Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

REVIEW REVIEW

mAbs 2:4, 365-378; July/August 2010; © 2010 Landes Bioscience

Mining human antibody repertoires


Roger R. Beerli1,† and Christoph Rader2
Cytos Biotechnology AG; Schlieren, Switzerland; 2Experimental Transplantation and Immunology Branch; Center for Cancer Research; National Cancer Institute;
1

National Institutes of Health; Bethesda, MD USA


Current Address: Intercell AG; Wagistrasse 25; CH-8952 Schlieren, Switzerland

Key words: human monoclonal antibodies, B cells, hybridoma technology, display technologies, antibody libraries,
antibody engineering

Abbreviations: Ig, immunoglobulin; mAbs, monoclonal antibodies; HAMA, human anti-mouse antibody; HARA, human anti-rat
antibody; HACA, human anti-chimeric antibody; HAHA, human anti-human antibody; FDA, Food and Drug Administration;
EMA, European Medicines Agency; GC, germinal centers; EBV, Epstein-Barr virus; AID, activation-induced deaminase;
PCR, polymerase chain reaction; RT, reverse transcriptase; CDR, complementarity-determining region; FACS,
fluorescence-activated cell sorting; GMP; good manufacturing practice; pAbs, polyclonal antibodies

Human monoclonal antibodies (mAbs) have become drugs of domains. Human mAbs (also referred to as fully human mAbs)
choice for the management of an increasing number of human are defined as having variable domains that are entirely derived
diseases. Human antibody repertoires provide a rich source for from human antibody repertoires, whereas chimeric and human-
human mAbs. Here we review the characteristics of natural and ized mAbs feature variable domains that originate from non-
non-natural human antibody repertoires and their mining with human antibody repertoires. A key feature of human mAbs is
non-combinatorial and combinatorial strategies. In particular, their ability to fully blend in with the human immune system.
we discuss the selection of human mAbs from naïve, immune, Being indistinguishable from endogenous human antibodies,
transgenic and synthetic human antibody repertoires using human mAbs are generally anticipated to have superior pharma-
methods based on hybridoma technology, clonal expansion cokinetic and pharmacodynamic properties compared to mAbs
of peripheral B cells, single-cell PCR, phage display, yeast
from nonhuman antibody repertoires. Ideally, human mAbs
display and mammalian cell display. Our reliance on different
strategies is shifting as we gain experience and refine methods
are not recognized as foreign by the human immune system,
to the efficient generation of human mAbs with superior i.e., are not immunogenic. Human and humanized mAbs have
pharmacokinetic and pharmacodynamic properties. been found to be less immunogenic than nonhuman and chi-
meric mAbs, the latter of which trigger human anti-mouse anti-
body (HAMA), human anti-rat antibody (HARA) and human
anti-chimeric antibody (HACA) responses.1 It is expected, but
Introduction remains to be substantiated, that human mAbs are generally less
immunogenic than humanized mAbs.2,3 Human anti-human
Human antibody repertoires are collections of human immuno- antibody (HAHA) responses are mainly due to idiotypic, allo-
globulin (Ig) genes that encode human heavy and light chains. typic and glycosylation differences between human mAbs and
These include V H, D, JH and CH gene segments of the heavy endogenous human antibodies. Although potentially beneficial
chain locus on chromosome 14; Vk, Jk and C k gene segments of for certain therapeutic applications,4 it is likely that fine tuning
the kappa light chain locus on chromosome 2; and Vl, Jl and Cl of human mAb engineering and manufacturing will reduce the
gene segments of the lambda light chain locus on chromosome remaining immunogenicity of human mAbs.
22 in the human genome. In addition, transgenic animals with Hybridoma technology 5 has been extremely successful in the
complete or partial human antibody repertoires have been gener- generation of mouse and rat mAbs; however, the initial failure
ated. Human antibody repertoires can be mined in vivo and in to robustly apply this method to the generation of human mAbs
vitro for human monoclonal antibodies (mAbs). paired with the prospect of lucrative intellectual property accel-
Chimeric, humanized and human mAbs, which collectively erated the development of a variety of alternative methods over
are the prevailing formats of therapeutic mAbs, share human the past three decades.2,6-8 The medical and commercial success
constant domains but are discerned by the origin of their variable of mAbs in the management of human disease drove this growth
and diversification.9,10 Although only six out of 28 mAbs that were
Correspondence to: Roger R. Beerli and Christoph Rader; approved by the US Food and Drug Administration (FDA) and/
Email: rbeerli@intercell.com and raderc@mail.nih.gov or the European Medicines Agency (EMA) are human mAbs
Submitted: 03/26/10; Accepted: 04/27/10 (Table 1), four of these antibodies were approved as recently
Previously published online: as 2009, and their proportion is likely to increase, owing to an
www.landesbioscience.com/journals/mabs/article/12187
unremitting pipeline of human mAbs in Phase 3 clinical trials.11

www.landesbioscience.com mAbs 365


Table 1. Human mAbs approved in the United States and the European Union
Generic name
Format (origin) Antigen Indications Approval
(trade name; company)
Rheumatoid arthritis; juvenile idiopathic
Adalimumab 2002 (FDA),
IgG1k (phage display) TNFa arthritis; psoriatic arthritis; plaque psoriasis;
(Humira®; Abbott) 2003 (EMA)
ankylosing spondylitis; Crohn’s disease
Panitumumab 2006 (FDA),
IgG2k (transgenic mouse) EGFR Metastatic colorectal cancer
(Vectibix®; Amgen) 2007 (EMA)
Golimumab Rheumatoid arthritis; psoriatic arthritis; 2009 (FDA),
IgG1k (transgenic mouse) TNFa
(Simponi®; Johnson & Johnson) ankylosing spondylitis 2009 (EMA)
Canakinumab Cryopyrin-associated periodic syndrome 2009 (FDA),
IgG1k (transgenic mouse) IL-1b
(Ilaris®; Novartis) (CAPS) 2009 (EMA)
Ustekinumab IL-12 and IL-23 2009 (FDA),
IgG1k (transgenic mouse) Plaque psoriasis
(Stelara®; Johnson & Johnson) ­(common p40 subunit) 2009 (EMA)
Ofatumumab 2009 (FDA),
IgG1k (transgenic mouse) CD20 Chronic lymphocytic leukemia
(Arzerra®; Genmab) 2010 (EMA)

Here we review the generation of human mAbs from naïve, mAbs, first with non-combinatorial and later with combina-
immune, transgenic and synthetic human antibody repertoires. torial strategies. By contrast, combinatorial strategies have
Strictly speaking, only human mAbs from naïve and immune dominated access to human mAbs from the naïve repertoire
repertoires, which we summarize as natural repertoires in the (Table 2).
next section, as well as human mAbs from transgenic reper- The bone marrow is a primary lymphoid tissue of interest
toires, which nonetheless are products of artificial immune sys- for mining natural human antibody repertoires. The principle
tems, meet the above stated criterion of having variable domains site of differentiation of hematopoietic stem cells into immature
that are entirely derived from human antibody repertoires and B cells,12 the bone marrow generates pro-B cells and pre-B cells
are therefore indistinguishable from endogenous human anti- with partially rearranged heavy and light chain genes. While
bodies. By contrast, human mAbs from synthetic repertoires these partially developed human antibody repertoires are gener-
generally contain artificial sequences that are not encoded in ally not suitable for mining human mAbs, surrobody libraries
the human genome and cannot be generated by natural pro- that pair rearranged heavy chains with surrogate light chains of
cesses such as V HDJH, VkJk and VlJl rearrangements and pre-B cells have been described recently.13 The ultimate products
somatic hypermutation. Transgenic and synthetic repertoires of B-cell development in the bone marrow are immature B cells
are summarized as non-natural repertoires in a subsequent sec- that express cell surface IgM. Upon exiting the bone marrow and
tion. Furthermore, we distinguish between non-combinatorial entering the circulation, immature B cells mature to naïve B cells,
and combinatorial strategies for mining human antibody rep- the latter of which express cell surface IgD in addition to IgM.
ertoires. Non-combinatorial strategies retrieve human mAbs Immature and naïve B cells collectively provide a fully developed
from single B-cells with the original heavy and light chain human antibody repertoire that has not been shaped, at least not
pairs (Fig. 1). Combinatorial strategies involve human anti- extensively, by antigen encounter and is therefore termed primary
body libraries with randomly combined heavy and light chains repertoire or naïve repertoire.
(Fig. 2). Hybridoma technology and phage display technology When naïve B cells encounter new antigens that bind to
exemplify non-combinatorial and combinatorial mining tools, their surface Ig they become activated B cells, proliferate and go
respectively. Both approaches have led to FDA- and EMA- through differentiation processes that typically involve somatic
approved human mAbs (Table 1). hypermutation and class switch recombination from the earliest
expressed IgM isotype to IgG, IgE and IgA isotypes. These pro-
Natural Repertoires cesses are T-cell-dependent and take place in germinal centers
(GC) of secondary lymphoid tissues such as lymph nodes, spleen
Each B cell encodes a single antibody with defined specificity. and tonsils. Post-GC B cells with affinity and isotype matured
The clonal diversity of the natural human antibody repertoire antibodies differentiate further into antibody secreting cells and
is defined by the number of B cells that encode antibodies with memory B cells.14 Antibody secreting cells can be divided into
unique specificities, i.e., the number of independent clones. In proliferating plasma blasts and fully differentiated plasma cells
an individual, this number is estimated to be at least 108. B that no longer proliferate. Plasma cells exit secondary lymphoid
cells from primary and secondary lymphoid tissues along with tissues, enter the circulation, and migrate to the bone marrow
B cells in circulation, i.e., in peripheral blood, constitute the where they may survive for weeks, months, or even years in spe-
natural human antibody repertoire. The natural repertoire cialized niches. Antibody titers in circulation are maintained by
can be divided into naïve and immune repertoires depending plasma cells residing in the bone marrow.15 In contrast to anti-
on whether or not it has been shaped by antigen encounter. body secreting cells, memory B cells do not secrete antibodies but
The immune repertoire has been extensively mined for human express cell surface Ig. Memory B cells are found in secondary

366 mAbs Volume 2 Issue 4


Figure 1. Non-combinatorial mining of human antibody repertoires. (A) Human antibody repertoires consist of polyclonal mixtures of a variety of B
cells that express functional human antibodies, including naive and post-GC B cells that represent naïve and immune repertoires, respectively.
Post-GC B cells, which differentiate into antibody secreting cells and memory B cells, provide a human antibody repertoire that has been extensively
mined with non-combinatorial strategies. (B) Hybridoma technology (top), EBV immortalization (center), and single-cell sorting (bottom) allow the
isolation and expansion of monoclonal B cells which are then screened with an antigen of interest. Cell surface Ig can be utilized to first enrich B cells
that express human antibodies to the antigen of interest. (C) Heavy and light chain cDNAs are amplified from mRNA of monoclonal B cells and B-cell
lines by RT-PCR. (D) After cloning heavy and light chain cDNAs into ectopic expression vectors, human mAbs are purified from the supernatant of
transfected mammalian cells (typically Chinese hamster ovary CHO or mouse myeloma NS0 cells) in high yields.

lymphoid tissues, in circulation and in the bone marrow. Upon Human immune repertoires are highly attractive because
recall antigen encounters, memory B cells rapidly differentiate they contain antibodies of high affinity with minimal immu-
into antibody secreting cells. Both memory B cells and plasma nogenicity and off-target reactivity. Unlike nonhuman immune
cells can be long-lived, maintaining immunological memory for repertoires that have been mined extensively for mAbs, the
a lifetime.16 human immune repertoire lacks both availability and accessi-
The secondary repertoire, or immune repertoire, is largely bility. First of all, humans cannot be exposed to antigens at
defined by the pool of post-GC B cells, plasma blasts, plasma will, limiting available immune repertoires to those induced
cells and memory B cells. Expression of CD27 distinguishes by infections, vaccinations, autoimmunity and alloimmunity.
this B-cell pool from other B cells. Antibody-secreting cells Furthermore, secondary lymphoid tissues such as the spleen,
and memory B cells can also arise from T-cell-independent which is a preferred source of mouse, rat and rabbit immune
processes that play a role in both adaptive and innate immunity. repertoires for mAb mining, are difficult to access in humans.
Adaptive immunity to T-cell-independent antigens such as Although tonsillectomy provides access to tonsils, it generally
carbohydrates and lipids contributes to the immune repertoire. does not coincide with an immune response of interest. Access
By contrast, cells expressing natural antibodies, which play a to the human immune repertoire is therefore generally limited
key role in innate immunity,17 can be considered to belong to to peripheral blood and, to a lesser extent, bone marrow. As dis-
the naïve repertoire. These two types are difficult to distinguish cussed above, these compartments also contain the naïve rep-
and typically share IgM isotype, low affinity and polyreactivity. ertoire. Combinatorial strategies allow the retrieval of human
They illustrate that immune and naïve repertoires often mAbs from the naïve repertoire. Typically, these mAbs need
overlap without clear anatomical localizations or functional to be improved by affinity maturation in vitro to compensate
distinctions. for their lack of efficient selection by antigens in vivo. The next

www.landesbioscience.com mAbs 367


Figure 2. Combinatorial mining of human antibody repertoires. Polyclonal or oligoclonal mixtures of human B cells (A) are used in bulk to isolate
mRNA and amplify cDNA of heavy and light chains (B). (C) Heavy and light chain cDNAs are cloned into display vectors that afford antibody libraries
with a physical linkage of genotype (cDNA) and phenotype (protein). Shown as examples are filamentous phage that encode and display Fab (top) and
virus-infected mammalian cells that express and display scFv (bottom). Note that heavy and light chains are randomly combined. (D) Antibody
libraries are then selected by several rounds of panning on immobilized antigen (top) or screened by a single round of FACS using fluorescently
labeled antigen (bottom). (See text for details). The physical linkage of genotype and phenotype allows the enrichment and amplification of displayed
antibodies. (E) Human mAbs from antibody libraries are manufactured the same way as human mAbs from monoclonal B cells and B-cell lines.

Table 2. Principal mining tools for human antibody repertoires


Mining tools
Repertoire
Non-combinatorial Combinatorial
Naïve Phage display; yeast display
Natural Hybridoma technology; clonal expansion; Phage display; yeast display; mammalian cell
Immune
single-cell PCR display
Transgenic Hybridoma technology
Non-natural
Synthetic Phage display; yeast display; ribosome display

two sections discuss various features of naïve and immune rep- available and can be produced in almost unlimited quantities due
ertoires that are exploited in non-combinatorial and combinato- to this robust underlying technology.5 Unfortunately, the therapeu-
rial strategies for mining human mAbs. tic use of rodent mAbs in humans is limited by their propensity to
induce strong HAMA and HARA responses that quickly neutral-
Non-Combinatorial Mining ize the rodent mAb. Given this, the mining of human antibody
repertoires using hybridoma technology was investigated exten-
Hybridoma technology. The advent of hybridoma technology had sively. Despite significant efforts, the adaptation of hybridoma
a tremendous impact on biomedical research, yielding mAbs that technology to the generation of human mAbs has been a slow and
have become an indispensable part of everyday laboratory work. difficult process due to the limited number of B cells stimulated
Today rodent mAbs against a plethora of antigens are widely with an antigen of interest and a lack of suitable drug-resistant

368 mAbs Volume 2 Issue 4


myeloma cell fusion partners for the immortalization of human involving EBV-immortalization of peripheral B cells followed by
B cells. Thus, mouse myeloma cells were initially used for the fusion with myeloma cells and cloning of hybridomas were estab-
generation of mouse-human heterohybridomas.18-20 Such hybrid lished.45-48 This method has yielded several human mAbs with
cells were shown to have a strong tendency to lose human chro- therapeutic potential, including neutralizing antibodies against
mosomes, including chromosome 2 carrying the kappa light chain the hemagglutinin protein derived from the 1918 H1N1 pan-
locus.21 While this precluded an extensive use of mouse myeloma demic influenza virus.49
cells for human mAb production, protocols have been established As an alternative to the formation of hybridomas, EBV-
that may ameliorate the undesirable behavior of heterohybridomas transformed cells have been infected with a retrovirus encoding
and allow long-term production of human mAbs.22 the ras oncogene, yielding cells with similar cloning efficiencies
Given the genetic instability of heterohybridomas, the use of and levels of antibody secretion.50 The most significant improve-
alternative fusion partners, particularly human lymphoblastoid ment of EBV immortalization came from the observation that
cell lines and human myeloma cells, has been explored exten- addition of the TLR9 ligand CpG to human B cells dramatically
sively.23-28 While in principle successful, the yield of viable hybrid- increases the efficiency of EBV immortalization and facilitates the
omas is typically too poor for practical applications. In addition, establishment of clonal lines stably secreting specific antibody. In
such hybridomas often secrete permutated Igs derived from this manner, mAbs potently neutralizing the SARS corona virus
both fusion partners. The use of certain mouse-human hybrid or the H5N1 Influenza A virus were isolated.51,52
cells including heteromyelomas and heterohybridomas as fusion Strategies allowing the long-term growth and cloning of
partner appears to resolve these problems, while at the same time human B cells, without immortalization by transformation with
generating stable antibody producing hybridomas.29-32 In addi- a virus or an oncogene, have also been explored. In initial experi-
tion, a promising human myeloma cell line allowing efficient ments, short-term B-cell clones were formed when cultured in the
generation of stable and highly productive human hybridomas presence of EL-4B5 thymoma cells in conjunction with human
was described, even though a more widespread use of this cell line T-cell plus macrophage supernatant.53,54 Improved proliferation
awaits the production of a subline lacking expression of an endog- and antibody production were found under more defined condi-
enous l chain.33 Finally, the use of genetically engineered mouse tions, when B cells were activated by IL-4 treatment and CD40
myeloma cells allows for highly efficient formation of stable het- triggering.55,56 A combination of both methods together with
erohybridomas,34 with a propensity to produce affinity-matured further refinements proved extremely efficient, allowing direct
IgG antibodies when appropriately pre-treated CD27-positive B establishment of viable B-cell cultures from single, antigen-
cells are used for fusion.35 specific B cells.57,58 Similar to EBV immortalization, these B-cell
As mentioned above, human hybridoma technology not only clones secrete sufficient amounts of antibody into the superna-
suffered from the lack of suitable fusion partners, but also from tant, and production of specific antibody can be validated prior
a shortage of antigen-stimulated B cells. Because ethical consid- to cloning of the variable domains. Further, high throughput
erations typically prevent antigen exposure at will in humans, screens based on short term clonal expansion of activated periph-
alternative methods to improve access to the human immune eral B cells are possible and have led to the identification of HIV-1
repertoire were developed. Human peripheral blood lymphocytes neutralizing antibodies.59
transplanted into immune-compromised mice were shown to Recently, a promising method involving genetic reprogram-
give rise to a secondary humoral immune response upon antigen ming of human memory B cells was described.60 Retrovirus-
stimulation,36,37 and provide access to the human memory com- mediated expression of Bcl-6 and Bcl-x L in peripheral memory B
partment through hybridoma technology.38,39 cells, combined with culturing on CD40L-expressing L cells in
In summary, each of the methods described is either labor- the presence of IL-21, converts them to highly proliferating, cell
intensive or suffers from drawbacks when compared to the classi- surface Ig-positive, antibody secreting B cells that can be cloned
cal hybridoma technology. Thus, hybridoma technology has not efficiently. The proteins Bcl-6 and Bcl-x L are typically expressed
become the method of choice for mining human antibody rep- in GC B cells, and the resulting cells have features of GC B cells,
ertoires, leading to tremendous efforts to develop the alternative including expression of activation-induced deaminase (AID). It
methods discussed in the following sections. is of interest whether this method will find broader application in
Clonal expansion of peripheral B cells. One of the most the generation of human mAbs.
popular methods for harnessing the human immune response Single-cell PCR. The most straightforward, though tech-
involves immortalization of peripheral B cells with Epstein-Barr nically challenging, non-combinatorial strategy for mining
virus (EBV). The first demonstration of EBV immortalization human antibody repertoires is single-cell polymerase chain
of human B cells into lymphoblastoid cell lines in vitro, was reaction (PCR). If both heavy and light chain variable domains
described almost 40 years ago.40 There have been numerous can be amplified in an efficient and reliable manner from single
reports since then, although initial success was limited due to low cells, then antibodies with original heavy and light chain pair-
efficiency of B-cell immortalization and cloning and the small ings can be cloned on a cell-by-cell basis without the need to
quantities of antibody secreted.41,42 Better cloning efficiency, sta- grow individual B-cell clones. In principle, either memory B
bility and productivity can be achieved when EBV-immortalized cells or plasma cells can serve as a source of rearranged anti-
antibody secreting cells are fused with mouse myeloma or mouse- gen-binding sites. As discussed, peripheral memory B cells are
human heteromyeloma cells.43,44 Thus, multi-step procedures known to persist for many years after antigen exposure, thereby

www.landesbioscience.com mAbs 369


offering a diverse and attractive repertoire of antigen specifici- Like other cDNA libraries, the first antibody libraries were
ties readily available in the blood. As memory B cells contain based on lambda phage vectors that carried heavy and light
only relatively small quantities of specific RNA, the successful chain cDNAs and facilitated their expression upon infection of
cloning of both heavy and light chain variable domains from Escherichia coli bacteria.72 Lysis plaques of lambda phage on bac-
a single B cell poses a significant technical challenge. In spite terial lawns were then screened with antigens of interest. This
of this, antibodies have successfully been cloned from single method was used to clone a diverse panel of human mAbs in
peripheral B cells, including mAbs against self-antigens,61,62 tet- Fab format to tetanus toxoid after generating an antibody library
anus toxoid57 and HIV-1 gp140.63 The efficiency of cloning rel- from peripheral blood lymphocytes isolated from human donors
evant antibodies has been improved, e.g., by enriching specific who had received a booster vaccination.73,74 A major drawback of
B cells using antigen-coated magnetic beads,57 or by directly this method is its limitation to screening as opposed to selecting
isolating antigen-specific B cells by fluorescence-activated cell antibody libraries. The number of independent clones, i.e., lysis
sorting (FACS).63 plaques, that can be screened, even if the antigen of interest is in
Compared to memory B cells, plasma cells have the disad- unlimited supply, is practically confined to 106.
vantage that they are present in the blood only for short periods Subsequent combinatorial strategies, e.g., phage, yeast and
of time after antigen exposure, after which they are only acces- mammalian cell display, replaced screening with selection, allow-
sible in the bone marrow. Nonetheless, a known antigen exposure ing the mining of much larger antibody libraries approaching 1011
or challenge, e.g., a booster immunization, induces circulating independent clones. Selection is facilitated by display technologies
plasma cells, of which a majority is specific for the antigen after that physically link a displayed antibody fragment to its cDNA
approximately one week.64 Although no highly plasma cell- in a defined particle such as a filamentous phage, a ribosome, or
specific marker is known to date, they are readily isolated by a cell. Millions to billions to, theoretically, trillions of indepen-
FACS, e.g., based on their high expression of CD19 and CD38, dent particles constitute an antibody library that can be mined
and intermediate expression of CD45.65 These cells represent with antigens of interest. Particles that display antibody frag-
dedicated antibody factories and contain huge amounts of spe- ments with high affinity to the antigen are isolated. Because of
cific heavy and light chain mRNAs, thereby facilitating antibody the simultaneous isolation of their cDNA, the displayed antibody
cloning by reverse transcriptase (RT)-PCR. Numerous human fragments can be copied, enabling multiple rounds of selection.
mAbs have been cloned from plasma cells to date, including In other words, the physical linkage of phenotype and genotype
mAbs against tetanus toxoid and influenza virus.64-66 Recently, effectively links recognition and replication. In addition, DNA
using a method related to microengraving,67 microwell array sequencing may readily identify phenotype and genotype.
chips were successfully used for high-throughput screening of Phage display. The generation and selection of peptide librar-
human antibodies secreted by single CD138 + plasma cells fol- ies displayed on filamentous phage marked the advent of display
lowed by RT-PCR cloning.68 technologies.75 The use of phage display for the generation and
selection of antibody libraries, displayed in either scFv76-78 or
Combinatorial Mining Fab79,80 format, revolutionized the field of mAbs and antibody
engineering in the early 1990s, arguably accelerated by an
Combinatorial mining of human antibody repertoires is inter- intense competition between researchers at the Medical Research
twined with the concept of antibody libraries.8,69 Antibody librar- Council (Cambridge, UK) and The Scripps Research Institute
ies randomly combine heavy and light chain cDNAs obtained (La Jolla, California, USA). Over the past 20 years, phage display
through RT-PCR from mRNA of B-cell pools into expression has proven to be the most robust and versatile mining tool for
vectors that afford a compartmental or physical linkage of phe- human antibody repertoires, yielding human mAbs to virtually
notype (protein) and genotype (cDNA). In contrast to non- any antigen of interest.8 The process of multiple rounds of selec-
combinatorial mining, the source of human antibody repertoires tion on a purified antigen or on antigen-expressing cells, referred
subjected to combinatorial mining is B-cell mRNA rather than B to as panning, can be adapted to positively or negatively select
cells. Thus, antibody secreting cells, due to their higher heavy and a range of desired antibody properties, such as affinity, specific-
light chain gene transcript levels compared to other B cells, can ity, manufacturability and catalytic activity. While scFv (~25
have much greater representation in antibody libraries. This bias kDa) and Fab (~50 kDa) still dominate the phage display format,
can be highly desirable for the combinatorial mining of immune, human antibody repertoires have also been mined through the
but not naïve, repertoires. To increase the diversity of indepen- display and selection of single variable domains (~12.5 kDa) of
dent clones in antibody libraries from naïve repertoires, RT-PCR heavy and light chain.81,82
of the heavy chain mRNA can be restricted to the IgM isotype. Typically, recombinant fusion to the N-terminus of phage
However, combinatorial strategies have inherent biases due to the protein pIII or a fragment of pIII provides the physical linkage
RT-PCR cloning of heavy and light chain cDNAs and due to of antibody fragment and phage, facilitating monovalent display
the expression and folding of non-natural antibody fragments in in phagemid systems and multivalent display in phage systems.83
non-B-cell environments. Nonetheless, high-throughput DNA Other multivalent display systems use phage proteins pVIII and
sequencing recently confirmed that large antibody libraries can pIX as fusion partners.79,84 For the generation of human mAbs
accurately mimic natural human antibody repertoires in terms of with high affinity, monovalent Fab display through phage protein
molecular diversity.70,71 pIII is generally preferred.85 Phage display relies on the expression

370 mAbs Volume 2 Issue 4


and folding of antibody fragments in the periplasm of gram- endogenous human antibodies involves the shuffling of heavy
negative bacteria, typically Escherichia coli. Although this com- and light chains.105 This method, known as chain shuffling, can
partment provides an oxidizing milieu suitable for disulfide bridge yield excellent human mAbs from moderately diverse antibody
formation and mimics the endoplasmic reticulum of eukaryotic libraries with 109 independent clones.106 Chain shuffling can also
cells surprisingly well, prokaryotic factors that skew the select- be used to convert mouse mAbs to human mAbs via sequen-
able diversity of antibody libraries are inevitable. For example, tial replacement of mouse heavy and light chains with human
the lack of chaperones in bacteria can lead to inadequate folding heavy and light chain libraries, respectively, followed by selection
of scFv and Fab and result in toxicity. through phage display.107 Adalimumab, the first human mAb
Human antibody libraries displayed on phage were generated approved by FDA and EMA (Table 1), was derived from a mouse
from immune and naïve repertoires at about the same time. The mAb to human TNFa via this method.108 A similar method,
first human immune repertoires mined by phage display were also based on phage display, preserves only the complementarity
based on peripheral B cells and bone marrow of vaccinated or determining region (CDR) 3 sequences of mouse heavy and light
infected human donors and yielded human mAbs to tetanus tox- chains, i.e., HCDR3 and LCDR3, while converting everything
oid, HIV-1 gp120, and other viral antigens.85-89 Notably, antibody else to human sequences.109
libraries derived from bone marrow of HIV-1-positive human A general concern about the de novo generation of human
donors with serum antibodies against a variety of other viruses mAbs from naïve and synthetic repertoires in vitro is their lack of
yielded human mAbs to each of these viruses following selection exposure to negative selection processes in vivo which are integral
by phage display.90 The bone marrow was conceptualized as the to immune tolerance mechanisms and efficiently remove undesir-
central bank for antigen-experienced B cells that carried a record able off-target reactivities. Addressing this concern, phage display
of antibody responses to past viral exposures. Autoimmune rep- has also been used to mine non-combinatorial antibody librar-
ertoires from human donors who have serum antibodies to DNA ies, i.e., antibody libraries that retain original heavy and light
and other autoantigens have also been accessed by phage dis- chain pairings of human antibody repertoires through in-cell
play.91-93 Tumor-infiltrating B cells provide a unique immune rep- RT-PCR.110 This method is of particular interest for the identifi-
ertoire in cancer patients that has been mined with phage display cation of human antibodies in autoimmune repertoires.
for potentially therapeutic human mAbs to tumor antigens.94-96 Yeast display. The expression, folding and display of antibody
Alternatively, peripheral B cells from cancer patients vaccinated fragments on the surface of cells from the yeast Saccharomyces
with autologous tumor cells have been used to generate and select cerevisiae involve eukaryotic processes that are similar to those
antibody libraries by phage display.97 Peripheral B cells and phage encountered in the natural B-cell environment. Antibody frag-
display were also employed to mine an alloimmune repertoire of ments are channeled through endoplasmic reticulum and Golgi
a cancer patient who had been treated with allogeneic hematopoi- apparatus, ensuring proper disulfide bridge formation and
etic stem cell transplantation, revealing serum antibodies to N-linked glycosylation.111,112 Thus, yeast display may address
tumor antigens that might have potential as therapeutic human some of the above mentioned biases prokaryotic systems impose
mAbs.98 on antibody libraries. In fact, in a direct comparison based on the
The first human naïve repertoire mined by phage display was same human antibody repertoire, yeast display yielded a more
based on peripheral B cells and yielded human mAbs to a vari- diverse set of human mAbs than phage display.113 The physical
ety of different antigens that had not been encountered in vivo.99 linkage of antibody fragment and cell is provided through recom-
Importantly, this method allowed the generation of human binant fusion to the C-terminus of the secreted protein Aga2,
mAbs to human antigens.100 These studies demonstrated that the which covalently associates with the cell surface protein Aga1.114
random combination of heavy and light chain can yield human Formats of antibody fragments that have been used for yeast
mAbs with new specificities. Due to the overlap of naïve and display include scFv,111 Fab115 and scFab.116
immune repertoires, as well as the bias of mRNA from antibody Initially limited by the number of independent clones that
secreting cells, antibody libraries from naïve repertoires typically could be transformed, yeast display has become a powerful
contain a mixture of heavy and light chain cDNAs with and mining tool for large antibody libraries from human naïve rep-
without somatic hypermutations. Human mAbs derived from ertoires.117-119 In addition, yeast display has been successfully
naïve repertoires generally have lower affinities compared to employed for the mining of human immune repertoires.113,120
human mAbs from immune repertoires, but this shortcoming Interestingly, the haploid/diploid lifecycle of yeast can be
can be overcome by increasing the number of independent clones, exploited in recombinatorial strategies that generate large anti-
thereby generating a larger antibody library.101-104 However, it is body libraries in diploid cells by mating a haploid cell pool har-
now common to mature the affinity of human mAbs derived boring a heavy chain library with a haploid cell pool harboring a
from naïve repertoires with subsequent antibody engineering light chain library.118,120 Such recombinatorial strategies based on
methods that are also based on display technologies. Methods yeast display have also been used for chain shuffling.121
that involve the randomization of certain sequences within heavy In contrast to phage display, antibody libraries displayed on
or light chain are discussed in a subsequent subsection on syn- yeast cells are simultaneously screened and selected with an anti-
thetic repertoires. gen of interest. Screening is carried out by flow cytometry and
A method that preserves natural V HDJH, VkJk and VlJl selection by FACS. The ability to detect and sort individual bind-
sequences and yields human mAbs indistinguishable from ing events during the selection process has made yeast display

www.landesbioscience.com mAbs 371


an exceptional method for the affinity maturation of mAbs,122 antibodies in a single round of FACS. Due to the highly repli-
including human mAbs derived from naïve repertoires by phage cation-competent nature of the Sindbis vector, viruses encoding
display.115 a specific antibody can readily be amplified from single-sorted
Mammalian cell display. A screening/selection system based cells. Thus, the binding properties of antibodies can be charac-
entirely on expression of human antibodies in their natural envi- terized prior to cloning, sequencing and subsequent production
ronment, i.e., the mammalian cell, may be best suited for the as whole IgG. Towards a clinical application of the technology,
mining of human antibody repertoires. Within this system, all therapeutic mAbs against nicotine and influenza A M2 protein
necessary components for human antibody synthesis and process- have been isolated, characterized and validated preclinically to
ing are available at physiological levels. Compared to yeast cells, date.125,127
antibody repertoires expressed in mammalian cells are less biased A drawback of mammalian cell display is that the number
by properties other than antigen binding. Several laboratories of cells that can be handled at the same time is limited. This is
have therefore developed strategies for human mAb mining by particularly true if the screening is done under conditions where
mammalian cell display. The most straightforward approach is one (or close to one) antibody species is expressed per cell, limit-
to transfect an antibody library with a vector directing antibody ing the diversity of libraries that can be conveniently handled to
expression to the cell surface by means of a C-terminal trans- about 107 independent clones. This is suboptimal for combinato-
membrane region, thereby allowing for selection of cells express- rial libraries and pre-selection of B cells for antigen specificity is
ing specific antibody based on antigen binding, e.g., by FACS. advisable. It is therefore no coincidence that the human mAbs
In a proof-of-concept study involving the expression of a mix- isolated by mammalian cell display to date are derived from pools
ture of two different CD22-specific scFv on the surface of human of B cells enriched for antigen specificity.125,127 Because these
embryonic kidney 293T cells, it was estimated that an up to enriched combinatorial libraries are of low diversity, there is a
240-fold enrichment can be achieved with only 2-fold affinity high likelihood that the resulting human mAbs contain natural
differences.123 It remains to be shown whether this approach will pairs of heavy and light chains, i.e., are derived from clonally
allow for isolation of antibodies from more complex mixtures, related B cells. An advantage common to all screening systems
such as a library. A general limitation of screening by plasmid based entirely on mammalian cell expression is that they have
transfection is the potential to deliver multiple plasmids per cell, a built-in selection for efficient expression in mammalian cells.
leading to the expression of an ill-defined number of different As antibody good manufacturing practice (GMP) processes typi-
antibodies on each cell. It would then require multiple rounds cally involve expression in mammalian cells, this is expected to
of selection in order to enrich for specific antibodies, even if the have a significant impact on the cost of goods.
library diversity is small. This problem can be circumvented with
viral expression vectors. As infection rates can be dosed precisely, Non-Natural Repertoires
conditions can be used where one cell expresses exactly one anti-
body species. Transgenic repertoires. More than 20 years ago, a proof of con-
One viral system suitable for screening of human antibody cept study first demonstrated that human Ig gene segments intro-
libraries on the surface of mammalian cells is based on vaccinia duced into a mouse can be accessed by the mouse immune system
virus.124 Using separate heavy and light chain libraries, African and used to express functional antibodies.128 In this approach, a
green monkey kidney BSC-1 cells are co-infected, human IgG human heavy chain mini locus containing V H, D and JH gene
antibodies expressed on the cell surface and cells encoding spe- segments linked to a C m gene was introduced into an embry-
cific antibody isolated by FACS. Because heavy and light chains onic stem cell that was used to develop a transgenic mouse. In
are expressed from separate vectors, infection rates are kept high this mouse, a large fraction of lymphoid cells expressed human
to ascertain that a substantial fraction of cells expresses func- antibody genes and this led to measurable levels of hybrid IgM
tional antibody. As a consequence, some of the cells isolated will with human heavy chains and mouse light chains in the serum.
express more than one heavy and light chain pair, and several Further, it was shown that conventional hybridoma technology
rounds of selection are beneficial. After screening, specific recom- could be used to recover recombinant monoclonal IgM, thus lay-
binants can be recovered from small numbers of selected cells, ing the ground for the production of human antibodies in so
perhaps even single cells.124 called humanized mice.
A two-step method for the isolation of human antibodies by Production of human antibodies in genetically modified mice
mammalian cell display has been described recently.125 Using was immediately recognized as a highly attractive approach for
virus like particles with highly repetitive arrays of antigen,126 the generation of human mAbs against human antigens, as mice
pools of B cells specific for an antigen of interest are first isolated can readily be immunized with any antigen of interest and are not
from peripheral blood lymphocytes of immunized or naturally tolerant to most human proteins.129 While the mice described by
immune human donors by FACS. Recombinant, antigen-focused Brüggemann et al.128 only expressed a human heavy chain mini
scFv libraries are then generated from these B cells and screened locus in the presence of the endogenous mouse heavy and light
by mammalian cell surface display using a Sindbis virus expres- chain loci, the production of fully human antibodies became
sion system. Infection of baby hamster kidney cells at a low feasible with the development of mice with additional genetic
multiplicity of infection ensures that one cell expresses only one modifications. In two different laboratories, mice were generated
antibody species, thus allowing identification of antigen-specific that combined disruptions of the endogenous mouse heavy and

372 mAbs Volume 2 Issue 4


kappa light chain loci and transgenes encoding human heavy and responses, transgenic mice were generated that carried, for the
kappa light chain loci.130,131 The endogenous mouse lambda light first time, large portions of all three human Ig loci, including the
chain locus was left unmodified by both groups because it only human lambda light chain locus, in a background in which the
contributes to 5–15% of the antibody repertoire.132 In the report endogenous mouse heavy and kappa light chain loci had been
by Lonberg et al.131 the human heavy chain locus comprised inactivated. Upon immunization, these mice generated antibod-
3 V H, 16 D, all 6 JH and the C m and C g1 constant region gene seg- ies containing both human kappa and lambda light chains.136
ments, whereas the light chain locus comprised 4 Vk, all 5 Jk and To generate more complete humanized mice, the to date larg-
the C k gene segments. The mice described by Green et al.130 car- est fraction of the human germline repertoire was introduced by
ried a repertoire of similar diversity, with the heavy chain locus microcell-mediated chromosome transfer. In this manner, tran-
including 5 V H, all 25 D, all 6 JH and the C m and C d constant schromosomal mice were generated that express the complete
region gene segments, and the light chain locus including 2 Vk, heavy chain and kappa light chain repertoire in absence of mouse
all 5 Jk and the C k gene segments. The human transgenes in such heavy and kappa light chains.137 However, instability of the chro-
mice had undergone V HDJH and VkJk rearrangements with N mosome fragment containing the kappa light chain locus led to a
region diversification, i.e., additions or deletions of nucleotides at substantial reduction in the generation of hybridomas, a problem
the V H-D-JH and Vk -Jk junctions, class switch recombination and that was solved by crossing IgH transchromosomal mice with Igk
somatic hypermutation.131 Despite the limited repertoire present transgenic mice.138 It is interesting to note that of the six human
in both transgenic mouse strains, immunization led to the gen- mAbs on the market today, five are derived from humanized
eration of specific human antibodies against several antigens that mice (Table 1). Of these, panitumumab is derived from Abgenix’
were accessible by conventional hybridoma technology.130,131 In XenoMouse,135 whereas the other four are derived from Medarex’
this respect, transgenic mice have led to a revival of hybridoma UltiMab platform, which is based on the HuMab mouse,134 the
technology for the mining of human antibody repertoires. Kirin TC mouse137 and a cross of the two, the KM mouse.138
The finding that transgenic mice containing human heavy Natural antibody responses are polyclonal. There are cases
and light chain mini loci are capable of mounting a specific in which immunotherapy using polyclonal antibodies (pAbs)
humoral immune response comprising human antibodies is sur- instead of mAbs is expected to be more efficient. This includes,
prising in at least two respects. First, it demonstrates that hybrid for example, the treatment or prevention of diseases caused by
B-cell receptor complexes consisting of human surface Ig and different viral or bacterial strains or combating viruses prone to
mouse Iga/Igb (CD79a/CD79b) chains are functional in vivo formation of escape mutants. In principle, human pAbs could
and permit largely normal development and function of B cells. be produced in humanized mice.138 While mice are well suited
Second, only a fraction of the natural heavy and light chain V for the generation of mAbs, their small body size makes them
gene repertoire appears to be sufficient for the generation of unsuitable for the production of useful amounts of therapeutic
antigen-specific antibodies, putting the relative importance of or preventive pAbs. One method of accessing the potential of
combinatorial diversity into perspective. For the production of human pAbs is to use recombinant pAbs, i.e., defined cocktails
antibodies against protein antigens using hyperimmunization of mAbs.139 In addition, due to advances into the understand-
protocols, the two non-germline encoded sources of clonal diver- ing of humoral immune systems of large farm animals combined
sity, i.e., junctional diversity and somatic hypermutation, appear with significant technological advances, it may soon be possible
to be sufficient. Indeed, it was shown that the HCDR3 repertoire to generate human pAbs by hyperimmunization of transchromo-
is in large part created by junctional diversity, and sufficient for somal cattle.140
most antigen specificities.133 Synthetic repertoires. Antibody libraries from synthetic rep-
Despite the initial success of the transgenic mice carrying ertoires are based on synthetic DNA sequences designed to diver-
mini loci, human mAbs against few antigens were isolated from sify the antibody sequence.69,141 Depending on the design, human
these mice. The immune responses in these mice were inferior mAbs from synthetic repertoires may or may not be distinguish-
to those of wild type mice, with reduced numbers of mature B able from endogenous human mAbs. The first human synthetic
cells and lower levels of circulating Igs; however, incorporation repertoire was generated by randomizing all of the 16 amino
of a significantly larger repertoire of Vk gene segments led to acids that comprised the HCDR3 sequence of a human anti-
increased levels of pre-B and B cells.134 Mice that incorporated tetanus toxoid mAb.142 Subsequent selection by phage display
the majority of the human heavy chain and kappa light chain loci against fluorescein yielded a panel of human mAbs that revealed
were found to recapitulate the human antibody response even a shift in specificity from tetanus toxoid to fluorescein. In addi-
better by restoring B-cell compartments to near normal levels.135 tion, synthetic repertoires allow the CDR grafting of a peptide
Compared to the transgenic mice carrying mini loci,130,131 the motif with known specificity followed by randomization of the
animals with near complete loci displayed higher levels of human flanking sequences and selection by phage display. This method
serum Ig, better class switch recombination efficiency and yielded human mAbs that bind human integrins aVb3 and aIIbb3
increased clonal diversity and magnitude of the human antibody with high affinity.143,144 The randomization of single or multiple
response. Accordingly, a panel of human mAbs with high affinity CDR sequences followed by selection with phage display has also
against three human antigens was isolated.135 In contrast to mice, been utilized for the affinity maturation of human mAbs that
approximately 40% of serum Igs in humans contains a lambda were derived from immune or naïve repertoires. This process can
light chain. To further enhance the diversity of humanized B-cell yield human mAbs with picomolar affinity.145,146

www.landesbioscience.com mAbs 373


Antibody libraries that are based on human naïve repertoires Another exciting advance in the area of human synthetic rep-
but introduce additional sequence diversification through ran- ertoires has been the generation and selection of antibody librar-
domizing single or multiple CDR sequences represent, by defini- ies that feature an expanded genetic code for the incorporation
tion, human synthetic repertoires;147-150 however, not all synthetic of unnatural amino acids into CDR sequences.175 Collectively,
repertoires are based on randomized CDR sequences. Using mas- synthetic repertoires have begun to outshine all other human
ter framework sequences derived from a single human V H and Vl antibody repertoires with respect to diversity and versatility.
gene segment, Söderlind et al.151 introduced and recombined a Human mAbs from synthetic repertoires are poised to become
complete set of CDR sequences derived by RT-PCR from human preferred reagents for diagnostic and basic applications; however,
naïve repertoires. Yet another approach of sequence diversification their utility for preventive and therapeutic interventions remains
in vitro is based on dispersing mutations throughout heavy and to be established in clinical trials.
light chain variable domain sequences, as opposed to focusing
mutations on defined segments such as CDR sequences. This can Outlook
be achieved through error-prone PCR or mutagenic Escherichia
coli strains. Like focused mutagenesis, dispersed mutagenesis has Increased availability and improved accessibility of human anti-
been utilized for the affinity (or catalytic activity) maturation of body repertoires have made human mAbs the entity of choice for
human mAbs, usually by phage display152,153 and, more recently, therapeutic mAbs. Today, human mAbs to virtually any antigen
by yeast display.115,154 of interest can be generated. The clinical performance and the
Owing to the fact that the discussed antibody libraries blend commercial viability of currently approved human mAbs provide
in vivo and in vitro diversity, they are sometimes referred to as a robust platform for the development of future generations of
semi-synthetic. By contrast, fully synthetic antibody libraries human mAbs. Not only will these broaden the scope of targeted
are entirely designed and engineered in vitro. A fully synthetic antigens and indications, but also improve the activity of inter-
antibody library that combined a single human V H and Vk gene vention through better pharmacodynamic and pharmacokinetic
segment with randomized HCDR3 and LCDR3 sequences properties. Although an initial goal in the development of human
was mined successfully for human mAbs to human antigens.155 mAbs was to render them indistinguishable from endogenous
Further restrictions that mimic natural CDR sequence diversity human antibodies, subtle antibody engineering and expression
in the heavy chain variable domain and use a fixed light chain strategies that retain low immunogenicity, but tune affinity,
variable domain yielded human mAbs that closely resemble effector functions and circulatory half-life, as well as lower the
endogenous human antibodies. These were improved further by cost of goods, are being employed in the development of the next
introducing natural CDR sequence diversity in the light chain generation of human mAbs.176
variable domain.156,157 CDR sequence diversity can be further Of the currently approved human mAbs (Table 1), one was
restricted to a four-amino-acid code (Ala, Asp, Ser and Tyr)158 derived from a naïve repertoire and five were derived from trans-
or even a two-amino-acid code (Ser and Tyr),159 demonstrating genic repertoires. It will be interesting to compare human mAbs
the versatility of synthetic repertoires and providing molecular from different sources with respect to their pharmacokinetic and
insights into specificity and affinity of antibody/antigen interac- pharmacodynamic performance. For example, the human anti-
tions. Restricting sequence diversity in fully synthetic and semi- human TNFa mAb, adalimumab, which was derived indirectly
synthetic repertoires has also been used for tailoring antibody from a naïve repertoire as described above, induces a HAHA
libraries toward the selection of human mAbs to a particular response in a significant percentage of patients, adversely affect-
class of antigens, such as peptides and carbohydrates.160,161 Fully ing serum concentrations and clinical responses.177 The recent
synthetic antibody libraries that are more complex and based on approval of human anti-human TNFa mAb, golimumab,178
>1010 independent clones (in scFv or Fab format) reassemble a which was derived from a transgenic repertoire, provides a first
variety of natural CDR and framework sequences and combine opportunity to clinically investigate whether human mAbs
these with randomized CDR sequences.162,163 A fully synthetic derived from different human antibody repertoires against the
antibody library with diversification in all six CDR sequences same antigen differ in terms of immunogenicity.
was reported more recently.164 The immune repertoire is a natural repertoire that has not been
In addition to phage display, fully synthetic antibody libraries fully exploited for the mining of human mAbs. Sophisticated in
have been mined successfully with ribosome display.165 Ribosome vivo processes that shape the immune repertoire afford endog-
display is an in vitro display technology166 that does not require enous human antibodies with high affinity, minimal immunoge-
transformation of prokaryotic or eukaryotic cells.167-169 Ribosome nicity and minimal off-target reactivity. The immune repertoire
display has been employed to both accomplish and guide the has become increasingly accessible through new methods that
affinity maturation of human mAbs.170 Other less common dis- facilitate the enrichment, isolation or expansion of antigen-spe-
play technologies, such as retroviral display,171 have also been used cific post-GC B cells, plasma blasts, plasma cells or memory B
for mining synthetic repertoires. In addition to the conventional cells from peripheral blood. The development of these methods
scFv and Fab formats, synthetic repertoires have facilitated the has been spurred by the discovery of endogenous human antibod-
display and selection of single human variable domains 81,172 and, ies of potential therapeutic utility in normal human donors, as
recently, human constant domains with diversified sequences in well as in those with inflammatory diseases, infectious diseases,
non-CDR loops of the Ig fold.173,174 and cancer. Endogenous human antibodies that are triggered

374 mAbs Volume 2 Issue 4


by preventive or therapeutic interventions, such as vaccination factor in future antibody therapy. Mining strategies that yield a
or allogeneic hematopoietic stem cell transplantation, are par- panel of human mAbs to a variety of different epitopes of an anti-
ticularly attractive. An ideal mining strategy for this exceptional gen of interest are suitable for the development of human pAbs.
source of human mAbs is mammalian cell display as it provides Nonetheless, it remains possible that cocktails of human mAbs
an environment that closely resembles B cells. Consequently, this that are individual products of conventional mining strategies are
platform is anticipated to contribute to the development of future still inferior to truly polyclonal human antibodies that emerged
generations of human mAbs. as complex mixtures in vivo. Thus, evolving mining strategies
Finally, it is important to keep in mind that endogenous from monoclonal to polyclonal screening and selection offers a
human antibodies are polyclonal whereas human mAbs, by worthy challenge for antibody engineers.
definition, are monoclonal. In general, polyclonal or even oligo-
clonal responses are thought to be more proficient in mounting Acknowledgements
effector functions compared to monoclonal responses.179 While We thank Drs. Gregg J. Silverman (University of California,
non-recombinant human pAbs purified from human donors San Diego) and Brian C. Shaffer (National Institutes of Health)
have long been used in preventive and therapeutic interventions, for critically reading the manuscript. This work was supported
recombinant human pAbs would afford much broader applica- in part by the Intramural Research Program of the Center for
bility.139 Thus, despite current practical and regulatory challenges Cancer Research, National Cancer Institute, National Institutes
in manufacturing and formulation, human pAbs will likely be a of Health.
17. Vollmers HP, Brandlein S. Natural IgM antibodies: the 30. Ostberg L, Pursch E. Human X (mouse X human)
orphaned molecules in immune surveillance. Adv Drug hybridomas stably producing human antibodies.
Conflict-of-interest and financial disclosure Deliv Rev 2006; 58:755-65. Hybridoma 1983; 2:361-7.
statements 18. Lane HC, Shelhamer JH, Mostowski HS, Fauci AS. 31. Posner MR, Elboim H, Santos D. The construction
Human monoclonal anti-keyhole limpet hemocyanin and use of a human-mouse myeloma analogue suit-
R.R.B. is an employee of Cytos antibody-secreting hybridoma produced from periph- able for the routine production of hybridomas secret-
Biotechnology AG and holds stock options eral blood B lymphocytes of a keyhole limpet hemocy- ing human monoclonal antibodies. Hybridoma 1987;
anin-immune individual. J Exp Med 1982; 155:333-8. 6:611-25.
in the company. 19. Nowinski R, Berglund C, Lane J, Lostrom M, Bernstein 32. Teng NN, Lam KS, Calvo Riera F, Kaplan HS.
References I, Young W, et al. Human monoclonal antibody against Construction and testing of mouse—human heteromy-
1. Hwang WY, Foote J. Immunogenicity of engineered Forssman antigen. Science 1980; 210:537-9. elomas for human monoclonal antibody production.
antibodies. Methods 2005; 36:3-10. 20. Schlom J, Wunderlich D, Teramoto YA. Generation Proc Natl Acad Sci USA 1983; 80:7308-12.
2. Weiner LM. Fully human therapeutic monoclonal of human monoclonal antibodies reactive with human 33. Karpas A, Dremucheva A, Czepulkowski BH. A human
antibodies. J Immunother 2006; 29:1-9. mammary carcinoma cells. Proc Natl Acad Sci USA myeloma cell line suitable for the generation of human
3. Swann PG, Tolnay M, Muthukkumar S, Shapiro MA, 1980; 77:6841-5. monoclonal antibodies. Proc Natl Acad Sci USA 2001;
Rellahan BL, Clouse KA. Considerations for the devel- 21. Croce CM, Shander M, Martinis J, Cicurel L, 98:1799-804.
opment of therapeutic monoclonal antibodies. Curr D’Ancona GG, Koprowski H. Preferential retention 34. Dessain SK, Adekar SP, Stevens JB, Carpenter KA,
Opin Immunol 2008; 20:493-9. of human chromosome 14 in mouse X human B cell Skorski ML, Barnoski BL, et al. High efficiency
4. Weiner LM, Dhodapkar MV, Ferrone S. Monoclonal hybrids. Eur J Immunol 1980; 10:486-8. creation of human monoclonal antibody-producing
antibodies for cancer immunotherapy. Lancet 2009; 22. Yoshinari K, Arai K, Kimura H, Matsumoto K, hybridomas. J Immunol Methods 2004; 291:109-22.
373:1033-40. Yamaguchi Y. Long-term production of human mono- 35. Adekar SP, Jones RM, Elias MD, Al-Saleem FH,
5. Kohler G, Milstein C. Continuous cultures of fused clonal antibodies by human-mouse heterohybridomas. Root MJ, Simpson LL, et al. Hybridoma populations
cells secreting antibody of predefined specificity. Nature J Immunol Methods 1995; 186:17-25. enriched for affinity-matured human IgGs yield high-
1975; 256:495-7. 23. Chiorazzi N, Wasserman RL, Kunkel HG. Use of affinity antibodies specific for botulinum neurotoxins.
6. Steinitz M. Three decades of human monoclonal anti- Epstein-Barr virus-transformed B cell lines for the J Immunol Methods 2008; 333:156-66.
bodies: past, present and future developments. Hum generation of immunoglobulin-producing human B 36. Duchosal MA, Eming SA, Fischer P, Leturcq D, Barbas
Antibodies 2009; 18:1-10. cell hybridomas. J Exp Med 1982; 156:930-5. CF 3rd, McConahey PJ, et al. Immunization of hu-
7. Lonberg N. Fully human antibodies from transgen- 24. Cote RJ, Morrissey DM, Houghton AN, Beattie EJ Jr, PBL-SCID mice and the rescue of human monoclonal
ic mouse and phage display platforms. Curr Opin Oettgen HF, Old LJ. Generation of human monoclonal Fab fragments through combinatorial libraries. Nature
Immunol 2008; 20:450-9. antibodies reactive with cellular antigens. Proc Natl 1992; 355:258-62.
8. Hoogenboom HR. Selecting and screening recom- Acad Sci USA 1983; 80:2026-30. 37. Carlsson R, Martensson C, Kalliomaki S, Ohlin M,
binant antibody libraries. Nat Biotechnol 2005; 23: 25. Croce CM, Linnenbach A, Hall W, Steplewski Z, Borrebaeck CA. Human peripheral blood lymphocytes
1105-16. Koprowski H. Production of human hybridomas transplanted into SCID mice constitute an in vivo
9. Reichert JM. Monoclonal antibodies in the clinic. Nat secreting antibodies to measles virus. Nature 1980; culture system exhibiting several parameters found in a
Biotechnol 2001; 19:819-22. 288:488-9. normal humoral immune response and are a source of
26. Houghton AN, Brooks H, Cote RJ, Taormina MC, immunocytes for the production of human monoclonal
10. Dimitrov DS, Marks JD. Therapeutic antibodies:
Oettgen HF, Old LJ. Detection of cell surface and antibodies. J Immunol 1992; 148:1065-71.
current state and future trends—is a paradigm change
coming soon? Methods Mol Biol 2009; 525:1-27. intracellular antigens by human monoclonal antibod- 38. Eren R, Lubin I, Terkieltaub D, Ben-Moshe O,
ies. Hybrid cell lines derived from lymphocytes of Zauberman A, Uhlmann R, et al. Human monoclonal
11. Reichert JM. Antibodies to watch in 2010. MAbs
patients with malignant melanoma. J Exp Med 1983; antibodies specific to hepatitis B virus generated in a
2010; 2:84-100.
158:53-65. human/mouse radiation chimera: the Trimera system.
12. LeBien TW, Tedder TF. B lymphocytes: how they Immunology 1998; 93:154-61.
develop and function. Blood 2008; 112:1570-80. 27. Larrick JW, Truitt KE, Raubitschek AA, Senyk G,
Wang JC. Characterization of human hybridomas 39. Sawada-Hirai R, Jiang I, Wang F, Sun SM, Nedellec
13. Xu L, Yee H, Chan C, Kashyap AK, Horowitz L, R, Ruther P, et al. Human anti-anthrax protective
secreting antibody to tetanus toxoid. Proc Natl Acad
Horowitz M, et al. Combinatorial surrobody libraries. antigen neutralizing monoclonal antibodies derived
Sci USA 1983; 80:6376-80.
Proc Natl Acad Sci USA 2008; 105:10756-61. from donors vaccinated with anthrax vaccine adsorbed.
28. Olsson L, Kaplan HS. Human-human hybridomas
14. McHeyzer-Williams LJ, McHeyzer-Williams MG. J Immune Based Ther Vaccines 2004; 2:5.
producing monoclonal antibodies of predefined anti-
Antigen-specific memory B cell development. Annu 40. Nilsson K, Klein G, Henle W, Henle G. The establish-
genic specificity. Proc Natl Acad Sci USA 1980;
Rev Immunol 2005; 23:487-513. ment of lymphoblastoid lines from adult and fetal
77:5429-31.
15. Manz RA, Hauser AE, Hiepe F, Radbruch A. human lymphoid tissue and its dependence on EBV.
29. Cooper MD, Kirkpatrick R. Production of stable het-
Maintenance of serum antibody levels. Annu Rev Int J Cancer 1971; 8:443-50.
erohybridomas producing human monoclonal antibod-
Immunol 2005; 23:367-86.
ies. Methods Mol Biol 1995; 45:29-39.
16. Lanzavecchia A, Sallusto F. Human B cell memory.
Curr Opin Immunol 2009; 21:298-304.

www.landesbioscience.com mAbs 375


41. Kozbor D, Roder JC. Requirements for the establish- 59. Walker LM, Phogat SK, Chan-Hui PY, Wagner D, 79. Kang AS, Barbas CF, Janda KD, Benkovic SJ, Lerner
ment of high-titered human monoclonal antibodies Phung P, Goss JL, et al. Broad and potent neutralizing RA. Linkage of recognition and replication functions
against tetanus toxoid using the Epstein-Barr virus antibodies from an African donor reveal a new HIV-1 by assembling combinatorial antibody Fab libraries
technique. J Immunol 1981; 127:1275-80. vaccine target. Science 2009; 326:285-9. along phage surfaces. Proc Natl Acad Sci USA 1991;
42. Steinitz M, Klein G, Koskimies S, Makel O. EB virus- 60. Kwakkenbos MJ, Diehl SA, Yasuda E, Bakker AQ, van 88:4363-6.
induced B lymphocyte cell lines producing specific Geelen CM, Lukens MV, et al. Generation of stable 80. Hoogenboom HR, Griffiths AD, Johnson KS, Chiswell
antibody. Nature 1977; 269:420-2. monoclonal antibody-producing B cell receptor-posi- DJ, Hudson P, Winter G. Multi-subunit proteins on
43. Bron D, Feinberg MB, Teng NN, Kaplan HS. tive human memory B cells by genetic programming. the surface of filamentous phage: methodologies for
Production of human monoclonal IgG antibodies Nat Med 2010; 16:123-8. displaying antibody (Fab) heavy and light chains.
against Rhesus (D) antigen. Proc Natl Acad Sci USA 61. Mietzner B, Tsuiji M, Scheid J, Velinzon K, Tiller T, Nucleic Acids Res 1991; 19:4133-7.
1984; 81:3214-7. Abraham K, et al. Autoreactive IgG memory antibod- 81. Holt LJ, Herring C, Jespers LS, Woolven BP, Tomlinson
44. Kozbor D, Roder JC, Chang TH, Steplewski Z, ies in patients with systemic lupus erythematosus arise IM. Domain antibodies: proteins for therapy. Trends
Koprowski H. Human anti-tetanus toxoid monoclonal from nonreactive and polyreactive precursors. Proc Natl Biotechnol 2003; 21:484-90.
antibody secreted by EBV-transformed human B cells Acad Sci USA 2008; 105:9727-32. 82. Holliger P, Hudson PJ. Engineered antibody fragments
fused with murine myeloma. Hybridoma 1982; 1: 62. Tiller T, Tsuiji M, Yurasov S, Velinzon K, Nussenzweig and the rise of single domains. Nat Biotechnol 2005;
323-8. MC, Wardemann H. Autoreactivity in human IgG+ 23:1126-36.
45. Geylis V, Kourilov V, Meiner Z, Nennesmo I, memory B cells. Immunity 2007; 26:205-13. 83. O’Connell D, Becerril B, Roy-Burman A, Daws M,
Bogdanovic N, Steinitz M. Human monoclonal 63. Scheid JF, Mouquet H, Feldhahn N, Seaman MS, Marks JD. Phage versus phagemid libraries for gen-
antibodies against amyloid-beta from healthy adults. Velinzon K, Pietzsch J, et al. Broad diversity of neutral- eration of human monoclonal antibodies. J Mol Biol
Neurobiol Aging 2005; 26:597-606. izing antibodies isolated from memory B cells in HIV- 2002; 321:49-56.
46. Lang AB, Furer E, Senyk G, Larrick JW, Cryz SJ Jr. infected individuals. Nature 2009; 458:636-40. 84. Shi L, Wheeler JC, Sweet RW, Lu J, Luo J, Tornetta M,
Systematic generation of antigen specific human mono- 64. Wrammert J, Smith K, Miller J, Langley WA, Kokko K, et al. De novo selection of high-affinity antibodies from
clonal antibodies with therapeutical activities using Larsen C, et al. Rapid cloning of high-affinity human synthetic fab libraries displayed on phage as pIX fusion
active immunization. Hum Antibodies Hybridomas monoclonal antibodies against influenza virus. Nature proteins. J Mol Biol 2010.
1990; 1:96-103. 2008; 453:667-71. 85. Barbas CF 3rd, Kang AS, Lerner RA, Benkovic SJ.
47. Ohlin M, Broliden PA, Danielsson L, Wahren B, Rosen 65. Meijer PJ, Andersen PS, Haahr Hansen M, Steinaa L, Assembly of combinatorial antibody libraries on phage
J, Jondal M, et al. Human monoclonal antibodies Jensen A, Lantto J, et al. Isolation of human antibody surfaces: the gene III site. Proc Natl Acad Sci USA
against a recombinant HIV envelope antigen produced repertoires with preservation of the natural heavy and 1991; 88:7978-82.
by primary in vitro immunization. Characterization light chain pairing. J Mol Biol 2006; 358:764-72. 86. Burton DR, Barbas CF 3rd, Persson MA, Koenig S,
and epitope mapping. Immunology 1989; 68:325-31. 66. Poulsen TR, Meijer PJ, Jensen A, Nielsen LS, Andersen Chanock RM, Lerner RA. A large array of human
48. Yu X, McGraw PA, House FS, Crowe JE Jr. An PS. Kinetic, affinity and diversity limits of human monoclonal antibodies to type 1 human immunodefi-
optimized electrofusion-based protocol for generat- polyclonal antibody responses against tetanus toxoid. J ciency virus from combinatorial libraries of asymptom-
ing virus-specific human monoclonal antibodies. J Immunol 2007; 179:3841-50. atic seropositive individuals. Proc Natl Acad Sci USA
Immunol Methods 2008; 336:142-51. 67. Love JC, Ronan JL, Grotenbreg GM, van der Veen 1991; 88:10134-7.
49. Yu X, Tsibane T, McGraw PA, House FS, Keefer CJ, AG, Ploegh HL. A microengraving method for rapid 87. Zebedee SL, Barbas CF 3rd, Hom YL, Caothien RH,
Hicar MD, et al. Neutralizing antibodies derived from selection of single cells producing antigen-specific Graff R, DeGraw J, et al. Human combinatorial anti-
the B cells of 1918 influenza pandemic survivors. antibodies. Nat Biotechnol 2006; 24:703-7. body libraries to hepatitis B surface antigen. Proc Natl
Nature 2008; 455:532-6. 68. Jin A, Ozawa T, Tajiri K, Obata T, Kondo S, Kinoshita Acad Sci USA 1992; 89:3175-9.
50. Shammah S, Mantovani TL, Dalla-Favera R, Casali P. K, et al. A rapid and efficient single-cell manipulation 88. Barbas CF 3rd, Crowe JE Jr, Cababa D, Jones TM,
Generation of human monoclonal antibodies by trans- method for screening antigen-specific antibody-secret- Zebedee SL, Murphy BR, et al. Human monoclonal
formation of lymphoblastoid B cells with ras oncogene. ing cells from human peripheral blood. Nat Med 2009; Fab fragments derived from a combinatorial library
J Immunol Methods 1993; 160:19-25. 15:1088-92. bind to respiratory syncytial virus F glycoprotein and
51. Simmons CP, Bernasconi NL, Suguitan AL, Mills K, 69. Rader C. Antibody libraries in drug and target discov- neutralize infectivity. Proc Natl Acad Sci USA 1992;
Ward JM, Chau NV, et al. Prophylactic and therapeu- ery. Drug Discov Today 2001; 6:36-43. 89:10164-8.
tic efficacy of human monoclonal antibodies against 70. Glanville J, Zhai W, Berka J, Telman D, Huerta G, 89. Barbas CF 3rd, Collet TA, Amberg W, Roben P, Binley
H5N1 influenza. PLoS Med 2007; 4:178. Mehta GR, et al. Precise determination of the diversity JM, Hoekstra D, et al. Molecular profile of an antibody
52. Traggiai E, Becker S, Subbarao K, Kolesnikova L, of a combinatorial antibody library gives insight into response to HIV-1 as probed by combinatorial libraries.
Uematsu Y, Gismondo MR, et al. An efficient method the human immunoglobulin repertoire. Proc Natl Acad J Mol Biol 1993; 230:812-23.
to make human monoclonal antibodies from memory Sci USA 2009; 106:20216-21. 90. Williamson RA, Burioni R, Sanna PP, Partridge LJ,
B cells: potent neutralization of SARS coronavirus. Nat 71. Persson MA. Twenty years of combinatorial antibody Barbas CF 3rd, Burton DR. Human monoclonal anti-
Med 2004; 10:871-5. libraries, but how well do they mimic the immuno- bodies against a plethora of viral pathogens from single
53. Wen L, Hanvanich M, Werner-Favre C, Brouwers N, globulin repertoire? Proc Natl Acad Sci USA 2009; combinatorial libraries. Proc Natl Acad Sci USA 1993;
Perrin LH, Zubler RH. Limiting dilution assay for 106:20137-8. 90:4141-5.
human B cells based on their activation by mutant EL4 72. Huse WD, Sastry L, Iverson SA, Kang AS, Alting-Mees 91. Barbas SM, Ditzel HJ, Salonen EM, Yang WP,
thymoma cells: total and antimalaria responder B cell M, Burton DR, et al. Generation of a large combinato- Silverman GJ, Burton DR. Human autoantibody
frequencies. Eur J Immunol 1987; 17:887-92. rial library of the immunoglobulin repertoire in phage recognition of DNA. Proc Natl Acad Sci USA 1995;
54. Zubler RH, Erard F, Lees RK, Van Laer M, Mingari lambda. Science 1989; 246:1275-81. 92:2529-33.
C, Moretta L, et al. Mutant EL-4 thymoma cells poly- 73. Mullinax RL, Gross EA, Amberg JR, Hay BN, Hogrefe 92. Roben P, Barbas SM, Sandoval L, Lecerf JM, Stollar
clonally activate murine and human B cells via direct HH, Kubitz MM, et al. Identification of human anti- BD, Solomon A, et al. Repertoire cloning of lupus anti-
cell interaction. J Immunol 1985; 134:3662-8. body fragment clones specific for tetanus toxoid in a DNA autoantibodies. J Clin Invest 1996; 98:2827-37.
55. Banchereau J, de Paoli P, Valle A, Garcia E, Rousset bacteriophage lambda immunoexpression library. Proc 93. Siegel DL. Translational applications of antibody phage
F. Long-term human B cell lines dependent on inter- Natl Acad Sci USA 1990; 87:8095-9. display. Immunol Res 2008; 42:118-31.
leukin-4 and antibody to CD40. Science 1991; 251: 74. Persson MA, Caothien RH, Burton DR. Generation 94. Hansen MH, Nielsen H, Ditzel HJ. The tumor-
70-2. of diverse high-affinity human monoclonal antibodies infiltrating B cell response in medullary breast cancer is
56. Rousset F, Garcia E, Banchereau J. Cytokine-induced by repertoire cloning. Proc Natl Acad Sci USA 1991; oligoclonal and directed against the autoantigen actin
proliferation and immunoglobulin production of 88:2432-6. exposed on the surface of apoptotic cancer cells. Proc
human B lymphocytes triggered through their CD40 75. Smith GP. Filamentous fusion phage: novel expression Natl Acad Sci USA 2001; 98:12659-64.
antigen. J Exp Med 1991; 173:705-10. vectors that display cloned antigens on the virion sur- 95. Coronella JA, Spier C, Welch M, Trevor KT, Stopeck
57. Lagerkvist AC, Furebring C, Borrebaeck CA. Single, face. Science 1985; 228:1315-7. AT, Villar H, et al. Antigen-driven oligoclonal expan-
antigen-specific B cells used to generate Fab fragments 76. McCafferty J, Griffiths AD, Winter G, Chiswell DJ. sion of tumor-infiltrating B cells in infiltrating ductal
using CD40-mediated amplification or direct PCR Phage antibodies: filamentous phage displaying anti- carcinoma of the breast. J Immunol 2002; 169:
cloning. Biotechniques 1995; 18:862-9. body variable domains. Nature 1990; 348:552-4. 1829-36.
58. Weitkamp JH, Kallewaard N, Kusuhara K, Feigelstock 77. Clackson T, Hoogenboom HR, Griffiths AD, Winter 96. Pavoni E, Monteriu G, Santapaola D, Petronzelli F,
D, Feng N, Greenberg HB, et al. Generation of recom- G. Making antibody fragments using phage display Anastasi AM, Pelliccia A, et al. Tumor-infiltrating
binant human monoclonal antibodies to rotavirus libraries. Nature 1991; 352:624-8. B lymphocytes as an efficient source of highly spe-
from single antigen-specific B cells selected with fluo- 78. Breitling F, Dubel S, Seehaus T, Klewinghaus I, Little cific immunoglobulins recognizing tumor cells. BMC
rescent virus-like particles. J Immunol Methods 2003; M. A surface expression vector for antibody screening. Biotechnol 2007; 7:70.
275:223-37. Gene 1991; 104:147-53.

376 mAbs Volume 2 Issue 4


97. Cai X, Garen A. Anti-melanoma antibodies from mela- 115. van den Beucken T, Pieters H, Steukers M, van der 134. Fishwild DM, O’Donnell SL, Bengoechea T, Hudson
noma patients immunized with genetically modified Vaart M, Ladner RC, Hoogenboom HR, et al. Affinity DV, Harding F, Bernhard SL, et al. High-avidity
autologous tumor cells: selection of specific antibodies maturation of Fab antibody fragments by fluorescent- human IgGkappa monoclonal antibodies from a novel
from single-chain Fv fusion phage libraries. Proc Natl activated cell sorting of yeast-displayed libraries. FEBS strain of minilocus transgenic mice. Nat Biotechnol
Acad Sci USA 1995; 92:6537-41. Lett 2003; 546:288-94. 1996; 14:845-51.
98. Baskar S, Suschak JM, Samija I, Srinivasan R, Childs 116. Walker LM, Bowley DR, Burton DR. Efficient recov- 135. Mendez MJ, Green LL, Corvalan JR, Jia XC, Maynard-
RW, Pavletic SZ, et al. A human monoclonal antibody ery of high-affinity antibodies from a single-chain Fab Currie CE, Yang XD, et al. Functional transplant of
drug and target discovery platform for B-cell chronic yeast display library. J Mol Biol 2009; 389:365-75. megabase human immunoglobulin loci recapitulates
lymphocytic leukemia based on allogeneic hematopoi- 117. Feldhaus MJ, Siegel RW, Opresko LK, Coleman human antibody response in mice. Nat Genet 1997;
etic stem cell transplantation and phage display. Blood JR, Feldhaus JM, Yeung YA, et al. Flow-cytometric 15:146-56.
2009; 114:4494-502. isolation of human antibodies from a nonimmune 136. Nicholson IC, Zou X, Popov AV, Cook GP, Corps EM,
99. Marks JD, Hoogenboom HR, Bonnert TP, McCafferty Saccharomyces cerevisiae surface display library. Nat Humphries S, et al. Antibody repertoires of four- and
J, Griffiths AD, Winter G. By-passing immunization. Biotechnol 2003; 21:163-70. five-feature translocus mice carrying human immu-
Human antibodies from V-gene libraries displayed on 118. Blaise L, Wehnert A, Steukers MP, van den Beucken noglobulin heavy chain and kappa and lambda light
phage. J Mol Biol 1991; 222:581-97. T, Hoogenboom HR, Hufton SE. Construction and chain yeast artificial chromosomes. J Immunol 1999;
100. Griffiths AD, Malmqvist M, Marks JD, Bye JM, diversification of yeast cell surface displayed libraries by 163:6898-906.
Embleton MJ, McCafferty J, et al. Human anti-self yeast mating: application to the affinity maturation of 137. Tomizuka K, Shinohara T, Yoshida H, Uejima H,
antibodies with high specificity from phage display Fab antibody fragments. Gene 2004; 342:211-8. Ohguma A, Tanaka S, et al. Double trans-chromosomic
libraries. EMBO J 1993; 12:725-34. 119. Benatuil L, Perez JM, Belk J, Hsieh CM. An improved mice: maintenance of two individual human chromo-
101. Vaughan TJ, Williams AJ, Pritchard K, Osbourn JK, yeast transformation method for the generation of very some fragments containing Ig heavy and kappa loci and
Pope AR, Earnshaw JC, et al. Human antibodies with large human antibody libraries. Protein Eng Des Sel expression of fully human antibodies. Proc Natl Acad
sub-nanomolar affinities isolated from a large non- 2010; 23:155-9. Sci USA 2000; 97:722-7.
immunized phage display library. Nat Biotechnol 1996; 120. Weaver-Feldhaus JM, Lou J, Coleman JR, Siegel RW, 138. Ishida I, Tomizuka K, Yoshida H, Tahara T, Takahashi
14:309-14. Marks JD, Feldhaus MJ. Yeast mating for combinato- N, Ohguma A, et al. Production of human monoclonal
102. Sheets MD, Amersdorfer P, Finnern R, Sargent P, rial Fab library generation and surface display. FEBS and polyclonal antibodies in TransChromo animals.
Lindquist E, Schier R, et al. Efficient construction of a Lett 2004; 564:24-34. Cloning Stem Cells 2002; 4:91-102.
large nonimmune phage antibody library: the produc- 121. Lou J, Geren I, Garcia-Rodriguez C, Forsyth CM, Wen 139. Haurum JS. Recombinant polyclonal antibodies: the
tion of high-affinity human single-chain antibodies W, Knopp K, et al. Affinity maturation of human botu- next generation of antibody therapeutics? Drug Discov
to protein antigens. Proc Natl Acad Sci USA 1998; linum neurotoxin antibodies by light chain shuffling Today 2006; 11:655-60.
95:6157-62. via yeast mating. Protein Eng Des Sel 2010. 140. Kuroiwa Y, Kasinathan P, Sathiyaseelan T, Jiao JA,
103. de Haard HJ, van Neer N, Reurs A, Hufton SE, Roovers 122. Boder ET, Midelfort KS, Wittrup KD. Directed evolu- Matsushita H, Sathiyaseelan J, et al. Nat Biotechnol
RC, Henderikx P, et al. A large non-immunized human tion of antibody fragments with monovalent femtomo- 2009; 27:173-81.
Fab fragment phage library that permits rapid isolation lar antigen-binding affinity. Proc Natl Acad Sci USA 141. Sidhu SS, Fellouse FA. Synthetic therapeutic antibod-
and kinetic analysis of high affinity antibodies. J Biol 2000; 97:10701-5. ies. Nat Chem Biol 2006; 2:682-8.
Chem 1999; 274:18218-30. 123. Ho M, Nagata S, Pastan I. Isolation of anti-CD22 Fv 142. Barbas CF 3rd, Bain JD, Hoekstra DM, Lerner
104. Ling MM. Large antibody display libraries for isola- with high affinity by Fv display on human cells. Proc RA. Semisynthetic combinatorial antibody libraries: a
tion of high-affinity antibodies. Comb Chem High Natl Acad Sci USA 2006; 103:9637-42. chemical solution to the diversity problem. Proc Natl
Throughput Screen 2003; 6:421-32. 124. Smith ES, Shi S, Zauderer M. Construction of cDNA Acad Sci USA 1992; 89:4457-61.
105. Marks JD, Griffiths AD, Malmqvist M, Clackson TP, libraries in vaccinia virus. Methods Mol Biol 2004; 143. Barbas CF 3rd, Languino LR, Smith JW. High-affinity
Bye JM, Winter G. By-passing immunization: build- 269:65-76. self-reactive human antibodies by design and selection:
ing high affinity human antibodies by chain shuffling. 125. Beerli RR, Bauer M, Buser RB, Gwerder M, Muntwiler targeting the integrin ligand binding site. Proc Natl
Biotechnology 1992; 10:779-83. S, Maurer P, et al. Isolation of human monoclonal anti- Acad Sci USA 1993; 90:10003-7.
106. Kwong KY, Baskar S, Zhang H, Mackall CL, Rader C. bodies by mammalian cell display. Proc Natl Acad Sci 144. Chung J, Rader C, Popkov M, Hur YM, Kim HK,
Generation, affinity maturation and characterization of USA 2008; 105:14336-41. Lee YJ, et al. Integrin alphaIIbbeta3-specific synthetic
a human anti-human NKG2D monoclonal antibody 126. Jennings GT, Bachmann MF. The coming of age of human monoclonal antibodies and HCDR3 peptides
with dual antagonistic and agonistic activity. J Mol Biol virus-like particle vaccines. Biol Chem 2008; 389: that potently inhibit platelet aggregation. FASEB J
2008; 384:1143-56. 521-36. 2004; 18:361-3.
107. Jespers LS, Roberts A, Mahler SM, Winter G, 127. Beerli RR, Bauer M, Schmitz N, Buser RB, Gwerder 145. Yang WP, Green K, Pinz-Sweeney S, Briones AT,
Hoogenboom HR. Guiding the selection of human M, Muntwiler S, et al. Prophylactic and therapeutic Burton DR, Barbas CF 3rd. CDR walking mutagenesis
antibodies from phage display repertoires to a single activity of fully human monoclonal antibodies directed for the affinity maturation of a potent human anti-
epitope of an antigen. Biotechnology 1994; 12:899- against influenza A M2 protein. Virol J 2009; 6:224. HIV-1 antibody into the picomolar range. J Mol Biol
903. 128. Bruggemann M, Caskey HM, Teale C, Waldmann 1995; 254:392-403.
108. Osbourn J, Groves M, Vaughan T. From rodent H, Williams GT, Surani MA, et al. A repertoire 146. Schier R, McCall A, Adams GP, Marshall KW, Merritt
reagents to human therapeutics using antibody guided of monoclonal antibodies with human heavy chains H, Yim M, et al. Isolation of picomolar affinity anti-c-
selection. Methods 2005; 36:61-8. from transgenic mice. Proc Natl Acad Sci USA 1989; erbB-2 single-chain Fv by molecular evolution of the
109. Rader C, Cheresh DA, Barbas CF 3rd. A phage display 86:6709-13. complementarity determining regions in the center
approach for rapid antibody humanization: designed 129. Lonberg N. Human antibodies from transgenic ani- of the antibody binding site. J Mol Biol 1996; 263:
combinatorial V gene libraries. Proc Natl Acad Sci USA mals. Nat Biotechnol 2005; 23:1117-25. 551-67.
1998; 95:8910-5. 130. Green LL, Hardy MC, Maynard-Currie CE, Tsuda 147. Hoogenboom HR, Winter G. By-passing immunisa-
110. Chapal N, Peraldi-Roux S, Bresson D, Pugniere M, H, Louie DM, Mendez MJ, et al. Antigen-specific tion. Human antibodies from synthetic repertoires of
Mani JC, Granier C, et al. Human anti-thyroid peroxi- human monoclonal antibodies from mice engineered germline VH gene segments rearranged in vitro. J Mol
dase single-chain fragment variable of Ig isolated from with human Ig heavy and light chain YACs. Nat Genet Biol 1992; 227:381-8.
a combinatorial library assembled in-cell: insights into 1994; 7:13-21. 148. Griffiths AD, Williams SC, Hartley O, Tomlinson
the in vivo situation. J Immunol 2000; 164:4162-9. 131. Lonberg N, Taylor LD, Harding FA, Trounstine M, IM, Waterhouse P, Crosby WL, et al. Isolation of high
111. Boder ET, Wittrup KD. Yeast surface display for screen- Higgins KM, Schramm SR, et al. Antigen-specific affinity human antibodies directly from large synthetic
ing combinatorial polypeptide libraries. Nat Biotechnol human antibodies from mice comprising four distinct repertoires. EMBO J 1994; 13:3245-60.
1997; 15:553-7. genetic modifications. Nature 1994; 368:856-9. 149. de Kruif J, Boel E, Logtenberg T. Selection and applica-
112. Gai SA, Wittrup KD. Yeast surface display for protein 132. Alter-Wolf S, Blomberg BB, Riley RL. Old mice retain tion of human single chain Fv antibody fragments from
engineering and characterization. Curr Opin Struct bone marrow B1 progenitors, but lose B2 precursors, a semi-synthetic phage antibody display library with
Biol 2007; 17:467-73. and exhibit altered immature B cell phenotype and designed CDR3 regions. J Mol Biol 1995; 248:97-
113. Bowley DR, Labrijn AF, Zwick MB, Burton DR. light chain usage. Mech Ageing Dev 2009; 130:401-8. 105.
Antigen selection from an HIV-1 immune antibody 133. Xu JL, Davis MM. Diversity in the CDR3 region 150. Hoet RM, Cohen EH, Kent RB, Rookey K,
library displayed on yeast yields many novel antibodies of V(H) is sufficient for most antibody specificities. Schoonbroodt S, Hogan S, et al. Generation of high-
compared to selection from the same library displayed Immunity 2000; 13:37-45. affinity human antibodies by combining donor-derived
on phage. Protein Eng Des Sel 2007; 20:81-90. and synthetic complementarity-determining-region
114. Feldhaus MJ, Siegel RW. Yeast display of antibody diversity. Nat Biotechnol 2005; 23:344-8.
fragments: a discovery and characterization platform. J
Immunol Methods 2004; 290:69-80.

www.landesbioscience.com mAbs 377


151. Soderlind E, Strandberg L, Jirholt P, Kobayashi N, 161. Schoonbroodt S, Steukers M, Viswanathan M, Frans 170. Thom G, Cockroft AC, Buchanan AG, Candotti CJ,
Alexeiva V, Aberg AM, et al. Recombining germline- N, Timmermans M, Wehnert A, et al. Engineering Cohen ES, Lowne D, et al. Probing a protein-protein
derived CDR sequences for creating diverse single- antibody heavy chain CDR3 to create a phage display interaction by in vitro evolution. Proc Natl Acad Sci
framework antibody libraries. Nat Biotechnol 2000; Fab library rich in antibodies that bind charged carbo- USA 2006; 103:7619-24.
18:852-6. hydrates. J Immunol 2008; 181:6213-21. 171. Urban JH, Schneider RM, Compte M, Finger C,
152. Low NM, Holliger PH, Winter G. Mimicking somatic 162. Knappik A, Ge L, Honegger A, Pack P, Fischer M, Cichutek K, Alvarez-Vallina L, et al. Selection of
hypermutation: affinity maturation of antibodies dis- Wellnhofer G, et al. Fully synthetic human combina- functional human antibodies from retroviral display
played on bacteriophage using a bacterial mutator torial antibody libraries (HuCAL) based on modular libraries. Nucleic Acids Res 2005; 33:35.
strain. J Mol Biol 1996; 260:359-68. consensus frameworks and CDRs randomized with 172. Chen W, Zhu Z, Feng Y, Xiao X, Dimitrov DS.
153. Cesaro-Tadic S, Lagos D, Honegger A, Rickard JH, trinucleotides. J Mol Biol 2000; 296:57-86. Construction of a large phage-displayed human anti-
Partridge LJ, Blackburn GM, et al. Turnover-based 163. Rauchenberger R, Borges E, Thomassen-Wolf E, Rom body domain library with a scaffold based on a newly
in vitro selection and evolution of biocatalysts from a E, Adar R, Yaniv Y, et al. Human combinatorial Fab identified highly soluble, stable heavy chain variable
fully synthetic antibody library. Nat Biotechnol 2003; library yielding specific and functional antibodies domain. J Mol Biol 2008; 382:779-89.
21:679-85. against the human fibroblast growth factor receptor 3. 173. Xiao X, Feng Y, Vu BK, Ishima R, Dimitrov DS. A
154. Zhou Y, Drummond DC, Zou H, Hayes ME, Adams J Biol Chem 2003; 278:38194-205. large library based on a novel (CH2) scaffold: iden-
GP, Kirpotin DB, et al. Impact of single-chain Fv 164. Rothe C, Urlinger S, Lohning C, Prassler J, Stark Y, tification of HIV-1 inhibitors. Biochem Biophys Res
antibody fragment affinity on nanoparticle targeting Jager U, et al. The human combinatorial antibody Commun 2009; 387:387-92.
of epidermal growth factor receptor-expressing tumor library HuCAL GOLD combines diversification of all 174. Wozniak-Knopp G, Bartl S, Bauer A, Mostageer M,
cells. J Mol Biol 2007; 371:934-47. six CDRs according to the natural immune system with Woisetschlager M, Antes B, et al. Introducing antigen-
155. Pini A, Viti F, Santucci A, Carnemolla B, Zardi L, a novel display method for efficient selection of high- binding sites in structural loops of immunoglobu-
Neri P, et al. Design and use of a phage display library. affinity antibodies. J Mol Biol 2008; 376:1182-200. lin constant domains: Fc fragments with engineered
Human antibodies with subnanomolar affinity against 165. Hanes J, Schaffitzel C, Knappik A, Pluckthun A. HER2/neu-binding sites and antibody properties.
a marker of angiogenesis eluted from a two-dimension- Picomolar affinity antibodies from a fully synthetic Protein Eng Des Sel 2010.
al gel. J Biol Chem 1998; 273:21769-76. naive library selected and evolved by ribosome display. 175. Liu CC, Mack AV, Tsao ML, Mills JH, Lee HS, Choe
156. Sidhu SS, Li B, Chen Y, Fellouse FA, Eigenbrot C, Nat Biotechnol 2000; 18:1287-92. H, et al. Protein evolution with an expanded genetic
Fuh G. Phage-displayed antibody libraries of synthetic 166. Rothe A, Hosse RJ, Power BE. In vitro display tech- code. Proc Natl Acad Sci USA 2008; 105:17688-93.
heavy chain complementarity determining regions. J nologies reveal novel biopharmaceutics. FASEB J 2006; 176. Carter PJ. Potent antibody therapeutics by design. Nat
Mol Biol 2004; 338:299-310. 20:1599-610. Rev Immunol 2006; 6:343-57.
157. Lee CV, Liang WC, Dennis MS, Eigenbrot C, Sidhu 167. Hanes J, Pluckthun A. In vitro selection and evolution 177. Aarden L, Ruuls SR, Wolbink G. Immunogenicity of
SS, Fuh G. High-affinity human antibodies from of functional proteins by using ribosome display. Proc anti-tumor necrosis factor antibodies-toward improved
phage-displayed synthetic Fab libraries with a single Natl Acad Sci USA 1997; 94:4937-42. methods of anti-antibody measurement. Curr Opin
framework scaffold. J Mol Biol 2004; 340:1073-93. 168. He M, Taussig MJ. Antibody-ribosome-mRNA (ARM) Immunol 2008; 20:431-5.
158. Fellouse FA, Wiesmann C, Sidhu SS. Synthetic anti- complexes as efficient selection particles for in vitro 178. Mazumdar S, Greenwald D. Golimumab. MAbs 2009;
bodies from a four-amino-acid code: a dominant role display and evolution of antibody combining sites. 1:422-31.
for tyrosine in antigen recognition. Proc Natl Acad Sci Nucleic Acids Res 1997; 25:5132-4. 179. Logtenberg T. Antibody cocktails: next-generation
USA 2004; 101:12467-72. 169. Schaffitzel C, Hanes J, Jermutus L, Pluckthun A. biopharmaceuticals with improved potency. Trends
159. Fellouse FA, Li B, Compaan DM, Peden AA, Hymowitz Ribosome display: an in vitro method for selection Biotechnol 2007; 25:390-4.
SG, Sidhu SS. Molecular recognition by a binary code. and evolution of antibodies from libraries. J Immunol
J Mol Biol 2005; 348:1153-62. Methods 1999; 231:119-35.
160. Cobaugh CW, Almagro JC, Pogson M, Iverson B,
Georgiou G. Synthetic antibody libraries focused
towards peptide ligands. J Mol Biol 2008; 378:622-33.

378 mAbs Volume 2 Issue 4

You might also like