You are on page 1of 10

REVIEWS

Building better monoclonal


antibody-based therapeutics
George J. Weiner
Abstract | For 20 years, monoclonal antibodies (mAbs) have been a standard component of
cancer therapy, but there is still much room for improvement. Efforts continue to build
better cancer therapeutics based on mAbs. Anticancer mAbs function through various
mechanisms, including directly targeting the malignant cells, modifying the host response,
delivering cytotoxic moieties and retargeting cellular immunity towards the malignant cells.
Characteristics of mAbs that affect their efficacy include antigen specificity, overall structure,
affinity for the target antigen and how a mAb component is incorporated into a construct
that can trigger target cell death. This Review discusses the various approaches to using
mAb-based therapeutics to treat cancer and the strategies used to take advantage of the
unique potential of each approach, and provides examples of current mAb-based treatments.

Cluster of differentiation
More than two centuries have passed since we began to their clinical utility. These problems included the develop­
numbers understand the remarkable characteristics and potential ment of an immune response against the therapeutic
Numbers assigned to cell of antibodies1. Indeed, polyclonal antisera have been used mAb itself, the rapid clearance of the mAb and the sub-
surface molecules on the basis in the treatment of certain infectious diseases for decades2. optimal ability of the murine mAb to interact with the
of immunophenotyping. They
Soon after the first description of monoclonal anti- human immune system in a manner that led to immune
are often used to identify
different monoclonal
bodies (mAbs) in 1975 (REF. 3) (a discovery that led to destruction of the cancer.
antibodies that bind to the a Nobel Prize 10 years later 4), mAbs were recognized Fortunately, persistent investigators continued to
same antigen. as unique biological tools and quickly became invalu- explore how mAbs could be used in cancer treatment.
able in pathological diagnosis and basic laboratory They evaluated various strategies, including using
investigation. Their ability to bind to specific antigenic immunoglobulin G (IgG) to target cancer directly, alter
epitopes allowed for the rapid assessment of the molecu- the host response to cancer, deliver cytotoxic substances
lar pheno­type of blood cells and, subsequently, of other to cancer and retarget the cellular immune response
tissues. Molecules that were identified by mAb binding towards cancer (TABLE 1).
were given cluster of differentiation numbers5 that are still The current era of successful mAb therapy began
used extensively in diagnosis. mAbs are now used widely with the development of techniques that allowed genetic
in immunohistochemistry, flow cytometry and related modification of murine mAbs to produce chimeric
technologies. Aside from the use of mAbs as tools in mouse–human mAbs that behave immunologically in
diagnosis, at the time they were first described there was most ways like naturally occurring human IgG8,9. Such
equal excitement about their therapeutic potential based mAbs are less likely to be recognized by the host immune
on the ability to manufacture mAbs of defined specificity system as a foreign antigen, have half-lives similar to
and class in essentially unlimited amounts. Theoretically, those of natural human IgG (generally 2–4 weeks) and
this would allow for highly specific targeting of cancer interact well with the effector arm of the human immune
cells on the basis of their molecular phenotype. system. They can be administered on a schedule that is
However, early clinical results exploring mAb-based practical for patients (in many cases weekly or monthly),
Holden Comprehensive
Cancer Center, University of therapeutics were disappointing 6, and until only 20 years and they are present in the circulation of patients at
Iowa Hospitals and Clinics, ago, some experts considered cancer treatment with therapeutic levels for months at a time. They distribute
200 Hawkins Drive, antibody-­based therapy a failed hypothesis. The first to both the intravascular and the extravascular com-
5970Z‑JPP, Iowa City, mAbs evaluated in the clinic as cancer treatments were partments and are present within the tumour mass for
Iowa 52242, USA.
e-mail:
murine mAbs. Although there were intriguing hints that long periods of time where they interact with malignant
george-weiner@uiowa.edu mAb therapy could be successful7, problems associated cells, stromal cells, benign lymphocytes, the extracellular
doi:10.1038/nrc3930 with administering murine mAbs to humans limited matrix and the vasculature.

NATURE REVIEWS | CANCER VOLUME 15 | JUNE 2015 | 361

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Table 1 | Monoclonal antibody-based therapeutics


mAb-based Structure Characteristics of target Example of major ongoing research
therapeutic antigen questions
Antitumour mAbs Unmodified IgG or IgG modified to Tumour-associated surface Are IgGs with enhanced affinity for Fc receptors
mediate enhanced ADCC antigen more clinically effective than unaltered IgG?
Angiogenesis Unmodified IgG Host molecules that control What is the best way to evaluate clinical
inhibition angiogenesis response in patients treated with angiogenesis
inhibitors?
T cell checkpoint IgG1 (blocks checkpoint and Molecules that limit the How should we combine checkpoint
blockade mediates ADCC) or IgG4 (blocks anticancer T cell response blockade mAbs with each other, with other
checkpoint without mediating immunotherapeutics and with other anticancer
extensive ADCC) agents?
Radioimmunotherapy Unmodified IgG or mAb fragment Tumour-associated antigen that How can the logistics of administering
is not shed or present in the successful radioimmunotherapeutic agents be
circulation simplified to enhance their clinical utility?
Antibody–drug IgG modified with cleavable linker Highly specific tumour-associated What is the best combination of linkers and
conjugate and drug antigen that can internalize when drugs with each mAb and target antigen?
bound by a mAb
Bispecific antibody Variable regions from cancer-specific Tumour-associated antigen that is Can effective bispecific constructs that have
mAbs linked to variable regions not commonly absent in antigen- modified kinetics (thereby avoiding the logistic
specific for activating receptors on loss-resistant cancer variants complexities of continuous infusion) be
T cells developed?
Chimeric antigen Gene therapy approach to modifying Highly tumour-specific antigen Can very promising preliminary results be
receptor T cell T cells by inserting DNA coding for that is not commonly absent in extended to solid tumours, or will toxicity
the mAb variable region fused to DNA antigen-loss-resistant cancer be associated even with low levels of target
coding for signalling peptides variants antigen expression by benign cells?
ADCC, antibody-dependent cellular cytotoxicity; IgG, immunoglobulin G; mAb, monoclonal antibody.

After many years of research, a variety of mAb-based target cells by complement-mediated cytotoxicity (CMC)12
approaches to cancer therapy are being used with con- and by inducing antibody-dependent cellular cytotoxicity
siderable success, and progress using mAb therapeutics (ADCC)13. Determining which of these mechanisms
that are based on various different strategies (FIG. 1) is (FIG. 2) is most important for a given mAb in a given
Transmembrane signalling continuing at a remarkable pace. Oncologists now see clinical scenario remains a challenge.
The process by which an mAb-based cancer therapies as a vital component of
extracellular signal, mediated state‑of‑the-art cancer care, and even better mAb-based mAb-mediated cell signalling. Some mAbs can induce the
by a natural ligand or an
treatments are on the horizon. death of malignant cells in vitro, and presumably in vivo, in
alternative agent such as a
monoclonal antibody, binds to This Review provides an overview of recent pro- the absence of immune effector mechanisms. The strength
a membrane receptor and gress in the development and application of mAb-based of this effect varies considerably depending on the mAb,
generates an intracellular approaches to cancer therapy, including a description of the target antigen and the target cell14,15. Assessment of
signal that can affect a broad mAbs and mAb-based constructs that directly target the whether a given mAb can mediate signalling-­induced
range of cellular functions,
including cell growth, cell
cancer, alter the host response to the cancer, deliver cyto- death in the target cell in  vitro sometimes requires
differentiation and cell death. toxic moieties to the cancer or redirect T cells towards crosslinking with secondary antibodies, such as those
the cancer. specific to human IgG16. Secondary crosslinking results
Complement-mediated in enhanced aggregation of receptors that can lead to a
cytotoxicity
Targeting the cancer cell more robust signal. This is not necessarily an artificial way
(CMC). Also known as
complement-­dependent mAbs have been used to target a wide variety of antigens of replicating the crosslinking and signalling observed
cytotoxicity. Cell death expressed on the surface of cancer cells10. Characteristics in vivo, in which crosslinking of mAbs bound to malig-
resulting from the activation of that make antigens attractive as targets for mAb therapy nant cells probably occurs to a certain extent through the
the complement cascade by a include the density and consistency of expression of the binding of the constant region of IgG to Fc receptors (FcRs)
monoclonal antibody that
leads to the formation of a
target molecule by malignant cells, limited expression of expressed by a variety of cells in the tumour microenvi-
membrane attack complex on the target molecule on physiologically vital benign cells, ronment. It is not clear whether artificial crosslinking of
the surface of the cell. lack of high levels of soluble target and limited tendency mAbs speeds up and strengthens the physiological effect
of antigen-negative tumour variants to emerge. Desirable of mAb signalling so that it can be measured in a quick
Antibody-dependent
characteristics of target tumour antigens vary depending laboratory assay, or by contrast, whether it provides such
cellular cytotoxicity
(ADCC). Lysis of a target cell by on the mAb construct being considered, the nature of the a strong signal that non-physiological changes are induced
an immune effector cell (such malignancy (for example, haematological versus solid that may not be clinically relevant.
as a natural killer cell, tumours) and the mechanism of action of such constructs. In vitro measurement of signalling needs to be inter-
monocyte, macrophage or In vitro and animal model data indicate that some preted with caution as it varies greatly from the clinical
granulocyte) induced by the
recognition of an antibody
mAbs that target antigens on the surface of malignant environment17. In vitro studies usually use cells that have
bound to the surface of the cells are able to induce apoptosis by direct transmem- been selected to grow rapidly and consistently outside the
target cell. brane signalling11. There is also evidence that mAbs kill body — that is, independent of their normal environment.

362 | JUNE 2015 | VOLUME 15 www.nature.com/reviews/cancer

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

a Tumour-specific IgG b Angiogenesis inhibition c Checkpoint blockade the mAb results from the mAb interrupting the inter­
Tumour action between an activating ligand and the receptor,
cell
T cell
from inhibiting the dimerization of the receptor, or from
Receptor
having a direct effect on receptor signalling 19 (FIG. 2b).
Tumour PDL1 This complexity has been most extensively studied with
antigen VEGFR
the ERBB tyrosine kinase family of receptors20 and their
VEGF CTLA4 PD1
Complement PDL1- various ligands, and with mAbs such as trastuzumab
Effector cell specific and pertuzumab that recognize various receptor family
Ipilimumab Nivolumab members21. Individual receptors in this family can have
IgG multiple ligands, mAbs can alter dimerization proper-
ties and a mAb can have different signalling properties
d Radioimmunotherapy g CAR T cells depending on whether it is targeting a homodimeric or a
heterodimeric receptor 22. Understanding this complexity
is not just an academic exercise, as it can have a major
impact on the development and clinical testing of novel
Immunoconjugates Antigen-based therapeutics, including mAb combinations23. Resistance
retargeting of
cellular immunity to small molecules that inhibit signalling pathways can
develop with the emergence of cells that activate alterna-
tive or compensatory signalling pathways24. Similar pro-
e Antibody–drug f Bispecific cesses can lead to the emergence of resistance to mAbs
conjugate antibody
therapy therapy that mediate their therapeutic effects by signalling 25. As
CD3 with small molecules, the use of combination therapy is
one strategy to overcome this mechanism of resistance.
Figure 1 | Monoclonal antibody-based cancer therapeutic strategies.  Successful
monoclonal antibody (mAb) therapeutics have been based on a number of strategies.
Nature Reviews | Cancer Complement. The first studies demonstrating the very
Immunoglobulin G (IgG) molecules that bind to target cancer cells (part a) can mediate potent ability of complement fixation to enhance antibody-
antibody-dependent cellular cytotoxicity (ADCC) by immune effector cells, induce
mediated cytolysis were carried out more than a cen-
complement-­mediated cytotoxicity (CMC) or result in the direct signalling-induced
death of cancer cells (for example, herceptin and rituximab). IgG mAbs can also be used
tury ago. They involved the evaluation of the effect of
to inhibit angiogenesis (part b) (for example, bevacizumab) or to block inhibitory signals cobra venom on the lysis of red cells and bacteria26, and
(part c), thereby resulting in a stronger antitumour T cell response (for example, took place when our understanding of both antibodies
ipilimumab and nivolumab). Radioimmunoconjugates (part d) (for example, 131I and complement was obviously fairly limited. We have
tositumomab and ibritumomab tiuxetan) deliver radioisotopes to the cancer cells, since learned an amazing amount about complement in
whereas antibody–drug conjugates (part e) (for example, brentuximab vedotin and general and about how complement can have an impact
trastuzumab emtansine) deliver highly potent toxic drugs to the cancer cells. mAb on the efficacy of mAbs in particular12. We now know
variable regions are also used to retarget immune effector cells towards cancer cells that some (but not all) mAbs can mediate CMC27. The
through the use of bispecific mAbs that recognize cancer cells with one arm and ability of a given mAb to fix complement and to induce
activating antigens on immune effector cells with the other arm (part f) (for example,
CMC is partly dependent on antigen concentration, the
blinatumomab) or through a gene therapy approach in which DNA for a mAb variable
region fused to signalling peptides is transferred to T cells, thereby rendering them
orientation of the antigen in the membrane, and whether
chimeric antigen receptor (CAR) T cells (part g) specific for the tumour. CD3, T cell the antigen is present on the surface as a monomer or a
surface glycoprotein CD3 ε-chain; CTLA4, cytotoxic T lymphocyte-associated antigen 4; polymer. CMC can also depend on the mAb isotype and
PD1, programmed cell death protein 1; PDL1, PD1 ligand; VEGF, vascular endothelial the characteristics of the target cell, including whether
growth factor; VEGFR, VEGF receptor. the cell expresses complement-neutralizing molecules.
Even mAbs that are the same isotype and that target
the same antigen can vary in their ability to fix comple-
This can affect cell sensitivity to a wide variety of sig- ment 28. Type I human IgG1 CD20-specific mAbs (for
nals, and studying primary cells obtained directly from example, rituximab) crosslink CD20 tetramers and also
patients can avoid this problem. However, the evaluation fix complement. By contrast, type II IgG1 CD20‑specific
of primary cells requires extensive previous manipulation, mAbs (such as obinutuzumab) do not crosslink tetram-
Fc receptors including mincing, filtering and washing. This obliterates ers or fix complement well29. Antibody engineering that
(FcRs). A family of protein
receptors specific for an
the normal architecture of the microenvironment, alters alters either the constant region protein sequence or the
epitope on the constant region cell growth properties and often leads to the activation glycosylation of the mAb, gives developers control over
of an antibody. When FcRs on of apoptotic signalling pathways independently of addi- a number of characteristics, including the ability to fix
immune effector cells come tional therapy. In vitro signalling assays are usually carried complement 30,31 (FIG. 3).
into contact with an
out over a timescale of minutes to hours, whereas clinical Many studies exploring CMC use serum as a source
antibody-coated target cell,
this can result in immune responses to mAbs are measured over weeks and months. of complement, which highlights the importance of
effector cell activation (FcRs Despite these limitations, the detection of signalling- CMC in the circulation. Indeed, CMC seems to contri­
with immunoreceptor induced apoptosis has become a common approach to the bute most to the therapeutic effect of mAb in haemato-
tyrosine-based activation in vitro analysis of potential signalling effects of mAbs18. logical malignancies, in which target cells are exposed to
motifs (ITAMs)) or inhibition
(FcRs with immunoreceptor
Even when a mAb is known to alter signalling proper- complement in the circulation32. However, complement
tyrosine-based inhibitory ties and is effective clinically, it is difficult to use in vitro binding to target cells does not necessarily result in the
motifs (ITIMs)). assays to determine whether the therapeutic effect of lysis of the target cell. When a CD20‑specific mAb binds

NATURE REVIEWS | CANCER VOLUME 15 | JUNE 2015 | 363

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

a Immune-mediated effects of tumour-specific IgG FcRs that then signal through ITAMs and induce effec-
tor cell activation13. Other cells express immuno­receptor
tyrosine-based inhibitory motifs (ITIMs), which can
NK cell Complement inhibit ADCC39. mAb-coated target cells can induce
ADCC
ITAM the production and release of cytokines by immune
CMC
effector cells that express FcRs40. These cytokines can
Tumour cell then activate other immune effector cells in the tumour
Tumour Lysis
antigen microenvironment 41. Thus, immune cell activation via
MAC
FcRs can contribute to direct ADCC, as well as to the
production of cytokines that contribute to the control
FcR of tumour growth in other ways.
Opsonization
Granulocyte Monocyte Interacting mechanisms of mAb-induced cytotoxicity.
There is growing evidence that there are extensive
b Direct effects of tumour-specific IgG
interactions — both synergistic and antagonistic
Inhibit receptor — between various mechanisms of action that can
dimerization
affect the antitumour effects of a single type of mAb.
Block ligand A mAb may be developed with one mechanism in
Induce apoptotic mind, but other mechanisms may also be important 42.
signalling Complement fixation has very complex effects43. On
the one hand, the CD20‑specific mAbs rituximab and
ofatumumab, and the CD52‑specific mAb alemtu-
Figure 2 | Mechanisms of action of monoclonal antibodies that target cancer
cells.  Monoclonal antibodies (mAbs) that bind directly to cancer cells can mediate zumab, rapidly kill target cells in vitro via CMC; on the
Nature Reviews
their antitumour effects through various mechanisms. These mechanisms | Cancer
are routinely other hand, complement fixation can block the inter-
identified in vitro but their relative contributions to clinical responses to mAb therapy action between mAbs and activating FcRs on NK cells,
is difficult to determine. a | mAbs can mediate antibody-dependent cellular and reduce ADCC as a consequence44. Thus, in some
cytotoxicity (ADCC) by immune effector cells that express immunoreceptor circumstances, complement fixation may induce CMC
tyrosine-based activation motifs (ITAMs), such as natural killer (NK) cells, monocytes, and enhance the response to mAbs, but in others it
macrophages and granulocytes. Fixation of complement can trigger the opsonization could inhibit ADCC, thereby blocking the response to
of the target cell and thereby enhance phagocytosis and lysis by monocytes and mAbs. Signalling that results from a mAb binding to a
granulocytes. Complement-mediated cytotoxicity (CMC) can directly result in target receptor on a cancer cell can alter the sensitivity of that
cell death through the development of a membrane attack complex (MAC). b | mAbs
target cell to ADCC11. mAb-induced cancer cell lysis
can also have direct effects on target cells by blocking the binding of an activating
ligand that is responsible for the survival of the cancer cell, inhibiting the dimerization can, at least in preclinical models, lead to enhanced
of a receptor, thereby blocking an activation signal or inducing an apoptotic signal by uptake of the target antigen and subsequent cross-
crosslinking a receptor. Such crosslinking of the receptor can be enhanced when a presentation by cells — such as dendritic cells —
mAb is bound to Fc receptor-expressing cells. IgG, immunoglobulin G. thereby leading to an enhanced T cell response 45.
Malignant B cells that express FcRs without activat-
ing motifs (that is, FcRs without ITAMs) may actu-
to a circulating chronic lymphocytic leukaemia (CLL) ally positively contribute to the antitumour effect of
cell and fixes complement, the entire antigen–mAb– a mAb. These FcRs can allow for autocrine crosslink-
complement complex may be sheared off the surface of ing of B cell-specific mAbs that bind both to target
the leukaemic cell as it circulates through the liver and antigens, through the variable region, and to the
spleen in a process known as ‘shaving’, or trogocytosis, FcRs expressed by the same cell or by neighbouring
which is mediated by FcR-expressing cells including malignant cells46.
Complement fixation
monocytes33. This results in circulating malignant cells An antigen that can be effectively targeted by a
Initiation of the complement
cascade by an antibody bound that temporarily lack the target antigen and therefore cancer-­specific mAb needs to be found on the surface
to an antigen. It can lead to evade mAbs specific for that antigen. The relative con- of the cancer cell in concentrations high enough to
complement-mediated centration of various complement components in the trigger one or more of the effector mechanisms out-
cytotoxicity of the target cell, extravascular fluid is not well understood and may be lined above once the mAb has bound to the surface.
as well as other complex
effects mediated by the
inadequate to mediate CMC34. It is generally accepted However, the antigen should not be expressed to a
activation of various that CMC has a limited role in the efficacy of mAbs that similar degree by a vital population of benign cells47 or
complement components. recognize target antigens on malignancies outside the found in high concentrations as a soluble antigen in the
vascular compartment: that is, solid tumours. circulation48. Efforts to identify mAbs that recognize
mAb isotype
novel tumour-associated antigens continue, although
The subtype of a monoclonal
antibody (mAb) based on the ADCC. mAbs can induce ADCC by binding to FcRs, it could be argued that most of the highly promising
amino acid sequence of the which are expressed by a variety of immune effector antigens that can be effectively targeted by unmodified
constant region. Isotypes cells, including natural killer (NK) cells, granulocytes, IgG have already been identified. Ongoing efforts to
(IgG1, IgG2, IgG3 and IgG4) monocytes and macrophages 35–38 (FIG.  2) . Some of enhance the efficacy of IgGs that directly target can-
vary in their ability to mediate
antibody-dependent cellular
these effector cells express FcRs with immunoreceptor cer cells involve the identification of mAbs with unique
cytotoxicity and complement- tyrosine-based activation motifs (ITAMs). To trigger signalling properties — such as HER2 (also known
mediated cytotoxicity. ADCC, mAbs bound to the target cell interact with as ERBB2) mAbs that vary in their ability to interfere

364 | JUNE 2015 | VOLUME 15 www.nature.com/reviews/cancer

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Glycosylation with receptor heterodimerization49 — or the modifi- T cell receptor-like mAbs. Many tumour-associated
The post-translational cation of the IgG constant region to enhance its abil- antigens are not expressed on the surface of the cancer
attachment of a carbohydrate ity to interact with the human immune system (FIG. 3). cell. This has led to the generation of mAbs that bind to
moiety to a protein. The Fc Approaches to enhancing this interaction include peptides expressed by human leukocyte antigen (HLA)
region of immunoglobulin G
includes carbohydrate
changing the amino acid sequence50 or the glycosylation molecules55. These mAbs recognize peptides, such as
moieties. Altering the pattern of the IgG in a way that enhances interaction PR1 and WT1, that are derived from intracellular onco-
enzymes responsible for with FcRs on effector cells51. mAbs with such modifi- proteins56,57. Such mAbs are HLA-restricted (that is, they
glycosylation in a cell line that cations can have enhanced in vitro efficacy and seem only bind to antigen expressed by cells from patients
produces a monoclonal
to be safe in clinical trials52. One glycomodified mAb, with a given HLA genotype) and are in early develop-
antibody (mAb) can alter the
glycosylation of the mAb,
obinutuzumab, was recently approved by the US Food ment; however, they represent a novel approach to
thereby altering its ability to and Drug Administration (FDA) for the treatment of broadening the potential targets for mAb-based therapy.
activate immune effector CLL53. Obinutuzumab is unique in other ways, such
cells. as the manner by which it crosslinks its target antigen Altering the host response
CD20, and the actual therapeutic contribution of chang- Inhibiting angiogenesis. In the early 1970s, Folkman
ing IgG glycosylation remains to be determined. There and others hypothesized that tumours require new
have also been studies exploring mAbs that are based blood vessels to grow, and that the growth of such ves-
on other antibody isotypes, such as IgA54. These agents sels is stimulated by pro-angiogenic factors produced
have some intriguing properties but are not currently in by the cancer 58. A number of substances that stimulate
wide clinical development. intratumoural blood vessel growth were subsequently
identified, including vascular endothelial growth factor
(VEGF). mAbs (including bevacizumab) were produced
Heavy chain to inhibit angiogenesis by interfering with the ability
Light chain of VEGF to stimulate new intratumoural blood vessel
Variable region growth (FIG. 1). This, in turn, has an antitumour effect
Carbohydrate Constant region
by depriving a growing tumour of nutrients and oxy-
gen provided by the new blood vessels59. As such angio-
g Crosslink regions genesis inhibitors do not directly target the cancer and
a Glycomodified mAb from two mAbs
are generally assumed to inhibit cancer growth rather
than induce cancer cell death, they are most often used
in combination with cytotoxic agents60. An important
advantage of this approach is that it is not dependent
b Alter amino f Insert DNA for mAb on expression of a specific target antigen by the tumour
acids in variable region fused
constant to signalling peptide cell, but is instead based on the role of VEGF in neo­
region into T cell to induce angiogenesis, which is vital for the growth of tumours
expression of CAR in general. Bevacizumab has been shown to be effective,
c Use different e Link drug
human mAb d Link isotope to mAb with at least in some clinical trials and scenarios, in various
isotype to mAb with cleavable linker cancers, including colorectal, lung, breast, renal, brain
(e.g. IgG4) stable linker
and ovarian cancer61.
Figure 3 | Modifying monoclonal antibody structure.  Monoclonal antibody (mAb) The unique mechanism of action of angiogenesis
structure can be modified on the basis of the desired mechanism of action. inhibition creates a particular challenge in evaluating
Nature Reviews | Cancer
Immunoglobulin G1 (IgG1) is the most effective naturally occurring human IgG isotype efficacy 62. A standard approach to assess the response
at mediating antibody-dependent cellular cytotoxicity (ADCC). Glycomodified to most anticancer agents involves radiological determi-
afucosylated mAbs (part a) (such as obinutuzumab) demonstrate enhanced binding to nation of tumour shrinkage. It has been proposed that
IgG Fc receptors (FcγRs) and enhanced ADCC. Afucosylated mAbs are produced using angiogenesis inhibitors may temporarily shrink the size
cell lines that lack the enzymes that are responsible for fucosylation. Modifying the of a cancer by affecting vascularity, without having a
amino acid sequence of mAb Fc (part b), as was done to produce ocaratuzumab50, can
significant impact on the growth of the malignant cells.
also result in enhanced binding to FcγRs and enhanced ADCC. For mechanisms of action
in which ADCC is not desirable, IgG4 is a more appropriate isotype, as IgG4 mAbs do not Thus, although it remains controversial, some experts
mediate ADCC to the same degree as IgG1 (part c). Nivolumab, an IgG4 mAb that blocks claim that the evaluation of efficacy of angiogenesis
programmed cell death protein 1 (PD1) on T cells, is one such example. Producing radio- inhibitors should be based on overall survival rather
immunoconjugates involves linking the radioisotope to the mAb. A stable linker is most than on radiographical determination of tumour shrink-
desirable (part d) to limit the leakage of the free radioactive isotope. Conversely, optimal age63. Trials with survival as an end point are more chal-
antibody–drug conjugates (ADCs) use a cleavable linker (part e). To avoid nonspecific lenging, and robust debate continues over the clinical
toxicity, it is desirable for drugs used in ADCs to be cytotoxic once inside the target cell value of angiogenesis inhibition in various cancer types.
but non-toxic when bound to the mAb in the circulation. Linkers that are pH-sensitive or
enzymatically cleaved are now a standard component of ADCs. Chimeric antigen T cell checkpoint blockade. Years of basic immunology
receptor (CAR) T cells get their specificity from mAb variable regions but are a form of
research exploring the exquisite control of T cell
gene, not protein, therapy. They are produced by inserting DNA coding for the mAb
variable region fused to DNA coding for signalling peptides into T cells (part f). Bispecific immunity have demonstrated that co‑regulatory
antibodies require the removal of a functional constant region so that they do not ligand–receptor pairs have a central role in the balance
nonspecifically crosslink activating receptors and activate T cells (part g). The lack of a between activation and inhibition of antigen-specific
constant region on such constructs results in a short half-life, thus requiring continuous (and tumour-specific) T cells64. Such precise control is
infusion to achieve the desired exposure. central to the ability of the immune system to respond

NATURE REVIEWS | CANCER VOLUME 15 | JUNE 2015 | 365

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Opsonization to infection and to avoid the autoimmunity that could This helps to explain the hallmark of Hodgkin lym-
The process by which a target result from an uncontrolled T cell response. Over the phoma that has intrigued haematologists for decades:
is marked for phagocytosis or past decade, mAbs that interfere with the inhibitory how malignant Reed–Sternberg cells are able to survive
for destruction by phagocytes. signals responsible for limiting T cell activation have despite being surrounded by benign immune cell infil-
This term is often used to
describe phagocytosis of
been developed. These mAbs, called checkpoint blockade trates. An early phase clinical trial of single-agent PD1
microbial pathogens. In the mAbs, can maintain the T cell activation phenotype and blockade in relapsed or refractory Hodgkin lymphoma
case of monoclonal antibody enhance T cell-mediated lysis (FIG. 1). In the case of can- demonstrated very positive results74.
therapy of cancer, the target is cer, such mAbs can induce a more robust and sustained
a cancer cell.
antitumour T cell response64,65. Delivering cytotoxic moieties
Checkpoint blockade Cytotoxic T lymphocyte-associated antigen 4 Military analogies have been a part of immunology since
Inhibitory pathways limit T cell (CTLA4; also known as CD152) is a receptor expressed the days of Paul Ehrlich and his description of anti­bodies
activation in order to maintain by activated T cells that results in the downregula- as “magic bullets” (REF. 1). In more recent years, the
self-tolerance and prevent tion of the T cell response. Physiologically, CTLA4 phrase ‘smart bombs’ has been used to describe mAbs
autoimmunity. Checkpoint
blockade involves blocking
and other negative regulators of T cell activation have that deliver cytotoxic moieties to cancer cells. The types
these inhibitory pathways, and a role in blunting the T cell response. Such blunting of payload that have been successfully delivered by mAb-
thereby allowing for to a more of the T cell response is important for avoiding auto­ based smart bombs are quite varied and include radio­
robust T cell response. immunity but is undesirable when it comes to the gen- active molecules, cytotoxic small molecules and cellular
eration and maintenance of an effective antitumour components of the immune system (FIG. 3).
Breakthrough therapy
designation T cell response66. Blocking this negative signal would
US Food and Drug be expected to enhance and maintain such a response, Radioimmunoconjugates. Treatment of thyroid can-
Administration (FDA) and this has proven to be the case. Ipilimumab, a mAb cer with 131I was the first highly effective targeted
designation if a new drug is that blocks CTLA4, has been approved by the FDA and cancer treatment and was successful because of the
intended to treat a serious or
life-threatening disease and
has had a major impact on the treatment of patients specificity of elemental iodine for the thyroid. 131I is a
preliminary clinical evidence with melanoma67. Clinical responses to ipilimumab β-particle emitter that also emits a modest amount
suggests that it provides a can be profound and long-lasting. Ongoing studies in of γ-radiation and so can be used in both diagnosis
substantial improvement over various cancers are exploring regimens that incorporate (imaging) and treatment. This success led investiga-
existing therapies.
ipilimumab, both alone and in combination with other tors to explore approaches to delivering radioisotopes
agents, particularly when there is reason to think that directly to target molecules expressed by other cancers
such therapy could be inducing a T cell response, such using various carrier molecules, including antibodies.
as in the case of vaccines or agents that induce antigen Attempts to use such radioimmunoconjugates to treat
release68. The toxicity of checkpoint blockade mAbs cancer began with polyclonal antibodies before mAb
is also unique and, as would be expected, includes technology was available. Order and colleagues75 pro-
autoimmunity 69. duced polyclonal antisera by immunizing various species
Several mAbs that interfere with a different T cell (including rabbits, pigs, monkeys and cows) with human
regulatory pathway, the programmed cell death pro- ferritin. Antisera from these animals were labelled with
tein 1 (PD1) receptor–PD1 ligand 1 (PDL1) axis, are 131
I and administered to patients with tumours that
also generating considerable excitement. These include were known to express high levels of ferritin receptor 75.
mAbs that bind to the ligand and others that bind to the The results, particularly the images demonstrating evi-
receptor 70,71. Both approaches have been very promis- dence for some targeting of the agent to the tumour,
ing in early clinical trials, and we are sure to learn much were intriguing, but the use of polyclonal antisera from
more about the relative value of these approaches in the various species demonstrated little clinical efficacy and
years ahead. Indeed, the FDA has recently approved two posed many problems. These included significant con-
PD1‑specific mAbs (pembrolizumab and nivolumab) for sistency and quality-control issues, and the development
the treatment of melanoma. A mAb against PDL1 has of host immune responses against the foreign proteins
been granted breakthrough therapy designation by the FDA (similar to classic serum sickness)76.
on the basis of its promise in early phase clinical trials70. mAb technology solved some of these problems and
Whereas CTLA4 regulates de novo immune responses, resulted in a renewed interest in radioimmunoconju-
the PD1–PDL1 axis has a greater influence on ongoing gates and radioimmunotherapy. However, challenges
T cell immune responses. Thus the concept of check- have persisted and have limited the clinical utility of
point blockade is similar, but the efficacy and toxicity radioimmunotherapy. Radioisotopes continually decay
of mAbs that target these two pathways are likely to be and cause nonspecific radiation damage to normal tis-
different. For instance, a PD1‑specific mAb has been sue when the agent is in the circulation or is taken up
found to be effective in patients with melanoma refrac- nonspecifically by normal tissues77. Bone marrow is
tory to CTLA4‑specific therapy 72. Combination trials particularly radiosensitive and is therefore affected by
exploring the inhibition of CTLA4 and PD1 together are circulating radioimmunoconjugate. The kidney and the
promising 73, although blocking both pathways does raise liver receive high doses of radiation because of their role
concerns about enhanced autoimmunity. in clearing the radioimmunoconjugate and free radio-
The PD1–PDL1 axis seems to be particularly impor- isotope, and only a small fraction (typically 0.01–0.001%
tant in classic Hodgkin lymphoma in which Reed– per gram of tumour) of the injected radiation dose ends
Sternberg cells have recently been shown to express up in the tumour, even for the most specific agents77.
PDL1 and PDL2, which allows them to turn off T cells. Optimizing the very narrow therapeutic window of

366 | JUNE 2015 | VOLUME 15 www.nature.com/reviews/cancer

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

radioimmunotherapy requires careful choosing of the a cancer cell, enter the cell by endocytosis and traffic
mAb, the radioisotope and the chemistry that links within the cell to the site where it can kill the cell. The
the two. Experience over the past 25 years has taught toxins need to be modified by removing the moieties
us that radioimmunoconjugates directed towards solid responsible for nonspecific binding to normal cells, so
tumours can be useful as diagnostic agents, but that they that cellular targeting and uptake is controlled by mAb
have limited therapeutic impact owing to radioresistance specificity. Initial studies of immunotoxins were carried
of the tumours78. A radioimmunoconjugate consisting of out using protein conjugation techniques, such as the
a radioiodinated mAb that targets necrotic tumour cells use of heterobifunctional reagents to link the mAb to
has been approved for clinical use in China79. Ongoing the toxin. Subsequent efforts involved the development
studies are evaluating radioimmunotherapy combined of recombinant immunotoxins. These agents were found
with chemotherapy. to be highly potent, but the immunogenicity and non-
Lymphoma is uniquely suited for radioimmuno­ specific toxicity of protein toxins posed major problems
therapy because of the availability of highly specific and limited further development 87.
target antigens and its relative radiosensitivity. Two Many of the important lessons learned from the study
radio­immunoconjugates, ibritumomab tiuxetan80 and of immunotoxins were applied to the development and
tositumomab81, have been approved by the FDA as testing of antibody–drug conjugates (ADCs), a field that
therapeutic agents for the treatment of lymphoma. They is showing great promise88. ADCs combine the cytotoxic
are both based on CD20‑specific mAbs but use differ- potential of drugs with the specifıcity of mAbs and theor­
ent isotopes (90Y for ibritumomab tiuxetan and 131I for etically overcome the limitations of both nonspecifıc
tositumomab). The logistics of administration, need cytotoxic drugs and specifıc but often ineffective mAbs.
for dosimetry and pharmacokinetics of these two agents When choosing the target antigen and the mAb for ideal
are different, but their clinical efficacies and toxic effects are ADCs, desirable characteristics are different from those
similar. Despite the clear clinical efficacy and limited used for unlabelled mAbs89. Tumour antigens for ADCs
toxicity of lymphoma radioimmunotherapy, clinical can be expressed at lower concentrations compared with
use of this modality has been surprisingly limited. This unlabelled mAbs because the delivery of a highly toxic
seems to be due to the complex logistics of providing drug by a small number of ADC molecules can result
radioimmunotherapy (it requires experienced nuclear- in the death of the target cancer cell. Depending on the
medicine physicians), and the emergence of a number drug used, only a few molecules delivered intracellularly
of other new therapies for lymphoma that are less com- may be capable of killing a target cell. Internalization
plex to deliver. Recent and ongoing studies are explor- properties are also important. For unlabelled mAbs, it is
ing additional clinical questions related to lymphoma helpful for the mAb–antigen complex to remain on the
radioimmunotherapy, including the timing of therapy surface of the cell so that it can be recognized by the host
(at diagnosis versus at relapse), which lymphoma sub- immune system. For ADCs, internalization needs to take
types should be treated (given that it is most effec- place so that the drug can be released inside the cancer
tive for follicular lymphoma) and how therapy should cell and mediate its toxic effect.
be combined with other treatments, including bone ADCs use small molecules instead of protein tox-
marrow transplantation82 and novel agents83. ins and thereby reduce immunogenicity. Drugs used
Ongoing studies are also exploring radioimmuno- as components of ADCs include potent drugs such as
conjugates that contain novel radioisotopes, including calicheamicin, which binds to the minor groove in DNA
α-emitters84. The advantage of α-emitters is that the and causes strand scission90; monomethyl auristatin E,
ionizing radiation is delivered very locally (within just which blocks polymerization of tubulin91; maitansine,
a few cell diameters), which limits radiation exposure which inhibits the assembly of microtubules92; and,
to cells that express the target antigen and spares benign most recently, pyrrolobenzodiazepines, which crosslink
neighbouring cells. This highly specific delivery of DNA93. These are extremely potent drugs; indeed, some
radiation allows the treatment of malignancies such as were evaluated as stand-alone chemotherapy agents and
leukaemia that are not geographically isolated but that rejected because of their toxicity at very low doses90,94.
are highly radiosensitive. The radiochemistry of work- The linker that connects the mAb to the drug is vital.
ing with these isotopes is challenging because of their It needs to attach the drug to the mAb in a manner that
very short half-life and because the production of iso- does not alter the specificity of the mAb, to render the
topes with promising properties, such as 211At, requires drug non-toxic while bound to the mAb, to remain
a cyclotron85. However, not all α-particle-emitting iso- stable in the circulation and to release the drug in the
topes are short-lived. Preliminary preclinical and clinical appropriate intracellular compartment when the ADC
results using α-particles are intriguing 86 and studies are is internalized, so that the drug can kill the target cell95.
ongoing. Linker motifs include disulfides, hydrazones, peptides
and thioethers. Some are cleavable, whereas others are
Antibody–drug conjugates. In the 1970s and 1980s, a not, based on the properties of the mAb and the desired
number of investigators began developing and testing distribution of the cytotoxic agent within the cancer cell.
immunotoxins composed of mAbs linked to a variety of As with radioimmunoconjugates, ADCs seem to be
very potent protein toxins, such as ricin, pseudomonas particularly effective in lymphoma, but they are also
exotoxin and diphtheria toxin. For an immunotoxin showing promise in solid tumours. Brentuximab vedo-
to be effective, it needs to bind to a surface antigen on tin (for lymphoma)96,97 and ado-trastuzumab emtansine

NATURE REVIEWS | CANCER VOLUME 15 | JUNE 2015 | 367

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

(for breast cancer)98 were the first ADCs to be approved are under development, including some with different
by the FDA. The field is rapidly expanding with more structures that have longer half-lives and that may not
than 30 ADCs currently in development and in clinical require continuous infusion106,107.
trials99, and many have shown encouraging preliminary
results100,101. The ADC field is currently in its infancy, Chimeric antigen receptor T cells. Another example
and major advances based on the design of new ADCs, of how persistence has paid off in mAb-based cancer
and a better understanding of how to use them, are sure immuno­therapy is demonstrated by the decades of
to come. research that preceded the current high level of excitement
An approach that combines targeting of the cancer surrounding chimeric antigen receptor (CAR) T cells108.
cell directly with altering the tumour microenviron- CAR T cells are genetically modified to respond to target
ment is the use of immunocytokines that can mediate cells expressing a given antigen, and consist of a mAb vari-
their antitumour effect through both the direct effect able region linked to a T cell-activating motif. A number
of the mAb and the ability to deliver a cytokine, such as of investigators worked for many years to refine various
interleukin (IL‑2) or granulocyte–macrophage colony- antigen-specific constructs and approaches to transferring
stimulating factor (GM‑CSF), to the tumour. Most notable those constructs into primary T cells109. Recent clinical
in this area of research have been immunocytokines based trials based on some of the newer constructs show that
on ganglioside-­specific GD2, with clinical development CAR T cells can lead to robust therapeutic responses110,111.
mostly centred on the treatment of neuroblastoma102,103. They also result in significant, but increasingly manage-
These agents have shown remarkable efficacy in this able, toxicity owing to cytokine storm that occurs as a
rare childhood malignancy. A modified version of this consequence of massive activation and proliferation of
immuno­cytokine, designed to mediate reduced comple- CAR T cells; activation and proliferation are induced
ment fixation that is thought to decrease its neurotoxicity, when CAR T cells come into contact with a large tumour
has also shown promise in an early phase clinical trial104. burden. CAR T cells are unique among mAb-based treat-
ment strategies in that the antitumour effect can expand
Retargeting T cells over time without additional therapy as the CAR T cells
Tumour immunology has taught us that T cell immu- divide and the tumour specificity of the CAR T cells is
nity is key to the immune rejection of many cancers. passed on to daughter cells.
The initial step of cell-mediated cytolysis involves the Indeed, long-lived CAR T memory cells have been
formation of conjugates between T cells and target observed in a number of patients 112. This memory
cells. The appropriate engagement of receptors and response represents both an advantage and a challenge.
co‑­receptors on T cells is followed by the triggering of On the one hand, it probably contributes to the very grati-
cytotoxic responses that result in the death of the cancer fying long-term clinical responses that are seen in some
cell. Two broad approaches to combining the specificity patients treated with CAR T cells. On the other hand, such
of mAbs with the power of T cell immune responses long-term immune memory can result in long-term, per-
have been explored with the hope of enhancing immune haps even permanent, depletion of any cells that express
Bispecific antibody
rejection of cancer (FIG. 3). Both of these approaches are the target antigen. In the case of B cell malignancies, long-
(Also known as bifunctional
antibody). An engineered designed to retarget large numbers of T cells towards the term loss of benign B cells that express CD19 may be an
monoclonal antibody (mAb) tumour by bypassing the need for the T cell receptor to acceptable toxicity. However, most tumour antigens are
that is composed of fragments recognize target antigens as processed and expressed by not as specific as CD19, and long-term toxicity to vital
of two different mAbs that bind target cell major histocompatibility complexes (MHCs). normal tissues can result if such tissues express even low
to two different antigens. In
cancer therapy, it typically
levels of the target antigen recognized by CAR T cells. This
binds to an activating antigen Bispecific antibodies. The first approach involves the may limit the number and types of cancers that can be
on an immune effector cell with creation of bispecific antibody-like molecules that have one treated with this approach.
one arm and to a tumour- arm that binds to the target cell and one arm that binds Bispecific antibodies and CAR T cells each have advan-
associated antigen on a cancer
to activating receptors on cytotoxic cells, such as T cells tages and disadvantages. Bispecific antibodies are ‘off-the-
cell with the other arm, thereby
retargeting the immune or NK cells. Most bispecific antibodies involve constructs shelf ’ reagents and can be stopped if autoimmune toxic
effector cell towards the target that bind to an antigen on a cancer cell with one arm effects are observed, but they are challenging to admin-
cancer cell. and to T cell surface glycoprotein CD3 ε-chain (CD3) on ister. CAR T cells need to be produced individually for
T cells with the other arm. This, as with many of the strat- each patient, but they expand in response to tumour and
Chimeric antigen receptor
(CAR). An engineered receptor
egies based on using mAbs to treat cancer, is not a new can result in long-term antitumour immune responses.
that grafts an alternative concept, but it has benefited from a steady improvement Both approaches can result in the rapid activation of large
specificity onto an immune in technology and lessons learned from earlier preclinical numbers of T cells interacting with large numbers of cells
effector cell, most often a and clinical studies105. For example, early studies demon- expressing the target antigen113. The resulting massive
T cell. Most of the current
strated that bispecific antibodies with intact Fc activate release of cytokines can result in a clinical cytokine storm
constructs for engineered
receptors include a single-chain T cells nonspecifically and result in unacceptable toxicity. with potentially devastating physiological effects, includ-
monoclonal antibody variable Smaller bispecific molecules lacking Fc have short half- ing cardiovascular collapse114. Cytokine storms can be
region that recognizes the lives and need to be given by continuous infusion, but limited by stopping infusion for bispecific antibodies or by
target cell and is linked to these molecules result in less nonspecific immune activa- using mAbs against certain cytokines such as IL-6‑specific
activating transmembrane
domains that activate the T cell
tion than do intact bispecific mAbs. Recent positive clini- mAbs for CAR T cells. Accumulating experience with
when it comes into contact cal trials with the bispecific mAb blinatumomab led to CAR T cells is leading to the development of protocols
with a target cell. its approval by the FDA. Additional bispecific antibodies to prevent or treat this challenging complication114.

368 | JUNE 2015 | VOLUME 15 www.nature.com/reviews/cancer

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Additional studies are needed to determine the relative Indeed, investment in mAb-based therapy of can-
efficacy, toxicity and value of these two exciting new cer from both the public and the private sectors and
strategies for using mAbs to treat cancer. the speed of progress have never been greater. Recent
advances and positive clinical results in checkpoint
Conclusion blockade, ADCs and retargeting T  cells via CAR
Cancer mAb therapy has come a long way since the days T cells and bispecific antibodies are particularly excit-
when unmodified murine mAbs were first explored as ing. We are in a period of unprecedented progress
anticancer agents. A wide variety of mAb-based strate- in mAb-based therapy with the rapid development
gies have proven to be useful in treating patients with of new therapeutic agents and constructs, enhanced
cancer, including: the use of unlabelled IgG that binds understanding of their biological effects, and grow-
directly to cancer cells, mAbs that alter the active host ing clinical experience based on both clinical trials
response to the cancer, immunoconjugates that deliver and community use of FDA-approved drugs. With
cytotoxic moieties to the cancer and constructs that use dedicated attention to basic, translational and clinical
the specificity of mAbs to retarget cellular immunity research geared towards additional progress, we will
towards the cancer cell. The demonstration of clinical certainly be able to build even better anticancer mAbs
efficacy for each of these approaches was made possible in the years ahead. Equally important will be studies
by the dedication and persistence of investigators who evaluating how best to use these agents, alone and in
would not give up on a good idea. combination, to better serve our patients.

1. Ehrlich, P. Collected Studies on Immunity 1st edn 16. Pedersen, I. M., Buhl, A. M., Klausen, P., Geisler, C. H. 32. Pawluczkowycz, A. W. et al. Binding of submaximal
(J. Wiley & Sons, 1906). & Jurlander, J. The chimeric anti‑CD20 antibody C1q promotes complement-dependent cytotoxicity
2. Keller, M. A. & Stiehm, E. R. Passive immunity in rituximab induces apoptosis in B‑cell chronic (CDC) of B cells opsonized with anti‑CD20 mAbs
prevention and treatment of infectious diseases. Clin. lymphocytic leukemia cells through a p38 mitogen ofatumumab (OFA) or rituximab (RTX): considerably
Microbiol. Rev. 13, 602–614 (2000). activated protein-kinase-dependent mechanism. Blood higher levels of CDC are induced by OFA than by RTX.
3. Kohler, G. & Milstein, C. Continuous cultures of fused 99, 1314–1319 (2002). J. Immunol. 183, 749–758 (2009).
cells secreting antibody of predefined specificity. 17. Weiner, G. J. Rituximab: mechanism of action. Semin. 33. Beum, P. V. et al. Loss of CD20 and bound CD20
Nature 256, 495 (1975). Hematol. 47, 115–123 (2010). antibody from opsonized B cells occurs more rapidly
This landmark publication outlines how to produce 18. Shan, D., Ledbetter, J. A. & Press, O. W. Signaling because of trogocytosis mediated by Fc receptor-
mAbs, as well as their therapeutic potential. events involved in anti‑CD20‑induced apoptosis of expressing effector cells than direct internalization by
4. Milstein, C. From the structure of antibodies to the malignant human B cells. Cancer Immunol. the B cells. J. Immunol. 187, 3438–3447 (2011).
diversification of the immune response. Nobel lecture, Immunother. 48, 673–683 (2000). 34. Wang, S. Y. et al. Depletion of the C3 component of
8 December 1984. Biosci. Rep. 5, 275–297 (1985). 19. Rimawi, M. F., Schiff, R. & Osborne, C. K. Targeting complement enhances the ability of rituximab-coated
5. Bernard, A. & Boumsell, L. The clusters of HER2 for the treatment of breast cancer. Annu. Rev. target cells to activate human NK cells and improves
differentiation (CD) defined by the First International Med. 66, 111–128 (2015). the efficacy of monoclonal antibody therapy in an
Workshop on Human Leucocyte Differentiation 20. Lavaud, P. & Andre, F. Strategies to overcome in vivo model. Blood 114, 5322–5330 (2009).
Antigens. Hum. Immunol. 11, 1–10 (1984). trastuzumab resistance in HER2‑overexpressing 35. Dall’Ozzo, S. et al. Rituximab-dependent cytotoxicity
6. Vaickus, L. & Foon, K. A. Overview of monoclonal breast cancers: focus on new data from clinical trials. by natural killer cells: influence of FCGR3A
antibodies in the diagnosis and therapy of cancer. BMC Med. 12, 132 (2014). polymorphism on the concentration–effect
Cancer Invest. 9, 195–209 (1991). 21. Yarden, Y. & Sliwkowski, M. X. Untangling the ErbB relationship. Cancer Res. 64, 4664–4669 (2004).
7. Meeker, T. C. et al. A clinical trial of anti-idiotype signalling network. Nature Rev. Mol. Cell Biol. 2, 36. Lefebvre, M. L., Krause, S. W., Salcedo, M. &
therapy for B cell malignancy. Blood 65, 1349–1363 127–137 (2001). Nardin, A. Ex vivo-activated human macrophages kill
(1985). 22. Yarden, Y. & Pines, G. The ERBB network: at last, chronic lymphocytic leukemia cells in the presence of
This early clinical trial demonstrated the potential cancer therapy meets systems biology. Nature Rev. rituximab: mechanism of antibody-dependent cellular
efficacy of mAb-based therapy and many of the Cancer 12, 553–563 (2012). cytotoxicity and impact of human serum.
challenges associated with it. 23. Franklin, M. C. et al. Insights into ErbB signaling from J. Immunother. 29, 388–397 (2006).
8. LoBuglio, A. F. et al. Mouse/human chimeric the structure of the ErbB2‑pertuzumab complex. 37. Hernandez-Ilizaliturri, F. J. et al. Neutrophils
monoclonal antibody in man: kinetics and immune Cancer Cell 5, 317–328 (2004). contribute to the biological antitumor activity of
response. Proc. Natl Acad. Sci. USA 86, 4220–4224 24. Le, X. F. et al. Differential signaling by an anti‑p185HER2 rituximab in a non-Hodgkin’s lymphoma severe
(1989). antibody and heregulin. Cancer Res. 60, 3522–3531 combined immunodeficiency mouse model. Clin.
9. Maloney, D. G. et al. Idec‑C2b8 (Rituximab) anti‑CD20 (2000). Cancer Res. 9, 5866–5873 (2003).
monoclonal antibody therapy patients with relapsed 25. Swain, S. M. et al. Pertuzumab, trastuzumab, and 38. Beers, S. A. & Glennie, M. J. Neutrophils: “neu players”
low-grade non-Hodgkins lymphoma. Blood 90, docetaxel for HER2‑positive metastatic breast cancer in antibody therapy? Blood 122, 3093–3094 (2013).
2188–2195 (1997). (CLEOPATRA study): overall survival results from a 39. Clynes, R. A., Towers, T. L., Presta, L. G. &
This article details the first clinical trial to randomised, double-blind, placebo-controlled, Ravetch, J. V. Inhibitory Fc receptors modulate in vivo
demonstrate the clear clinical efficacy of a chimeric Phase 3 study. Lancet Oncol. 14, 461–471 (2013). cytotoxicity against tumor targets. Nature Med. 6,
human–murine mAb. 26. Flexner, S. & Noguchi, H. Snake venom in relation to 443–446 (2000).
10. Scott, A. M., Wolchok, J. D. & Old, L. J. Antibody haemolysis, bacteriolysis, and toxicity. J. Exp. Med. 6, This important publication illustrates the
therapy of cancer. Nature Rev. Cancer 12, 278–287 277–301 (1902). importance of both activating and inhibitory FcRs
(2012). 27. Wang, S. Y. & Weiner, G. Complement and cellular in mediating the antitumour effects of mAbs.
11. Tutt, A. L. et al. Monoclonal antibody therapy of B cell cytotoxicity in antibody therapy of cancer. Expert 40. Bowles, J. A. & Weiner, G. J. CD16 polymorphisms and
lymphoma: signaling activity on tumor cells appears Opin. Biol. Ther. 8, 759–768 (2008). NK activation induced by monoclonal antibody-coated
more important than recruitment of effectors. 28. Teeling, J. L. et al. The biological activity of human target cells. J. Immunol. Methods 304, 88–99 (2005).
J. Immunol. 161, 3176–3185 (1998). CD20 monoclonal antibodies is linked to unique 41. Hoffmeyer, F., Witte, K. & Schmidt, R. E. The high-
12. Taylor, R. P. Of mice and mechanisms: identifying the epitopes on CD20. J. Immunol. 177, 362–371 affinity FcγRI on PMN: regulation of expression and
role of complement in monoclonal antibody-based (2006). signal transduction. Immunology 92, 544–552 (1997).
immunotherapy. Haematologica 91, 146a (2006). 29. Alduaij, W. et al. Novel type II anti‑CD20 monoclonal 42. Glennie, M. J., French, R. R., Cragg, M. S. & Taylor, R. P.
13. Clynes, R., Takechi, Y., Moroi, Y., Houghton, A. & antibody (GA101) evokes homotypic adhesion and Mechanisms of killing by anti‑CD20 monoclonal
Ravetch, J. V. Fc receptors are required in passive and actin-dependent, lysosome-mediated cell death in antibodies. Mol. Immunol. 44, 3823–3837 (2007).
active immunity to melanoma. Proc. Natl Acad. Sci. B‑cell malignancies. Blood 117, 4519–4529 (2011). 43. Rogers, L. M., Veeramani, S. & Weiner, G. J.
USA 95, 652–656 (1998). 30. Natsume, A., Shimizu-Yokoyama, Y., Satoh, M., Complement in monoclonal antibody therapy of
14. Beers, S. A. et al. Type II (tositumomab) anti‑CD20 Shitara, K. & Niwa, R. Engineered anti‑CD20 cancer. Immunol. Res. 59, 203–210 (2014).
monoclonal antibody out performs type I (rituximab- antibodies with enhanced complement-activating 44. Wang, S. Y., Racila, E., Taylor, R. P. & Weiner, G. J.
like) reagents in B‑cell depletion regardless of capacity mediate potent anti-lymphoma activity. NK‑cell activation and antibody-dependent cellular
complement activation. Blood 112, 4170–4177 Cancer Sci. 100, 2411–2418 (2009). cytotoxicity induced by rituximab-coated target cells is
(2008). 31. Mossner, E. et al. Increasing the efficacy of CD20 inhibited by the C3b component of complement.
15. Shan, D., Ledbetter, J. A. & Press, O. W. Apoptosis of antibody therapy through the engineering of a new Blood 111, 1456–1463 (2008).
malignant human B cells by ligation of CD20 type II anti‑CD20 antibody with enhanced direct and 45. Harbers, S. O. et al. Antibody-enhanced cross-
monoclonal antibodies. Blood 91, 1644–1652 immune effector cell-mediated B‑cell cytotoxicity. presentation of self antigen breaks T cell tolerance.
(1998). Blood 115, 4393–4402 (2010). J. Clin. Invest. 117, 1361–1369 (2007).

NATURE REVIEWS | CANCER VOLUME 15 | JUNE 2015 | 369

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

46. Vaughan, A. T. et al. Inhibitory FcγRIIb (CD32b) 69. Sondak, V. K., Smalley, K. S., Kudchadkar, R., 93. Cooper, N. et al. Synthesis of novel C2‑aryl
becomes activated by therapeutic mAb in both cis and Grippon, S. & Kirkpatrick, P. Ipilimumab. Nature Rev. pyrrolobenzodiazepines (PBDs) as potential antitumour
trans and drives internalization according to antibody Drug Discov. 10, 411–412 (2011). agents. Chem. Commun. 21, 1764–1765 (2002).
specificity. Blood 123, 669–677 (2014). 70. Brahmer, J. R. et al. Safety and activity of anti‑PD‑L1 94. Sievers, E. L. & Senter, P. D. Antibody–drug conjugates
47. Feldman, A. M., Lorell, B. H. & Reis, S. E. Trastuzumab antibody in patients with advanced cancer. N. Engl. in cancer therapy. Annu. Rev. Med. 64, 15–29 (2013).
in the treatment of metastatic breast cancer: J. Med. 366, 2455–2465 (2012). 95. Shen, B. Q. et al. Conjugation site modulates the in vivo
anticancer therapy versus cardiotoxicity. Circulation 71. Robert, C. et al. Anti-programmed-death-receptor‑1 stability and therapeutic activity of antibody–drug
102, 272–274 (2000). treatment with pembrolizumab in ipilimumab- conjugates. Nature Biotech. 30, 184–189 (2012).
48. Nadler, L. M. et al. Serotherapy of a patient with a refractory advanced melanoma: a randomised dose- 96. Younes, A. et al. Results of a pivotal Phase II study of
monoclonal antibody directed against a human comparison cohort of a Phase 1 trial. Lancet 384, brentuximab vedotin for patients with relapsed or
lymphoma-associated antigen. Cancer Res. 40, 1109–1117 (2014). refractory Hodgkin’s lymphoma. J. Clin. Oncol. 30,
3147–3154 (1980). 72. Wolchok, J. D. et al. Nivolumab plus ipilimumab in 2183–2189 (2012).
49. de Goeij, B. E. et al. HER2 monoclonal antibodies that advanced melanoma. N. Engl. J. Med. 369, 97. Gopal, A. K. et al. Durable remissions in a pivotal
do not interfere with receptor heterodimerization- 122–133 (2013). Phase 2 study of brentuximab vedotin in relapsed or
mediated signaling induce effective internalization and 73. Topalian, S. L. et al. Safety, activity, and immune refractory Hodgkin lymphoma. Blood 125,
represent valuable components for rational antibody– correlates of anti‑PD‑1 antibody in cancer. N. Engl. 1236–1243 (2015).
drug conjugate design. mAbs 6, 392–402 (2014). J. Med. 366, 2443–2454 (2012). 98. Verma, S. et al. Trastuzumab emtansine for
50. Bowles, J. A. et al. Anti‑CD20 monoclonal antibody 74. Ansell, S. M. et al. PD‑1 blockade with nivolumab in HER2‑positive advanced breast cancer. N. Engl.
with enhanced affinity for CD16 activates NK cells at relapsed or refractory Hodgkin’s lymphoma. N. Engl. J. Med. 367, 1783–1791 (2012).
lower concentrations and more effectively than J. Med. 372, 311–319 (2015). This clinical trial demonstrated the potential efficacy
rituximab. Blood 108, 2648–2654. (2006). This article describes an early phase clinical trial of an ADC in a solid tumour.
51. Dalle, S. et al. Preclinical studies on the mechanism of that demonstrates how an enhanced understanding 99. Mullard, A. Maturing antibody–drug conjugate pipeline
action and the anti-lymphoma activity of the novel of the immune response can lead to the hits 30. Nature Rev. Drug Discov. 12, 329–332 (2013).
anti‑CD20 antibody GA101. Mol. Cancer Ther. 10, development of a new therapeutic approach. 100. Bendell, J. et al. Phase I/II study of the antibody–drug
178–185 (2011). 75. Order, S. E. The history and progress of serologic conjugate glembatumumab vedotin in patients with
52. Salles, G. et al. Phase 1 study results of the type II immunotherapy and radiodiagnosis. Radiology 118, locally advanced or metastatic breast cancer. J. Clin.
glycoengineered humanized anti‑CD20 monoclonal 219–223 (1976). Oncol. 32, 3619–3625 (2014).
antibody obinutuzumab (GA101) in B‑cell lymphoma 76. Order, S. E. et al. Radiolabeled antibody in the 101. Ott, P. A. et al. Phase I/II study of the antibody–drug
patients. Blood 119, 5126–5132 (2012). treatment of primary and metastatic liver conjugate glembatumumab vedotin in patients with
53. Cartron, G. et al. Obinutuzumab (GA101) in relapsed/ malignancies. Recent Results Cancer Res. 1986, advanced melanoma. J. Clin. Oncol. 32, 3659–3666
refractory chronic lymphocytic leukemia: final data 100307–100314 (1986). (2014).
from the Phase 1/2 GAUGUIN study. Blood 124, 77. Pouget, J. P. et al. Clinical radioimmunotherapy — the 102. Yu, A. L. et al. Anti‑GD2 antibody with GM‑CSF,
2196–2202 (2014). role of radiobiology. Nature Rev. Clin. Oncol. 8, interleukin‑2, and isotretinoin for neuroblastoma.
54. Lohse, S. et al. Recombinant dimeric IgA antibodies 720–734 (2011). N. Engl. J. Med. 363, 1324–1334 (2010).
against the epidermal growth factor receptor mediate 78. Jain, M., Venkatraman, G. & Batra, S. K. Optimization 103. Shusterman, S. et al. Antitumor activity of
effective tumor cell killing. J. Immunol. 186, of radioimmunotherapy of solid tumors: biological hu14.18‑IL2 in patients with relapsed/refractory
3770–3778 (2011). impediments and their modulation. Clin. Cancer Res. neuroblastoma: a Children’s Oncology Group (COG)
55. Dao, T., Liu, C. & Scheinberg, D. A. Approaching 13, 1374–1382 (2007). Phase II study. J. Clin. Oncol. 28, 4969–4975 (2010).
untargetable tumor-associated antigens with 79. Chen, S. et al. Pivotal study of iodine‑131‑labeled 104. Navid, F. et al. Phase I trial of a novel anti‑GD2
antibodies. Oncoimmunology 2, e24678 (2013). chimeric tumor necrosis treatment monoclonal antibody, Hu14.18K322A, designed to
56. Dubrovsky, L. et al. A TCR-mimic antibody to WT1 radioimmunotherapy in patients with advanced lung decrease toxicity in children with refractory or recurrent
bypasses tyrosine kinase inhibitor resistance in human cancer. J. Clin. Oncol. 23, 1538–1547 (2005). neuroblastoma. J. Clin. Oncol. 32, 1445–1452 (2014).
BCR–ABL+ leukemias. Blood 123, 3296–3304 80. Witzig, T. E. et al. Randomized controlled trial of 105. Segal, D. M., Weiner, G. J. & Weiner, L. M. Bispecific
(2014). yttrium‑90‑labeled ibritumomab tiuxetan antibodies in cancer therapy. Curr. Opin. Immunol. 11,
57. Sergeeva, A. et al. An anti‑PR1/HLA‑A2 T‑cell receptor- radioimmunotherapy versus rituximab immunotherapy 558–562 (1999).
like antibody mediates complement-dependent for patients with relapsed or refractory low-grade, 106. Nagorsen, D. et al. Immunotherapy of lymphoma and
cytotoxicity against acute myeloid leukemia progenitor follicular, or transformed B‑cell non-Hodgkin’s leukemia with T‑cell engaging BiTE antibody
cells. Blood 117, 4262–4272 (2011). lymphoma. J. Clin. Oncol. 20, 2453–2463 (2002). blinatumomab. Leukemia Lymphoma 50, 886–891
58. Folkman, J. Tumor angiogenesis: therapeutic 81. Kaminski, M. S. et al. Radioimmunotherapy of B‑cell (2009).
implications. N. Engl. J. Med. 285, 1182–1186 lymphoma with 131I anti‑B1 (anti‑CD20) antibody. 107. Topp, M. S. et al. Targeted therapy with the T‑cell-
(1971). N. Engl. J. Med. 329, 459–465 (1993). engaging antibody blinatumomab of chemotherapy-
This article is a description of the potential This early clinical trial demonstrated the efficacy of refractory minimal residual disease in B‑lineage acute
therapeutic impact of altering angiogenesis. radioimmunotherapy. lymphoblastic leukemia patients results in high
59. Ferrara, N., Hillan, K. J., Gerber, H. P. & Novotny, W. 82. Kolstad, A. et al. Nordic MCL3 study: response rate and prolonged leukemia-free survival.
Discovery and development of bevacizumab, an anti- 90Y‑ibritumomab-tiuxetan added to BEAM/C in J. Clin. Oncol. 29, 2493–2498 (2011).
VEGF antibody for treating cancer. Nature Rev. Drug non‑CR patients before transplant in mantle cell This clinical trial demonstrated the potential
Discov. 3, 391–400 (2004). lymphoma. Blood 123, 2953–2959 (2014). efficacy of bispecific antibody therapy.
60. Sennino, B. & McDonald, D. M. Controlling escape 83. Witzig, T. E. et al. A Phase I trial of immunostimulatory 108. Barrett, D. M., Singh, N., Porter, D. L., Grupp, S. A. &
from angiogenesis inhibitors. Nature Rev. Cancer 12, CpG 7909 oligodeoxynucleotide and yttrium June, C. H. Chimeric antigen receptor therapy for
699–709 (2012). ibritumomab tiuxetan radioimmunotherapy for cancer. Annu. Rev. Med. 65, 333–347 (2014).
61. Lima, A. B., Macedo, L. T. & Sasse, A. D. Addition of relapsed B‑cell non-Hodgkin lymphoma. 109. June, C., Rosenberg, S. A., Sadelain, M. & Weber, J. S.
bevacizumab to chemotherapy in advanced non-small Am. J. Hematol. 53, 211–217 (2012). T‑cell therapy at the threshold. Nature Biotech. 30,
cell lung cancer: a systematic review and meta- 84. Allen, B. J. Can α-radioimmunotherapy increase 611–614 (2012).
analysis. PLoS ONE 6, e22681 (2011). efficacy for the systemic control of cancer? 110. Lee, D. W. et al. T cells expressing CD19 chimeric
62. Wen, P. Y. et al. Updated response assessment criteria Immunotherapy 3, 455–458 (2011). antigen receptors for acute lymphoblastic leukaemia
for high-grade gliomas: response assessment in neuro- 85. Zalutsky, M. R. & Pruszynski, M. Astatine‑211: in children and young adults: a Phase 1 dose-
oncology working group. J. Clin. Oncol. 28, production and availability. Curr. Radiopharm. 4, escalation trial. Lancet 385, 517–528 (2014).
1963–1972 (2010). 177–185 (2011). 111. Maude, S. L. et al. Chimeric antigen receptor T cells
63. Cristofanilli, M., Charnsangavej, C. & 86. Scheinberg, D. A. & McDevitt, M. R. Actinium‑225 in for sustained remissions in leukemia. N. Engl. J. Med.
Hortobagyi, G. N. Angiogenesis modulation in cancer targeted α-particle therapeutic applications. Curr. 371, 1507–1517 (2014).
research: novel clinical approaches. Nature Rev. Drug Radiopharm. 4, 306–320 (2011). 112. Kalos, M. et al. T cells with chimeric antigen receptors
Discov. 1, 415–426 (2002). 87. Ghetie, V. & Vitetta, E. Immunotoxins in the therapy of have potent antitumor effects and can establish
64. Pardoll, D. M. The blockade of immune checkpoints in cancer: from bench to clinic. Pharmacol. Ther. 63, memory in patients with advanced leukemia. Sci.
cancer immunotherapy. Nature Rev. Cancer 12, 209–234 (1994). Transl Med. 3, 95ra73 (2011).
252–264 (2012). 88. Zolot, R. S., Basu, S. & Million, R. P. Antibody–drug This early demonstration of the potential of CAR
65. Page, D. B., Postow, M. A., Callahan, M. K., conjugates. Nature Rev. Drug Discov. 12, 259–260 T cells included the potential to develop CAR
Allison, J. P. & Wolchok, J. D. Immune modulation in (2013). T memory cells.
cancer with antibodies. Annu. Rev. Med. 65, 89. Mathur, R. & Weiner, G. J. Picking the optimal target 113. Maude, S. L., Barrett, D., Teachey, D. T. & Grupp, S. A.
185–202 (2014). for antibody–drug conjugates. American Society of Managing cytokine release syndrome associated with
66. Peggs, K. S., Quezada, S. A., Korman, A. J. & Clinical Oncology [online], http://meetinglibrary.asco. novel T cell-engaging therapies. Cancer J. 20,
Allison, J. P. Principles and use of anti‑CTLA4 antibody org/content/17-132 (2013). 119–122 (2014).
in human cancer immunotherapy. Curr. Opin. 90. Hinman, L. M. et al. Preparation and characterization 114. DeFrancesco, L. CAR‑T cell therapy seeks strategies to
Immunol. 18, 206–213 (2006). of monoclonal antibody conjugates of the harness cytokine storm. Nature Biotech. 32, 604 (2014).
67. Hodi, F. S. et al. Improved survival with ipilimumab in calicheamicins: a novel and potent family of antitumor
patients with metastatic melanoma. N. Engl. J. Med. antibiotics. Cancer Res. 53, 3336–3342 (1993). Acknowledgements
363, 711–723 (2010). 91. Pettit, G. R. et al. Antineoplastic agents 337. The author would like to acknowledge support from the
This article documents a successful clinical trial of Synthesis of dolastatin 10 structural modifications. US National Institutes of Health grants P30 CA86862 and
checkpoint blockade in cancer. Anticancer Drug Des. 10, 529–544 (1995). P50 CA97274.
68. Robert, C. et al. Ipilimumab plus dacarbazine for 92. Kupchan, S. M. et al. Maytansine, a novel antileukemic
previously untreated metastatic melanoma. N. Engl. ansa macrolide from Maytenus ovatus. J. Am. Chem. Competing interests statement
J. Med. 364, 2517–2526 (2011). Soc. 94, 1354–1356 (1972). The author declares no competing interests.

370 | JUNE 2015 | VOLUME 15 www.nature.com/reviews/cancer

© 2015 Macmillan Publishers Limited. All rights reserved

You might also like