Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect
ACS Publications. Most Trusted. Most Cited. Most Read
Site-Specific Conjugation of the Indolinobenzodiazepine DGN549 to Antibodies Affords Antibody–Drug Conjugates with an Improved Therapeutic Index as Compared with Lysine Conjugation
My Activity

Figure 1Loading Img
  • Open Access
Article

Site-Specific Conjugation of the Indolinobenzodiazepine DGN549 to Antibodies Affords Antibody–Drug Conjugates with an Improved Therapeutic Index as Compared with Lysine Conjugation
Click to copy article linkArticle link copied!

  • Chen Bai
    Chen Bai
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    More by Chen Bai
  • Emily E. Reid
    Emily E. Reid
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
  • Alan Wilhelm
    Alan Wilhelm
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    More by Alan Wilhelm
  • Manami Shizuka
    Manami Shizuka
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
  • Erin K. Maloney
    Erin K. Maloney
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
  • Rassol Laleau
    Rassol Laleau
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
  • Lauren Harvey
    Lauren Harvey
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
  • Katie E. Archer
    Katie E. Archer
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
  • Dilrukshi Vitharana
    Dilrukshi Vitharana
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
  • Sharlene Adams
    Sharlene Adams
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
  • Yelena Kovtun
    Yelena Kovtun
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
  • Michael L. Miller
    Michael L. Miller
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
  • Ravi Chari
    Ravi Chari
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    More by Ravi Chari
  • Thomas A. Keating
    Thomas A. Keating
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
  • Nicholas C. Yoder*
    Nicholas C. Yoder
    Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    *E-mail: [email protected]
Open PDFSupporting Information (1)

Bioconjugate Chemistry

Cite this: Bioconjugate Chem. 2020, 31, 1, 93–103
Click to copy citationCitation copied!
https://doi.org/10.1021/acs.bioconjchem.9b00777
Published November 20, 2019

Copyright © 2019 American Chemical Society. This publication is licensed under these Terms of Use.

Abstract

Click to copy section linkSection link copied!

Antibody–drug conjugates have elicited great interest recently as targeted chemotherapies for cancer. Recent preclinical and clinical data have continued to raise questions about optimizing the design of these complex therapeutics. Biochemical methods for site-specific antibody conjugation have been a design feature of recent clinical ADCs, and preclinical reports suggest that site-specifically conjugated ADCs generically offer improved therapeutic indices (i.e., the fold difference between efficacious and maximum tolerated doses). Here we present the results of a systematic preclinical comparison of ADCs embodying the DNA-alkylating linker-payload DGN549 generated with both heterogeneous lysine-directed and site-specific cysteine-directed conjugation chemistries. Importantly, the catabolites generated by each ADC are the same regardless of the conjugation format. In two different model systems evaluated, the site-specific ADC showed a therapeutic index benefit. However, the therapeutic index benefit is different in each case: both show evidence of improved tolerability, though with different magnitudes, and in one case significant efficacy improvement is also observed. These results support our contention that conjugation chemistry of ADCs is best evaluated in the context of a particular antibody, target, and linker-payload, and ideally across multiple disease models.

Copyright © 2019 American Chemical Society

Introduction

Click to copy section linkSection link copied!

Antibody–drug conjugates (ADCs) are a class of biotherapeutics for targeted cancer chemotherapy that consist of a cytotoxic payload chemically conjugated to a monoclonal antibody via a linker. (1,2) Upon binding to receptors expressed on the surface of cancer cells, the ADC is internalized as a complex with the receptor and processed to release cytotoxic catabolites that induce cell death.
By combining the tumor specificity of the antibody with the potent cytotoxicity of the payload, ADCs offer the potential to selectively deliver cytotoxic drugs to cancer cells while minimizing systemic toxicity. To date, five have been approved: brentuximab vedotin (3) for Hodgkin’s lymphoma and some non-Hodgkin’s lymphomas; ado-trastuzumab emtansine (2) for metastatic HER2+ breast cancer; gemtuzumab ozogamicin (4) for acute myeloid leukemia; inotuzumab ozogamicin (5) for acute lymphoblastic leukemia; and, recently, polatuzumab vedotin (6) as part of triple combination therapy for diffuse large B-cell lymphoma. However, the promise of ADCs has yet to be fully realized, particularly in solid tumor indications. Inadequate therapeutic indices remain a barrier to the development of many ADCs. (7)
Recent advances in payload, antibody, and linker technologies have yielded ADCs with improved in vivo pharmacologic properties. (8) In addition to these innovations, there has been much interest in developing novel conjugation methods to produce more active and better tolerated ADCs. Traditionally, ADCs have been synthesized by linkage of the payload to cysteines derived from reduction of interchain disulfides, or to lysine residues. These processes yield heterogeneous conjugates that are mixtures of species with different numbers of payload molecules linked at different sites on the antibody surface. Newer methods involve conjugation of the payload at defined sites on the antibody to give homogeneous conjugates with uniform drug to antibody ratio (DAR). (9−11) In the past decade, numerous studies comparing ADCs prepared by heterogeneous chemistries to those synthesized by novel site-specific methods have concluded that the latter exhibit improved in vivo pharmacologic properties. (12−21)
We recently conducted a direct comparison of ADCs that were synthesized by conjugating the same maytansinoid effector to the same antibody via either heterogeneous lysine conjugation or site-specific, engineered cysteine conjugation. (22) These ADCs were similarly potent in vitro and were shown to release very similar catabolites. In contrast to previous reports, the lysine-linked ADC was more efficacious than the site-specific ADC at matched payload doses in a mouse model of cervical cancer. This result suggests that the benefits of site-specific conjugation may not be general to all combinations of antibody, linker, and payload.
In this study, we further evaluate the advantages and disadvantages of site-specific conjugation by comparing heterogeneous lysine- and site-specific engineered cysteine-conjugated (termed CYSMAB) ADCs of the DNA alkylator DGN549. (23) Anticipating, in light of the findings from our maytansinoid study, that the results of such a comparison may depend on the exact system being studied, we tested conjugates of two different antibodies that bind to different cell surface receptors. Our results show that, unlike the maytansinoid case study, site-specific conjugation can lead to improved efficacy or tolerability in the context of DGN549 ADCs. However, the advantages do not appear to translate across different antibodies. We conclude that the choice of conjugation chemistry defies general rulemaking and must be considered in every specific case.

Results and Discussion

Click to copy section linkSection link copied!

Chemistry

The structures of DGN549-L (1a) and DGN549-C (1b) are shown in Figure 1. (23) Compound 1a, bearing an N-hydroxysuccinimide ester, was utilized for antibody conjugation at lysine residues. Compound 1b, bearing a maleimide, was utilized for antibody conjugation at engineered cysteine residues, and was generated from 1a via acylation of aminoethylmaleimide hydrochloride (SI Figure 1). Additional chemical synthetic details can be found in the Supporting Information.

Figure 1

Figure 1. (A) Structures of DGN549-L (1a), used for lysine-conjugation; and DGN549-C (1b), used for cysteine conjugation. (B) Structural representations of lysine-linked and CYSMAB ADCs.

Antibody Conjugation

Cysteine residues were introduced into the sequence of human IgG1s mAb1 and mAb2 via mutation as previously described. (22) Variants of FRα-targeting mAb1 with Cys mutations LC-V205C (14) and at HC-S442C (mAb1-C442) (24) were generated, as well as a double mutant containing both mutations (mAb1-C205-C442). Likewise, the CD123-targeting antibody mAb2 was used in a wild-type humanized IgG1 format (mAb2) or as an HC-S442C mutant (mAb2-C442). None of these mutants showed any reduction in antibody titer (compared to the wild-type antibodies) at the ∼10 mg scale.
Lysine conjugation of 1a to mAb1 and mAb2 was carried out as previously described to give ADCs 2a and 3a, respectively (Figure 1B). (23) ADCs site-specifically conjugated at HC-C442 are referred to as CYSMAB ADCs. In order to produce CYSMAB DGN549-C ADCs 2b and 3b (Figure 1C), global reduction and selective reoxidation of the Cys mutant antibodies were carried out as previously described. (22) The resulting partially reduced antibodies were conjugated with 1b in an aqueous buffer containing a small amount of dimethylacetamide (DMA), as well as a relatively large proportion of propylene glycol as nondenaturing cosolvents. Higher proportions of DMA supported conjugation with some antibodies, but other antibodies suffered from aggregate formation. As with lysine conjugation of 1a, free 1b could be readily purified away from the product ADC using disposable gel filtration columns. Preparation of a ∼4 DAR site-specific ADC via conjugation of mAb1-C205-C442 with 1b was carried out to give ADC 2c, using the same protocol used to produce ∼2 DAR ADCs 2b and 3b.

ADC Characterization

As previously reported, lysine conjugation of 1a could be titrated to give ADCs with DAR between 2.5 and 3.0, which was consistent across UV–vis, SEC, and LC-MS analyses. (23) Likewise, CYSMAB conjugation of 1b gave the expected DAR of approximately 2.0 by UV–vis spectroscopy. Analysis by LC-MS and HIC showed profiles consistent with conjugation largely at a single site, with some evidence for DAR 1 species and other ADC variants as reported by others. (12,14,25) Engineered cysteine conjugation to produce ADC 2c proceeded to an average DAR of 3.5. No attempt was made to optimize the conjugation reaction to increase the DAR. Unoptimized conjugation of 1b to a single mutant LC-V205C antibody achieved a DAR of 1.6, so we speculate that the cysteine in this position is simply less reactive to this linker-payload due to steric or electronic properties. (14,26)
Representative examples of lysine-directed and site-specific LC-MS profiles are shown in Figure 2, and a full data set, including the 3.5 DAR DGN549-C conjugate of mAb1-C205-C442 is shown in SI Figure 2. Both lysine-directed and site-specific conjugation gave the expected high monomer as measured by SEC (SI Table 1). A previous report showed site-specific conjugation to be an effective strategy for mitigating aggregation observed for heterogeneous pyrrolobenzodiazepine ADCs. (27) In some cases, site-specific conjugation may be useful to limit high DAR species which have undesirable biophysical or pharmacological properties. (28,29) In the present, and in some other (30) cases, there is little biophysical advantage to be gained from site-specific conjugation, since ADCs with favorable quality attributes can be produced by heterogeneous chemistries, at least at the DARs we report here. Sodium bisulfite was added to the formulation to reversibly sulfonate the imine functionality of DGN549 to improve its water solubility and thereby the biophysical properties of the ADCs, without compromising their potency. Preparation of ADC 2b without sodium bisulfite in the formulation was possible but required more extensive purification and the use of formulation buffer with high excipient concentrations (data not shown). The use of disposable gel filtration columns, sometimes with the additional step of dialysis, was enough to reduce free IGN impurities to the levels shown in SI Table 1 without the use of activated charcoal as has been reported for other hydrophobic ADC payloads. (27)

Figure 2

Figure 2. SEC-MS data for mAb1 ADCs: (A) ADC 2a and (B) ADC 2b.

HIC

HIC has historically been a useful method for ADC characterization and measuring ADC hydrophobicity. (31,32) Some literature reports have claimed a correlation between hydrophobicity and the pharmacokinetic properties of ADCs, (18,33−36) with the hypothesis that the same properties that lead to binding to a HIC column can lead to nonspecific uptake in animal tissues. Therefore, a method for analyzing DGN549 ADCs using HIC was developed, which required the use of a relatively less hydrophobic stationary phase than that used for auristatin (37) or maytansinoid (38) ADCs. In this method, the lysine-linked DGN549 ADCs eluted as a broad unresolved peak over ∼45 min of the HIC gradient, as expected due to the heterogeneity in both DAR species as well as conjugation sites. (38) In contrast, the CYSMAB DGN549 ADCs appeared much less heterogeneous by HIC, eluting largely in a single primary peak with additional minor peaks for DAR 1 and other apparent minor impurities. Interestingly, the primary CYSMAB ADC peak eluted almost at the very end of the HIC gradient spanned by the lysine conjugate, suggesting that conjugation at this site results in relatively hydrophobic ADCs, at least as measured by this assay (Figure 3). Finally, we note that the HIC analyses are conducted after the formulation of ADCs with sodium bisulfite, but show little evidence of heterogeneity that might be expected from reversible sulfonation of DGN549. This suggests that the sulfonation equilibrium lies to one side under the conditions of the experiment.

Figure 3

Figure 3. (A) HIC chromatograms of ADCs 2a and 2b. (B) HIC chromatograms of ADCs 3a and 3b.

In Vitro Catabolite Identification

Many previous studies comparing site-specific and heterogeneously conjugated ADCs have been confounded by differences in the expected catabolite profile of comparator ADCs. (13,15,16,39,40) In order to ensure that the catabolites generated by each method of DGN549 conjugation are similar, we subjected CYSMAB ADC 1b to the cell-based LC-MS catabolite identification assay previously described. (23) The only major catabolite species identified was the monoimine indolinobenzodiazepine aniline species 4 (Figure 4, SI Figure 3), which was previously identified as the catabolite of DGN549-L ADCs. (23)

Figure 4

Figure 4. Processing of ADCs 2a and 2b to release the same catabolite FGN849 (4).

In Vitro Binding Cytotoxicity

Binding of mAb1 and mAb2 to FRα-positive and CD123-positive cell lines, respectively, was confirmed using a flow cytometry-based assay. For all ADCs, binding EC50’s were comparable to those of the parent antibodies (SI Table 2) indicating that conjugation did not affect antigen binding. Cytotoxicity comparison of lysine-linked and CYSMAB DGN549 ADCs was carried out with both mAb1 and mAb2-derived ADCs in FRα-positive and CD123-positive cell lines, respectively, and the cytotoxic IC50’s are summarized in Table 1 and Table 2. In all cases, potency was strongly specific with respect to antibody–antigen binding, as shown with either antigen blocking by excess unconjugated targeting antibody, or by comparison with a nontargeted ADC of the same format. Consistent with what we (22,41) and others (12,18,30,35) have generally observed, in vitro potency of ADCs derived from the same antibody and linker-payload generally correlates with the DAR. Thus, the cytotoxic potency derived on a molar DGN549 basis (Table 1, Table 2, right-hand columns) generally appears similar across each cell line, though in some cases the CYSMAB ADCs 2b and 3b were slightly more potent than the lysine-linked comparators 2a and 3a. Bystander activity (cytotoxic activity of an ADC toward antigen-negative cells in the presence of antigen-positive cells) of ADCs 2a and 2b was evaluated in a mixed population of FRα-negative and FRα-positive cells. Both ADCs effected killing of FRα-negative cells at subnanomolar concentrations (SI Figure 4). This observation is consistent with the release of matched cell-permeable catabolites from the lysine-linked and CYSMAB ADCs, independent of conjugation chemistry.
Table 1. Cytotoxic IC50 Values as a Function of [ADC] and [DGN549] for mAb1 ADCs against FRα-Expressing Cells in the Absence or Presence of Antigen Blocking by Excess Unconjugated Targeting Antibody
  IC50 (M ADC)IC50 (M DGN549)
Cell LineADC– Block+ Block– Block+ Block
KB2a5 × 10–129 × 10–101 × 10–112 × 10–9
2b5 × 10–122 × 10–97 × 10–125 × 10–9
2c2 × 10–121 × 10–95 × 10–125 × 10–9
T47D2a2 × 10–119 × 10–96 × 10–113 × 10–8
2b1 × 10–111 × 10–83 × 10–112 × 10–8
NCI-H21102a4 × 10–112 × 10–91 × 10–105 × 10–9
2b4 × 10–112 × 10–89 × 10–114 × 10–8
Table 2. Cytotoxic IC50 Values as a Function of [ADC] and [DGN549] for mAb2 ADCs and Nontargeting Controls against CD123-Expressing Cells
Cell LineADCIC50 (M ADC)IC50 (M DGN549)
EOL-13a2 × 10–126 × 10–12
3b2 × 10–124 × 10–12
Non-Targeting Lysine-Linked ADC8 × 10–102 × 10–9
Non-Targeting CYSMAB ADC2 × 10–94 × 10–9
HNT-343a8 × 10–132 × 10–12
3b1 × 10–122 × 10–12
Non-Targeting Lysine-Linked ADC7 × 10–102 × 10–9
Non-Targeting CYSMAB ADC1 × 10–92 × 10–9
MV4-113a5 × 10–132 × 10–12
3b8 × 10–132 × 10–12
Non-Targeting Lysine-Linked ADC3 × 10–116 × 10–11
Non-Targeting CYSMAB ADC2 × 10–114 × 10–11

In Vivo Efficacy

Efficacy comparison of lysine-linked and site-specific DGN549 ADCs of both mAb1 and mAb2 was carried out in xenograft models that express their respective antigens. Prior literature comparing site-specific ADCs with heterogeneous comparators has reported a variety of outcomes. Moreover, interpretation of many of the studies is confounded by differences in catabolism of the comparator ADCs. (13,15,16,39,40) For studies with similar catabolite profiles, most have reported similar or marginally better efficacy for site-specific ADCs, provided the stability of the conjugation chemistry is adequate to maintain PK exposure of the resulting ADC. (12,14,21) By contrast, our previous investigation of peptide-linked maytansinoid ADCs found superior efficacy of a heterogeneous lysine-linked conjugate compared to site-specific cysteine-linked counterparts. (22) Finally, a recent survey of the literature has claimed that site-specific ADCs trend toward improved efficacy. (42) While some of the underlying published reports utilize ADCs that generate different catabolites, this article provides a physiologically based mathematical model that suggests that the decreased DAR leads to increased antibody doses, which in turn results in improvements in tumor delivery that more than compensate for the decrease in payload delivered per binding event.
We evaluated anti-FRα ADCs 2a and 2b in the NCI-H2110 mouse xenograft model of NSCLC (Figure 5, Table 3). The ADCs were dosed at equivalent quantities of the payload DGN549. At 3 μg/kg, both ADCs were minimally active with similar percent tumor growth inhibition (%T/C), while at the higher dose of 9 μg/kg, both ADCs induced durable complete regressions.

Figure 5

Figure 5. Activity of mAb1 ADCs in SCID mice bearing NCI-H2110 non-small cell lung cancer xenografts expressing FRα.

Table 3. Activity of mAb1 ADCs in SCID Mice Bearing NCI-H2110 Non-Small Cell Lung Cancer Xenografts Expressing FRα
 Dose Regressions
Treatmentμg DGN549/kgμg ADC/kgT/C Day 22 (%)PartialComplete
Vehicle---0/60/6
ADC 2a3160320/60/6
951016/66/6
ADC 2b3230390/60/6
968026/66/6
Anti-CD123 ADCs 3a and 3b were compared at equivalent payload doses in a disseminated model of AML derived from the cell line MOLM-13 (Figure 6, Table 4). In this survival model, both ADCs showed potent activity, with doses of 1 μg/kg of DGN549 or greater generating sustained lifespan increase in a majority of animals. Even at doses as low as 0.1 μg/kg, activity was observed, though it was discernibly improved for CYSMAB ADC 3b, showing a lifespan increase of 262% as compared with 59% for the equivalent dose of lysine-linked ADC 3a. Thus, in this case CYSMAB conjugation affords a significant increase in activity, while in the case of the FRα ADCs the two modes of conjugation give ADCs that appear similarly active.

Figure 6

Figure 6. Activity of mAb2 ADCs and nontargeting controls in SCID mice bearing Molm-13 acute myeloid leukemia xenografts expressing CD123.

Table 4. Activity of mAb2 ADCs and Non-Targeting Controls in SCID Mice Bearing Molm-13 Acute Myeloid Leukemia Xenografts Expressing CD123
 Dose   
Treatmentμg DGN549/kgμg ADC/kgMedian Survival (Days)Tumor Growth DelayIncreased Life Span (%)
Vehicle- 28--
ADC 3a0.15.844.516.559
0.317.4>152>124>443
1.058>152>124>443
Non-Targeting Lysine-Linked ADC1.05431311
ADC 3b0.17.6101.573.5262.5
0.322.8>152>124>443
1.076>152>124>443
Non-Targeting CYSMAB ADC1.06.826.5--

Mouse Tolerability

Previous reports have claimed that site-specific conjugation can improve the tolerability of ADCs. (13,18,19,40) Although these data have primarily been established by comparison with auristatin ADCs conjugated via interchain disulfides, we reasoned that this benefit might be observed with other payload classes as well. Interestingly, for ADCs of mAb1, the CYSMAB ADC 2b was approximately 2-fold better tolerated in mouse on a DGN549 payload basis than the heterogeneous ADC 2a (Figure 7). In the case of mAb2, the MTD for both ADCs was ∼100 μg/kg (Figure 8). However, comparison of the 125 μg/kg doses shows that for site-specific ADC 3b, the majority of animals recovered and gained weight following the acute toxicity post-dose, while for 3a the majority of animals had to be euthanized. So, while the MTD was formally the same, the tolerability of 3b appears slightly improved over 3a. Two potential hypotheses for the improved tolerability of site-specific CYSMAB DGN549 ADCs 2b and 3b over their lysine-linked comparators are (1) the improved pharmacologic properties of the conjugate made only at HC-C442 relative to the amalgamated properties of all the lysine-linked species; and (2) the decreased DAR of the CYSMAB ADCs eliminating the population of higher DAR, potentially more toxic species within the heterogeneous ADCs. In order to examine these hypotheses further, we tested the MTD of the ∼4 DAR site-specific ADC 2c in mice. The MTD of 125 μg/kg (Figure 9) is intermediate between DAR 2 CYSMAB 2b and heterogeneous DAR 2.5 2a, suggesting that both the site-specificity and the lower DAR of 2b contribute to its greater tolerability in mouse. Neither mAb1 nor mAb2 shows significant cross-reactivity to their respective mouse antigens, indicating that antigen-mediated effects do not play a role in toxicity of the ADCs. Thus, the fact that site-specific ADC 3b shows a lower tolerability advantage over heterogeneous conjugate 3a than does site-specific 2b over 2a suggests that antibody-specific factors significantly influence the in vivo outcomes. While this result may appear surprising due to the conserved sequence and structural features of human IgG1s such as mAb1 and mAb2, it has been observed that many biophysical factors can lead to disparate PK properties and tolerability of monoclonal Abs. (43)

Figure 7

Figure 7. Tolerability of mAb1 ADCs in non-tumor-bearing CD-1 mice. (A) ADC 2a and (B) ADC 2b

Figure 8

Figure 8. Tolerability of mAb2 ADCs in non tumor-bearing CD-1 mice: (A) ADC 3a and (B) ADC 3b.

Figure 9

Figure 9. Tolerability of ADC 2c in non tumor-bearing CD-1 mice.

Mouse Pharmacokinetics

Previous reports have claimed that site-specific ADCs can exhibit increased pharmacokinetic (PK) exposure because of reduced hydrophobicity. This has been linked to improved tolerability, with the rationale that increased ADC exposure reduces payload delivery to sensitive tissues. (12,18,21,33,34,36) For site-specific ADCs, site-specific conjugation near the C-terminus of the IgG1 heavy chain has been reported to be problematic with regard to both hydrophobicity and exposure. (33,35) ADC 2b therefore presented a bit of an enigma, being better tolerated in mouse but appearing more hydrophobic in toto than ADC 2a. We therefore assessed the PK of ADCs 2a and 2b in non-tumor-bearing CD1 mice (Figure 10, Table 5).

Figure 10

Figure 10. Pharmacokinetics of mAb1 ADCs 2a and 2b in non-tumor-bearing CD-1 mice, assayed by ELISA methods for total antibody and conjugated antibody.

Table 5. Pharmacokinetic Parameters for mAb1 ADCs in Non-Tumor-Bearing CD-1 Mice
ADCCmax (μg/mL)AUC0-∞ (h·μg/mL)Cl (mL/h/kg)t1/2 (h/days)t1/2 (days)Vss (mL/kg)
2aTotal Antibody40.346550.542219.2163
2aConjugated Antibody45.355780.452329.7142
2bTotal Antibody52.1107950.2338315.6124
2bConjugated Antibody65.491590.2732213.4121
Despite its apparent hydrophobicity as read out by HIC, site-specific ADC 2b exhibits 1.6-fold greater exposure as well as 1.7-fold slower clearance in mice than heterogeneous ADC 2a. Because the clearance as read out by total antibody ELISA is slower for 2b, we speculate that there is some pharmacological advantage of ADC 2b, despite its higher apparent hydrophobicity (Figure 3, above). It is possible that DGN549 conjugation at the relatively solvent-exposed HC-C442 position may enable noncovalent binding of albumin to the conjugated DGN (44) effectively solubilizing the ADC. Alternatively, the HIC assay may not completely capture the ADC properties relevant to pharmacological disposition. Finally, it should be noted that several prior reports (21,33,35,36) evaluated PK in rat, rather than in mouse as presented here. One of the prior reports (33) observed significantly more rapid clearance in rat than in mouse for a site-specific ADC, so we cannot rule out the possibility of species-specific pharmacokinetic effects playing a role here.
Cysteine-maleimide-linked ADCs have been shown to undergo degradation in vivo via elimination of the maleimide which is subsequently scavenged by thiols present in the biological milieu. (45) Linkage at some cysteine mutation sites appear to be particularly prone to this “retro-Michael” mechanism of instability, leading to rapid formation of unconjugated antibody. (14,26,46) Although the ELISA method used to determine [ADC] is not DAR-sensitive, the accumulation of unconjugated antibody can be inferred from the difference between the total antibody and ADC ELISA readouts. By this metric, conjugation at HC-C442 appears quite stable chemically. In order to validate this further, we examined the intact LC-MS of ADC 2b affinity purified from mouse plasma 3 days after administration. The resulting LC-MS profile (SI Figure 5) showed only minor increase in the thiol adducts to the ADC, providing additional evidence for chemical stability of maleimide conjugation at HC-C442.

Conclusions

Click to copy section linkSection link copied!

Although some ADCs have had clinical success in treating cancer, development of these therapies has also been notoriously challenging. It is clear that the rules for generating successful therapies with this class of cancer therapeutic are not completely understood. (8,47) While publications in the field have emphasized the benefits of second-generation site-specific conjugation technologies versus the earlier heterogeneous conjugation chemistries used, both our prior report (22) and this work suggest that the benefits are highly context dependent.
In our previous report, site-specific 2 DAR conjugation reduced the efficacy of a maytansinoid ADC compared with a lysine-conjugated 3.5 DAR ADC with a similar catabolite profile. In the present case, site-specific conjugation of a highly potent, the peptide linked IGN payload gave an improvement in preclinical therapeutic index as compared with lysine conjugation. Both site-specific ADCs were better tolerated in mouse, and one antibody/antigen system also showed improved efficacy. The greater hydrophobicity of the IGN payload may contribute to the benefits observed with site-specific conjugation, although a uniform 4 DAR site-specific ADC was still somewhat better tolerated than the lysine linked with an average DAR of 2.5. Moreover, the ADC hydrophobicity as measured by HIC did not appear to correlate with either therapeutic index or PK exposure.
As long as site-specific conjugation does not decrease therapeutic index of a given ADC, it remains of interest for ADC design. Controlled, uniform DAR can enable higher antibody dosing, which may improve ADC targeting either by improved transport and delivery through tumor vasculature or by saturating a normal tissue antigen sink, (42,48,49) as demonstrated both preclinically and in a clinical program targeting MUC16. (47) While the present case clouds the relationship between PK exposure and ADC hydrophobicity, prior examples suggest that reduction in hydrophobicity via site-specific conjugation, or using other technologies, may help improve pharmacological properties. For example, while 8 DAR interchain cysteine-directed conjugates of the widely studied mc-Val-Cit-PAB-MMAE linker-payload show poor PK properties, (28) second-generation auristatin payloads can be used in this conjugation format. (34,36) Notably, camptothecin ADCs (50,51) can also be deployed as 8 DAR interchain cysteine conjugates, likely due to their modest hydrophobicity, and have seen encouraging clinical activity as such.
Our results reinforce our general conviction that site-specific conjugation is a useful tool to be applied in the development of new conjugate therapies. However, rather than providing a generic benefit, its use must be carefully considered in combination with each individual target, linker-payload, antibody, and disease in order to make an optimally safe and effective therapy.

Materials and Methods

Click to copy section linkSection link copied!

Preparation and Characterization of ADCs

Antibodies

Human IgG1 antibodies against FRα and CD123 were produced at ImmunoGen, Inc. IgG1 antibodies against Kunitz trypsin inhibitor (KTI), used as nontargeting controls that do not bind any human antigen, were also produced at ImmunoGen, Inc. Antibody mutants with engineered cysteines were produced by modifying the heavy chain gene sequence to give a serine to cysteine mutation at position 442 (EU numbering).

DGN549 Payloads

Synthesis of a lysine-reactive form of DGN549-L (1a) was previously described. (52) Synthesis of a cysteine-reactive form of DGN549-C (1b) from 1a is described in the Supporting Information.

Synthesis of Lysine-Linked ADCs

DGN549-L was reversibly sulfonated at the imine by treatment with a 5-fold molar excess of sodium bisulfite in a 9:1 mixture of N,N-dimethylacetamide (DMA) and 50 mM sodium succinate, pH 5.0. The sulfonated DGN549 was added in a 3–5-fold molar excess to a solution of antibody in 15 mM 4-(2-hydroxyethyl)-1-piperazine ethanesulfonic acid (HEPES) pH 8.5 and 10% DMA. The reaction was allowed to proceed for 3–4 h at 25 °C. The conjugate was purified into 10 mM succinate, 50 mM sodium chloride, 8.5% sucrose, 0.01% Tween-20, and 50 μM sodium bisulfite pH 6.2 formulation buffer using Sephadex G25 desalting columns.

Synthesis of CYSMAB ADCs

Antibodies bearing engineered cysteines at position 442 in the heavy chain in phosphate-buffered saline (PBS) pH 7.4 were treated with 50 mol equiv of tris(2-carboxyethyl)phosphine and incubated for 1 h at 37 °C to reduce the solvent-accessible cysteines. The reduced interchain disulfides were reoxidized by treating the crude reduced antibody with 100 mol equiv of dehydroascorbic acid for 90 min at 25 °C to afford an antibody intermediate with the two engineered cysteines in reduced state. This intermediate was purified into PBS pH 6.0 with 5 mM N,N,N′,N′-ethylenediaminetetraacetic acid (EDTA) by gel filtration using Sephadex G25 columns. Conjugation was then effected by adding propylene glycol and DGN549-C as a stock solution in DMA to give a reaction mixture with a final solvent composition of 2% v/v DMA and 38% v/v propylene glycol in PBS pH 6.0 containing 5 mM EDTA. The reaction was allowed to proceed overnight at 25 °C, then purified into 20 mM histidine, 50 mM sodium chloride, 8.5% sucrose, 0.01% Tween-20, and 50 μM sodium bisulfite pH 6.2 formulation buffer using Sephadex G25 desalting columns, and then dialyzed against the same buffer using a membrane with 10 kDa molecular weight cutoff to remove residual unconjugated DGN549 and any other side products.

Characterization of ADCs

Concentrations of antibody and DGN549, and DAR, were determined by UV–vis spectroscopy on an Agilent 8543 spectrophotometer. Percent monomeric ADC was determined by size exclusion chromatography (SEC) using a TSKGgel G3000SWXL column (Tosoh Bioscience #08541). ADCs were eluted isocratically with 400 mM sodium perchlorate, 50 mM sodium phosphate pH 7.0, 5% isopropanol mobile phase run at 0.5 mL/min for 30 min. Unconjugated DGN549 was assayed by tandem size exclusion and reversed phase chromatography using an Acquity UPLC (Waters), equipped with a Protein BEH SEC guard column (Waters Corporation #186005793) and an Acquity UPLC BEH C18 column (Waters Corporation #176000863) run in series. Mobile phases were 0.1% trifluoroacetic acid in water (mobile phase A) and 0.1% trifluoroacetic acid in acetonitrile (mobile phase B). The flow rate was 0.35 mL/min and the column temperature was set to 30 °C. The ADC was eluted from the SEC column isocratically over 2.5 min with 30% mobile phase B. Unconjugated DGN549 species retained in the SEC column were then separated on the C18 column on a linear gradient of 30% to 80% mobile phase B from 2.5 to 8.0 min and quantified based on a DGN549 standard curve. Intact ADCs were analyzed by SEC coupled to mass spectrometry as previously described. (53) Hydrophobic interaction chromatography (HIC) was performed on a Proteomix HIC Ethyl NP5 column (Sepax Technologies #432NP5-7810) using mobile phases consisting of 1.5 M ammonium sulfate, 50 mM potassium phosphate pH 5.8 (mobile phase A), and 50 mM potassium phosphate pH 6.6 with 25% isopropanol (mobile phase B). Analytes were eluted on a gradient of 0% to 100% B over 45 min at a flow rate of 1.0 mL/min.

In Vitro Experiments

Binding

Binding of mAb1 ADCs to FRα on T47D cells and of mAb2 ADCs to CD123 on HNT-34 cells wereconfirmed by flow cytometry as previously described. (29)

Cytotoxicity

Cytotoxic potencies were assayed by treating tumor cells in 96-well plates (1000 to 5000 cells per well, depending on the cell line) in appropriate culture media with ADC over a range concentrations for 5 days at 37 °C and determining cell viability using the WST-8 assay (Dojindo Molecular Technologies) as previously described. (54) Data were fit using Prism (GraphPad, San Diego CA), with independent IC50 values generated for ADC and DGN549 concentrations.

Bystander Cytotoxicity Assay

Bystander activity of mAb1 ADCs was determined by treating FRα-positive 300.19 cells (generated by transfection with cDNA encoding the human FRα gene), FRα-negative parental 300.19 cells, or a mixed population of transfected and parental cells with ADC over a range of concentrations such that the ADC was nontoxic to the antigen-negative cells, but cytotoxic to the antigen-positive cells. Cell viability after 4 days at 37 °C was determined by the CellTiter-Glo (Promega) luminescent assay. (55)

Identification of Catabolites from mAb1-DGN549 CYSMAB ADC

Catabolites from mAb1-DGN549 CYSMAB ADC were identified as previously described. (23) Briefly, KB cells expressing FRα were treated with a saturating concentration of ADC for 24 h at 37 °C. Catabolites in the media were concentrated by binding to an anti-IGN antibody (ImmuoGen, Inc.) immobilized on Protein A resin, extracted into acetone, and dried in vacuo. The residue was redissolved in 80:20 water:acetonitrile and analyzed using a Dionex UltiMate 3000 UPLC (Thermo Fisher, USA) fitted with a reverse phase column (BEH C8, 1.7 μm, 100 mm × 2.1 mm, Waters) coupled to a Q-Exactive high resolution mass spectrometer (Thermo Fisher, USA). A 30 min gradient elution method was applied with a flow rate of 0.35 mL/min (mobile phase A: 0.1% formic acid in water; mobile phase B: 0.1% formic acid in acetonitrile; gradient: 20% B for 1 min, 20–80% B in 19 min, 80–100% B in 0.4 min, 100% B for 4.6 min, 100–20% B in 2 min and 20% B for 3 min) and a column temperature of 30 °C. The MS method was set at positive ion mode with full MS scan. Extracted ion chromatograms were generated for a set of plausible catabolites, and signal above an established confidence threshold was considered positive for presence of a given mass. Positive hits were confirmed by comparison with a spiked synthetic standard.

In Vivo Studies

Efficacy of mAb1 ADCs in NCI-H2110 Xenografts

Female CB.17 SCID mice were inoculated with 1 × 107 NCI-H2110 cells in 0.1 mL serum free medium + Matrigel (1:1) by subcutaneous injection in the area on the right hind flank. The mice were randomized by tumor volume into groups of 6 and single doses of ADC were administered intravenously on day 8 post-inoculation when the mean tumor volume reached ∼100 mm3. Tumor size was measured two times per week in three dimensions using a caliper. The tumor volume was expressed in mm3 using the formula V = Length × Width × Height × 1/2. Percent tumor growth inhibition (% T/C), defined as T/C × 100 where T is the median tumor volume in treated mice and C is the median tumor volume in the control group, was calculated on day 22 post-inoculation when the median tumor volume in the control group reached 1000 mm3. A mouse was determined to have a partial regression (PR) when tumor volume was reduced by 50% or greater and complete tumor regression (CR) when no palpable tumor could be detected.

Efficacy of mAb2 ADCs in Molm-13 Xenografts

Female athymic nude mice were inoculated with 1 × 107 Molm-13 tumor cells suspended in 0.1 mL serum-free culture media by intravenous injection into the tail vein. The mice were randomized by body weight into groups of 10 and single doses of ADC were administered intravenously on day 7 post-inoculation. To block Fc receptors, single 400 mg/kg doses of KTI-targeting IgG1 antibody (this antibody binds human Fc receptor but does not bind any human antigen) were administered intraperitoneally 24 h before ADC dosing. To maintain Fc receptor blockade, single intraperitoneal injections of 100 mg/kg KTI-targeting antibody were then given on days 4 and 9 after ADC administration. Animals were monitored and sacrificed if (1) body weight dropped by more than 20% of initial body weight at any time during the study, (2) one or both hind legs were paralyzed, (3) tumor growth occurred anywhere on the animal, or (4) the animal was too sick to reach food and water. Mice found dead were recorded on the day of discovery. Tumor growth delay was defined as T – C, where T is median survival time in the treated group and C is the median survival time in the control group. Percent increased life span was defined as (T – C)/C × 100.

Tolerability

Non-tumor-bearing CD-1 mice were randomized by body weight into groups of 8, and single doses of ADC were administered intravenously. Mice were weighed once daily for the duration of the study. Toxicity was assessed based on body weight and observation. Results were reported as percent body weight change relative to the start of treatment. The maximum tolerated dose (MTD) was defined as the maximum dose at which no animals died or were euthanized due to >20% body weight loss.

Pharmacokinetics of mAb1 ADCs

Non-tumor-bearing CD-1 mice were randomized by body weight and single 2.5 mg/kg doses of ADC were administered intravenously. Plasma samples were collected at 2 and 30 min; 2, 4, and 8 h; and 1, 2, 3, 5, 7, 10, 14, 21, and 28 days post-administration. ADC concentrations in plasma were assayed by two ELISA methods, one specific for the antibody component and another specific for the DGN549 component. Costar high-binding plates (Corning) were coated with goat anti-human IgG1 antibody (Jackson ImmunoResearch) for capturing total antibody or murine anti-IGN antibody (ImmunoGen Inc.) for capturing DGN549-conjugated antibody. After blocking with casein, plasma samples were added and captured ADC was detected with anti-human IgG Fc antibody conjugated to horseradish peroxidase (Jackson ImmunoResearch) using 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) as the substrate. Concentrations were determined from a standard curve generated using purified ADC and reported as the mean of samples from three individual mice. Pharmacokinetic parameters were calculated by noncompartmental analysis using Phoenix WinNonlin software (Certara).

Supporting Information

Click to copy section linkSection link copied!

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.bioconjchem.9b00777.

  • Detailed experimental procedures for synthesis of compound 1b, supplementary characterization data for ADCs, and supplementary methods (PDF)

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

Click to copy section linkSection link copied!

  • Corresponding Author
  • Authors
    • Chen Bai - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    • Emily E. Reid - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    • Alan Wilhelm - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    • Manami Shizuka - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesPresent Address: Pharmaron,404 Wyman Street, Waltham, MA 02451
    • Erin K. Maloney - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    • Rassol Laleau - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    • Lauren Harvey - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesPresent Address: Kaleido Biosciences, 65 Hayden Avenue, Lexington, MA 02421
    • Katie E. Archer - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    • Dilrukshi Vitharana - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesPresent Address: Anokion, 50 Hampshire Street, Cambridge, MA 02139
    • Sharlene Adams - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    • Yelena Kovtun - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    • Michael L. Miller - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesOrcidhttp://orcid.org/0000-0003-3762-3319
    • Ravi Chari - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
    • Thomas A. Keating - Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
  • Notes
    The authors declare no competing financial interest.

References

Click to copy section linkSection link copied!

This article references 55 other publications.

  1. 1
    Chari, R. V. J., Miller, M. L., and Widdison, W. C. (2014) Antibody–Drug Conjugates: An Emerging Concept in Cancer Therapy. Angew. Chem., Int. Ed. 53 (15), 37963827,  DOI: 10.1002/anie.201307628
  2. 2
    Lambert, J. M. and Chari, R. V. (2014) Ado-trastuzumab Emtansine (T-DM1): an antibody-drug conjugate (ADC) for HER2-positive breast cancer. J. Med. Chem. 57 (16), 69496964,  DOI: 10.1021/jm500766w
  3. 3
    Senter, P. D. and Sievers, E. L. (2012) The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nat. Biotechnol. 30 (7), 631637,  DOI: 10.1038/nbt.2289
  4. 4
    Appelbaum, F. R. and Bernstein, I. D. (2017) Gemtuzumab ozogamicin for acute myeloid leukemia. Blood 130 (22), 23732376,  DOI: 10.1182/blood-2017-09-797712
  5. 5
    Yurkiewicz, I. R., Muffly, L., and Liedtke, M. (2018) Inotuzumab ozogamicin: a CD22 mAb-drug conjugate for adult relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Drug Des., Dev. Ther. 12, 22932300,  DOI: 10.2147/DDDT.S150317
  6. 6
    Dornan, D., Bennett, F., Chen, Y., Dennis, M., Eaton, D., Elkins, K., French, D., Go, M. A., Jack, A., Junutula, J. R. (2009) Therapeutic potential of an anti-CD79b antibody-drug conjugate, anti-CD79b-vc-MMAE, for the treatment of non-Hodgkin lymphoma. Blood 114 (13), 27212729,  DOI: 10.1182/blood-2009-02-205500
  7. 7
    Donaghy, H. (2016) Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugates. MAbs 8 (4), 659671,  DOI: 10.1080/19420862.2016.1156829
  8. 8
    Chari, R. V. J. (2016) Expanding the Reach of Antibody-Drug Conjugates. ACS Med. Chem. Lett. 7 (11), 974976,  DOI: 10.1021/acsmedchemlett.6b00312
  9. 9
    Behrens, C. R. and Liu, B. (2014) Methods for site-specific drug conjugation to antibodies. MAbs 6 (1), 4653,  DOI: 10.4161/mabs.26632
  10. 10
    Agarwal, P. and Bertozzi, C. R. (2015) Site-specific antibody-drug conjugates: the nexus of bioorthogonal chemistry, protein engineering, and drug development. Bioconjugate Chem. 26 (2), 176192,  DOI: 10.1021/bc5004982
  11. 11
    Panowski, S., Bhakta, S., Raab, H., Polakis, P., and Junutula, J. R. (2014) Site-specific antibody drug conjugates for cancer therapy. MAbs 6 (1), 3445,  DOI: 10.4161/mabs.27022
  12. 12
    Junutula, J. R., Raab, H., Clark, S., Bhakta, S., Leipold, D. D., Weir, S., Chen, Y., Simpson, M., Tsai, S. P., Dennis, M. S. (2008) Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat. Biotechnol. 26 (8), 925932,  DOI: 10.1038/nbt.1480
  13. 13
    Junutula, J. R., Flagella, K. M., Graham, R. A., Parsons, K. L., Ha, E., Raab, H., Bhakta, S., Nguyen, T., Dugger, D. L., Li, G. (2010) Engineered Thio-Trastuzumab-DM1 Conjugate with an Improved Therapeutic Index to Target Human Epidermal Growth Factor Receptor 2–Positive Breast Cancer. Clin. Cancer Res. 16 (19), 47694778,  DOI: 10.1158/1078-0432.CCR-10-0987
  14. 14
    Shen, B. Q., Xu, K., Liu, L., Raab, H., Bhakta, S., Kenrick, M., Parsons-Reponte, K. L., Tien, J., Yu, S. F., Mai, E. (2012) Conjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugates. Nat. Biotechnol. 30 (2), 184189,  DOI: 10.1038/nbt.2108
  15. 15
    Pillow, T. H., Tien, J., Parsons-Reponte, K. L., Bhakta, S., Li, H., Staben, L. R., Li, G., Chuh, J., Fourie-O’Donohue, A., Darwish, M. (2014) Site-specific trastuzumab maytansinoid antibody-drug conjugates with improved therapeutic activity through linker and antibody engineering. J. Med. Chem. 57 (19), 78907899,  DOI: 10.1021/jm500552c
  16. 16
    Tian, F., Lu, Y., Manibusan, A., Sellers, A., Tran, H., Sun, Y., Phuong, T., Barnett, R., Hehli, B., Song, F. (2014) A general approach to site-specific antibody drug conjugates. Proc. Natl. Acad. Sci. U. S. A. 111 (5), 17661771,  DOI: 10.1073/pnas.1321237111
  17. 17
    Jackson, D., Atkinson, J., Guevara, C. I., Zhang, C., Kery, V., Moon, S. J., Virata, C., Yang, P., Lowe, C., Pinkstaff, J. (2014) In vitro and in vivo evaluation of cysteine and site specific conjugated herceptin antibody-drug conjugates. PLoS One 9 (1), e83865  DOI: 10.1371/journal.pone.0083865
  18. 18
    Strop, P., Delaria, K., Foletti, D., Witt, J. M., Hasa-Moreno, A., Poulsen, K., Casas, M. G., Dorywalska, M., Farias, S., Pios, A. (2015) Site-specific conjugation improves therapeutic index of antibody drug conjugates with high drug loading. Nat. Biotechnol. 33 (7), 694696,  DOI: 10.1038/nbt.3274
  19. 19
    Behrens, C. R., Ha, E. H., Chinn, L. L., Bowers, S., Probst, G., Fitch-Bruhns, M., Monteon, J., Valdiosera, A., Bermudez, A., Liao-Chan, S. (2015) Antibody-Drug Conjugates (ADCs) Derived from Interchain Cysteine Cross-Linking Demonstrate Improved Homogeneity and Other Pharmacological Properties over Conventional Heterogeneous ADCs. Mol. Pharmaceutics 12 (11), 39863998,  DOI: 10.1021/acs.molpharmaceut.5b00432
  20. 20
    Hamblett, K. J., Le, T., Rock, B. M., Rock, D. A., Siu, S., Huard, J. N., Conner, K. P., Milburn, R. R., O’Neill, J. W., Tometsko, M. E. (2016) Altering Antibody–Drug Conjugate Binding to the Neonatal Fc Receptor Impacts Efficacy and Tolerability. Mol. Pharmaceutics 13 (7), 23872396,  DOI: 10.1021/acs.molpharmaceut.6b00153
  21. 21
    Lhospice, F., Bregeon, D., Belmant, C., Dennler, P., Chiotellis, A., Fischer, E., Gauthier, L., Boedec, A., Rispaud, H., Savard-Chambard, S. (2015) Site-Specific Conjugation of Monomethyl Auristatin E to Anti-CD30 Antibodies Improves Their Pharmacokinetics and Therapeutic Index in Rodent Models. Mol. Pharmaceutics 12 (6), 18631871,  DOI: 10.1021/mp500666j
  22. 22
    Yoder, N. C., Bai, C., Tavares, D., Widdison, W. C., Whiteman, K. R., Wilhelm, A., Wilhelm, S. D., McShea, M. A., Maloney, E. K., Ab, O. (2019) A Case Study Comparing Heterogeneous Lysine- and Site-Specific Cysteine-Conjugated Maytansinoid Antibody-Drug Conjugates (ADCs) Illustrates Benefits of Lysine Conjugation. Mol. Pharmaceutics 16 (9), 39263937,  DOI: 10.1021/acs.molpharmaceut.9b00529
  23. 23
    Miller, M. L., Shizuka, M., Wilhelm, A., Salomon, P., Reid, E. E., Lanieri, L., Sikka, S., Maloney, E. K., Harvey, L., Qiu, Q. (2018) A DNA-Interacting Payload Designed to Eliminate Cross-Linking Improves the Therapeutic Index of Antibody-Drug Conjugates (ADCs). Mol. Cancer Ther. 17 (3), 650660,  DOI: 10.1158/1535-7163.MCT-17-0940
  24. 24
    Stimmel, J. B., Merrill, B. M., Kuyper, L. F., Moxham, C. P., Hutchins, J. T., Fling, M. E., and Kull, F. C. (2000) Site-specific conjugation on serine -> cysteine variant monoclonal antibodies. J. Biol. Chem. 275 (39), 3044530450,  DOI: 10.1074/jbc.M001672200
  25. 25
    Cao, M., De Mel, N., Jiao, Y., Howard, J., Parthemore, C., Korman, S., Thompson, C., Wendeler, M., and Liu, D. (2019) Site-specific antibody-drug conjugate heterogeneity characterization and heterogeneity root cause analysis. MAbs 11 (6), 10641076,  DOI: 10.1080/19420862.2019.1624127
  26. 26
    Vollmar, B. S., Wei, B., Ohri, R., Zhou, J., He, J., Yu, S. F., Leipold, D., Cosino, E., Yee, S., Fourie-O’Donohue, A. (2017) Attachment Site Cysteine Thiol pKa Is a Key Driver for Site-Dependent Stability of THIOMAB Antibody-Drug Conjugates. Bioconjugate Chem. 28 (10), 25382548,  DOI: 10.1021/acs.bioconjchem.7b00365
  27. 27
    Jeffrey, S. C., Burke, P. J., Lyon, R. P., Meyer, D. W., Sussman, D., Anderson, M., Hunter, J. H., Leiske, C. I., Miyamoto, J. B., Nicholas, N. D. (2013) A potent anti-CD70 antibody-drug conjugate combining a dimeric pyrrolobenzodiazepine drug with site-specific conjugation technology. Bioconjugate Chem. 24 (7), 12561263,  DOI: 10.1021/bc400217g
  28. 28
    Hamblett, K. J., Senter, P. D., Chace, D. F., Sun, M. M. C., Lenox, J., Cerveny, C. G., Kissler, K. M., Bernhardt, S. X., Kopcha, A. K., Zabinski, R. F. (2004) Effects of Drug Loading on the Antitumor Activity of a Monoclonal Antibody Drug Conjugate. Clin. Cancer Res. 10 (20), 70637070,  DOI: 10.1158/1078-0432.CCR-04-0789
  29. 29
    Sun, X., Ponte, J. F., Yoder, N. C., Laleau, R., Coccia, J., Lanieri, L., Qiu, Q., Wu, R., Hong, E., Bogalhas, M. (2017) Effects of Drug-Antibody Ratio on Pharmacokinetics, Biodistribution, Efficacy, and Tolerability of Antibody-Maytansinoid Conjugates. Bioconjugate Chem. 28 (5), 13711381,  DOI: 10.1021/acs.bioconjchem.7b00062
  30. 30
    Thompson, P., Fleming, R., Bezabeh, B., Huang, F., Mao, S., Chen, C., Harper, J., Zhong, H., Gao, X., Yu, X.-Q. (2016) Rational design, biophysical and biological characterization of site-specific antibody-tubulysin conjugates with improved stability, efficacy and pharmacokinetics. J. Controlled Release 236, 100116,  DOI: 10.1016/j.jconrel.2016.06.025
  31. 31
    Ouyang, J. (2013) Drug-to-antibody ratio (DAR) and drug load distribution by hydrophobic interaction chromatography and reversed phase high-performance liquid chromatography. Methods Mol. Biol. (N. Y., NY, U. S.) 1045, 275283,  DOI: 10.1007/978-1-62703-541-5_17
  32. 32
    Fekete, S., Veuthey, J. L., Beck, A., and Guillarme, D. (2016) Hydrophobic interaction chromatography for the characterization of monoclonal antibodies and related products. J. Pharm. Biomed. Anal. 130, 318,  DOI: 10.1016/j.jpba.2016.04.004
  33. 33
    Strop, P., Liu, S. H., Dorywalska, M., Delaria, K., Dushin, R. G., Tran, T. T., Ho, W. H., Farias, S., Casas, M. G., Abdiche, Y. (2013) Location matters: site of conjugation modulates stability and pharmacokinetics of antibody drug conjugates. Chem. Biol. 20 (2), 161167,  DOI: 10.1016/j.chembiol.2013.01.010
  34. 34
    Lyon, R. P., Bovee, T. D., Doronina, S. O., Burke, P. J., Hunter, J. H., Neff-LaFord, H. D., Jonas, M., Anderson, M. E., Setter, J. R., and Senter, P. D. (2015) Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic index. Nat. Biotechnol. 33 (7), 733735,  DOI: 10.1038/nbt.3212
  35. 35
    Tumey, L. N., Li, F., Rago, B., Han, X., Loganzo, F., Musto, S., Graziani, E. I., Puthenveetil, S., Casavant, J., Marquette, K. (2017) Site Selection: a Case Study in the Identification of Optimal Cysteine Engineered Antibody Drug Conjugates. AAPS J. 19, 113,  DOI: 10.1208/s12248-017-0083-7
  36. 36
    Burke, P. J., Hamilton, J. Z., Jeffrey, S. C., Hunter, J. H., Doronina, S. O., Okeley, N. M., Miyamoto, J. B., Anderson, M. E., Stone, I. J., Ulrich, M. L. (2017) Optimization of a PEGylated Glucuronide-Monomethylauristatin E Linker for Antibody-Drug Conjugates. Mol. Cancer Ther. 16 (1), 116123,  DOI: 10.1158/1535-7163.MCT-16-0343
  37. 37
    Lyon, R. P., Meyer, D. L., Setter, J. R., and Senter, P. D. (2012) Conjugation of anticancer drugs through endogenous monoclonal antibody cysteine residues. Methods Enzymol. 502, 123138,  DOI: 10.1016/B978-0-12-416039-2.00006-9
  38. 38
    Bhakta, S., Raab, H., and Junutula, J. R. (2013) Engineering THIOMABs for site-specific conjugation of thiol-reactive linkers. Methods Mol. Biol. 1045, 189203,  DOI: 10.1007/978-1-62703-541-5_11
  39. 39
    Garcia-Echeverria, C. (2014) Developing second generation antibody-drug conjugates: the quest for new technologies. J. Med. Chem. 57 (19), 78887889,  DOI: 10.1021/jm501298k
  40. 40
    Drake, P. M., Albers, A. E., Baker, J., Banas, S., Barfield, R. M., Bhat, A. S., de Hart, G. W., Garofalo, A. W., Holder, P., Jones, L. C. (2014) Aldehyde Tag Coupled with HIPS Chemistry Enables the Production of ADCs Conjugated Site-Specifically to Different Antibody Regions with Distinct in Vivo Efficacy and PK Outcomes. Bioconjugate Chem. 25 (7), 13311341,  DOI: 10.1021/bc500189z
  41. 41
    Catcott, K. C., McShea, M. A., Bialucha, C. U., Miller, K. L., Hicks, S. W., Saxena, P., Gesner, T. G., Woldegiorgis, M., Lewis, M. E., Bai, C. (2016) Microscale screening of antibody libraries as maytansinoid antibody-drug conjugates. MAbs 8 (3), 513523,  DOI: 10.1080/19420862.2015.1134408
  42. 42
    Cilliers, C., Guo, H., Liao, J., Christodolu, N., and Thurber, G. M. (2016) Multiscale Modeling of Antibody-Drug Conjugates: Connecting Tissue and Cellular Distribution to Whole Animal Pharmacokinetics and Potential Implications for Efficacy. AAPS J. 18 (5), 11171130,  DOI: 10.1208/s12248-016-9940-z
  43. 43
    Bumbaca, D., Boswell, C. A., Fielder, P. J., and Khawli, L. A. (2012) Physiochemical and biochemical factors influencing the pharmacokinetics of antibody therapeutics. AAPS J. 14 (3), 554558,  DOI: 10.1208/s12248-012-9369-y
  44. 44
    Bohnert, T. and Gan, L. S. (2013) Plasma protein binding: from discovery to development. J. Pharm. Sci. 102 (9), 29532994,  DOI: 10.1002/jps.23614
  45. 45
    Alley, S. C., Benjamin, D. R., Jeffrey, S. C., Okeley, N. M., Meyer, D. L., Sanderson, R. J., and Senter, P. D. (2008) Contribution of linker stability to the activities of anticancer immunoconjugates. Bioconjugate Chem. 19 (3), 759765,  DOI: 10.1021/bc7004329
  46. 46
    Ohri, R., Bhakta, S., Fourie-O’Donohue, A., Dela Cruz-Chuh, J., Tsai, S. P., Cook, R., Wei, B., Ng, C., Wong, A. W., Bos, A. B. (2018) High-Throughput Cysteine Scanning To Identify Stable Antibody Conjugation Sites for Maleimide- and Disulfide-Based Linkers. Bioconjugate Chem. 29 (2), 473485,  DOI: 10.1021/acs.bioconjchem.7b00791
  47. 47
    Lyon, R. (2018) Drawing lessons from the clinical development of antibody-drug conjugates. Drug Discovery Today: Technol. 30, 105109,  DOI: 10.1016/j.ddtec.2018.10.001
  48. 48
    Khera, E., Cilliers, C. M., Bhatnagar, S., and Thurber, G. M. (2018) Computation transport analysis of antibody-drug conjugate bystander effects and payload tumoral distribution: implications for therapy. Mol. Syst. Des. Eng. 3, 7388,  DOI: 10.1039/C7ME00093F
  49. 49
    Cilliers, C., Menezes, B., Nessler, I., Linderman, J., and Thurber, G. M. (2018) Improved Tumor Penetration and Single-Cell Targeting of Antibody-Drug Conjugates Increases Anticancer Efficacy and Host Survival. Cancer Res. 78 (3), 758768,  DOI: 10.1158/0008-5472.CAN-17-1638
  50. 50
    Sahota, S. and Vahdat, L. T. (2017) Sacituzumab govitecan: an antibody-drug conjugate. Expert Opin. Biol. Ther. 17 (8), 10271031,  DOI: 10.1080/14712598.2017.1331214
  51. 51
    Nakada, T., Sugihara, K., Jikoh, T., Abe, Y., and Agatsuma, T. (2019) The Latest Research and Development into the Antibody-Drug Conjugate, [fam-] Trastuzumab Deruxtecan (DS-8201a), for HER2 Cancer Therapy. Chem. Pharm. Bull. 67 (3), 173185,  DOI: 10.1248/cpb.c18-00744
  52. 52
    Reid, E. E., Archer, K. E., Shizuka, M., Wilhelm, A., Yoder, N. C., Bai, C., Fishkin, N. E., Harris, L., Maloney, E. K., E, H. (2019) Effect of linker stereochemistry on the antitumor activity of anti-body-drug conjugates (ADCs) containing indolinobenzodiazepine payloads. ACS Med. Chem. Lett. 10 (8), 11931197,  DOI: 10.1021/acsmedchemlett.9b00240
  53. 53
    Lazar, A. C., Wang, L., Blättler, W. A., Amphlett, G., Lambert, J. M., and Zhang, W. (2005) Analysis of the composition of immunoconjugates using size-exclusion chromatography coupled to mass spectrometry. Rapid Commun. Mass Spectrom. 19 (13), 18061814,  DOI: 10.1002/rcm.1987
  54. 54
    Kovtun, Y. V., Audette, C. A., Mayo, M. F., Jones, G. E., Doherty, H., Maloney, E. K., Erickson, H. K., Sun, X., Wilhelm, S., Ab, O. (2010) Antibody-maytansinoid conjugates designed to bypass multidrug resistance. Cancer Res. 70 (6), 25282537,  DOI: 10.1158/0008-5472.CAN-09-3546
  55. 55
    Ab, O., Whiteman, K. R., Bartle, L. M., Sun, X., Singh, R., Tavares, D., LaBelle, A., Payne, G., Lutz, R. J., Pinkas, J. (2015) IMGN853, a Folate Receptor-α (FRα)-Targeting Antibody-Drug Conjugate, Exhibits Potent Targeted Antitumor Activity against FRα-Expressing Tumors. Mol. Cancer Ther. 14 (7), 16051613,  DOI: 10.1158/1535-7163.MCT-14-1095

Cited By

Click to copy section linkSection link copied!

This article is cited by 21 publications.

  1. Amissi Sadiki, Shanshan Liu, Shefali R. Vaidya, Eric M. Kercher, Ryan T. Lang, James McIsaac, Bryan Q. Spring, Jared R. Auclair, Zhaohui Sunny Zhou. Site-Specific Conjugation of Native Antibody: Transglutaminase-Mediated Modification of a Conserved Glutamine While Maintaining the Primary Sequence and Core Fc Glycan via Trimming with an Endoglycosidase. Bioconjugate Chemistry 2024, 35 (4) , 465-471. https://doi.org/10.1021/acs.bioconjchem.4c00013
  2. Thomas T. C. Yue, Ying Ge, Francesco A. Aprile, Michelle T. Ma, Truc T. Pham, Nicholas J. Long. Site-Specific 68Ga Radiolabeling of Trastuzumab Fab via Methionine for ImmunoPET Imaging. Bioconjugate Chemistry 2023, 34 (10) , 1802-1810. https://doi.org/10.1021/acs.bioconjchem.3c00344
  3. Samantha M. Sarrett, Cindy Rodriguez, Grzegorz Rymarczyk, Meena M. Hosny, Outi Keinänen, Samantha Delaney, Sarah Thau, Benjamin A. Krantz, Brian M. Zeglis. Lysine-Directed Site-Selective Bioconjugation for the Creation of Radioimmunoconjugates. Bioconjugate Chemistry 2022, 33 (9) , 1750-1760. https://doi.org/10.1021/acs.bioconjchem.2c00354
  4. Shin Hye Ahn, Brett A. Vaughn, Willy A. Solis, Mark L. Lupher, Jr., Trevor J. Hallam, Eszter Boros. Site-Specific 89Zr- and 111In-Radiolabeling and In Vivo Evaluation of Glycan-free Antibodies by Azide–Alkyne Cycloaddition with a Non-natural Amino Acid. Bioconjugate Chemistry 2020, 31 (4) , 1177-1187. https://doi.org/10.1021/acs.bioconjchem.0c00100
  5. Qi Ma, Puro Durga, Frederick X.C. Wang, Hang-Ping Yao, Ming-Hai Wang. Pharmaceutical innovation and advanced biotechnology in the biotech-pharmaceutical industry for antibody–drug conjugate development. Drug Discovery Today 2024, 29 (8) , 104057. https://doi.org/10.1016/j.drudis.2024.104057
  6. Yi-Ching Lin, Chao-Wei Yang, Shih-Chuan Tsai, Shiou-Shiow Farn, Fang-Yu Ou Yang, Wei-Lin Lo, Liang-Cheng Chen, Kuo-Ting Chen, Mao-Chi Weng, Jui-Yin Kung, Xin-Yu Qiu, Ching-Chun Lu, Feng-Yun J. Huang. Random and site-specific radiolabeling of [89Zr]Zr-DFO-anti-PD-L1-mAb iPET tracer. Journal of Radioanalytical and Nuclear Chemistry 2024, 11 https://doi.org/10.1007/s10967-024-09401-0
  7. Viktoriia Postupalenko, Léo Marx, Mathilde Pantin, David Viertl, Nadège Gsponer, Gaëlle Giudice, Natalia Gasilova, Margret Schottelius, Frédéric Lévy, Patrick Garrouste, Jean-Manuel Segura, Origène Nyanguile. Site-selective template-directed synthesis of antibody Fc conjugates with concomitant ligand release. Chemical Science 2024, 15 (4) , 1324-1337. https://doi.org/10.1039/D3SC04324J
  8. Karoline Bechtold-Peters, Andrea Ruggiero, Nienke Vriezen, Nathan Ihle, Armin Klein, Charles Morgan, Daniel Schweizer, Dengfeng Liu, Fred Jacobson, Jakob Buecheler, Mark Panek, Naomi Duggan, Padma Malyala, Philippe Dupraz, Priyanka Desai, Shufang Niu, Yiqing Feng, Xiangyang Wang. CMC Regulatory Considerations for Antibody-Drug Conjugates. Journal of Pharmaceutical Sciences 2023, 112 (12) , 2965-2980. https://doi.org/10.1016/j.xphs.2023.09.007
  9. Ryan J. Pakula, Peter J. H. Scott. Applications of radiolabeled antibodies in neuroscience and neuro‐oncology. Journal of Labelled Compounds and Radiopharmaceuticals 2023, 66 (9) , 269-285. https://doi.org/10.1002/jlcr.4049
  10. Cindy Rodriguez, Samantha Delaney, Joni Sebastiano, Samantha M. Sarrett, Mike A. Cornejo, Sarah Thau, Meena M. Hosny, Brian M. Zeglis. Site-selective radiolabeling using mushroom tyrosinase and the strain-promoted oxidation-controlled 1,2-quinone cycloaddition. RSC Advances 2023, 13 (26) , 17705-17709. https://doi.org/10.1039/D3RA03486K
  11. Rita Khoury, Khalil Saleh, Nadine Khalife, Mohamad Saleh, Claude Chahine, Rebecca Ibrahim, Axel Lecesne. Mechanisms of Resistance to Antibody-Drug Conjugates. International Journal of Molecular Sciences 2023, 24 (11) , 9674. https://doi.org/10.3390/ijms24119674
  12. Arnaud C. Tiberghien, Balakumar Vijayakrishnan, Arman Esfandiari, Mahammad Ahmed, Raul Pardo, John Bingham, Lauren Adams, Kathleen Santos, Gyoung-Dong Kang, Kathryn M. Pugh, Shameen Afif-Rider, Kapil Vashisht, Kemal Haque, Ravinder Tammali, Edward Rosfjord, Adriana Savoca, John A. Hartley, Philip W. Howard. Comparison of Pyrrolobenzodiazepine Dimer Bis-imine versus Mono-imine: DNA Interstrand Cross-linking, Cytotoxicity, Antibody–Drug Conjugate Efficacy and Toxicity. Molecular Cancer Therapeutics 2023, 22 (2) , 254-263. https://doi.org/10.1158/1535-7163.MCT-21-0693
  13. Juniper A. Scribner, Stuart W. Hicks, Kerstin W. Sinkevicius, Nicholas C. Yoder, Gundo Diedrich, Jennifer G. Brown, Jacquelynn Lucas, Megan E. Fuller, Thomas Son, Anahita Dastur, Jeff Hooley, Christopher Espelin, Marian Themeles, Francine Z. Chen, Ying Li, Michael Chiechi, Jenny Lee, Bhaswati Barat, Lusiana Widjaja, Sergey Gorlatov, James Tamura, Valentina Ciccarone, Olga Ab, Kerry A. McEachem, Scott Koenig, Eric H. Westin, Paul A. Moore, Thomas Chittenden, Richard J. Gregory, Ezio Bonvini, Deryk Loo. Preclinical Evaluation of IMGC936, a Next-Generation Maytansinoid-based Antibody–drug Conjugate Targeting ADAM9-expressing Tumors. Molecular Cancer Therapeutics 2022, 21 (7) , 1047-1059. https://doi.org/10.1158/1535-7163.MCT-21-0915
  14. Ning Zou, Amy Han. Application of Pyrrolobenzodiazepines in Antibody Drug Conjugates. 2022, 293-339. https://doi.org/10.1002/9781119627784.ch13
  15. Charlotte Sornay, Valentine Vaur, Alain Wagner, Guilhem Chaubet. An overview of chemo- and site-selectivity aspects in the chemical conjugation of proteins. Royal Society Open Science 2022, 9 (1) https://doi.org/10.1098/rsos.211563
  16. M. W. Giese, R. H. Woodman, G. T. Hermanson, P. D. Davis. The Use of Uniform PEG Compounds in the Design of ADCs. 2021, 286-376. https://doi.org/10.1039/9781839165153-00286
  17. Anna Kaempffe, Stephan Dickgiesser, Nicolas Rasche, Andrea Paoletti, Elisa Bertotti, Ilse De Salve, Federico Riccardi Sirtori, Roland Kellner, Doreen Könning, Stefan Hecht, Jan Anderl, Harald Kolmar, Christian Schröter. Effect of Conjugation Site and Technique on the Stability and Pharmacokinetics of Antibody-Drug Conjugates. Journal of Pharmaceutical Sciences 2021, 110 (12) , 3776-3785. https://doi.org/10.1016/j.xphs.2021.08.002
  18. Jeffrey Y.-K. Wong, Raja Mukherjee, Jiayuan Miao, Olena Bilyk, Vivian Triana, Mark Miskolzie, Antoine Henninot, John J. Dwyer, Serhii Kharchenko, Anna Iampolska, Dmitriy M. Volochnyuk, Yu-Shan Lin, Lynne-Marie Postovit, Ratmir Derda. Genetically-encoded discovery of proteolytically stable bicyclic inhibitors for morphogen NODAL. Chemical Science 2021, 12 (28) , 9694-9703. https://doi.org/10.1039/D1SC01916C
  19. Ming-Chen Yang, Chi-Sheng Shia, Wan-Fen Li, Chun-Chung Wang, I-Ju Chen, Teng-Yi Huang, Yu-Jung Chen, Hui-Wen Chang, Chi-Huan Lu, Yueh-Chin Wu, Nan-Hsuan Wang, Jiann-Shiun Lai, Cheng-Der Yu, Ming-Tain Lai. Preclinical Studies of OBI-999: A Novel Globo H–Targeting Antibody–Drug Conjugate. Molecular Cancer Therapeutics 2021, 20 (6) , 1121-1132. https://doi.org/10.1158/1535-7163.MCT-20-0763
  20. Jose F. Ponte, Leanne Lanieri, Eshita Khera, Rassol Laleau, Olga Ab, Christopher Espelin, Neeraj Kohli, Bahar Matin, Yulius Setiady, Michael L. Miller, Thomas A. Keating, Ravi Chari, Jan Pinkas, Richard Gregory, Greg M. Thurber. Antibody Co-Administration Can Improve Systemic and Local Distribution of Antibody–Drug Conjugates to Increase In Vivo Efficacy. Molecular Cancer Therapeutics 2021, 20 (1) , 203-212. https://doi.org/10.1158/1535-7163.MCT-20-0451
  21. Amissi Sadiki, Shefali R Vaidya, Mina Abdollahi, Gunjan Bhardwaj, Michael E Dolan, Harpreet Turna, Varnika Arora, Athul Sanjeev, Timothy D Robinson, Andrea Koid, Aashka Amin, Zhaohui Sunny Zhou. Site-specific conjugation of native antibody. Antibody Therapeutics 2020, 3 (4) , 271-284. https://doi.org/10.1093/abt/tbaa027
Open PDF

Bioconjugate Chemistry

Cite this: Bioconjugate Chem. 2020, 31, 1, 93–103
Click to copy citationCitation copied!
https://doi.org/10.1021/acs.bioconjchem.9b00777
Published November 20, 2019

Copyright © 2019 American Chemical Society. This publication is licensed under these Terms of Use.

Article Views

7783

Altmetric

-

Citations

Learn about these metrics

Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.

  • Abstract

    Figure 1

    Figure 1. (A) Structures of DGN549-L (1a), used for lysine-conjugation; and DGN549-C (1b), used for cysteine conjugation. (B) Structural representations of lysine-linked and CYSMAB ADCs.

    Figure 2

    Figure 2. SEC-MS data for mAb1 ADCs: (A) ADC 2a and (B) ADC 2b.

    Figure 3

    Figure 3. (A) HIC chromatograms of ADCs 2a and 2b. (B) HIC chromatograms of ADCs 3a and 3b.

    Figure 4

    Figure 4. Processing of ADCs 2a and 2b to release the same catabolite FGN849 (4).

    Figure 5

    Figure 5. Activity of mAb1 ADCs in SCID mice bearing NCI-H2110 non-small cell lung cancer xenografts expressing FRα.

    Figure 6

    Figure 6. Activity of mAb2 ADCs and nontargeting controls in SCID mice bearing Molm-13 acute myeloid leukemia xenografts expressing CD123.

    Figure 7

    Figure 7. Tolerability of mAb1 ADCs in non-tumor-bearing CD-1 mice. (A) ADC 2a and (B) ADC 2b

    Figure 8

    Figure 8. Tolerability of mAb2 ADCs in non tumor-bearing CD-1 mice: (A) ADC 3a and (B) ADC 3b.

    Figure 9

    Figure 9. Tolerability of ADC 2c in non tumor-bearing CD-1 mice.

    Figure 10

    Figure 10. Pharmacokinetics of mAb1 ADCs 2a and 2b in non-tumor-bearing CD-1 mice, assayed by ELISA methods for total antibody and conjugated antibody.

  • References


    This article references 55 other publications.

    1. 1
      Chari, R. V. J., Miller, M. L., and Widdison, W. C. (2014) Antibody–Drug Conjugates: An Emerging Concept in Cancer Therapy. Angew. Chem., Int. Ed. 53 (15), 37963827,  DOI: 10.1002/anie.201307628
    2. 2
      Lambert, J. M. and Chari, R. V. (2014) Ado-trastuzumab Emtansine (T-DM1): an antibody-drug conjugate (ADC) for HER2-positive breast cancer. J. Med. Chem. 57 (16), 69496964,  DOI: 10.1021/jm500766w
    3. 3
      Senter, P. D. and Sievers, E. L. (2012) The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nat. Biotechnol. 30 (7), 631637,  DOI: 10.1038/nbt.2289
    4. 4
      Appelbaum, F. R. and Bernstein, I. D. (2017) Gemtuzumab ozogamicin for acute myeloid leukemia. Blood 130 (22), 23732376,  DOI: 10.1182/blood-2017-09-797712
    5. 5
      Yurkiewicz, I. R., Muffly, L., and Liedtke, M. (2018) Inotuzumab ozogamicin: a CD22 mAb-drug conjugate for adult relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Drug Des., Dev. Ther. 12, 22932300,  DOI: 10.2147/DDDT.S150317
    6. 6
      Dornan, D., Bennett, F., Chen, Y., Dennis, M., Eaton, D., Elkins, K., French, D., Go, M. A., Jack, A., Junutula, J. R. (2009) Therapeutic potential of an anti-CD79b antibody-drug conjugate, anti-CD79b-vc-MMAE, for the treatment of non-Hodgkin lymphoma. Blood 114 (13), 27212729,  DOI: 10.1182/blood-2009-02-205500
    7. 7
      Donaghy, H. (2016) Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugates. MAbs 8 (4), 659671,  DOI: 10.1080/19420862.2016.1156829
    8. 8
      Chari, R. V. J. (2016) Expanding the Reach of Antibody-Drug Conjugates. ACS Med. Chem. Lett. 7 (11), 974976,  DOI: 10.1021/acsmedchemlett.6b00312
    9. 9
      Behrens, C. R. and Liu, B. (2014) Methods for site-specific drug conjugation to antibodies. MAbs 6 (1), 4653,  DOI: 10.4161/mabs.26632
    10. 10
      Agarwal, P. and Bertozzi, C. R. (2015) Site-specific antibody-drug conjugates: the nexus of bioorthogonal chemistry, protein engineering, and drug development. Bioconjugate Chem. 26 (2), 176192,  DOI: 10.1021/bc5004982
    11. 11
      Panowski, S., Bhakta, S., Raab, H., Polakis, P., and Junutula, J. R. (2014) Site-specific antibody drug conjugates for cancer therapy. MAbs 6 (1), 3445,  DOI: 10.4161/mabs.27022
    12. 12
      Junutula, J. R., Raab, H., Clark, S., Bhakta, S., Leipold, D. D., Weir, S., Chen, Y., Simpson, M., Tsai, S. P., Dennis, M. S. (2008) Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat. Biotechnol. 26 (8), 925932,  DOI: 10.1038/nbt.1480
    13. 13
      Junutula, J. R., Flagella, K. M., Graham, R. A., Parsons, K. L., Ha, E., Raab, H., Bhakta, S., Nguyen, T., Dugger, D. L., Li, G. (2010) Engineered Thio-Trastuzumab-DM1 Conjugate with an Improved Therapeutic Index to Target Human Epidermal Growth Factor Receptor 2–Positive Breast Cancer. Clin. Cancer Res. 16 (19), 47694778,  DOI: 10.1158/1078-0432.CCR-10-0987
    14. 14
      Shen, B. Q., Xu, K., Liu, L., Raab, H., Bhakta, S., Kenrick, M., Parsons-Reponte, K. L., Tien, J., Yu, S. F., Mai, E. (2012) Conjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugates. Nat. Biotechnol. 30 (2), 184189,  DOI: 10.1038/nbt.2108
    15. 15
      Pillow, T. H., Tien, J., Parsons-Reponte, K. L., Bhakta, S., Li, H., Staben, L. R., Li, G., Chuh, J., Fourie-O’Donohue, A., Darwish, M. (2014) Site-specific trastuzumab maytansinoid antibody-drug conjugates with improved therapeutic activity through linker and antibody engineering. J. Med. Chem. 57 (19), 78907899,  DOI: 10.1021/jm500552c
    16. 16
      Tian, F., Lu, Y., Manibusan, A., Sellers, A., Tran, H., Sun, Y., Phuong, T., Barnett, R., Hehli, B., Song, F. (2014) A general approach to site-specific antibody drug conjugates. Proc. Natl. Acad. Sci. U. S. A. 111 (5), 17661771,  DOI: 10.1073/pnas.1321237111
    17. 17
      Jackson, D., Atkinson, J., Guevara, C. I., Zhang, C., Kery, V., Moon, S. J., Virata, C., Yang, P., Lowe, C., Pinkstaff, J. (2014) In vitro and in vivo evaluation of cysteine and site specific conjugated herceptin antibody-drug conjugates. PLoS One 9 (1), e83865  DOI: 10.1371/journal.pone.0083865
    18. 18
      Strop, P., Delaria, K., Foletti, D., Witt, J. M., Hasa-Moreno, A., Poulsen, K., Casas, M. G., Dorywalska, M., Farias, S., Pios, A. (2015) Site-specific conjugation improves therapeutic index of antibody drug conjugates with high drug loading. Nat. Biotechnol. 33 (7), 694696,  DOI: 10.1038/nbt.3274
    19. 19
      Behrens, C. R., Ha, E. H., Chinn, L. L., Bowers, S., Probst, G., Fitch-Bruhns, M., Monteon, J., Valdiosera, A., Bermudez, A., Liao-Chan, S. (2015) Antibody-Drug Conjugates (ADCs) Derived from Interchain Cysteine Cross-Linking Demonstrate Improved Homogeneity and Other Pharmacological Properties over Conventional Heterogeneous ADCs. Mol. Pharmaceutics 12 (11), 39863998,  DOI: 10.1021/acs.molpharmaceut.5b00432
    20. 20
      Hamblett, K. J., Le, T., Rock, B. M., Rock, D. A., Siu, S., Huard, J. N., Conner, K. P., Milburn, R. R., O’Neill, J. W., Tometsko, M. E. (2016) Altering Antibody–Drug Conjugate Binding to the Neonatal Fc Receptor Impacts Efficacy and Tolerability. Mol. Pharmaceutics 13 (7), 23872396,  DOI: 10.1021/acs.molpharmaceut.6b00153
    21. 21
      Lhospice, F., Bregeon, D., Belmant, C., Dennler, P., Chiotellis, A., Fischer, E., Gauthier, L., Boedec, A., Rispaud, H., Savard-Chambard, S. (2015) Site-Specific Conjugation of Monomethyl Auristatin E to Anti-CD30 Antibodies Improves Their Pharmacokinetics and Therapeutic Index in Rodent Models. Mol. Pharmaceutics 12 (6), 18631871,  DOI: 10.1021/mp500666j
    22. 22
      Yoder, N. C., Bai, C., Tavares, D., Widdison, W. C., Whiteman, K. R., Wilhelm, A., Wilhelm, S. D., McShea, M. A., Maloney, E. K., Ab, O. (2019) A Case Study Comparing Heterogeneous Lysine- and Site-Specific Cysteine-Conjugated Maytansinoid Antibody-Drug Conjugates (ADCs) Illustrates Benefits of Lysine Conjugation. Mol. Pharmaceutics 16 (9), 39263937,  DOI: 10.1021/acs.molpharmaceut.9b00529
    23. 23
      Miller, M. L., Shizuka, M., Wilhelm, A., Salomon, P., Reid, E. E., Lanieri, L., Sikka, S., Maloney, E. K., Harvey, L., Qiu, Q. (2018) A DNA-Interacting Payload Designed to Eliminate Cross-Linking Improves the Therapeutic Index of Antibody-Drug Conjugates (ADCs). Mol. Cancer Ther. 17 (3), 650660,  DOI: 10.1158/1535-7163.MCT-17-0940
    24. 24
      Stimmel, J. B., Merrill, B. M., Kuyper, L. F., Moxham, C. P., Hutchins, J. T., Fling, M. E., and Kull, F. C. (2000) Site-specific conjugation on serine -> cysteine variant monoclonal antibodies. J. Biol. Chem. 275 (39), 3044530450,  DOI: 10.1074/jbc.M001672200
    25. 25
      Cao, M., De Mel, N., Jiao, Y., Howard, J., Parthemore, C., Korman, S., Thompson, C., Wendeler, M., and Liu, D. (2019) Site-specific antibody-drug conjugate heterogeneity characterization and heterogeneity root cause analysis. MAbs 11 (6), 10641076,  DOI: 10.1080/19420862.2019.1624127
    26. 26
      Vollmar, B. S., Wei, B., Ohri, R., Zhou, J., He, J., Yu, S. F., Leipold, D., Cosino, E., Yee, S., Fourie-O’Donohue, A. (2017) Attachment Site Cysteine Thiol pKa Is a Key Driver for Site-Dependent Stability of THIOMAB Antibody-Drug Conjugates. Bioconjugate Chem. 28 (10), 25382548,  DOI: 10.1021/acs.bioconjchem.7b00365
    27. 27
      Jeffrey, S. C., Burke, P. J., Lyon, R. P., Meyer, D. W., Sussman, D., Anderson, M., Hunter, J. H., Leiske, C. I., Miyamoto, J. B., Nicholas, N. D. (2013) A potent anti-CD70 antibody-drug conjugate combining a dimeric pyrrolobenzodiazepine drug with site-specific conjugation technology. Bioconjugate Chem. 24 (7), 12561263,  DOI: 10.1021/bc400217g
    28. 28
      Hamblett, K. J., Senter, P. D., Chace, D. F., Sun, M. M. C., Lenox, J., Cerveny, C. G., Kissler, K. M., Bernhardt, S. X., Kopcha, A. K., Zabinski, R. F. (2004) Effects of Drug Loading on the Antitumor Activity of a Monoclonal Antibody Drug Conjugate. Clin. Cancer Res. 10 (20), 70637070,  DOI: 10.1158/1078-0432.CCR-04-0789
    29. 29
      Sun, X., Ponte, J. F., Yoder, N. C., Laleau, R., Coccia, J., Lanieri, L., Qiu, Q., Wu, R., Hong, E., Bogalhas, M. (2017) Effects of Drug-Antibody Ratio on Pharmacokinetics, Biodistribution, Efficacy, and Tolerability of Antibody-Maytansinoid Conjugates. Bioconjugate Chem. 28 (5), 13711381,  DOI: 10.1021/acs.bioconjchem.7b00062
    30. 30
      Thompson, P., Fleming, R., Bezabeh, B., Huang, F., Mao, S., Chen, C., Harper, J., Zhong, H., Gao, X., Yu, X.-Q. (2016) Rational design, biophysical and biological characterization of site-specific antibody-tubulysin conjugates with improved stability, efficacy and pharmacokinetics. J. Controlled Release 236, 100116,  DOI: 10.1016/j.jconrel.2016.06.025
    31. 31
      Ouyang, J. (2013) Drug-to-antibody ratio (DAR) and drug load distribution by hydrophobic interaction chromatography and reversed phase high-performance liquid chromatography. Methods Mol. Biol. (N. Y., NY, U. S.) 1045, 275283,  DOI: 10.1007/978-1-62703-541-5_17
    32. 32
      Fekete, S., Veuthey, J. L., Beck, A., and Guillarme, D. (2016) Hydrophobic interaction chromatography for the characterization of monoclonal antibodies and related products. J. Pharm. Biomed. Anal. 130, 318,  DOI: 10.1016/j.jpba.2016.04.004
    33. 33
      Strop, P., Liu, S. H., Dorywalska, M., Delaria, K., Dushin, R. G., Tran, T. T., Ho, W. H., Farias, S., Casas, M. G., Abdiche, Y. (2013) Location matters: site of conjugation modulates stability and pharmacokinetics of antibody drug conjugates. Chem. Biol. 20 (2), 161167,  DOI: 10.1016/j.chembiol.2013.01.010
    34. 34
      Lyon, R. P., Bovee, T. D., Doronina, S. O., Burke, P. J., Hunter, J. H., Neff-LaFord, H. D., Jonas, M., Anderson, M. E., Setter, J. R., and Senter, P. D. (2015) Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic index. Nat. Biotechnol. 33 (7), 733735,  DOI: 10.1038/nbt.3212
    35. 35
      Tumey, L. N., Li, F., Rago, B., Han, X., Loganzo, F., Musto, S., Graziani, E. I., Puthenveetil, S., Casavant, J., Marquette, K. (2017) Site Selection: a Case Study in the Identification of Optimal Cysteine Engineered Antibody Drug Conjugates. AAPS J. 19, 113,  DOI: 10.1208/s12248-017-0083-7
    36. 36
      Burke, P. J., Hamilton, J. Z., Jeffrey, S. C., Hunter, J. H., Doronina, S. O., Okeley, N. M., Miyamoto, J. B., Anderson, M. E., Stone, I. J., Ulrich, M. L. (2017) Optimization of a PEGylated Glucuronide-Monomethylauristatin E Linker for Antibody-Drug Conjugates. Mol. Cancer Ther. 16 (1), 116123,  DOI: 10.1158/1535-7163.MCT-16-0343
    37. 37
      Lyon, R. P., Meyer, D. L., Setter, J. R., and Senter, P. D. (2012) Conjugation of anticancer drugs through endogenous monoclonal antibody cysteine residues. Methods Enzymol. 502, 123138,  DOI: 10.1016/B978-0-12-416039-2.00006-9
    38. 38
      Bhakta, S., Raab, H., and Junutula, J. R. (2013) Engineering THIOMABs for site-specific conjugation of thiol-reactive linkers. Methods Mol. Biol. 1045, 189203,  DOI: 10.1007/978-1-62703-541-5_11
    39. 39
      Garcia-Echeverria, C. (2014) Developing second generation antibody-drug conjugates: the quest for new technologies. J. Med. Chem. 57 (19), 78887889,  DOI: 10.1021/jm501298k
    40. 40
      Drake, P. M., Albers, A. E., Baker, J., Banas, S., Barfield, R. M., Bhat, A. S., de Hart, G. W., Garofalo, A. W., Holder, P., Jones, L. C. (2014) Aldehyde Tag Coupled with HIPS Chemistry Enables the Production of ADCs Conjugated Site-Specifically to Different Antibody Regions with Distinct in Vivo Efficacy and PK Outcomes. Bioconjugate Chem. 25 (7), 13311341,  DOI: 10.1021/bc500189z
    41. 41
      Catcott, K. C., McShea, M. A., Bialucha, C. U., Miller, K. L., Hicks, S. W., Saxena, P., Gesner, T. G., Woldegiorgis, M., Lewis, M. E., Bai, C. (2016) Microscale screening of antibody libraries as maytansinoid antibody-drug conjugates. MAbs 8 (3), 513523,  DOI: 10.1080/19420862.2015.1134408
    42. 42
      Cilliers, C., Guo, H., Liao, J., Christodolu, N., and Thurber, G. M. (2016) Multiscale Modeling of Antibody-Drug Conjugates: Connecting Tissue and Cellular Distribution to Whole Animal Pharmacokinetics and Potential Implications for Efficacy. AAPS J. 18 (5), 11171130,  DOI: 10.1208/s12248-016-9940-z
    43. 43
      Bumbaca, D., Boswell, C. A., Fielder, P. J., and Khawli, L. A. (2012) Physiochemical and biochemical factors influencing the pharmacokinetics of antibody therapeutics. AAPS J. 14 (3), 554558,  DOI: 10.1208/s12248-012-9369-y
    44. 44
      Bohnert, T. and Gan, L. S. (2013) Plasma protein binding: from discovery to development. J. Pharm. Sci. 102 (9), 29532994,  DOI: 10.1002/jps.23614
    45. 45
      Alley, S. C., Benjamin, D. R., Jeffrey, S. C., Okeley, N. M., Meyer, D. L., Sanderson, R. J., and Senter, P. D. (2008) Contribution of linker stability to the activities of anticancer immunoconjugates. Bioconjugate Chem. 19 (3), 759765,  DOI: 10.1021/bc7004329
    46. 46
      Ohri, R., Bhakta, S., Fourie-O’Donohue, A., Dela Cruz-Chuh, J., Tsai, S. P., Cook, R., Wei, B., Ng, C., Wong, A. W., Bos, A. B. (2018) High-Throughput Cysteine Scanning To Identify Stable Antibody Conjugation Sites for Maleimide- and Disulfide-Based Linkers. Bioconjugate Chem. 29 (2), 473485,  DOI: 10.1021/acs.bioconjchem.7b00791
    47. 47
      Lyon, R. (2018) Drawing lessons from the clinical development of antibody-drug conjugates. Drug Discovery Today: Technol. 30, 105109,  DOI: 10.1016/j.ddtec.2018.10.001
    48. 48
      Khera, E., Cilliers, C. M., Bhatnagar, S., and Thurber, G. M. (2018) Computation transport analysis of antibody-drug conjugate bystander effects and payload tumoral distribution: implications for therapy. Mol. Syst. Des. Eng. 3, 7388,  DOI: 10.1039/C7ME00093F
    49. 49
      Cilliers, C., Menezes, B., Nessler, I., Linderman, J., and Thurber, G. M. (2018) Improved Tumor Penetration and Single-Cell Targeting of Antibody-Drug Conjugates Increases Anticancer Efficacy and Host Survival. Cancer Res. 78 (3), 758768,  DOI: 10.1158/0008-5472.CAN-17-1638
    50. 50
      Sahota, S. and Vahdat, L. T. (2017) Sacituzumab govitecan: an antibody-drug conjugate. Expert Opin. Biol. Ther. 17 (8), 10271031,  DOI: 10.1080/14712598.2017.1331214
    51. 51
      Nakada, T., Sugihara, K., Jikoh, T., Abe, Y., and Agatsuma, T. (2019) The Latest Research and Development into the Antibody-Drug Conjugate, [fam-] Trastuzumab Deruxtecan (DS-8201a), for HER2 Cancer Therapy. Chem. Pharm. Bull. 67 (3), 173185,  DOI: 10.1248/cpb.c18-00744
    52. 52
      Reid, E. E., Archer, K. E., Shizuka, M., Wilhelm, A., Yoder, N. C., Bai, C., Fishkin, N. E., Harris, L., Maloney, E. K., E, H. (2019) Effect of linker stereochemistry on the antitumor activity of anti-body-drug conjugates (ADCs) containing indolinobenzodiazepine payloads. ACS Med. Chem. Lett. 10 (8), 11931197,  DOI: 10.1021/acsmedchemlett.9b00240
    53. 53
      Lazar, A. C., Wang, L., Blättler, W. A., Amphlett, G., Lambert, J. M., and Zhang, W. (2005) Analysis of the composition of immunoconjugates using size-exclusion chromatography coupled to mass spectrometry. Rapid Commun. Mass Spectrom. 19 (13), 18061814,  DOI: 10.1002/rcm.1987
    54. 54
      Kovtun, Y. V., Audette, C. A., Mayo, M. F., Jones, G. E., Doherty, H., Maloney, E. K., Erickson, H. K., Sun, X., Wilhelm, S., Ab, O. (2010) Antibody-maytansinoid conjugates designed to bypass multidrug resistance. Cancer Res. 70 (6), 25282537,  DOI: 10.1158/0008-5472.CAN-09-3546
    55. 55
      Ab, O., Whiteman, K. R., Bartle, L. M., Sun, X., Singh, R., Tavares, D., LaBelle, A., Payne, G., Lutz, R. J., Pinkas, J. (2015) IMGN853, a Folate Receptor-α (FRα)-Targeting Antibody-Drug Conjugate, Exhibits Potent Targeted Antitumor Activity against FRα-Expressing Tumors. Mol. Cancer Ther. 14 (7), 16051613,  DOI: 10.1158/1535-7163.MCT-14-1095
  • Supporting Information

    Supporting Information


    The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.bioconjchem.9b00777.

    • Detailed experimental procedures for synthesis of compound 1b, supplementary characterization data for ADCs, and supplementary methods (PDF)


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.