluB Mice · Single dose protects against lethal challenge
· Coordinated delivery of mAb resulting in exceptionally broad protection against both influenza A and B
Elliott et al., 2017
HIV-1 Envelope Protein VRC01 Mice · First report of use of strategies to enhance Fab expression for antibody-encoding plasmids, like codon optimization and improved EP conditions
· A peak serum conc of 2–3 μg/mL at day 12 post-injection
· Single EP enhanced administration results in rapid production of Fabs in vivo, which neutralized a panel of different viral tier 1 and 2 isolates.
Muthumani et al., 2013
VRC01, PGT151, PGDM1400, PGT121, PGT145, 3BNC117, 10–1074 Mice and Rhesus macaque · First report- DMAb platform for delivering bnAbs
· Delivery of multiple DMAbs to a single animal neutralized the entire global panel of HIV-1.
· High peak-circulating levels and broad neutralization activity.
· 6-34 mg/ml expression levels in NHPs
Wise et al., 2020
Dengue E Protein, DIII DV87.1 Mice · First report- protection against multiple serotypes into any animal model
· Single dose- prevents Antibody-dependent enhancement (ADE)
Flingai et al., 2015
Chikungunya Envelope Protein CVM1 Mice · First report of DMAb and DNA vaccine
· Single dose- Protects against lethal challenge
Muthumani et al., 2016
P. aeruginosa PcrV and Psl exopolysaccharide αPcrV, MEDI3902 Mice · Bispecific DMAb- (MEDI3902) exhibits enhanced protective activity with antibiotic treatment in a lethal pneumonia model Patel et al., 2017
Ebola EBOV GP ZMapp (2G4, 4G7,13C6) Mice · Sustained expression for 15 weeks
· oligoclonal and anti-ebola mAbs protection
Andrews et al., 2017
EBOV GP Fusion Loop and Heptad Repeat 2 DMAb-11, DMAb-34 Mice · Fully human DMAb confers 100% protection Patel et al., 2018
B.burgdorferi OspA 319–44 Mice · First report- DNA transfer as a delivery system for antibodies that block transmission of Borrelia in animal models (HuMAb)
· protection against an acute challenge by Borrelia-infected ticks
Wang et al., 2019
Zika E Protein DMAb-ZK190 Mice and Rhesus macaque · First report- infectious disease control in NHPs following in vivo delivery of a nucleic acid-encoded antibody
· Expression levels persisting >10 weeks in mice and >3 weeks in non-human primate (NHPs)
· Protection against infectious challenge in NHPs
Esquivel et al., 2019
Env Protein 1C2A6, 1D4G7, 2B707, 3F12E9, 4D6E8, 5E6D9, 6F9D1, D10F4, 8A9F9, 9F7E1 Mice and Rhesus macaque · First report- in vivo expression of anti-ZIKV antibodies from a vector system
· Single dose protects against infectious challenge
· co-formulated with an anti-ZIKV DNA vaccine provides immediate and persistent anti-ZIKV immune responses
Choi et al., 2020
Respiratory Syncytial virus fusion protein (F) pGX9369 Mice and Cotton rat · Single administration of single-chain fragment variable-constant fragment (scFv-Fc) RSV-F dMAb demonstrated long-lasting immunity and effective biodistribution
· In vivo protection in viral challenge
Schultheis et al., 2020
N. gonorrhoeae Oligosaccharide on Lipooligosacchride 2C7 Mice · Complement-engaging variants facilitated rapid clearance following primary challenge with longer duration of protection Parzych et al., 2021
P. falciparum Circumsporozoite Surface Protein human mAb clones CIS43, 317, and L9 Mice · Long-term serological expression
· In vivo efficacy of CIS43 and 317 (germ line modified variants) in mosquito bite challenge
Tursi et al., 2022
SARS-CoV-2 Spike Protein COV2-2196 and COV2-2130 Mouse, Hamster · First report- cryo EM structure of polyclonal in vivo produced DMAbs
· High peak DMAb serum titers and long-term expression. DMAbs exhibited prolonged kinetics relative to protein IgG
· Reduction in lung viral burden by >4-6 logs in a lethal challenge.
· Protection was similar to protein mAbs
Parzych et al., 2022
Cancer Breast Cancer HER2 mumAb4D5 Mice · High and sustained expression
· Effective inhibition of tumor growth
Kim et al., 2016
Anti-HER2 Mice · Expression for several months, boosting expression by pDNA re-dosing
· Complete tumor regressions
Hollevoet et al., 2018
Ovarian Cancer HER2 Anti-HER2 Mice · High serum expression for long duration
· Ovarian tumor control and prolonging survival
Perales-Puchalt et al., 2019
FSHR Anti-FSHR (D2AP11) Mice · D2AP11 can identify resistant ovarian cancer cell lines
· Development of T cell engagers.
Bordoloi et al., 2022
Prostate Cancer PSMA Anti-PSMA Mice · First application of enhanced synthetic DNA for vivo production of human MAb for cancer immunotherapy
· Robust expression, controlled tumor growth and prolonged survival
Muthumani et al., 2017
Fibrosarcoma CTLA-4 Anti CTLA-4 Mice · Single dose - expression for several months
· high serum levels and tumor regression
Duperret et al., 2018
Colorectal cancer (along with many others) Cancer Embryonic Antigen OVAC Sheep · Robust and prolonged in vivo production
· Dose dependent response observed
Hollevoet et al., 2022
Glioblastoma Multiforme DBTE EGFRvIII-targeting DBTE Mice · First as a monotherapy for direct in vivo treatment for GBM in both peripheral and orthotopic challenge animal models
· Durable in vivo expression and demonstrated potent tumor regression and clearance in mice
Park et al., 2023

An important area for HIV research is the delivery of cocktails of broadly neutralizing antibodies as a means of preventing infections or as therapy. Wise et al. designed a large panel of DMAbs optimized for in vivo expression and evaluated whether these antibodies could be delivered in combinations to neutralize a global viral HIV panel. Specific combinations of 2 to 4 DMAbs (PGDM1400, PGT121, VRC01, and PGT151) selected from a larger panel in DMAb formats were tested in combinations of DMAb and found that cocktails were able to neutralize the 12-member global panel. A dramatic increase in neutralization breadth was described, with IC50 levels below 0.1μg/ml for all 12 global panel viruses [29]. Two anti-HIV-1 DMAbs, PGDM1400 and PGT121 alone or in combination, were also tested in NHPs, with peak serum concentrations ranging between 6-34 μg/ml and no safety concerns being reported in NHPs. These studies demonstrate that the in vivo produced DMAbs retained neutralization properties equivalent to the original bnAbs [29]. These studies serve as a stepping stone for further development of bnAbs as DMAbs. Future efforts are needed to enhance their immunological properties as well as their half-life, which would improve the pharmacokinetic profile and reduce the need for frequent infusions.

DNA as a gene delivery platform has unique advantages, including its inherent stability and simplicity of production, as well as the ease of dissemination worldwide. The simplicity of combining multiple therapeutic proteins/antibodies is likely to be important for the successful application of this approach, particularly for HIV and other infectious diseases. Recent studies using engineered forms of biologics, such as DMAb encoded Bispecific T cell Engagers (BiTEs) to treat diverse cancers, further support the important applications of the DNA platform [44,48,49]. Future investigations will focus on the development and use of next-generation DNA platforms in the context of HIV therapy and other infectious diseases of global importance.

Modified mRNA for Delivery of bNAbs

The administration of exogenous mRNA for the expression of proteins by the host has made great strides in the last few years [50]. Notably, the COVID-19 pandemic greatly accelerated the clinical development and widespread use of mRNA-based vaccines to induce the expression of SARS-CoV-2 spike antigen, which stimulated an effective immune response against this virus [51]. Rapid and high-level expression, proven scalability, and an inability to integrate into the host genome, are among the significant advantages that mRNA has over other platforms [52,53]. However, if delivered alone, mRNA can induce the activation of Pattern Recognition Receptors (PRRs) [54], such as toll-like receptor (TLR)-3, −7, and −8 [55]; retinoic acid-inducible gene I (RIG-I) [56]; melanoma differentiation-associated protein 5 (MDA-5) [57]. In addition, mRNA molecules are degraded by intra- and extra-cellular ribonucleases and cannot easily enter the host cell [58], requiring the incorporation of modified nucleosides to side-step these challenges [59]. Multiple mRNA formats and routes of administration are currently being studied [60].

The use of mRNA to encode antibody transgenes has been tested for a multitude of indications, including pathogens [52,61-76], toxins [67,77,78], and cancers [67,79-84] (Table 2. Current approaches for mRNA delivery of antibodies). Despite these promising reports, most studies of mRNA relating to HIV have been focused on the expression of recombinant viral antigens to promote adaptive humoral immunity rather than to produce bNAbs. The first report on mRNA administration to induce the expression of bNAbs was published in 2017 by Pardi et al. [61]. Therein, the expression of VRC01, a bNAb targeting the CD4 binding site of the HIV envelope (Env), was achieved in vivo by lipid nanoparticle (LNP)-encapsulated and nucleoside-modified mRNA. The antibody levels achieved by a single injection of 30μg of mRNA resulted in higher levels of circulating antibodies than those detected for a single administration of 600μg of recombinant VRC01, culminating in the protection of two humanized mouse models from infection with HIV-1 [61]. Antibody levels following mRNA administration peaked at 24 hours, with a gradual decrease over five days and a sharp decrease on day 7. Subsequently, a report by Lindsay et al. showed that aerosol administration of synthetic mRNA coding for an optimized PGT121, a bNAb targeting the V3-glycan of HIV Env, led to high expression levels in the reproductive tract of sheep and rhesus macaques [62]. This was sufficient to protect against simian-human immunodeficiency virus (SHIV) infection, with antibody expression lasting up to 28 days [62]. A recent study by Narayanan et al. sought to induce the simultaneous expression of three bNAbs, PGDM1400, PGT121, and N6, which also target HIV Env, using an mRNA-LNP platform [63]. Simultaneous expression of multiple antibodies could lead to mismatched combinations of heavy- and light-chains, thereby yielding aberrant antibodies [85]. To prevent this, the authors engineered single-chain (sc)Fv-Fc molecules in which the heavy- and light-chain variable domains from each antibody were bound by flexible linkers. This construct was linked to an Fc constant region to eliminate potential mismatches. While in vitro expressed PGDM1400 and PGT121 scFv-Fc proteins exhibited similar neutralizing potency as natural antibodies, this strategy was not always effective, requiring full-length IgG sequences for the N6 bNAb to retain activity. In vivo administration of an mRNA cocktail led to high expression levels of all three antibodies in human neonatal Fc receptor (FcRn) transgenic mice (Tg32), a model chosen to more accurately predict the pharmacokinetics of human antibodies [63].

Table 2.

Current approaches for mRNA delivery of antibodies.

Type of disease Disease Target Antibodies expressed Organism tested Comments References
Infectious Pathogen Chikungunya E2 glycoprotein CHKV-24 (mRNA-1994) Mouse, NHP, and humans. First ever clinical trial for an mRNA-expressed bNAb. Pre-clinical data shows protection against infection upon mRNA administration and antibody expression. Kose et al., 2019 and August et al., 2021
HBV HBV surface antigens G12-scFv, G12-scFv-Fc, and G12-IgG Mouse The expression of these three antibodies upon I.V. injection of the mRNA to C57BL/6 mice led to long-term clearance of HBV antigens from circulation. Chen et al., 2022
HIV Envelope protein VRC01 Mouse The I.V. mRNA administration led to over 170μg/mL of antibodies in BALB/c or BLT mice. Pardi et al., 2017
BHK cells The mRNA-expressed antibody exhibited neutralizing potency comparable to recombinant protein in vitro. Thran et al., 2017
PGT121 Sheep and NHP There was a marked antibody expression upon aerosol delivery of the mRNA either as full-length or only heavy chain in the reproductive tract. Lindsay et al., 2020
N6, PGDM1400, and PGT121 Mouse The I.V. administration of the mRNA mixture led to the concomitant expression of scFv-Fc antibodies retaining neutralization potency in Tg32. Narayanan et al., 2022
Influenza A Influenza A antigen Human IgG anti-Influenza A NHP The I.V. infusion of improved LNP led to increased antibody expression from the mRNA cargo. Sabnis et al., 2018
M2e and FcɣRIV RiboBiFE Mouse The expression of bispecific nanobody (VHHs) with protective features against viral infection was reported upon I.T. administration of the mRNA in BALB/c and C57BL/6. Van Hoecke et al., 2020
Influenza B HA CR8033 Mouse The I.V. mRNA administration led to up to 2μg/mL of circulating antibody in Swiss-Albino mice. However, this therapy could not protect from lethal infection. Thran et al., 2017
Rabies G glycoprotein S057 Mouse The I.M. injection of the mRNA led to the expression of the antibody as early as 2h in Swiss-Albino mice, which protected them from viral infection even as post-expsosure prophylaxis. Thran et al., 2017
Pseudomonas aeruginosa PsI (Biofilm component) CAM003 Mouse The expression of this antibody as an hIgA1 protein was achieved upon I.V. administration of the mRNA. This therapy led to similar levels of protection as those reported for the recombinant protein in BALB/c mice. Deal et al., 2023
Poxviruses Enveloped virions (EV) and mature virions (MV) c7D11 (anti-L1 MV), c8A (anti-B5 EV), and c6C (anti-A33 EV) Rabbits All three antibodies could be expressed concomitantly upon I.M. injection of the mRNA mix. However, based on empirical projections, the therapy in its current state was unlikely to protect against viral infection. Mucker et al., 2022
RSV Fusion glycoprotein Secreted Palivizumab (sPali) and RSV-neutralizing VHH camelid antibody (RSVaVHH) Mouse The prophylactic I.T. administration of the mRNA to BALB/c mice reduced viral loads and prevented severe disease. Tiwari et al., 2018
Salmonella enterica Typhimurium O5-antigen of LPS Sal4 Mouse The I.V. administration of the mRNA led to the expression of this antibody as an hIgA2, which protected BALB/c mice from infection to similar levels as those reported for the recombinant protein. Deal et al., 2023
SARS-CoV-2 Spike protein (RBD) CB6 Mouse Testing of self-replicating mRNA I.N. administered. Exhibited protection against viral infection in BALB/c. Li et al., 2021
HB27 Mouse and Hamster This antibody (not the mRNA-expressing therapy) is currently being tested in human trials. IV-administration of the mRNA led to long-term protection from infection in BALB/c mice and Syrian hamster. Deng et al., 2022
COV2-2832 and DH1041 Hamster The nebulized prophylactic administration of the mRNA significantly protected Syrian hamsters from SARS-CoV-2. Vanover et al., 2022
hACE 3E8 Hamster This self-replicating mRNA I.N.-administered to Syrian hamster protected against Beta, Delta, Gamma, and Omicron variants. Zhang et al., 2023
Zika Envelope protein ZIKV-117 Mouse The pre- or post-exposure I.M. administration of self-amplifying mRNA to C57BL/6 protected from lethal dose. Erasmus et al., 2020
Cancer Non-hodgkin lymphoma CD20 Rituximab Mouse There was a significant deceleration or abolishment of tumor growth in NOD/SCID mice upon I.V. administration of the mRNA. Thran et al., 2017
Lymphoblastic leukemia CD3 and tumor associated proteins (CLDN6, CLDN18.2, and EpCAM) Three different RiboMAbs Mouse The I.V. administration of mRNA encoding for these bispecific ScFv to NSG mice cleared advanced tumor as effectively as the recombinant protein. Stadler et al., 2017
Breast cancer HER-2 (ERBB2) Trastuzumab Mouse The I.V. injection to C57BL/6 mice of the mRNA led to high levels of expression of the antibody and a selective reduction of HER2-positive tumors, with improved survival. Rybakova et al., 2019
Hepatocellular carcinoma CCL2 and CCL5 BisCCL2/5i Mouse The I.V. administration of the mRNA coding for this bispecific single-domain VH antibody led to long-term survival of CD57BL/6 and BALB/c mice with primary, colorectal, and pancreatic metastasized hepatocellular carcinoma. Wang et al., 2021
Colon carcinoma PD-1 and PD-L1 XA-1 Mouse A marked expression of this bispecific antibody was seen upon I.V. injection of the mRNA coding in C57BL/6 and NOD/SCID mice. There was also a marked tumor growth inhibition of MC38 cells implanted to NOD/SCID mice. Wu et al., 2021
PD-1 Pembrolizumab Mouse The I.V. administration of the mRNA coding for this antibody effectively reduced intestinal tumor growth and improved C57BL/6 NOD/SCID mice survival. Wu et al., 2022
Acute myeloid leukemia and Subcutaneous melanoma CD3 and B7H3 B7 homolog 3 protein (B7H3 or CD276) and CD3 bispecific T-cell engaging (BiTE) antibody Mouse The I.V. adminstration of the mRNA coding for the BiTE antibody led to high levels of its expression, with extended half-life compared to the recombinant protein. This therapy also resulted in a marked and long-lived antitumor efficacy in NSG mice. Huan et al., 2023
Toxins Escherichia coli (O157:H7) Shiga toxin 2 (Stx2) VNA-Stx2 Mouse The mRNA expression of the VNA protected cell viability against Shiga Toxin 2 (Stx2) and can be expressed in CD1 mice. Thran et al., 2017
Clostridium botulinum Botulinum neurotoxin serotype A (BoNTA) VH domain-based neutralizing agent (VNA)-BonTA Mouse The mRNA-expressed VNA neutralized BoNTA in vitro to levels comparable to the recombinant protein and can be expressed in CD1 mice. Thran et al., 2017
BoNTA, BoNTB, and BoNTE Heterohexamer VHH Mouse The I.M. administration of the mRNA-expressing heterohexamer expressed efficiently and protected CD1 mice from lethal toxin doses for all serotypes of toxins tested. Mukherjee et al., 2022
BoNTA B11-Fc Mouse The I.M. administration of the mRNA-expressing nanobody protected BALB/c mice from high lethal doses of serotype A toxin. Panova et al., 2023

Of note is the first report of LNP-mRNA encoding IgA isotype antibodies targeting antigens from Salmonella Typhimurium and Pseudomonas aeruginosa [52]. The report by Deal et al. confirms the successful production of dimeric IgA antibodies by mRNA injection, which preferentially accumulates at mucosal surfaces and exhibits a longer half-life relative to its recombinant counterpart [52]. This valuable proof of concept demonstrates the versatility of mRNA for antibody delivery. Clinically, an ongoing phase 1 study for LNP-encapsulated mRNA encoding for CHKV-24, a monoclonal neutralizing antibody against the Chikungunya virus, is of particular interest [65]. This first-ever clinical trial of in vivo expression of a monoclonal antibody through the mRNA platform shows that administration of this mRNA is safe and well tolerated, resulting in therapeutically relevant concentrations and robust neutralizing activity of the circulating antibody [65]. Translating this progress to the clinical use of mRNA to encode HIV-targeting bNAbs is clearly warranted.

Adeno Associated Virus for Delivery of bNAbs

Vectored antibody delivery, or vectored immunoprophylaxis (VIP), has emerged as a promising tool for the delivery of broadly neutralizing antibodies. Adeno-associated virus (AAV) remains one of the most widely used vectors for antibody delivery (Table 3. Current approaches for AAV delivery of antibodies). AAV-mediated antibody delivery has several advantages over other approaches, including very high expression levels, persistence of expression lasting for years, and clinically proven safety and tolerability. The first demonstration of antibody delivery using AAV was by Lewis et al. in 2002 [86]. After administering a single intramuscular injection of rAAV vector expressing the HIV Env-binding antibody b12, they observed HIV-neutralizing activity in the sera of mice for over 6 months. Important contributions were made by Fang et al., who significantly optimized expression from AAV-based antibody delivery systems [87,88]. They showed that antibodies could be expressed via a single open reading frame by linking heavy and light chains with a picornavirus-based 2A self-processing peptide sequence [87]. Our lab further improved this strategy to achieve higher levels of bNAb using a muscle-optimized CASI promoter and added a post-transcriptional regulatory element (WPRE) downstream of the transgene [89]. This optimized AAV expression cassette resulted in several fold higher levels of bNAb expression (20-250 μg/mL) compared to non-optimized vectors in both immunocompetent and immunodeficient mouse models [89]. We have shown that VIP is capable of protecting humanized mice from intravenous [89] as well as repeated vaginal challenges with diverse HIV strains [90,91]. Others have shown that 6 out of 7 humanized mice could sustain suppression of HIV-1 using AAV8 to express 10-1074, an antibody targeting the V3 glycan in Env [92].

Table 3.

Current approaches for AAV delivery of antibodies.

Disease AAV seroype Antibody Animal Model Route AAV Dose (vector genomes;
vg)
Antibody concentration
achieved
Anti-drug antibody
response
Notes Reference
HIV AAV2 B12 Rag1 Mice Intramuscular 5E+10 to 5E+11 0.5 - 8 μg/mL Lewis et al., 2002
AAV1 Immunoadhesins ( 4L6, 8S, 5L7, 3V0 Rhesus macaques Intramuscular 2.00E+13 4L6 (100-190 μg/mL )
5L7 (50- 175 μg/mL)
Johnson et al., 2009
AAV8 B12 NSG, B6 and Balb/C Intramuscular 1.00E+11 20-250 μg/mL Balazs et al., 2011
AAV8 3BNC117, 10-1074 Humanized mice 2.50E+11 200 μg/mL Horwitz et al., 2013
AAV8 multiple anitbodies including PG9, VRC07, 3BNC117 NSG and BLT mice Intramuscular 0.05 - 390 μg/mL Mice receiving AAV-VROC7 were completely resistant to repetitive intravaginal challenge Balazs et al., 2014
AAV8 Simianized VRC07 Rhesus macaques Intramuscular 30? All animals unless Cyclosporin First to show that immunosuppression could alleviate ADA responses Saunders et al., 2015
AAV1 Immunoadhesin with rhesus IgG1 Rhesus macaques Intramuscular 0.8E+13 to 2.5E+13 1-270 μg/mL 9/ 12 animals In one animal, the concetration of antibody was 270 μg/mL and the levels persisted for 2 years Fuchs et al., 2015
AAV1 eCD4-Ig Rhesus macaques Intramuscular 2.50E+13 17- 77 μg/mL Rhesus eCD4-Ig was less immunogenic than rhesus forms of bNAbs Gardner et al., 2015
AAV1 4L6, 5L7, 1NC9, 8ANC195, 3BNC117 Rhesus macaques Intramuscular 1.6E+13 to 3E+13 NA 17/20 animals Martinez-Navio et al., 2016
AAV8 anti-SIV Env mAb ITS01 and ITS06.02 Rhesus macaques Intramuscular 1.00E+13 8-21 μg/mL ADA in 20% animals Welles et al., 2018
AAV1 and AAV8 10E8, 3BNC117, 10-1074 Rhesus macaques Intramuscular 2E+12 vg/kg 2-200 μg/mL Varying magnitidue of ADA present in most animals Viremia undetectable in one monkey for over 3 years Martinez-Navio et al., 2019
AAV1 3BNC117, NIH45-46, 10-1074 and PGT121 Rhesus macaques Intramuscular 2E+11 to 1E+13 3-69 μg/mL 12/ 12 animals Antibodies isotyped with IgG2 were found to be less immunogenic than IgG1 isotyped antibodies Gardner et al., 2019
AAV1 and AAV8 4L6 Rhesus macaques Intramuscular /Intravenous 0.25E+12 vg/kg Intramuscular : 1-7 μg/mL
Intravenous : 0.3- 2.3 μg/mL
AAV8 priming and AAV1 boost : 186-302 μg/mL
ADA in 9/9 animals in intramuscular group; no ADA detected (0/3) in intravenous group Muscle-specific or liver-specific promoters were used Fuchs et al., 2019
AAV1 PG9 Human Intramuscular 4E+12 to 1.2E+14 Undectectable (< 2 μg/mL) 10/16 detectable ADA First human clinical trial Priddy et al., 2019
AAV8 VRC07 Human Intramuscular 5E+10 to 2.5E+12 vg/kg <1 - 3.3 μg/mL 3/8 detected ADA (2/8 lost transgene) Clinical trial ongoing Casazza et al., 2022
AAV8 VRC07 containing Fc region of different human IgG subclass huPBMC and BLT mice Intramuscluar <1 - 70 μg/mL VRC07-IgG2 exhibited redued protection in vivo relative to other IgG subclasses. Brady et al., 2022
Cancer AAV1 anti-EGFR antibody 14E1 A431 xenograft tumor model Intramuscular 1E+11 to 5E+11 over 1000 μg/mL Ho et al., 2009
Malaria AAV8 2A10, 2C11 C57BL/6(6NCr) Intramuscular 1.00E+11 50-1000 μg/mL Deal et al., 2014
C. difficile AAV6.2FF actoxumab, bezlotoxumab Mice and Syrian Hamsters Intramuscualar Mice : 1E+11
Hamsters : 1E+12
90-195 μg/mL Guilleman et al., 2021
Parkinson's Disease AAV8 anti-Synuclein (NAC32) Rats (DAT-Cre) Intracerebral 2E+12 vg per injection site Chen et al., 2021
Ebola AAV6.2FF 2G4, 5D2 (murine IgG2a ebola virus mAbs)
EBOV mAb 100, 114, FVM04, ADI-15876, CA45 (as human IgG1)
Mice (BALB/c) Intramuscular 8E+9 to 4E+11 Dose dependent (<1 to 900 μg/mL) Sustained expression in mice for more than 400 days. Minimum serum antibody level of 2 μg/mL was found to be protective. Leishout et al., 2022
AAV9 2G4, 4G7, c13C6 Mice Intramusclar/ Intranasal 1.00E+11 9 μg/mL in serum; 3 μg/mL in BALF Humanization of mouse antibodies improved expression profile Limberis et al., 2016
AAV9 c2G4, c4G7, c13C6 Mice Intramuscular/ Intravenous/ Intranasal 2.7E+10 to 3E+11 Intramuscular : <1 - 26 μg/mL
Intravenous : 5.3 - 33 μg/mL
Intranasal : not detected
Robert et al., 2018
Herpes simplex virus (HSV) AAV8 CH42, CH43, E317 (HSV mAbs targeting gD) Mice (C57BL/6) Intramuscular 1.00E+11 Passive transfer of HSV-specific mAbs delivered via AAV from dams to their offspring Backes et al., 2022
Respiratory syncytial virus (RSV) AAV6.2FF Palivizumab, hRSV90 Mice Intramuscular / Intranasal Mice : 1E+11 174 - 397 μg/mL ( on day 70) Antibody detected in the serum and at various mucosal surfaces. Maternal passive transfer of antibodies observed. Rghei et al., 2022
Keratitis ichthyosis deafness AAV8 abEC1.1 Cx26G45E mouse Intravenous 1.25E+12 50 μg/mL Peres et al., 2023
SARS-CoV-2 AAV8 and AAV9 NC0321 hACE2-expressing mice Intramuscular/Intranasal 1.00E+11 AAV8 given IM : 3.9 μg/mL in serum; 18 μg/mL in BALF
AAV9 given IN : 0.9 μg/mL ; 65 μg/mL in BALF
Du et al., 2022
Influenza AAV8 F10, CR6261 Mice (BALB/c and NSG) Intramuscular 1.00E+11 F10 :100-200 μg/mL
CR6261 : 0.1-100 μg/mL
Balazs et al., 2013
AAV9 FI6 Mice Intranasal 1.00E+11 Adam et al., 2014
AAV9 MD3606 Mice Intranasal 4E+7 to 5E+9 Laursen et al., 2018
AAV8 R1a-B6 Mice Intramuscular 1.00E+11 0.5 - 1100 μg/mL Del Rosario et al., 2020

The efficacy of VIP has also been evaluated in non-human primate (NHP) models by several groups. Johnson et al. showed that a single intramuscular injection of an AAV1 encoding antibody-like immunoadhesin molecules in monkeys resulted in long-term (>1 year) expression of the biologically active protein that blocked SIV challenge [93]. Saunders et al. were the first to demonstrate delivery of an HIV bNAb (VRC07) in the non-human primate model, however these efforts yielded short-lived expression and significant anti-drug antibodies (ADA) targeting the simianized VRC07 transgene [94]. Immunosuppression with Cyclosporine during VIP was found to improve expression and reduce ADA responses [94]. When NHP-derived immunoadhesins (5L7 and 4L6) were converted to authentic IgG1 and delivered to rhesus monkeys using AAV, persisting levels of antibodies were achieved ranging from 1-270 μg/mL [95], but almost all animals exhibited ADA responses against at least one of the two antibodies. Notably, the monkey with the highest level of antibody (270 μg/mL of 5L7) in serum completely resisted six successive I.V. challenges. Recently, the authors reported that this monkey maintained 240-350 μg/mL of 5L7 antibody for over 6 years and still remains protected despite receiving multiple SIVmac239 challenges [96]. Reports from NHP have shown that host ADA can limit the concentration of delivered antibodies. The ADAs bind both heavy and light chains, but they predominantly target variable regions of delivered antibodies [97,98]. It has also been demonstrated that the magnitude of the ADA response correlates with the degree of sequence divergence of the delivered antibody to the germline sequence [98]. Interestingly, the isotype of the antibody has also been shown to influence the ADA response, as IgG2-Fc isotyped bNAbs induced significantly lower ADA and better protection against SHIV-AD8 challenges than their IgG1-Fc counterparts in the NHP model [99]. However, a recent study reported that VRC07-IgG2 exhibited reduced protection compared to other IgG subclasses in BLT mice. In fact, VRC07-IgG1 provided better protection relative to other IgG subclasses against vaginal challenge of HIV in BLT mice [91]. Additionally, intravenous administration of AAV8 using a liver-specific promoter to direct expression of the transgene in the liver has been reported to mitigate ADA response in macaques [100]. In addition to bNABs, AAV has been used to deliver eCD4-Ig, a fusion of CD4-Ig with a small CCR5-mimetic sulfopeptide to rhesus macaques [101]. Stable expression of rhesusized eCD4-Ig (17-77 μg/mL) was obtained in these animals, which were protected from multiple infectious challenges with SHIV-AD8. In a follow-up study, the authors reported that AAV1 inoculation of rh-CD4-Ig provided complete protection of macaques from intravenous challenge with SIVmac239 [102]. However, animals eventually succumbed to infection when the challenge dose was escalated. Other studies have also demonstrated long-term virologic suppression using AAV-mediated bNAb delivery. Martinez-Navio et al. showed in rhesus monkeys infected with SHIV-AD8, that a combination of AAV1s encoding three bNAbs (3BNC117, 10-1074, and 10E8), resulted in one monkey exhibiting 50-150 μg/mL of 3BNC117 and 10-1074 for over 2 years. Impressively, plasma viremia remained undetectable in this monkey for over 3 years [103]. The authors then extended this study with 12 monkeys using different combinations of antibodies and vectors. Long-term virologic suppression was observed in two monkeys that received a cocktail of four bNABs (N6, 35022, PGT128, and PGT145) delivered using AAV [103]. Overall, findings from the aforementioned NHP studies highlight the possibility of achieving a continued viral suppression from a single AAV-bNAb administration.

Based on the promising results of vectored immunoprophylaxis obtained from preclinical studies in mouse and NHP models, two Phase I clinical trials have been conducted to evaluate its safety and efficacy in humans. In the first human clinical trial, 16 healthy men aged 18-45 years were given an I.M. injection of AAV1 expressing PG9 [104]. Four different doses of AAV1-PG9 were tested, the lowest being 4x1012 vector genome copies and the highest being 1.2x1014 vector genome copies. No severe reactions or adverse effects were observed in these individuals indicating that antibody-expressing vectors are safe in humans. Although PG9 antibody was not detectable in the serum of these individuals by quantitative ELISA, the serum from four individuals showed detectable neutralizing activity against HIV pseudovirus. It is worth noting, however, that 10 out of 16 (62.5%) recipients in this study developed anti-PG9 antibodies, which could potentially have contributed to low expression or clearance of the transgene. It is also important to note that the lower limit of quantification of the assay used in this study was 2.5 μg/mL. It is plausible that some individuals may have exhibited PG9 levels below the detection limit and were therefore not measurable by the assay.

The results from a second Phase I clinical trial (VRC 603), which utilized AAV8-VRC07 have recently been published [97]. In this study, eight adults living with HIV on a stable antiretroviral regimen were enrolled and remained on ART throughout the study period. The participants received one of the three doses 5x1010 or 5x1011 or 2.5x1012 vector genome copies/kg intramuscularly. All eight individuals produced measurable amounts of serum VRC07, and in three individuals, the VRC07 concentration was >1 μg/mL. One participant receiving 5x1012 vg/kg achieved a VRC07 concentration of 3.3 μg/mL 1.5 years after AAV administration. In six of eight individuals, VRC07 concentrations remained stable near maximal concentration for up to 3 years of follow-up. The neutralizing activity of VRC07 in the serum was found to be equivalent to that of VRC07 produced in vitro, indicating that antibodies produced in vivo retained full biological activity. ADA responses were observed in three of the eight participants (38%), with responses primarily targeting the Fab portion of VRC07. Interestingly, one of these individuals continued to express VRC07 despite ADA, suggesting that these are not necessarily directly responsible for the loss of transgene expression. As in pre-clinical NHP studies, the choice of vector and transgene can influence ADA responses, perhaps explaining the less frequent ADA observed in the VRC 603 trial that employed AAV8-VRC07 as compared to the IAVI trial that used AAV1-PG9. Although challenges associated with host immune responses remain, these two human trials have clearly demonstrated the feasibility of vectored immunoprophylaxis as a means of producing long-lived bNAb expression in humans.

B cells for Delivery of bNAbs

Recent advances in genome engineering, largely stemming from the widespread use of lentiviral vectors and CRISPR-mediated gene targeting, have created new avenues for the delivery of antibodies by engineering the genome of B cells (Table 4. Current approaches for B cell-mediated delivery of antibodies).

Table 4.

Current approaches for B cell-mediated delivery of antibodies.

Type of therapy Antibody/Protein
targeted/expressed
Study type Cell/Organism
targeted
Comments References
Lentivirus b12 (anti-HIV bNAb) In vitro HSPCs-derived human B cells A secretion of over 1μg/mL was registered in culture supernatants upon transfection of these cells. Luo et al., 2009
mRNA and AAV CCR5 Ex vivo Primary human T cells and adult mobilized CD34+ PBSCs The authors modified and optimized the gene editing of the CCR5 locus as a potential therapy approaches against HIV. Sather et al., 2015
Lentivirus ARA3 (anti-HCV antibody) Ex vivo Primary human B cells The adoptive transfer of transduced B cells into NSG mice led to high levels of expression of the antibody Fusil et al., 2015
CRISPR-Cas9 Human factor IX (FIX) or B cell activating factor (BAFF) Ex vivo Primary human B cells Through several editions of primary cells, the authors achieved, among others, their differentiation protein-secreting plasma B cells. The expression of BAFF also led to the engraftment of these plasma cells into NSG mice. Hung et al., 2017
Lentivirus Targeting of TCR for the expression of several HIV scFv bNAbs (PGT145, VRC07, PGT128, and 10E8) Ex vivo Primary human T cells The cells were engineered to express chimeric antigen receptors (CAR) based on HIV bNAbs, leading to its activation and the killing of HIV-infected cells. Hale et al., 2017
CRISPR-Cas9 and AAV Human β-globin Ex vivo HSCs The collective results from this report set the basis for a CRISPR-based editing therapy for β-hemoglobinopathies. Dever et al., 2016
Lentivirus PGT128 and VRC01 (anti-HIV bNAb) Ex vivo HSPCs Transduction of cells and engraftment into humanized NSG mice resulted in the expression of these bNAs for the 9 months that this study lasted. PGT128 was also able to reduce HIV viremia and CD4+ T cells decline. Kuhlmann et al., 2019
CRISPR-Cas9 Targeting of CXCR4 and expression of ozoralizumab (anti-TNF-α nanobody) or adalimumab (anti-TNF-α mAb) Ex vivo Primary human B cells The editing performed in this report focused on the homologous recombination of the BCR loci, which actually results in the replacement of the original BCR by the new antibody. Greiner et al., 2019
CRISPR-Cas9 3BNC60 and 10-1074 (anti-HIV bNAb) Ex vivo Primary human and mouse B cells The adoptive transfer into mice led to high antibody titers with marked neutralizing potency. Hartweger et al., 2019
CRISPR-Cas9 and AAV 3BNC117 (anti-HIV bNAb) Ex vivo Primary mouse B cells Immunization with the antigen led to an increased accumulation of engineered cells in the germinal centers and increased rates of class switch recombination. A booster immunization also led to a memory response with a clonal selection pattern. Nahmad et al., 2020
CRISPR-Cas9 VRC01 (anti-HIV bNAb) Ex vivo Primary mouse B cells The adoptive transfer of the engineered cells to immunocompetent mice resulted in the establishment of memory and long-lived plasma cells able to secrete the bNAb properly and even undergo somatic hypermutation. Huang et al., 2020
CRISPR-Cas9 and AAV 3BNC117 (anti-HIV bNAb) In vivo Mouse Through the use of two different AAVs, one for Cas9 and a sgRNA for the IgH locus, and another one for 3BNC117, the expression of the antibody as the membrane-bound BCR was achieived. The bNAb-expressing B cells differentiated into memory and plasma cells in C57BL/6 mice. The in vivo expressed antibody exhibited a marked neutralizing potency. Nahmad et al., 2022
Lentivirus eCD4-Ig immunoadhesin (anti-HIV therapy) Ex vivo Primary human B cells Using this optimized lentivirus with a B cell-specific led to the high-efficient expression of this protein, capable of neutralizing HIV in vitro. Vamva et al., 2023

In 2009, Luo et al. reported the transduction of in vitro matured human B cells with lentiviruses coding for one of the first-identified bNAbs, b12 [105]. This transduction led to the secretion of over 1μg/mL of b12 in vitro. In 2015, Fusil et al. demonstrated that ex vivo lentiviral transduction of B cells, and the subsequent adoptive transfer of these cells into NSG mice, led to high levels of a hepatitis C virus-specific antibody [106]. While these early steps were promising, the efficiency of this approach improved dramatically with the emergence of CRIPSR-mediated gene targeting. In 2017, Hung et al. reported the first ex vivo transduction of proliferating B cells with CRISPR-Cas9 editing techniques, leading to significant secretion of the recombinant protein and the differentiation of these cells into plasma cells [107].

These findings, along with several others in hematopoietic stem cells, as well as primary human T, and B cells [108-112], led to the first reports of engineered human and mouse B cells expressing HIV bNAbs through CRISPR editing [113-115]. Hartweger et al. achieved the expression of 3BNC60 and 10-1074, both anti-HIV bNAbs, in primary human and mouse B cells through CRISPR-Cas9 editing [113]. The adoptive transfer of these cells back into B6 mice resulted in high serum concentrations of these antibodies that retained significant neutralizing capacities. Nahmad et al. and Huang et al. took these approaches a step further and demonstrated the establishment of long-lasting plasma cells, exhibiting affinity maturation, isotype switching, and clonal selection after the adoptive transfer of B cells engineered to express 3BNC117 or VRC01 [114,115]. These cells accumulated in germinal centers and, upon exposure to their antigen (HIV gp120), showed high rates of class switch recombination and affinity maturation, an adaptive immune response that was improved from that originally conferred to the adoptively transferred mice. This milestone for the induction of an evolving humoral response opened new possibilities for the adaption of bNAbs and B cells into in situ enhanced therapies [114,115].

Given that ex vivo transduction and adoptive transfer of engineered B cells into humans presents significant barriers to translation, Nahmad et al. used two different AAVs, one coding for the Staphylococcus aureus Cas9 and a guide RNA targeting the IgH locus, and the other containing the sequence for 3BNC117 flanked by homology arms matching the IgH locus [116]. This strategy led to the expression of 3BNC117 as a membrane-bound BCR of the transduced B cells. Joint administration of these AAVs promoted the clonal expansion and differentiation of bNAb-expressing B cells into memory and plasma cells in C57BL/6 mice. Upon immunization with the HIV gp120 antigen, circulating 3BNC117 reached up to 2μg/mL. However, to achieve these levels of transduction, B cells had to be previously primed to induce their activation. Of note, the authors reported unwanted cleavage of off-target genome sites with this approach, albeit at low frequency [116]. Finally, of significant note is the lentiviral-mediated B cell transduction strategy reported recently by Vamva et al. to express the eCD4-Ig immunoadhesin [117]. Through the use of an optimized lentivirus containing the B cell-specific promoter EμB29, the authors achieved efficient expression of this protein in human B cells, capable of neutralizing HIV in vitro. Further studies are needed to test the feasibility of this approach in vivo and its protection efficacy [117]. While significantly less mature than other platforms, the field of B cell engineering is making rapid advances towards clinical translation of these technologies.

CONCLUSION

Given the promise of bNAbs for HIV prevention and therapy, multiple efforts are under development to efficiently and conveniently deliver these proteins to patients. While each of the reviewed approaches has intrinsic benefits, they will all need to achieve certain parameters to be clinically useful. A successful approach will need to be capable of eliciting sufficiently high titers of antibodies to be clinically useful. Recent studies of antibody-mediated prevention in humans have suggested that this could require a steady-state concentration of as much as 10 μg/mL of VRC01 [5]. Similarly, a successful approach will need to provide bNAb expression lasting substantially longer than what can be achieved via passive transfer studies. Given the remarkable safety of bNAb passive transfer, any competing approaches will need to demonstrate at least equivalent metrics before they can be deployed widely.

Key points.

  1. Different gene-based delivery approaches have been developed for the expression of bNAbs.

  2. The administration of bNAbs through DNA-based platforms has proven to be effective in animal models due to its simplicity, rapid manufacturing, and the lack of vector-directed immune responses.

  3. The COVID-19 pandemic resulted in rapid translation of mRNA-mediated gene delivery for vaccination; this technology is now being tested for bNAb expression.

  4. AAV mediated bNAb delivery has achieved long-term antibody expression in humans and is the furthest developed approach.

  5. Lentiviral- and CRISPR-mediated engineering of bNAb expression by B cells leads to class switch recombination and further affinity maturation.

Acknowledgements

This work was supported by NIAID K22AI102769 to A.B.B., NIDA Avenir New Innovator Award DP2DA040254 to A.B.B., the MGH Transformative Scholars award to A.B.B., and funding from the Charles H. Hood Foundation to A.B.B. This independent research was supported by the Gilead Sciences Research Scholars Program in HIV to A.B.B. Subaward to DBW on UM1 AI164570 co-funded by NHLBI, NINDS, NIDDK, NIDA; WW Smith distinguished professor in cancer research to DBW and NIAID/ Martin Delaney Collaboratories for HIV Cure Research and NIH R01 AI141236 to DBW

Footnotes

Financial support and sponsorship

None.

Conflicts of interest

A.B.B. is a named inventor on patent US9527904B2 held by the California Institute of Technology describing AAV-mediated antibody delivery and holds equity in the following commercial partners: Cure Systems (Founder). D.B.W. has received grant funding, participates in industry collaborations, has received speaking honoraria, and has received fees for consulting, including serving on scientific review committees. Remunerations received by D.B.W. include direct payments and equity/options. D.B.W. also discloses the following associations with commercial partners: Geneos (consultant/advisory board), AstraZeneca (advisory board, speaker), Inovio (board of directors, consultant), Sanofi (advisory board), BBI (advisory board), Pfizer (advisory board), Flagship (consultant), and Advaccine (consultant). The other authors declare that they have no competing interests.

REFERENCES:

Recent references of additional notoriety published within the past 18 months are bulleted and annotated.

References have one bullet (*) for special interest and two bullets (**) for outstanding interest.

Annotations provide a brief description of the paper's importance.

Bibliography

  • 1.Haynes BF, Wiehe K, Borrow P, Saunders KO, Korber B, Wagh K, McMichael AJ, Kelsoe G, Hahn BH, Alt F, et al. : Strategies for HIV-1 vaccines that induce broadly neutralizing antibodies. Nat Rev Immunol 2023, 23:142–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kumar S, Singh S, Luthra K: An Overview of Human Anti-HIV-1 Neutralizing Antibodies against Diverse Epitopes of HIV-1. ACS Omega 2023, 8:7252–7261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Corey L, Gilbert PB, Juraska M, Montefiori DC, Morris L, Karuna ST, Edupuganti S, Mgodi NM, deCamp AC, Rudnicki E, et al. : Two Randomized Trials of Neutralizing Antibodies to Prevent HIV-1 Acquisition. N Engl J Med 2021, 384:1003–1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Julg B, Barouch DH: Neutralizing antibodies for HIV-1 prevention. Curr Opin HIV AIDS 2019, 14:318–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Gilbert PB, Huang Y, deCamp AC, Karuna S, Zhang Y, Magaret CA, Giorgi EE, Korber B, Edlefsen PT, Rossenkhan R, et al. : Neutralization titer biomarker for antibody-mediated prevention of HIV-1 acquisition. Nat Med 2022, 28:1924–1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Caskey M, Klein F, Lorenzi JCC, Seaman MS, West AP, Buckley N, Kremer G, Nogueira L, Braunschweig M, Scheid JF, et al. : Viraemia suppressed in HIV-1-infected humans by broadly neutralizing antibody 3BNC117. Nature 2015, doi: 10.1038/nature14411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bar KJ, Sneller MC, Harrison LJ, Justement JS, Overton ET, Petrone ME, Salantes DB, Seamon CA, Scheinfeld B, Kwan RW, et al. : Effect of HIV Antibody VRC01 on Viral Rebound after Treatment Interruption. The New England journal of medicine 2016, 375:2037–2050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Caskey M, Schoofs T, Gruell H, Settler A, Karagounis T, Kreider EF, Murrell B, Pfeifer N, Nogueira L, Oliveira TY, et al. : Antibody 10-1074 suppresses viremia in HIV-1-infected individuals. Nature Medicine 2017, 23:185–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mendoza P, Gruell H, Nogueira L, Pai JA, Butler AL, Millard K, Lehmann C, Suárez I, Oliveira TY, Lorenzi JCC, et al. : Combination therapy with anti-HIV-1 antibodies maintains viral suppression. Nature 2018, doi: 10.1038/s41586-018-0531-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Danko I, Wolff JA: Direct gene transfer into muscle. Vaccine 1994, 12:1499–1502. [DOI] [PubMed] [Google Scholar]
  • 11.Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A, Felgner PL: Direct gene transfer into mouse muscle in vivo. Science 1990, 247:1465–1468. [DOI] [PubMed] [Google Scholar]
  • 12.Sung Y, Kim S: Recent advances in the development of gene delivery systems. Biomater Res 2019, 23:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Gary EN, Weiner DB: DNA vaccines: prime time is now. Current Opinion in Immunology 2020, 65:21–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Blakney AK, Bekker L-G: DNA vaccines join the fight against COVID-19. The Lancet 2022, 399:1281–1282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Flingai S, Plummer EM, Patel A, Shresta S, Mendoza JM, Broderick KE, Sardesai NY, Muthumani K, Weiner DB: Protection against dengue disease by synthetic nucleic acid antibody prophylaxis/immunotherapy. Sci Rep 2015, 5:12616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Aihara H, Miyazaki J: Gene transfer into muscle by electroporation in vivo. Nat Biotechnol 1998, 16:867–870. [DOI] [PubMed] [Google Scholar]
  • 17.Perez N, Bigey P, Scherman D, Danos O, Piechaczyk M, Pelegrin M: Regulatable systemic production of monoclonal antibodies by in vivo muscle electroporation. Genetic vaccines and therapy 2004, 2:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sheets RL, Stein J, Manetz TS, Andrews C, Bailer R, Rathmann J, Gomez PL: Toxicological Safety Evaluation of DNA Plasmid Vaccines against HIV-1, Ebola, Severe Acute Respiratory Syndrome, or West Nile Virus Is Similar Despite Differing Plasmid Backbones or Gene-Inserts. Toxicological Sciences 2006, 91:620–630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Morrow MP, Pankhong P, Weiner DB: Design and characterization of a plasmid vector system capable of rapid generation of antibodies of multiple isotypes and specificities. Biotechnol Lett 2009, 31:13–22. [DOI] [PubMed] [Google Scholar]
  • 20.Yamazaki T, Nagashima M, Ninomiya D, Arai Y, Teshima Y, Fujimoto A, Ainai A, Hasegawa H, Chiba J: Passive immune-prophylaxis against influenza virus infection by the expression of neutralizing anti-hemagglutinin monoclonal antibodies from plasmids. Jpn J Infect Dis 2011, 64:40–49. [PubMed] [Google Scholar]
  • 21.Lopes A, Vanvarenberg K, Préat V, Vandermeulen G: Codon-Optimized P1A-Encoding DNA Vaccine: Toward a Therapeutic Vaccination against P815 Mastocytoma. Molecular Therapy - Nucleic Acids 2017, 8:404–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Andrews CD, Luo Y, Sun M, Yu J, Goff AJ, Glass PJ, Padte NN, Huang Y, Ho DD: In Vivo Production of Monoclonal Antibodies by Gene Transfer via Electroporation Protects against Lethal Influenza and Ebola Infections. Molecular Therapy - Methods & Clinical Development 2017, 7:74–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Elliott STC, Kallewaard NL, Benjamin E, Wachter-Rosati L, McAuliffe JM, Patel A, Smith TRF, Schultheis K, Park DH, Flingai S, et al. : DMAb inoculation of synthetic cross reactive antibodies protects against lethal influenza A and B infections. npj Vaccines 2017, 2:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Patel A, Park DH, Davis CW, Smith TRF, Leung A, Tierney K, Bryan A, Davidson E, Yu X, Racine T, et al. : In Vivo Delivery of Synthetic Human DNA-Encoded Monoclonal Antibodies Protect against Ebolavirus Infection in a Mouse Model. Cell Reports 2018, 25:1982–1993.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wang Y, Esquivel R, Flingai S, Schiller ZA, Kern A, Agarwal S, Chu J, Patel A, Sullivan K, Wise MC, et al. : Anti-OspA DNA-Encoded Monoclonal Antibody Prevents Transmission of Spirochetes in Tick Challenge Providing Sterilizing Immunity in Mice. The Journal of Infectious Diseases 2019, 219:1146–1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Esquivel RN, Patel A, Kudchodkar SB, Park DH, Stettler K, Beltramello M, Allen JW, Mendoza J, Ramos S, Choi H, et al. : In Vivo Delivery of a DNA-Encoded Monoclonal Antibody Protects Non-human Primates against Zika Virus. Molecular Therapy 2019, 27:974–985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Andrews CD, Huang Y, Ho DD, Liberatore RA: In vivo expressed biologics for infectious disease prophylaxis: rapid delivery of DNA-based antiviral antibodies. Emerging Microbes & Infections 2020, 9:1523–1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Choi H, Kudchodkar SB, Reuschel EL, Asija K, Borole P, Agarwal S, Van Gorder L, Reed CC, Gulendran G, Ramos S, et al. : Synthetic nucleic acid antibody prophylaxis confers rapid and durable protective immunity against Zika virus challenge. Human Vaccines & Immunotherapeutics 2020, 16:907–918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wise MC, Xu Z, Tello-Ruiz E, Beck C, Trautz A, Patel A, Elliott STC, Chokkalingam N, Kim S, Kerkau MG, et al. : In vivo delivery of synthetic DNA–encoded antibodies induces broad HIV-1–neutralizing activity. Journal of Clinical Investigation 2020, 130:827–837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.McNee A, Smith TRF, Holzer B, Clark B, Bessell E, Guibinga G, Brown H, Schultheis K, Fisher P, Ramos S, et al. : Establishment of a Pig Influenza Challenge Model for Evaluation of Monoclonal Antibody Delivery Platforms. The Journal of Immunology 2020, 205:648–660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Muthumani K, Flingai S, Wise M, Tingey C, Ugen KE, Weiner DB: Optimized and enhanced DNA plasmid vector based in vivo construction of a neutralizing anti-HIV-1 envelope glycoprotein Fab. Human vaccines & immunotherapeutics 2013, 9:2253–2262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Muthumani K, Block P, Flingai S, Muruganantham N, Chaaithanya IK, Tingey C, Wise M, Reuschel EL, Chung C, Muthumani A, et al. : Rapid and Long-Term Immunity Elicited by DNA-Encoded Antibody Prophylaxis and DNA Vaccination Against Chikungunya Virus. J Infect Dis 2016, 214:369–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Patel A, DiGiandomenico A, Keller AE, Smith TRF, Park DH, Ramos S, Schultheis K, Elliott STC, Mendoza J, Broderick KE, et al. : An engineered bispecific DNA-encoded IgG antibody protects against Pseudomonas aeruginosa in a pneumonia challenge model. Nat Commun 2017, 8:637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Andrews CD, Luo Y, Sun M, Yu J, Goff AJ, Glass PJ, Padte NN, Huang Y, Ho DD: In Vivo Production of Monoclonal Antibodies by Gene Transfer via Electroporation Protects against Lethal Influenza and Ebola Infections. Molecular Therapy - Methods & Clinical Development 2017, 7:74–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Patel A, Park DH, Davis CW, Smith TRF, Leung A, Tierney K, Bryan A, Davidson E, Yu X, Racine T, et al. : In Vivo Delivery of Synthetic Human DNA-Encoded Monoclonal Antibodies Protect against Ebolavirus Infection in a Mouse Model. Cell Reports 2018, 25:1982–1993.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Esquivel RN, Patel A, Kudchodkar SB, Park DH, Stettler K, Beltramello M, Allen JW, Mendoza J, Ramos S, Choi H, et al. : In Vivo Delivery of a DNA-Encoded Monoclonal Antibody Protects Non-human Primates against Zika Virus. Molecular Therapy 2019, 27:974–985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Schultheis K, Pugh HM, Oh J, Nguyen J, Yung B, Reed C, Cooch N, Chen J, Yan J, Muthumani K, et al. : Active immunoprophylaxis with a synthetic DNA-encoded monoclonal anti-respiratory syncytial virus scFv-Fc fusion protein confers protection against infection and durable activity. Human Vaccines & Immunotherapeutics 2020, 16:2165–2175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Parzych EM, Gulati S, Zheng B, Bah MA, Elliott STC, Chu JD, Nowak N, Reed GW, Beurskens FJ, Schuurman J, et al. : Synthetic DNA Delivery of an Optimized and Engineered Monoclonal Antibody Provides Rapid and Prolonged Protection against Experimental Gonococcal Infection. mBio 2021, 12:e00242–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Tursi NJ, Reeder SM, Flores-Garcia Y, Bah MA, Mathis-Torres S, Salgado-Jimenez B, Esquivel R, Xu Z, Chu JD, Humeau L, et al. : Engineered DNA-encoded monoclonal antibodies targeting Plasmodium falciparum circumsporozoite protein confer single dose protection in a murine malaria challenge model. Sci Rep 2022, 12:14313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Parzych EM, Du J, Ali AR, Schultheis K, Frase D, Smith TRF, Cui J, Chokkalingam N, Tursi NJ, Andrade VM, et al. : DNA-delivered antibody cocktail exhibits improved pharmacokinetics and confers prophylactic protection against SARS-CoV-2. Nat Commun 2022, 13:5886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kim H, Danishmalik SN, Hwang H, Sin J-I, Oh J, Cho Y, Lee H, Jeong M, Kim S-H, Hong HJ: Gene therapy using plasmid DNA-encoded anti-HER2 antibody for cancers that overexpress HER2. Cancer Gene Ther 2016, 23:341–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hollevoet K, De Smidt E, Geukens N, Declerck P: Prolonged in vivo expression and anti-tumor response of DNA-based anti-HER2 antibodies. Oncotarget 2018, 9:13623–13636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Perales-Puchalt A, Duperret EK, Yang X, Hernandez P, Wojtak K, Zhu X, Jung S-H, Tello-Ruiz E, Wise MC, Montaner LJ, et al. : DNA-encoded bispecific T cell engagers and antibodies present long-term antitumor activity. JCI Insight 2019, 4:e126086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bordoloi D, Bhojnagarwala PS, Perales-Puchalt A, Kulkarni AJ, Zhu X, Liaw K, O’Connell RP, Park DH, Kulp DW, Zhang R, et al. : A mAb against surface-expressed FSHR engineered to engage adaptive immunity for ovarian cancer immunotherapy. JCI Insight 2022, 7:e162553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Muthumani K, Marnin L, Kudchodkar SB, Perales-Puchalt A, Choi H, Agarwal S, Scott VL, Reuschel EL, Zaidi FI, Duperret EK, et al. : Novel prostate cancer immunotherapy with a DNA-encoded anti-prostate-specific membrane antigen monoclonal antibody. Cancer Immunol Immunother 2017, 66:1577–1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Duperret EK, Trautz A, Stoltz R, Patel A, Wise MC, Perales-Puchalt A, Smith T, Broderick KE, Masteller E, Kim JJ, et al. : Synthetic DNA-Encoded Monoclonal Antibody Delivery of Anti–CTLA-4 Antibodies Induces Tumor Shrinkage In Vivo. Cancer Research 2018, 78:6363–6370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hollevoet K, Thomas D, Compernolle G, Vermeire G, De Smidt E, De Vleeschauwer S, Smith TRF, Fisher PD, Dewilde M, Geukens N, et al. : Clinically relevant dosing and pharmacokinetics of DNA-encoded antibody therapeutics in a sheep model. Front Oncol 2022, 12:1017612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Park DH, Liaw K, Bhojnagarwala P, Zhu X, Choi J, Ali AR, Bordoloi D, Gary EN, O’Connell RP, Kulkarni A, et al. : Multivalent in vivo delivery of DNA-encoded bispecific T cell engagers effectively controls heterogeneous GBM tumors and mitigates immune escape. Molecular Therapy - Oncolytics 2023, 28:249–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Bhojnagarwala PS, O’Connell RP, Park D, Liaw K, Ali AR, Bordoloi D, Cassel J, Tursi NJ, Gary E, Weiner DB: In vivo DNA-launched bispecific T cell engager targeting IL-13Rα2 controls tumor growth in an animal model of glioblastoma multiforme. Mol Ther Oncolytics 2022, 26:289–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Barbier AJ, Jiang AY, Zhang P, Wooster R, Anderson DG: The clinical progress of mRNA vaccines and immunotherapies. Nat Biotechnol 2022, 40:840–854. [DOI] [PubMed] [Google Scholar]
  • 51.Chaudhary N, Weissman D, Whitehead KA: mRNA vaccines for infectious diseases: principles, delivery and clinical translation. Nat Rev Drug Discov 2021, 20:817–838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Deal CE, Richards AF, Yeung T, Maron MJ, Wang Z, Lai Y-T, Fritz BR, Himansu S, Narayanan E, Liu D, et al. : mRNA delivery of dimeric human IgA protects mucosal tissues from bacterial infection. Immunology; 2023. [Google Scholar]
  • 53. Fortner A, Bucur O: mRNA-based vaccine technology for HIV. Discoveries (Craiova) 2022, 10:e150. ** This was the first use of mRNA to express dimeric IgA antibody.
  • 54.Nelson J, Sorensen EW, Mintri S, Rabideau AE, Zheng W, Besin G, Khatwani N, Su SV, Miracco EJ, Issa WJ, et al. : Impact of mRNA chemistry and manufacturing process on innate immune activation. Sci Adv 2020, 6:eaaz6893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Alexopoulou L, Holt AC, Medzhitov R, Flavell RA: Recognition of double-stranded RNA and activation of NF-κB by Toll-like receptor 3. Nature 2001, 413:732–738. [DOI] [PubMed] [Google Scholar]
  • 56.Rehwinkel J, Gack MU: RIG-I-like receptors: their regulation and roles in RNA sensing. Nat Rev Immunol 2020, 20:537–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Duic I, Tadakuma H, Harada Y, Yamaue R, Deguchi K, Suzuki Y, Yoshimura SH, Kato H, Takeyasu K, Fujita T: Viral RNA recognition by LGP2 and MDA5, and activation of signaling through step-by-step conformational changes. Nucleic Acids Research 2020, 48:11664–11674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Houseley J, Tollervey D: The Many Pathways of RNA Degradation. Cell 2009, 136:763–776. [DOI] [PubMed] [Google Scholar]
  • 59.Kim SC, Sekhon SS, Shin W-R, Ahn G, Cho B-K, Ahn J-Y, Kim Y-H: Modifications of mRNA vaccine structural elements for improving mRNA stability and translation efficiency. Mol Cell Toxicol 2022, 18:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Zhang C, Maruggi G, Shan H, Li J: Advances in mRNA Vaccines for Infectious Diseases. Front Immunol 2019, 10:594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Pardi N, Secreto AJ, Shan X, Debonera F, Glover J, Yi Y, Muramatsu H, Ni H, Mui BL, Tam YK, et al. : Administration of nucleoside-modified mRNA encoding broadly neutralizing antibody protects humanized mice from HIV-1 challenge. Nat Commun 2017, 8:14630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lindsay KE, Vanover D, Thoresen M, King H, Xiao P, Badial P, Araínga M, Park SB, Tiwari PM, Peck HE, et al. : Aerosol Delivery of Synthetic mRNA to Vaginal Mucosa Leads to Durable Expression of Broadly Neutralizing Antibodies against HIV. Mol Ther 2020, 28:805–819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Narayanan E, Falcone S, Elbashir SM, Attarwala H, Hassett K, Seaman MS, Carfi A, Himansu S: Rational Design and In Vivo Characterization of mRNA-Encoded Broadly Neutralizing Antibody Combinations against HIV-1. Antibodies 2022, 11:67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kose N, Fox JM, Sapparapu G, Bombardi R, Tennekoon RN, de Silva AD, Elbashir SM, Theisen MA, Humphris-Narayanan E, Ciaramella G, et al. : A lipid-encapsulated mRNA encoding a potently neutralizing human monoclonal antibody protects against chikungunya infection. Sci Immunol 2019, 4:eaaw6647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.August A, Attarwala HZ, Himansu S, Kalidindi S, Lu S, Pajon R, Han S, Lecerf J-M, Tomassini JE, Hard M, et al. : A phase 1 trial of lipid-encapsulated mRNA encoding a monoclonal antibody with neutralizing activity against Chikungunya virus. Nat Med 2021, 27:2224–2233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chen B, Chen Y, Li J, Wang C, Song W, Wen Y, Lin J, Wu Y, Ying T: A Single Dose of Anti-HBsAg Antibody-Encoding mRNA-LNPs Suppressed HBsAg Expression: a Potential Cure of Chronic Hepatitis B Virus Infection. mBio 2022, 13:e0161222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Thran M, Mukherjee J, Pönisch M, Fiedler K, Thess A, Mui BL, Hope MJ, Tam YK, Horscroft N, Heidenreich R, et al. : mRNA mediates passive vaccination against infectious agents, toxins, and tumors. EMBO Mol Med 2017, 9:1434–1447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Sabnis S, Kumarasinghe ES, Salerno T, Mihai C, Ketova T, Senn JJ, Lynn A, Bulychev A, McFadyen I, Chan J, et al. : A Novel Amino Lipid Series for mRNA Delivery: Improved Endosomal Escape and Sustained Pharmacology and Safety in Non-human Primates. Mol Ther 2018, 26:1509–1519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Van Hoecke L, Verbeke R, De Vlieger D, Dewitte H, Roose K, Van Nevel S, Krysko O, Bachert C, Schepens B, Lentacker I, et al. : mRNA Encoding a Bispecific Single Domain Antibody Construct Protects against Influenza A Virus Infection in Mice. Mol Ther Nucleic Acids 2020, 20:777–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Mucker EM, Thiele-Suess C, Baumhof P, Hooper JW: Lipid nanoparticle delivery of unmodified mRNAs encoding multiple monoclonal antibodies targeting poxviruses in rabbits. Molecular Therapy - Nucleic Acids 2022, 28:847–858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Tiwari PM, Vanover D, Lindsay KE, Bawage SS, Kirschman JL, Bhosle S, Lifland AW, Zurla C, Santangelo PJ: Engineered mRNA-expressed antibodies prevent respiratory syncytial virus infection. Nat Commun 2018, 9:3999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Li J-Q, Zhang Z-R, Zhang H-Q, Zhang Y-N, Zeng X-Y, Zhang Q-Y, Deng C-L, Li X-D, Zhang B, Ye H-Q: Intranasal delivery of replicating mRNA encoding neutralizing antibody against SARS-CoV-2 infection in mice. Signal Transduct Target Ther 2021, 6:369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Deng Y-Q, Zhang N-N, Zhang Y-F, Zhong X, Xu S, Qiu H-Y, Wang T-C, Zhao H, Zhou C, Zu S-L, et al. : Lipid nanoparticle-encapsulated mRNA antibody provides long-term protection against SARS-CoV-2 in mice and hamsters. Cell Res 2022, 32:375–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Vanover D, Zurla C, Peck HE, Orr-Burks N, Joo JY, Murray J, Holladay N, Hobbs RA, Jung Y, Chaves LCS, et al. : Nebulized mRNA-Encoded Antibodies Protect Hamsters from SARS-CoV-2 Infection. Adv Sci (Weinh) 2022, 9:e2202771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zhang Y-N, Zhang H-Q, Wang G-F, Zhang Z-R, Li J-Q, Chen X-L, Hu Y-Y, Zeng X-Y, Shi Y-J, Wang J, et al. : Intranasal delivery of replicating mRNA encoding hACE2-targeting antibody against SARS-CoV-2 Omicron infection in the hamster. Antiviral Res 2023, 209:105507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Erasmus JH, Archer J, Fuerte-Stone J, Khandhar AP, Voigt E, Granger B, Bombardi RG, Govero J, Tan Q, Durnell LA, et al. : Intramuscular Delivery of Replicon RNA Encoding ZIKV-117 Human Monoclonal Antibody Protects against Zika Virus Infection. Mol Ther Methods Clin Dev 2020, 18:402–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Mukherjee J, Ondeck CA, Tremblay JM, Archer J, Debatis M, Foss A, Awata J, Erasmus JH, McNutt PM, Shoemaker CB: Intramuscular delivery of formulated RNA encoding six linked nanobodies is highly protective for exposures to three Botulinum neurotoxin serotypes. Sci Rep 2022, 12:11664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Panova EA, Kleymenov DA, Shcheblyakov DV, Bykonia EN, Mazunina EP, Dzharullaeva AS, Zolotar AN, Derkaev AA, Esmagambetov IB, Sorokin II, et al. : Single-domain antibody delivery using an mRNA platform protects against lethal doses of botulinum neurotoxin A. Front Immunol 2023, 14:1098302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Stadler CR, Bähr-Mahmud H, Celik L, Hebich B, Roth AS, Roth RP, Karikó K, Türeci Ö, Sahin U: Elimination of large tumors in mice by mRNA-encoded bispecific antibodies. Nat Med 2017, 23:815–817. [DOI] [PubMed] [Google Scholar]
  • 80.Rybakova Y, Kowalski PS, Huang Y, Gonzalez JT, Heartlein MW, DeRosa F, Delcassian D, Anderson DG: mRNA Delivery for Therapeutic Anti-HER2 Antibody Expression In Vivo. Mol Ther 2019, 27:1415–1423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wang Y, Tiruthani K, Li S, Hu M, Zhong G, Tang Y, Roy S, Zhang L, Tan J, Liao C, et al. : mRNA Delivery of a Bispecific Single-Domain Antibody to Polarize Tumor-Associated Macrophages and Synergize Immunotherapy against Liver Malignancies. Adv Mater 2021, 33:e2007603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Wu L, Wang W, Tian J, Qi C, Cai Z, Yan W, Xuan S, Shang A: Engineered mRNA-expressed bispecific antibody prevent intestinal cancer via lipid nanoparticle delivery. Bioengineered 2021, 12:12383–12393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Wu L, Wang W, Tian J, Qi C, Cai Z, Yan W, Xuan S, Shang A: Intravenous Delivery of RNA Encoding Anti-PD-1 Human Monoclonal Antibody for Treating Intestinal Cancer. J Cancer 2022, 13:579–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Huang C, Duan X, Wang J, Tian Q, Ren Y, Chen K, Zhang Z, Li Y, Feng Y, Zhong K, et al. : Lipid Nanoparticle Delivery System for mRNA Encoding B7H3-redirected Bispecific Antibody Displays Potent Antitumor Effects on Malignant Tumors. Adv Sci (Weinh) 2023, 10:e2205532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Krah S, Kolmar H, Becker S, Zielonka S: Engineering IgG-Like Bispecific Antibodies—An Overview. Antibodies 2018, 7:28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lewis AD, Chen R, Montefiori DC, Johnson PR, Clark KR: Generation of Neutralizing Activity against Human Immunodeficiency Virus Type 1 in Serum by Antibody Gene Transfer. Journal of Virology 2002, 76:8769–8775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Fang J, Qian J-J, Yi S, Harding TC, Tu GH, VanRoey M, Jooss K: Stable antibody expression at therapeutic levels using the 2A peptide. Nature Biotechnology 2005, 23:584–590. [DOI] [PubMed] [Google Scholar]
  • 88.Fang J, Yi S, Simmons A, Tu GH, Nguyen M, Harding TC, VanRoey M, Jooss K: An Antibody Delivery System for Regulated Expression of Therapeutic Levels of Monoclonal Antibodies In Vivo. Molecular therapy: the journal of the American Society of Gene Therapy 2007, 15:1153–1159. [DOI] [PubMed] [Google Scholar]
  • 89.Balazs AB, Chen J, Hong CM, Rao DS, Yang L, Baltimore D: Antibody-based protection against HIV infection by vectored immunoprophylaxis. Nature 2011, 481:81–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Balazs AB, Ouyang Y, Hong CM, Chen J, Nguyen SM, Rao DS, An DS, Baltimore D: Vectored immunoprophylaxis protects humanized mice from mucosal HIV transmission. Nature Medicine 2014, 20:296–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Brady JM, Phelps M, MacDonald SW, Lam EC, Nitido A, Parsons D, Boutros CL, Deal CE, Garcia-Beltran WF, Tanno S, et al. : Antibody-mediated prevention of vaginal HIV transmission is dictated by IgG subclass in humanized mice. Sci Transl Med 2022, 14:eabn9662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Horwitz JA, Halper-Stromberg A, Mouquet H, Gitlin AD, Tretiakova A, Eisenreich TR, Malbec M, Gravemann S, Billerbeck E, Dorner M, et al. : HIV-1 suppression and durable control by combining single broadly neutralizing antibodies and antiretroviral drugs in humanized mice. Proc Natl Acad Sci U S A 2013, 110:16538–16543. ** First use of AAV to deliver different IgG subclasses in vivo which demonstrated the contribution of innate immunity towards HIV prevention in humanized mice.
  • 93.Johnson PR, Schnepp BC, Zhang J, Connell MJ, Greene SM, Yuste E, Desrosiers RC, Clark KR: Vector-mediated gene transfer engenders long-lived neutralizing activity and protection against SIV infection in monkeys. Nature Medicine 2009, 15:901–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Saunders KO, Wang L, Joyce MG, Yang Z-Y, Balazs AB, Cheng C, Ko S-Y, Kong W-P, Rudicell RS, Georgiev IS, et al. : Broadly Neutralizing Human Immunodeficiency Virus Type 1 Antibody Gene Transfer Protects Nonhuman Primates from Mucosal Simian-Human Immunodeficiency Virus Infection. Journal of Virology 2015, 89:8334–8345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Fuchs SP, Martinez-Navio JM, Piatak M, Lifson JD, Gao G, Desrosiers RC: AAV-Delivered Antibody Mediates Significant Protective Effects against SIVmac239 Challenge in the Absence of Neutralizing Activity. PLoS Pathogens 2015, 11:e1005090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Martinez-Navio JM, Fuchs SP, Mendes DE, Rakasz EG, Gao G, Lifson JD, Desrosiers RC: Long-Term Delivery of an Anti-SIV Monoclonal Antibody With AAV. Frontiers in Immunology 2020, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Casazza JP, Cale EM, Narpala S, Yamshchikov GV, Coates EE, Hendel CS, Novik L, Holman LA, Widge AT, Apte P, et al. : Safety and tolerability of AAV8 delivery of a broadly neutralizing antibody in adults living with HIV: a phase 1, dose-escalation trial. Nat Med 2022, 28:1022–1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Martinez-Navio JM, Fuchs SP, Pedreño-López S, Rakasz EG, Gao G, Desrosiers RC: Host Anti-antibody Responses Following Adeno-associated Virus–mediated Delivery of Antibodies Against HIV and SIV in Rhesus Monkeys. Molecular Therapy 2016, 24:76–86. ** This was the first human study (VRC603) to demonstrate sustained expression of a broadly neutralizing antibody in humans. This was achieved using a single administration of AAV8 expressing VRC07 which reached up to 3 μg/mL for at least three years.
  • 99.<J/>Gardner MR: Anti-drug Antibody Responses Impair Prophylaxis Mediated by AAV-Delivered HIV-1 Broadly Neutralizing Antibodies ∣ Elsevier Enhanced Reader. 2019, doi: 10.1016/j.ymthe.2019.01.004. [DOI] [PMC free article] [PubMed]
  • 100.Fuchs SP, Martinez-Navio JM, Rakasz EG, Gao G, Desrosiers RC: Liver-directed but not muscle-directed AAV-antibody gene transfer limits humoral immune responses in rhesus monkeys. Molecular Therapy — Methods & Clinical Development 2019, doi: 10.1016/j.omtm.2019.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Gardner MR, Kattenhorn LM, Kondur HR, von Schaewen M, Dorfman T, Chiang JJ, Haworth KG, Decker JM, Alpert MD, Bailey CC, et al. : AAV-expressed eCD4-Ig provides durable protection from multiple SHIV challenges. Nature 2015, doi: 10.1038/nature14264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Gardner MR, Fellinger CH, Kattenhorn LM, Davis-Gardner ME, Weber JA, Alfant B, Zhou AS, Prasad NR, Kondur HR, Newton WA, et al. : AAV-delivered eCD4-Ig protects rhesus macaques from high-dose SIVmac239 challenges. Science Translational Medicine 2019, 11:eaau5409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Martinez-Navio JM, Fuchs SP, Pantry SN, Lauer WA, Duggan NN, Keele BF, Rakasz EG, Gao G, Lifson JD, Desrosiers RC: Adeno-Associated Virus Delivery of Anti-HIV Monoclonal Antibodies Can Drive Long-Term Virologic Suppression. Immunity 2019, doi: 10.1016/j.immuni.2019.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Priddy FH, Lewis DJM, Gelderblom HC, Hassanin H, Streatfield C, LaBranche C, Hare J, Cox JH, Dally L, Bendel D, et al. : Adeno-associated virus vectored immunoprophylaxis to prevent HIV in healthy adults: a phase 1 randomised controlled trial. Lancet HIV 2019, 6:e230–e239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Luo XM, Maarschalk E, O’Connell RM, Wang P, Yang L, Baltimore D: Engineering human hematopoietic stem/progenitor cells to produce a broadly neutralizing anti-HIV antibody after in vitro maturation to human B lymphocytes. Blood 2009, 113:1422–1431. [DOI] [PubMed] [Google Scholar]
  • 106.Fusil F, Calattini S, Amirache F, Mancip J, Costa C, Robbins JB, Douam F, Lavillette D, Law M, Defrance T, et al. : A Lentiviral Vector Allowing Physiologically Regulated Membrane-anchored and Secreted Antibody Expression Depending on B-cell Maturation Status. Molecular Therapy 2015, 23:1734–1747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Hung KL, Meitlis I, Hale M, Chen C-Y, Singh S, Jackson SW, Miao CH, Khan IF, Rawlings DJ, James RG: Engineering Protein-Secreting Plasma Cells by Homology-Directed Repair in Primary Human B Cells. Molecular Therapy 2018, 26:456–467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Hale M, Mesojednik T, Romano Ibarra GS, Sahni J, Bernard A, Sommer K, Scharenberg AM, Rawlings DJ, Wagner TA: Engineering HIV-Resistant, Anti-HIV Chimeric Antigen Receptor T Cells. Molecular Therapy 2017, 25:570–579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Sather BD, Romano Ibarra GS, Sommer K, Curinga G, Hale M, Khan IF, Singh S, Song Y, Gwiazda K, Sahni J, et al. : Efficient modification of CCR5 in primary human hematopoietic cells using a megaTAL nuclease and AAV donor template. Sci Transl Med 2015, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Dever DP, Bak RO, Reinisch A, Camarena J, Washington G, Nicolas CE, Pavel-Dinu M, Saxena N, Wilkens AB, Mantri S, et al. : CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells. Nature 2016, 539:384–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Greiner V, Bou Puerto R, Liu S, Herbel C, Carmona EM, Goldberg MS: CRISPR-Mediated Editing of the B Cell Receptor in Primary Human B Cells. iScience 2019, 12:369–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kuhlmann A-S, Haworth KG, Barber-Axthelm IM, Ironside C, Giese MA, Peterson CW, Kiem H-P: Long-Term Persistence of Anti-HIV Broadly Neutralizing Antibody-Secreting Hematopoietic Cells in Humanized Mice. Molecular Therapy 2019, 27:164–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Hartweger H, McGuire AT, Horning M, Taylor JJ, Dosenovic P, Yost D, Gazumyan A, Seaman MS, Stamatatos L, Jankovic M, et al. : HIV-specific humoral immune responses by CRISPR/Cas9-edited B cells. Journal of Experimental Medicine 2019, 216:1301–1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Nahmad AD, Raviv Y, Horovitz-Fried M, Sofer I, Akriv T, Nataf D, Dotan I, Carmi Y, Burstein D, Wine Y, et al. : Engineered B cells expressing an anti-HIV antibody enable memory retention, isotype switching and clonal expansion. Nat Commun 2020, 11:5851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Huang D, Tran JT, Olson A, Vollbrecht T, Tenuta M, Guryleva MV, Fuller RP, Schiffner T, Abadejos JR, Couvrette L, et al. : Vaccine elicitation of HIV broadly neutralizing antibodies from engineered B cells. Nat Commun 2020, 11:5850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Nahmad AD, Lazzarotto CR, Zelikson N, Kustin T, Tenuta M, Huang D, Reuveni I, Nataf D, Raviv Y, Horovitz-Fried M, et al. : In vivo engineered B cells secrete high titers of broadly neutralizing anti-HIV antibodies in mice. Nat Biotechnol 2022, 40:1241–1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Vamva E, Ozog S, Leaman DP, Yu-Hong Cheng R, Irons NJ, Ott A, Stoffers C, Khan I, Goebrecht GKE, Gardner MR, et al. : A lentiviral vector B cell gene therapy platform for the delivery of the anti-HIV-1 eCD4-Ig-Knob-in-Hole-Reversed immunoadhesin. Molecular Therapy - Methods & Clinical Development 2023, doi: 10.1016/j.omtm.2023.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES