Sunteți pe pagina 1din 16

Expert Review of Vaccines

ISSN: 1476-0584 (Print) 1744-8395 (Online) Journal homepage: http://www.tandfonline.com/loi/ierv20

In situ production of therapeutic monoclonal


antibodies

Todd J Suscovich & Galit Alter

To cite this article: Todd J Suscovich & Galit Alter (2015) In situ production of
therapeutic monoclonal antibodies, Expert Review of Vaccines, 14:2, 205-219, DOI:
10.1586/14760584.2015.1001375

To link to this article: http://dx.doi.org/10.1586/14760584.2015.1001375

Published online: 12 Jan 2015.

Submit your article to this journal

Article views: 327

View related articles

View Crossmark data

Citing articles: 3 View citing articles

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=ierv20

Download by: [Cornell University Library] Date: 02 November 2017, At: 12:47
SPECIAL FOCUS y RNA Vaccines Review

In situ production of
therapeutic monoclonal
antibodies
Expert Rev. Vaccines 14(2), 205–219 (2015)

Todd J Suscovich* and The use of antibodies as a treatment for disease has it origins in experiments performed in
the 1890s, and since these initial experiments, monoclonal antibodies (mAbs) have become
Downloaded by [Cornell University Library] at 12:47 02 November 2017

Galit Alter
one of the fastest growing therapeutic classes for the treatment of cancer, autoimmune
Ragon Institute of MGH, MIT, and
Harvard, Cambridge, MA 02139, USA disease, and infectious diseases. However, treatment with therapeutic mAbs often requires
*Author for correspondence: high doses given via long infusions or multiple injections, which, coupled with the
TJSuscovich@mgh.harvard.edu prohibitively high cost associated with the production of clinical-grade proteins and the
transient serum half-lives that necessitate multiple administrations to gain therapeutic
benefits, makes large-scale treatment of patients, especially patients in the developing world,
difficult. Due to their low-cost and rapid scalability, nucleic acid-based approaches to deliver
antibody gene sequences for in situ mAb production have gained substantial traction. In this
review, we discuss new approaches to produce therapeutic mAbs in situ to overcome the
need for the passive infusion of purified protein.

KEYWORDS: adeno-associated virus . adenovirus . ex vivo gene transfer . gene therapy . in situ production
. monoclonal antibody . non-vectored gene delivery

However, technological advances made over


Antibodies as therapeutic proteins the last 40 years in areas including mAb dis-
While monoclonal antibody (mAb)-based ther- covery, recombinant DNA technology and
apeutics account for over one-third of all bio- large-scale cell culture have fully unlocked the
logics currently in development [1], the use of therapeutic potential of antibodies.
antibodies as a therapeutic was first explored
in the 1890s. In 1890, von Behring and Kita- Antibody discovery techniques
sato demonstrated that immunity to diphtheria In 1975, Köhler and Milstein reported the
and tetanus could be transferred from animals generation of a cell line producing an antibody
given small doses of diphtheria or tetanus toxin of a single specificity, that is, a mAb [5]. This
to naı̈ve animals via the transfer of serum [2]. cell line was generated by fusing splenocytes
These early experiments provided the founda- from a vaccinated mouse with a mouse mye-
tion for the use of polyclonal antisera as a ther- loma cell line, and the resulting hybridoma
apeutic, enabling the development of the continuously produced antibodies against the
11 US FDA-approved polyclonal antisera cur- antigen used for vaccination. Suddenly, this
rently licensed for use, largely as post-exposure method made the production of single anti-
prophylaxis in high-risk, immunocompromised bodies a reality, thereby enabling the screening,
patients [3]. These antibodies are purified and selection and optimization of the most potent
pooled from vaccinated individuals (e.g., teta- antibodies. Furthermore, using this technology,
nus) or animals (e.g., botulism) with high titers it now became possible to develop antibodies
of antibodies [4]. However, the identification of targeting a specific protein or antigen, thus
immune individuals and the collection of plas- allowing the targeted development of therapeu-
maphereses to generate these polyclonal sera tic antibodies.
are time consuming and expensive, strongly As a direct result of this technology, in
limiting the widespread use of antisera as either 1986, the first therapeutic mAb was approved
a pre-exposure prophylactic or a therapeutic. for use in humans (muromonab-CD3) [6].

informahealthcare.com 10.1586/14760584.2015.1001375  2015 Informa UK Ltd ISSN 1476-0584 205


Review Suscovich & Alter

chimeric and humanized mAbs is human, these second-


generation therapeutics are significantly less immunogenic than
their mouse counterparts. Furthermore, using current mAb
discovery techniques, it is now possible to rapidly clone and
produce high-affinity, fully human antibodies. Importantly,
even fully human antibodies can be immunogenic due to the
recognition of complementarity-determining region-derived
epitopes by CD4 T cells; however, using antibody engineering,
Mouse antibody Chimeric antibody it may be possible to even further reduce the immunogenicity
of fully human antibodies [8]. Since the approval of muromo-
nab-CD3, 36 additional mAbs have been approved for use in
either the EU or the USA (TABLE 1), with over 300 additionally
in the pipeline [1]. Of these new mAbs therapeutics, only a
small subset is fully mouse antibodies without any degree of
humanization.
Downloaded by [Cornell University Library] at 12:47 02 November 2017

While the advent of hybridoma technology rapidly expanded


the therapeutic potential of mAbs, recent advances in antibody
discovery techniques, including antibody display (e.g., phage
Humanized antibody Human antibody display [9], yeast display [10] and ribosome display [11]), vaccina-
tion of humanized mice and single-cell RNA sequencing will
Figure 1. Humanization of therapeutic monoclonal anti- expand this potential even further. Antibody display utilizes a
bodies. Mouse antibody sequences, indicated in red, can induce library of carriers (e.g., bacteriophages, yeast or ribosomes)
an immune response in humans, decreasing the therapeutic effi- engineered to display human antibody fragments. These anti-
cacy of the monoclonal antibody. To prevent the induction of
body fragment-expressing carriers are then exposed repeatedly
this immune response, chimeric and humanized antibodies have
been developed. To generate a chimeric antibody, the entire vari- to antigens of interest, enabling the selection/propagation (also
able domain of the mouse heavy and light chains are grafted referred to as panning) of carriers displaying antibodies capable
onto a human antibody backbone, indicated in blue. To generate of binding to the target antigen with remarkably high affinity.
a humanized antibody, the specific antigen recognition domains Importantly, the library of antibodies displayed can be derived
(complementarity determining regions) are grafted onto a human
from any source, including vaccinated or immune individuals.
antibody backbone. As the humanized antibody has a smaller
proportion of mouse antibody sequences, they are significantly Therefore, by varying the antibody library, antibody display
less immunogenic than chimeric antibodies. allows for the identification of human antibodies targeting vir-
tually any antigen. Of the 12 fully human mAbs currently
approved for use, four were identified using phage display (ada-
Muromonab-CD3 is a mouse mAb isolated using hybridoma limumab, belimumab, raxibacumab and ramucirumab), with
technology that targets CD3". As CD3" is present on the adalimumab, the first fully human, antibody phage display-
surface of T cells, this antibody was used as an immuno- isolated mAb approved for use in 2002 [12].
suppressant for the treatment of acute transplant rejection. While antibody phage display allows for the in vitro selection
While effective, patients treated with muromonab-CD3 gener- of human mAbs, humanized transgenic mice, such as the Xen-
ated anti-mouse antibodies, as it is a fully mouse mAb [7]. omouse [13] or the Kymouse [14], allow for the directed in vivo
These newly generated antibodies against muromonab-CD3 induction and selection of human antibodies. In these trans-
resulted in the rapid clearance of the mAb from the circulation, genic mice, the endogenous mouse antibody variable regions are
thereby reducing its clinical potency with repeat administration. deleted using gene targeting, and the human variable regions are
To circumvent the generation of an immune response inserted using artificial chromosomes. Normal B-cell develop-
against the infused antibody, subsequent therapeutic mAbs are ment and IGH and IGK/IGL loci recombination occurs in these
often chimeric, humanized or fully human (FIGURE 1). Specifically, mice, and following immunization, normal antibody maturation
with rapid advances in recombinant DNA technology over the occurs, except that the antibodies produced contain fully human
same timeframe, it became possible to isolate the genes encod- variable regions. Hybridomas can then be generated or B-cell
ing the mouse mAb and manipulate them to make the result- clonal repertoires can be sequenced and expressed (as described
ing antibody less immunogenic while retaining its function. To below) from the immunized mice to generate fully human
accomplish this, either the entire antigen-binding domain (i.e., mAbs. Transgenic humanized mice have proven to be extremely
the variable domains) or the individual sequences responsible useful in the identification of human mAbs as eight of 12 human
for antigen recognition (i.e., the complementarity-determining mAb therapeutics currently approved for use were generated in
regions) from the mouse antibody are grafted onto a human humanized transgenic mice (panitumumab, golimumab, canaki-
antibody backbone, generating chimeric or humanized antibod- numab, ustekinumab, ofatumumab, denosumab, ipilimumab
ies, respectively. As the majority of the protein sequence from and nivolumab), and the first human mAb generated from

206 Expert Rev. Vaccines 14(2), (2015)


Downloaded by [Cornell University Library] at 12:47 02 November 2017

Table 1. Therapeutic monoclonal antibodies approved for use in the USA and/or EU.
International Trade name Target Type Approval Indication
name
US EU
Muromonab- Orthoclone CD3 Mouse IgG2a 1986 1986 Acute organ transplant rejection
CD3 Okt3

informahealthcare.com
Abciximab Reopro GPIIb/IIIa Chimeric IgG1 Fab 1994 1995 Prevention of cardiac ischemia
(integrin aIIbb3)
Rituximab MabThera, CD20 Chimeric IgG1 1997 1998 Non-Hodgkin’s lymphoma
Rituxan
Daclizumab Zenapax IL-2R Humanized IgG1 1997 1999 Acute organ transplant rejection
Basiliximab Simulect IL-2R Chimeric IgG1 1998 1998 Acute organ transplant rejection
Palivizumab Synagis RSV F protein Humanized IgG1 1998 1999 Respiratory syncytial virus infection
Infliximab Remicade TNF-a Chimeric IgG1 1998 1999 Crohn’s disease, ulcerative colitis, rheumatoid
arthritis, ankylosing spondylitis, psoriatic
arthritis, plaque psoriasis
Trastuzumab Herceptin HER2 Humanized IgG1 1998 2000 Breast cancer

Gemtuzumab Mylotarg CD33 Antibody drug conjugate 2000 NA Acute myelogenous leukemia
ozogamicin (gemtuzumab [Humanized
IgG4]: calicheamicin)
Alemtuzumab MabCampath, CD52 Humanized IgG1 2001 2001 Chronic lymphocytic leukemia, cutaneous
Campath-1H T-cell lymphoma, and T-cell lymphoma
Brentuximab Adcetris CD30 Antibody drug conjugate 2001 2012 Hodgkin’s lymphoma, systemic anaplastic
vedotin (brentuximab [chimeric IgG1]: large cell lymphoma
monomethyl auristatin E)
Adalimumab Humira TNF-a Human IgG1 2002 2003 Rheumatoid arthritis, juvenile idiopathic
arthritis, psoriatic arthritis, ankylosing
spondylitis, Crohn’s disease, and plaque
psoriasis
Ibritumomab Zevalin CD20 Radiolabeled mouse IgG1 2002 2004 Non-Hodgkin’s lymphoma
tiuxetan
Efalizumab Raptiva CD11a Humanized IgG1 2003 2004 Plaque psoriasis
Omalizumab Xolair IgE Humanized IgG1 2003 2005 Asthma
In situ production of therapeutic mAbs

Tositumomab- Bexxar CD20 Radiolabeled mouse IgG2a 2003‡ NA Non-Hodgkin’s lymphoma


I131

Withdrawn from market in 2010.

Withdrawn from market in 2014.
§
Approved for use in Japan.
Review

NA: Not approved.

207
Downloaded by [Cornell University Library] at 12:47 02 November 2017

208
Table 1. Therapeutic monoclonal antibodies approved for use in the USA and/or EU (cont.).
International Trade name Target Type Approval Indication
name
Review

US EU
Cetuximab Erbitux EGFR Chimeric IgG1 2004 2004 Colorectal cancer, non-small-cell lung cancer,
squamous cell carcinoma of the head and
neck
Bevacizumab Avastin VEGF Humanized IgG1 2004 2005 Colorectal, lung, breast (outside the USA),
glioblastoma (USA only), kidney and ovarian
cancer
Suscovich & Alter

Natalizumab Tysabri Integrin a4 Humanized IgG4 2004 2006 Multiple sclerosis and Crohn’s disease
Ranibizumab Lucentis VEGF Humanized IgG1 2006 2007 Macular degeneration
Panitumumab Vectibix EGFR Human IgG2 2006 2007 Colorectal cancer
Eculizumab Soliris C5 Humanized IgG2/4 2007 2007 Paroxysmal nocturnal hemoglobinuria
Certolizumab Cimzia TNF-a Pegylated humanized Fab 2008 2009 Crohn’s disease and rheumatoid arthritis
pegol
Catumaxomab Removab EPCAM Rat/mouse bispecific NA 2009 Malignant ascites
CD3
Golimumab Simponi TNF-a Human IgG1 2009 2009 Rheumatoid arthritis, psoriatic arthritis,
ankylosing spondylitis and ulcerative colitis
Canakinumab Ilaris IL-1b Human IgG1 2009 2009 Cryopyrin-associated periodic syndrome
Ustekinumab Stelara IL-12 Human IgG1 2009 2009 Plaque psoriasis and psoriatic arthritis
IL-23
Ofatumumab Arzerra CD20 Human IgG1 2009 2010 Chronic lymphocytic leukemia
Tocilizumab RoActemra, IL-6R Humanized IgG1 2010 2009 Rheumatoid arthritis
Actemra
Denosumab Prolia RANK-L Human IgG2 2010 2010 Bone loss
Belimumab Benlysta BAFF Human IgG1 2011 2011 Systemic lupus erythematosus
Ipilimumab Yervoy CTLA-4 Human IgG1 2011 2011 Metastatic melanoma
Pertuzumab Perjeta HER2 Humanized IgG1 2012 2013 Breast cancer
Raxibacumab Abthrax Protective Human IgG1 2012 NA Inhaled anthrax
antigen of
anthrax toxin

Withdrawn from market in 2010.

Withdrawn from market in 2014.
§
Approved for use in Japan.
NA: Not approved.

Expert Rev. Vaccines 14(2), (2015)


In situ production of therapeutic mAbs Review

transgenic humanized mice was approved for use in humans in


2006 (panitumumab) [12].

Uveitis, rheumatoid arthritis and psoriasis


The newest technique that has been adapted to isolate

Ulcerative colitis and Crohn’s disease


human mAb sequences is the high-throughput sequencing of

Multicentric Castleman’s disease


the antibody repertoire. With this technology, DNA or RNA is
Chronic lymphocytic leukemia isolated from bulk B-cell populations and sequenced using
next-generation sequencing. The matching heavy- and light-
chain sequences can be bioinformatically paired, and the anti-

Metastatic melanoma
body produced in vitro and screened for function. Importantly,
recent advances in single-cell sequencing of DNA/RNA has rev-

Neuroblastoma
Gastric cancer
Breast cancer

olutionized this application dramatically, as paired heavy- and


Indication

Melanoma
light-chain sequences can be directly identified and screened [15].
While no therapeutic human mAbs have been produced using
the technology, this technology is only in its infancy and its
therapeutic potential likely remains untapped and will explode.
Downloaded by [Cornell University Library] at 12:47 02 November 2017

One area in which high-throughput sequencing of antibody


In review

In review
In review

repertoires will be particularly useful is in the treatment/


2013

2014

2014

2014
2014

prevention of infectious diseases, where immune repertoires can


NA§
EU
Approval

be sequenced in real time at the start of an epidemic/pandemic


Table 1. Therapeutic monoclonal antibodies approved for use in the USA and/or EU (cont.).

in immune individuals to rapidly develop protective monoclo-


nal therapeutics.
In review
In review
2013

2013

2014
2014
2014
2014

2014
US

Production of therapeutic mAbs


Even though advances in antibody discovery technologies made
it possible to produce antibodies targeting specific antigens,
therapeutic mAbs often require relatively high doses to be effec-
Glycoengineered humanized

tive for the treatment of solid tumors (up to hundreds of milli-


Antibody drug conjugate

grams per kilogram), largely due to their poor penetrance into


(Herceptin:mertansine)

tumors [16]. Initially, therapeutic mAbs were purified from the


Humanized IgG1
Humanized IgG4

ascitic fluid of mice injected with hybridomas. While the ascitic


Chimeric IgG1

Chimeric IgG1
Human IgG1

Human IgG1

Human IgG4

fluid contained relatively high concentrations of antibodies (on


the order of 1 g/l), large-scale production of therapeutic mAbs
would require an excessively large number of mice. Further-
Type

IgG1

more, ascites induction in mice causes a significant amount of


pain and distress to the animal and is currently considered
unethical. Therefore, therapeutic mAb production was quickly
moved into mammalian cell culture expression systems, and
a4b7 integrin

virtually all therapeutic mAbs approved to date are produced in


mammalian cell culture systems. While the initial cell lines pro-
VEGFR2
Target

IL-17a

ducing therapeutic mAbs produced antibodies on the order of


CD20
HER2

PD-1

PD-1
GD2
IL-6

100–500 mg of antibody per liter of tissue culture [17], advan-


ces in antibody production technology, including production
cell line generation and optimization, large-scale bioreactor-
based tissue culture and fed-batch production, have resulted in
Trade name

significant increases in antibody production [18]. As a result of


(Pending)
Keytruda

these advances, therapeutic mAbs are often produced on the


Cyramza

Unituxin
Kadcyla

Gazyva

Entyvio

Opdivo
Sylvant

Withdrawn from market in 2010.


Withdrawn from market in 2014.

order of 5 g or more per liter of tissue culture in 15,000–


20,000 l bioreactors.
Approved for use in Japan.

Although these advances in antibody production technology


have reduced the cost of producing therapeutic mAbs from
NA: Not approved.
Pembrolizumab
International

Obinutuzumab

US$ 300/g to approximately US$ 100/g these costs, the down-


Ramucirumab

Secukinumab
Vedolizumab
Trastuzumab

Dinutuximab

stream quality control requirements and the large dose of anti-


Nivolumab
emtansine

Siltuximab

bodies required for efficacy still make the production of


name

therapeutic mAbs extremely costly [19,20]. This expense signifi-


cantly limits the use of therapeutic mAbs in the developing


§

informahealthcare.com 209
Review Suscovich & Alter

Heavy chain be replaced with a single or regularly


Vectored cDNA Ex vivo spaced injections resulting in the continu-
delivery Light chain modification ous and sustained production of thera-
cDNA peutically efficacious doses of mAbs. To
Non-vectored date, three main approaches have been
delivery used to ectopically produce mAbs in situ
(FIGURE 2): ex vivo genetic modification of
cells, viral vector-mediated gene transfer
and non-vectored gene transfer.

Ex vivo genetic manipulation of cells


In vivo, antibodies are produced by tran-
sient plasmablasts and long-lived plasma
cells, which are capable of secreting over
3000 antibody molecules a second [23].
Downloaded by [Cornell University Library] at 12:47 02 November 2017

While this naturally high secretion rate


and long persistence in vivo [24] make
plasma cells ideal candidates for ex vivo
manipulation and engineering, these cells
are difficult to access (bone marrow or
Tumor tissue localized). Furthermore, these cells
already produce antibodies, making pro-
Pathogen Cytokines duction of additional antibodies in these
cells difficult, but not impossible [25].
However, a wide array of non-B cells has
Figure 2. Strategies to produce therapeutic monoclonal antibodies in situ. Three
been demonstrably modified to secrete
approaches have been used to produce therapeutic monoclonal antibodies in situ:
ex vivo modification of primary cells, vectored gene delivery and non-vectored gene therapeutic mAbs in vivo.
delivery. During ex vivo modification of primary cells, cells are harvested from the sub- The ideal target cell type for ex vivo
ject, transduced ex vivo with antibody-encoding vectors or transfected with antibody- modification should be easily accessible,
encoding nucleic acids, expanded and re-implanted into the subject. Alternatively, the manipulatable and engraftable. Early work
modified cells can be used to seed a bioscaffold and implanted as a pseudo-organ.
in this field demonstrated that many cell
During vectored gene delivery, the antibody-encoding genes are cloned into a viral vec-
tor (e.g., adenovirus or adeno-associated virus), which is then directly injected into the types can be modified to secrete mAbs
subject. By contrast, during non-vectored gene delivery, the antibody-encoding genes in vitro. Noël et al. demonstrated that
are directly injected into the subject as either plasmid DNA or RNA. DNA/RNA uptake myoblast, hepatocyte, keratinocyte and
can be increased using electroporation or packaging into lipid nanoparticles or polymers. fibroblast cell lines could all produce
mAbs at levels similar to or greater than
world where they could have the greatest impact. In particular, hybridomas in vitro following transduction with retroviral con-
one area in which mAbs are currently in development is as a structs and that similar result could be seen using primary
prophylactic against HIV infection [21]. However, to gain wide- cells [26,27]. Importantly, these cells retained their ability to pro-
spread utility as a prophylactic, it has been estimated that the duce mAbs following implantation in vivo, with consistent
cost of production of a mAb would have to be reduced to the secretion seen for over a year [28].
level of dollars per gram [19]. As these reductions are likely While the clinical efficacy of these antibodies was not tested
beyond what can be accomplished via advances in cell culture in these models, therapeutic effects following the ex vivo engi-
technology, novel approaches to produce therapeutic mAbs are neering of cells to secrete mAbs have been observed. Using a
imminently needed. The remainder of this review focuses on severe combined immunodeficiency mouse model of cancer,
one promising approach – in situ production of therapeutic human peripheral blood mononuclear cells were transduced
mAbs – and the strategies being used to accomplish this. with an antibody-encoding lentivirus and injected into mice [29].
Mice receiving cells transduced with the antibody-encoding len-
In situ production of therapeutic mAbs tivirus demonstrated a significant reduction in tumor growth
Given the high cost associated with the production of therapeu- compared to the control mice. Additionally, fibroblasts modified
tic mAbs, in situ mAb production has been a long-term goal of to secrete an HIV-specific mAb demonstrated antiviral activity
the gene therapy field. Using gene therapy-based approaches to following implantation into humanized mice [30], resulting in a
turn cells of the body into ‘in situ bioreactors’ producing thera- 100,000-fold lower viral load following HIV infection.
peutic mAbs, the high doses, long infusion times and side Intriguingly, while some early studies examining ectopic,
effects typically associated with mAb-based treatment [22] could in situ antibody production simply injected modified cells into

210 Expert Rev. Vaccines 14(2), (2015)


In situ production of therapeutic mAbs Review

the mouse, the HIV-specific antibody-secreting fibroblasts were have pro-angiogenic [38] and immunomodulatory effects [39].
injected not as a suspension of cells, but were rather implanted These effects can increase tumor growth and metastasis [40],
as a pseudo-organ generated in vitro using bioscaffolds [30]. In potentially limiting the effectiveness of ex vivo-engineered stem
multiple studies examining the ectopic expression of therapeutic cells as a therapeutic. To bypass this limitation, ex vivo-
proteins other than mAbs, implantation of transduced cells in a engineered stem cells could be implanted in association with a
bioscaffold resulted in increased protein expression and a pro- bioscaffold, limiting their dissemination throughout the body.
longed therapeutic effect when compared with transduced cells In support of this, stem cells engineered to secrete an antibody
injected alone, strongly suggesting that implantation of mAb- targeting a tumor antigen expressed on the cell surface and
secreting pseudo-organs derived from a patient’s own cells has implanted in a bioscaffold at a site distal to the tumor still
therapeutic potential. demonstrated a therapeutic effect: tumor growth in mice receiv-
As a proof of concept for the engineering of therapeutic ing the antibody-secreting stem cells was significantly reduced
mAb-secreting tissues, Compte et al. generated antibody- in comparison to the mice receiving the control stem cells [32].
secreting engineered blood vessels using primary human endo-
thelial cells and implanted these tissues into nude mice [31]. Vectored production of therapeutic mAbs
Stable antibody production was observed for at least 7 weeks. Although the ex vivo engineering of autologous cells to secrete
Downloaded by [Cornell University Library] at 12:47 02 November 2017

Importantly, a clear therapeutic effect was observed in the therapeutic mAbs has shown potential, the cost and labor
mice receiving the antibody-secreting blood vessels, with a sig- required to collect, engineer and implant the modified cells will
nificantly reduced tumor growth rate and a significantly likely limit the scalability of this approach. Therefore, addi-
increased survival time. As additional advancements are made tional strategies to produce antibodies in situ are needed. Vec-
in bioscaffold development and tissue engineering, the potential tored gene therapy, that is, the delivery of foreign genes using
of antibody-secreting engineered pseudo-organs may become viral vectors, has a long history of being used to express thera-
fully realized. peutic proteins in vivo, including antibodies. Even though strat-
While many of the early studies reporting the ex vivo engi- egies employing other viruses [41–43] and bacteria [44] have been
neering of cells to secrete mAbs focused on manipulating used, the vectors typically used in vectored gene therapy are
terminally differentiated cell types ex vivo, terminally differenti- adenovirus (AdV), adeno-associated virus (AAV) and retrovi-
ated cells often have a finite lifespan, potentially limiting their ruses. While retroviruses have been used to ectopically express
use for the persistent, long-term production of mAbs in situ. mAbs in vivo with therapeutic effects [45,46], because of the
By contrast, the self-renewal capacity of stem cells affords them inherent risk of insertional mutagenesis due to integration of
a significantly longer lifespan, making them strong candidates the retroviral genome, most of the research harnessing viral vec-
for modification ex vivo. To date, adult mesenchymal [32,33], tors has used AdVs or AAVs.
hematopoietic [34] and neural [35] stem cells have all been suc-
cessfully engineered in vitro to secrete mAbs in vivo. Hemato- Adenoviral-mediated gene delivery
poietic stem cells engineered to secrete an HIV-specific antibody AdVs are non-enveloped, double-stranded DNA viruses that
readily differentiated into B cells that secreted the transduced infect a large variety of cell types. AdVs are one of the most
antibody in vivo [34], resulting in an 80-fold reduction in viral frequently used and best-studied viral vectors for the expression
load and a 200-fold reduction in the number of infected cells of therapeutic proteins in vivo, and multiple studies have dem-
following challenge with HIV. Similarly, mice injected with onstrated in vivo production of mAbs using antibody-encoding
mesenchymal stem cell-like stem cells engineered to secrete a AdVs [47–58]. Furthermore, in many of these studies, the
trastuzumab-like antibody demonstrated a significantly increased expressed mAbs had a demonstrable therapeutic effect, includ-
survival rate compared with the control animals [32]. ing inhibition of tumor growth [50–53]; protection from infec-
In addition to the increased lifespan of stem cells making tion with Yersinia pestis [54,55], respiratory syncytial virus [57]
them an attractive candidate for ex vivo engineering, both mes- and West Nile virus [58]; and the suppression of pulmonary
enchymal stem cells and neural stem cells can home to and edema [56].
seed tumors in vivo following injection [36,37]. As therapeutic While therapeutic effects of AdV-encoded mAbs have been
mAbs penetrate into solid tumors poorly [16], the homing and observed, the expression of mAbs in vivo following infection
seeding of the tumor with cells engineered to secrete antibodies with AdVs is highly variable. Following systemic administration
targeting the tumor allows for more effective eradication of the of antibody-encoding AdVs, peak concentrations greater than
malignant cells. In fact, neural stem cells engineered to secrete 1 mg/ml have been reported early (Day 3) after AdV deliv-
trastuzumab readily homed to the tumor and secreted anti- ery [54]. However, by Day 5, the antibody titer had begun to
body [35]. Importantly, the intratumoral concentration of trastu- decline, and long-term concentrations ranging from 20 ng/ml
zumab was significantly increased in mice treated with the to 40 mg/ml have been reported [48,51], illustrating the transient
antibody-secreting neural stem cells compared with mice nature of AdV-mediated gene delivery. Importantly, in the
directly treated with trastuzumab [35]. majority of the studies in which a therapeutic or protective
It should be noted that while the tumor-homing potential of effect was observed, the challenge was carried out near the peak
stem cells can be beneficial, stem cells have also been shown to antibody titer. Moreover, because many AdVs are highly

informahealthcare.com 211
Review Suscovich & Alter

prevalent, pre-existing immunity can greatly affect the clinical its limitations. First, unlike AdV-mediated antibody expression,
efficacy of AdV-mediated antibody gene transfer [59,60]. Thus, which occurs rapidly, the expression of mAbs using AAV can
the prevalence of pre-existing anti-AdV immunity coupled with take weeks to reach therapeutically efficacious or protective
the transient nature of AdV antibody expression significantly concentrations. De et al. treated two groups of mice with either
limits enthusiasm for using AdVs as a delivery vehicle for the an AdV expressing a mAb against Bacillus anthracis lethal toxin
in situ expression of mAbs [61]. or an AAV expressing the same mAb [82]. The mice were then
challenged with B. anthracis lethal toxin at different time points
AAV-mediated gene delivery (1, 3, 7, 14, 28, 64 and 182 days) post-AdV/AAV treatment.
In contrast to the limited durability of AdV-mediated ectopic In contrast to the AdV-treated mice, which were protected
protein expression, expression of proteins from AAVs can per- from the toxin when challenged 1 day post-treatment, AAV-
sist for years [62]. AAVs are small, single-stranded DNA viruses treated mice were only protected beginning 14 days post-treat-
that are capable of infecting both dividing and non-dividing ment. However, the protection in the AdV-treated mice waned
cells [63]. Furthermore, because AAVs are not associated with between Week 8 and Week 26; the AAV-treated mice were
any known disease and AAV-based vectors (which lack the viral completely protected from the toxin even at Week 26. These
rep and cap genes required for integration) do not integrate results suggest that in instances where immediate, but not long-
Downloaded by [Cornell University Library] at 12:47 02 November 2017

into the genome, AAVs have become the most attractive viral term, antibody expression is required, such as during an infec-
vector for gene therapy [64–67], and the first AAV-based gene tious pandemic or bioterrorism event, AdV-mediated antibody
therapy product was recently approved for use in Europe expression may be superior to AAV-mediated antibody expres-
(Glybera; [68]). sion. By contrast, AAV may be the vector of choice when
In 2002, Lewis et al. reported the first instance of a mAb slower, but persistent, antibody expression is required, such as
being produced from an AAV-based vector [69]. Although for cancer immunotherapy. When both rapid and persistent
only low-level antibody production was observed in vivo antibody expression is required, co-delivery of antibody-
(<10 mg/ml), this low antibody titer persisted for at least encoding AdVs and AAVs will likely prove effective, as demon-
6 months. While low antibody levels were observed in this ini- strated by De et al. [82].
tial experiment, follow-up experiments using optimized anti-
body expression cassettes significantly enhanced antibody Non-vectored production of therapeutic mAbs
levels [70]. Further refinements to the antibody-encoding cas- Vector-mediated expression of mAbs has shown success in ani-
sette, including the utilization of a self-cleaving picornavirus 2A mal models and has begun Phase I clinical trials; however,
peptide, resulted in a peak serum antibody level greater than vector-mediated delivery has several disadvantages that are
8 mg/ml, with sustained production of 1 mg/ml up to 4 months shared among vectors, including the development of or pre-
post-AAV delivery [71]. Not surprisingly, the expression of a existing anti-vector immunity, a limited coding capacity and
therapeutic mAb (anti-mouse VEGFR2) at this level had signifi- difficulties in producing clinical-grade products, which may
cant therapeutic effects: tumor dormancy was observed in over limit its broad use in clinical settings, especially for the treat-
half of the treated mice for 3 months, with complete regression ment of otherwise healthy individuals. Unlike vectored delivery,
of even large tumors observed in a subset of treated mice. non-vectored gene delivery (i.e., direct delivery of DNA or RNA)
Since these initial descriptions of AAV-mediated in situ is not confined by many of these limitations as an immune
expression of mAbs, a large number of additional studies have response does not develop against transferred DNA/RNA, there
demonstrated the therapeutic effectiveness of AAV-expressed are no restrictions on the size of the construct, it can be produced
mAbs. AAV-mediated expression of mAbs has now been shown easily and cheaply in a cell-free manner and it has an excellent
to suppress tumor growth in mouse models of cutaneous T-cell safety profile.
lymphoma [72], colon cancer [73], liver cancer [73,74] and lung
cancer [74–76]. Furthermore, AAV-mediated antibody expression DNA as a vehicle
has been effective against tumors, both prophylactically and However, in spite of these apparent advantages, in situ expres-
therapeutically [77]. Beyond tumor models, AAV-mediated sion of mAbs via direct delivery of DNA has gained limited
expression of mAbs has also been shown to provide protection traction largely due to the poor expression observed following
from infection with HIV [70,78,79], respiratory syncytial virus [57], injection. Tjelle et al. were the first to report the in situ pro-
influenza [80], prion disease [81], anthrax [82] and malaria [83] and duction of mAbs by the direct transfection of plasmid DNA in
the first clinical trials utilizing AAV-mediated expression of a 2004 [90]. Following a single injection of DNA and in vivo
mAb (two different HIV-specific mAbs) are currently under- electroporation, low levels of a human mAb (50–200 ng/ml)
way [84,85]. Intriguingly, AAV-mediated expression of mAbs has could be detected in the mouse. While the antibody titers
demonstrated therapeutic potential in areas in which mAbs have in vivo declined after 7–14 days, this decline was associated
only recently begun to be exploited, including addiction [86,87], with the development of an immune response against the
Alzheimer’s disease [88] and macular degeneration [89]. human mAb, and the plasmid DNA-mediated production of a
While AAV has become the de facto vector of choice for the fully mouse version of the same antibody persisted in the
expression of therapeutic antibodies in vivo, it is not without serum for over 7 months. Furthermore, when injected into

212 Expert Rev. Vaccines 14(2), (2015)


In situ production of therapeutic mAbs Review

severe combined immunodeficiency mice, which lack T or approximately 5 mg/ml after 70 days, delivery into three differ-
B cells, increased antibody titers were observed, with no decrease ent sites resulted in peak expression levels of approximately
in antibody expression observed after 28 days [91]. 30 mg/ml. Furthermore, following injection into three sites,
Similarly, Perez et al. also reported in situ mouse mAb the titer of the antibody remained over 10 mg/ml over the
expression following the delivery of DNA via in vivo electropo- course of 70 days [94]. These results suggest that with further
ration [92]. A peak level of 1.5 mg/ml of antibody was observed; refinement of the delivery strategy, it may be possible to
however, this concentration decreased over time, with average increase in situ antibody production even higher.
levels of approximately 200 ng/ml observed 5 months post-
plasmid delivery. Interestingly, by incorporating tetracycline- RNA as a vehicle
responsive elements into the promoter controlling the expression While several studies have demonstrated the production of
of the antibody, they demonstrated that it was possible to regu- therapeutic mAbs in situ following delivery of plasmid DNA,
late the expression of the antibody in vivo via treatment with the to date, there have been no studies demonstrating production
antibiotic doxycycline. Similar regulated in vivo antibody expres- of mAbs directly from RNA. Importantly, RNA has several
sion following AAV-mediated gene delivery has also recently advantages over DNA as a non-vectored delivery strategy,
been reported [93]. Overall, these studies demonstrate that injec- including reduced immunogenicity and no risk of genomic
Downloaded by [Cornell University Library] at 12:47 02 November 2017

tion of mAb-encoding DNA can result in durable mAb expres- integration. Furthermore, unlike DNA, RNA does not require
sion, albeit at low levels. entry into the nucleus to be effective, potentially eliminating a
Importantly, despite the low levels of mAbs produced significant limitation of DNA [95].
in vivo, protection from infection has been observed following Although there have been no studies demonstrating the
plasmid DNA-mediated antibody delivery [94]. Following injec- in situ production of mAbs using RNA, RNA has been success-
tion of a plasmid encoding an influenza-specific mAb, serum fully used to produce secreted proteins in situ [96–98]. Using a
levels of 4–20 mg/ml and pulmonary levels of 50–300 ng/ml chemically modified mRNA encoding erythropoietin,
were observed 20 days after injection. Despite these relatively Kormann et al. demonstrated that a single intramuscular injec-
low antibody titers, following challenge with a lethal dose of tion of 5 mg of mRNA resulted in a fourfold increase in the
influenza on Day 20, all of the plasmid-treated mice demon- serum erythropoietin level 14 days post-delivery, with the
strated a significant reduction in lung viral load and weight erythropoietin level remaining significantly increased 28 days
loss, with a corresponding increase in survival. Furthermore, post-injection [98]. This increased erythropoietin level conse-
the protection afforded by these antibodies persisted even quently resulted in a significantly increased hematocrit level in
though the circulating and tissue titers declined over time. One the mice both 14 and 28 days following delivery of the
hundred twenty days after plasmid injection, the antibody levels mRNA. Similarly, Kariko et al. demonstrated that intraperito-
in the blood and lung decreased to 600 ng–4 mg/ml and 4 ng– neal injection of 100 ng of HPLC-purified, chemically modi-
60 ng/ml, respectively. However, even with these low antibody fied, erythropoietin-encoding mRNA resulted in a significant
titers, a significant reduction in lung viral load was observed increase in the serum erythropoietin level that persisted for
following challenge on Day 120. Combined, these data demon- 4 days post-injection, with corresponding increases in hemato-
strate that plasmid DNA can be used as a delivery modality to crit persisting for 20 days post-injection [96]. Importantly, injec-
produce persistent levels of functional mAbs in vivo. tion of recombinant erythropoietin protein resulted in only a
While only relatively low levels of mAb expression following transient (6 h) increase in the serum erythropoietin level, dem-
delivery of plasmid DNA in vivo have been reported, the con- onstrating that the persistent increase in erythropoietin levels
centration of antibody required for a therapeutic effect differs following delivery of mRNA was due to the continued produc-
drastically depending on the disease being targeted. While ther- tion of erythropoietin in situ. Furthermore, as little as 1 ng of
apeutic mAbs used for the treatment of cancer often require this mRNA was shown to have a therapeutic effect, illustrating
high doses, this is largely due to the poor penetrance of mAbs the potency of mRNA as a delivery modality [96].
into solid tumors [16]. By contrast, mAbs used for the treatment Beyond erythropoietin, both surfactant protein B (SFTPB) [98]
of autoimmune disease, such as ustekinumab (anti-IL-12/IL-23) and VEGF [97] have been produced in situ using mRNA, with
and adalimumab (anti-TNF-a), require significantly lower significant therapeutic effects. Following delivery of a SFTPB-
serum concentrations to be effective (6 mg/ml for adalimumab encoding mRNA directly to the lung of mice with a condi-
and 0.31 mg/ml for ustekinumab) [12]. Therefore, while the tional deficiency of SFTPB using an endotracheally inserted
serum concentrations currently attainable following delivery of microsprayer, near wildtype levels of SFTPB were detected [98].
antibody-encoding plasmid DNA may not be sufficient for the This level of SFTPB resulted in a significant increase in the
treatment of cancer, these levels are above or near the concen- survival of the conditionally deficient mice compared to the
trations associated with a therapeutic effect in the treatment of control mice. Furthermore, repeated delivery of the SFTPB-
autoimmune disease. expressing mRNA (twice a week) protected the mice from
Importantly, injections at multiple sites can boost in vivo pulmonary failure until the end of the study, further demon-
antibody titers. While delivery into one site resulted in peak strating the therapeutic effects of the delivery of mRNAs
expression levels of approximately 10 mg/ml, declining to in vivo. In a different study, Zangi et al. demonstrated the

informahealthcare.com 213
Review Suscovich & Alter

therapeutic effectiveness of mRNA-delivered VEGF in a model manufacturing difficulties. Conversely, synthetic approaches,
of myocardial infarction [97]. The intramyocardial injection of including the delivery of non-vectored nucleic acids, offer
the VEGF-encoding mRNA at the time of infarct induction remarkable flexibility and the greatest speed of production, have
resulted in a significant increase in both the short-term and shown promise in animal models and, while still in their
long-term survival of the mRNA-treated mice compared to the infancy, are currently being vetted in clinical trials. However,
untreated controls. each of the approaches discussed in this review has their advan-
While there have only been a handful of studies expressing tages and disadvantages. Importantly, as the range of indications
therapeutic proteins in situ using RNA, and no studies demon- for which therapeutic mAbs can be used is diverse and the dos-
strating in situ production of mAbs, the therapeutic potential ing requirements vary considerably based on the indication, it is
of RNA is likely to be greater than that of DNA, especially likely that no single strategy will be ideal, but rather, distinct
when transient expression is needed. For example, prolonged strategies for antibody gene transfer will be of utility for spe-
exposure to VEGF can have detrimental effects, including cific clinical indications.
increased vascular permeability [99]. Therefore, while transient
expression of VEGF can be beneficial, persistent expression Expert commentary
may be harmful. In support of this, while mRNA-mediated Despite their promise, key technological advances are still
Downloaded by [Cornell University Library] at 12:47 02 November 2017

expression of VEGF resulted in prolonged survival of mice fol- required to make either vectored or non-vectored antibody gene
lowing myocardial infarct induction, plasmid DNA-mediated delivery approaches a reality. To gain widespread acceptance
expression of VEGF actually reduced survival, highlighting the for vectored gene delivery, particularly with AAV, advances
benefits of transient expression from RNA [97]. must be made in limiting/subverting anti-vector immunity,
However, while the transient nature of RNA can be an high-throughput production methodologies and strategies to
advantage, it can also significantly limit broader therapeutic turn off the vector. For non-vectored gene delivery, delivery
potential. To circumvent this, self-replicating RNAs (or repli- approaches must improve.
cons) based on single-stranded RNA viruses (e.g., flaviviruses Vectored delivery of mAb-encoding genes has proven to be
and alphaviruses) have been developed [100–103]. These replicons quite effective as a prophylactic against a number of infectious
encode all of the viral proteins required for viral replication, diseases, including HIV and influenza [79,80]. However, while
but none of the viral structural proteins, which have been AAV-based vectors are largely considered safe, with the first
replaced with one or more genes of interest. Following delivery AAV vector-based gene therapy approved for use in Europe,
into a cell, the viral non-structural proteins are produced, treatment with AAV induces both a humoral [105] and T-cell
allowing for the replication of the replicon and the expression [106] response against the vector itself. Furthermore, while mul-
of the gene of interest. Importantly, the gene of interest is tiple AAV subtypes have been identified, a significant portion
expressed at an extremely high level, accounting for 10–20% of of the population has already been exposed to these viruses and
the total protein of the cell [104]. Furthermore, due to the self- thus harbors pre-existing immunity against many of these sub-
propagation of the replicon, the gene of interest continues to types [107]. Because even low levels of anti-AAV antibodies can
be expressed in cells irrespective of cell division, and long-term limit transduction and gene expression [108], the presence of
(>2 months) replicon-mediated gene expression has been pre-existing antibodies targeting AAV can significantly decrease
observed [101,102]. Therefore, via the use of replicons, it will the efficacy of AAV-based mAb expression strategies, greatly
likely be possible to durably express high levels of therapeutic limiting the utility of AAV in particular geographic areas or for
proteins, including mAbs, in situ. applications requiring repeated administration. While several
promising approaches to subvert this immunity have been
Conclusions developed, including chemical [109] and genetic [110] modifica-
Since the first serum transfer experiments were performed in tion of the viral capsid, in order to enable the wide use of
the 1890s, antibody-based therapeutics have evolved rapidly. AAV-based gene therapy, additional strategies to evade the pre-
While therapeutic antibody infusion and ex vivo cell manipula- existing anti-vector immunity and prevent the induction of
tion have shown clinical efficacy in humans and animal models, de novo anti-vector immune responses must be developed.
respectively, these techniques are costly and slow. Therefore, for Similar to the mAb therapeutics currently in use, vectored
indications in which mAbs must be rapidly delivered, such as gene delivery approaches are produced using eukaryotic cell cul-
during a pandemic, this approach cannot be used. To date, vec- ture, which can make manufacturing costly and compli-
tored gene delivery has demonstrated the most success in the cated [111,112]. Currently, it is estimated that 104–105 viral
in situ production of mAbs, and approaches utilizing this tech- particles can be produced from a singly transfected cell [113].
nique have advanced to clinical trials. Depending on the vector However, one of the ongoing AAV-mediated, in situ antibody
used, either rapid, short-term antibody production (e.g., AdV production clinical trials will deliver 4  1012–1.2  1014 viral
vectors) or delayed, long-term antibody production (e.g., AAV particles per subject. Therefore, to gain widespread use, large
vectors) can be observed in situ. However, viral vectors are lim- amounts of virus must be generated [85]. As approximately 2 
ited by pre-existing immunity against the vector, the relatively 1014 viral particles can be grown per 1 liter culture [114], large-
low coding capacity of the commonly used vectors and scale batch cultures must be generated and quality controlled

214 Expert Rev. Vaccines 14(2), (2015)


In situ production of therapeutic mAbs Review

to produce sufficient clinical-grade product. While significant field; a goal that has not been met. However, with the recent
advances have been made in producing recombinant viral vec- advancements in both vectored and non-vectored gene delivery
tors, including the use of HSV-complemented cultures [115] and approaches, we are closer than ever in attaining this goal. The
insect production cell lines [114], further advances will be first clinical trials for the in situ production of mAbs are cur-
required to make production of viral vectors economical. rently underway, scheduled for completion by January
While vectored gene delivery has been exploited more widely 2015 [85]. These trials make the critical step from animal mod-
than non-vectored gene delivery, non-vectored gene delivery els to humans and will provide the pivotal proof-of-concept
promises to provide a flexible, non-immunogenic, high- studies that in situ production of therapeutic mAbs is feasible.
throughput, cost-effective solution for vectored-antibody deliv- Over the coming 5 years, the expected advancements in both
ery approaches. Currently, the greatest limitation associated vector design/manufacturing and vehicles for the delivery of
with non-vectored gene delivery is the poor titers obtained fol- non-vectored DNA/RNA, coupled with advancements in anti-
lowing plasmid DNA delivery. However, all of the initial stud- body discovery techniques and antibody engineering, including
ies have been carried out using suboptimal electroporation the use of recombinant antibody fragments (e.g., single-chain
conditions/equipment. Yet, delivery strategies are rapidly evolv- Fv fragments, minibodies, camelid-based nanobodies and
ing, already showing great promise in the expression of a mAb immunoadhesins) [21], will allow the potential of the in situ
Downloaded by [Cornell University Library] at 12:47 02 November 2017

fragment at levels greater than 3 mg/ml [116]. Additionally, production of mAbs to become fully realized. Furthermore, as
non-electroporation-based strategies have improved dramati- the indications for the administration of mAbs broaden
cally over the past several years [117], with lipid-based delivery beyond cancer, autoimmune disease and infection, in situ pro-
vehicles showing great promise for RNA delivery. Thus, with duction of mAbs will gain increased widespread use.
further technical improvements in electroporation and advan-
ces in material sciences, novel delivery strategies will likely Financial & competing interests disclosure
increase the in situ expression of non-vectored antibody gene The authors have no relevant affiliations or financial involvement with
delivery strategies further. any organization or entity with a financial interest in or financial conflict
with the subject matter or materials discussed in the manuscript. This
Five-year view includes employment, consultancies, honoraria, stock ownership or options,
The in situ production of therapeutic proteins, including thera- expert testimony, grants or patents received or pending, or royalties.
peutic mAbs, has been a long-term goal of the gene therapy No writing assistance was utilized in the production of this manuscript.

Key issues
. Monoclonal antibody (mAb)-based therapeutics are one of the fastest growing classes of therapeutics, with over 300 currently in
various stages of clinical development. However, the large doses required, coupled with the high cost of production and long-
administration times, limits the use of mAb-based therapeutics as a widespread prophylactic or therapeutic treatment in resource-poor
settings.
. Three main approaches have been used for the in situ production of therapeutic mAbs: ex vivo cell manipulation, vectored gene
delivery and non-vectored gene delivery.
. Multiple cell types have been successfully modified ex vivo to produce mAbs with demonstrable therapeutic effects, and advancements
in tissue engineering will likely increase the titer of antibodies obtainable. However, the therapeutic usefulness of this approach is limited
by the cost and labor required to collect, engineer and implant the modified cells as well as the mutagenic risk associated with ex vivo
manipulation of cells.
. Vectored gene delivery has demonstrated the greatest success for in situ mAb production due to its ability to generate high titers of
mAbs. These antibodies have shown significant therapeutic benefits, particularly as a prophylaxis against infectious diseases, and the first
clinical trials using this approach have begun. However, the therapeutic usefulness of this approach is limited by the development of or
pre-existing immunity against the vector, the relatively low coding capacity of the commonly used vectors and
manufacturing difficulties.
. Non-vectored gene delivery utilizing plasmid DNA has shown limited success for the in situ production of mAbs, while RNA
has been successfully used to express therapeutic secreted proteins in situ, but not antibodies. While this approach still struggles
to achieve clinically effective titers, recent advances in in vivo nucleic acid delivery approaches may drastically improve this
therapeutic approach.

informahealthcare.com 215
Review Suscovich & Alter

References 13. Green LL. Antibody engineering via genetic heterodimerization. Protein Eng 1996;9(7):
engineering of the mouse: xenoMouse 617-21
Papers of special note have been highlighted as:
. of interest
strains are a vehicle for the facile generation 26. Noël D, Pelegrin M, Marin M, et al. In
.. of considerable interest of therapeutic human monoclonal vitro and in vivo secretion of cloned
antibodies. J Immunol Methods 1999; antibodies by genetically modified myogenic
1. Pharmaceutical Research and Manufacturers 231(1-2):11-23 cells. Hum Gene Ther 1997;8(10):1219-29
of America. Medicines in development:
14. Lee EC, Liang Q, Ali H, et al. Complete 27. Noel D, Dazard JE, Pelegrin M, et al. Skin
biologics. In: 2013 Biopharmaceutical
humanization of the mouse as a potential organ for ectopic monoclonal
Research Industry Profile. PhRMA;
immunoglobulin loci enables efficient antibody production. J Invest Dermatol
Washington, DC: 2013. Available from:
therapeutic antibody discovery. Nat 2002;118(2):288-94
http://www.phrma.org/sites/default/files/pdf/
Biotechnol 2014;32(4):356-63
biologics2013.pdf 28. Noel D, Pelegrin M, Brockly F, et al.
15. Georgiou G, Ippolito GC, Beausang J, et al. Sustained systemic delivery of monoclonal
2. von Behring E, Kitasato S. [The mechanism
The promise and challenge of antibodies by genetically modified skin
of diphtheria immunity and tetanus
high-throughput sequencing of the antibody fibroblasts. J Invest Dermatol 2000;115(4):
immunity in animals. 1890]. Mol Immunol
repertoire. Nat Biotechnol 2014;32(2): 740-5
1991;28(12):1317; 1319-1320
158-68
3. Robbins JB, Schneerson R, Szu SC. Specific 29. Compte M, Blanco B, Serrano F, et al.
Downloaded by [Cornell University Library] at 12:47 02 November 2017

16. Carter P. Improving the efficacy of Inhibition of tumor growth in vivo by


acquired immunity. In: Baron S, editor.
antibody-based cancer therapies. Nat Rev in situ secretion of bispecific anti-CEA x
Medical microbiology. University of Texas
Cancer 2001;1(2):118-29 anti-CD3 diabodies from lentivirally
Medical Branch, Galveston, TX: 1996
17. Jones SD, Castillo FJ, Levine HL. Advances transduced human lymphocytes. Cancer
4. Bregenholt S, Haurum J. Pathogen-specific
in the development of therapeutic Gene Ther 2007;14(4):380-8
recombinant human polyclonal antibodies:
monclonal antibodies. BioPharm 30. Sanhadji K, Grave L, Touraine JL, et al.
biodefence applications. Expert Opin Biol
International; 2007;20(10):97-113 Gene transfer of anti-gp41 antibody and
Ther 2004;4(3):387-96
18. Li F, Vijayasankaran N, Shen AY, et al. Cell CD4 immunoadhesin strongly reduces the
5. Kohler G, Milstein C. Continuous cultures
culture processes for monoclonal antibody HIV-1 load in humanized severe combined
of fused cells secreting antibody of
production. MAbs 2010;2(5):466-79 immunodeficient mice. AIDS 2000;14(18):
predefined specificity. Nature 1975;
19. Dumiak M. Making it to manufacturing. 2813-22
256(5517):495-7
IAVI Rep 2014;18(2):4-7 31. Compte M, Alonso-Camino V,
6. Goldstein G. Overview of the development
20. Farid SS. Process economic drivers in Santos-Valle P, et al. Factory neovessels:
of Orthoclone OKT3: monoclonal antibody
industrial monoclonal antibody engineered human blood vessels secreting
for therapeutic use in transplantation.
manufacture. In: Process scale purification therapeutic proteins as a new drug delivery
Transplant Proc 1987;19(2 Suppl 1):1-6
of antibodies. John Wiley & Sons, Inc; system. Gene Ther 2010;17(6):745-51
7. Kimball JA, Norman DJ, Shield CF, et al.
2008. p. 239-61 32. Compte M, Cuesta AM, Sanchez-Martin D,
The OKT3 Antibody Response Study:
21. Winnall WR, Beasley MD, Center RJ, et al. et al. Tumor immunotherapy using
a multicentre study of human anti-mouse
The maturation of antibody technology for gene-modified human mesenchymal stem
antibody (HAMA) production following
the HIV epidemic. Immunol Cell Biol cells loaded into synthetic extracellular
OKT3 use in solid organ transplantation.
2014;92(7):570-7 matrix scaffolds. Stem Cells 2009;27(3):
Transpl Immunol 1995;3(3):212-21
753-60
8. Harding FA, Stickler MM, Razo J, .. This review provides an excellent
overview of recombinant antibody 33. Kasuya K, Shimazu M, Suzuki M, et al.
DuBridge RB. The immunogenicity of
Bispecific anti-HER2 and CD16
humanized and fully human antibodies: fragments and their potential use in HIV
single-chain antibody production prolongs
residual immunogenicity resides in the CDR infection.
the use of stem cell-like cell transplantation
regions. MAbs 2010;2(3):256-65 22. Maloney DG, Grillo-Lopez AJ, White CA, against HER2-overexpressing cancer. Int J
9. Hammers CM, Stanley JR. Antibody phage et al. IDEC-C2B8 (Rituximab) Mol Med 2010;25(2):209-15
display: technique and applications. J Invest anti-CD20 monoclonal antibody therapy in
34. Joseph A, Zheng JH, Chen K, et al.
Dermatol 2014;134(2):e17 patients with relapsed low-grade non-
Inhibition of in vivo HIV infection in
10. Boder ET, Raeeszadeh-Sarmazdeh M, Hodgkin’s lymphoma. Blood 1997;90(6):
humanized mice by gene therapy of human
Price JV. Engineering antibodies by yeast 2188-95
hematopoietic stem cells with a lentiviral
display. Arch Biochem Biophys 2012; 23. Salmon SE, Smith BA. Immunoglobulin vector encoding a broadly neutralizing
526(2):99-106 synthesis and total body tumor cell number anti-HIV antibody. J Virol 2010;84(13):
11. Schaffitzel C, Hanes J, Jermutus L, in IgG multiple myeloma. J Clin Invest 6645-53
Pluckthun A. Ribosome display: an in vitro 1970;49(6):1114-21
. This study is a representative study
method for selection and evolution of 24. Slifka MK, Antia R, Whitmire JK, detailing the use of ex vivo cell
antibodies from libraries. J Immunol Ahmed R. Humoral immunity due to modification for the production of
Methods 1999;231(1-2):119-35 long-lived plasma cells. Immunity 1998;
monoclonal antibodies in situ.
12. Ho RJY, Gibaldi M. Antibodies and 8(3):363-72
35. Frank RT, Edmiston M, Kendall SE, et al.
derivatives. In: Ho RJY, editor. 25. Ridgway JB, Presta LG, Carter P.
Neural stem cells as a novel platform for
Biotechnology and biopharmaceuticals. ‘Knobs-into-holes’ engineering of antibody
tumor-specific delivery of therapeutic
Wiley-Blackwell, Hoboken, NJ 2013. CH3 domains for heavy chain
antibodies. PLoS One 2009;4(12):e8314
p. 139-210

216 Expert Rev. Vaccines 14(2), (2015)


In situ production of therapeutic mAbs Review

36. Reagan MR, Kaplan DL. Concise review: antibody on adenoviral gene transfer. Hum 59. Fausther-Bovendo H, Kobinger GP.
mesenchymal stem cell tumor-homing: Gene Ther 2002;13(12):1483-93 Pre-existing immunity against Ad vectors:
detection methods in disease model systems. 49. Whittington HA, Ashworth LJ, humoral, cellular and innate response,
Stem Cells 2011;29(6):920-7 Hawkins RE. Recombinant adenoviral what’s important? Hum Vaccin
37. Aboody KS, Brown A, Rainov NG, et al. delivery for in vivo expression of scFv Immunother 2014;
Neural stem cells display extensive tropism antibody fusion proteins. Gene Ther 1998; 10(9). [Epub ahead of print]
for pathology in adult brain: evidence from 5(6):770-7 60. Hareendran S, Balakrishnan B, Sen D, et al.
intracranial gliomas. Proc Natl Acad Sci 50. Afanasieva TA, Wittmer M, Vitaliti A, et al. Adeno-associated virus (AAV) vectors in
USA 2000;97(23):12846-51 Single-chain antibody and its derivatives gene therapy: immune challenges and
38. Sanz L, Santos-Valle P, Alonso-Camino V, directed against vascular endothelial growth strategies to circumvent them. Rev Med
et al. Long-term in vivo imaging of human factor: application for antiangiogenic gene Virol 2013;23(6):399-413
angiogenesis: critical role of bone therapy. Gene Ther 2003;10(21):1850-9 .. This review provides an excellent
marrow-derived mesenchymal stem cells for 51. Jiang M, Shi W, Zhang Q, et al. Gene overview of the challenges limiting the
the generation of durable blood vessels. therapy using adenovirus-mediated full- widespread use of adeno-associated virus-
Microvasc Res 2008;75(3):308-14 length anti-HER-2 antibody for mediated gene transfer.
Nauta AJ, Kruisselbrink AB, Lurvink E, HER-2 overexpression cancers. Clin Cancer
Downloaded by [Cornell University Library] at 12:47 02 November 2017

39. 61. Giacca M. Gene therapy. Springer;


et al. Mesenchymal stem cells inhibit Res 2006;12(20 Pt 1):6179-85 Dordrecht; New York: 2010
generation and function of both CD34 52. Liu X, Wu J, Zhang S, et al. Novel 62. Jiang H, Pierce GF, Ozelo MC, et al.
+-derived and monocyte-derived dendritic strategies to augment genetically delivered Evidence of multiyear factor IX expression
cells. J Immunol 2006;177(4):2080-7 immunotoxin molecular therapy for cancer by AAV-mediated gene transfer to skeletal
40. Karnoub AE, Dash AB, Vo AP, et al. therapy. Cancer Gene Ther 2009;16(11): muscle in an individual with severe
Mesenchymal stem cells within tumour 861-72 hemophilia B. Mol Ther 2006;14(3):452-5
stroma promote breast cancer metastasis. 53. Watanabe M, Boyer JL, Hackett NR, et al. 63. Daya S, Berns KI. Gene therapy using
Nature 2007;449(7162):557-63 Genetic delivery of the murine equivalent of adeno-associated virus vectors. Clin
41. Lundstrom K. Gene therapy applications of bevacizumab (Avastin), an anti-vascular Microbiol Rev 2008;21(4):583-93
viral vectors. Technol Cancer Res Treat endothelial growth factor monoclonal
64. Flotte TR, Trapnell BC, Humphries M,
2004;3(5):467-77 antibody, to suppress growth of human
et al. Phase 2 clinical trial of a recombinant
42. Poliani PL, Brok H, Furlan R, et al. tumors in immunodeficient mice. Hum
adeno-associated viral vector expressing
Delivery to the central nervous system of a Gene Ther 2008;19(3):300-10
alpha1-antitrypsin: interim results. Hum
nonreplicative herpes simplex type 1 vector 54. Sofer-Podesta C, Ang J, Hackett NR, et al. Gene Ther 2011;22(10):1239-47
engineered with the interleukin 4 gene Adenovirus-mediated delivery of an anti-V
65. Maguire AM, Simonelli F, Pierce EA, et al.
protects rhesus monkeys from hyperacute antigen monoclonal antibody protects mice
Safety and efficacy of gene transfer for
autoimmune encephalomyelitis. Hum Gene against a lethal Yersinia pestis challenge.
Leber’s congenital amaurosis. N Engl J Med
Ther 2001;12(8):905-20 Infect Immun 2009;77(4):1561-8
2008;358(21):2240-8
43. Furlan R, Poliani PL, Marconi PC, et al. . This study is a representative study
66. Cideciyan AV, Hauswirth WW, Aleman TS,
Central nervous system gene therapy with detailing the use of adenovirus-mediated et al. Vision 1 year after gene therapy for
interleukin-4 inhibits progression of ongoing gene transfer for the production of Leber’s congenital amaurosis. N Engl J Med
relapsing-remitting autoimmune monoclonal antibodies in situ. 2009;361(7):725-7
encephalomyelitis in Biozzi AB/H mice.
55. Van Blarcom TJ, Sofer-Podesta C, Ang J, 67. Nathwani AC, Tuddenham EG,
Gene Ther 2001;8(1):13-19
et al. Affinity maturation of an anti-V Rangarajan S, et al. Adenovirus-associated
44. Kruger C, Hu Y, Pan Q, et al. In situ antigen IgG expressed in situ through virus vector-mediated gene transfer in
delivery of passive immunity by lactobacilli adenovirus gene delivery confers enhanced hemophilia B. N Engl J Med 2011;365(25):
producing single-chain antibodies. Nat protection against Yersinia pestis challenge. 2357-65
Biotechnol 2002;20(7):702-6 Gene Ther 2010;17(7):913-21
68. Gaudet D, Methot J, Dery S, et al. Efficacy
45. Li M, Wu Y, Qiu Y, et al. 56. Watanabe M, Boyer JL, Crystal RG. and long-term safety of alipogene tiparvovec
2A peptide-based, lentivirus-mediated anti- Genetic delivery of bevacizumab to suppress (AAV1-LPLS447X) gene therapy for
death receptor 5 chimeric antibody vascular endothelial growth factor-induced lipoprotein lipase deficiency: an open-label
expression prevents tumor growth in nude high-permeability pulmonary edema. Hum trial. Gene Ther 2013;20(4):361-9
mice. Mol Ther 2012;20(1):46-53 Gene Ther 2009;20(6):598-610
69. Lewis AD, Chen R, Montefiori DC, et al.
46. Vigna E, Pacchiana G, Mazzone M, et al. 57. Skaricic D, Traube C, De B, et al. Genetic Generation of neutralizing activity against
‘Active’ cancer immunotherapy by anti-Met delivery of an anti-RSV antibody to protect human immunodeficiency virus type 1 in
antibody gene transfer. Cancer Res 2008; against pulmonary infection with RSV. serum by antibody gene transfer. J Virol
68(22):9176-83 Virology 2008;378(1):79-85 2002;76(17):8769-75
47. Jiang MH, Chen L, Li LF, et al. A gutless 58. Pereboev A, Borisevich V, Tsuladze G, et al. 70. Johnson PR, Schnepp BC, Zhang J, et al.
adenoviral vector expressing full-length anti- Genetically delivered antibody protects Vector-mediated gene transfer engenders
Her2 antibody. Clin Exp Pharmacol Physiol against West Nile virus. Antiviral Res 2008; long-lived neutralizing activity and
2009;36(9):e26-31 77(1):6-13 protection against SIV infection in
48. Noel D, Pelegrin M, Kramer S, et al. High monkeys. Nat Med 2009;15(8):901-6
in vivo production of a model monoclonal

informahealthcare.com 217
Review Suscovich & Alter

71. Fang J, Qian JJ, Yi S, et al. Stable antibody 82. De BP, Hackett NR, Crystal RG, Boyer JL. 93. Fang J, Yi S, Simmons A, et al. An
expression at therapeutic levels using the Rapid/sustained anti-anthrax passive antibody delivery system for regulated
2A peptide. Nat Biotechnol 2005;23(5): immunity mediated by co-administration of expression of therapeutic levels of
584-90 Ad/AAV. Mol Ther 2008;16(1):203-9 monoclonal antibodies in vivo. Mol Ther
72. Han T, Abdel-Motal UM, Chang DK, .. This study compares adenovirus- and 2007;15(6):1153-9
et al. Human anti-CCR4 minibody gene adeno-associated virus-mediated gene 94. Yamazaki T, Nagashima M, Ninomiya D,
transfer for the treatment of cutaneous transfer appoaches for the production of et al. Passive immune-prophylaxis against
T-cell lymphoma. PLoS One 2012;7(9): monoclonal antibodies in situ. influenza virus infection by the expression
e44455 of neutralizing anti-hemagglutinin
83. Deal C, Balazs AB, Espinosa DA, et al.
73. Lv F, Qiu Y, Zhang Y, et al. monoclonal antibodies from plasmids. Jpn J
Vectored antibody gene delivery protects
Adeno-associated virus-mediated anti- Infect Dis 2011;64(1):40-9
against Plasmodium falciparum sporozoite
DR5 chimeric antibody expression challenge in mice. Proc Natl Acad Sci USA . This study is a representative study
suppresses human tumor growth in nude 2014;111(34):12528-32 detailing the use of DNA for the
mice. Cancer Lett 2011;302(2):119-27 production of monoclonal antibodies
84. Balazs AB, West AP Jr. Antibody gene
74. Shi J, Liu Y, Zheng Y, et al. Therapeutic transfer for HIV immunoprophylaxis. Nat in situ.
expression of an anti-death receptor
Downloaded by [Cornell University Library] at 12:47 02 November 2017

Immunol 2013;14(1):1-5 95. Zabner J, Fasbender AJ, Moninger T, et al.


5 single-chain fixed-variable region prevents Cellular and molecular barriers to gene
85. A Phase 1, Randomized, Blinded,
tumor growth in mice. Cancer Res 2006; transfer by a cationic lipid. J Biol Chem
Dose-escalation Study of rAAV1-PG9DP
66(24):11946-53 1995;270(32):18997-9007
Recombinant AAV Vector Coding for
75. Wang G, Qiu J, Wang R, et al. Persistent PG9 Antibody in Healthy Male Adults. 96. Kariko K, Muramatsu H, Keller JM,
expression of biologically active Available from: http://clinicaltrials.gov/ct2/ Weissman D. Increased erythropoiesis in
anti-HER2 antibody by AAVrh.10-mediated show/NCT01937455 mice injected with submicrogram quantities
gene transfer. Cancer Gene Ther 2010; of pseudouridine-containing
86. Hicks MJ, Rosenberg JB, De BP, et al.
17(8):559-70 mRNA encoding erythropoietin. Mol Ther
AAV-directed persistent expression of a gene
76. Watanabe M, Boyer JL, Crystal RG. encoding anti-nicotine antibody for smoking 2012;20(5):948-53
AAVrh.10-mediated genetic delivery of cessation. Sci Transl Med 2012;4(140): 97. Zangi L, Lui KO, von Gise A, et al.
bevacizumab to the pleura to provide local 140ra187 Modified mRNA directs the fate of heart
anti-VEGF to suppress growth of metastatic progenitor cells and induces vascular
87. Rosenberg JB, Hicks MJ, De BP, et al.
lung tumors. Gene Ther 2010;17(8): regeneration after myocardial infarction. Nat
AAVrh.10-mediated expression of an
1042-51 Biotechnol 2013;31(10):898-907
anti-cocaine antibody mediates persistent
77. Ho DT, Wykoff-Clary S, Gross CS, et al. passive immunization that suppresses 98. Kormann MS, Hasenpusch G, Aneja MK,
Growth inhibition of an established cocaine-induced behavior. Hum Gene Ther et al. Expression of therapeutic proteins
A431 xenograft tumor by a full-length anti- 2012;23(5):451-9 after delivery of chemically modified
EGFR antibody following gene delivery by mRNA in mice. Nat Biotechnol 2011;
88. Yang J, Pattanayak A, Song M, et al.
AAV. Cancer Gene Ther 2009;16(2):184-94 29(2):154-7
Muscle-directed anti-Abeta single-chain
78. Balazs AB, Ouyang Y, Hong CM, et al. antibody delivery via AAV1 reduces cerebral . This study is a representative study
Vectored immunoprophylaxis protects Abeta load in an Alzheimer’s disease mouse detailing the use of RNA for the
humanized mice from mucosal HIV model. J Mol Neurosci 2013;49(2):277-88 production of therapeutic proteins in situ.
transmission. Nat Med 2014;20(3):296-300
89. Mao Y, Kiss S, Boyer JL, et al. Persistent 99. Su H, Takagawa J, Huang Y, et al. Additive
. This study is a representative study suppression of ocular neovascularization effect of AAV-mediated angiopoietin-1 and
detailing the use of adeno-associated with intravitreal administration of VEGF expression on the therapy of
virus-mediated gene transfer for the AAVrh.10 coding for bevacizumab. Hum infarcted heart. Int J Cardiol 2009;133(2):
production of monoclonal antibodies Gene Ther 2011;22(12):1525-35 191-7
in situ. 90. Tjelle TE, Corthay A, Lunde E, et al. 100. Deering RP, Kommareddy S, Ulmer JB,
79. Balazs AB, Chen J, Hong CM, et al. Monoclonal antibodies produced by muscle et al. Nucleic acid vaccines: prospects for
Antibody-based protection against HIV after plasmid injection and electroporation. non-viral delivery of mRNA vaccines.
infection by vectored immunoprophylaxis. Mol Ther 2004;9(3):328-36 Expert Opin Drug Deliv 2014;11(6):885-99
Nature 2012;481(7379):81-4 91. Kitaguchi K, Toda M, Takekoshi M, et al. 101. Brito LA, Chan M, Shaw CA, et al.
80. Balazs AB, Bloom JD, Hong CM, et al. Immune deficiency enhances expression of A cationic nanoemulsion for the delivery of
Broad protection against influenza infection recombinant human antibody in mice after next-generation RNA vaccines. Mol Ther
by vectored immunoprophylaxis in mice. nonviral in vivo gene transfer. Int J Mol 2014;22(12):2118-29
Nat Biotechnol 2013;31(7):647-52 Med 2005;16(4):683-8
102. Geall AJ, Verma A, Otten GR, et al.
81. Zuber C, Mitteregger G, Schuhmann N, 92. Perez N, Bigey P, Scherman D, et al. Nonviral delivery of self-amplifying
et al. Delivery of single-chain antibodies Regulatable systemic production of RNA vaccines. Proc Natl Acad Sci USA
(scFvs) directed against the 37/67 kDa monoclonal antibodies by in vivo muscle 2012;109(36):14604-9
laminin receptor into mice via recombinant electroporation. Genet Vaccines Ther 2004;
adeno-associated viral vectors for prion 2(1):2
disease gene therapy. J Gen Virol 2008;
89(Pt 8):2055-61

218 Expert Rev. Vaccines 14(2), (2015)


In situ production of therapeutic mAbs Review

103. Geall AJ, Mandl CW, Ulmer JB. RNA: the 108. Scallan CD, Jiang H, Liu T, et al. Human 113. Aucoin MG, Perrier M, Kamen AA. Critical
new revolution in nucleic acid vaccines. immunoglobulin inhibits liver transduction assessment of current adeno-associated viral
Semin Immunol 2013;25(2):152-9 by AAV vectors at low AAV2 neutralizing vector production and quantification
.. This review provides an excellent titers in SCID mice. Blood 2006;107(5): methods. Biotechnol Adv 2008;26(1):73-88
overview of the therpeutic potential of 1810-17 114. Mietzsch M, Grasse S, Zurawski C, et al.
both mRNA and self-replicating RNA. 109. Lee GK, Maheshri N, Kaspar B, OneBac: platform for scalable and high-titer
Schaffer DV. PEG conjugation moderately production of adeno-associated virus
104. Kamrud KI, Coffield VM, Owens G, et al.
protects adeno-associated viral vectors serotype 1-12 vectors for gene therapy.
In vitro and in vivo characterization of
against antibody neutralization. Biotechnol Hum Gene Ther 2014;25(3):212-22
microRNA-targeted alphavirus replicon and
Bioeng 2005;92(1):24-34 115. Clement N, Knop DR, Byrne BJ.
helper RNAs. J Virol 2010;84(15):7713-25
110. Huttner NA, Girod A, Perabo L, et al. Large-scale adeno-associated viral vector
105. Tseng YS, Agbandje-McKenna M. Mapping
Genetic modifications of the production using a herpesvirus-based system
the AAV capsid host antibody response
adeno-associated virus type 2 capsid reduce enables manufacturing for clinical studies.
toward the development of second
the affinity and the neutralizing effects of Hum Gene Ther 2009;20(8):796-806
generation gene delivery vectors. Front
human serum antibodies. Gene Ther 2003; 116. Muthumani K, Flingai S, Wise M, et al.
Immunol 2014;5:9
10(26):2139-47 Optimized and enhanced DNA plasmid
Downloaded by [Cornell University Library] at 12:47 02 November 2017

106. Basner-Tschakarjan E, Mingozzi F.


111. Kotin RM. Large-scale recombinant vector based in vivo construction of a
Cell-mediated immunity to AAV vectors,
adeno-associated virus production. Hum neutralizing anti-HIV-1 envelope
evolving concepts and potential solutions.
Mol Genet 2011;20(R1):R2-6 glycoprotein Fab. Hum Vaccin Immunother
Front Immunol 2014;5:350
112. Wright JF, Wellman J, High KA. 2013;9(10):2253-62
107. Louis Jeune V, Joergensen JA, Hajjar RJ,
Manufacturing and regulatory strategies for 117. Yin H, Kanasty RL, Eltoukhy AA, et al.
Weber T. Pre-existing anti-adeno-associated
clinical AAV2-hRPE65. Curr Gene Ther Non-viral vectors for gene-based therapy.
virus antibodies as a challenge in AAV gene
2010;10(5):341-9 Nat Rev Genet 2014;15(8):541-55
therapy. Hum Gene Ther Methods 2013;
24(2):59-67

informahealthcare.com 219

S-ar putea să vă placă și