Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

MOLECULAR CANCER THERAPEUTICS | LARGE MOLECULE THERAPEUTICS

Design and Evaluation of Phosphonamidate-Linked


Exatecan Constructs for Highly Loaded, Stable, and
Efficacious Antibody–Drug Conjugates
Saskia Schmitt1, Paul Machui1, Isabelle Mai1, Sarah Herterich1, Swetlana Wunder1, Philipp Cyprys1,
Marcus Gerlach1, Philipp Ochtrop1, Christian P.R. Hackenberger2,3, Dominik Schumacher1, Jonas Helma1,
Annette M. Vogl1, and Marc-Andre  Kasper1

ABSTRACT

Topoisomerase I (TOP1) Inhibitors constitute an emerging that the identified linker–payload structure enables the construction
payload class to engineer antibody–drug conjugates (ADC) as of highly loaded DAR8 ADCs with excellent solubility properties.
next-generation biopharmaceutical for cancer treatment. Existing Head-to-head comparison with Enhertu, an approved camptothe-
ADCs are using camptothecin payloads with lower potency and cin-based ADC, revealed improved target-mediated killing of tumor
suffer from limited stability in circulation. With this study, we cells, excellent bystander killing, drastically improved linker stabil-
introduce a novel camptothecin-based linker–payload platform ity in vitro and in vivo and superior in vivo efficacy over four tested
based on the highly potent camptothecin derivative exatecan. First, dose levels in a xenograft model. Moreover, we show that ADCs
we describe general challenges that arise from the hydrophobic based on the novel exatecan linker–payload platform exhibit anti-
combination of exatecan and established dipeptidyl p-aminobenzyl- body-like pharmacokinetic properties, even when the ADCs are
carbamate (PAB) cleavage sites such as reduced antibody conju- highly loaded with eight drug molecules per antibody. This ADC
gation yields and ADC aggregation. After evaluating several linker– platform constitutes a new and general solution to deliver TOP1
payload structures, we identified ethynyl-phosphonamidates in inhibitors with highest efficiency to the site of the tumor, indepen-
combination with a discrete PEG24 chain to compensate for the dent of the antibody and its target, and is thereby broadly applicable
hydrophobic PAB–exatecan moiety. Furthermore, we demonstrate to various cancer indications.

Introduction irinotecan used in Trodelvy, have been contributing to the success


of the TOP1 inhibitor payload family as ADC payloads (see Fig. 1
With a total of 12 approved antibody–drug conjugates (ADC) up
for structures). Trodelvy has been shown to significantly prolong
to the present day and not less than 8 approvals by the FDA only in
overall survival (OS) compared with single-agent chemotherapy in
the last four years, ADCs for the treatment of cancer are coming of
difficult to treat patients with estrogen receptor–positive, HER2-
age (1, 2). Drug classes that are being used in those marketed ADCs
negative breast cancer (9). Enhertu demonstrated superior efficacy,
include DNA-damaging and microtubule-disrupting agents (3).
with a significantly longer progression-free survival in patients with
However, one class that stands out in terms of clinical efficacy
breast cancer compared with trastuzumab-DM1 (Kadcyla) treat-
and market success is of topoisomerase I (TOP1) inhibitors, used
ment (10). Notably, this tremendous advantage is achieved with the
in trastuzumab deruxtecan (Enhertu), sacituzumab govitecan
same antibody and target, only changing the linker–payload from
(Trodelvy) (4) and many other preclinically and clinically inves-
the tubulin-disrupting agent DM1 to a TOP1 inhibitor. Moreover,
tigated ADCs (5–7). Next to the application as ADC payloads, which
Enhertu has been changing the paradigm of HER2-positivity,
this publication is focusing on, camptothecin and its derivatives have
because it showed remarkable activity in patients with HER2-low
been studied for decades as small-molecule therapeutics in cancer
breast cancer, a patient population previously considered as being
treatment (8).
not amenable to HER2-based treatments (11).
In particular, camptothecin analogs such as DXd, a derivative of
Despite those remarkable achievements in the treatment of
exatecan used in Enhertu, and SN38, the active metabolite of
patients with cancer with ADCs using camptothecin-based pay-
loads, there is also still room for improvement. Even though ADCs
1
Tubulis GmbH, Planegg-Martinsried, Germany. 2Chemical Biology Department,
are in theory highly targeted medications, treated patients are still
Leibniz-Forschungsinstitut fu€r Molekulare Pharmakologie (FMP), Berlin, suffering from marked signs of toxicity (12) with neutropenia and
Germany. 3Department of Chemistry, Humboldt Universit€at zu Berlin, Berlin, severe diarrhea being the most common adverse event leading
Germany. to dose reductions for Trodelvy and pneumonitis and interstitial
S. Schmitt and P. Machui contributed equally as co-authors of this article. lung disease being the most common reasons for treatment dis-
continuation for Enhertu (1). Even though mechanisms of ADC-
Corresponding Authors: Marc-Andre  Kasper, Tubulis GmbH, Am Klopferspitz
19a, Planegg-Martinsried, Bavaria 82152, Germany. E-mail:
mediated toxicities are complex and not yet fully understood, it is
marc.kasper@tubulis.com; and Annette M. Vogl, annette.vogl@tubulis.com discussed throughout the literature that the linker that connects the
cytotoxic drug with the antibody plays a crucial role in how efficacy
Mol Cancer Ther 2023;XX:XX–XX
and toxicity is balanced within a given ADC (13–16). Critical
doi: 10.1158/1535-7163.MCT-23-0359
requirements on a linker include plasma stability (15, 17) and
This open access article is distributed under the Creative Commons Attribution- inclusion of hydrophilicity to compensate for the typically hydro-
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license. phobic drug to prevent undesired ADC aggregation and accelerated
2023 The Authors; Published by the American Association for Cancer Research plasma clearance (18, 19).

AACRJournals.org | OF1
Schmitt et al.

Figure 1.
Overview of TOP1 inhibitor-based linker–payloads used in marketed ADCs Enhertu and Trodelvy (top), based on the payloads DXd and SN38 (2) and the structure of
the linker–payloads described herein based on exatecan with an overview over basic features of the ADCs originating from those linker systems. Payload structures
are highlighted in orange.

Despite being a highly promising ADC payload class, the conju- Furthermore, ethynylphosphonamidates provided in combination
gation of the camptothecin moiety, including its derivatives has been with solubility enhancers high DAR ADCs of hydrophobic VC–
shown to be in particular challenging in the past, especially in higher PAB–MMAE linker–payloads without increased in vivo clear-
drug-to-antibody ratios (DAR). Burke and colleagues (20) have ance (31, 32). High DARs are important for camptothecin payloads,
shown that antibody conjugates with valine–citrulline–PAB [VC– in particular, because high DAR can compensate for the lower
PAB (dipeptidyl p-aminobenzyl-carbamate)]–camptothecin deriva- potency compared with other ADC payloads such as auristatins or
tives exhibit a strong tendency to form higher molecular weight species calicheamycins (4).
(HMWS). This issue could only be solved by DAR reduction Our goal in the current study was to design a novel TOP1 inhibitor-
and substitution of the VC-cleavage site with a more polar glucuro- based linker–payload platform that facilitates aggregation-free con-
nide (20). Similar challenges in induction of aggregation, once con- struction of highly loaded DAR8 ADCs with excellent serum stability,
jugated to an antibody, have been described for several exatecan- without enhanced in vivo clearance rates and potent and selective
based derivatives (21). Further improvements in the linker structure in vitro and in vivo efficacy. For this, we used exatecan as payload and
such as the implementation of a more polar hemiaminal-based self- conducted chemical optimization of the linker to facilitate aggrega-
immolative moiety led to the discovery of the DXd-containing linker tion-free ADCs even at high DARs of 8. The optimal linker–payload
used in Enhertu (22, 23). This linker strategy has also been applied to structure identified carries the well-established VC–PAB cleavage site
similar camptothecin analogs, all of which showing decent activity for traceless intracellular release of the payload (33) and ethynylpho-
in vitro, including bystander killing (24). In another study, hydrophilic sphonamidates equipped with polyethylene glycol (PEG) chains of
polysarcosins have been added to the linker to compensate for discrete length of 24 units. The PEG chain is thereby attached in a
the hydrophobic exatecan moiety (25). It should be noted that all branched fashion to ensure close distance between antibody and
those examples exhibit maleimide linker chemistry for conjugation, payload. We demonstrate that DAR8 ADCs based on this linker–
whereas a camptothecin-based linker–payload platform that provides payload structure are superior in head-to-head comparisons with
fully in vivo-stable antibody conjugates of a high DAR has still been Enhertu in a broad panel of in vitro and in vivo experiments. Moreover,
unprecedented until this study. we observed excellent bystander killing as well as release of damage-
Unsaturated phosphorous(V)-based electrophiles have been associated molecular patterns (DAMP) as an indirect measure of
recently introduced as new cystein-selective bioconjugation immunogenic cell death (ICD) of our linker–payload, two features
reagents for the generation of highly efficacious ADCs. We have that are discussed as key drivers of efficacy of ADCs (34–37). Finally,
shown that they can easily be incorporated even into complex we demonstrate excellent in vivo features of our newly identified
molecules and they yield highly stable cysteine adducts with a high linker–payload structure, such as a tremendous increase in efficacy
selectivity for thiols (no modification of other amino acids than compared with Enhertu and antibody-like pharmacokinetics (PK)
cysteine can be observed) and fast reaction kinetics in the range of properties of the highly loaded ADCs with retained DAR8 even after
bioorthogonal strain promoted cycloaddition reactions (26–30). 21 days of circulation.

OF2 Mol Cancer Ther; 2023 MOLECULAR CANCER THERAPEUTICS


Efficacious Exatecan-based DAR8 Antibody–Drug Conjugates

Materials and Methods Bystander killing


For the co-culture–based bystander experiment, 10.000 HER2-
In vitro cytotoxicity
positive SKBR-3 and 2.000 HER2-negative MDA-MB-468 cells were
To investigate the cytotoxicity of the unconjugated small-molecule
treated with trastuzumab-LP5 DAR8 or Enhertu at concentrations
TOP1 inhibitors and ADCs, respective cells were incubated for 4 days
ranging from 0.05 to 3 mg/mL. After 5 days, cells were stained
(small molecules) and 7 days (ADCs) with increasing concentrations
with LIVE/DEAD Fixable Aqua (Thermo Fisher Scientific) and
of small molecules (0.015–1,000 nmol/L) and ADCs (0.05–3 mg/mL) to
HER2-PE (BioLegend) to distinguish between target-positive and
generate a dose–response curve. Killing was analyzed using resa-
-negative cells. The percentage of viable HER2-positive and -neg-
zurin cell viability dye at a final concentration of 55 mmol/L (Merck)
ative cells was analyzed by flow cytometry on a CytoFLEX S flow
by dividing the fluorescence from control cells in medium by the
cytometer (Beckman Coulter).
fluorescence of ADC-treated cells. Fluorescence emission at
590 nmol/L was measured on a microplate reader Infinite 200 Pro
DNA damage
(Tecan Group Ltd.).
The upregulation of DNA damage markers as response to TOP1-
inhibiting ADCs were analyzed by a flow cytometry–based readout.
Optimized procedure for the conjugation of LP5 to antibodies to
For this, 30,000 HER2-positive SKBR-3 cells were incubated for
achieve DAR8
24 hours to 72 hours with 0.5 or 5 mg/mL of ADCs. After the end
50 mL of the antibody solution at 10.0 mg/mL in P5-conjugation
of the incubation time, cells were stained with LIVE/DEAD Fixable
buffer (50 mmol/L Tris, 1 mmol/L EDTA, 100 mmol/L NaCl, pH 8.3 at
Aqua (Thermo Fisher Scientific) followed by fixation and permeabi-
room temperature) were mixed with 1.66 mL of a 10 mmol/L TCEP
lization using the BD Cytofix/Cytoperm Kit (BD Biosciences) accord-
[Tris(2-carboxyethyl)phosphine] solution (5 eq.) in P5-conjugation
ing to the manufacturer’s instructions. Intracellular staining of DNA
buffer. Directly afterwards, 0.83 mL of a 40 mmol/L solution of LP5
damage markers was performed using anti-cleaved PARP (Asp214)
dissolved in DMSO (10 eq.) were added. The mixture was shaken at
PE, anti-H2AX (pSer139) AF647, and anti-active caspase 3 FITC (all
350 rpm and 25 C for 16 hours. The reaction mixtures were purified by
BD Biosciences) or respective isotype controls. Cells were acquired on
preparative size-exclusion chromatography (SEC) with a Superdex 200
a CytoFLEX S flow cytometer (Beckman Coulter) and the percentage
Increase 10/300GL (Cytiva) and a flow of 0.8 mL/min eluting with
of positive cells was determined.
sterile PBS (Merck). The antibody-containing fractions were pooled
and concentrated by spin-filtration (Amicon Ultra 2mL MWCO:
Cell-cycle analysis
30 kDa, Merck).
To investigate changes in cell cycle upon TOP1 inhibitor treatment,
Determination of antibodies bound per cell 30,000 HER2-positive SKBR-3 cells were incubated for 24 to 48 hour
Antibodies bound per cell (ABC) were determined by staining of cell with 20 nmol/L of ADCs (3 mg/mL) or small molecules if not otherwise
lines with saturating concentrations of HER2-PE (BioLegend) or the indicated. After the end of the incubation time, cells were permeabi-
respective isotype control and acquisition by flow cytometry on a lized in 70% ethanol on ice, followed by staining in FxCycle PI/RNAse
CytoFLEX S flow cytometer. Quantification of ABCs as an estimation Staining Solution (Thermo Fisher Scientific). Cells were acquired on a
for surface receptor expression was performed by simultaneous acqui- CytoFLEX S flow cytometer and PI fluorescence was analyzed on a
sition of QuantiBRITE beads (BD Biosciences) conjugated with four linear scale (Beckman Coulter).
levels of PE and generation of a standard curve.
Release of DAMPs as a surrogate marker for ICD in vitro
Analysis of HER2-binding of trastuzumab-LP5 and enhertu As an indirect measure for ICD, calreticulin-positive cells, HMGB1
To determine equilibrium-binding constants (KD; as an avidity and ATP release were quantified. For all readouts, 30,000 HER2-
measurement), HER2-positive SKBR-3 cells were incubated with positive cells were incubated with 20 nmol/L of exatecan, DXd or
antibodies at increasing concentrations up to saturation and stained derived ADCs (3 mg/mL) for 24 to 48 hours if not otherwise indicated.
with an AF488-labeled goat anti-human IgG (HþL) secondary anti- Calreticulin staining of cells was performed using PE anti-calreticulin
body (Thermo Fisher Scientific). Cells were acquired by flow cyto- antibody (Abcam) or respective isotype control. Lumit HMGB1
metry on a CytoFLEX S flow cytometer (Beckman Coulter). Mean Human/Mouse Immunoassay (Promega) and RealTime-Glo Extra-
fluorescence intensity (MFI) ratios were determined by dividing the cellular ATP Assay (Promega) were used to quantify secreted HMGB1
MFI of the antibody-incubated cells by the fluorescence of the sec- and ATP into the media according to the manufacturer’s instructions.
ondary antibody control. Data points were analyzed by a non-linear Measurement of luminescence was performed on a microplate reader
regression using a one-site–specific binding model. Infinite 200 Pro (Tecan Group Ltd.).

Analysis of HER2-mediated internalization of trastuzumab-LP5 In vivo efficacy experiments


and enhertu The experiments were conducted in accordance with German
For pHrodo-based investigation of internalization, antibodies were animal welfare law and approved by local authorities. A total of
labeled with the pHrodo Deep Red Antibody Labeling Kit (Thermo 2106 NCI-N87 cells were subcutaneously injected into the flank of
Fisher Scientific) according to the manufacturer’s instructions. HER2- female CB17-Scid mice. Treatment was initiated when tumors reached
positive and -negative cell lines were incubated with 5 mg/mL pHrodo a mean tumor volume of 0.1–0.15 cm3. 10 animals per group were
Deep Red-antibodies for 1 hour at 37 C and acquired by flow treated once with either 0.25, 0.5, 1 or 2 mg/kg, trastuzumab-LP5
cytometry on a CytoFLEX S flow cytometer (Beckman Coulter). An DAR8 or Enhertu. 5 animals per group were treated with vehicle or
increase in MFI indicates the presence of aHER2 antibodies in late with 2 mg/kg palivizumab-LP5 as Isotype control, as intravenous
endosomal and lysosomal compartments. The MFI ratio was deter- injection after randomization into treatment and control groups.
mined by dividing the MFI of pHrodo-incubated cells by the MFI of Tumor volumes, body weights, and general health conditions were
unstained cells. recorded throughout the whole study.

AACRJournals.org Mol Cancer Ther; 2023 OF3


Schmitt et al.

Data availability immolative PAB moiety that facilitates traceless release of exate-
The data generated in this study are available upon request from the can (38). The linker–payload LP6 based on maleimide chemistry as
corresponding author. the most commonly used conjugation strategy was generated as a
control. To assess whether the exatecan moiety must be tracelessly and
efficiently cleaved to unleash its full activity, we also synthesized
Results construct LP7 without the PAB moiety and the non-cleavable LP8.
Design and evaluation of TOP1-based linker–payloads for highly The absence of the lipophilic PAB moiety in LP7 and LP8 has the
loaded and efficacious ADCs advantage of reduced linker–payload hydrophobicity to potentially
We started our investigation with the selection of an appropriate reduce ADC aggregation.
camptothecin derivative. For this, we tested the unconjugated pay- With LP1–8 in hands, we continued our studies with conjugation
loads, highlighted in orange in the linker–payload structures depicted of the linker–payload to trastuzumab, a HER2-targeting antibody
in Fig. 1, for their antiproliferative potency on a range of different cell of the same amino acid sequence that is used in the antibody of
lines (Fig. 2; Supplementary Table S1).The figure clearly shows that of Enhertu (39). Antibody conjugation has been conducted in accordance
the three TOP1 inhibitors tested in this study, exatecan is the most to previously published procedures (31). A large excess of the linker–
potent with a factor of 2–10-fold potency difference in the IC50 value of payloads and TCEP reduction agents has been used in those first
cell viability compared with SN38 and DXd. Next, a series of different conjugation experiments to force the conjugation reaction and ensure
linker–payloads, shown in Fig. 3 based on exatecan, including a the highest possible conjugation yield. After conjugation and purifi-
maleimide-control ADC, has been synthesized. The dipeptidyl- cation from residual linker–payload, TCEP, and HMWS via semi-
based linker–exatecan constructs LP1–5 carry the well-known VC preparative SEC into PBS, the corresponding ADCs were analyzed for
and valine–alanine (VA) peptides in combination with the self- yield based on protein concentration before and after the conjugation,

Figure 2.
Dose–response curves for DXd, SN38, and exatecan on a range of different cell lines.

OF4 Mol Cancer Ther; 2023 MOLECULAR CANCER THERAPEUTICS


Efficacious Exatecan-based DAR8 Antibody–Drug Conjugates

Figure 3.
Depiction of the linker–payload (LP) structures used herein. Functional elements are highlighted. Hydrophilic substituent, purple; conjugation handle, gray; cleavage
side, green; release handle, orange; exatecan, black.

DAR via mass spectrometry (MS), formation of undesired HMWS via Table 1 shows that incorporating PEG chains into the VC–PAB–
SEC and targeted antiproliferative effect on two different HER2- exatecan-based linker–payload systems can improve the conjugation
positive (SKBR-3 and HCC-78) and one HER2-negative cell line efficiency and simultaneously reduce aggregation of the derived ADCs.
(Table 1; Supplementary Fig. S1). Both effects are depending on the PEG length, as shown by linker–
Interestingly, after conjugation of the phosphonamidate-O-ethyl– payloads LP2, LP3, and LP5, with 2, 12, and 24 PEG monomer units,
substituent linker–payload LP1 to trastuzumab, we observed only a respectively. It could be demonstrated that an increase in PEG length
minor conversion of the antibody to a DAR of 0.28, even with a large led to a higher DAR, less aggregation during the conjugation process
excess of 30 equivalents of LP1 with respect to the antibody. In addition and consequently to an increase in the yield of monomeric protein after
to this, we observed a high percentage of HMWS formation of almost conjugation and to an increase in in vitro efficacy on the targeted cell
10% during the conjugation process (Table 1). To exclude slower lines (Table 1; Supplementary Fig. S1). Switching from a VC LP3 to a
reaction kinetics of thiol addition to ethynylphosphonamidates, VA LP4 did not improve the conjugation behavior (Table 1). Only the
compared with very fast maleimide conjugation (30, 40), being the PEG24-carrying linker–payload LP5 led to full conjugation of tras-
reason for this low conjugation yield, we also tried the maleimide- tuzumab to a DAR8 ADC without any sign of HMWS being formed
based linker–payload LP6 for conjugation. However, also LP6 only during the conjugation process resulting in the highest in vitro cyto-
resulted in a very minor conjugation to a DAR of 0.12 in the toxicity on HER2-positive SKBR-3 and HCC-78 among all tested
monomer fraction after conjugation with almost 20% of HMWS linker–payloads. We also synthesized ADCs from linker–payload LP7,
being formed (Table 1). which is lacking the PAB moiety. Interestingly, LP7 yields ADCs with a

Table 1. Overview of the properties of the ADCs, synthesized from the HER2-targeting trastuzumab conjugated to LP1–8.

Yield after DAR (MS) Cell killing SKBR-3 Cell killing HCC-78
purification After
based on purification Max. Max.
protein from HMWS after EC50 amount of EC50 amount of
LP Linker–payload concentration HMWS conjugation (%) (ng/mL) dead cells (ng/mL) dead cells

LP1 P5(OEt)–VC–PAB–exatecan 59% 0.28 9.9% 251 37% >3,000 N/A


LP2 P5(PEG2)–VC–PAB–exatecan 82% 5.51 2.3% n.d. n.d. n.d. n.d.
LP3 P5(PEG12)–VC–PAB–exatecan 80% 6.36 3.9% 15.5 86% 144.1 91%
LP4 P5(PEG12)–VA–PAB–exatecan 84% 4.39 7.5% 18.0 78% 121.2 68%
LP5 P5(PEG24)–VC–PAB–exatecan 99% 8.00 0.9% 12.5 96% 97.6 88%
LP6 MC–VC–PAB–exatecana 75% 0.12 19.2% 265 14% >3,000 N/A
LP7 P5(PEG12)–VA–exatecan 77% 7.89 1.0% 114.7 44% >3,000 N/A
LP8 P5(PEG12)–exatecan 97% 7.19 4.0% 182.4 35% >3,000 N/A

Note: The conjugation has been performed with excess linker–payload (30 eq. with respect to the antibody) and TCEP (10 eq. with respect to the antibody). After
purification, the ADCs have been analyzed for yield based on protein concentration before and after the conjugation, DAR via MS, ADC aggregation via analytic
SEC and antiproliferative effect on HER2-positive cell-lines (SKBR-3 and HCC-78). The antiproliferative potency of the ADCs on the HER2-positive cells is reported
as an EC50 value of cell viability in ng/mL and the total amount of affected cells in percentage. None of the ADCs showed any effect on the HER2-negative cell line
(MDA-MB-468, Supplementary Fig. S1).
a
Conjugation conditions for LP6 were slightly adjusted to more typical maleimide reaction conditions: Reaction was conducted in PBS at pH 7.4 at room temperature
for 2 hours; n.d., not determined; N/A, not applicable.

AACRJournals.org Mol Cancer Ther; 2023 OF5


Schmitt et al.

very high DAR and almost no HMWS being formed during conju- showed the least tendency to form HMWS under the tested conditions,
gation, in contrast with LP4, which is the same linker–payload even compared with Enhertu.
structure but includes the PAB moiety. However, we observed that To compare ADCs from LP3 and LP5 in their in vivo PK behavior,
ADCs from LP7 and LP8, even though high in DAR, showed only Sprague Dawley rats were treated with ADCs carrying 8 linker–
modest anticancer activity in the antigen-positive cell lines (Table 1; payload molecules, and their clearance behavior was monitored over
Supplementary Fig. S1). time via analysis of blood samples. It must be noted that both
experiments have been performed with different antibodies conjugat-
Biophysical and in vivo characterization of the two best ed to LP3 and LP5. However, because both antibodies are not cross-
performing exatecan-based linker–payloads reactive in rodents, it is anticipated that the influence on the clearance
Next, we continued our efforts in designing novel camptothecin- behavior of the ADCs can be neglected. The samples at various time
based linker–payload structures with a more thorough in vitro and points were analyzed by ELISA for total ADC with an antibody capture
in vivo investigation of ADCs originating from linker–payload LP3 step and for intact ADC, capturing with an anti-exatecan antibody.
and LP5, carrying either 12 or 24 PEG units, respectively. To allow for a The high overlap between those two curves again highlights the great
fair comparison between the linker systems, DAR8 ADCs were serum stability of the ehynylphosphonamidate linker structure and is
synthesized out of both linker–payload structures (Fig. 4A). For this, in line with previous reports (31, 32). Interestingly, the stronger
equivalents of LP3 were increased from 30, that were used to produce aggregation tendency of ADCs synthesized from LP3 did not drasti-
the results in Table 1, to 50 and the monomeric ADCs were isolated via cally increase the in vivo clearance of the ADCs (Fig. 4D). Moreover,
preparative SEC. The desired ADCs were characterized via SEC and HIC analysis of ADCs from LP3 showed only a modest increase in
hydrophobic interaction chromatography (HIC; Fig. 4B). Trastuzu- overall hydrophobicity compared with MMAE-based ADCs (Supple-
mab-LP5 DAR8 demonstrated best characteristics, because it had the mentary Fig. S3).
highest degree of monomer content compared with trastuzumab-LP3
and Enhertu, judged by SEC. Interestingly, the overall hydrophobicity In vitro head-to-head comparison of the best performing linker–
increase of the DAR8 constructs compared with the unconjugated payload to enhertu
antibody was negligible, judged by the HIC retention time (Supple- With our main goal being to develop a novel superior camptothecin-
mentary Fig. S2) and similar HIC retention times were measured for based linker–payload structure, we continued our investigations with
trastuzumab-LP3 DAR8, trastuzumab-LP5, and Enhertu (Fig. 4B). ADCs that originate from LP5, because this linker–payload structure
The aggregation behavior of trastuzumab-based DAR8 ADCs from combines high conjugation yields in DAR and antibody recovery, very
LP3 and LP5 was more thoroughly monitored over several weeks at low aggregation tendency, highest in vitro activity and excellent in vivo
low (4 C) and physiological temperatures (37 C) and Enhertu was stability and clearance behavior. To ensure manufacturability also on a
added as a comparison (Fig. 4C; Supplementary Fig. S2). The results higher scale, we first investigated how many equivalents of LP5 are
clearly show that ADCs conjugated to LP3 have a stronger tendency to needed to synthesize DAR8 ADCs. On the basis of previous experience,
aggregate under both conditions compared with Enhertu and ADCs we determined the DAR as a function of equivalents of LP5 and
conjugated to LP5. It should be noted that DAR8 ADCs from LP5 equivalents of TCEP in the conjugation reactions in a pH 8.3 buffer

Figure 4.
Head-to-head comparison between DAR8 ADCs from LP3 and LP5 and Enhertu. A, Structure of DAR8 ADCs from LP3 and LP5. B, Comparison of the physicochemical
properties of trastuzumab-LP5 (orange), trastuzumab-LP3 (purple), and Enhertu (gray). SEC analysis for the formation of aggregation (top) and HIC analysis
to investigate overall hydrophilicity of the DAR8 constructs (bottom). C, Monitoring of increase in aggregation over 4 weeks of trastuzumab-LP3 (orange),
trastuzumab-LP5 (purple), and Enhertu (gray). All three ADCs were formulated in the same buffer system at 1 mg/mL. Samples were analyzed by SEC for HMWS after
1, 2, and 4 weeks of incubation at 4 C (left) and 37 C, right. Raw SEC chromatograms are shown in Supplementary Fig. S3. Aggregates were normalized to day 0 by
subtraction, because trastuzumab-LP5, already contained 8% HMWS at day 0. D, PK analysis of Sprague Dawley rats that were treated at day 0 with brentuximab-LP5
(left, purple) or IgG1-LP5 (right, orange) at day 0. Samples were drawn after several time points throughout the 3 week study period (9 animals per group, 3 animals
were sampled per time point). Shown are mean and SD out of the three animals. The serum samples were analyzed by ELISA for total mAb (dotted line) and intact
ADC (straight line). Black dotted line corresponds to the lower limit of quantification of the assays.

OF6 Mol Cancer Ther; 2023 MOLECULAR CANCER THERAPEUTICS


Efficacious Exatecan-based DAR8 Antibody–Drug Conjugates

system at 10 mg/mL (31). We found out that under those conditions, as described earlier (30–32) and could be confirmed here in the direct
less as 10 equivalents with respect to the mAb (1.25 eq. per Cys) are comparison of retained DAR after several days of incubation in rat
enough to generate a DAR8 ADC (Supplementary Fig. S4). Moreover, serum (Fig. 6A). Moreover, we performed an in vivo stability head-to-
we tried to reduce the pH of the conjugation reaction. Here, it could be head comparison between trastuzumab-LP5 DAR8 and Enhertu.
shown that the reaction kinetics are pH-dependent. Full conversion to Here, mice were treated with 20 mg/kg of each ADC at day 0 and
DAR8 is still possible at pH 7.8, when the reaction time is increased to blood samples were drawn after 1, 3, 5, and 7 days after treatment.
24 hours (Supplementary Fig. S4). Serum samples were analyzed by ELISA for total ADC exposure,
After optimization of the conjugation conditions with LP5, the showing a similar in vivo clearance behavior of trastuzumab-LP5
conjugation to trastuzumab was scaled to 10 grams of mAb and the compared with Enhertu, with a slightly higher exposure for tras-
DAR8 ADC was purified via tangential flow filtration only. Analytic tuzumab-LP5, 7 days after treatment. Moreover, the ADCs were
data of the upscaling runs were identical to what has been observed on captured from the sera by immunoprecipitation and analyzed via
small scale. The DAR8 trastuzumab-LP5 ADC has been further MS for DAR. The results clearly indicate that the ex vivo stability
investigated in head-to-head comparisons with Enhertu. For this also translates to an in vivo setting, showing that trastuzumab-LP5
purpose, Enhertu has been purchased in pharmaceutical grade. First, is still connected to 8 linker–payload molecules after one week of
a broader panel of HER2-positive cell lines and one HER2-negative cell circulation, whereas Enhertu’s DAR is reduced to approximately 5
line were treated with either trastuzumab-LP5 or Enhertu. The results (Fig. 6B). The main route of drug loss that has been observed for
show that the in vitro efficacy is equal or superior in all HER2-positive Enhertu is via retro-Michael addition, leading to the detection of the
cell lines (Fig. 5A). Less unspecific killing was observed in the non- unmodified cysteines by MS.
targeted antigen-negative cell line L-540. In general, we were able to Encouraged by these results, we wanted to investigate the influence
observe an expected correlation between ABCs and in vitro cytotox- of the reduction in DAR on the in vitro efficacy of the constructs,
icity of both ADCs (Fig. 5B). Of note, only minor differences in because we have already shown in Table 1 that a lower DAR can
binding on SKBR-3 (KD trastuzumab-LP5: 475.1 ng/mL, Enhertu: lead to a reduction in cell killing efficiency. Therefore, we took the
420.6 ng/mL) and internalization on HER2-positive and -negative cell serum-stressed samples containing trastuzumab-LP5 DAR8 and
lines were observed between trastuzumab-LP5 and Enhertu (Fig. 5C). Enhertu and performed a cell viability experiment on a HER2-positive
Next, we investigated the bystander capacity, a potential feature of and a HER2-negative cell line (Fig. 6C). The results show that the
ADCs to compensate for heterogenous target expression, in depen- reduction in DAR that originates from a retro-Michael–based insta-
dency of the concentration of trastuzumab-LP5 DAR8 and Enhertu. bility of Enhertu also directly leads to a reduction in cell killing of the
Thereby, non-targeted HER2-negative MDA-MB-468 cells (47 ABCs) antigen-positive cell line after prolonged serum incubation. As
were co-cultured with HER2-positive SKBR-3 (790520 ABCs) fol- expected, this effect is much less pronounced for the stably conjugated
lowed by the assessment of cell viability of the HER2-negative cell only trastuzumab-LP5 DAR8.
via flow cytometry. The MDA-MB-468 cells were only affected in
viability when co-cultured with SKBR-3. A tendency toward lower IC50 In vivo efficacy head-to-head comparison with enhertu and PK
values and higher number of cells being affected was visible for analysis
trastuzumab-LP5 compared with Enhertu (Fig. 5D). Moreover, we Next, we performed an in vivo experiment, in which we compared
characterized the mechanism of cell killing induced by trastuzumab- the efficacy of trastuzumab-LP5 DAR8 to that of Enhertu over 4
LP5 and Enhertu in more detail. We have shown by flow cytometry different dose levels. 10 mice per group were implanted with tumors
that trastuzumab-LP5 DAR8 and Enhertu are both inducing DNA derived from the HER2-positive N87-cell line and treated once with
damage resulting from TOP1 inhibition to a similar extent after either 0.25, 0.5, 1 or 2 mg/kg at day 0, once the tumors have reached a
treatment of SKBR-3 cells with 0.5 mg/mL of the ADCs as shown by volume of 0.1–0.15 cm3. The N87-cell line was chosen because no
the upregulation of phosphorylated histon 2AX, activated caspase 3, significant difference in in vitro efficacy between both ADCs was
and cleaved PARP (Fig. 5E; Supplementary Fig. S6). Another mech- observed (Fig. 5A). We anticipated that thereby the true impact of the
anism of action of TOP1 inhibition is the induction of cell-cycle arrest, linker technology can be assessed without being biased by the more
which has been shown in cells treated with trastuzumab-LP5 DAR8, potent payload that was chosen in LP5 compared with deruxtecan. The
Enhertu and also free payloads (Fig. 5F; Supplementary Fig. S6). Cells results show a clear benefit in efficacy for trastuzumab-LP5 over all 4
were arrested mainly in S phase, but also in G2–M phase at selected dose levels tested (Fig. 6D). It should be highlighted that trastuzumab-
concentrations and incubation times. Another important feature for LP5 DAR8 is more effective at 0.5 mg/kg than Enhertu at 1 mg/kg.
cytotoxic payloads (also described for TOP1 inhibitors) is the Moreover, at 1 mg/kg, no complete remissions were observed in
induction of ICD, a process by which the drug-induced apoptosis Enhertu in contrast with 80% for trastuzumab-LP5 DAR8. At the
of a tumor cell releases DAMPs and thereby stimulates the immune highest dose level of 2 mg/kg, a non-targeted isotype control has been
system. For this purpose, SKBR-3 cells were treated with Enhertu, included, which did not show an effect with similar tumor growths
trastuzumab-LP5 or the respective payloads DXd and exatecan, than in the vehicle control, clearly underlining the target selectivity of
followed by the quantification of cells exposing calreticulin on the LP5 when conjugated to an antibody.
surface and releasing ATP and HMGB1 (Fig. 5G; Supplementary Finally, we conducted a more comprehensive PK analysis of ADCs
Fig. S7), all of which are DAMPs and surrogate markers for ICD. based on LP5 in Sprague Dawley rats. Previously, high DAR ADCs of
Indication for induction of ICD was shown by an increase in payloads such as MMAE have been shown to rapidly clear from
exposed calreticulin on the surface and ATP and HMGB1 release circulation (41). Here, two groups of rats were treated with 10 mg/kg
for both ADCs in a similar manner. of either an unconjugated IgG1 or a DAR8 conjugate of that antibody
(IgG1-LP5 DAR8). Blood samples from those groups were taken over a
Stability and in vivo PK head-to-head comparison with enhertu time course of three weeks and analyzed via ELISA for total antibody
The high stability of ethynylphosphonamidate-linked ADCs, in and total and intact ADC. Remarkably, we observed a very similar
particular in comparison with maleimide chemistry, has been clearance profile of the IgG1 antibody, and the same antibody

AACRJournals.org Mol Cancer Ther; 2023 OF7


Schmitt et al.

Figure 5.
Cell-based in vitro efficacy experiments of trastuzumab-LP5 DAR8 (orange) or isotype-LP5 DAR8 (black) compared with Enhertu (gray). A, Cell viability on seven
HER2-positive cell lines (N87, SKBR-3, HCC-78, SK-OV-3, OE-19, SK-GT-2, and HCC-1569) and one HER2-negative cell line L-540. B, Correlation between HER2-
receptor level on the tested cell lines and antiproliferative effect (EC50 in ng/mL) for trastuzumab-LP5 DAR8 (left, orange) and Enhertu (gray, right). C, Binding of
trastuzumab-LP5 and Enhertu on SKBR-3 and internalization on SKBR-3, N87 (HER2þ) and MDA-MB-468 (HER2). D, Bystander killing of non-targeted HER2-
negative MDA-MB-468 cells in co-culture with HER2-positive SKBR-3 (right). Shown is cell viability in dependency of the ADC concentrations. No effect was observed
in MDA-MB-468 only cultures (left). EC) Relative quantification of histone H2AX phosphorylation (left), active caspase 3 (middle) and cleaved PARP (right) after
treatment of SKBR-3-cells with 500 ng/mL of the ADCs for 3 days versus untreated. D–F, Relative quantification of cells in S-, sub-G0, G0–G1, and G2–M phase
after treatment with 3,000 ng/mL of the ADCs for 2 days versus untreated. E–G, Secretion of DAMPs as a surrogate marker for measurement of immunogenic
cell death as indicated by calrecticulin exposure on cell surface (left), ATP release (middle), and HMGB1 release (right). Shown are results after treatment
with 3,000 ng/mL (¼ 20 nmol/L) of the trastuzumab-LP5 DAR8 (orange, solid) or Enhertu (gray, solid) and with 20 nmol/L of unconjugated exatecan (orange,
checkered) or DXd (gray, checkered) for 2 days versus untreated (black). Graphs show means of n ¼ 2  SEM.

OF8 Mol Cancer Ther; 2023 MOLECULAR CANCER THERAPEUTICS


Efficacious Exatecan-based DAR8 Antibody–Drug Conjugates

Figure 6.
Stability experiments, in vivo efficacy and PK analysis of trastuzumab-LP5 DAR8, (orange) compared with isotype-LP5 DAR8 (yellow), vehicle (black), and
Enhertu (gray). A, DAR ratio measured by MS after 0, 1, 3, and 7 days of incubation in at least 80% rat serum at 37 C. B, PK analysis of blood samples from mice
being treated with 20 mg/kg. Samples have been drawn after 1, 3, 5, and 7 days of circulation. Exposure by total ADC ELISA (top). DAR measured by MS
(bottom). C, Cell viability experiments of ADC samples after 0, 1, 3, and 7 days of incubation in at least 80% rat serum at 37 C on the HER2-positive cell line
SKBR-3 (left) or HER2-negative cell line MDA-MB-468 (right). D, Female SCID mice have been implanted with cell-culture–derived xenograft model based on
N87 cells in Matrigel. Treatment was started once when tumor volumes reached 0.1–0.15 cm3. Mice were treated at day 0 with a single dose of either vehicle, 2,
1, 0.5, and 0.25 mg/kg trastuzumab-LP5 DAR8, or Enhertu. At the highest dose of 2 mg/kg, a non-targeting isotype control ADC Isotype-LP5 DAR8 was
included. Each group consisted of n ¼ 10 females each. F, Sprague Dawley rats were treated at day 0 with 10 mg/kg of either IgG1 (left) or IgG1-LP5 DAR8
(right) at day 0. Samples were drawn after several time points throughout the 3 week study period (9 animals per group, 3 animals were sampled per time
point). Shown are mean and SD out of the three animals. The serum samples were analyzed by ELISA for total mAb/total ADC (dotted line) and intact ADC
(straight line). G, DAR measured by MS of blood samples from rats being treated with 10 mg/kg of IgG1-LP5 DAR8. Black dotted line corresponds to the lower
limit of quantification of the assays.

conjugated to 8 molecules of LP5, judged by the similarity of the


profiles for total ADC and total antibody and of the AUC values Discussion
(Fig. 6E). This again highlights the negligible influence of the With this study, we aimed to develop a superior TOP1 Inhib-
conjugation of 8 molecules of linker–payload LP5 to the antibody itor-based linker–payload platform for the construction of high
leading to a high DAR ADC with mAb-like properties. Once more, DAR ADCs. Our studies demonstrate antibody-like properties
excellent stability could be confirmed by overlapping total ADC and for highly loaded phosphonamidate-linked exatecan-containing
intact ADC. This could be verified by an MS assay, which revealed a ADCs, thus providing a promising alternative for existing link-
DAR8 circulating in rats even 21 days after administration of the er–payload structures used in marketed ADCs such as Trodelvy
ADC (Fig. 6F). and Enhertu.

AACRJournals.org Mol Cancer Ther; 2023 OF9


Schmitt et al.

On the basis of our findings of exatecan being more potent than reduced in in vitro efficacy in comparison with conjugates that carry
SN38 and DXd and with the intention to design TOP1 inhibitor-based the PAB release handle. This observation is in line with early inves-
ADCs with superior efficacy, we decided to construct linker–payload tigations of Trail and coworkers that the PAB moiety can be crucial for
systems that use exatecan as the cytotoxic payload. With the aim to efficient release of the payload (33). Very similar behavior was
compensate for a potential toxicity risk, which is often associated with observed for the non-cleavable linker–payload LP8, clearly indicating
ADCs based on more potent payloads and linker instability (13, 14, 42), the necessity of an efficient release handle for a potent antibody-
we decided to make use of ethynylphosphonamidates for antibody mediated delivery of exatecan into the targeted cell.
conjugation. This linker class has been previously shown to exhibit In a head-to-head comparison of the suboptimal PEG12-based LP3
superior stability in head-to-head comparisons with approved with the superior PEG24-based LP5, we have furthermore shown that
ADCs (32, 43, 44). Hence, we synthesized several linker–exatecan conjugation of 8 VC–PAB–exatecan molecules to an antibody is
constructs with and without the traceless PAB-release unit in the neither drastically increasing the overall hydrophobicity of the anti-
presence or absence of different dipeptidyl cleavage sides, with the bodies, judged by HIC retention, nor the in vivo clearance rates, which
addition of PEG solubilizing groups of various lengths. VC–PAB– makes aggregation during conjugation and low conjugation yields the
exatecan constructs with shorter PEG chains than 12 units showed a main challenge to be solved for VC–PAB–exatecan. This is contrary to
strong aggregation tendency and led to low conjugation yields. This the observation by us and others that VC–PAB–MMAE DAR8 ADCs
stands in strong contrast with what has previously been observed with clear rapidly from circulation in vivo, when not adequately balanced
other ethynylphosphonamidate-based linker–payloads (30, 32, 40) with solubility enhancers (18, 41). We interpret this as being another
and led to our conclusion that the VC–PAB–exatecan moiety, as a hint toward our hypothesis that not the overall hydrophobicity of VC–
very hydrophobic linker–payload group itself, exhibits the following PAB–exatecan-based ADCs lead to aggregation and low conjugation
challenges: (i) Low aqueous solubility leading to decreased available but rather the strong tendency of larger aromatic molecules to build
linker–payload concentrations under the aqueous conjugation con- strong intermolecular p–p interactions. In previous work, we have
ditions and (ii) ADCs, once formed, tend to form HMWS, leading mainly used the P5(PEG) moieties to improve in vivo PK of high-DAR
again to reduced DARs in the monomeric fraction. Attempts to reduce ADCs (31, 32). Here, we demonstrate that such P5(PEG) building
this aggregation by switching from a VC LP3 to a VA linker–payload blocks can also be applied to efficiently reduce ADC aggregation and
LP4 did not improve the conjugation behavior, which is in agreement facilitate conjugation, which generally underlines the beneficial fea-
with previous reports on the differences between those two dipep- tures of branched PEG-based linker systems.
tides (41). To reduce the overall hydrophobicity of the linker–pay- With the identification of phosphonamidate-linked P5(PEG24)–
loads, we incorporated PEG chains at the phosphonamidates-O- VC–PAB–exatecan LP5 as an optimal linker–payload system, we
substituent (31). This strategy has been previously reported for ADCs improved the conjugation conditions to be able to use as little as
to improve the PK profile of MMAE-based ADCs, because it reduces 1.25 equivalents per cysteine to generate a DAR8 ADC from an IgG1
clearance from circulation mediated by the overall hydrophobicity of antibody. Next, a comprehensive in vitro head-to-head comparison
the ADC (18, 19). Notably, selective potency on the two tested HER2- between trastuzumab-LP5 DAR8 and Enhertu has been performed.
positive, targeted cancer cell lines also increased with longer PEG We could confirm that both ADCs exhibit the same mechanism
chains, observable by a shift of the IC50 value to lower concentration of cell killing as a result of TOP1 inhibition, demonstrated by upre-
and an increase in the maximum amount of cells that have been gulation of DNA damage markers such as phosphorylated histon 2AX,
affected by the ADC. Most likely this is a function of the higher drug- activated caspase 3, and cleaved PARP (47). Moreover, in accordance
load that could be achieved with higher PEG length and is in line with with literature data, we showed both S and G2–M arrest as response
previous reports on higher DAR-ADCs having a higher potency to TOP1 inhibition (48, 49). We also investigated the capability of
in vitro (41, 45). TOP1-based ADCs to elicit ICD, a process by which a cytotoxic drug
ADC aggregation arising during the conjugation process was a induces apoptosis of a tumor cell that causes stimulation of the
major issue that prevented the formation of high DAR ADCs for some immune system. Increase of immunostimulatory DAMPs has been
of the constructs. It should be noted that HMWS formation was not previously described to accompany and precede ICD and serves as a
observed in the past when VC–PAB–MMAE or VC–PAB–MMAF was surrogate marker for the latter (50, 51). Immunostimulatory activity
used instead of VC–PAB–exatecan, even with shorter PEG substitu- has been described for Enhertu and could be confirmed in the
ents at the ethynylphosphonamidate (30, 32). The observation on current study via an in vitro experiment. Such activation of the
aggregation being induced by the VC–PAB–exatecan moiety is sup- immune system can lead to a long-lasting protective antitumor
ported by studies of Burke and colleagues (20), who have also reported immunity and opens a highly promising road for combination of
strong tendencies for HMWS formation with other maleimide–VC– ADCs with immuno-oncology agents (52, 53). The release of
PAB–camptothecin constructs. For deruxtecan, the linker–payload of DAMPs from trastuzumab-LP5 in similar quantities than Enhertu
Enhertu with a very similar camptothecin core structure compared indicates the immunostimulatory effect of exatecan when delivered
with exatecan but different in linker, there have not been reports on via LP5 conjugated to a targeted antibody.
problematic aggregation behavior even at high DARs (22). We there- Higher in vitro potency of trastuzumab-LP5 compared with
fore suspected that the origin of the aggregation and solubility issue Enhertu could be observed in some cell lines. Because we demon-
lays in the combination of exatecan with the p-aminobenzyl moiety, strated similar HER2 binding for both ADCs, we attribute this to
leading to moieties that combine several p-electron systems, poten- differences in potency of the attached payloads, but not to an
tially leading to flat, ridged PAB–exatecan moieties. Strong p–p influence of the attached linker on the properties of the antibody.
interactions between molecules that carry several aromatic cores are Bystander killing is considered to play a key role in the effective
generally known to impair solubility (46). Indeed, LP7, which is in treatment of solid tumors to compensate for heterogenous target
comparison with LP4 lacking the PAB moiety is not inducing aggre- expression (54). Enhertu has been described to exhibit highly potent
gation even at high DAR. However, ADCs from LP7 are also clearly bystander activity, in particular when compared with ADCs with

OF10 Mol Cancer Ther; 2023 MOLECULAR CANCER THERAPEUTICS


Efficacious Exatecan-based DAR8 Antibody–Drug Conjugates

other camptothecin-based linker–payload systems and to trastuzu- Affairs and Energy (German Accelerator), Federal Ministry of Education and
mab-DM1 (23). Here, we even observed a slightly higher bystander Research (Rahmenprogramm Gesundheitsforschung), and LMU Munich outside
the submitted work; and Tubulis GmbH sponsored the work and is owner of the
killing effect for trastuzumab-LP5 over Enhertu. Most importantly,
relevant patents listed above; as well as reports employment with Tubulis GmbH. P.
we were able to show a high increase of trastuzumab-LP5 in serum Cyprys reports grants from Federal Ministry for Economic Affairs and Energy (EXIST
stability of the linker in vitro and in vivo. This results in retained Forschungstransfer II), Federal Ministry for Economic Affairs and Energy (German
in vitro activity of trastuzumab-LP5 DAR8 even after several days of Accelerator), Federal Ministry of Education and Research (Rahmenprogramm
serum stress, whereas cell killing was drastically reduced for Gesundheitsforschung), and LMU Munich outside the submitted work; and Tubulis
Enhertu in this setting. The effect of DAR reduction and unstable GmbH sponsored the work and is owner of the relevant patents listed above; as well as
reports employment with Tubulis GmbH. M. Gerlach reports grants from Federal
linker technology in circulation on the safety of an ADC is still
Ministry for Economic Affairs and Energy (EXIST Forschungstransfer II), Federal
under debate (1). However, those results rather suggest a retained Ministry for Economic Affairs and Energy (German Accelerator), Federal Ministry of
efficacy over time in circulation of ADCs conjugated to LP5, in Education and Research (Rahmenprogramm Gesundheitsforschung), and LMU
particular when compared with unstably conjugated maleimides as Munich outside the submitted work; and Tubulis GmbH sponsored the work and
used in Enhertu. is owner of the relevant patents listed above; as well as reports employment
Moreover, we were able to show that the superiority of trastu- with Tubulis GmbH. P. Ochtrop reports grants from Federal Ministry for Economic
Affairs and Energy (EXIST Forschungstransfer II), Federal Ministry for Economic
zumab-LP5 in cell killing, bystander activity, and serum stability of
Affairs and Energy (German Accelerator), Federal Ministry of Education and
the linker in vitro and in vivo also translates into improved in vivo Research (Rahmenprogramm Gesundheitsforschung), and LMU Munich outside
efficacy. Trastuzumab-LP5 DAR8 revealed a more potent targeted the submitted work; and Tubulis GmbH sponsored the work and is owner of the
effect over four tested dose levels, with a drastically increased relevant patents; as well as reports employment by Tubulis GmbH. C.P.R. Hack-
activity even when half of the dose has been administered. We enberger reports grants from Federal Ministry of Education and Research (Rahmen-
explain this with a slightly better PK profile, with less clearance and programm Gesundheitsforschung), Leibniz Competition (SAW), and German Sci-
ence Foundation (DFG) outside the submitted work; as well as reports a patent for
very importantly higher in vivo stability, which taken together helps
WO2018041985 issued. D. Schumacher reports grants from Federal Ministry for
to deliver the payload more efficiently to the site of the tumor. We Economic Affairs and Energy (EXIST Forschungstransfer II), Federal Ministry for
conclude that those features can be attributed to the linker prop- Economic Affairs and Energy (German Accelerator), Federal Ministry of Education
erties rather than exatecan being more potent payload compared and Research (Rahmenprogramm Gesundheitsforschung), and LMU Munich outside
with DXd, because we chose a cell line in which both ADCs had a the submitted work; as well as reports a patent for WO2018041985 issued and
highly comparable efficacy in vitro. WO2023083919 pending; and Tubulis GmbH sponsored the work and is owner of the
relevant patents listed above; and reports employment with Tubulis GmbH. J. Helma
Finally, it could be shown in an in vivo PK study that LP5 can be used
reports grants from Federal Ministry for Economic Affairs and Energy (EXIST
to synthesize DAR8 ADCs with antibody-like clearance behavior in Forschungstransfer II), Federal Ministry for Economic Affairs and Energy (German
combination with highest stability, judged by the detection of a fully Accelerator), Federal Ministry of Education and Research (Rahmenprogramm
conjugated DAR8 ADC after 21 days of circulation in a living Gesundheitsforschung), and LMU Munich outside the submitted work; as well as
organism. We believe that phosphonamidate-linked exatecan pay- reports a patent for WO2018041985 issued and WO2023083919 pending; and
loads LP5 can be used to reduce last drawbacks of the already well- Tubulis GmbH sponsored the work and is owner of the relevant patents listed above;
and reports employment with Tubulis GmbH. A.M. Vogl reports grants from Federal
functioning deruxtecan platform used in Enhertu for a next-
Ministry for Economic Affairs and Energy (EXIST Forschungstransfer II), Federal
generation of TOP1-based ADCs of highest therapeutic activity over Ministry for Economic Affairs and Energy (German Accelerator), Federal Ministry of
a broad span of different tumor antigen targets. Education and Research (Rahmenprogramm Gesundheitsforschung), and LMU
Munich outside the submitted work; as well as reports a patent for WO2023083919
pending; and Tubulis GmbH sponsored the work and is owner of the relevant patents
Authors’ Disclosures listed above; and reports employment with Tubulis GmbH. M.-A. Kasper reports
S. Schmitt reports grants from Federal Ministry for Economic Affairs and Energy grants from Federal Ministry for Economic Affairs and Energy (EXIST Forschung-
(EXIST Forschungstransfer II), Federal Ministry for Economic Affairs and Energy stransfer II), Federal Ministry for Economic Affairs and Energy (German Acceler-
(German Accelerator), Federal Ministry of Education and Research (Rahmenpro- ator), Federal Ministry of Education and Research (Rahmenprogramm Gesundheits-
gramm Gesundheitsforschung), and LMU Munich outside the submitted work; as forschung), and LMU Munich outside the submitted work; as well as reports a patent
well as reports a patent for WO2023083919 pending; and Tubulis GmbH sponsored for WO2023083919 pending and WO2018041985 issued; and Tubulis GmbH
the work and is owner of the relevant patents listed above; and reports employment sponsored the work and is owner of the relevant patents listed above; and reports
with Tubulis GmbH. P. Machui reports grants from Federal Ministry for Economic employment with Tubulis GmbH.
Affairs and Energy (EXIST Forschungstransfer II), Federal Ministry for Economic
Affairs and Energy (German Accelerator), Federal Ministry of Education and Authors’ Contributions
Research (Rahmenprogramm Gesundheitsforschung), and LMU Munich outside
S. Schmitt: Data curation, formal analysis, investigation, writing–review and
the submitted work; as well as reports a patent for WO2023083919 pending; and editing. P. Machui: Formal analysis, investigation. I. Mai: Data curation, investiga-
Tubulis GmbH sponsored the work and is owner of the relevant patents listed above; tion. S. Herterich: Data curation, investigation. S. Wunder: Investigation. P. Cyprys:
and reports employment with Tubulis GmbH. I. Mai reports grants from Federal Investigation. M. Gerlach: Methodology. P. Ochtrop: Conceptualization.
Ministry for Economic Affairs and Energy (EXIST Forschungstransfer II), Federal
C.P.R. Hackenberger: Conceptualization, methodology, writing–review and edit-
Ministry for Economic Affairs and Energy (German Accelerator), Federal Ministry of
ing. D. Schumacher: Supervision, funding acquisition, methodology, project admin-
Education and Research (Rahmenprogramm Gesundheitsforschung), and LMU
istration. J. Helma: Conceptualization, supervision, funding acquisition, methodol-
Munich outside the submitted work; as well as reports a patent for WO2023083919
ogy. A.M. Vogl: Conceptualization, supervision, methodology, writing–review and
pending; and Tubulis GmbH sponsored the work and is owner of the relevant patents editing. M.-A. Kasper: Conceptualization, data curation, supervision, investigation,
listed above; and reports employment with Tubulis GmbH. S. Herterich reports grants visualization, methodology, writing-original draft.
from Federal Ministry for Economic Affairs and Energy (EXIST Forschungstransfer
II), Federal Ministry for Economic Affairs and Energy (German Accelerator), Federal
Ministry of Education and Research (Rahmenprogramm Gesundheitsforschung),
Acknowledgments
and LMU Munich outside the submitted work; and Tubulis GmbH sponsored the The authors thank Danila Hauswald for excellent technical assistance in antibody
work and is owner of the relevant patents listed above; as well as reports employment purification. This work was supported by grants from the German Federal Ministry
with Tubulis GmbH. S. Wunder reports grants from Federal Ministry for Economic for Economic Affairs and Energy and the European Social Fund with grants (to
Affairs and Energy (EXIST Forschungstransfer II), Federal Ministry for Economic D. Schumacher and J. Helma; EXIST FT I); and the Bavarian Ministry of Economic

AACRJournals.org Mol Cancer Ther; 2023 OF11


Schmitt et al.

Affairs, Regional Development and Energy with grants (to D. Schumacher, J. Helma, Note
and C.P.R. Hackenberger; m4-Award). Supplementary data for this article are available at Molecular Cancer Therapeutics
Online (http://mct.aacrjournals.org/).
The publication costs of this article were defrayed in part by the payment of
publication fees. Therefore, and solely to indicate this fact, this article is hereby Received June 10, 2023; revised August 30, 2023; accepted October 10, 2023;
marked “advertisement” in accordance with 18 USC section 1734. published first October 12, 2023.

References
1. Nguyen TD, Bordeau BM, Balthasar JP. Mechanisms of ADC toxicity and 21. Nakada T, Masuda T, Naito H, Yoshida M, Ashida S, Morita K, et al. Novel
strategies to increase ADC tolerability. Cancers 2023;15:713. antibody–drug conjugates containing exatecan derivative-based cytotoxic pay-
2. Kostova V, Desos P, Starck JB, Kotschy A. The chemistry behind ADCs. loads. Bioorg Med Chem Lett 2016;26:1542–5.
Pharmaceuticals 2021;14:442. 22. Ogitani Y, Aida T, Hagihara K, Yamaguchi J, Ishii C, Harada N, et al. DS-8201a, a
3. Marei HE, Cenciarelli C, Hasan A. Potential of antibody–drug conjugates novel HER2-targeting ADC with a novel DNA topoisomerase I inhibitor,
(ADCs) for cancer therapy. Cancer Cell Int 2022;22:255. demonstrates a promising antitumor efficacy with differentiation from T-DM1.
4. Conilh L, Sadilkova L, Viricel W, Dumontet C. Payload diversification: a key Clin Cancer Res 2016;22:5097–108.
step in the development of antibody–drug conjugates. J Hematol Oncol 2023; 23. Ogitani Y, Hagihara K, Oitate M, Naito H, Agatsuma T. Bystander killing effect of
16:3. DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody–
5. Okajima D, Yasuda S, Maejima T, Karibe T, Sakurai K, Aida T, et al. Datopo- drug conjugate, in tumors with human epidermal growth factor receptor 2
tamab deruxtecan, a novel TROP2-directed antibody–drug conjugate, demon- heterogeneity. Cancer Sci 2016;107:1039–46.
strates potent antitumor activity by efficient drug delivery to tumor cells. 24. Li W, Veale KH, Qiu Q, Sinkevicius KW, Maloney EK, Costoplus JA, et al.
Mol Cancer Ther 2021;20:2329–40. Synthesis and evaluation of camptothecin antibody–drug conjugates. ACS Med
6. Kinneer K, Wortmann P, Cooper ZA, Dickinson NJ, Masterson L, Chem Lett 2019;10:1386–92.
Cailleau T, et al. Design and preclinical evaluation of a Novel B7- 25. Conilh L, Fournet G, Fourmaux E, Murcia A, Matera E-L, Joseph B, et al.
H4–directed antibody–drug conjugate, AZD8205, alone and in combina- Exatecan antibody–drug conjugates based on a hydrophilic polysarcosine drug-
tion with the PARP1-selective inhibitor AZD5305. Clin Cancer Res 2023; linker platform. Pharmaceuticals 2021;14:247.
29:1086–101. 26. Stieger CE, Park Y, de Geus MAR, Kim D, Huhn C, Slenczka JS, et al. DFT-guided
7. Lawn S, Rojas AH, Colombo R, Siddappa D, Wong J, Wu K, et al. Abstract 2641: discovery of ethynyl-triazolyl-phosphinates as modular electrophiles for che-
ZW191, a novel FRa-targeting antibody–drug conjugate bearing a topoisom- moselective cysteine bioconjugation and profiling. Angew Chem Int Ed 2022;61:
erase 1 inhibitor payload. Cancer Res 2023;83:2641-. e202205348.
8. Ulukan H, Swaan PW. Camptothecins. Drugs 2002;62:2039–57. 27. Stieger CE, Franz L, K€orlin F, Hackenberger CPR. Diethynyl phosphinates for
9. Rugo HS, Bardia A, Marme F, Cortes J, Schmid P, Loirat D, et al. Primary results cysteine-selective protein labeling and disulfide rebridging. Angew Chem Int Ed
from TROPiCS-02: a randomized phase 3 study of sacituzumab govitecan (SG) 2021;60:15359–64.
versus treatment of physician’s choice (TPC) in patients (Pts) with hormone 28. Baumann AL, Schwagerus S, Broi K, Kemnitz-Hassanin K, Stieger CE, Trieloff N,
receptor–positive/HER2-negative (HRþ/HER2) advanced breast cancer. J Clin et al. Chemically induced vinylphosphonothiolate electrophiles for thiol–thiol
Oncol 2022;40:LBA1001–LBA. bioconjugations. J Am Chem Soc 2020;142:9544–52.
10. Cortes J, Kim S-B, Chung W-P, Im S-A, Park YH, Hegg R, et al. Trastuzumab 29. Kasper M-A, Glanz M, Oder A, Schmieder P, von Kries JP, Hackenberger CPR.
deruxtecan versus trastuzumab emtansine for breast cancer. N Engl J Med 2022; Vinylphosphonites for staudinger-induced chemoselective peptide cyclization
386:1143–54. and functionalization. Chem Sci 2019;10:6322–9.
11. Corti C, Giugliano F, Nicolo E, Tarantino P, Criscitiello C, Curigliano G. HER2- 30. Kasper M-A, Glanz M, Stengl A, Penkert M, Klenk S, Sauer T, et al. Cysteine-
low breast cancer: a new subtype? Curr Treat Options Oncol 2023;24:468–78. selective phosphonamidate electrophiles for modular protein bioconjugations.
12. Zhu Y, Liu K, Wang K, Zhu H. Treatment-related adverse events of antibody– Angew Chem Int Ed Engl 2019;58:11625–30.
drug conjugates in clinical trials: a systematic review and meta-analysis. Cancer 31. Ochtrop P, Jahzerah J, Machui P, Mai I, Schumacher D, Helma J, et al. Compact
2023;129:283–95. hydrophilic electrophiles enable highly efficacious high DAR ADCs with excel-
13. Beck A, Goetsch L, Dumontet C, Corva€a N. Strategies and challenges for the lent in vivo PK profile. Chem Sci 2023;14:2259–66.
next-generation of antibody–drug conjugates. Nat Rev Drug Discovery 2017;16: 32. Kasper M-A, Stengl A, Ochtrop P, Gerlach M, Stoschek T, Schumacher D,
315–37. et al. Ethynylphosphonamidates for the rapid and cysteine-selective gener-
14. Su Z, Xiao D, Xie F, Liu L, Wang Y, Fan S, et al. Antibody–drug conjugates: recent ation of efficacious antibody–drug conjugates. Angew Chem Int Ed Engl
advances in linker chemistry. Acta Pharmaceutica Sinica B 2021;11:3889–907. 2019;58:11631–6.
15. Sheyi R, de la Torre BG, Albericio F. Linkers: an assurance for controlled delivery 33. Dubowchik GM, Firestone RA, Padilla L, Willner D, Hofstead SJ, Mosure
of antibody–drug conjugate. Pharmaceutics 2022;14:396. K, et al. Cathepsin B-labile dipeptide linkers for lysosomal release of
16. Srinivasarao M, Low PS. Ligand-targeted drug delivery. Chem Rev 2017;117: doxorubicin from internalizing immunoconjugates: model studies of enzy-
12133–64. matic drug release and antigen-specific in vitro anticancer activity.
17. Su D, Zhang D. Linker design impacts antibody–drug conjugate pharmacoki- Bioconjugate Chem 2002;13:855–69.
netics and efficacy via modulating the stability and payload release efficiency. 34. Bauzon M, Drake PM, Barfield RM, Cornali BM, Rupniewski I, Rabuka D.
Front Pharmacol 2021;12:687926. Maytansine-bearing antibody–drug conjugates induce in vitro hallmarks of
18. Lyon RP, Bovee TD, Doronina SO, Burke PJ, Hunter JH, Neff-LaFord HD, immunogenic cell death selectively in antigen-positive target cells. Oncoimmu-
et al. Reducing hydrophobicity of homogeneous antibody–drug conjugates nology 2019;8:e1565859.
improves pharmacokinetics and therapeutic index. Nat Biotechnol 2015;33: 35. Muller P, Martin K, Theurich S, Schreiner J, Savic S, Terszowski G, et al.
733–5. Microtubule-depolymerizing agents used in antibody–drug conjugates induce
19. Burke PJ, Hamilton JZ, Jeffrey SC, Hunter JH, Doronina SO, Okeley NM, et al. antitumor immunity by stimulation of dendritic cells. Cancer Immunol Res
Optimization of a PEGylated glucuronide-monomethylauristatin E Linker for 2014;2:741–55.
antibody–drug conjugates. Mol Cancer Ther 2017;16:116–23. 36. Rios-Doria J, Harper J, Rothstein R, Wetzel L, Chesebrough J, Marrero A,
20. Burke PJ, Senter PD, Meyer DW, Miyamoto JB, Anderson M, Toki BE, et al. et al. Antibody–drug conjugates bearing pyrrolobenzodiazepine or tubulysin
Design, synthesis, and biological evaluation of antibodydrug conjugates payloads are immunomodulatory and synergize with multiple immunother-
comprised of potent camptothecin analogues. Bioconjugate Chem 2009;20: apiessynergy of ADCs with cancer immunotherapies. Cancer Res 2017;77:
1242–50. 2686–98.

OF12 Mol Cancer Ther; 2023 MOLECULAR CANCER THERAPEUTICS


Efficacious Exatecan-based DAR8 Antibody–Drug Conjugates

37. Staudacher AH, Brown MP. Antibody–drug conjugates and bystander 46. Cho HJ, Kim SW, Kim S, Lee S, Lee J, Cho Y, et al. Suppressing p–p stacking
killing: is antigen-dependent internalisation required? Br J Cancer 2017; interactions for enhanced solid-state emission of flat aromatic molecules via
117:1736–42. edge functionalization with picket-fence-type groups. J Mater Chem C 2020;
38. Wang Y, Fan S, Zhong W, Zhou X, Li S. Development and properties of valine– 8:17289–96.
alanine-based antibody–drug conjugates with monomethyl auristatin E as the 47. Pommier Y. Topoisomerase I inhibitors: camptothecins and beyond. Nat Rev
potent payload. Int J Mol Sci 2017;18:1860. Cancer 2006;6:789–802.
39. Indini A, Rijavec E, Grossi F. Trastuzumab deruxtecan: changing the destiny of 48. Cliby WA, Lewis KA, Lilly KK, Kaufmann SH. S phase and G2 arrests induced by
HER2 expressing solid tumors. Int J Mol Sci 2021;22:4774. topoisomerase I poisons are dependent on ATR kinase function. J Biol Chem
40. Saito F, Noda H, Bode JW. Critical evaluation and rate constants of chemose- 2002;277:1599–606.
lective ligation reactions for stoichiometric conjugations in water. ACS Chem 49. Tolis C, Peters GJ, Ferreira CG, Pinedo HM, Giaccone G. Cell-cycle disturbances
Biol 2015;10:1026–33. and apoptosis induced by topotecan and gemcitabine on human lung cancer cell
41. Hamblett KJ, Senter PD, Chace DF, Sun MMC, Lenox J, Cerveny CG, et al. Effects lines. Eur J Cancer 1999;35:796–807.
of drug loading on the antitumor activity of a monoclonal antibody–drug 50. Fucikova J, Kepp O, Kasikova L, Petroni G, Yamazaki T, Liu P, et al. Detection of
conjugate. Clin Cancer Res 2004;10:7063–70. immunogenic cell death and its relevance for cancer therapy. Cell Death Dis
42. Khongorzul P, Ling CJ, Khan FU, Ihsan AU, Zhang J. Antibody–drug conjugates: 2020;11:1013.
a comprehensive review. Mol Cancer Res 2020;18:3–19. 51. Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, et al.
43. Kasper M-A, Lassak L, Vogl AM, Mai I, Helma J, Schumacher D, et al. Bis- Consensus guidelines for the detection of immunogenic cell death. Oncoimmu-
ethynylphosphonamidates as an modular conjugation platform to generate nology 2014;3:e955691.
multi-functional protein- and antibody–drug conjugates. Eur J Org Chem 52. Smyth MJ, Ngiow SF, Ribas A, Teng MW. Combination cancer immu-
2022;2022:e202101389. notherapies tailored to the tumour microenvironment. Nat Rev Clin Oncol
44. Kasper M-A, Gerlach M, Schneider AFL, Groneberg C, Ochtrop P, Boldt S, 2016;13:143–58.
et al. N-hydroxysuccinimide-modified ethynylphosphonamidates enable the 53. Zhou J, Wang G, Chen Y, Wang H, Hua Y, Cai Z. Immunogenic cell death in
synthesis of configurationally defined protein conjugates. ChemBioChem cancer therapy: present and emerging inducers. J Cell Mol Med 2019;23:
2020;21:113–9. 4854–65.
45. Nadkarni DV, Lee J, Jiang Q, Patel V, Sriskanda V, Dutta K, et al. Impact of drug 54. Drago JZ, Modi S, Chandarlapaty S. Unlocking the potential of anti-
conjugation and loading on target antigen binding and cytotoxicity in cysteine body–drug conjugates for cancer therapy. Nat Rev Clin Oncol 2021;18:
antibody–drug conjugates. Mol Pharmaceutics 2021;18:889–97. 327–44.

AACRJournals.org Mol Cancer Ther; 2023 OF13

You might also like