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
Ado-trastuzumab Emtansine (T-DM1): An Antibody–Drug Conjugate (ADC) for HER2-Positive Breast Cancer
My Activity
  • Free to Read
Drug Annotation

Ado-trastuzumab Emtansine (T-DM1): An Antibody–Drug Conjugate (ADC) for HER2-Positive Breast Cancer
Click to copy article linkArticle link copied!

View Author Information
ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
*J.M.L.: e-mail, [email protected]; phone, 781-895-0600.
*R.V.J.C.: e-mail, [email protected]; phone, 781-895-0600.
Open PDF

Journal of Medicinal Chemistry

Cite this: J. Med. Chem. 2014, 57, 16, 6949–6964
Click to copy citationCitation copied!
https://doi.org/10.1021/jm500766w
Published June 26, 2014

Copyright © 2014 American Chemical Society. This publication is available under these Terms of Use.

Abstract

Click to copy section linkSection link copied!

Ado-trastuzumab emtansine (T-DM1) is an antibody–drug conjugate that combines the antitumor properties of the humanized anti-human epidermal growth factor receptor 2 (HER2) antibody, trastuzumab, with the maytansinoid, DM1, a potent microtubule-disrupting agent, joined by a stable linker. Upon binding to HER2, the conjugate is internalized via receptor-mediated endocytosis, and an active derivative of DM1 is subsequently released by proteolytic degradation of the antibody moiety within the lysosome. Initial clinical evaluation led to a phase III trial in advanced HER2-positive breast cancer patients who had relapsed after prior treatment with trastuzumab and a taxane, which showed that T-DM1 significantly prolonged progression-free and overall survival with less toxicity than lapatinib plus capecitabine. In 2013, T-DM1 received FDA approval for the treatment of patients with HER2-positive metastatic breast cancer who had previously received trastuzumab and a taxane, separately or in combination, the first ADC to receive full approval based on a randomized study.

This publication is licensed for personal use by The American Chemical Society.

Copyright © 2014 American Chemical Society

Introduction

Click to copy section linkSection link copied!

The HER2/neu proto-oncogene (also called c-erbB-2), shares extensive sequence homology with the epidermal growth factor receptor gene and other members of the epidermal growth factor receptor family. The gene was first identified in 1981 and was subsequently found to encode a 185 kDa transmembrane glycoprotein with tyrosine kinase activity. Amplification of the gene occurs in about 20–25% of human breast cancers and is associated with aggressive tumor growth and poor clinical outcome. (1) A panel of murine monoclonal antibodies against the extracellular domain of HER2 was derived from immunization of mice with a cell line that had been transfected with the erbB-2 gene. From more than 100 antibodies that were obtained, 4D5 was chosen as the lead. This antibody was shown to specifically bind to HER2 and induced cytostatic growth inhibition of a number of HER2 overexpressing cell lines in vitro. This antibody also inhibited growth of HER2-expressing human breast tumor xenografts in athymic mice. In addition, treatment of tumor-bearing mice with a combination of 4D5 and the chemotherapeutic drug cis-platin resulted in synergistic anti-tumor activity. (2) Since the 4D5 antibody was of murine origin and had the potential to be immunogenic in humans, a series of humanized versions were produced by grafting the complementarity determining regions, involved in antigen binding, from the murine antibody into the human antibody framework. A humanized monoclonal antibody of the IgG1 isotype, trastuzumab, was selected as the lead based on its high binding affinity to the extracellular domain of HER2 and on the retention in the humanized antibody of the ability to inhibit growth of HER2-overexpressing cell lines and xenografts.
There are multiple mechanisms through which trastuzumab is believed to effect cell kill. These include inhibition of constitutive HER2 signaling and disruption of HER2/HER3 interactions in HER2-overexpressing cells resulting in inhibition of cellular proliferation, together with activation of immune effector systems via binding of tumor cell-bound trastuzumab to the FcγRIII receptor on immune effector cells, leading to antibody-dependent cell mediated cytotoxicity (ADCC). (3, 4) Trastuzumab was also shown to enhance the cytotoxic effect of paclitaxel, in a dose-dependent manner, toward HER2 overexpressing breast cancer cells in vitro and also increase the antitumor activity of paclitaxel in breast carcinoma xenografts in vivo. On the basis of these preclinical data, trastuzumab, was advanced into clinical evaluation and ultimately received marketing approval by the U.S. Food and Drug Administration (FDA) in 1998 for use as a single agent for the treatment of patients with metastatic breast cancer (MBC) whose tumors overexpress the HER2 protein and who had received one or more prior chemotherapy regimens. When added to chemotherapy, trastuzumab (Herceptin) was shown to improve time to disease progression and overall survival in patients with HER2-positive MBC compared to chemotherapy alone. (5) Thus, trastuzumab was also approved in combination with paclitaxel in a first-line setting for the treatment of patients with MBC whose tumors overexpress the HER2 protein and who had not received prior chemotherapy.
Although trastuzumab has had a major impact in the treatment of patients with HER2-positive MBC, a subset of patients do not respond to treatment, and most patients who are initially responsive to treatment ultimately experience disease progression. (6) The finding that tumors in these patients continued to express HER2 set off a wave of further research to develop additional approaches to target HER2. These included the development of small molecule kinase inhibitors of HER2, such as lapatinib, and antibodies such as pertuzumab that bind to different sites on HER2 than trastuzumab. Pertuzumab acts by binding to the extracellular dimerization domain II of HER2, thus inhibiting the binding of HER2 to other HER family members, especially HER3, preventing HER2–HER3 dimerization which is a robust activator of the PI3 kinase signaling pathway. (7) Despite these noteworthy advances in HER2-targeted therapy, breast cancer in these patients will eventually progress, underscoring the need for alternative therapies. These new HER2-targeted agents are generally given together with chemotherapeutic agents, thus retaining the toxic effects of chemotherapy in the therapeutic regimen.
The concept of antibody–drug conjugates (ADCs) evolved as a means either to improve the tumor selectivity of cytotoxic drugs or to confer higher potency to monoclonal antibodies that display preferential binding to tumor cells but lack sufficient cytotoxicity. The FDA approvals of two ADCs, brentuximab vedotin and ado-trastuzumab emtansine, have provided proof of concept to this approach and have generated tremendous excitement. There are over 30 ADCs currently in clinical evaluation, and almost every major pharmaceutical company has embraced this technology. (8) The majority of ADCs in clinical evaluation utilize the highly potent tubulin-interacting agents, maytansinoids or auristatins. A few ADCs in the clinic have incorporated other potent effector molecules, such as the topoisomerase 1 inhibitor SN-38, and the DNA interacting agents, calicheamicin and pyrrolobenzodiazepines.
Targeted delivery of a cytotoxic agent to HER2-positive tumors in the form of an ADC, using trastuzumab as the targeting antibody, affords the potential of improved therapeutic activity with lower systemic toxicity. This annotation will review the rationale and preclinical data that drove the linker design and cytotoxic agent selection leading to the development of ado-trastuzumab emtansine (T-DM1). (9) Clinical data that led to the approval of T-DM1 by the U.S. FDA, for the treatment of patients with HER2-positive metastatic breast who have received prior treatment with trastuzumab and a taxane, will be briefly discussed.

Design of T-DM1

Click to copy section linkSection link copied!

(a) Selection of the Cytotoxic Agent

Since taxanes were already used with trastuzumab in the clinic, an obvious ADC design would have consisted of linking taxanes directly to trastuzumab. However, at the time of the selection of the appropriate cytotoxic agent for use in an ADC with trastuzumab, clinical results with ADCs prepared utilizing conventional anticancer drugs such as methotrexate, doxorubicin, and vinblastine had failed to demonstrate therapeutic benefit. A careful retrospective analysis of the preclinical data that were used to support clinical development of these early ADCs revealed several shortcomings. (A) Poor in vitro potency is one shortcoming. Conjugation often led to decreased potency compared to the parent free drug. For example, an ADC with des-acetylvinblastine was reported to be 8-fold less potent than the unconjugated desacetylvinblastine drug. Similarly, a doxorubicin conjugate with the BR96 antibody was about 8-fold less potent than doxorubicin. Unlike the unconjugated dug that can freely diffuse into cells, delivery of the cytotoxic molecule by an antibody is limited by the relatively moderate number of antigen molecules on the cell surface to which the antibody can bind (typically ∼105 receptors/cell). In addition, internalization of cell-surface bound antigen–antibody complex, or intracellular processing to release the active drug moiety, may be inefficient. (B) Modest in vivo activity is another shortcoming. Although these early ADCs were shown to be more active than the corresponding unconjugated drugs in tumor xenograft models, long-term tumor regressions in such xenograft models were only achieved when large doses of ADC were used. For example, the doxorubicin conjugate BR96-Dox was used at a dose of 1 g/kg in vivo, while the clinically achievable dose was only ∼19 mg/kg. (C) Localization in human tumors is also a shortcoming. Dosimetry studies with radiolabeled antibodies in cancer patients have revealed that uptake by the tumor was quite low, ranging from 0.003% to 0.01% injected dose/g tumor. (10) On the basis of these findings, a cytotoxic compound with considerably higher potency (IC50 ≈ 10–11 M) than the drugs used in the early ADCs was set as a key requirement. In addition to high potency, an important requirement that is often overlooked is that cytotoxic molecules for use in ADCs have to be stable and adequately soluble in the aqueous milieu of the antibody. (8, 11)
Maytansine (1), a benzoansamacrolide natural product originally derived from the bark of the African shrub Maytenus ovatus, fits these requirements. (12) Maytansine was found to be a potent inhibitor of tubulin polymerization. Although maytansine bound to the same site on tubulin as the Vinca alkaloids, with similar in vitro inhibition constants, it was considerably more potent as a cell-killing agent. In a direct comparison of in vitro cytotoxicity toward the Burkitt’s lymphoma cell line Namalwa, maytansine was several orders of magnitude more potent than clinically used anticancer drugs such as vinblastine, methotrexate, mitomycin C, and daunorubicin (Figure 1). Further testing of the cytotoxicity of maytansine on a panel of solid tumor cell lines typically showed IC50 values between 30 and 100 pM, with the two tested breast cancer cell lines being among the most sensitive (IC50 values of 30 and 44 pM for SK-Br-3 and MCF-7, respectively). (13) Since breast cancer in humans is known to be sensitive to tubulin agents, with paclitaxel and docetaxel often used in first-line treatment regimens, and given that maytansine is a tubulin agent with high potency toward breast tumor cell lines, it was deemed to be ideally suited for use in an ADC of trastuzumab. Maytansine had been extensively evaluated in phase I and II clinical trials in humans and was discontinued because of an insufficient therapeutic index. (14) However, the wealth of safety data from these trials provided some level of comfort in the selection of maytansine as an effector molecule for ADCs. In addition, maytansine had excellent stability and acceptable solubility in aqueous solutions for use in ADCs.

Figure 1

Figure 1. Comparison of the in vitro potency of cytotoxic drugs toward the Burkitt lymphoma cell line Namalwa: maytansine (blue circle), vinblastine (red triangle), daunorubicin (green diamond), methotrexate (brown square), mitomycin C (purple triangle).

(b) Design and Synthesis of Maytansinoids for Linkage to Antibodies

Although maytansine met all the biochemical and biological criteria for use as an effector molecule in ADCs, it lacked a suitable functional group that could facilitate conjugation to an antibody. Introduction of a thiol functionality into maytansine would provide the opportunity to exploit the two most efficient coupling chemistries available at, or near, neutral pH in the dilute aqueous solution necessitated by the antibody, namely, (a) thiol–disulfide exchange and (b) thioether formation with a reactive Michael acceptor, such as a maleimide. In order to introduce a thiol substituent, without affecting potency, a careful analysis of the SAR studies of maytansine was conducted to determine the best site for such incorporation. The aminoacyl side chain at C3 appeared to be ideal, as it was reported to be quite tolerant to different acyl chain lengths and was synthetically amenable to alterations. (15) Thus, synthetic methods were developed for the incorporation of new ester side chains bearing a terminal thiol group to enable linkage to antibodies (see Scheme 1). Aminoacyl side chains at the C3 position of the maytansinoid molecule are typically installed by first hydrolyzing the C3 ester of maytansine to provide maytansinol and then re-esterification with different side chains to produce maytansinoids of interest. The first challenge was to find a source of maytansine, a plant product in short supply. Fortunately, the ansamitocins, which are a mixture of C3 esters (∼80% isobutanoyl and smaller amounts of n-propanoyl, isopropanoyl, and n-butanoyl) of maytansinol, could be obtained readily by fermentation of the microorganism Actinosynnema pretiosum. Since the maytansinoid molecule was known to be susceptible to β-elimination of the ester under mild alkaline conditions, controlled reduction with LiAl(OMe)3H at −40 °C was employed to convert ansamitocins into maytansinol. Esterification with a carboxylic acid that contained a disulfide in the presence of a coupling agent N-ethyl-N′-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC) and a Lewis acid (zinc chloride) provided maytansinoid disulfides. A panel of disulfide-containing maytansinoids were prepared to examine the effect of length of the acyl side chain on potency. (16) In addition, one or two methyl groups were introduced on the carbon atom geminal to the disulfide to evaluate the effect of steric hindrance on potency. (13) The in vitro cytotoxicity data of a representative set of these new maytansinoids on human cancer cell lines are shown in Table 1. All these maytansine analogues had similar or higher potency than maytansine, indicating that the disulfide substituent did not adversely affect potency. Reduction of the disulfide bond with dithiothreitol gave the desired thiol-bearing maytansinoids for linkage to antibodies.

Scheme 1

Scheme 1. Synthesis of Thiol-Containing Maytansinoidsa

Scheme aReaction conditions: (a) LiAlH(OMe)3/THF, −40 °C, (b) EDC/ZnCl2/CH2Cl2, rt, (c) dithiothreitol, rt.

Table 1. In Vitro Potency of Maytansinoids toward KB (Human Epidermal Carcinoma) and SK-Br-3 (Human Breast Tumor) Cell Lines, Using a Clonogenic Assay and a 72 h Exposure to the Drugs
 IC50, nM
maytansinoidKB cellsSK-Br-3 cells
1 (maytansine)0.0340.030
4a (DM0-SMe)0.1900.180
4b (DM1-SMe)0.0290.014
4c (DM2′-SMe)0.0090.038
4d (DM3-SMe)0.0110.004
4e (DM4-SMe)0.0010.003

(c) Selection of the Linker

The availability of a reactive moiety in the form a sulfhydryl group on the maytansinoid molecule afforded the opportunity to link it to trastuzumab via a disulfide linker or a thioether linker. The disulfide bond strength of the conjugate could be modulated by using a maytansinoid with a sterically hindered thiol and reacting it with an antibody that had been modified with a linker bearing a sterically hindered disulfide bond. (17) The bifunctional cross-linking agent, succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC) was used to introduce maleimido group on the antibody to enable linkage of the maytansinoid via a nonreducible thioether bond. Linker structures used in these conjugations are shown (Figure 2).

Figure 2

Figure 2. Structures of cross-linkers used in conjugate preparation.

(d) Conjugation Methods

Lysine residues on the antibody were selected as the conjugation sites, since a portion of their surface-accessible amino groups can be modified without disturbing the structural integrity and native function of the antibody, along with preserving its favorable pharmacokinetic properties. Briefly, treatment of the antibody with the disulfide-containing linkers resulted in aminolysis of the N-hydroxysuccinimide esters of the linker by lysine residues on the antibody, leading to amide formation and the incorporation of reactive disulfide groups (Scheme 2a). Disulfide exchange between the modified antibody and the thiol-containing maytansinoids resulted in the incorporation of an average of 3.5 maytansinoid molecules linked per antibody molecule. In order to test the role of linker, a trastuzumab conjugate bearing a nonreducible thioether link was prepared by reaction of the antibody with SMCC to introduce maleimido groups. Reaction with the thiol-containing maytansinoid DM1 proceeded smoothly to give a trastuzumab conjugate with an average of 3.5 DM1 molecules linked via thioether bonds (Scheme 2b). Mass spectrometric analysis of T-DM1 shows molecular masses corresponding to a distribution of different number of maytansinoid molecules linked per antibody. The maytansinoid to antibody ratio for T-DM1 was selected based on the desire to (a) minimize the amount of unconjugated antibody, which would diminish the cytotoxic potency of the conjugate, and (b) avoid species with higher maytansinoid load, which might pose manufacturing challenges because of lower solubility. Thus, an average maytansinoid load of ∼3.5 was found to be optimal, as the conjugate maintained good biochemical characteristics with a minimal amount of unconjugated antibody. (18, 19)

Scheme 2

Scheme 2. Representative Conjugation Processes for Trastuzumab-Maytansinoid Conjugates

Preclinical Evaluation Leading to the Selection of Maytansinoid and Linker Component of T-DM1

Click to copy section linkSection link copied!

(a) In Vitro Studies

The five trastuzumab–maytansinoid conjugates (four with a disulfide linker and one with a thioether linker) shown in Figure 3 were evaluated for their in vitro potency toward the HER2-amplified cell lines BT-474 and SK-BR-3. (20) The nature of the linker did not affect potency, as all five conjugates displayed similar high cytotoxicity, with IC50 values ranging from 0.085 to 0.148 μg/mL toward BT-474 cells and from 0.007 to 0.018 μg/mL (4.7 × 10–11 to 1.2 × 10–10 M) toward SK-BR-3 cells, after 3 days of exposure to the ADCs. In contrast, unconjugated trastuzumab displayed only a modest cytotoxic effect (IC50 > 10 μg/mL), indicating that conversion of trastuzumab into an ADC greatly enhanced its cell killing power. Breast tumor cell lines that had little (MCF-7) or no (MDA-MB-468) HER2 expression were much less susceptible to killing by any of the trastuzumab maytansinoid conjugates, with IC50 values of >10 μg/mL for MCF-7 cells and about 3–5 μg/mL for MDA-MB-468 cells, demonstrating good antigen specificity of the cytotoxic effect, regardless of linker choice.

Figure 3

Figure 3. Structural representation of trastuzumab-maytansinoid conjugates with a disulfide linker (8ad) or a thioether linker (8e). Adapted by permission from the American Association for Cancer Research (Lewis Phillips. G. D.; Li, G.; Dugger, D. L.; Crocker, L. M.; Parsons, K. L.; Mai, E.; Lambert, J. M.; Chari, R. V.; Lutz, R. J.; Wong, W. L.; Jacobson, F. S.; Koeppen, H.; Schwall, R. H.; Kenkare-Mitra, S. R.; Spencer, S. D.; Sliwkowski, M. X.Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 2008, 68, 9280–9290). (20)

(b) In Vivo Pharmacokinetic Studies

Since the in vitro potency assay did not distinguish between the ADCs bearing different linkers, all were submitted for in vivo evaluation. A pharmacokinetic study was conducted in CD1 mice to determine the serum concentration of the ADCs at various time points and thus gauge the relative stabilities of the linkers in vivo. In contrast to the in vitro cytotoxicity data, there were clear differences between the conjugates, with a clear correlation between the rate of reduction of the disulfide bond of the ADC as measured in vitro, and the in vivo half-life and exposure. As expected, the trastuzumab ADC with the least hindered disulfide (T-SPDP-DM1) showed the fastest clearance, with no detectable conjugate after 3 days in circulation. (20) For the remaining ADCs, the clearance rate was also determined by the degree of steric hindrance of the disulfide bond. Thus, the ADC with one methyl group on each side of the disulfide bond (T-SSNPP-DM1) gave a higher serum concentration than the ADC with a methyl group just on one side of the disulfide bond. The clearance rate of the ADC with a high degree of steric hindrance (T-SSNPP-DM4) bearing three methyl groups adjacent to the disulfide bond mirrored that of the conjugate bearing the nonreducible thioether link (T-MCC-DM1). Overall, the stability ranking of the trastuzumab–maytansinoid conjugates determined in vivo matched well with the disulfide cleavage rate of a set of non-trastuzumab–maytansinoid ADCs, upon treatment with the reducing agent dithiothreitol. (17)

(c) In Vivo Antitumor Activity

The in vivo efficacy of the trastuzumab ADCs with different linkers was evaluated in the trastuzumab-resistant MMTV-HER2 Fo5 mammary tumor model, a syngeneic tumor-transplant model where murine tumors cells express the human erbB-2 gene. Although the tumors express high levels of human HER2 (3+ expression by immunohistochemistry), they do not respond to trastuzumab alone, making it a suitable model to assess the value of arming the antibody with a maytansinoid. Four of the five linker-maytansinoid ADC formats evaluated in the pharmacokinetic study were tested (the ADC with the most labile linker (SPDP) was not included in the study). From this in vivo efficacy study, it was concluded that higher linker stability correlated with increased antitumor activity, since the nonreducible conjugate with the SMCC linker displayed a statistically significant improvement in activity over the ADC with the disulfide linker, SPP, at the single tested dose (Figure 04). (20) Subsequently, however, a more thorough comparison, at three different doses, of trastuzumab conjugated to DM1 via an SMCC linker or an SPP linker failed to show a measurable difference in antitumor activity in a BT474-EEI tumor model. (21)

Figure 4

Figure 4. Comparison of the in vivo efficacy of trastuzumab–maytansinoid conjugates with a disulfide linker (8b (green triangle), 8c (purple cross), 8d (red boxed cross)) or a thioether linker (8e (blue square)) at a single iv dose of 10 mg/kg conjugate and a vehicle control (×). Adapted by permission from the American Association for Cancer Research (Lewis Phillips. G. D.; Li, G.; Dugger, D. L.; Crocker, L. M.; Parsons, K. L.; Mai, E.; Lambert, J. M.; Chari, R. V.; Lutz, R. J.; Wong, W. L.; Jacobson, F. S.; Koeppen, H.; Schwall, R. H.; Kenkare-Mitra, S. R.; Spencer, S. D.; Sliwkowski, M. X.Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 2008, 68, 9280–9290). (20)

Metabolism Studies

Click to copy section linkSection link copied!

(a) In Vitro Studies

In order to study the intracellular fate of T-DM1 and the role of linker on the amount and nature of catabolites formed, trastuzumab conjugates with the noncleavable SMCC linker (T-DM1) and a disulfide linker (T-SPP-DM1) were prepared using [3H]DM1, with the radiolabel incorporated at the C20 methoxy group of DM1. The HER2-overexpressing cell line BT474-EEI was exposed to the radiolabeled conjugates, and the cells were harvested at various time points, followed by extraction of protein-free maytansinoid catabolites using acetone. The extracts were subjected to HPLC analysis. Exposure to T-DM1 resulted in the formation of a sole metabolite that was identified as lysine-Nε-MCC-DM1, presumably arising from lysosomal degradation of the antibody component leaving the lysine residue of the antibody attached to DM1 via the linker. The peak size increased significantly from the first time point (3 h) to the last one (24 h). Representative chromatograms at the 6 and 24 h time points are shown (Figure 05A). Treatment of cells with the disulfide-linked conjugate, T-SPP-DM1, gave the expected lysine- Nε-SPP-DM1 catabolite resulting from lysosomal degradation of the antibody. However, in this case, subsequent cleavage of the disulfide bond gave rise to the maytansinoid thiol, DM1, which was isolated as a stabilized N-ethylmaleimide (NEM) adduct. At the 6 h time point, the peak area for lysine-SPP-DM1 is somewhat greater than that of DM1, but at the 24 h time point further reduction has taken place to give DM1 as the major peak (Figure 05B). The rate and extent of processing of T-SPP-DM1 by BT474-EEI cells in vitro were compared to those of T-DM1. The protein-free maytansinoid catabolites produced at various time points after exposure to the conjugates were quantified. Processing of both conjugates was found to be equally efficient and approached ∼70% at 24 h (Figure 05C). (21)

Figure 5

Figure 5. (A) In vitro catabolism by BT474-EEI cells of the trastuzumab-MCC-DM1 conjugate 8e (thioether linker): top panel, 6 h time point; bottom panel, 24 h time point. (B) In vitro catabolism by BT474-EEI cells of the trastuzumab-SPP-DM1 conjugate 8b (disulfide linker): top panel, 6 h time point; bottom panel, 24 h time point. (C) Comparison of the rate and extent of in vitro processing of trastuzumab-SPP-DM1 conjugate 8b (disulfide linker: blue squares) and trastuzumab-MCC-DM1 conjugate 8e (thioether linker, red squares) by BT474-EEI cells.

(b) In Vivo Studies

In order to determine how the results from the in vitro metabolism studies related to the in vivo situation, tumor xenografts were established in mice with the same cell line used for the in vitro studies (BT474-EEI). Mice bearing these tumor xenografts were treated with either radiolabeled T-DM1 or T-SPP-DM1 prepared with [3H]DM1. Tumors were excised at various time points and analyzed for both total radioactivity (antibody-bound plus small molecular weight catabolites) and protein-free maytansinoid catabolites by HPLC analysis. Although the T-SPP-DM1 conjugate displayed a faster plasma clearance than T-DM1, the amount of maytansinoid catabolites in the tumor xenograft at the various time points was quite similar, with maximum accumulation occurring between 2 and 4 days (Figure 6A). It was thus unsurprising that the antitumor activity of the two compounds was also similar in this study. The nature of the catabolites mirrored the pattern observed with the same cell line in vitro, with T-DM1 giving just one catabolite (lysine-Nε-MCC-DM1) and with T-SPP-DM1 yielding the same two catabolites observed in vitro (lysine- Nε-SPP-DM1 and DM1, stabilized as N-ethylmaleimide adduct). The catabolite profiles for the 2-day time-point are shown (Figure 6B), with T-SPP-DM1 giving predominantly DM1 (isolated as an N-ethylmaleimide adduct) after reduction of the SPP linker within tumor cells and T-DM1 showing a single peak of its sole catabolite. (21)

Figure 6

Figure 6. (A) Time dependent formation of catabolite: comparison of in vivo catabolism of trastuzumab–maytansinoid conjugates in BT474-EEI tumor xenografts in mice: trastuzumab-SPP-DM1 conjugate 8b (disulfide linker, blue square) and trastuzumab-MCC-DM1 conjugate 8e (thioether linker, red square). (B) In vivo catabolism of trastuzumab–maytansinoid conjugates in BT474-EEI tumor xenografts in mice at 2-day time-point: top panel, 8b, disulfide linker (SPP); bottom panel, 8e, thioether linker (SMCC).

In Vivo Tolerability Studies

Click to copy section linkSection link copied!

In order to select the final linker format for the maytansinoid conjugated trastuzumab for ADC development, the tolerability of the two conjugates (SMCC linker versus SPP linker) was evaluated in Sprague–Dawley rats. In this single dose acute toxicity study, the ADC with the nonreducible SMCC linker was found to be at least about 2-fold better tolerated than the conjugate with the disulfide (SPP) linker. Thus, treatment at a dose of 22 mg/kg with the SPP conjugate resulted in significant (∼10%) body weight loss, while the SMCC conjugate could be dosed at 50 mg/kg without any weight loss. On the basis of these preliminary acute toxicity results, the SMCC linker afforded at least about a 2-fold greater therapeutic index in mice to a trastuzumab-DM1 conjugate compared with that provided by the disulfide linker, and so SMCC was the linker of choice for T-DM1, the development candidate ADC. (20)

Biological Activity of T-DM1

Click to copy section linkSection link copied!

(a) In Vitro Cytotoxicity

Testing of the in vitro cytotoxicity of T-DM1 (8e) in comparison to unconjugated trastuzumab was expanded to a panel of cell lines with different levels of expression of HER2 and varying sensitivity to trastuzumab. (20) This panel also included HER2-positive cell lines from tumor types other than breast, such as the lung carcinoma cell line Calu-3, the ovarian cancer cell line SK-OV-3, and the gastric carcinoma cell line MKN7. In all cases, T-DM1 was considerably more potent than free trastuzumab (Table 2) toward the cancer cell lines. In contrast, the normal human mammary epithelial cells (HMEC) and normal human epidermal keratinocytes (NHEK) were much less sensitive to trastuzumab and T-DM1. (20) Since the small molecule kinase inhibitor lapatinib is used in the treatment of trastuzumab refractory breast cancer, it was important to test the potency of T-DM1 for HER2-amplified cells that were resistant to lapatinib. As a first test, SK-Br-3 cells, which are highly sensitive to T-DM1 and lapatinib, were exposed to increasing concentrations of lapitinib over a 9-month period. The resulting lapitinib-resistant clone was ∼200-fold less sensitive to lapatinib but remained as sensitive to T-DM1 as the parent clone. Another mechanism of resistance to lapatinib involves activation of the PI3 kinase pathway, either through the acquisition of a PIKCA mutation or loss of PTEN. The MCF7-neo/HER2 cell line cell line, which has an activated PI3 kinase pathway, was found to be resistant to lapatinb (IC50 ≈ 20 μM) and to trastuzumab (IC50 > 66 nM) but was quite sensitive to T-DM1 (IC50 ≈ 0.16 nM). These results demonstrate that T-DM1 is a highly potent agent even against cells that are resistant to the approved therapy options (trastuzumab and lapatinib) for HER2-positive breast cancer. (22, 23)
Table 2. Comparison of the in Vitro Potency of Trastuzumab and Trastuzumab-MCC-DM1 Conjugate toward Human Tumor and Normal Cell Lines after a 3-Day Exposure to the Antibody or Conjugate
  IC50, μg/mL
cell lineorigintrastuzumab-MCC-DM1trastuzumab
SK-Br-3human breast tumor0.011>10
BT-474-EEIhuman breast tumor0.004>10
HCC1954human breast tumor0.015>10
KPL-4human breast tumor0.011>10
Calu-3human lung carcinoma0.062>10
MKN-7human gastric carcinoma0.266>10
SK-OV-3human ovarian cancer0.009>10
HMEChuman mammary epithelial cells4.0>10
NHEKnormal human epidermal keratinocytes10.0>10
Maytansinoids are reported to be substrates for the MDR1/PgP transporter leading to poorer killing of multidrug resistant (MDR) cells. MDR1/PgP is believed to confer resistance to maytansinoids via two mechanisms: (a) mediating efflux of the drug that diffuses into the plasma membrane from the extracellular space, thereby preventing the compound from reaching the cytoplasm and (b) effluxing any drug that does enter the cytoplasm back to the outside of the cell. (24) ADCs, such as T-DM1, deliver the maytansinoids into cells via antigen-mediated endocytosis, thus bypassing the first MDR1 mechanism. Upon intracellular processing, T-DM1 is processed into a charged cytotoxic metabolite, lysine-Nε-MCC-DM1, which is less susceptible to MDR1/PgP-mediated efflux. Only high levels of MDR1/PgP expression appear to confer resistance to DM1 and T-DM1, and the free maytansinoid and ADC were not substrates for BCRP/ABCG2. Breast cancer lines expressing a range of levels of MRP3/ABCC3 were not resistant to DM1 or T-DM1. (25)

(b) In Vivo Evaluation

The in vivo antitumor activity of T-DM1 was more thoroughly evaluated in multiple HER2-positive models in mice. For example, in a breast tumor xenograft model established with KPL-4 cells, a single iv administration of T-DM1 at 15 mg/kg gave complete tumor regression lasting for the duration of the experiment (125 days), while treatment with four weekly doses of trastuzumab at 15 mg/kg only achieved a modest delay in tumor growth (Figure 7A). (20) In another example using the BT474-EEI breast tumor xenograft model, T-DM1 displayed dose-dependent antitumor activity, with tumor growth delay at the lower doses (0.3–3 mg/kg, q3w × 3), tumor regression at the 10 mg/kg dose, and complete regression at the 15 mg/kg dose (Figure 7B). (20) Unconjugated trastuzumab showed little activity in this model. Similar results were obtained in the MMTV-HER2 Fo5 trastuzumab-resistant transgenic transplant model. Treatment with a control, nonbinding conjugate gave no tumor growth inhibition, demonstrating the antigen specificity of the activity of T-DM1. (20, 26)

Figure 7

Figure 7. (A) Comparison of the in vivo antitumor activity of trastuzumab (red circle, 15 mg/kg × 4) and T-DM1 (blue square, 15 mg/kg, single dose), vehicle control (×), in the KPL-4 model. (Excerpted from Lewis-Phillips et al. (20)). (B) Dose-dependent antitumor activity of T-DM1 in the BT474-EEI model (dosing q3w × 3). Doses of T-DM1, are the following: 0.3 mg/kg (purple-outline triangle), 1 mg/kg (green triangle), 3 mg/kg (blue-outline triangle), 10 mg/kg (red triangle), 15 mg/kg (blue square), vehicle control (×); unconjugated trastuzumab (red circle, 15 mg/kg). Parts A and B are adapted by permission from the American Association for Cancer Research (Lewis Phillips. G. D.; Li, G.; Dugger, D. L.; Crocker, L. M.; Parsons, K. L.; Mai, E.; Lambert, J. M.; Chari, R. V.; Lutz, R. J.; Wong, W. L.; Jacobson, F. S.; Koeppen, H.; Schwall, R. H.; Kenkare-Mitra, S. R.; Spencer, S. D.; Sliwkowski, M. X.Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 2008, 68, 9280–9290). (20)

Following up on the in vitro study described above, wherein T-DM1 was found to be active against lapatinib-resistant cell lines, an in vivo study was conducted to compare the activity of T-DM1 and lapatinib. Mice bearing established Fo5 tumors were found to insensitive to lapatinib, and regressions could not be attained even after daily treatment for 3 weeks. Treatment with T-DM1, however, was quite effective, resulting in complete regression in 3/10 animals and partial regression in 6/10 mice. (22)
A pharmacokinetic study was conducted in rats to compare the levels of T-DM1 and total trastuzumab. Serum concentration of total antibody was determine using a specific ELISA for trastuzumab, while intact T-DM1 conjugate was assayed by ELISA using an antibody against DM1. Circulating serum concentrations of T-DM1 measured for 1 week were similar to the measurement of total serum trastuzumab concentrations, demonstrating that there was negligible release of maytansinoid from the conjugate over 7 days in circulation in rats. (20) The anti-DM1 antibody used in the ELISA was found to bind appropriately with antibodies with different maytansinoid load, including one maytansinoid per antibody. Plasma samples spiked with T-DM1 were also analyzed by capillary LC–MS, using anti-DM1 antibody as the capture probe, and the results matched those obtained through ELISA measurements. (27)

(c) Functional Activity

Since trastuzumab is by itself an active antibody with biological function, it was important to show that conjugation to DM1 did not affect its functional properties. In a head to head comparison, T-DM1 was shown to retain the binding affinity of trastuzumab to the HER2 antigen. Thus, in a competition binding assay, T-DM1 competed as well as trastuzumab with the binding of radioiodinated trastuzumab to the antigen (HER2 extracellular domain) coated on a plate. Some of the functional activity of trastuzumab is attributed to its ability to cause ADCC. Both T-DM1 and trastuzumab were shown to mediate ADCC equally well in vitro. In control experiments, a nonbinding antibody–DM1 conjugate and a trastuzumab mutant that been engineered to abolish binding to FcγR on immune cells did not mediate ADCC. The cytotoxic effect of trastuzumab has been attributed, in part, to its ability to inhibit the phosphorylation of the HER family member HER3 resulting in inhibition of the PI3 kinase pathway. T-DM1 and trastuzumab were found to show similar levels of inhibition of AKT phosphorylation, demonstrating that conjugation to DM1 did not affect trastuzumab’s ability to inhibit AKT phosphorylation. (22)

Preclinical Toxicology and Toxicokinetics of T-DM1

Click to copy section linkSection link copied!

Single- and repeat-dose safety toxicology studies were performed with T-DM1 in both rats and cynomolgus monkeys. (28) Trastuzumab recognized HER2 in cynomolgus monkeys but does not bind to the rat antigen. Thus, rats allowed assessment of antigen-independent toxicity of T-DM1, while cynomolgus monkeys allowed evaluation of the potential for any antigen-mediated effects in addition to antigen-independent toxicity. The toxicity of the free maytansinoid, DM1, was assessed in rats only. (28) T-DM1 was well-tolerated at single intravenous doses up to 46 mg/kg (protein dose, equivalent to ∼4400 μg DM1/m2) in rats and 30 mg/kg (∼6000 μg DM1/m2) in monkeys. Rats given 60 mg/kg T-DM1 (∼6800–7800 μg DM1/m2) showed clinical signs of morbidity and/or mortality. DM1 was only tolerated up to 0.2 mg/kg (∼1600 μg DM1/m2) in rats, suggesting that 2–3 times higher doses of maytansinoid are tolerated in rats given T-DM1 versus free DM1. As noted above, the major low molecular weight maytansinoid metabolite of T-DM1 is the lysine adduct, lysine-Nε-MCC-DM1. (21) The hydrophilic nature of the charged amino and carboxyl groups of the lysine greatly reduces the cytotoxicity of the adduct relative to DM1 and other noncharged maytansinoids and likely alters its distribution in vivo relative to noncharged maytansinoids, with a favorable effect on the tolerable dose. (13, 21) Thus, the hydrophilic metabolite lysine-Nε-MCC-DM1 is >200-fold less potent (IC50 ≈ 7.5–17 nM) when applied to cancer cells in vitro, compared to noncharged maytansinoids such as maytansine (IC50 ≈ 0.03–0.09 nM). (29)
The principal acute toxicities in rats were comparable between T-DM1 and DM1 and were associated with dose-dependent effects in bone marrow/hematologic systems, lymphoid organs, and liver. Both agents induced minimal to moderate reductions in lymphocyte counts and reticulocyte counts and minimal to mild reductions in platelets, as well as an increase in absolute neutrophil counts. (28) These clinical laboratory observations were consistent with histologic findings of bone marrow and lymphoid organ toxicities, although such findings were more severe for DM1 than for T-DM1 at their respective maximum tolerated doses (MTD), 1600 and 4400 μg DM1/m2, respectively. Elevations of serum transaminase levels were in the range of 2- to 4-fold at the MTD of both T-DM1 and DM1, consistent with histologic findings of liver toxicity. (28) At the tolerated doses, all findings were partially or completely reversed during the recovery period. Histopathology revealed an increase in mitotic figures in a variety of tissues in rats dosed with T-DM1, notably in adrenals, liver, kidney, eye (corneal epithelium), and skin (epidermis), findings consistent with the expected pharmacologic action of maytansinoids in disrupting microtubule dynamicity resulting in cell cycle arrest. (28) Mitotic figures were found only in liver and kidney in rats dosed with free DM1.
While the toxicity profiles of T-DM1 and DM1 were similar, the differences in severity in some findings (e.g., reduction in reticulocytes was more severe for DM1 versus T-DM1; different distribution of mitotic figures) are likely related to differences of pharmacokinetics, distribution, and metabolism between T-DM1 and DM1. Separate ELISA methods were used to measure T-DM1 and total trastuzumab in serum of rats dosed with T-DM1. The terminal t1/2 of T-DM1 in rats was about 3–5 days, with a slow clearance (CL) of 13–15 mL day–1 kg–1 and a volume of distribution approximating the plasma volume, characteristics of an antibody-based biologic agent. (28) On the other hand, in rats dosed with DM1, pharmacokinetics showed a large volume of distribution (>5000 mL/kg) and rapid CL (20–55 mL min–1 kg–1), as expected for the small molecule “payload”. The clearance of total trastuzumab (all conjugated species and any unconjugated antibody) in rats dosed with T-DM1 was about 2-fold slower than that of T-DM1, a finding that could be accounted for by one (or both) of two mechanisms: a certain rate of deconjugation in vivo to form species with a lower DM1-to-antibody ratio (DAR) or a somewhat faster clearance of species with higher-than-average DAR versus species with lower-than-average DAR. Free serum DM1 concentrations in rats treated with T-DM1 were low, approximately 50-fold lower than that of conjugated DM1 at any time-point. (28) Even this low level of free DM1 may be an overestimate of in vivo plasma concentrations, given that sample preparation utilized treatment with a reducing agent, which may result in ex vivo cleavage of any oxidized derivatives of the thioether. (30)
T-DM1 was well tolerated in cynomolgus monkeys upon repeat dosing at 3-week intervals with 4 doses of up to 30 mg/kg (∼6000 μg DM1/m2) or with 8 doses at 10 mg/kg (∼2000 μg DM1/m2). Most adverse findings were similar to those described for rats, with liver and bone marrow/hematologic systems being the primary target organs for toxicity. A minimal to mild decrease in platelets was noted on day 3 after each dose, as was a reversible 2- to 4-fold increase in serum hepatic transaminase levels. Microscopic findings showed an increase in the number of cells in mitotic arrest in a variety of tissues in addition to liver findings. However, despite the fact that epithelial cells of many tissues express HER2, the incidence of cellular mitotic arrest in the rat studies was noted to be more widespread than in monkey, indicating that antigen-independent mechanisms predominate in the uptake and intracellular catabolism of T-DM1 and that antigen-dependent binding and uptake by normal tissue may not be a major safety concern. (28) That antigen-mediated uptake does occur in cynomolgus monkeys was clearly demonstrated by the nonlinear pharmacokinetics, with the CL in animals treated at 3 mg/kg being 50% faster than those in the higher dose groups. At doses of ≥10 mg/kg, the t1/2 of T-DM1 in cynomolgus monkeys was approximately 3–5 days (as in rat), with CL in the range of 9.4–11.5 mL day–1 kg–1. There was no accumulation of T-DM1 upon repeat dosing at 3-week intervals. As in the rat, free DM1 concentrations were always at least 50-fold lower than conjugated DM1 at any time-point. Also as observed in rats, the CL of the intact T-DM1 conjugate was faster than that of total trastuzumab. In 4 of 36 monkeys (11%) given repeated doses of T-DM1, antiproduct antibodies were detected, although there were no apparent effects on the toxicokinetic profiles in these animals.
Dose-dependent axonal degeneration was observed in the repeat-dose cynomolgus monkey studies, although these histopathologic findings were without any clinical observations of neurologic deficit. (28) These finding were not reversible within the 6-week recovery period. Neuropathy is a common toxicity finding with microtubule-acting agents such as taxanes and the Vinca alkaloids, especially with the longer exposures of repeat-dose schedules of administration. There were no cardiovascular safety signals in the monkey studies. (28) The overall conclusions of the toxicity evaluation of T-DM1 were that the mechanism of toxicity is consistent with the pharmacology of DM1 as a microtubule-acting compound and that the observed toxicities were primarily through antigen-independent mechanisms of T-DM1 catabolism. The starting dose level of T-DM1 for the first-in-human phase I clinical trial was 0.3 mg/kg based on 1/12 of the highest nonseverely toxic dose in cynomolgus monkeys. (28, 31)

Clinical Evaluation of T-DM1

Click to copy section linkSection link copied!

(a) Phase I Studies

The safety, tolerability, and pharmacokinetics of T-DM1 was evaluated in a phase I dose-escalation trial in patients with HER2-positive metastatic breast cancer (HER2-positive MBC) who had previously received a trastuzumab-containing regimen. Twenty-four patients were given T-DM1 at doses ranging from 0.3 to 4.8 mg/kg as an intravenous infusion once every 21 days. (31) At the 4.8 mg/kg dose, two of three subjects had dose-limiting thrombocytopenia (grade 4). A total of 15 patients were treated at the maximum tolerated dose (MTD) of 3.6 mg/kg, where nearly all patients exhibited thrombocytopenia, generally grade 1 or 2 and rapidly reversible, with a nadir by about day 8 and recovery typically by day 15 of each cycle. Other commonly reported adverse events (AEs) included elevated hepatic transaminases (41.5% of patients), fatigue (37.5%), anemia (29.2%), and nausea (25.0%), generally grade 1 or 2, and reversible. There were no reports of greater than grade 1 nausea, vomiting, alopecia, or neuropathy or any cardiac observations that required dose modification.
Pharmacokinetics were nonlinear in the dose range 0.3–4.8 mg/kg, consistent with the anticipated antigen-mediated clearance at these doses. (31, 32) The t1/2 of T-DM1 at the 3.6 mg/kg dose (n = 15) was about 3.5 days. (31) Six of the 24 patients had an objective partial response (five confirmed). All the responding patients had previously received at least one tubulin-acting agent among their prior chemotherapy treatments. In the 15 patients treated at the MTD, the confirmed response rate in patients with measurable disease (n = 9) was 44%, while the clinical benefit rate (CBR), including patients with stable disease of ≥6 months, was 73%, a substantial level of clinical activity in a heavily pretreated patient population.
Once MTD had been established, a second arm of the study was opened to evaluate weekly administration. Twenty-eight patients received weekly T-DM1, from 1.2 to 2.9 mg/kg (5 dose levels). Two of three patients dosed at 2.9 mg/kg experienced dose-limiting toxicities (DLTs) preventing dosing on day 8, one case of grade 3 thrombocytopenia, and one case of grade 3 hepatic transaminase (AST) levels. (33) The MTD for T-DM1 administered weekly was established at 2.4 mg/kg, for a cumulative dose of 7.2 mg/kg over 3 weeks which was double that given as a single dose q3 weeks. The most common drug-related grade ≥3 adverse events seen in >10% of patients were increased AST (11%) and thrombocytopenia (11%). There was only a low incidence of any gastrointestinal toxicity, very few reports of infusion reactions, and no significant cardiac abnormalities in the study. Thirteen patients experienced eye disorders, two of which were grade 3. (33) In general, the tolerability of T-DM1 on the weekly schedule was similar to that of the q3 week schedule, with patients receiving a somewhat longer median duration of treatment (18.9 weeks versus 16.7 weeks). The DLT, reversible thrombocytopenia, was the same for both regimens, together with reversible minimal to moderate increases in hepatic transaminases consistently observed on both schedules. The incidence of adverse events associated with administration of maytansine, severe diarrhea, vomiting, and sensory neuropathy were low and of low grade when reported, consistent with minimal systemic exposure to free maytansinoid and demonstrating the benefit of the concept of an antibody–drug conjugate. (14)
Confirmed partial responses were reported in 46% (13 of 28) of patients enrolled in the weekly dosing arm of the phase I study, and the six-month CBR was 57% (16 of 28 patients). (33) Both the weekly and the q3 week schedules demonstrate that T-DM1 has substantial clinical activity in patients with previously treated HER2-positive MBC. The q3 week schedule was chosen for the subsequent phase II clinical evaluation of T-DM1.

(b) Phase II Clinical Trials of T-DM1 as a Single Agent

A single arm phase II study (TDM4258g) was conducted in 112 patients with HER2-positive MBC utilizing the 3.6 mg/kg q3 week dosing schedule. (34) Patients had received a median of eight prior anticancer agents including trastuzumab (100%), a taxane (84%), an anthracycline (71%), capecitabine (66%), and lapatanib (60%). In this heavily pretreated patient population, 29 patients (25.9%) had objective partial responses by independent assessment. The median progression-free survival (PFS) was 4.6 months, while the medial duration of response was 9.4 months (investigator assessment). The overall response rate (ORR) was higher (33.8%) in patients whose HER2-overexpression status on archival primary tumor specimens was confirmed by a central laboratory (74 confirmed positive of 95 patients reassessed). The ORR in the 21 patients reassessed as HER2-normal was only 4.8%. T-DM1 was well-tolerated in this study, with safety findings similar to those observed in the phase I study. The median dose intensity (dose delivered/expected dose) was 99.7%, and dose modifications or discontinuations due to AEs were infrequent. (34) Twenty-one patients (18.8%) completed at least 1 year on treatment, suggesting that long-term administration was tolerated.
A subsequent confirmatory single arm phase II study was conducted in 110 patients with HER2-positive MBC who had previously received a taxane, an anthracycline, capecitabine, as well as two HER2-targeted agents, trastuzumab and lapatinib, for a median of seven prior anticancer agents for their metastatic disease. (35) The ORR was 34.5% (38 of 110 patients). The median PFS was 6.9 months, and the median duration of response was 7.2 months. With inclusion of patients in stable disease for at least 6 months, the overall CBR was 48.2%. The HER2-positive (HER2-overexpression) status was confirmed upon reassessment by a central laboratory in 84% of patients, and in this subgroup of 80 patients, the ORR was 41% with a median PFS of 7.3 months. There were no new safety signals in this confirmatory phase II study, most adverse events being grade 1 or 2. Reversible thrombocytopenia was observed in 38.2% of patients (only 9.1% of grade ≥3); as with the prior studies, platelet transfusions were infrequent, hemorrhagic adverse events were generally mild, and no patient discontinued T-DM1 due to a bleeding event. (35) This trial also confirmed the observation of generally mild transaminase elevations indicative of hepatic toxicities that appeared in temporal relationship with T-DM1 dosing (AST increases were seen in 26.4% of patients), although a few patients (8.2%) experienced a grade ≥3 adverse event. (35)
Given the encouraging efficacy signals and tolerable safety profile of T-DM1 in these single arm, single agent studies, a phase II randomized study was performed to directly compare T-DM1 as a single agent with an active regimen for first-line treatment of HER2-positive MBC, trastuzumab plus docetaxel. (36) After a median follow-up of about 14 months, there was a significant improvement in PFS in patients treated with T-DM1 (14.2 months) compared with those treated with the standard-of-care (9.2 months). The overall response rates were similar, 64.2% in those patients treated with T-DM1 (n = 67), including seven complete responses (CRs), versus 58% in those patients treated with trastuzumab/docetaxel (n = 70) including three CRs. (36) Regarding safety in the first-line setting, the T-DM1 arm had about half the number of grade ≥3 adverse events than the trastuzumab/docetaxel arm (46.4% versus 90.9%, respectively). Only 7.2% of AEs led to discontinuation of treatment in patients treated with T-DM1, compared with 40.9% for patients treated with standard-of-care. There were no cases of clinically significant cardiac events, a concern due to the known association of trastuzumab with cardiac toxicity, although three patients on each arm had decreased left ventricular ejection fraction. (36, 37) Not only was the incidence of grade ≥3 AEs lower in the T-DM1 arm in comparison to the trastuzumab/docetaxel arm, but their nature was also different. The most common grade ≥3 AEs in the latter arm were neutropenia (62.1%), leukopenia (24.2%), and febrile neutropenia (13.6%), together with a high incidence of alopecia (66.7%), while grade ≥3 AEs in patients treated with T-DM1 were increases in hepatic transaminases (AST 8.7%; ALT 10.1%), thrombocytopenia (7.2%), and neutropenia (5.8%) with only a low incidence of alopecia (4.3%). This randomized study demonstrates the potential for an antibody–drug conjugate to deliver improved clinical benefit and decreased toxicity risk in these patients that should be validated in phase III trials.

(c) Phase III Clinical Trials of T-DM1

The phase I and phase II studies led to a pivotal phase III trial (“EMILIA”) in which 991 patients with advanced HER2-positive MBC whose disease had progressed following treatment with trastuzumab and a taxane were randomized to receive either T-DM1 (single agent at 3.6 mg/kg, q3 weeks) or lapatinib plus capecitabine. (38) Preliminary results were first reported at the 2012 Annual Meeting of the American Society of Clinical Oncology. The median ORR, PFS, and overall survival all significantly favored the T-DM1 arm of the study, as summarized in Table 03. The number of AEs of grade ≥3 was less in the T-DM1 arm (40.8%) than in the lapatinib/capecitabine arm (57.0%). As anticipated from the phase I and phase II experience, the incidence of thrombocytopenia, and of increased serum hepatic transaminase levels, were higher in patients treated with T-DM1, while the incidence of diarrhea, nausea, vomiting, and hand–foot syndrome were higher in patients treated with lapatinib/capecitabine (Table 03). The rates of cardiac adverse events were low, about 1.6–1.7% in both arms.
Table 3. Efficacy and Safety Results Reported for the Phase III Clinical Trial, “EMILIA”, Which Randomized HER2-Positive Breast Cancer Patients Who Had Previously Received Trastuzumab and a Taxane for Treatment of Their Metastatic Disease To Receive Either T-DM1 (single agent) or Lapatinib + Capecitabine (38)
EfficacyT-DM1lapatinib + capecitabine
no. of patients evaluablen = 397n = 389
   
objective response rate43.6%30.8%
complete response1% (n = 4)0.5% (n = 2)
partial response42.6% (n = 169)30.3% (n = 118)
median duration of response12.6 months6.5 months
estimated 1-year survival85.2%78.4%
estimated 2-year survival64.7%51.8%
median progression-free survival9.6 months6.4 months
median overall survival30.9 months25.1 months
SafetyT-DM1lapatinib + capecitabine
no. of subjects evaluablen = 490n = 488
 
adverse events (≥15% of patients)any gradegrade ≥3any gradegrade ≥3
     
any adverse event95.9%40.8%97.7%57.0%
diarrhea23.3%1.6%79.7%20.7%
hand–foot syndrome1.2%058.0%16.4%
vomiting19.0%0.8%29.3%4.5%
fatigue35.1%2.4%27.9%3.5%
nausea39.2%0.8%44.7%2.5%
mucosal inflammation6.7%0.2%19.1%2.3%
thrombocytopenia28.0%12.9%2.5%0.2%
elevated serum ALT16.9%2.9%8.8%1.4%
elevated serum AST22.4%4.3%9.4%0.8%
The “EMILIA” trial established the safety and effectiveness of T-DM1, and it was approved as a new therapy for patients with HER2-positive late stage (metastatic) breast cancer, previously treated with trastuzumab and taxane chemotherapy, by the U.S. FDA on February 22, 2013. T-DM1 (ado-trastuzumab emtansine, Kadcyla) is the first antibody–drug conjugate to receive full approval from FDA on the basis of a randomized study for any indication.
First results of another phase III trial (“TH3RESA”), in which 602 patients with advanced HER2-positive MBC previously treated with at least two HER2-directed therapies were randomized 2:1 to receive either T-DM1 or physician’s choice of treatment, were reported at the European Cancer Congress in September 2013. (39) The median PFS increased from 3.3 months in the reference arm to 6.2 months in patients receiving T-DM1. The ORR was 31.3% in the T-DM1 arm compared with only 8.6% in these very heavily pretreated patients receiving physician’s choice of treatment. Overall survival showed a trend in favor of T-DM1 (although it did not reach statistical significance at the reported interim analysis), and as with “EMILIA”, there were fewer grade ≥3 AEs in the T-DM1 arm.
A phase III trial comparing single agent T-DM1 to trastuzumab plus docetaxel in first-line treatment of HER2-positive MBC is ongoing. This trial, called “MARIANNE”, builds upon the experience of the earlier phase II trial in patients previously untreated for metastatic disease and also includes a comparison of single agent T-DM1 to T-DM1 plus pertuzumab, another HER2-targeting antibody that binds to a distinct site on the HER2 extracellular domain. (36) Pertuzumab is approved for use in combination with trastuzumab and docetaxel to treat patients with HER2-positive breast cancer. (40) Including a T-DM1 plus pertuzumab arm in the MARIANNE trial was based on preclinical studies suggesting increased activity of the combination and an earlier phase Ib trial of the safety and preliminary efficacy of the combination in HER2-positive MBC patients. (41, 42) The results of the “MARIANNE” trial are expected in 2014 and are eagerly awaited.
Brain metastases are common in patients with HER2-positive MBC and are associated with poor prognosis. (43) Up to now, patients with symptomatic brain metastases were excluded from enrollment in clinical trials of T-DM1. (31, 34, 38) However, a preliminary report documents a finding that brain metastases of a HER2-postive breast cancer patient can respond to systemic therapy with single-agent T-DM1, suggesting that the blood–brain barrier is impaired in the vicinity of the tumor and can allow passage of antibodies into the metastatic tumor tissue. (43) Further investigation of the potential of T-DM1 for treating HER2-positive MBC patients with brain metastases is warranted.
Besides development of T-DM1 in MBC, there is an ongoing phase II/III clinical trial to evaluate T-DM1 versus taxane therapy in patients with previously treated, advanced HER2-positive gastric cancer (www.clinicaltrials.gov). Preclinical studies have demonstrated that T-DM1 is active in HER2-positive gastric cancer models. (23)

Clinical Pharmacokinetics and Metabolism of T-DM1

Click to copy section linkSection link copied!

The pharmacokinetic properties of T-DM1 were assessed utilizing two analytical methodologies, those typically used for large proteinaceous molecules (e.g., antibodies), as well as those used for quantifying small molecules. The analytes included total trastuzumab concentrations (unconjugated antibody as well as conjugated antibody irrespective of the number of linked DM1 molecules) and DM1-conjugated trastuzumab concentrations (T-DM1), both measured by specific ELISA methods, as well as free DM1 and DM1 catabolites which were measured using LC–MS/MS methodology. (44)
T-DM1 showed linear pharmacokinetics across doses ranging from 2.4 to 4.8 mg/kg. Lower doses (<1.2 mg/kg) administered to patients in the phase I trial showed evidence of nonlinearity, consistent with a contribution of concentration-dependent target-mediated clearance to the overall clearance. (44) In an analysis of 272 patients dosed at 3.6 mg/kg across four clinical studies, the Cmax for T-DM1 was 75.6–80.9 μg/mL, the t1/2 was about 4.0 days, and the CL was 7.0–13 mL day–1 kg–1. (44) There was no significant accumulation of T-DM1 on the q3 week schedule. The total trastuzumab measurements showed a longer t1/2 (about 9–11 days) and a slower CL (about 4–5 mL day–1 kg–1) than T-DM1, which can be explained by a slow rate of deconjugation. (44, 45) Furthermore, given that T-DM1 is a mixture of molecules from one to eight linked DM1 molecules per antibody, a slightly faster clearance of T-DM1 species bearing higher amounts of linked DM1 (e.g., species with ≥5 DM1/antibody) versus species with lower amounts of linked DM1 (e.g., ≤3 DM1/antibody) could also contribute to the observed slow decrease in the average ratio of DM1/antibody in circulation over time. (45, 46) Population pharmacokinetic analysis suggests that T-DM1 exposure was relatively constant among patients, although body weight did have the greatest impact on the interindividual variability in some of the pharmacokinetic parameters. (47) Markers of liver function of renal function had no clinically meaningful effect on the pharmacokinetics of T-DM1.
Plasma DM1 concentrations were consistently low, with an average level of about 5 ng/mL in patients dosed with T-DM1 at 3.6 mg/kg, levels of free DM1 that correspond to only about 0.3% of the total conjugated DM1 in circulation, demonstrating the good stability of the linker chemistry in vivo. (44, 47) Exploratory analysis of maytansinoid catabolites from T-DM1 showed low concentrations of the linker–DM1 adduct (MCC-DM1) and lysine–linker–DM1 (lysine-Nε-MCC-DM1) in plasma (<1.9–122 ng/mL and <1.08–6.38 ng/mL, respectively). The MCC-DM1 levels were highest immediately after dosing, while the lysine-Nε-MCC-DM1 levels peaked at later time-points as might be expected for a catabolite generated via intracellular proteolytic degradation of T-DM1 within lysosomes of cells. (21, 48) The maytansinoid metabolites detected in clinical samples were as expected from metabolic studies in rats of T-DM1 made with tritium-labeled DM1. The fecal/biliary route appears to be the major pathway of elimination of the DM1-containing catabolites. (29, 48)
The incidence of a human anti-T-DM1 antibody response has been low (<5%) in patients exposed to repeated doses of T-DM1. Where they were observed in clinical trials (in 13 of 286 patients), there were no obvious changes in pharmacokinetics, safety profiles, or efficacy outcomes. (44)

Safety and Tolerability Profile of T-DM1

Click to copy section linkSection link copied!

Nearly all patients receiving T-DM1 at 3.6 mg/kg, q3 weeks, experience transient declines in platelets, often beginning 1 day after each dose and reaching a nadir on about day 8 with recovery by about day 15. (31, 34, 35, 38, 47, 49) Most such AEs were grade 1 or 2, while grade ≥3 events were infrequent, seen in about 10% of patients across most studies, and were generally rapidly reversible and noncumulative. (31, 34, 35, 47) Reports of any clinically significant bleeding events were uncommon, and only rarely were there any reports of the need for platelet transfusions. Nonclinical studies suggest that T-DM1 can be taken up by megakaryocytes via nontarget mediated uptake mechanisms (perhaps pinocytosis), whereupon intracellular generation of the active catabolite, lysine-Nε-MCC-DM1, can result in disruption of microtubules and proplatelet production, pointing to a possible mechanism for thrombocytopenia in patients. (49) It is interesting to note that while thrombocytopenia defined the DLT of T-DM1 in human, the severity of thrombocytopenia in the non-human primate toxicology studies was minimal at much higher doses and did not define MTD in the preclinical safety studies (see Preclinical Toxicology and Toxicokinetics of T-DM1, above).
Similar to thrombocytopenia, reversible elevations in serum levels of hepatic transaminases were seen in many patients upon dosing with T-DM1, the majority of such events being grade 1 or 2 with grade 3 or 4 AEs seen in less than 10% of patients and one grade 5 event reported to date in a patient with underlying fatty liver disease. (35, 38) Other potential toxicities common to microtubule-disrupting agents were notable by their infrequency: alopecia was almost never seen, while neutropenia, leukopenia, and nervous system toxicities were not reported at clinically significant grades or frequencies. (31, 34, 36) While a serious adverse event of trastuzumab treatment is cardiotoxicity seen in <5% of patients, (50) the clinical studies thus far suggest that the risk of cardiotoxicity of T-DM1 is low, (31, 32, 34, 38) although most evidence reported to date is for patients who previously received trastuzumab, so susceptible patients may have been excluded from most of the reported clinical studies. (34, 38)

Conclusion

Click to copy section linkSection link copied!

T-DM1, ado-trastuzumab emtansine, is the first antibody–drug conjugate (ADC) to receive full approval from FDA (February 22, 2013). It was approved as a new therapy for patients with HER2-positive late stage (metastatic) breast cancer, previously treated with trastuzumab and taxane chemotherapy. It fulfils the long-sought objective of ADC development, that of better tolerated, more active anticancer agents, and its early promise encourages the development of other agents based on similar ADC platform technologies for a wide array of cancers.

Author Information

Click to copy section linkSection link copied!

  • Corresponding Authors
    • John M. Lambert - ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States Email: [email protected]
    • Ravi V. J. Chari - ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States Email: [email protected]
    • Notes
      The authors declare the following competing financial interest(s): The authors are employees of ImmunoGen, Inc., the developer of the maytansinoid ADC platform utilized in T-DM1.

    Acknowledgment

    Click to copy section linkSection link copied!

    We thank Dr. Gail Lewis Phillips from Genentech, a member of the Roche group, for kindly providing us some figures for inclusion in this paper.

    Abbreviations Used

    Click to copy section linkSection link copied!

    T-DM1

    ado-trastuzumab emtansine

    DM1

    N2′-deacetyl-N2′-(3-mercapto-1-oxopropyl)maytansine

    SPDP

    N-succinimidyl 3-(2-pyridyldithio)propionate

    SPP

    N-succinimidyl 4-(2-pyridyldithio)pentanoate

    SSNPP

    N-sulfosuccinimidyl 4-(5-nitro-2-pyridyldithio)pentanoate

    SMCC

    N-succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate

    MCC

    4-(N-maleimidomethyl)cyclohexane-1-carboxylate

    EDC

    N-ethyl-N′-(3-dimethylaminopropyl) carbodiimide hydrochloride

    rt

    room temperature

    HER2

    human epidermal growth factor receptor 2

    ADC

    antibody–drug conjugate

    ADCC

    antibody-dependent cell-mediated cytotoxicity

    ELISA

    enzyme-linked immunosorbent assay

    FDA

    Food and Drug Administration

    MBC

    metastatic breast cancer

    SAR

    structure–activity relationship

    MDR

    multidrug resistance

    DAR

    DM1-to-antibody ratio

    MTD

    maximum tolerated dose

    DLT

    dose-limiting toxicity

    AE

    adverse event

    CBR

    clinical benefit rate

    ORR

    overall response rate

    CR

    complete response

    PFS

    progression-free survival

    AST

    aspartate aminotransferase

    ALT

    alanine aminotransferase

    References

    Click to copy section linkSection link copied!

    This article references 50 other publications.

    1. 1
      Colomer, R.; Shamon, L. A.; Tsai, M. S.; Lupu, R. Herceptin: from the bench to the clinic Cancer Invest. 2001, 19, 49 56
    2. 2
      Pegram, M. D.; Konecny, G. E.; O’Callaghan, C.; Beryt, M.; Pietras, R.; Slamon, D. J. Rational combinations of trastuzumab with chemotherapeutic drugs used in the treatment of breast cancer J. Natl. Cancer Inst. 2004, 96, 739 749
    3. 3
      Shepard, H. M.; Lewis, G. D.; Sarup, J. C.; Fendly, B. M.; Maneval, D.; Mordenti, J.; Figari, I.; Kotts, C. E.; Palladino, M. A., Jr.; Ullrich, A. Monoclonal antibody therapy of human cancer: taking the HER2 protooncogene to the clinic J. Clin. Immunol. 1991, 11, 117 127
    4. 4
      Spector, N. L.; Blackwell, K. L. Understanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancer J. Clin. Oncol. 2009, 27, 5838 5847
    5. 5
      Slamon, D. J.; Leyland-Jones, B.; Shak, S.; Fuchs, H.; Paton, V.; Bajamonde, A.; Fleming, T.; Eiermann, W.; Wolter, J.; Pegram, M.; Baselga, J.; Norton, L. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2 N. Engl. J. Med. 2001, 344, 783 792
    6. 6
      Nahta, R.; Esteva, F. J. HER-2-targeted therapy: lessons learned and future directions Clin. Cancer Res. 2003, 9, 5078 5084
    7. 7
      Metzger-Filho, O.; Winer, E. P.; Krop, I. Pertuzumab: optimizing HER2 blockade Clin. Cancer Res. 2013, 19, 5552 5556
    8. 8
      Chari, R. V.; Miller, M. L.; Widdison, W. C. Antibody–drug conjugates: an emerging concept in cancer therapy Angew. Chem., Int. Ed. 2014, 53, 3796 3827
    9. 9
      Blattler, W. A.; Chari, R. V. J. Methods of treatment using anti-ERBB antibody–maytansinoid conjugates. U.S. Patent 8,337,856, 2012.
    10. 10
      Sedlacek, H. H. Antibodies as Carriers of Cytotoxicity; Karger: Basel, Switzerland, 1992; pp 1 145.
    11. 11
      Chari Ravi, V. J. Targeted cancer therapy: conferring specificity to cytotoxic drugs Acc. Chem. Res. 2008, 41, 98 107
    12. 12
      Kupchan, S. M.; Komoda, Y.; Branfman, A. R.; Sneden, A. T.; Court, W. A.; Thomas, G. J.; Hintz, H. P.; Smith, R. M.; Karim, A.; Howie, G. A.; Verma, A. K.; Nagao, Y.; Dailey, R. G., Jr.; Zimmerly, V. A.; Sumner, W. C., Jr. The maytansinoids. Isolation, structural elucidation, and chemical interrelation of novel ansa macrolides J. Org. Chem. 1977, 42, 2349 2357
    13. 13
      Widdison, W. C.; Wilhelm, S. D.; Cavanagh, E. E.; Whiteman, K. R.; Leece, B. A.; Kovtun, Y.; Goldmacher, V. S.; Xie, H.; Steeves, R. M.; Lutz, R. J.; Zhao, R.; Wang, L.; Blattler, W. A.; Chari, R. V. Semisynthetic maytansine analogues for the targeted treatment of cancer J. Med. Chem. 2006, 49, 4392 4408
    14. 14
      Issell, B. F.; Crooke, S. T. Maytansine Cancer Treat. Rev. 1978, 5, 199 207
    15. 15
      Kupchan, S. M.; Sneden, A. T.; Branfman, A. R.; Howie, G. A.; Rebhun, L. I.; McIvor, W. E.; Wang, R. W.; Schnaitman, T. C. Structural requirements for antileukemic activity among the naturally occurring and semisynthetic maytansinoids J. Med. Chem. 1978, 21, 31 37
    16. 16
      Chari, R. V.; Martell, B. A.; Gross, J. L.; Cook, S. B.; Shah, S. A.; Blattler, W. A.; McKenzie, S. J.; Goldmacher, V. S. Immunoconjugates containing novel maytansinoids: promising anticancer drugs Cancer Res. 1992, 52, 127 131
    17. 17
      Kellogg, B. A.; Garrett, L.; Kovtun, Y.; Lai, K. C.; Leece, B.; Miller, M.; Payne, G.; Steeves, R.; Whiteman, K. R.; Widdison, W.; Xie, H.; Singh, R.; Chari, R. V.; Lambert, J. M.; Lutz, R. J. Disulfide-linked antibody-maytansinoid conjugates: optimization of in vivo activity by varying the steric hindrance at carbon atoms adjacent to the disulfide linkage Bioconjugate Chem. 2011, 22, 717 727
    18. 18
      Wakankar, A.; Chen, Y.; Gokarn, Y.; Jacobson, F. S. Analytical methods for physicochemical characterization of antibody drug conjugates mAbs 2011, 3, 161 172
    19. 19
      Wakankar, A. A.; Feeney, M. B.; Rivera, J.; Chen, Y.; Kim, M.; Sharma, V. K.; Wang, Y. J. Physicochemical stability of the antibody–drug conjugate trastuzumab-DM1: changes due to modification and conjugation processes Bioconjugate Chem. 2010, 21, 1588 1595
    20. 20
      Lewis Phillips, G. D.; Li, G.; Dugger, D. L.; Crocker, L. M.; Parsons, K. L.; Mai, E.; Blattler, W. A.; Lambert, J. M.; Chari, R. V.; Lutz, R. J.; Wong, W. L.; Jacobson, F. S.; Koeppen, H.; Schwall, R. H.; Kenkare-Mitra, S. R.; Spencer, S. D.; Sliwkowski, M. X. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate Cancer Res. 2008, 68, 9280 9290
    21. 21
      Erickson, H. K.; Lewis Phillips, G. D.; Leipold, D. D.; Provenzano, C. A.; Mai, E.; Johnson, H. A.; Gunter, B.; Audette, C. A.; Gupta, M.; Pinkas, J.; Tibbitts, J. The effect of different linkers on target cell catabolism and pharmacokinetics/pharmacodynamics of trastuzumab maytansinoid conjugates Mol. Cancer Ther. 2012, 11, 1133 1142
    22. 22
      Junttila, T. T.; Li, G.; Parsons, K.; Phillips, G. L.; Sliwkowski, M. X. Trastuzumab-DM1 (T-DM1) retains all the mechanisms of action of trastuzumab and efficiently inhibits growth of lapatinib insensitive breast cancer Breast Cancer Res. Treat. 2011, 128, 347 356
    23. 23
      Barok, M.; Tanner, M.; Koninki, K.; Isola, J. Trastuzumab-DM1 is highly effective in preclinical models of HER2-positive gastric cancer Cancer Lett. 2011, 306, 171 179
    24. 24
      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.; Lai, K. C.; Widdison, W. C.; Kellogg, B.; Johnson, H.; Pinkas, J.; Lutz, R. J.; Singh, R.; Goldmacher, V. S.; Chari, R. V. Antibody–maytansinoid conjugates designed to bypass multidrug resistance Cancer Res. 2010, 70, 2528 2537
    25. 25
      Guo, J.; Li, G.; Lewis Phillips, G. D. Role of multidrug resistance transporters in the biological response to trastuzumab–cytotoxic drug conjugates Cancer Res. 2010, 70 (Suppl. 1) Abstract 618
    26. 26
      Jumbe, N. L.; Xin, Y.; Leipold, D. D.; Crocker, L.; Dugger, D.; Mai, E.; Sliwkowski, M. X.; Fielder, P. J.; Tibbitts, J. Modeling the efficacy of trastuzumab–DM1, an antibody drug conjugate, in mice J. Pharmacokinet. Pharmacodyn. 2010, 37, 221 242
    27. 27
      Kaur, S.; Xu, K.; Saad, O. M.; Dere, R. C.; Carrasco-Triguero, M. Bioanalytical assay strategies for the development of antibody–drug conjugate biotherapeutics Bioanalysis 2013, 5, 201 226
    28. 28
      Poon, K. A.; Flagella, K.; Beyer, J.; Tibbitts, J.; Kaur, S.; Saad, O.; Yi, J. H.; Girish, S.; Dybdal, N.; Reynolds, T. Preclinical safety profile of trastuzumab emtansine (T-DM1): mechanism of action of its cytotoxic component retained with improved tolerability Toxicol. Appl. Pharmacol. 2013, 273, 298 313
    29. 29
      Sun, X.; Widdison, W.; Mayo, M.; Wilhelm, S.; Leece, B.; Chari, R.; Singh, R.; Erickson, H. Design of antibody–maytansinoid conjugates allows for efficient detoxification via liver metabolism Bioconjugate Chem. 2011, 22, 728 735
    30. 30
      Fishkin, N.; Maloney, E. K.; Chari, R. V.; Singh, R. A novel pathway for maytansinoid release from thioether linked antibody–drug conjugates (ADCs) under oxidative conditions Chem. Commun. 2011, 47, 10752 10754
    31. 31
      Krop, I. E.; Beeram, M.; Modi, S.; Jones, S. F.; Holden, S. N.; Yu, W.; Girish, S.; Tibbitts, J.; Yi, J. H.; Sliwkowski, M. X.; Jacobson, F.; Lutzker, S. G.; Burris, H. A. Phase I study of trastuzumab–DM1, an HER2 antibody–drug conjugate, given every 3 weeks to patients with HER2-positive metastatic breast cancer J. Clin. Oncol. 2010, 28, 2698 2704
    32. 32
      Baselga, J. Phase I and II clinical trials of trastuzumab Ann. Oncol. 2001, 12 (Suppl. 1) S49 S55
    33. 33
      Beeram, M.; Krop, I. E.; Burris, H. A.; Girish, S. R.; Yu, W.; Lu, M. W.; Holden, S. N.; Modi, S. A phase 1 study of weekly dosing of trastuzumab emtansine (T-DM1) in patients with advanced human epidermal growth factor 2-positive breast cancer Cancer 2012, 118, 5733 5740
    34. 34
      Burris, H. A., 3rd; Rugo, H. S.; Vukelja, S. J.; Vogel, C. L.; Borson, R. A.; Limentani, S.; Tan-Chiu, E.; Krop, I. E.; Michaelson, R. A.; Girish, S.; Amler, L.; Zheng, M.; Chu, Y. W.; Klencke, B.; O’Shaughnessy, J. A. Phase II study of the antibody drug conjugate trastuzumab–DM1 for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer after prior HER2-directed therapy J. Clin. Oncol. 2011, 29, 398 405
    35. 35
      Krop, I. E.; LoRusso, P.; Miller, K. D.; Modi, S.; Yardley, D.; Rodriguez, G.; Guardino, E.; Lu, M.; Zheng, M.; Girish, S.; Amler, L.; Winer, E. P.; Rugo, H. S. A phase II study of trastuzumab emtansine in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer who were previously treated with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine J. Clin. Oncol. 2012, 30, 3234 3241
    36. 36
      Hurvitz, S. A.; Dirix, L.; Kocsis, J.; Bianchi, G. V.; Lu, J.; Vinholes, J.; Guardino, E.; Song, C.; Tong, B.; Ng, V.; Chu, Y. W.; Perez, E. A. Phase II randomized study of trastuzumab emtansine versus trastuzumab plus docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer J. Clin. Oncol. 2013, 31, 1157 1163
    37. 37
      Suter, T. M.; Procter, M.; van Veldhuisen, D. J.; Muscholl, M.; Bergh, J.; Carlomagno, C.; Perren, T.; Passalacqua, R.; Bighin, C.; Klijn, J. G.; Ageev, F. T.; Hitre, E.; Groetz, J.; Iwata, H.; Knap, M.; Gnant, M.; Muehlbauer, S.; Spence, A.; Gelber, R. D.; Piccart-Gebhart, M. J. Trastuzumab-associated cardiac adverse effects in the herceptin adjuvant trial J. Clin. Oncol. 2007, 25, 3859 3865
    38. 38
      Verma, S.; Miles, D.; Gianni, L.; Krop, I. E.; Welslau, M.; Baselga, J.; Pegram, M.; Oh, D. Y.; Dieras, V.; Guardino, E.; Fang, L.; Lu, M. W.; Olsen, S.; Blackwell, K. Trastuzumab emtansine for HER2-positive advanced breast cancer N. Engl. J. Med. 2012, 367, 1783 1791
    39. 39
      Krop, I. E.; Kim, S. B.; Gonzalez-Martin, A.; LoRusso, P. M.; Ferrero, J. M.; Smitt, M.; Yu, R.; Leung, A.; Wildiers, H. Trastuzumab emtansine versus treatment of physician’s choice for pretreated HER2-positive advanced breast cancer (TH3RESA): a randomized open-label phase 3 trial Lancet Oncol. 2014, 15, 689 699
    40. 40
      Baselga, J.; Cortes, J.; Kim, S. B.; Im, S. A.; Hegg, R.; Im, Y. H.; Roman, L.; Pedrini, J. L.; Pienkowski, T.; Knott, A.; Clark, E.; Benyunes, M. C.; Ross, G.; Swain, S. M. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer N. Engl. J. Med. 2012, 366, 109 119
    41. 41
      Phillips, G. D.; Fields, C. T.; Li, G.; Dowbenko, D.; Schaefer, G.; Miller, K.; Andre, F.; Burris, H. A., 3rd; Albain, K. S.; Harbeck, N.; Dieras, V.; Crivellari, D.; Fang, L.; Guardino, E.; Olsen, S. R.; Crocker, L. M.; Sliwkowski, M. X. Dual targeting of HER2-positive cancer with trastuzumab emtansine and pertuzumab: critical role for neuregulin blockade in antitumor response to combination therapy Clin. Cancer Res. 2014, 20, 456 468
    42. 42
      Lu, D.; Burris, H. A., 3rd; Wang, B.; Dees, E. C.; Cortes, J.; Joshi, A.; Gupta, M.; Yi, J. H.; Chu, Y. W.; Shih, T.; Fang, L.; Girish, S. Drug interaction potential of trastuzumab emtansine (T-DM1) combined with pertuzumab in patients with HER2-positive metastatic breast cancer Curr. Drug Metab. 2012, 13, 911 922
    43. 43
      Bartsch, R.; Berghoff, A. S.; Preusser, M. Breast cancer brain metastases responding to primary systemic therapy with T-DM1 J. Neuro-Oncol. 2014, 116, 205 206
    44. 44
      Girish, S.; Gupta, M.; Wang, B.; Lu, D.; Krop, I. E.; Vogel, C. L.; Burris, H. A., III; LoRusso, P. M.; Yi, J. H.; Saad, O.; Tong, B.; Chu, Y. W.; Holden, S.; Joshi, A. Clinical pharmacology of trastuzumab emtansine (T-DM1): an antibody–drug conjugate in development for the treatment of HER2-positive cancer Cancer Chemother. Pharmacol. 2012, 69, 1229 1240
    45. 45
      Lu, D.; Joshi, A.; Wang, B.; Olsen, S.; Yi, J. H.; Krop, I. E.; Burris, H. A.; Girish, S. An integrated multiple-analyte pharmacokinetic model to characterize trastuzumab emtansine (T-DM1) clearance pathways and to evaluate reduced pharmacokinetic sampling in patients with HER2-positive metastatic breast cancer Clin. Pharmacokinet. 2013, 52, 657 672
    46. 46
      Chudasama, V. L.; Schaedeli Stark, F.; Harrold, J. M.; Tibbitts, J.; Girish, S. R.; Gupta, M.; Frey, N.; Mager, D. E. Semi-mechanistic population pharmacokinetic model of multivalent trastuzumab emtansine in patients with metastatic breast cancer Clin. Pharmacol. Ther. 2012, 92, 520 527
    47. 47
      Bender, B. C.; Schaedeli-Stark, F.; Koch, R.; Joshi, A.; Chu, Y. W.; Rugo, H.; Krop, I. E.; Girish, S.; Friberg, L. E.; Gupta, M. A population pharmacokinetic/pharmacodynamic model of thrombocytopenia characterizing the effect of trastuzumab emtansine (T-DM1) on platelet counts in patients with HER2-positive metastatic breast cancer Cancer Chemother. Pharmacol. 2012, 70, 591 601
    48. 48
      Shen, B. Q.; Bumbaca, D.; Saad, O.; Yue, Q.; Pastuskovas, C. V.; Khojasteh, S. C.; Tibbitts, J.; Kaur, S.; Wang, B.; Chu, Y. W.; LoRusso, P. M.; Girish, S. Catabolic fate and pharmacokinetic characterization of trastuzumab emtansine (T-DM1): an emphasis on preclinical and clinical catabolism Curr. Drug Metab. 2012, 13, 901 910
    49. 49
      Thon, J. N.; Devine, M. T.; Begonja, A. J.; Tibbitts, J.; Italiano, J. E., Jr. High-content live-cell imaging assay used to establish mechanism of trastuzumab emtansine (T-DM1)-mediated inhibition of platelet production Blood 2012, 120, 1975 1984
    50. 50
      Sendur, M. A.; Aksoy, S.; Altundag, K. Cardiotoxicity of novel HER2-targeted therapies Curr. Med. Res. Opin. 2013, 29, 1015 1024

    Cited By

    Click to copy section linkSection link copied!

    This article is cited by 394 publications.

    1. Meiling Qi, Chenyue Zhu, Yi Chen, Chenxi Wang, Xinyuan Ye, Sen Li, Zhongzhe Cheng, Hongliang Jiang, Zhifeng Du. Site-Specific Stability Evaluation of Antibody–Drug Conjugate in Serum Using a Validated Liquid Chromatography–Mass Spectrometry Method. Journal of Proteome Research 2024, Article ASAP.
    2. Michael Cochran, Isaac Marks, Tyler Albin, Danny Arias, Philip Kovach, Beatrice Darimont, Hanhua Huang, Usue Etxaniz, Hae Won Kwon, Yunyu Shi, Matthew Diaz, Olecya Tyaglo, Arthur Levin, Venkata Ramana Doppalapudi. Structure–Activity Relationship of Antibody–Oligonucleotide Conjugates: Evaluating Bioconjugation Strategies for Antibody–Phosphorodiamidate Morpholino Oligomer Conjugates for Drug Development. Journal of Medicinal Chemistry 2024, 67 (17) , 14868-14884. https://doi.org/10.1021/acs.jmedchem.4c00803
    3. Mahadeva M. M. Swamy, Yuta Murai, Kenji Monde, Setsuko Tsuboi, Aravind K. Swamy, Takashi Jin. Biocompatible and Water-Soluble Shortwave-Infrared (SWIR)-Emitting Cyanine-Based Fluorescent Probes for In Vivo Multiplexed Molecular Imaging. ACS Applied Materials & Interfaces 2024, 16 (14) , 17253-17266. https://doi.org/10.1021/acsami.4c01000
    4. Xiao Zhang, Bowen Zhao, Shiwei Fu, Ronald S. Seruya, John F. Madey, III, Eva Bukhryakova, Fuwu Zhang. Rapid and Versatile Synthesis of Glutathione-Responsive Polycarbonates from Activated Cyclic Carbonates. Macromolecules 2024, 57 (6) , 2858-2867. https://doi.org/10.1021/acs.macromol.3c02633
    5. Qingqing Liu, Yu Wang, Xin Xia, Zhongyue Li, Yaoyao Li, Yuemao Shen, Haoxin Wang. Combinatorial Biosynthesis of 3-O-Carbamoylmaytansinol by Rational Engineering of the Tailoring Steps of Ansamitocins. ACS Synthetic Biology 2024, 13 (3) , 721-727. https://doi.org/10.1021/acssynbio.3c00575
    6. Benqian Wei, Carter Lantz, Rachel R. Ogorzalek Loo, Iain D. G. Campuzano, Joseph A. Loo. Internal Fragments Enhance Middle-Down Mass Spectrometry Structural Characterization of Monoclonal Antibodies and Antibody-Drug Conjugates. Analytical Chemistry 2024, 96 (6) , 2491-2499. https://doi.org/10.1021/acs.analchem.3c04526
    7. Afruja Ahad, Hiwa K. Saeed, Virginia del Solar, Javier E. López-Hernández, Alexa Michel, Joshua Mathew, Jason S. Lewis, Maria Contel. Shifting the Antibody–Drug Conjugate Paradigm: A Trastuzumab-Gold-Based Conjugate Demonstrates High Efficacy against Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer Mouse Model. ACS Pharmacology & Translational Science 2023, 6 (12) , 1972-1986. https://doi.org/10.1021/acsptsci.3c00270
    8. Vlad Bacauanu, Zoe N. Merz, Zhong L. Hua, Simon B. Lang. Nickel-Catalyzed Antibody Bioconjugation. Journal of the American Chemical Society 2023, 145 (47) , 25842-25849. https://doi.org/10.1021/jacs.3c10185
    9. Benqian Wei, Carter Lantz, Weijing Liu, Rosa Viner, Rachel R. Ogorzalek Loo, Iain D. G. Campuzano, Joseph A. Loo. Added Value of Internal Fragments for Top-Down Mass Spectrometry of Intact Monoclonal Antibodies and Antibody–Drug Conjugates. Analytical Chemistry 2023, 95 (24) , 9347-9356. https://doi.org/10.1021/acs.analchem.3c01426
    10. Natalia Danielewicz, Francesca Rosato, Jana Tomisch, Jonas Gräber, Birgit Wiltschi, Gerald Striedner, Winfried Römer, Juergen Mairhofer. Clickable Shiga Toxin B Subunit for Drug Delivery in Cancer Therapy. ACS Omega 2023, 8 (17) , 15406-15421. https://doi.org/10.1021/acsomega.3c00667
    11. Peidong Wu, Theeraphop Prachyathipsakul, Uyen Huynh, Jingyi Qiu, D. Joseph Jerry, S. Thayumanavan. Optimizing Conjugation Chemistry, Antibody Conjugation Site, and Surface Density in Antibody–Nanogel Conjugates (ANCs) for Cell-Specific Drug Delivery. Bioconjugate Chemistry 2023, 34 (4) , 707-718. https://doi.org/10.1021/acs.bioconjchem.3c00034
    12. Ki Bum Hong, Hongchan An. Degrader–Antibody Conjugates: Emerging New Modality. Journal of Medicinal Chemistry 2023, 66 (1) , 140-148. https://doi.org/10.1021/acs.jmedchem.2c01791
    13. Douglas S. MacPherson, Dobeen Hwang, Samantha M. Sarrett, Outi Keinänen, Cindy Rodriguez, Christoph Rader, Brian M. Zeglis. Leveraging a Dual Variable Domain Immunoglobulin to Create a Site-Specifically Modified Radioimmunoconjugate. Molecular Pharmaceutics 2023, 20 (1) , 775-782. https://doi.org/10.1021/acs.molpharmaceut.2c00700
    14. Siddharth S. Matikonda, Ryan McLaughlin, Pradeep Shrestha, Carol Lipshultz, Martin J. Schnermann. Structure–Activity Relationships of Antibody-Drug Conjugates: A Systematic Review of Chemistry on the Trastuzumab Scaffold. Bioconjugate Chemistry 2022, 33 (7) , 1241-1253. https://doi.org/10.1021/acs.bioconjchem.2c00177
    15. Mei Zhu, Lei Zhou, Shangjiu Hu, Qingfang Miao, Jianhua Gong, Na Zhang, Guoning Zhang, Minghua Wang, Juxian Wang, Hongwei He, Yucheng Wang. Rational Design and Systemic Appraisal of an EGFR-Targeting Antibody–Drug Conjugate LR-DM1 for Pancreatic Cancer. Journal of Medicinal Chemistry 2022, 65 (10) , 7141-7153. https://doi.org/10.1021/acs.jmedchem.1c01920
    16. TaeJin Lee, Ju Hwan Kim, Se Jeong Kwon, Jin Woo Seo, Sun Hee Park, Jinyoung Kim, Jonghwa Jin, Ji Hye Hong, Hyo Jin Kang, Chiranjeev Sharma, Ji Hoon Choi, Sang J. Chung. Site-Selective Antibody–Drug Conjugation by a Proximity-Driven S to N Acyl Transfer Reaction on a Therapeutic Antibody. Journal of Medicinal Chemistry 2022, 65 (7) , 5751-5759. https://doi.org/10.1021/acs.jmedchem.2c00084
    17. Zeru Tian, Chenfei Yu, Weijie Zhang, Kuan-Lin Wu, Chenhang Wang, Ruchi Gupta, Zhan Xu, Ling Wu, Yuda Chen, Xiang H.-F. Zhang, Han Xiao. Bone-Specific Enhancement of Antibody Therapy for Breast Cancer Metastasis to Bone. ACS Central Science 2022, 8 (3) , 312-321. https://doi.org/10.1021/acscentsci.1c01024
    18. Setsuko Tsuboi, Takashi Jin. In Vitro and In Vivo Fluorescence Imaging of Antibody–Drug Conjugate-Induced Tumor Apoptosis Using Annexin V–EGFP Conjugated Quantum Dots. ACS Omega 2022, 7 (2) , 2105-2113. https://doi.org/10.1021/acsomega.1c05636
    19. Lin Zhang, Zewei Wang, Zhiyuan Wang, Fang Luo, Mingfeng Guan, Meimei Xu, Yundong Li, Yaoyang Zhang, Zhaoyin Wang, Wenyuan Wang. A Simple and Efficient Method to Generate Dual Site-Specific Conjugation ADCs with Cysteine Residue and an Unnatural Amino Acid. Bioconjugate Chemistry 2021, 32 (6) , 1094-1104. https://doi.org/10.1021/acs.bioconjchem.1c00134
    20. Ryo Okajima, Shuichi Hiraoka, Takefumi Yamashita. Environmental Effects on Salt Bridge Stability in the Protein–Protein Interface: The Case of Hen Egg-White Lysozyme and Its Antibody, HyHEL-10. The Journal of Physical Chemistry B 2021, 125 (6) , 1542-1549. https://doi.org/10.1021/acs.jpcb.0c09248
    21. Heinz Fabian Raber, Thomas Heerde, Suzanne Nour El Din, Carolin Flaig, Fabienne Hilgers, Nora Bitzenhofer, Karl-Erich Jäger, Thomas Drepper, Kay-Eberhard Gottschalk, Nicholas Emil Bodenberger, Tanja Weil, Dennis Horst Kubiczek, Frank Rosenau. Azulitox—A Pseudomonas aeruginosa P28-Derived Cancer-Cell-Specific Protein Photosensitizer. Biomacromolecules 2020, 21 (12) , 5067-5076. https://doi.org/10.1021/acs.biomac.0c01216
    22. Yam B. Poudel, Naidu S. Chowdari, Heng Cheng, Christiana I. Iwuagwu, H. Dalton King, Srikanth Kotapati, David Passmore, Richard Rampulla, Arvind Mathur, Gregory Vite, Sanjeev Gangwar. Chemical Modification of Linkers Provides Stable Linker–Payloads for the Generation of Antibody–Drug Conjugates. ACS Medicinal Chemistry Letters 2020, 11 (11) , 2190-2194. https://doi.org/10.1021/acsmedchemlett.0c00325
    23. Hans-Georg Lerchen, Beatrix Stelte-Ludwig, Anette Sommer, Sandra Berndt, Anne-Sophie Rebstock, Sarah Johannes, Christoph Mahlert, Simone Greven, Lisa Dietz, Hannah Jörißen. Tailored Linker Chemistries for the Efficient and Selective Activation of ADCs with KSPi Payloads. Bioconjugate Chemistry 2020, 31 (8) , 1893-1898. https://doi.org/10.1021/acs.bioconjchem.0c00357
    24. María A. Oliva, Andrea E. Prota, Javier Rodríguez-Salarichs, Youssef L. Bennani, Jesús Jiménez-Barbero, Katja Bargsten, Ángeles Canales, Michel O. Steinmetz, J. Fernando Díaz. Structural Basis of Noscapine Activation for Tubulin Binding. Journal of Medicinal Chemistry 2020, 63 (15) , 8495-8501. https://doi.org/10.1021/acs.jmedchem.0c00855
    25. Marı́a Maneiro, Nafsika Forte, Maria M. Shchepinova, Cyrille S. Kounde, Vijay Chudasama, James Richard Baker, Edward W. Tate. Antibody–PROTAC Conjugates Enable HER2-Dependent Targeted Protein Degradation of BRD4. ACS Chemical Biology 2020, 15 (6) , 1306-1312. https://doi.org/10.1021/acschembio.0c00285
    26. Jiaxun Wan, Yongjing Li, Ke Jin, Jia Guo, Jiangtao Xu, Changchun Wang. Robust Strategy for Antibody–Polymer–Drug Conjugation: Significance of Conjugating Orientation and Linker Charge on Targeting Ability. ACS Applied Materials & Interfaces 2020, 12 (21) , 23717-23725. https://doi.org/10.1021/acsami.0c04471
    27. Fang Dong, Jian-Quan Liu, Xiang-Shan Wang. CuBr-Catalyzed α-Arylation and Aerobic Oxidative Dehydrogenative C–N Coupling for the Synthesis of Spiro[cyclohexane-1,12′-isoindolo[1,2-b]quinazolin]-10′-one Derivatives. Organic Letters 2020, 22 (8) , 2887-2891. https://doi.org/10.1021/acs.orglett.0c00497
    28. Eiko Seki, Tatsuo Yanagisawa, Mitsuo Kuratani, Kensaku Sakamoto, Shigeyuki Yokoyama. Fully Productive Cell-Free Genetic Code Expansion by Structure-Based Engineering of Methanomethylophilus alvus Pyrrolysyl-tRNA Synthetase. ACS Synthetic Biology 2020, 9 (4) , 718-732. https://doi.org/10.1021/acssynbio.9b00288
    29. Iván Ramos-Tomillero, Gema Pérez-Chacon, Beatriz Somovilla-Crespo, Francisco Sánchez-Madrid, Carmen Cuevas, Juan Manuel Zapata, Juan Manuel Domínguez, Hortensia Rodríguez, Fernando Albericio. From Ugi Multicomponent Reaction to Linkers for Bioconjugation. ACS Omega 2020, 5 (13) , 7424-7431. https://doi.org/10.1021/acsomega.0c00099
    30. Arnab Rudra, Junwei Li, Rameen Shakur, Sachin Bhagchandani, Robert Langer. Trends in Therapeutic Conjugates: Bench to Clinic. Bioconjugate Chemistry 2020, 31 (3) , 462-473. https://doi.org/10.1021/acs.bioconjchem.9b00828
    31. Chen Bai, Emily E. Reid, Alan Wilhelm, Manami Shizuka, Erin K. Maloney, Rassol Laleau, Lauren Harvey, Katie E. Archer, Dilrukshi Vitharana, Sharlene Adams, Yelena Kovtun, Michael L. Miller, Ravi Chari, Thomas A. Keating, Nicholas C. Yoder. Site-Specific Conjugation of the Indolinobenzodiazepine DGN549 to Antibodies Affords Antibody–Drug Conjugates with an Improved Therapeutic Index as Compared with Lysine Conjugation. Bioconjugate Chemistry 2020, 31 (1) , 93-103. https://doi.org/10.1021/acs.bioconjchem.9b00777
    32. Vasilii F. Otvagin, Natalia S. Kuzmina, Lubov V. Krylova, Arthur B. Volovetsky, Alexander V. Nyuchev, Andrei E. Gavryushin, Ivan N. Meshkov, Yulia G. Gorbunova, Yuliya V. Romanenko, Oscar I. Koifman, Irina V. Balalaeva, Alexey Yu Fedorov. Water-Soluble Chlorin/Arylaminoquinazoline Conjugate for Photodynamic and Targeted Therapy. Journal of Medicinal Chemistry 2019, 62 (24) , 11182-11193. https://doi.org/10.1021/acs.jmedchem.9b01294
    33. Alexei S. Karpov, Cristina M. Nieto-Oberhuber, Tinya Abrams, Edwige Beng-Louka, Enrique Blanco, Sylvie Chamoin, Patrick Chene, Isabelle Dacquignies, Dylan Daniel, Michael P. Dillon, Lionel Doumampouom-Metoul, Nikolaos Drosos, Pavel Fedoseev, Markus Furegati, Brian Granda, Robert M. Grotzfeld, Suzanna Hess Clark, Emilie Joly, Darryl Jones, Marion Lacaud-Baumlin, Stephanie Lagasse-Guerro, Edward G. Lorenzana, William Mallet, Piotr Martyniuk, Andreas L. Marzinzik, Yannick Mesrouze, Sandro Nocito, Yoko Oei, Francesca Perruccio, Grazia Piizzi, Etienne Richard, Patrick J. Rudewicz, Patrick Schindler, Mélanie Velay, Kristine Venstrom, Peiyin Wang, Mauro Zurini, Marc Lafrance. Discovery of Potent and Selective Antibody–Drug Conjugates with Eg5 Inhibitors through Linker and Payload Optimization. ACS Medicinal Chemistry Letters 2019, 10 (12) , 1674-1679. https://doi.org/10.1021/acsmedchemlett.9b00468
    34. Paulin L. Salomon, Emily E. Reid, Katie E. Archer, Luke Harris, Erin K. Maloney, Alan J. Wilhelm, Michael L. Miller, Ravi V. J. Chari, Thomas A. Keating, Rajeeva Singh. Optimizing Lysosomal Activation of Antibody–Drug Conjugates (ADCs) by Incorporation of Novel Cleavable Dipeptide Linkers. Molecular Pharmaceutics 2019, 16 (12) , 4817-4825. https://doi.org/10.1021/acs.molpharmaceut.9b00696
    35. Joshua A. Walker, John J. Bohn, Francis Ledesma, Michelle R. Sorkin, Sneha R. Kabaria, Dana N. Thornlow, Christopher A. Alabi. Substrate Design Enables Heterobifunctional, Dual “Click” Antibody Modification via Microbial Transglutaminase. Bioconjugate Chemistry 2019, 30 (9) , 2452-2457. https://doi.org/10.1021/acs.bioconjchem.9b00522
    36. Nicholas C. Yoder, Chen Bai, Daniel Tavares, Wayne C. Widdison, Kathleen R. Whiteman, Alan Wilhelm, Sharon D. Wilhelm, Molly A. McShea, Erin K. Maloney, Olga Ab, Lintao Wang, Shan Jin, Hans K. Erickson, Thomas A. Keating, John M. Lambert. A Case Study Comparing Heterogeneous Lysine- and Site-Specific Cysteine-Conjugated Maytansinoid Antibody-Drug Conjugates (ADCs) Illustrates the Benefits of Lysine Conjugation. Molecular Pharmaceutics 2019, 16 (9) , 3926-3937. https://doi.org/10.1021/acs.molpharmaceut.9b00529
    37. Kelsey E. Knewtson, Chamani Perera, David Hymel, Zhe Gao, Molly M. Lee, Blake R. Peterson. Antibody–Drug Conjugate that Exhibits Synergistic Cytotoxicity with an Endosome–Disruptive Peptide. ACS Omega 2019, 4 (7) , 12955-12968. https://doi.org/10.1021/acsomega.9b01585
    38. Dian Su, Jinhua Chen, Ely Cosino, Josefa dela Cruz-Chuh, Helen Davis, Geoffrey Del Rosario, Isabel Figueroa, Leanne Goon, Jintang He, Amrita V. Kamath, Surinder Kaur, Katherine R. Kozak, Jeffrey Lau, Donna Lee, M. Violet Lee, Douglas Leipold, Luna Liu, Peter Liu, Guo-Liang Lu, Chris Nelson, Carl Ng, Thomas H. Pillow, Paul Polakis, Andrew G. Polson, Rebecca K. Rowntree, Ola Saad, Brian Safina, Nicola J. Stagg, Moana Tercel, Richard Vandlen, Breanna S. Vollmar, John Wai, Tao Wang, BinQing Wei, Keyang Xu, Juanjuan Xue, Zijin Xu, Gang Yan, Hui Yao, Shang-Fan Yu, Donglu Zhang, Fiona Zhong, Peter S. Dragovich. Antibody–Drug Conjugates Derived from Cytotoxic seco-CBI-Dimer Payloads Are Highly Efficacious in Xenograft Models and Form Protein Adducts In Vivo. Bioconjugate Chemistry 2019, 30 (5) , 1356-1370. https://doi.org/10.1021/acs.bioconjchem.9b00133
    39. Arnaud C. Tiberghien, Philip W. Howard, William R. F. Goundry, Marc McCormick, Jeremy S. Parker. An Alternative Focus for Route Design for the Synthesis of Antibody–Drug Conjugate Payloads. The Journal of Organic Chemistry 2019, 84 (8) , 4830-4836. https://doi.org/10.1021/acs.joc.8b02876
    40. Brian H. White, Kerry Whalen, Kristina Kriksciukaite, Rossitza Alargova, Tsun Au Yeung, Patrick Bazinet, Adam Brockman, Michelle DuPont, Haley Oller, Charles-Andre Lemelin, Patrick Lim Soo, Benoît Moreau, Samantha Perino, James M. Quinn, Gitanjali Sharma, Rajesh Shinde, Beata Sweryda-Krawiec, Richard Wooster, Mark T. Bilodeau. Discovery of an SSTR2-Targeting Maytansinoid Conjugate (PEN-221) with Potent Activity in Vitro and in Vivo. Journal of Medicinal Chemistry 2019, 62 (5) , 2708-2719. https://doi.org/10.1021/acs.jmedchem.8b02036
    41. Xun Liu, Fan Wu, Yong Ji, Lichen Yin. Recent Advances in Anti-cancer Protein/Peptide Delivery. Bioconjugate Chemistry 2019, 30 (2) , 305-324. https://doi.org/10.1021/acs.bioconjchem.8b00750
    42. Anokha S. Ratnayake, Li-ping Chang, L. Nathan Tumey, Frank Loganzo, Joseph A. Chemler, Melissa Wagenaar, Sylvia Musto, Fengping Li, Jeffrey E. Janso, T. Eric Ballard, Brian Rago, Greg L. Steele, WeiDong Ding, Xidong Feng, Christine Hosselet, Vlad Buklan, Judy Lucas, Frank E. Koehn, Christopher J. O’Donnell, Edmund I. Graziani. Natural Product Bis-Intercalator Depsipeptides as a New Class of Payloads for Antibody–Drug Conjugates. Bioconjugate Chemistry 2019, 30 (1) , 200-209. https://doi.org/10.1021/acs.bioconjchem.8b00843
    43. Hyungjun Kim, Dobeen Hwang, Minsuk Choi, Soyoung Lee, Sukmo Kang, Yonghyun Lee, Sunghyun Kim, Junho Chung, Sangyong Jon. Antibody-Assisted Delivery of a Peptide–Drug Conjugate for Targeted Cancer Therapy. Molecular Pharmaceutics 2019, 16 (1) , 165-172. https://doi.org/10.1021/acs.molpharmaceut.8b00924
    44. Zhengyang Jiang, Zhen Yang, Feng Li, Zheng Li, Nathan Fishkin, Kevin Burgess. Targeted Maytansinoid Conjugate Improves Therapeutic Index for Metastatic Breast Cancer Cells. Bioconjugate Chemistry 2018, 29 (9) , 2920-2926. https://doi.org/10.1021/acs.bioconjchem.8b00340
    45. Alexei S. Karpov, Tinya Abrams, Suzanna Clark, Ankita Raikar, Joseph A. D’Alessio, Michael P. Dillon, Thomas G. Gesner, Darryl Jones, Marion Lacaud, William Mallet, Piotr Martyniuk, Erik Meredith, Morvarid Mohseni, Cristina M. Nieto-Oberhuber, Daniel Palacios, Francesca Perruccio, Grazia Piizzi, Mauro Zurini, Carl Uli Bialucha. Nicotinamide Phosphoribosyltransferase Inhibitor as a Novel Payload for Antibody–Drug Conjugates. ACS Medicinal Chemistry Letters 2018, 9 (8) , 838-842. https://doi.org/10.1021/acsmedchemlett.8b00254
    46. Hang-Ping Yao, Liang Feng, Tian-Hao Weng, Chen-Yu Hu, Sreedhar Reddy Suthe, A. G. M. Mostofa, Ling-Hui Chen, Zhi-Gang Wu, Wei-Lin Wang, Ming-Hai Wang. Preclinical Efficacy of Anti-RON Antibody–Drug Conjugate Zt/g4-MMAE for Targeted Therapy of Pancreatic Cancer Overexpressing RON Receptor Tyrosine Kinase. Molecular Pharmaceutics 2018, 15 (8) , 3260-3271. https://doi.org/10.1021/acs.molpharmaceut.8b00298
    47. Dian Su, Katherine R. Kozak, Jack Sadowsky, Shang-Fan Yu, Aimee Fourie-O’Donohue, Christopher Nelson, Richard Vandlen, Rachana Ohri, Luna Liu, Carl Ng, Jintang He, Helen Davis, Jeff Lau, Geoffrey Del Rosario, Ely Cosino, Josefa dela Cruz-Chuh, Yong Ma, Donglu Zhang, Martine Darwish, Wenwen Cai, Chunjiao Chen, Hongxiang Zhou, Jiawei Lu, Yichin Liu, Surinder Kaur, Keyang Xu, Thomas H. Pillow. Modulating Antibody–Drug Conjugate Payload Metabolism by Conjugation Site and Linker Modification. Bioconjugate Chemistry 2018, 29 (4) , 1155-1167. https://doi.org/10.1021/acs.bioconjchem.7b00785
    48. Jeremy S. Parker, Marc McCormick, David W. Anderson, Beatrice A. Maltman, Lakshmaiah Gingipalli, and Dorin Toader . The Development and Scale-Up of an Antibody Drug Conjugate Tubulysin Payload. Organic Process Research & Development 2017, 21 (10) , 1602-1609. https://doi.org/10.1021/acs.oprd.7b00232
    49. Dorian M. Cheff and Matthew D. Hall . A Drug of Such Damned Nature.1 Challenges and Opportunities in Translational Platinum Drug Research. Journal of Medicinal Chemistry 2017, 60 (11) , 4517-4532. https://doi.org/10.1021/acs.jmedchem.6b01351
    50. Xiuxia Sun, Jose F. Ponte, Nicholas C. Yoder, Rassol Laleau, Jennifer Coccia, Leanne Lanieri, Qifeng Qiu, Rui Wu, Erica Hong, Megan Bogalhas, Lintao Wang, Ling Dong, Yulius Setiady, Erin K. Maloney, Olga Ab, Xiaoyan Zhang, Jan Pinkas, Thomas A. Keating, Ravi Chari, Hans K. Erickson, and John M. Lambert . Effects of Drug–Antibody Ratio on Pharmacokinetics, Biodistribution, Efficacy, and Tolerability of Antibody–Maytansinoid Conjugates. Bioconjugate Chemistry 2017, 28 (5) , 1371-1381. https://doi.org/10.1021/acs.bioconjchem.7b00062
    51. Nazzareno Dimasi, Ryan Fleming, Haihong Zhong, Binyam Bezabeh, Krista Kinneer, Ronald J. Christie, Christine Fazenbaker, Herren Wu, and Changshou Gao . Efficient Preparation of Site-Specific Antibody–Drug Conjugates Using Cysteine Insertion. Molecular Pharmaceutics 2017, 14 (5) , 1501-1516. https://doi.org/10.1021/acs.molpharmaceut.6b00995
    52. Chethana Kulkarni, James E. Finley, Andrew J. Bessire, Xiaotian Zhong, Sylvia Musto, and Edmund I. Graziani . Development of Fluorophore-Labeled Thailanstatin Antibody-Drug Conjugates for Cellular Trafficking Studies. Bioconjugate Chemistry 2017, 28 (4) , 1041-1047. https://doi.org/10.1021/acs.bioconjchem.6b00718
    53. April L. Risinger, Jing Li, Lin Du, Raymond Benavides, Andrew J. Robles, Robert H. Cichewicz, John G. Kuhn, and Susan L. Mooberry . Pharmacokinetic Analysis and in Vivo Antitumor Efficacy of Taccalonolides AF and AJ. Journal of Natural Products 2017, 80 (2) , 409-414. https://doi.org/10.1021/acs.jnatprod.6b00944
    54. Jose F. Ponte, Xiuxia Sun, Nicholas C. Yoder, Nathan Fishkin, Rassol Laleau, Jennifer Coccia, Leanne Lanieri, Megan Bogalhas, Lintao Wang, Sharon Wilhelm, Wayne Widdison, Jan Pinkas, Thomas A. Keating, Ravi Chari, Hans K. Erickson, and John M. Lambert . Understanding How the Stability of the Thiol-Maleimide Linkage Impacts the Pharmacokinetics of Lysine-Linked Antibody–Maytansinoid Conjugates. Bioconjugate Chemistry 2016, 27 (7) , 1588-1598. https://doi.org/10.1021/acs.bioconjchem.6b00117
    55. David Y. Jackson . Processes for Constructing Homogeneous Antibody Drug Conjugates. Organic Process Research & Development 2016, 20 (5) , 852-866. https://doi.org/10.1021/acs.oprd.6b00067
    56. Hannes Merten, Fabian Brandl, Andreas Plückthun, and Uwe Zangemeister-Wittke . Antibody–Drug Conjugates for Tumor Targeting—Novel Conjugation Chemistries and the Promise of non-IgG Binding Proteins. Bioconjugate Chemistry 2015, 26 (11) , 2176-2185. https://doi.org/10.1021/acs.bioconjchem.5b00260
    57. Kohta Mohri, Naoki Morimoto, Megumi Maruyama, Norimasa Nakamoto, Emi Hayashi, Kengo Nagata, Kohei Miyata, Kyohei Ochiai, Ken-ichiro Hiwatari, Kazufumi Tsubaki, Etsuo Tobita, Yuki Ishimaru, Sadaaki Maeda, and Shinji Sakuma . Potential of d-Octaarginine-Linked Polymers as an in Vitro Transfection Tool for Biomolecules. Bioconjugate Chemistry 2015, 26 (8) , 1782-1790. https://doi.org/10.1021/acs.bioconjchem.5b00323
    58. Paulin L. Salomon and Rajeeva Singh . Sensitive ELISA Method for the Measurement of Catabolites of Antibody–Drug Conjugates (ADCs) in Target Cancer Cells. Molecular Pharmaceutics 2015, 12 (6) , 1752-1761. https://doi.org/10.1021/acs.molpharmaceut.5b00028
    59. Serengulam V. Govindan, Thomas M. Cardillo, Edmund A. Rossi, Preeti Trisal, William J. McBride, Robert M. Sharkey, and David M. Goldenberg . Improving the Therapeutic Index in Cancer Therapy by Using Antibody–Drug Conjugates Designed with a Moderately Cytotoxic Drug. Molecular Pharmaceutics 2015, 12 (6) , 1836-1847. https://doi.org/10.1021/mp5006195
    60. Andreas Maderna and Carolyn A. Leverett . Recent Advances in the Development of New Auristatins: Structural Modifications and Application in Antibody Drug Conjugates. Molecular Pharmaceutics 2015, 12 (6) , 1798-1812. https://doi.org/10.1021/mp500762u
    61. Ronald C. Elgersma, Ruud G. E. Coumans, Tijl Huijbregts, Wiro M. P. B. Menge, John A. F. Joosten, Henri J. Spijker, Franciscus M. H. de Groot, Miranda M. C. van der Lee, Ruud Ubink, Diels J. van den Dobbelsteen, David F. Egging, Wim H. A. Dokter, Gijs F. M. Verheijden, Jacques M. Lemmens, C. Marco Timmers, and Patrick H. Beusker . Design, Synthesis, and Evaluation of Linker-Duocarmycin Payloads: Toward Selection of HER2-Targeting Antibody–Drug Conjugate SYD985. Molecular Pharmaceutics 2015, 12 (6) , 1813-1835. https://doi.org/10.1021/mp500781a
    62. Kelly E. Burns, Matthew K. Robinson, and Damien Thévenin . Inhibition of Cancer Cell Proliferation and Breast Tumor Targeting of pHLIP–Monomethyl Auristatin E Conjugates. Molecular Pharmaceutics 2015, 12 (4) , 1250-1258. https://doi.org/10.1021/mp500779k
    63. Carlos Garcia-Echeverria . Developing Second Generation Antibody–Drug Conjugates: The Quest for New Technologies. Journal of Medicinal Chemistry 2014, 57 (19) , 7888-7889. https://doi.org/10.1021/jm501298k
    64. Dan-dan Zhou, Xiao-tian Zhai, Lan-wen Zhang, Zi-hui Xie, Ying Wang, Yong-su Zhen, Rui-juan Gao, Qing-fang Miao. A new TROP2-targeting antibody-drug conjugate shows potent antitumor efficacy in breast and lung cancers. npj Precision Oncology 2024, 8 (1) https://doi.org/10.1038/s41698-024-00584-z
    65. Elena Giordani, Matteo Allegretti, Alberto Sinibaldi, Francesco Michelotti, Gianluigi Ferretti, Elena Ricciardi, Giovanna Ziccheddu, Fabio Valenti, Simona Di Martino, Cristiana Ercolani, Diana Giannarelli, Grazia Arpino, Stefania Gori, Claudia Omarini, Alberto Zambelli, Emilio Bria, Ida Paris, Simonetta Buglioni, Patrizio Giacomini, Alessandra Fabi. Monitoring changing patterns in HER2 addiction by liquid biopsy in advanced breast cancer patients. Journal of Experimental & Clinical Cancer Research 2024, 43 (1) https://doi.org/10.1186/s13046-024-03105-9
    66. T. Tedeschini, B. Campara, A. Grigoletto, I. Zanotto, L. Cannella, D. Gabbia, Y. Matsuno, A. Suzuki, H. Yoshioka, A. Armirotti, S. De Martin, G. Pasut. Optimization of a pendant-shaped PEGylated linker for antibody-drug conjugates. Journal of Controlled Release 2024, 375 , 74-89. https://doi.org/10.1016/j.jconrel.2024.08.049
    67. Antonella Giorgia Becchetti, Anna Martini, Giovanna Scroccaro, Roberta Joppi. History of trastuzumab: a case study in health technology reassessment and natural disinvestment in Veneto Region. Frontiers in Pharmacology 2024, 15 https://doi.org/10.3389/fphar.2024.1406351
    68. Joo-Hyun Jeon, Seo Woo Kim, Yoon-Jung Kim, Jang-Woo Park, Jee Eun Moon, Yong Beom Lee, Hana Yu, Geon-Ho Lee, Sung-Ha Jin, Jin-Hyun Jeong. Synthesis and evaluation of antibody-drug conjugates with high drug-to-antibody ratio using dimaleimide-DM1 as a linker- payload. Bioorganic Chemistry 2024, 149 , 107504. https://doi.org/10.1016/j.bioorg.2024.107504
    69. Fan-jie Qu, Yan Kong, Xin Yan, Hai Wang. Elderly patients with advanced HER2-positive breast cancer with liver metastases benefit from low dose disitamab vedotin (RC48): case series and literature review. Anti-Cancer Drugs 2024, 35 (7) , 658-665. https://doi.org/10.1097/CAD.0000000000001613
    70. Yiran Tao, Ying Lu, Ting Xue, Qinhuai Lai, Hengrui Song, Xiaofeng Chen, Cuiyu Guo, Jinliang Yang, Yuxi Wang. Antibody-drug conjugates targeting DDR1 as a novel strategy for treatment of breast cancer. Journal of Drug Targeting 2024, , 1-21. https://doi.org/10.1080/1061186X.2024.2386621
    71. Qian Zhang, Kaiyan Chen, Xiaoqing Yu, Yun Fan. Spotlight on the treatment of non‐small cell lung cancer with rare genetic alterations and brain metastasis: Current status and future perspectives. International Journal of Cancer 2024, 18 https://doi.org/10.1002/ijc.35070
    72. Peyton High, Cara Guernsey, Shraddha Subramanian, Joan Jacob, Kendra S. Carmon. The Evolving Paradigm of Antibody–Drug Conjugates Targeting the ErbB/HER Family of Receptor Tyrosine Kinases. Pharmaceutics 2024, 16 (7) , 890. https://doi.org/10.3390/pharmaceutics16070890
    73. Adriana Aurelia Chis, Carmen Maximiliana Dobrea, Anca Maria Arseniu, Adina Frum, Luca-Liviu Rus, Gabriela Cormos, Cecilia Georgescu, Claudiu Morgovan, Anca Butuca, Felicia Gabriela Gligor, Andreea Loredana Vonica-Tincu. Antibody–Drug Conjugates—Evolution and Perspectives. International Journal of Molecular Sciences 2024, 25 (13) , 6969. https://doi.org/10.3390/ijms25136969
    74. Weifeng Xu, Nazneen Bano, Olguitza Guzman-Valdes, Jessica Amberman, Elisha Bandlamudi, Pooja Khanna, Rebecca Carmean, Roy Helmy. Development and Validation of a Cell-Based Binding Neutralizing Antibody Assay for an Antibody–Drug Conjugate. The AAPS Journal 2024, 26 (3) https://doi.org/10.1208/s12248-024-00909-7
    75. Yan Zhao, Haolin Jiang, Hang Chen, Jiazhen Yu, Luyao Wang, Wen Zhou, Juanjuan Du. Charge-guided masking of a membrane-destabilizing peptide enables efficient endosomal escape for targeted intracellular delivery of proteins. Acta Pharmaceutica Sinica B 2024, 40 https://doi.org/10.1016/j.apsb.2024.06.022
    76. Khalil Saleh, Rita Khoury, Nadine Khalife, Claude Chahine, Rebecca Ibrahim, Zamzam Tikriti, Axel Le Cesne. Mechanisms of action and resistance to anti-HER2 antibody-drug conjugates in breast cancer. Cancer Drug Resistance 2024, https://doi.org/10.20517/cdr.2024.06
    77. Mustafa Emre Gedik, Ozge Saatci, Nathaniel Oberholtzer, Meral Uner, Ozge Akbulut Caliskan, Metin Cetin, Mertkaya Aras, Kubra Ibis, Burcu Caliskan, Erden Banoglu, Stefan Wiemann, Ayşegül Üner, Sercan Aksoy, Shikhar Mehrotra, Ozgur Sahin. Targeting TACC3 Induces Immunogenic Cell Death and Enhances T-DM1 Response in HER2-Positive Breast Cancer. Cancer Research 2024, 84 (9) , 1475-1490. https://doi.org/10.1158/0008-5472.CAN-23-2812
    78. Waleed H. Almalki. An Up-to-date Review on Protein-based Nanocarriers in the Management of Cancer. Current Drug Delivery 2024, 21 (4) , 509-524. https://doi.org/10.2174/1567201820666230509101020
    79. Chia-Hsien Shih, Yu-Hua Lin, Hao-Lun Luo, Wen-Wei Sung. Antibody-drug conjugates targeting HER2 for the treatment of urothelial carcinoma: potential therapies for HER2-positive urothelial carcinoma. Frontiers in Pharmacology 2024, 15 https://doi.org/10.3389/fphar.2024.1326296
    80. Meng Zhang, Yuanye Zuo, Siyi Chen, Yaonan Li, Yang Xing, Lei Yang, Hong Wang, Rui Guo. Antibody–drug conjugates in urothelial carcinoma: scientometric analysis and clinical trials analysis. Frontiers in Oncology 2024, 14 https://doi.org/10.3389/fonc.2024.1323366
    81. Giuseppe Saltalamacchia, Rosalba Torrisi, Rita De Sanctis, Giovanna Masci, Chiara Miggiano, Mariangela Gaudio, Chiara Benvenuti, Flavia Jacobs, Riccardo Gerosa, Armando Santoro, Alberto Zambelli. Charting the Course in Sequencing Antibody-Drug Conjugates in Breast Cancer. Biomedicines 2024, 12 (3) , 500. https://doi.org/10.3390/biomedicines12030500
    82. Conner Hartupee, Bolni Marius Nagalo, Chiswili Y. Chabu, Mulu Z. Tesfay, Joycelynn Coleman-Barnett, John T. West, Omeed Moaven. Pancreatic cancer tumor microenvironment is a major therapeutic barrier and target. Frontiers in Immunology 2024, 15 https://doi.org/10.3389/fimmu.2024.1287459
    83. Afruja Ahad, Fatima Aftab, Alexa Michel, Jason S. Lewis, Maria Contel. Development of immunoliposomes containing cytotoxic gold payloads against HER2-positive breast cancers. RSC Medicinal Chemistry 2024, 15 (1) , 139-150. https://doi.org/10.1039/D3MD00334E
    84. Liteng Shen, Xiuna Sun, Zhen Chen, Yu Guo, Zheyuan Shen, Yi Song, Wenxiu Xin, Haiying Ding, Xinyue Ma, Weiben Xu, Wanying Zhou, Jinxin Che, Lili Tan, Liangsheng Chen, Siqi Chen, Xiaowu Dong, Luo Fang, Feng Zhu. ADCdb: the database of antibody–drug conjugates. Nucleic Acids Research 2024, 52 (D1) , D1097-D1109. https://doi.org/10.1093/nar/gkad831
    85. Shivangi Kumari, Sonam Raj, M. Arockia Babu, Gurjit Kaur Bhatti, Jasvinder Singh Bhatti. Antibody-drug conjugates in cancer therapy: innovations, challenges, and future directions. Archives of Pharmacal Research 2024, 47 (1) , 40-65. https://doi.org/10.1007/s12272-023-01479-6
    86. Anindita De, Sonam Patel, K. Gowthamarajan. Next-Generation Therapies for Breast Cancer. 2024, 119-145. https://doi.org/10.1007/978-981-99-6597-7_5
    87. Sagar, Malemnganba Takhellambam, Aditi Rattan, Vijay Kumar Prajapati. Unleashing the power of antibodies: Engineering for tomorrow’s therapy. 2024, 1-36. https://doi.org/10.1016/bs.apcsb.2023.12.009
    88. Seema Yadav, Narahari Narayan Palei, Subas Chandra Dinda, Arghya Kusum Dhar. Drug Delivery in Biotechnology: Present and Future. 2024, 103-138. https://doi.org/10.1007/978-981-97-1148-2_7
    89. Mohsin Khan. Nanoparticle-Mediated Cancer Chemotherapy. 2024, 183-216. https://doi.org/10.1007/978-981-97-3545-7_9
    90. Lie Chen, Cui-Cui Liu, Si-Yuan Zhu, Jing-Yu Ge, Yu-Fei Chen, Ding Ma, Zhi-Ming Shao, Ke-Da Yu. Multiomics of HER2-low triple-negative breast cancer identifies a receptor tyrosine kinase–relevant subgroup with therapeutic prospects. JCI Insight 2023, 8 (22) https://doi.org/10.1172/jci.insight.172366
    91. Sandra Kannampuzha, Reshma Murali, Abilash Valsala Gopalakrishnan, Anirban Goutam Mukherjee, Uddesh Ramesh Wanjari, Arunraj Namachivayam, Alex George, Abhijit Dey, Balachandar Vellingiri. Novel biomolecules in targeted cancer therapy: a new approach towards precision medicine. Medical Oncology 2023, 40 (11) https://doi.org/10.1007/s12032-023-02168-6
    92. Zhi Zhou, Yuang Gu, Liexin Wu, Yan Wang, Huiying Xu, Lei Ma, Zhaoyong Zhang, Jincun Zhao, Wei Zhang, Wei Peng, Guang Yang, Xiyong Yu, Hongtao Xu, Wei Yi. Organo-Se BTSAs-enabled performance: From racemic and asymmetric synthesis to click chemistry application. Chem 2023, 9 (11) , 3335-3346. https://doi.org/10.1016/j.chempr.2023.07.022
    93. Zhilin Liu, Honglei Zhang, Jiali Sun, Mengfei Zheng, Linjie Cui, Yu Zhang, Jianjun Cheng, Zhaohui Tang, Xuesi Chen. Organic‐Solvent‐Free “Lego‐Like” Modular Preparation of Fab‐Nondestructive Antibody‐Drug Conjugates with Ultrahigh Drug‐to‐Antibody Ratio. Advanced Materials 2023, 35 (46) https://doi.org/10.1002/adma.202300377
    94. Funda Meric-Bernstam, Emiliano Calvo, Keun Seok Lee, Victor Moreno, Yeon Hee Park, Sun Young Rha, Pavani Chalasani, Wei Zhong, Li Zhou, Steven Pirie-Shepherd, Abraham C.F. Leung, Giuseppe Curigliano. Safety and Tolerability of a Novel Anti-HER2 Antibody–Drug Conjugate (PF-06804103) in Patients with HER2-Expressing Solid Tumors: A Phase 1 Dose-Escalation Study. Molecular Cancer Therapeutics 2023, 22 (10) , 1191-1203. https://doi.org/10.1158/1535-7163.MCT-23-0101
    95. Honglei Zhang, Jiali Sun, Yu Zhang, Zhenqian Zhang, Xiaoshuang Wang, Zhilin Liu, Xuefei Zhang, Zhaohui Tang, Xuesi Chen. Preparation of an Ultrahigh-DAR PDL1 monoclonal antibody-polymeric-SN38 conjugate for precise colon cancer therapy. Biomaterials 2023, 301 , 122285. https://doi.org/10.1016/j.biomaterials.2023.122285
    96. Ilana Schlam, Ruth Moges, Stefania Morganti, Sara M. Tolaney, Paolo Tarantino. Next-generation antibody-drug conjugates for breast cancer: Moving beyond HER2 and TROP2. Critical Reviews in Oncology/Hematology 2023, 190 , 104090. https://doi.org/10.1016/j.critrevonc.2023.104090
    97. Federico Riccardi, Michele Dal Bo, Paolo Macor, Giuseppe Toffoli. A comprehensive overview on antibody-drug conjugates: from the conceptualization to cancer therapy. Frontiers in Pharmacology 2023, 14 https://doi.org/10.3389/fphar.2023.1274088
    98. Bharat Goel, Shreyans K. Jain. Natural products as a source of cytotoxic warheads in antibody-drug conjugates. Natural Product Research 2023, 37 (17) , 2973-2985. https://doi.org/10.1080/14786419.2022.2138872
    99. Devesh Aggarwal, Jie Yang, Md. Abdus Salam, Sagnik Sengupta, Md. Yusuf Al-Amin, Saad Mustafa, Mohammad Aasif Khan, Xun Huang, Jogendra Singh Pawar. Antibody-drug conjugates: the paradigm shifts in the targeted cancer therapy. Frontiers in Immunology 2023, 14 https://doi.org/10.3389/fimmu.2023.1203073
    100. Fengjiao Yao, Yacong An, Xialian Lai, Xundou Li, Zhen Yu, Xian-Da Yang. Novel nanotherapeutics for cancer immunotherapy by CTLA-4 aptamer-functionalized albumin nanoparticle loaded with antihistamine. Journal of Cancer Research and Clinical Oncology 2023, 149 (10) , 7515-7527. https://doi.org/10.1007/s00432-023-04698-y
    Load more citations
    Open PDF

    Journal of Medicinal Chemistry

    Cite this: J. Med. Chem. 2014, 57, 16, 6949–6964
    Click to copy citationCitation copied!
    https://doi.org/10.1021/jm500766w
    Published June 26, 2014

    Copyright © 2014 American Chemical Society. This publication is available under these Terms of Use.

    Article Views

    35k

    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. Comparison of the in vitro potency of cytotoxic drugs toward the Burkitt lymphoma cell line Namalwa: maytansine (blue circle), vinblastine (red triangle), daunorubicin (green diamond), methotrexate (brown square), mitomycin C (purple triangle).

      Scheme 1

      Scheme 1. Synthesis of Thiol-Containing Maytansinoidsa

      Scheme aReaction conditions: (a) LiAlH(OMe)3/THF, −40 °C, (b) EDC/ZnCl2/CH2Cl2, rt, (c) dithiothreitol, rt.

      Figure 2

      Figure 2. Structures of cross-linkers used in conjugate preparation.

      Scheme 2

      Scheme 2. Representative Conjugation Processes for Trastuzumab-Maytansinoid Conjugates

      Figure 3

      Figure 3. Structural representation of trastuzumab-maytansinoid conjugates with a disulfide linker (8ad) or a thioether linker (8e). Adapted by permission from the American Association for Cancer Research (Lewis Phillips. G. D.; Li, G.; Dugger, D. L.; Crocker, L. M.; Parsons, K. L.; Mai, E.; Lambert, J. M.; Chari, R. V.; Lutz, R. J.; Wong, W. L.; Jacobson, F. S.; Koeppen, H.; Schwall, R. H.; Kenkare-Mitra, S. R.; Spencer, S. D.; Sliwkowski, M. X.Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 2008, 68, 9280–9290). (20)

      Figure 4

      Figure 4. Comparison of the in vivo efficacy of trastuzumab–maytansinoid conjugates with a disulfide linker (8b (green triangle), 8c (purple cross), 8d (red boxed cross)) or a thioether linker (8e (blue square)) at a single iv dose of 10 mg/kg conjugate and a vehicle control (×). Adapted by permission from the American Association for Cancer Research (Lewis Phillips. G. D.; Li, G.; Dugger, D. L.; Crocker, L. M.; Parsons, K. L.; Mai, E.; Lambert, J. M.; Chari, R. V.; Lutz, R. J.; Wong, W. L.; Jacobson, F. S.; Koeppen, H.; Schwall, R. H.; Kenkare-Mitra, S. R.; Spencer, S. D.; Sliwkowski, M. X.Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 2008, 68, 9280–9290). (20)

      Figure 5

      Figure 5. (A) In vitro catabolism by BT474-EEI cells of the trastuzumab-MCC-DM1 conjugate 8e (thioether linker): top panel, 6 h time point; bottom panel, 24 h time point. (B) In vitro catabolism by BT474-EEI cells of the trastuzumab-SPP-DM1 conjugate 8b (disulfide linker): top panel, 6 h time point; bottom panel, 24 h time point. (C) Comparison of the rate and extent of in vitro processing of trastuzumab-SPP-DM1 conjugate 8b (disulfide linker: blue squares) and trastuzumab-MCC-DM1 conjugate 8e (thioether linker, red squares) by BT474-EEI cells.

      Figure 6

      Figure 6. (A) Time dependent formation of catabolite: comparison of in vivo catabolism of trastuzumab–maytansinoid conjugates in BT474-EEI tumor xenografts in mice: trastuzumab-SPP-DM1 conjugate 8b (disulfide linker, blue square) and trastuzumab-MCC-DM1 conjugate 8e (thioether linker, red square). (B) In vivo catabolism of trastuzumab–maytansinoid conjugates in BT474-EEI tumor xenografts in mice at 2-day time-point: top panel, 8b, disulfide linker (SPP); bottom panel, 8e, thioether linker (SMCC).

      Figure 7

      Figure 7. (A) Comparison of the in vivo antitumor activity of trastuzumab (red circle, 15 mg/kg × 4) and T-DM1 (blue square, 15 mg/kg, single dose), vehicle control (×), in the KPL-4 model. (Excerpted from Lewis-Phillips et al. (20)). (B) Dose-dependent antitumor activity of T-DM1 in the BT474-EEI model (dosing q3w × 3). Doses of T-DM1, are the following: 0.3 mg/kg (purple-outline triangle), 1 mg/kg (green triangle), 3 mg/kg (blue-outline triangle), 10 mg/kg (red triangle), 15 mg/kg (blue square), vehicle control (×); unconjugated trastuzumab (red circle, 15 mg/kg). Parts A and B are adapted by permission from the American Association for Cancer Research (Lewis Phillips. G. D.; Li, G.; Dugger, D. L.; Crocker, L. M.; Parsons, K. L.; Mai, E.; Lambert, J. M.; Chari, R. V.; Lutz, R. J.; Wong, W. L.; Jacobson, F. S.; Koeppen, H.; Schwall, R. H.; Kenkare-Mitra, S. R.; Spencer, S. D.; Sliwkowski, M. X.Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 2008, 68, 9280–9290). (20)

    • References


      This article references 50 other publications.

      1. 1
        Colomer, R.; Shamon, L. A.; Tsai, M. S.; Lupu, R. Herceptin: from the bench to the clinic Cancer Invest. 2001, 19, 49 56
      2. 2
        Pegram, M. D.; Konecny, G. E.; O’Callaghan, C.; Beryt, M.; Pietras, R.; Slamon, D. J. Rational combinations of trastuzumab with chemotherapeutic drugs used in the treatment of breast cancer J. Natl. Cancer Inst. 2004, 96, 739 749
      3. 3
        Shepard, H. M.; Lewis, G. D.; Sarup, J. C.; Fendly, B. M.; Maneval, D.; Mordenti, J.; Figari, I.; Kotts, C. E.; Palladino, M. A., Jr.; Ullrich, A. Monoclonal antibody therapy of human cancer: taking the HER2 protooncogene to the clinic J. Clin. Immunol. 1991, 11, 117 127
      4. 4
        Spector, N. L.; Blackwell, K. L. Understanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancer J. Clin. Oncol. 2009, 27, 5838 5847
      5. 5
        Slamon, D. J.; Leyland-Jones, B.; Shak, S.; Fuchs, H.; Paton, V.; Bajamonde, A.; Fleming, T.; Eiermann, W.; Wolter, J.; Pegram, M.; Baselga, J.; Norton, L. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2 N. Engl. J. Med. 2001, 344, 783 792
      6. 6
        Nahta, R.; Esteva, F. J. HER-2-targeted therapy: lessons learned and future directions Clin. Cancer Res. 2003, 9, 5078 5084
      7. 7
        Metzger-Filho, O.; Winer, E. P.; Krop, I. Pertuzumab: optimizing HER2 blockade Clin. Cancer Res. 2013, 19, 5552 5556
      8. 8
        Chari, R. V.; Miller, M. L.; Widdison, W. C. Antibody–drug conjugates: an emerging concept in cancer therapy Angew. Chem., Int. Ed. 2014, 53, 3796 3827
      9. 9
        Blattler, W. A.; Chari, R. V. J. Methods of treatment using anti-ERBB antibody–maytansinoid conjugates. U.S. Patent 8,337,856, 2012.
      10. 10
        Sedlacek, H. H. Antibodies as Carriers of Cytotoxicity; Karger: Basel, Switzerland, 1992; pp 1 145.
      11. 11
        Chari Ravi, V. J. Targeted cancer therapy: conferring specificity to cytotoxic drugs Acc. Chem. Res. 2008, 41, 98 107
      12. 12
        Kupchan, S. M.; Komoda, Y.; Branfman, A. R.; Sneden, A. T.; Court, W. A.; Thomas, G. J.; Hintz, H. P.; Smith, R. M.; Karim, A.; Howie, G. A.; Verma, A. K.; Nagao, Y.; Dailey, R. G., Jr.; Zimmerly, V. A.; Sumner, W. C., Jr. The maytansinoids. Isolation, structural elucidation, and chemical interrelation of novel ansa macrolides J. Org. Chem. 1977, 42, 2349 2357
      13. 13
        Widdison, W. C.; Wilhelm, S. D.; Cavanagh, E. E.; Whiteman, K. R.; Leece, B. A.; Kovtun, Y.; Goldmacher, V. S.; Xie, H.; Steeves, R. M.; Lutz, R. J.; Zhao, R.; Wang, L.; Blattler, W. A.; Chari, R. V. Semisynthetic maytansine analogues for the targeted treatment of cancer J. Med. Chem. 2006, 49, 4392 4408
      14. 14
        Issell, B. F.; Crooke, S. T. Maytansine Cancer Treat. Rev. 1978, 5, 199 207
      15. 15
        Kupchan, S. M.; Sneden, A. T.; Branfman, A. R.; Howie, G. A.; Rebhun, L. I.; McIvor, W. E.; Wang, R. W.; Schnaitman, T. C. Structural requirements for antileukemic activity among the naturally occurring and semisynthetic maytansinoids J. Med. Chem. 1978, 21, 31 37
      16. 16
        Chari, R. V.; Martell, B. A.; Gross, J. L.; Cook, S. B.; Shah, S. A.; Blattler, W. A.; McKenzie, S. J.; Goldmacher, V. S. Immunoconjugates containing novel maytansinoids: promising anticancer drugs Cancer Res. 1992, 52, 127 131
      17. 17
        Kellogg, B. A.; Garrett, L.; Kovtun, Y.; Lai, K. C.; Leece, B.; Miller, M.; Payne, G.; Steeves, R.; Whiteman, K. R.; Widdison, W.; Xie, H.; Singh, R.; Chari, R. V.; Lambert, J. M.; Lutz, R. J. Disulfide-linked antibody-maytansinoid conjugates: optimization of in vivo activity by varying the steric hindrance at carbon atoms adjacent to the disulfide linkage Bioconjugate Chem. 2011, 22, 717 727
      18. 18
        Wakankar, A.; Chen, Y.; Gokarn, Y.; Jacobson, F. S. Analytical methods for physicochemical characterization of antibody drug conjugates mAbs 2011, 3, 161 172
      19. 19
        Wakankar, A. A.; Feeney, M. B.; Rivera, J.; Chen, Y.; Kim, M.; Sharma, V. K.; Wang, Y. J. Physicochemical stability of the antibody–drug conjugate trastuzumab-DM1: changes due to modification and conjugation processes Bioconjugate Chem. 2010, 21, 1588 1595
      20. 20
        Lewis Phillips, G. D.; Li, G.; Dugger, D. L.; Crocker, L. M.; Parsons, K. L.; Mai, E.; Blattler, W. A.; Lambert, J. M.; Chari, R. V.; Lutz, R. J.; Wong, W. L.; Jacobson, F. S.; Koeppen, H.; Schwall, R. H.; Kenkare-Mitra, S. R.; Spencer, S. D.; Sliwkowski, M. X. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate Cancer Res. 2008, 68, 9280 9290
      21. 21
        Erickson, H. K.; Lewis Phillips, G. D.; Leipold, D. D.; Provenzano, C. A.; Mai, E.; Johnson, H. A.; Gunter, B.; Audette, C. A.; Gupta, M.; Pinkas, J.; Tibbitts, J. The effect of different linkers on target cell catabolism and pharmacokinetics/pharmacodynamics of trastuzumab maytansinoid conjugates Mol. Cancer Ther. 2012, 11, 1133 1142
      22. 22
        Junttila, T. T.; Li, G.; Parsons, K.; Phillips, G. L.; Sliwkowski, M. X. Trastuzumab-DM1 (T-DM1) retains all the mechanisms of action of trastuzumab and efficiently inhibits growth of lapatinib insensitive breast cancer Breast Cancer Res. Treat. 2011, 128, 347 356
      23. 23
        Barok, M.; Tanner, M.; Koninki, K.; Isola, J. Trastuzumab-DM1 is highly effective in preclinical models of HER2-positive gastric cancer Cancer Lett. 2011, 306, 171 179
      24. 24
        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.; Lai, K. C.; Widdison, W. C.; Kellogg, B.; Johnson, H.; Pinkas, J.; Lutz, R. J.; Singh, R.; Goldmacher, V. S.; Chari, R. V. Antibody–maytansinoid conjugates designed to bypass multidrug resistance Cancer Res. 2010, 70, 2528 2537
      25. 25
        Guo, J.; Li, G.; Lewis Phillips, G. D. Role of multidrug resistance transporters in the biological response to trastuzumab–cytotoxic drug conjugates Cancer Res. 2010, 70 (Suppl. 1) Abstract 618
      26. 26
        Jumbe, N. L.; Xin, Y.; Leipold, D. D.; Crocker, L.; Dugger, D.; Mai, E.; Sliwkowski, M. X.; Fielder, P. J.; Tibbitts, J. Modeling the efficacy of trastuzumab–DM1, an antibody drug conjugate, in mice J. Pharmacokinet. Pharmacodyn. 2010, 37, 221 242
      27. 27
        Kaur, S.; Xu, K.; Saad, O. M.; Dere, R. C.; Carrasco-Triguero, M. Bioanalytical assay strategies for the development of antibody–drug conjugate biotherapeutics Bioanalysis 2013, 5, 201 226
      28. 28
        Poon, K. A.; Flagella, K.; Beyer, J.; Tibbitts, J.; Kaur, S.; Saad, O.; Yi, J. H.; Girish, S.; Dybdal, N.; Reynolds, T. Preclinical safety profile of trastuzumab emtansine (T-DM1): mechanism of action of its cytotoxic component retained with improved tolerability Toxicol. Appl. Pharmacol. 2013, 273, 298 313
      29. 29
        Sun, X.; Widdison, W.; Mayo, M.; Wilhelm, S.; Leece, B.; Chari, R.; Singh, R.; Erickson, H. Design of antibody–maytansinoid conjugates allows for efficient detoxification via liver metabolism Bioconjugate Chem. 2011, 22, 728 735
      30. 30
        Fishkin, N.; Maloney, E. K.; Chari, R. V.; Singh, R. A novel pathway for maytansinoid release from thioether linked antibody–drug conjugates (ADCs) under oxidative conditions Chem. Commun. 2011, 47, 10752 10754
      31. 31
        Krop, I. E.; Beeram, M.; Modi, S.; Jones, S. F.; Holden, S. N.; Yu, W.; Girish, S.; Tibbitts, J.; Yi, J. H.; Sliwkowski, M. X.; Jacobson, F.; Lutzker, S. G.; Burris, H. A. Phase I study of trastuzumab–DM1, an HER2 antibody–drug conjugate, given every 3 weeks to patients with HER2-positive metastatic breast cancer J. Clin. Oncol. 2010, 28, 2698 2704
      32. 32
        Baselga, J. Phase I and II clinical trials of trastuzumab Ann. Oncol. 2001, 12 (Suppl. 1) S49 S55
      33. 33
        Beeram, M.; Krop, I. E.; Burris, H. A.; Girish, S. R.; Yu, W.; Lu, M. W.; Holden, S. N.; Modi, S. A phase 1 study of weekly dosing of trastuzumab emtansine (T-DM1) in patients with advanced human epidermal growth factor 2-positive breast cancer Cancer 2012, 118, 5733 5740
      34. 34
        Burris, H. A., 3rd; Rugo, H. S.; Vukelja, S. J.; Vogel, C. L.; Borson, R. A.; Limentani, S.; Tan-Chiu, E.; Krop, I. E.; Michaelson, R. A.; Girish, S.; Amler, L.; Zheng, M.; Chu, Y. W.; Klencke, B.; O’Shaughnessy, J. A. Phase II study of the antibody drug conjugate trastuzumab–DM1 for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer after prior HER2-directed therapy J. Clin. Oncol. 2011, 29, 398 405
      35. 35
        Krop, I. E.; LoRusso, P.; Miller, K. D.; Modi, S.; Yardley, D.; Rodriguez, G.; Guardino, E.; Lu, M.; Zheng, M.; Girish, S.; Amler, L.; Winer, E. P.; Rugo, H. S. A phase II study of trastuzumab emtansine in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer who were previously treated with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine J. Clin. Oncol. 2012, 30, 3234 3241
      36. 36
        Hurvitz, S. A.; Dirix, L.; Kocsis, J.; Bianchi, G. V.; Lu, J.; Vinholes, J.; Guardino, E.; Song, C.; Tong, B.; Ng, V.; Chu, Y. W.; Perez, E. A. Phase II randomized study of trastuzumab emtansine versus trastuzumab plus docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer J. Clin. Oncol. 2013, 31, 1157 1163
      37. 37
        Suter, T. M.; Procter, M.; van Veldhuisen, D. J.; Muscholl, M.; Bergh, J.; Carlomagno, C.; Perren, T.; Passalacqua, R.; Bighin, C.; Klijn, J. G.; Ageev, F. T.; Hitre, E.; Groetz, J.; Iwata, H.; Knap, M.; Gnant, M.; Muehlbauer, S.; Spence, A.; Gelber, R. D.; Piccart-Gebhart, M. J. Trastuzumab-associated cardiac adverse effects in the herceptin adjuvant trial J. Clin. Oncol. 2007, 25, 3859 3865
      38. 38
        Verma, S.; Miles, D.; Gianni, L.; Krop, I. E.; Welslau, M.; Baselga, J.; Pegram, M.; Oh, D. Y.; Dieras, V.; Guardino, E.; Fang, L.; Lu, M. W.; Olsen, S.; Blackwell, K. Trastuzumab emtansine for HER2-positive advanced breast cancer N. Engl. J. Med. 2012, 367, 1783 1791
      39. 39
        Krop, I. E.; Kim, S. B.; Gonzalez-Martin, A.; LoRusso, P. M.; Ferrero, J. M.; Smitt, M.; Yu, R.; Leung, A.; Wildiers, H. Trastuzumab emtansine versus treatment of physician’s choice for pretreated HER2-positive advanced breast cancer (TH3RESA): a randomized open-label phase 3 trial Lancet Oncol. 2014, 15, 689 699
      40. 40
        Baselga, J.; Cortes, J.; Kim, S. B.; Im, S. A.; Hegg, R.; Im, Y. H.; Roman, L.; Pedrini, J. L.; Pienkowski, T.; Knott, A.; Clark, E.; Benyunes, M. C.; Ross, G.; Swain, S. M. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer N. Engl. J. Med. 2012, 366, 109 119
      41. 41
        Phillips, G. D.; Fields, C. T.; Li, G.; Dowbenko, D.; Schaefer, G.; Miller, K.; Andre, F.; Burris, H. A., 3rd; Albain, K. S.; Harbeck, N.; Dieras, V.; Crivellari, D.; Fang, L.; Guardino, E.; Olsen, S. R.; Crocker, L. M.; Sliwkowski, M. X. Dual targeting of HER2-positive cancer with trastuzumab emtansine and pertuzumab: critical role for neuregulin blockade in antitumor response to combination therapy Clin. Cancer Res. 2014, 20, 456 468
      42. 42
        Lu, D.; Burris, H. A., 3rd; Wang, B.; Dees, E. C.; Cortes, J.; Joshi, A.; Gupta, M.; Yi, J. H.; Chu, Y. W.; Shih, T.; Fang, L.; Girish, S. Drug interaction potential of trastuzumab emtansine (T-DM1) combined with pertuzumab in patients with HER2-positive metastatic breast cancer Curr. Drug Metab. 2012, 13, 911 922
      43. 43
        Bartsch, R.; Berghoff, A. S.; Preusser, M. Breast cancer brain metastases responding to primary systemic therapy with T-DM1 J. Neuro-Oncol. 2014, 116, 205 206
      44. 44
        Girish, S.; Gupta, M.; Wang, B.; Lu, D.; Krop, I. E.; Vogel, C. L.; Burris, H. A., III; LoRusso, P. M.; Yi, J. H.; Saad, O.; Tong, B.; Chu, Y. W.; Holden, S.; Joshi, A. Clinical pharmacology of trastuzumab emtansine (T-DM1): an antibody–drug conjugate in development for the treatment of HER2-positive cancer Cancer Chemother. Pharmacol. 2012, 69, 1229 1240
      45. 45
        Lu, D.; Joshi, A.; Wang, B.; Olsen, S.; Yi, J. H.; Krop, I. E.; Burris, H. A.; Girish, S. An integrated multiple-analyte pharmacokinetic model to characterize trastuzumab emtansine (T-DM1) clearance pathways and to evaluate reduced pharmacokinetic sampling in patients with HER2-positive metastatic breast cancer Clin. Pharmacokinet. 2013, 52, 657 672
      46. 46
        Chudasama, V. L.; Schaedeli Stark, F.; Harrold, J. M.; Tibbitts, J.; Girish, S. R.; Gupta, M.; Frey, N.; Mager, D. E. Semi-mechanistic population pharmacokinetic model of multivalent trastuzumab emtansine in patients with metastatic breast cancer Clin. Pharmacol. Ther. 2012, 92, 520 527
      47. 47
        Bender, B. C.; Schaedeli-Stark, F.; Koch, R.; Joshi, A.; Chu, Y. W.; Rugo, H.; Krop, I. E.; Girish, S.; Friberg, L. E.; Gupta, M. A population pharmacokinetic/pharmacodynamic model of thrombocytopenia characterizing the effect of trastuzumab emtansine (T-DM1) on platelet counts in patients with HER2-positive metastatic breast cancer Cancer Chemother. Pharmacol. 2012, 70, 591 601
      48. 48
        Shen, B. Q.; Bumbaca, D.; Saad, O.; Yue, Q.; Pastuskovas, C. V.; Khojasteh, S. C.; Tibbitts, J.; Kaur, S.; Wang, B.; Chu, Y. W.; LoRusso, P. M.; Girish, S. Catabolic fate and pharmacokinetic characterization of trastuzumab emtansine (T-DM1): an emphasis on preclinical and clinical catabolism Curr. Drug Metab. 2012, 13, 901 910
      49. 49
        Thon, J. N.; Devine, M. T.; Begonja, A. J.; Tibbitts, J.; Italiano, J. E., Jr. High-content live-cell imaging assay used to establish mechanism of trastuzumab emtansine (T-DM1)-mediated inhibition of platelet production Blood 2012, 120, 1975 1984
      50. 50
        Sendur, M. A.; Aksoy, S.; Altundag, K. Cardiotoxicity of novel HER2-targeted therapies Curr. Med. Res. Opin. 2013, 29, 1015 1024