Antibody-Drug Conjugates Recent Advances in Conjug
Antibody-Drug Conjugates Recent Advances in Conjug
Antibody-Drug Conjugates Recent Advances in Conjug
REVIEW
Antibody-drug conjugates: recent advances
in conjugation and linker chemistries
Kyoji Tsuchikama& , Zhiqiang An
Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science
Center at Houston, Houston, TX 77054, USA
& Correspondence: Kyoji.Tsuchikama@uth.tmc.edu (K. Tsuchikama)
Received July 5, 2016 Accepted August 6, 2016
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
REVIEW Kyoji Tsuchikama, Zhiqiang An
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
Recent ADC conjugation and linker chemistries REVIEW
A B
Conjugation site
lysine coupling, cysteine alkylation,
enzymatic reaction, etc.
(6)
(1) Cell death
Binding to
mAb cell-surface
humanized or Linker antigen
fully human cleavable or Payload
non-cleavable antimitotic agent
(5)
DNA or microtubule disruption
Key factors
- High potency - High cancer cell specificity (2)
- Low immunogenicity - Long circulating life Endocytosis of
- Low cytotoxicity to off-target cells ADC-antigen
complex
(4)
Release of
(3) active payload
Lysosomal degradation
been explored for ADC payloads: duocarmycins, maytansinoid-based ADCs and adverse toxicity (Drake and
pyrrolobenzodiazepine dimers (PBDs), amanitins, and tubu- Rabuka, 2015). Therefore, it is important to identify ADC
lysin analogs are such examples (Chari et al., 2014; Perez linkers with optimal linker stability for each combination of
et al., 2014). antigen, target tumor type, and payload. (2) At the same
time, the linker needs to possess the ability to be rapidly
cleaved and to release free and toxic payload once the ADC
THE CONJUGATION AND LINKER CHEMISTRIES
is internalized into the target tumor cell. (3) Another property
FOR ADC
to be considered in the linker design is hydrophobicity.
Though it is important to select optimal target-specific anti- Hydrophobic linkers coupled with hydrophobic payloads
bodies and potent payloads based on type of cancer cells, often promote aggregation of ADC molecules. For example,
the conjugation and linker chemistries are also crucial King and co-workers observed non-covalent dimerization of
components for successful construction of an ADC and the the monoclonal antibody BR96 conjugated with doxorubicin
major topics of this review. The linker moiety covalently through a multi-loading, hydrophobic dipeptide linker (King
tethers the antibody and payload components. Its molecular et al., 2002). Such molecules are unfavorable in the pursuit
design and properties are critical determinant factors for of therapeutically useful ADCs; aggregated proteins tend to
ADC efficacy in terms of pharmacokinetics (PK)/pharmaco- be rapidly sequestered in the liver and cleared by the retic-
dynamics (PD) and therapeutic window. To maximize these uloendothelial system, resulting in hepatotoxicity (Finbloom
parameters, various types of linkers have been developed et al., 1980). In addition, aggregated proteins are likely to
and evaluated in vitro and in vivo. Several criteria must be function as immunogenic substances, provoking undesired
met for successful ADC construction. (1) The linker needs to immune response during circulation in bloodstream. This
possess sufficient stability in plasma so that ADC molecules problem can be overcome by employing hydrophilic linkers
can circulate in the bloodstream and localize to the tumor containing negatively charged sulfonate groups (Zhao et al.,
site without premature cleavage. Instability of the linker 2011), polyethylene glycol (PEG) groups (Lyon et al., 2015),
causes premature liberation of the toxic payload and unde- or pyrophosphate diester groups (Kern et al., 2016).
sired damage to non-target healthy cells, which can lead to Based on the above-mentioned criteria, tremendous effort
systemic toxicity and adverse effects. However, a clinical has been put toward developing conjugation methods and
study revealed reverse correlation between linker stability of ADC linker structures. Chemical conjugation and enzymatic
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
REVIEW Kyoji Tsuchikama, Zhiqiang An
O O
O S N
N O HO
H NHCO 2Me O
N S
N S NH N O
αCD33 mAb H H O O O O Cl
O
I O H αHER2 O N OMe
S O N O mAb
O O
HO O
O OMe OH O
OMe Et
O O O N
HO AcN H OH
MeO OMe
MeO
OH
Gemtuzumab ozogamicin (Mylotarg ®) Trastuzumab emtansine (Kadcyla ®)
for CD33-positive acute myelogenous leukemia treatment for Her2-positive breast cancer treatment
FDA-approved in 2000; withdrawn in 2010 FDA-approved in 2013
Et
O O OH
H H
O N N N
O O O N N
S H
N N O OMe O OMe O
N N
O H H
αCD30 mAb O
Protein & Cell
Figure 2. Structures of FDA-approved ADCs: Mylotarg®, Adcetris®, and Kadcyla® (blue: linker, red: payload).
conjugation are two methods for tethering the antibody and However, there are about 80 lysine residues on a typical
payload components that are currently in use. Linker struc- antibody and about 10 residues are chemically accessible
ture is categorized into two major classes based on the (Chari, 2008). Thus, this conjugation modality often gives
payload release mechanism: cleavable or non-cleavable multiple ADC species with variable DARs and conjugation
linker. Herein, we review modern conjugation methods and sites. In the case of a maytansinoid-type ADC, the average
ADC linker technologies in detail. DAR was 3.5–4 with distribution between 0–7 (Lazar et al.,
2005). As described above, DAR and its distribution critically
Chemical conjugation impact PK/PD and cytotoxicity of ADCs. Furthermore, some
lysine residues that are critical in antibody-antigen interac-
In ADC chemical conjugation, accessible amino acid resi-
tions may be modified, resulting in reduced binding affinity.
dues on the surface of the antibody undergo a controlled
As such, heterogeneous mixtures of ADCs constructed
reaction with a reaction handle installed on the linker.
using this conjugation method could potentially lead to a
Depending on the chemical conjugation method selected,
poor therapeutic index. While achievable as seen in the
this process affords a mixture of ADC species with variable
FDA-approved Kadcyla® and clinically tested ADCs, the
Drug-Antibody Ratios (DARs) and tethering sites. In general,
lysine-based conjugation requires effort to develop repro-
a broad distribution of DAR can lead to reduced efficacy, and
ducible manufacturing processes ensuring controlled DAR
thus the distribution needs to be tightly controlled. High DAR
and distribution within a target range (typically 3–4 as a
can increase not only potency but also the risk of aggrega-
major species).
tion, clearance rate, and premature release of the toxic
payload during circulation. This risk can be reduced by
Cysteine coupling
employing hydrophilic, sufficiently stable linkers. Overall, it is
crucial to identify an optimal DAR value with a controlled Cysteine-based conjugation methods rely on a specific
distribution for each ADC that can maximize the balance of reaction between cysteine residues of the antibody and a
efficacy, tolerability, and cytotoxicity profiles. thiol-reactive functional group installed on the payload
(Fig. 4A). In general, antibodies do not possess free thiols,
and all cysteine residues form disulfide bonds. In human
Lysine amide coupling
IgG1, which is most commonly used in modern ADCs, there
Amide coupling is a major ADC conjugation method con- are 4 interchain and 12 intrachain disulfide bonds. The 4
necting a payload and solvent accessible lysine residues on interchain disulfides, which are generally not critical for
the antibody using linkers containing activated carboxylic structural stability of IgG1, can be selectively reduced under
acid esters (Fig. 3). Amide coupling of an amine and an mild conditions to give 2, 4, 6, or 8 free thiols while keeping
activated carboxylic acid is one of the most reliable, high- the 12 intrachain disulfides intact. Due to the limited number
yielding chemical conversions in organic synthesis. of conjugation sites and the distinct reactivity of the thiol
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
Recent ADC conjugation and linker chemistries REVIEW
O
-
O 3S
+ N O
NH 2 NH
O O Payload or another
O
reaction handle Average DAR: 3.5-4
DAR distribution: 0-7
Figure 3. Lysine amide coupling. An activated carboxylic acid moiety reacts with a lysine residue, which results in amide bond
linkage between mAb and the payload. Optimized conjugation conditions give an average drug-to-antibody ratio (DAR) value of 3.5–4
with distribution between 0–7.
group, cysteine-based conjugation is superior to lysine- recombinant HER2 (Kd = 0.1–0.5 nmol/L) comparable to that
based conjugation in terms of controlled DAR and hetero- of the parent trastuzumab. Another advantage of this method
geneity. As is the case with the lysine-based conjugation, is that the resulting aryl-cysteine conjugates are stable to-
this conjugation method was a major choice for ADC con- wards acids, bases, oxidants, and externally added thiols.
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
REVIEW Kyoji Tsuchikama, Zhiqiang An
A O
+ N O
SH Cys S
alkylation
O N
O
DAR distribution: 2, 4, 6, and 8
(pedominant at 2 using THIOMAB)
O
Br
B O N O
N
Br
S S S S O S S
Reduction of H H Cys
interchain disulfide rebridging
DAR: predominant at 4
Protein & Cell
C
PR3
+ X Pd
SH Cys-aryl S
coupling
X = Cl, Br, I, OTf Average DAR: 4.4
Figure 4. Cysteine coupling. (A) Maleimide alkylation. A maleimide moiety reacts with a reduced cysteine residue of a mAb
(distribution of DAR: 2, 4, 6, and 8 or predominant at 2 with THIOMAB technology). (B) Rebridging of interchain disulfide bonds. The
dibromo (or disulfonate) reagent reacts with the reduced interchain disulfides to provide rebridged mAbs (DAR: predominant at 4).
(C) Cysteine arylation using palladium complexes. Aryl-palladium complex reagents undergo aryl-thiol coupling, which affords mAbs
containing arylcysteines (average DAR: 4.4).
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
Recent ADC conjugation and linker chemistries REVIEW
A O O
O p-acetyl-Phe O N N
H 2N
+ O
Oxime
formation
B
Copper N N
catalyst N N
N3 N3 + N N
Click
chemistry
p-azidomethyl-Phe
or
N6-((2-azidoethoxy)carbonyl)-Lys
C N N N N
Figure 5. Non-natural amino acid incorporation by genetic engineering into mAbs and subsequent chemical conjugation.
(A) Oxime ligation. (B) Copper-catalyzed or (C) strain-promoted (copper-free) azide-alkyne cyclization. The site-specific conjugation
method gives a defined DAR value depending on the number of non-natural amino acid residues that are genetically incorporated.
content: >96%) showed no adverse effect on antibody is genetically incorporated, resulting in site-specific antibody-
binding to the counterpart antigens. Further, these conju- drug conjugation. Another advantage of this LLQG-specific
gates exerted in-vitro cell killing activities comparable to the bacterial transglutaminase is that conjugation sites can be
corresponding conjugates generated by traditional ADC flexibly laid by inserting this short peptide motif within the
conjugation methods, including the FDA-approved ADCs antibody structure. They demonstrated the potential of this
Adcetris® and Kadcyla®. This method can also be used for strategy by preparing two ADCs that showed tightly con-
site-specific conjugation of the single-chain variable frag- trolled DARs (∼1.9) and comparable cytotoxicity and tolera-
ment (scFv) derived from mAbs (Madej et al., 2012). bility profiles.
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
REVIEW Kyoji Tsuchikama, Zhiqiang An
A
Sortase
+ GGGGG
B Microbial
Q295 transglutaminase
H 2N NH 2 + H 2N HN NH
O O O O
deglycosylated mAb DAR: 1.8-2
C Gal Sial
GalT, SialT
O
N
Protein & Cell
NaIO4 H
H 2N H
O
N Average DAR: ~1.6
O
D
Endo-
glycosidase GalNAz
then N3 N3
GalT (Y289L)
N3 UDP
N N
N N
Strain-promoted N N
copper-free
click chemistry
DAR: 2
Figure 6. Site-specific (chemo)enzymatic conjugation. (A) Sortase-mediated conjugation. Sortase attaches oligoglycine-
functionalized linkers to LPETG tags on the mAb. (B) Microbial transglutaminase-mediated conjugation. The enzyme attaches an
ADC linker possessing a primary amine to Q295 of the heavy chain (DAR: 1.8–2, high homogeneity). (C) Conjugation using β-1,4-
galactosyltransferase (GalT) and α-2,6-sialyltransferase (SialT) (light green square: β-1,4-galactose, magenta circle: sialic acid). The
aldehyde groups installed react with alkoxyamine-functionalized linkers (average DAR: ∼1.6). (D) GlycoConnect technology using
endoglycosidase, galactosyltransferase, and N-azidoacetylgalactosamine (GalNAz, light blue square). The azide groups installed
react with strained cyclooctyne-functionalized linkers (DAR: 2, high homogeneity).
approach is to incorporate non-natural saccharides pos- this technology is that it gives consistent results regardless
sessing orthogonal reaction handles into the antibody. One of the heterogeneity of the N-Glycan forms, meaning that it
of the latest technologies based on this strategy is the Gly- can be used for any IgG isotypes with various N-glycosyla-
coConnect technology developed by van Delft and co- tion profiles.
workers (van Geel et al., 2015) (Fig. 6D). The glycan chain at
Asn297 was trimmed using the endoglycosidase Endo S2
Cleavable linkers
and then azide groups were introduced using a mutant
galactosyl transferase GalT(Y289L) and N-azidoacetyl- A major class of ADC linkers is the cleavable linker (Fig. 7).
galactosamine (GalNAz). The azide handles were used for a Cleavable linkers are designed to be cleaved by responding
strain-promoted click reaction with payloads, resulting in to an environmental difference between the extracellular and
stable and homogeneous ADCs with tightly controlled DARs intracellular environments (pH, redox potential, etc.) or by
(predominant at 2 in most cases). The biggest advantage of specific lysosomal enzymes. In most cases, the linkers in
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
Recent ADC conjugation and linker chemistries REVIEW
H O O O O N
N N H H
Acidic N
N N
O pH H H
O
O
Val-Cit H 2N
N NH 2
H
O O
HS HO
S Intracellular P P Sequential
S reduction O O O O lysosomal
HO HO processing
Figure 7. Cleavable linkers. (A) Hydrazone linker. This linker is cleaved in the acidic environment (i.e., endosome and lysosome).
this class are designed to release parental payload mole- Cathepsin B-responsive linker
cules after bond cleavage. Such traceless drug release
Cathepsin B is a lysosomal protease that is over-expressed
mechanisms allow researchers to estimate cytotoxic potency
in various cancer cells and involved in numerous oncogenic
of the conjugated payload based on known pharmacological
processes in humans (Gondi and Rao, 2013). Cathepsin B
parameters of the free payload.
has a relatively broad scope of substrate, but it preferentially
recognizes certain sequences such as phenylalanine-lysine
Hydrazone linker (Phe-Lys) and valine-citrulline (Val-Cit) and cleaves a pep-
tide bond on the C-terminal side of such sequences. In
Hydrazone, an acid-labile group, is used as a cleavable
particular, Val-Cit and Val-Ala linkers coupled with p-
linker that releases free drug through hydrolysis once an
aminobenzyloxycarbonyl (Val-Cit-PABC and Val-Ala-PABC)
ADC is transported to acidic endosomes (pH 5.0–6.0) and
are the most successful cleavable linkers for ADCs
lysosomes (pH about 4.8) (Fig. 7A). The chimeric antibody
(Dubowchik et al., 2002; Hartley, 2011) (Fig. 7B). Upon
BR96-doxorubicin conjugate (BR96-DOX) was developed
internalization through endocytosis and transportation to
with the hydrazone conjugation strategy. BR96-DOX was
lysosomes, cathepsin B selectively cleaves this linker and
advanced to a Phase II human clinical trial in metastatic
cytotoxic payloads are released from the ADC in a traceless
breast cancer (Tolcher et al., 1999). The toxicity profile of the
manner. The PABC moiety functions as a spacer between
conjugate was considerably improved compared to free
Val-Cit moiety and the payload, allowing cathepsin B to
doxorubicin administration. However, gastrointestinal toxicity
exhibit its full protease activity to the linker connected to a
was still prominent and clinical outcomes were not satisfying
bulky payload molecule such as doxorubicin (Dubowchik
due to its low tolerability. Another example is the anti-CD33
et al., 2002). This linker was used to construct the chimeric
antibody calicheamicin conjugate, Mylotarg® (Linenberger
anti-CD30 antibody-MMAE conjugate, or brentuximab
et al., 2001; Sievers et al., 2001). Mylotarg® showed
vedotin (Adcetris®) (Younes et al., 2010).
encouraging clinical results and was approved in 2000.
However, as mentioned earlier, it was withdrawn from the
Disulfide linker
market in 2010 due to a lack of clinically significant
improvement of patient outcome. Both unsuccessful ADCs Glutathione sensitive linker is another common cleavable
suffered from toxicities and low tolerability, which seems to linker (Fig. 7C). This strategy relies on the higher concen-
be attributed to lability of the hydrazone linker during circu- tration of reducing molecules such as glutathione in the
lation. Indeed, ADCs with the hydrazone linker undergo slow cytoplasm (1–10 mmol/L) (Wu et al., 2004) compared to the
hydrolysis under physiological conditions (pH 7.4, 37°C), extracellular environment (about 5 µmol/L in blood) (Mills
resulting in a slow release of the toxic payload (Laguzza and Lang, 1996). A disulfide bond is embedded within the
et al., 1989). linker and resists reductive cleavage in circulation. However,
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
REVIEW Kyoji Tsuchikama, Zhiqiang An
S
S O
O
NH N
NH N +
H 3N O
O Lysosomal
processing -
O
O O3C
Lys-payload
O
O S
S +
H 3N
N
N Lysosomal CO 2-
processing
O
O Cys-payload
Figure 8. Non-cleavable linker. The chemical stability of the non-cleavable linker withstands proteolytic degradation. Cytosolic/
lysosomal degradation of the mAb moiety liberates the payload molecule linked to an amino acid residue derived from the degraded
mAb.
Protein & Cell
upon internalization, abundant intracellular glutathione demonstrate the potential of the pyrophosphate diester linker
reductively cleaves the disulfide bond to release the free for the future development of therapeutically practical ADCs.
payload molecule. To further enhance stability during circu-
lation, methyl groups are often installed next to the disulfide
Non-cleavable Linkers
bond (Saito et al., 2003). This class of linker has been
employed in Mylotarg® (Sievers et al., 2001), and more Non-cleavable linkers consist of stable bonds that are
recently, in several maytansine-based candidates in clinical resistant to proteolytic degradation, ensuring greater stability
trials (Widdison et al., 2015). than that of cleavable linkers. Non-cleavable linkers rely on
complete degradation of the antibody component of ADC by
cytosolic and lysosomal proteases, which eventually liber-
Pyrophosphate diester linker
ates a payload molecule linked to an amino acid residue
Recently, Garbaccio and co-workers developed a novel derived from the degraded antibody (Fig. 8). As such, when
cleavable linker with a pyrophosphate diester structure coupled with a non-cleavable linker, the payload structure
(Fig. 7D) (Kern et al., 2016). This anionic linker has greater must be carefully selected and designed so that payload can
aqueous solubility than traditional linkers and excellent cir- exert comparable or even better anti-tumor potency in such a
culatory stability. Furthermore, upon internalization, the modified form. For that purpose, it may be necessary to
pyrophosphate diester gets promptly cleaved through the examine PK/PD and toxicity profiles of all possible metabo-
endosomal-lysosomal pathway to liberate unmodified pay- lites of ADCs with non-cleavable linkers. A successful
load molecules. The authors speculate that the pyrophos- example of ADCs using a non-cleavable linker is the
phate diester goes through a two-step enzymatic linker humanized anti-HER2 antibody-maytansine conjugate tras-
cleavage that releases a payload-monophosphate molecule tuzumab emtansine (T-DM1, or Kadcyla®) (LoRusso et al.,
and then a free payload, although the enzyme(s) involved in 2011; Verma et al., 2012).
this process have not yet been identified. With this encour-
aging result, they set out to construct conjugates of the anti-
CONCLUDING REMARKS AND OUTLOOK
human CD70 antibody and various glucocorticoids using this
linker. The ADCs constructed showed great stability in In this article, we have reviewed the concept and clinical
human plasma (intact in vitro up to 7 days) and fast linker potential of ADCs and various conjugation/linker strategies
cleavage and release of free payload molecules in lyso- for constructing this new class of molecules (Table 1).
somes. Interestingly, each conjugate released a free payload Compared to traditional small molecule-based chemother-
at different rates, depending on the substituent group prox- apy, well-designed ADCs have several distinct features and
imal to the pyrophosphate moiety. This result suggests that clinical advantages, including preferable PK/PD and biodis-
the rate of release could be fine-tuned by further structural tribution (which are generally similar to that of native IgGs),
modifications. In addition, one of the ADCs containing fluti- broader therapeutic window, and flexibility of molecular
casone propionate exerted remarkable potency (EC50: 0.37 customization. As exemplified in the successes of the FDA
nmol/L) in CD70-positive 786-O cells, comparable to free approved Adcetris® and Kadcyla®, this new therapeutic
fluticasone propionate (EC50: 0.25 nmol/L). These results modality has huge potential for anti-cancer therapy and has
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
Recent ADC conjugation and linker chemistries REVIEW
Table 1. Advantages and disadvantages of the conjugation and linker chemistries described
attracted a great deal of attention from researchers and optimization for clinical application and eventual failure in
clinicians. Indeed, significant advances have been made in clinical trials. To overcome these problems, current efforts
ADC technologies, with about 60 ADCs currently in clinical are directed toward developing novel stable linkers (with or
trials. This emerging molecular platform is expected to without a payload release mechanism) and site-specific
become mainstream in anti-cancer therapeutics in the near conjugation methods enabling construction of homogeneous
future. Despite its potential, further understanding biochem- ADCs. Further investigations along this line will provide
ical, immunological, pharmacological, and molecular aspects greater insights and sophisticated strategies from medicinal
of ADCs must be pursued to better design and develop chemistry and pharmacology standpoints, leading to inno-
effective ADCs. While choice of target antigens and pay- vative cancer therapeutics in the future.
loads is important, antibody-payload conjugation methods
and linker chemistry are also crucial elements for producing ACKNOWLEDGEMENTS
successful ADCs. In particular, instability of the linker and
We gratefully acknowledge Dr. Yasuaki Anami and Dr. Yasuhiro
heterogeneity of the product (i.e., broad distribution of DARs)
Shimamoto for insightful discussions and proofreading the article.
often negatively impacts ADC efficacy and therapeutic win- We thank Dr. Georgina T. Salazar for editing the article. This work
dow, which often leads to difficulty or limitation in the
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
REVIEW Kyoji Tsuchikama, Zhiqiang An
was supported in part by the Welch Foundation Grant AU00024 and Bryant P, Pabst M, Badescu G, Bird M, McDowell W, Jamieson E,
the CPRIT grant RP150551. Swierkosz J, Jurlewicz K, Tommasi R, Henseleit K et al (2015)
In vitro and in vivo evaluation of cysteine rebridged trastuzumab–
ABBREVIATIONS MMAE antibody drug conjugates with defined drug-to-antibody
ratios. Mol Pharm 12:1872–1879
ADC, antibody-drug conjugate; CD, cluster of differentiation; DAR,
Bryden F, Maruani A, Savoie H, Chudasama V, Smith MEB, Caddick
drug-antibody ratio; DOX, doxorubicin; Fc, fragment crystallizable;
S, Boyle RW (2014) Regioselective and stoichiometrically con-
FDA, U.S. Food and Drug Administration; Gal, β-1,4-galactose;
trolled conjugation of photodynamic sensitizers to a HER2
GalNAz, N-azidoacetylgalactosamine; GalT, β-1,4-galactosyltrans-
targeting antibody fragment. Bioconjugate Chem 25:611–617
ferase; HER2, human epidermal growth factor receptor 2; hmAb,
Chari RVJ (2008) Targeted cancer therapy: conferring specificity to
humanized/human monoclonal antibody; IgG, immunoglobulin G;
cytotoxic drugs. Acc Chem Res 41:98–107
MMAE, monomethylauristatin E; MMAF, monomethylauristatin F;
Chari RVJ, Miller ML, Widdison WC (2014) Antibody-drug conju-
PABC, para-aminobenzyloxycarbonyl; PBD, pyrrolobenzodiazepine;
gates: an emerging concept in cancer therapy. Angew Chem Int
PEG, polyethylene glycol; PK/PD, pharmacokinetics/pharmacody-
Ed Engl 53:3796–3827
namics; scFv, single-chain variable fragment; Sial, sialic acid; SialT,
Chudasama V, Maruani A, Caddick S (2016) Recent advances in the
α-2,6-sialyltransferase.
construction of antibody-drug conjugates. Nat Chem 8:114–119
Damelin M, Zhong W, Myers J, Sapra P (2015) Evolving strategies
for target selection for antibody-drug conjugates. Pharm Res
Protein & Cell
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
Recent ADC conjugation and linker chemistries REVIEW
modification of antibodies by bacterial transglutaminase. Angew globulins from hamsters inoculated with such leukemia by
Chem Int Ed Engl 49:9995–9997 heterografts. C R Hebd Seances Acad Sci 246:1626–1628
Junutula JR, Raab H, Clark S, Bhakta S, Leipold DD, Weir S, Chen McCombs JR, Owen SC (2015) Antibody drug conjugates: design
Y, Simpson M, Tsai SP, Dennis MS et al (2008) Site-specific and selection of linker, payload and conjugation chemistry. AAPS
conjugation of a cytotoxic drug to an antibody improves the J 17:339–351
therapeutic index. Nat Biotechnol 26:925–932 Mills BJ, Lang CA (1996) Differential distribution of free and bound
Kern JC, Cancilla M, Dooney D, Kwasnjuk K, Zhang R, Beaumont glutathione and cyst(e)ine in human blood. Biochem Pharmacol
M, Figueroa I, Hsieh S, Liang L, Tomazela D et al (2016) 52:401–406
Discovery of pyrophosphate diesters as tunable, soluble, and Mullard A (2013) Maturing antibody-drug conjugate pipeline hits 30.
bioorthogonal linkers for site-specific antibody-drug conjugates. Nat Rev Drug Discov 12:329–332
J Am Chem Soc 138:1430–1445 Perez HL, Cardarelli PM, Deshpande S, Gangwar S (2014)
King HD, Dubowchik GM, Mastalerz H, Willner D, Hofstead SJ, Antibody–drug conjugates: current status and future directions.
Firestone RA, Lasch SJ, Trail PA (2002) Monoclonal antibody Drug Discov Today 19:869–881
conjugates of doxorubicin prepared with branched peptide Polson AG, Ho WY, Ramakrishnan V (2011) Investigational anti-
linkers: inhibition of aggregation by methoxytriethyleneglycol body-drug conjugates for hematological malignancies. Expert
chains. J Med Chem 45:4336–4343 Opin Investig Drugs 20:75–85
Kung Sutherland MS, Walter RB, Jeffrey SC, Burke PJ, Yu C, Popp MWL, Antos JM, Ploegh HL (2009) Site-specific protein
Kostner H, Stone I, Ryan MC, Sussman D, Lyon RP et al (2013) labeling via sortase-mediated transpeptidation. Curr Protoc
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn
REVIEW Kyoji Tsuchikama, Zhiqiang An
Randomized phase II study of BR96-doxorubicin conjugate in Witte MD, Cragnolini JJ, Dougan SK, Yoder NC, Popp MW, Ploegh
patients with metastatic breast cancer. J Clin Oncol 17:478–484 HL (2012) Preparation of unnatural N-to-N and C-to-C protein
van Geel R, Wijdeven MA, Heesbeen R, Verkade JMM, Wasiel AA, fusions. Proc Natl Acad Sci USA 109:11993–11998
van Berkel SS, van Delft FL (2015) Chemoenzymatic conjugation Wu G, Fang Y-Z, Yang S, Lupton JR, Turner ND (2004) Glutathione
of toxic payloads to the globally conserved n-glycan of native metabolism and its implications for health. J Nutr 134:489–492
mAbs provides homogeneous and highly efficacious antibody- Younes A, Bartlett NL, Leonard JP, Kennedy DA, Lynch CM, Sievers
drug conjugates. Bioconjugate Chem 26:2233–2242 EL, Forero-Torres A (2010) Brentuximab vedotin (SGN-35) for
VanBrunt MP, Shanebeck K, Caldwell Z, Johnson J, Thompson P, relapsed CD30-positive lymphomas. N Engl J Med 363:1812–
Martin T, Dong H, Li G, Xu H, D’Hooge F et al (2015) Genetically 1821
encoded azide containing amino acid in mammalian cells Zhao RY, Wilhelm SD, Audette C, Jones G, Leece BA, Lazar AC,
enables site-specific antibody-drug conjugates using click Goldmacher VS, Singh R, Kovtun Y, Widdison WC et al (2011)
cycloaddition chemistry. Bioconjugate Chem 26:2249–2260 Synthesis and evaluation of hydrophilic linkers for antibody-
Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, Pegram maytansinoid conjugates. J Med Chem 54:3606–3623
M, Oh D-Y, Diéras V, Guardino E et al (2012) Trastuzumab Zhou Q, Stefano JE, Manning C, Kyazike J, Chen B, Gianolio DA,
emtansine for HER2-positive advanced breast cancer. N Engl J Park A, Busch M, Bird J, Zheng X et al (2014) Site-specific
Med 367:1783–1791 antibody-drug conjugation through glycoengineering. Bioconju-
Vinogradova EV, Zhang C, Spokoyny AM, Pentelute BL, Buchwald gate Chem 25:510–520
SL (2015) Organometallic palladium reagents for cysteine Zimmerman ES, Heibeck TH, Gill A, Li X, Murray CJ, Madlansacay
Protein & Cell
bioconjugation. Nature 526:687–691 MR, Tran C, Uter NT, Yin G, Rivers PJ et al (2014) Production of
Widdison W, Wilhelm S, Veale K, Costoplus J, Jones G, Audette C, site-specific antibody-drug conjugates using optimized non-nat-
Leece B, Bartle L, Kovtun Y, Chari R (2015) Metabolites of ural amino acids in a cell-free expression system. Bioconjugate
antibody-maytansinoid conjugates: characteristics and in vitro Chem 25:351–361
potencies. Mol Pharm 12:1762–1773
© The Author(s) 2016. This article is published with open access at Springerlink.com and journal.hep.com.cn