Ado-trastuzumab Emtansine (T-DM1): An Antibody–Drug Conjugate (ADC) for HER2-Positive Breast CancerClick to copy article linkArticle link copied!
Abstract
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.
Introduction
Design of T-DM1
(a) Selection of the Cytotoxic Agent
(b) Design and Synthesis of Maytansinoids for Linkage to Antibodies
IC50, nM | ||
---|---|---|
maytansinoid | KB cells | SK-Br-3 cells |
1 (maytansine) | 0.034 | 0.030 |
4a (DM0-SMe) | 0.190 | 0.180 |
4b (DM1-SMe) | 0.029 | 0.014 |
4c (DM2′-SMe) | 0.009 | 0.038 |
4d (DM3-SMe) | 0.011 | 0.004 |
4e (DM4-SMe) | 0.001 | 0.003 |
(c) Selection of the Linker
(d) Conjugation Methods
Preclinical Evaluation Leading to the Selection of Maytansinoid and Linker Component of T-DM1
(a) In Vitro Studies
(b) In Vivo Pharmacokinetic Studies
(c) In Vivo Antitumor Activity
Metabolism Studies
(a) In Vitro Studies
(b) In Vivo Studies
In Vivo Tolerability Studies
Biological Activity of T-DM1
(a) In Vitro Cytotoxicity
IC50, μg/mL | |||
---|---|---|---|
cell line | origin | trastuzumab-MCC-DM1 | trastuzumab |
SK-Br-3 | human breast tumor | 0.011 | >10 |
BT-474-EEI | human breast tumor | 0.004 | >10 |
HCC1954 | human breast tumor | 0.015 | >10 |
KPL-4 | human breast tumor | 0.011 | >10 |
Calu-3 | human lung carcinoma | 0.062 | >10 |
MKN-7 | human gastric carcinoma | 0.266 | >10 |
SK-OV-3 | human ovarian cancer | 0.009 | >10 |
HMEC | human mammary epithelial cells | 4.0 | >10 |
NHEK | normal human epidermal keratinocytes | 10.0 | >10 |
(b) In Vivo Evaluation
(c) Functional Activity
Preclinical Toxicology and Toxicokinetics of T-DM1
Clinical Evaluation of T-DM1
(a) Phase I Studies
(b) Phase II Clinical Trials of T-DM1 as a Single Agent
(c) Phase III Clinical Trials of T-DM1
Efficacy | T-DM1 | lapatinib + capecitabine |
---|---|---|
no. of patients evaluable | n = 397 | n = 389 |
objective response rate | 43.6% | 30.8% |
complete response | 1% (n = 4) | 0.5% (n = 2) |
partial response | 42.6% (n = 169) | 30.3% (n = 118) |
median duration of response | 12.6 months | 6.5 months |
estimated 1-year survival | 85.2% | 78.4% |
estimated 2-year survival | 64.7% | 51.8% |
median progression-free survival | 9.6 months | 6.4 months |
median overall survival | 30.9 months | 25.1 months |
Safety | T-DM1 | lapatinib + capecitabine | ||
---|---|---|---|---|
no. of subjects evaluable | n = 490 | n = 488 | ||
adverse events (≥15% of patients) | any grade | grade ≥3 | any grade | grade ≥3 |
any adverse event | 95.9% | 40.8% | 97.7% | 57.0% |
diarrhea | 23.3% | 1.6% | 79.7% | 20.7% |
hand–foot syndrome | 1.2% | 0 | 58.0% | 16.4% |
vomiting | 19.0% | 0.8% | 29.3% | 4.5% |
fatigue | 35.1% | 2.4% | 27.9% | 3.5% |
nausea | 39.2% | 0.8% | 44.7% | 2.5% |
mucosal inflammation | 6.7% | 0.2% | 19.1% | 2.3% |
thrombocytopenia | 28.0% | 12.9% | 2.5% | 0.2% |
elevated serum ALT | 16.9% | 2.9% | 8.8% | 1.4% |
elevated serum AST | 22.4% | 4.3% | 9.4% | 0.8% |
Clinical Pharmacokinetics and Metabolism of T-DM1
Safety and Tolerability Profile of T-DM1
Conclusion
Acknowledgment
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.
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
This article references 50 other publications.
- 1Colomer, R.; Shamon, L. A.; Tsai, M. S.; Lupu, R. Herceptin: from the bench to the clinic Cancer Invest. 2001, 19, 49– 56Google ScholarThere is no corresponding record for this reference.
- 2Pegram, 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– 749Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXkt1Sqsbo%253D&md5=1fb831bd3fbb62277de025c4a99f383dRational Combinations of Trastuzumab With Chemotherapeutic Drugs Used in the Treatment of Breast CancerPegram, Mark D.; Konecny, Gottfried E.; O'Callaghan, Carminda; Beryt, Malgorzata; Pietras, Richard; Slamon, Dennis J.Journal of the National Cancer Institute (2004), 96 (10), 739-749CODEN: JNCIEQ; ISSN:0027-8874. (Oxford University Press)Trastuzumab, a humanized anti-HER2 antibody, increases the clin. benefit of first-line chemotherapy in patients with metastatic breast cancers that overexpress HER2. We characterized interactions between trastuzumab and chemotherapeutic agents commonly used in the treatment of breast cancer. Multiple drug effect/combination index isobologram anal. was used to study the efficacy of chemotherapeutic drug plus trastuzumab combinations tested against four HER2-overexpressing breast cancer cell lines (SK-BR-3, BT-474, MDA-MB-361, and MDA-MB-453). Combination index values were derived from parameters of the median effect plots, and statistical tests were used to det. whether the mean combination index values at multiple effect levels were statistically significantly different from a combination index value of 1.0. Values less than 1.0 indicate synergistic interactions, values greater than 1.0 indicate antagonistic interactions, and values equal to 1.0 indicate additive interactions. At a wide range of clin. achievable drug concns., synergistic interactions were obsd. in all four breast cancer cell lines for trastuzumab plus carboplatin (mean combination index values ranged from 0.32 [P<.001] to 0.53 [P<.001]), 4-hydroxycyclophosphamide (mean combination index values ranged from 0.38 [P<.001] to 0.73 [P = .010]), docetaxel (mean combination index values ranged from 0.30 [P<.001] to 0.62 [P<.001]), and vinorelbine (mean combination index values ranged from 0.24 [P<.001] to 0.78 [P<.034]). Additive interactions were obsd. in all four cell lines with trastuzumab plus doxorubicin, epirubicin, and paclitaxel. Interactions between trastuzumab and gemcitabine were synergistic at low concns. of gemcitabine and antagonistic at high concns. A synergistic interaction was obsd. with a three-drug combination of docetaxel plus carboplatin plus trastuzumab in SK-BR-3 cells (mean combination index value = 0.09; P<.001). Consistent synergistic interactions of trastuzumab plus carboplatin, 4-hydroxycyclophosphamide, docetaxel, or vinorelbine across a wide range of clin. relevant concns. in HER2-overexpressing breast cancer cells indicate that these are rational combinations to test in human clin. trials.
- 3Shepard, 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– 127Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXmsFent7w%253D&md5=8c323a0c6d081fc00a2b3328a8711f75Monoclonal antibody therapy of human cancer: taking the HER2 protooncogene to the clinicShepard, H. Michael; Lewis, Gail D.; Sarup, Jay C.; Fendly, Brian M.; Maneval, Daniel; Mordenti, Joyce; Figari, Irene; Kotts, Claire E.; Palladino, Michael A., Jr.; et al.Journal of Clinical Immunology (1991), 11 (3), 117-27CODEN: JCIMDO; ISSN:0271-9142.A review, with 35 refs., on: the HER2 protoncogene and human cancer; derivation of muMAb 4D5; in vivo preclin. efficacy; and mechanism of action.
- 4Spector, 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– 5847Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtVWitro%253D&md5=d54f228c3b69c973238fb1279ee8572dUnderstanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancerSpector, Neil L.; Blackwell, Kimberly L.Journal of Clinical Oncology (2009), 27 (34), 5838-5847CODEN: JCONDN; ISSN:0732-183X. (American Society of Clinical Oncology)A review. Purpose: Targeted therapy with the humanized monoclonal antibody trastuzumab has become a mainstay for human epidermal growth factor receptor 2 (HER2) -pos. breast cancer (BC). The mechanisms of action of trastuzumab have not been fully elucidated, and data available to date are reviewed here. The impact of the mechanisms of action on clin. benefit also is discussed. Methods: An extensive literature review of trastuzumab and proposed mechanisms of action was performed. Results: At least five potential extracellular and intracellular antitumor mechanisms of trastuzumab have been identified in the preclin. setting. These include activation of antibody-dependent cellular cytotoxicity, inhibition of extracellular domain cleavage, abrogation of intracellular signaling, redn. of angiogenesis, and decreased DNA repair. These effects lead to tumor cell stasis and/or death. Clin. benefit from trastuzumab-based therapy in both early and advanced BC has been demonstrated. The benefit of trastuzumab use beyond progression has also been shown, which indicates the need for continuous suppression of the HER2 pathway. Targeting both HER2, with various approaches, and other pathways may enhance the clin. benefit obsd. with trastuzumab and overcome potential resistance. Novel combinations include pertuzumab (a HER2 dimerization inhibitor), lapatinib (a HER1/HER2 tyrosine kinase inhibitor), bevacizumab (an antiangiogenic agent), tanespimycin (a heat shock protein inhibitor), antiestrogen therapies, and an antibody-drug conjugate (trastuzumab-DM1). Conclusion: Trastuzumab is the foundation of care for patients with HER2-pos. BC. Emerging data from studies of other targeted agents may provide alternative treatment combinations to maximize the clin. benefit from trastuzumab and prevent or delay resistance. The continued development of trastuzumab highlights promising treatment approaches for the future.
- 5Slamon, 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– 792Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXisVGktrc%253D&md5=81112f11ffc66fc3a898b61e104322b6Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2Slamon, Dennis J.; Leyland-Jones, Brian; Shak, Steven; Fuchs, Hank; Paton, Virginia; Bajamonde, Alex; Fleming, Thomas; Eiermann, Wolfgang; Wolter, Janet; Pegram, Mark; Baselga, Jose; Norton, LarryNew England Journal of Medicine (2001), 344 (11), 783-792CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)The HER2 gene, which encodes the growth factor receptor HER2, is amplified and HER2 is overexpressed in 25 to 30 % of breast cancers, increasing the aggressiveness of the tumor. We evaluated the efficacy and safety of trastuzumab, a recombinant monoclonal antibody against HER2, in women with metastatic breast cancer that overexpressed HER2. We randomly assigned 234 patients to receive std. chemotherapy plus trastuzumab. Patients who had not previously received adjuvant (postoperative) therapy with an anthracycline were treated with doxorubicin (or epirubicin in the case of 36 women) and cyclophosphamide with (143 women) or without trastuzumab (138 women). Patients who had previously received adjuvant anthracycline were treated with paclitaxel alone (96 women) or paclitaxel with trastuzumab (92 women). The addn. of trastuzumab to chemotherapy was assocd. with a longer time to disease progression (median, 7.4 vs. 4.6 mo; P<0.001), a higher rate of objective response (50 % vs. 32 %, P<0.001), a longer duration of response (median, 9.1 vs. 6.1 mo; P<0.001), a lower rate of death at 1 yr (22 % vs. 33 %, P=0.008), longer survival (median survival, 25.1 vs. 20.3 mo; P=0.046), and a 20 % redn. in the risk of death. The most important adverse event was cardiac dysfunction, which occurred in 27 % of the group given an anthracycline, cyclophosphamide, and trastuzumab; 8 % of the group given an anthracycline and cyclophosphamide alone; 13 % of the group given paclitaxel and trastuzumab; and 1 % of the group given paclitaxel alone. Although the cardiotoxicity was potentially severe and, in some cases, life-threatening, the symptoms generally improved with std. medical management. Trastuzumab increases the clin. benefit of first-line chemotherapy in metastatic breast cancer that overexpresses HER2.
- 6Nahta, R.; Esteva, F. J. HER-2-targeted therapy: lessons learned and future directions Clin. Cancer Res. 2003, 9, 5078– 5084Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXovVOgtb4%253D&md5=0c48944fbc257ee35c8197d04b31595fHER-2-Targeted Therapy: Lessons Learned and Future DirectionsNahta, Rita; Esteva, Francisco J.Clinical Cancer Research (2003), 9 (14), 5078-5084CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)A review. HER-2 is overexpressed in 20-25% of invasive breast cancers and is assocd. with an aggressive tumor phenotype and reduced survival rates. The HER-2 status of a tumor is the crit. determinant of response to the HER-2-targeted antibody trastuzumab. Thus, accurate assessment of HER-2 expression levels is essential for identifying breast cancer patients who will benefit from HER-2-targeted therapy. Trastuzumab combined with chemotherapy increases response rates, time to progression, and survival. However, the majority of cancers that initially respond to trastuzumab begin to progress again within 1 yr. This minireview describes HER-2 targeting strategies currently in use or in stages of development for the treatment of breast cancer.
- 7Metzger-Filho, O.; Winer, E. P.; Krop, I. Pertuzumab: optimizing HER2 blockade Clin. Cancer Res. 2013, 19, 5552– 5556Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1ahurzK&md5=a72443fe22216b25ad4b438bd604f44ePertuzumab: Optimizing HER2 BlockadeMetzger-Filho, Otto; Winer, Eric P.; Krop, IanClinical Cancer Research (2013), 19 (20), 5552-5556CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)A review. Pertuzumab has been approved by the U.S. Food and Drug Administration for use in combination with trastuzumab and docetaxel for the first-line treatment of patients with advanced HER2-pos. (HER2+) breast cancer. Pertuzumab is a recombinant humanized monoclonal antibody that binds to the extracellular dimerization domain II of HER2 and inhibits heterodimerization of HER2 with other HER family members, including EGF receptor (EGFR), HER3, and HER4. The HER2-HER3 heterodimer is a robust activator of phosphoinositide 3-kinase (PI3K) pathway signaling and functions as the most transforming and mitogenic of the receptor complexes formed by the HER family of proteins; thus, blockade of HER2-HER3 likely represents the most relevant action of pertuzumab. In the seminal phase III study, patients with HER2+ metastatic breast cancer were randomized to receive trastuzumab and docetaxel, with or without pertuzumab: Addn. of pertuzumab significantly prolonged progression-free survival with an increase of 6.1 mo (12.4 vs. 18.5 mo, resp.). In a subsequent anal. with 30 mo of median follow-up, pertuzumab conferred a 34% redn. in the risk of mortality. Here, we review the mechanism of action of pertuzumab, the rationale for combining it with trastuzumab/pertuzumab, clin. data, and future directions for this work. Clin Cancer Res; 19(20); 5552-6. ©2013 AACR.
- 8Chari, R. V.; Miller, M. L.; Widdison, W. C. Antibody–drug conjugates: an emerging concept in cancer therapy Angew. Chem., Int. Ed. 2014, 53, 3796– 3827Google ScholarThere is no corresponding record for this reference.
- 9Blattler, W. A.; Chari, R. V. J. Methods of treatment using anti-ERBB antibody–maytansinoid conjugates. U.S. Patent 8,337,856, 2012.Google ScholarThere is no corresponding record for this reference.
- 10Sedlacek, H. H. Antibodies as Carriers of Cytotoxicity; Karger: Basel, Switzerland, 1992; pp 1– 145.Google ScholarThere is no corresponding record for this reference.
- 11Chari Ravi, V. J. Targeted cancer therapy: conferring specificity to cytotoxic drugs Acc. Chem. Res. 2008, 41, 98– 107Google ScholarThere is no corresponding record for this reference.
- 12Kupchan, 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– 2357Google ScholarThere is no corresponding record for this reference.
- 13Widdison, 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– 4408Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XlvFent7o%253D&md5=a06c19d2d5166b0ca806babf9624c48bSemisynthetic Maytansine Analogs for the Targeted Treatment of CancerWiddison, Wayne C.; Wilhelm, Sharon D.; Cavanagh, Emily E.; Whiteman, Kathleen R.; Leece, Barbara A.; Kovtun, Yelena; Goldmacher, Victor S.; Xie, Hongsheng; Steeves, Rita M.; Lutz, Robert J.; Zhao, Robert; Wang, Lintao; Blaettler, Walter A.; Chari, Ravi V. J.Journal of Medicinal Chemistry (2006), 49 (14), 4392-4408CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Maytansine, a highly cytotoxic natural product, failed as an anticancer agent in human clin. trials because of unacceptable systemic toxicity. The potent cell killing ability of maytansine can be used in a targeted delivery approach for the selective destruction of cancer cells. A series of new maytansinoids, bearing a disulfide or thiol substituent were synthesized. The chain length of the ester side chain and the degree of steric hindrance on the carbon atom bearing the thiol substituent were varied. Several of these maytansinoids were found to be even more potent in vitro than maytansine. The targeted delivery of these maytansinoids, using monoclonal antibodies, resulted in a high, specific killing of the targeted cells in vitro and remarkable antitumor activity in vivo.
- 14Issell, B. F.; Crooke, S. T. Maytansine Cancer Treat. Rev. 1978, 5, 199– 207Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaE1M7gsFektw%253D%253D&md5=73ec733c0581edc2085d87aa26291f26MaytansineIssell B F; Crooke S TCancer treatment reviews (1978), 5 (4), 199-207 ISSN:0305-7372.There is no expanded citation for this reference.
- 15Kupchan, 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– 37Google ScholarThere is no corresponding record for this reference.
- 16Chari, 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– 131Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38Xht1Snu7s%253D&md5=33e7613da2a71800fc13e0b2bc6e21f1Immunoconjugates containing novel maytansinoids: promising anticancer drugsChari, Ravi V. J.; Martell, Bridget A.; Gross, Jonathan L.; Cook, Sherrilyn B.; Shah, Sudhir A.; Blattler, Walter A.; McKenzie, Sara J.; Goldmacher, Victor S.Cancer Research (1992), 52 (1), 127-31CODEN: CNREA8; ISSN:0008-5472.The potential of immunoconjugates of cytotoxic drugs for the treatment of cancer has not yet been realized owing to the difficulty of delivering therapeutic concns. of these drugs to the target cells. In an effort to overcome this problem the authors have synthesized maytansinoids that have 100- to 1000-fold higher cytotoxic potency than clin. used anticancer drugs. These maytansinoids are linked to antibodies via disulfide bonds, which ensures the release of fully active drug inside the cells. The conjugates show high antigen-specific cytotoxicity for cultured human cancer cells (50% inhibiting concn., 10 to 40 pM), low systemic toxicity in mice, and good pharmacokinetic behavior.
- 17Kellogg, 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– 727Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjsFKnsro%253D&md5=ef2969ceaaea23e81da2edf06f62ea7eDisulfide-linked antibody-maytansinoid conjugates: optimization of in vivo activity by varying the steric hindrance at carbon atoms adjacent to the disulfide linkageKellogg, Brenda A.; Garrett, Lisa; Kovtun, Yelena; Lai, Katharine C.; Leece, Barbara; Miller, Michael; Payne, Gillian; Steeves, Rita; Whiteman, Kathleen R.; Widdison, Wayne; Xie, Hongsheng; Singh, Rajeeva; Chari, Ravi V. J.; Lambert, John M.; Lutz, Robert J.Bioconjugate Chemistry (2011), 22 (4), 717-727CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)In this report, we describe the synthesis of a panel of disulfide-linked huC242 (anti-CanAg) antibody maytansinoid conjugates (AMCs), which have varying levels of steric hindrance around the disulfide bond, in order to investigate the relationship between stability to redn. of the disulfide linker and antitumor activity of the conjugate in vivo. The conjugates were first tested for stability to redn. by dithiothreitol in vitro and for plasma stability in CD1 mice. It was found that the conjugates having the more sterically hindered disulfide linkages were more stable to reductive cleavage of the maytansinoid in both settings. When the panel of conjugates was tested for in vivo efficacy in two human colon cancer xenograft models in SCID mice, it was found that the conjugate with intermediate disulfide bond stability having two Me groups on the maytansinoid side of the disulfide bond and no Me groups on the linker side of the disulfide bond (huC242-SPDB-DM4) displayed the best efficacy. The ranking of in vivo efficacies of the conjugates was not predicted by their in vitro potencies, since all conjugates were highly active in vitro, including a huC242-SMCC-DM1 conjugate with a noncleavable linkage which showed only marginal activity in vivo. These data suggest that factors in addn. to intrinsic conjugate potency and conjugate half-life in plasma influence the magnitude of antitumor activity obsd. for an AMC in vivo. We provide evidence that bystander killing of neighboring nontargeted tumor cells by diffusible cytotoxic metabolites produced from target cell processing of disulfide-linked antibody-maytansinoid conjugates may be one addnl. factor contributing to the activity of these conjugates in vivo.
- 18Wakankar, A.; Chen, Y.; Gokarn, Y.; Jacobson, F. S. Analytical methods for physicochemical characterization of antibody drug conjugates mAbs 2011, 3, 161– 172Google ScholarThere is no corresponding record for this reference.
- 19Wakankar, 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– 1595Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXpvVyltbk%253D&md5=6c67fb904a35f97adfeb62ec69b7d4bePhysicochemical Stability of the Antibody-Drug Conjugate Trastuzumab-DM1: Changes due to Modification and Conjugation ProcessesWakankar, Aditya A.; Feeney, Maria B.; Rivera, Javier; Chen, Yan; Kim, Michael; Sharma, Vikas K.; Wang, Y. JohnBioconjugate Chemistry (2010), 21 (9), 1588-1595CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)In the manuf. of the antibody-drug conjugate Trastuzumab-DM1 (T-DM1), the lysine residues on the antibody trastuzumab (Tmab) are modified to form the intermediate Tmab-MCC (T-MCC) and then conjugated with the drug DM1. Our goal is to understand the effects of modification and conjugation steps on the physicochem. stability of the antibody. The structural stability of Tmab relative to its modified and conjugated forms was assessed, employing thermally induced stress conditions to formulations contg. Tmab, T-MCC, and T-DM1. DSC, SEC, CE-SDS, and LC-MS were used to study the stability of Tmab, T-MCC, and T-DM1 to thermal stress. The DSC thermograms show a decrease in melting temp. for the CH2 transition, in the order Tmab > T-MCC > T-DM1. As per SEC anal., a significant increase in level of aggregation was detected in T-MCC (∼32%) and T-DM1 (∼5%) after 14 days at 40°. Tmab did not show significant aggregate formation. CE-SDS and LC-MS data demonstrate that the aggregation in the case of T-MCC is largely covalent and involves mechanisms other than formation of intermol. disulfide crosslinks. The aggregation obsd. for T-MCC was significantly inhibited upon addn. of amino acids with nucleophilic side chains contg. thiol (Cys) and hydroxyl moieties (Ser, Tyr). The covalent aggregation obsd. for T-MCC and the ability of nucleophilic amino acids, particularly Cys, to inhibit it indicate that the maleimide moiety in the MCC linker may react to form intermol. covalent crosslinks between T-MCC mols., possibly through a Michael addn. mechanism. In addn., DSC results demonstrate that the conjugation of the drug moiety DM1 to Tmab results in destabilization of the CH2 domain of the antibody.
- 20Lewis 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– 9290Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlOmt7rK&md5=e86e8687d5f8884f8e28514299dcbbf1Targeting HER2-Positive Breast Cancer with Trastuzumab-DM1, an Antibody-Cytotoxic Drug ConjugateLewis Phillips, Gail D.; Li, Guangmin; Dugger, Debra L.; Crocker, Lisa M.; Parsons, Kathryn L.; Mai, Elaine; Blaettler, Walter A.; Lambert, John M.; Chari, Ravi V. J.; Lutz, Robert J.; Wong, Wai Lee T.; Jacobson, Frederic S.; Koeppen, Hartmut; Schwall, Ralph H.; Kenkare-Mitra, Sara R.; Spencer, Susan D.; Sliwkowski, Mark X.Cancer Research (2008), 68 (22), 9280-9290CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)HER2 is a validated target in breast cancer therapy. Two drugs are currently approved for HER2-pos. breast cancer: trastuzumab (Herceptin), introduced in 1998, and lapatinib (Tykerb), in 2007. Despite these advances, some patients progress through therapy and succumb to their disease. A variation on antibody-targeted therapy is utilization of antibodies to deliver cytotoxic agents specifically to antigen-expressing tumors. We detd. in vitro and in vivo efficacy, pharmacokinetics, and toxicity of trastuzumab-maytansinoid (microtubule-depolymg. agents) conjugates using disulfide and thioether linkers. Antiproliferative effects of trastuzumab-maytansinoid conjugates were evaluated on cultured normal and tumor cells. In vivo activity was detd. in mouse breast cancer models, and toxicity was assessed in rats as measured by body wt. loss. Surprisingly, trastuzumab linked to DM1 through a nonreducible thioether linkage (SMCC), displayed superior activity compared with unconjugated trastuzumab or trastuzumab linked to other maytansinoids through disulfide linkers. Serum concns. of trastuzumab-MCC-DM1 remained elevated compared with other conjugates, and toxicity in rats was negligible compared with free DM1 or trastuzumab linked to DM1 through a reducible linker. Potent activity was obsd. on all HER2-overexpressing tumor cells, whereas nontransformed cells and tumor cell lines with normal HER2 expression were unaffected. In addn., trastuzumab-DM1 was active on HER2-overexpressing, trastuzumab-refractory tumors. In summary, trastuzumab-DM1 shows greater activity compared with nonconjugated trastuzumab while maintaining selectivity for HER2-overexpressing tumor cells. Because trastuzumab linked to DM1 through a nonreducible linker offers improved efficacy and pharmacokinetics and reduced toxicity over the reducible disulfide linkers evaluated, trastuzumab-MCC-DM1 was selected for clin. development.
- 21Erickson, 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– 1142Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xms1yrtLs%253D&md5=e7485e8164a5bf3210f6c68f174df112The Effect of Different Linkers on Target Cell Catabolism and Pharmacokinetics/Pharmacodynamics of Trastuzumab Maytansinoid ConjugatesErickson, Hans K.; Lewis Phillips, Gail D.; Leipold, Douglas D.; Provenzano, Carmela A.; Mai, Elaine; Johnson, Holly A.; Gunter, Bert; Audette, Charlene A.; Gupta, Manish; Pinkas, Jan; Tibbitts, JayMolecular Cancer Therapeutics (2012), 11 (5), 1133-1142CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate consisting of the anti-HER2 antibody trastuzumab linked via a nonreducible thioether linker to the maytansinoid antitubulin agent DM1. T-DM1 has shown favorable safety and efficacy in patients with HER2-pos. metastatic breast cancer. In previous animal studies, T-DM1 exhibited better pharmacokinetics (PK) and slightly more efficacy than several disulfide-linked versions. The efficacy findings are unique, as other disulfide-linked antibody-drug conjugates (ADC) have shown greater efficacy than thioether-linked designs. To explore this further, the in vitro and in vivo activity, PK, and target cell activation of T-DM1 and the disulfide-linked T-SPP-DM1 were examd. Both ADCs showed high in vitro potency, with T-DM1 displaying greater potency in two of four breast cancer cell lines. In vitro target cell processing of T-DM1 and T-SPP-DM1 produced lysine-N.vepsiln.-MCC-DM1, and lysine-N.vepsiln.-SPP-DM1 and DM1, resp.; in vivo studies confirmed these results. The in vitro processing rates for the two conjugate to their resp. catabolites were similar. In vivo, the potencies of the conjugates were similar, and T-SPP-DM1 had a faster plasma clearance than T-DM1. Slower T-DM1 clearance translated to higher overall tumor concns. (conjugate plus catabolites), but unexpectedly, similar levels of tumor catabolite. These results indicate that, although the ADC linker can have clear impact on the PK and the chem. nature of the catabolites formed, both linkers seem to offer the same payload delivery to the tumor. Mol Cancer Ther; 11(5); 1133-42.
- 22Junttila, 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– 356Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXnslehsbo%253D&md5=f3b04d9b9bb099007f338c619dd65f00Trastuzumab-DM1 (T-DM1) retains all the mechanisms of action of trastuzumab and efficiently inhibits growth of lapatinib insensitive breast cancerJunttila, Teemu T.; Li, Guangmin; Parsons, Kathryn; Phillips, Gail Lewis; Sliwkowski, Mark X.Breast Cancer Research and Treatment (2011), 128 (2), 347-356CODEN: BCTRD6; ISSN:0167-6806. (Springer)Trastuzumab (Herceptin) is currently used as a treatment for patients whose breast tumors overexpress HER2/ErbB2. Trastuzumab-DM1 (T-DM1, trastuzumab emtansine) is designed to combine the clin. benefits of trastuzumab with a potent microtubule-disrupting drug, DM1 (a maytansine deriv.). Currently T-DM1 is being tested in multiple clin. trials. The mechanisms of action for trastuzumab include inhibition of PI3K/AKT signaling pathway, inhibition of HER-2 shedding and Fcγ receptor mediated engagement of immune cells, which may result in antibody-dependent cellular cytotoxicity (ADCC). Here we report that T-DM1 retains the mechanisms of action of unconjugated trastuzumab and is active against lapatinib resistant cell lines and tumors.
- 23Barok, 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– 179Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXlsV2nt7k%253D&md5=b3c549e901da81d69ccdf85af4808310Trastuzumab-DM1 is highly effective in preclinical models of HER2-positive gastric cancerBarok, Mark; Tanner, Minna; Koeninki, Katri; Isola, JormaCancer Letters (New York, NY, United States) (2011), 306 (2), 171-179CODEN: CALEDQ; ISSN:0304-3835. (Elsevier)Background: A novel antibody-drug conjugate (trastuzumab-DM1, T-DM1) is currently in clin. trials for patients with trastuzumab resistant HER2-pos. breast cancer. Since no clin. data is available from gastric cancer, we studied T-DM1 on HER2-pos. human gastric cancer cells and xenograft tumors. Methods: Effects of T-DM1 were studied in four HER2-pos. gastric cancer cell lines (N-87, OE-19, SNU-216 and MKN-7) in vitro. Xenograft tumors from N-87 and OE-19 were studied to det. the effect of T-DM1 in vivo. Results: T-DM1 was found more effective than trastuzumab in N-87 and OE-19, and moderately effective in MKN-7 cells. On SNU-216 cells both trastuzumab and T-DM1 showed limited efficacy. In xenograft tumor expts., complete pathol. response was obsd. in all OE-19 xenografted mice and in half of the N-87 xenografted mice. The results were equally good irresp. of the tumor burden at therapy initiation, or preceding trastuzumab treatment. T-DM1 treatment showed direct effects (apoptotic cell death and aberrant mitosis) as well as it mediated antibody-dependent cellular cytotoxcity (ADCC). Conclusions: T-DM1 showed a promising anti-tumor effect in HER2-pos. gastric cancer cell lines in vitro and in vivo, even in tumors which had developed resistance to trastuzumab. T-DM1 therapy may warrant clin. trials for HER2-pos. gastric cancer patients.
- 24Kovtun, 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– 2537Google ScholarThere is no corresponding record for this reference.
- 25Guo, 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 618Google ScholarThere is no corresponding record for this reference.
- 26Jumbe, 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– 242Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnslyktLw%253D&md5=28bac6e680530a3aeba7b44607669c8cModeling the efficacy of trastuzumab-DM1, an antibody drug conjugate, in miceJumbe, Nelson L.; Xin, Yan; Leipold, Douglas D.; Crocker, Lisa; Dugger, Debra; Mai, Elaine; Sliwkowski, Mark X.; Fielder, Paul J.; Tibbitts, JayJournal of Pharmacokinetics and Pharmacodynamics (2010), 37 (3), 221-242CODEN: JPPOAH; ISSN:1567-567X. (Springer)Trastuzumab-DM1 (T-DM1) is a novel antibody-drug conjugate under investigation for the treatment of human epidermal growth factor receptor 2 (HER2)-pos. metastatic breast cancer. One challenge in oncol. drug development is detg. the optimal dose and treatment schedule. A novel dose regimen-finding strategy was developed for T-DM1 using exptl. data and pharmacokinetic/pharmacodynamic modeling. To characterize the disposition of T-DM1, pharmacokinetic studies were conducted in athymic nude and beige nude mice. The pharmacokinetics of T-DM1 were described well by a two-compartment model. Tumor response data were obtained from single-dose, multiple-dose and time-dose-fractionation studies of T-DM1 in animal models of HER2-pos. breast cancer, specifically engineered to be insensitive to trastuzumab. A sequential population-based pharmacokinetic/pharmacodynamic modeling approach was developed to describe the anti-tumor activity of T-DM1. A cell-cycle-phase nonspecific tumor cell kill model incorporating transit compartments captured well the features of tumor growth and the activity of T-DM1. Key findings of the model were that tumor cell growth rate played a significant role in the sensitivity of tumors to T-DM1; anti-tumor activity was schedule independent; and tumor response was linked to the ratio of exposure to a concn. required for tumor stasis.
- 27Kaur, 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– 226Google ScholarThere is no corresponding record for this reference.
- 28Poon, 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– 313Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslGlsbbO&md5=f2ae6f6fc865d8ad027752e1301be3ccPreclinical safety profile of trastuzumab emtansine (T-DM1): Mechanism of action of its cytotoxic component retained with improved tolerabilityPoon, Kirsten Achilles; Flagella, Kelly; Beyer, Joseph; Tibbitts, Jay; Kaur, Surinder; Saad, Ola; Yi, Joo-Hee; Girish, Sandhya; Dybdal, Noel; Reynolds, TheresaToxicology and Applied Pharmacology (2013), 273 (2), 298-313CODEN: TXAPA9; ISSN:0041-008X. (Elsevier Inc.)Trastuzumab emtansine (T-DM1) is the first antibody-drug conjugate (ADC) approved for patients with human epidermal growth factor receptor 2 (HER2)-pos. metastatic breast cancer. The therapeutic premise of ADCs is based on the hypothesis that targeted delivery of potent cytotoxic drugs to tumors will provide better tolerability and efficacy compared with non-targeted delivery, where poor tolerability can limit efficacious doses. Here, we present results from preclin. studies characterizing the toxicity profile of T-DM1, including limited assessment of unconjugated DM1. T-DM1 binds primate ErbB2 and human HER2 but not the rodent homolog c-neu. Therefore, antigen-dependent and non-antigen-dependent toxicity was evaluated in monkeys and rats, resp., in both single- and repeat-dose studies; toxicity of DM1 was assessed in rats only. T-DM1 was well tolerated at doses up to 40 mg/kg (~ 4400 μg DM1/m2) and 30 mg/kg (~ 6000 μg DM1/m2) in rats and monkeys, resp. In contrast, DM1 was only tolerated up to 0.2 mg/kg (1600 μg DM1/m2). This suggests that at least two-fold higher doses of the cytotoxic agent are tolerated in T-DM1, supporting the premise of ADCs to improve the therapeutic index. In addn., T-DM1 and DM1 safety profiles were similar and consistent with the mechanism of action of DM1 (i.e., microtubule disruption). Findings included hepatic, bone marrow/hematol. (primarily platelet), lymphoid organ, and neuronal toxicities, and increased nos. of cells of epithelial and phagocytic origin in metaphase arrest. These adverse effects did not worsen with chronic dosing in monkeys and are consistent with those reported in T-DM1-treated patients to date.
- 29Sun, 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– 735Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXivFyitL4%253D&md5=042dafa46e287f5a8a76af8714c124ceDesign of Antibody-Maytansinoid Conjugates Allows for Efficient Detoxification via Liver MetabolismSun, Xiuxia; Widdison, Wayne; Mayo, Michele; Wilhelm, Sharon; Leece, Barbara; Chari, Ravi; Singh, Rajeeva; Erickson, HansBioconjugate Chemistry (2011), 22 (4), 728-735CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Antibody-maytansinoid conjugates (AMCs) are targeted chemotherapeutic agents consisting of a potent microtubule-depolymg. maytansinoid (DM1 or DM4) attached to lysine residues of a monoclonal antibody (mAb) using an uncleavable thioether linker or a stable disulfide linker. Most of the administered dose of an antibody-based therapeutic is slowly catabolized by the liver and other tissues of the reticuloendothelial system. Maytansinoids released from an AMC during this catabolic process could potentially be a source of toxicity. To investigate this, we isolated and identified liver metabolites in mice for three different [3H]AMCs with structures similar to those currently undergoing evaluation in the clinic. We then synthesized each metabolite to confirm the identification and assessed their cytotoxic potencies when added extracellularly. We found that the uncleavable mAb-SMCC-[3H]DM1 conjugate was degraded to a single major maytansinoid metabolite, lysine-SMCC-[3H]DM1, that was nearly 50-fold less cytotoxic than maytansine. The two disulfide-linked conjugates, mAb-SPP-[3H]DM1 and mAb-SPDB-[3H]DM4, were also catabolized to the analogous lysine-linked maytansinoid metabolites. However, subsequent redn., S-methylation, and NADPH-dependent oxidn. steps in the liver yielded the corresponding S-Me sulfoxide and S-Me sulfone derivs. The cytotoxic potencies of the oxidized maytansinoids toward several human carcinoma cell lines were 5- to 50-fold less potent than maytansine. Our results suggest that liver plays an important role in the detoxification of both cleavable and uncleavable AMCs.
- 30Fishkin, 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– 10754Google ScholarThere is no corresponding record for this reference.
- 31Krop, 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– 2704Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXoslykt7c%253D&md5=98ba29a289f4ac0ea38c37a9b6bd0c5dPhase I study of trastuzumab-DM1, an HER2 antibody-drug conjugate, given every 3 weeks to patients with HER2-positive metastatic breast cancerKrop, Ian E.; Beeram, Muralidhar; Modi, Shanu; Jones, Suzanne F.; Holden, Scott N.; Yu, Wei; Girish, Sandhy; Tibbitts, Jay; Yi, Joo-Hee; Sliwkowski, Mark X.; Jacobson, Fred; Lutzker, Stuart G.; Burris, Howard A.Journal of Clinical Oncology (2010), 28 (16), 2698-2704CODEN: JCONDN; ISSN:0732-183X. (American Society of Clinical Oncology)Purpose: Trastuzumab-DM1 (T-DM1) is an antibody-drug conjugate that uses trastuzumab to specifically deliver the maytansinoid antimicrotubule agent DM1 to HER2-pos. cells. This first-in-human study of T-DM1 evaluated safety, pharmacokinetics, and preliminary activity of T-DM1 in patients with advanced HER2-pos. breast cancer. Patients and Methods: Successive cohorts of patients who had progressed on trastuzumab-based therapy received escalating doses of T-DM1. Outcomes were assessed by std. solid-tumor phase I methods. Results: Twenty-four patients who had received a median of four prior chemotherapeutic agents for metastatic disease received T-DM1 at 0.3 mg/kg to 4.8 mg/kg on an every-3-wk schedule. Transient thrombocytopenia was dose-limiting at 4.8 mg/kg; the max.-tolerated dose (MTD) was 3.6 mg/kg. The half-life of T-DM1 at the MTD was 3.5 days, with peak DM1 levels < 10 ng/mL. Clearance at doses < 1.2 mg/kg was faster than at higher doses. Common drug-related adverse events (AEs) included grade ≤ 2 thrombocytopenia, elevated transaminases, fatigue, nausea, and anemia. No grade > 1 nausea, vomiting, alopecia, or neuropathy events and no cardiac effects requiring dose modification were reported. The clin. benefit rate (objective response plus stable disease at 6 mo) among 15 patients treated at the MTD was 73%, including five objective responses. The confirmed response rate in patients with measurable disease at the MTD (n = 9) was 44%. Conclusion: At the MTD of 3.6 mg/kg every 3 wk, T-DM1 was assocd. with mild, reversible toxicity and substantial clin. activity in a heavily pretreated population. Phase II and III trials in patients with advanced HER2-pos. breast cancer are under way.
- 32Baselga, J. Phase I and II clinical trials of trastuzumab Ann. Oncol. 2001, 12 (Suppl. 1) S49– S55Google ScholarThere is no corresponding record for this reference.
- 33Beeram, 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– 5740Google ScholarThere is no corresponding record for this reference.
- 34Burris, 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– 405Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXivFWmsrc%253D&md5=94f0c939a12a7875590fc02f3f056d65Phase 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 therapyBurris, Howard A., III; Rugo, Hope S.; Vukelja, Svetislava J.; Vogel, Charles L.; Borson, Rachel A.; Limentani, Steven; Tan-Chiu, Elizabeth; Krop, Ian E.; Michaelson, Richard A.; Girish, Sandhya; Amler, Lukas; Zheng, Maoxia; Chu, Yu-Waye; Klencke, Barbara; O'Shaughnessy, Joyce A.Journal of Clinical Oncology (2011), 29 (4), 398-405CODEN: JCONDN; ISSN:0732-183X. (American Society of Clinical Oncology)The antibody-drug conjugate trastuzumab-DM1 (T-DM1) combines the biol. activity of trastuzumab with targeted delivery of a potent antimicrotubule agent, DM1, to human epidermal growth factor receptor 2 (HER2) -overexpressing cancer cells. Based on results from a phase I study that showed T-DM1 was well tolerated at the max.-tolerated dose of 3.6 mg/kg every 3 wk, with evidence of efficacy, in patients with HER2-pos. metastatic breast cancer (MBC) who were previously treated with trastuzumab, we conducted a phase II study to further define the safety and efficacy of T-DM1 in this patient population. This report describes a single-arm phase II study (TDM4258g) that assessed efficacy and safety of i.v. T-DM1 (3.6 mg/kg every 3 wk) in patients with HER2-pos. MBC who had tumor progression after prior treatment with HER2-directed therapy and who had received prior chemotherapy. With a follow-up of ≥ 12 mo among 112 treated patients, the objective response rate by independent assessment was 25.9% (95% CI, 18.4% to 34.4%). Median duration of response was not reached as a result of insufficient events (lower limit of 95% CI, 6.2 mo), and median progression-free survival time was 4.6 mo (95% CI, 3.9 to 8.6 mo). The response rates were higher among patients with confirmed HER2-pos. tumors (immunohistochem. 3+ or fluorescent in situ hybridization pos.) by retrospective central testing (n = 74). Higher response rates were also obsd. in patients whose tumors expressed ≥ median HER2 levels by quant. reverse transcriptase polymerase chain reaction for HER2 expression, compared with patients who had less than median HER2 levels. T-DM1 was well tolerated with no dose-limiting cardiotoxicity. Most adverse events (AEs) were grade 1 or 2; the most frequent grade ≥ 3 AEs were hypokalemia (8.9%), thrombocytopenia (8.0%), and fatigue (4.5%). T-DM1 has robust single-agent activity in patients with heavily pretreated, HER2-pos. MBC and is well tolerated at the recommended phase II dose.
- 35Krop, 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– 3241Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsVKrsL%252FJ&md5=e3c54c63b8a2c6a585818e35c0e4aab9A 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 capecitabineKrop, Ian E.; LoRusso, Patricia; Miller, Kathy D.; Modi, Shanu; Yardley, Denise; Rodriguez, Gladys; Guardino, Ellie; Lu, Michael; Zheng, Maoxia; Girish, Sandhya; Amler, Lukas; Winer, Eric P.; Rugo, Hope S.; Krop, Ian E.; Winer, Eric P.Journal of Clinical Oncology (2012), 30 (26), 3234-3241CODEN: JCONDN; ISSN:0732-183X. (American Society of Clinical Oncology)Purpose. To det. whether the antibody-drug conjugate trastuzumab emtansine (T-DM1), which combines human epidermal growth factor receptor 2 (HER2) -targeted delivery of the potent antimicrotubule agent DM1 with the antitumor activity of trastuzumab, is effective in patients with HER2-pos. metastatic breast cancer (MBC) who have previously received all std. HER2-directed therapies. Patients and Methods. In this single-arm phase II study, T-DM1 3.6 mg/kg was administered i.v. every 3 wk to patients with HER2-pos. MBC who had prior treatment with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine. The primary objectives were overall response rate (ORR) by independent review and safety. Results. Among 110 pretreated patients (median, seven prior agents for MBC; median follow-up, 17.4 mo), the ORR was 34.5% (95% CI, 26.1% to 43.9%), clin. benefit rate was 48.2% (95% CI, 38.8% to 57.9%), median progression-free survival (PFS) was 6.9 mo (95% CI, 4.2 to 8.4 mo), and median duration of response was 7.2 mo (95% CI, 4.6 mo to not estimable). In patients with confirmed HER2-pos. tumors (n = 80 by retrospective central testing), the response rate was 41.3% (95% CI, 30.4% to 52.8%), and median PFS was 7.3 mo (95% CI, 4.6 to 12.3 mo). Most adverse events were grades 1 to 2; the most frequent grade ≥ 3 events were thrombocytopenia (9.1%), fatigue (4.5%), and cellulitis (3.6%). Conclusion. T-DM1 is well tolerated and has single-agent activity in patients with HER2-pos. MBC who have previously received both approved HER2-directed therapies and multiple chemotherapy agents. T-DM1 may be an effective new treatment for this patient population.
- 36Hurvitz, 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– 1163Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXntVyhtbs%253D&md5=95cd6eb05ce016e971b2af745d1b587fPhase II randomized study of trastuzumab emtansine versus trastuzumab plus docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancerHurvitz, Sara A.; Dirix, Luc; Kocsis, Judit; Bianchi, Giulia V.; Lu, Janice; Vinholes, Jeferson; Guardino, Ellie; Song, Chunyan; Tong, Barbara; Ng, Vivian; Chu, Yu-Waye; Perez, Edith A.Journal of Clinical Oncology (2013), 31 (9), 1157-1163CODEN: JCONDN; ISSN:0732-183X. (American Society of Clinical Oncology)Purpose: Trastuzumab emtansine (T-DM1), an antibody-drug conjugate composed of the cytotoxic agent DM1 conjugated to trastuzumab via a stable thioether linker, has shown clin. activity in single-arm studies enrolling patients with human epidermal growth factor receptor 2 (HER2)-pos. metastatic breast cancer (MBC) whose disease had progressed on HER2-targeted therapy in the metastatic setting. Patients and Methods: Patients (N = 137) with HER2-pos. MBC or recurrent locally advanced breast cancer were randomly assigned to trastuzumab plus docetaxel (HT; n = 70) or T-DM1 (n = 67) as first-line treatment until disease progression or unacceptable toxicity. Primary end points were investigator-assessed progression-free survival (PFS) and safety. Key secondary end points included overall survival (OS), objective response rate (ORR), duration of objective response, clin. benefit rate, and quality of life. Results: Median PFS was 9.2 mo with HT and 14.2 mo with T-DM1 (hazard ratio, 0.59; 95% CI, 0.36 to 0.97); median follow-up was approx. 14 mo in both arms. ORR was 58.0% (95% CI, 45.5% to 69.2%) with HT and 64.2% (95% CI, 51.8% to 74.8%) with T-DM1. T-DM1 had a favorable safety profile vs. HT, with fewer grade ≥ 3 adverse events (AEs; 46.4% v 90.9%), AEs leading to treatment discontinuations (7.2% v 34.8%), and serious AEs (20.3% v 25.8%). Preliminary OS results were similar between treatment arms; median follow-up was approx. 23 mo in both arms. Conclusion: In this randomized phase II study, first-line treatment with T-DM1 for patients with HER2-pos. MBC provided a significant improvement in PFS, with a favorable safety profile, vs. HT.
- 37Suter, 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– 3865Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtFanu7zE&md5=9aa4ecb50b0566677b2552e7c645b2f2Trastuzumab-associated cardiac adverse effects in the herceptin adjuvant trialSuter, Thomas M.; Procter, Marion; van Veldhuisen, Dirk J.; Muscholl, Michael; Bergh, Jonas; Carlomagno, Chiara; Perren, Timothy; Passalacqua, Rodolfo; Bighin, Claudia; Klijn, Jan G. M.; Ageev, Fail T.; Hitre, Erika; Greetz, Juergen; Iwata, Hiroji; Knap, Malgorzata; Gnant, Michael; Muehlbauer, Susanne; Spence, Alison; Gelber, Richard D.; Piccart-Gebhart, Martine J.Journal of Clinical Oncology (2007), 25 (25), 3859-3865CODEN: JCONDN; ISSN:0732-183X. (American Society of Clinical Oncology)Purpose: The purpose of this anal. was to investigate trastuzumab-assocd. cardiac adverse effects in breast cancer patients after completion of (neo)adjuvant chemotherapy with or without radiotherapy. Patients and Methods: The Herceptin Adjuvant (HERA) trial is a three-group, multicenter, open-label randomized trial that compared 1 or 2 years of trastuzumab given once every 3 wk with observation in patients with HER-2-pos. breast cancer. Only patients who after completion of (neo)adjuvant chemotherapy with or without radiotherapy had normal left ventricular ejection fraction (LVEF ≥ 55%) were eligible. A repeat LVEF assessment was performed in case of cardiac dysfunction. Results: Data were available for 1,693 patients randomly assigned to 1 yr trastuzumab and 1,693 patients randomly assigned to observation. The incidence of trastuzumab discontinuation due to cardiac disorders was low (4.3%). The incidence of cardiac end points was higher in the trastuzumab group compared with observation (severe congestive heart failure [CHF], 0.60% v 0.00%; symptomatic CHF, 2.15% v 0.12%; confirmed significant LVEF drops, 3.04% v 0.53%). Most patients with cardiac dysfunction recovered in fewer than 6 mo. Patients with trastuzumab-assocd. cardiac dysfunction were treated with higher cumulative doses of doxorubicin (287 mg/m2 v 257 mg/m2) or epirubicin (480 mg/m2 v 422 mg/m2) and had a lower screening LVEF and a higher body mass index. Conclusion: Given the clear benefit in disease-free survival, the low incidence of cardiac adverse events, and the suggestion that cardiac dysfunction might be reversible, adjuvant trastuzumab should be considered for treatment of breast cancer patients who fulfill the HERA trial eligibility criteria.
- 38Verma, 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– 1791Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1ekt73M&md5=3be5bac27a81b530442dc8a6b4a1f82eTrastuzumab emtansine for HER2-positive advanced breast cancerVerma, Sunil; Miles, David; Gianni, Luca; Krop, Ian E.; Welslau, Manfred; Baselga, Jose; Pegram, Mark; Oh, Do-Youn; Dieras, Veronique; Guardino, Ellie; Fang, Liang; Lu, Michael W.; Olsen, Steven; Blackwell, KimNew England Journal of Medicine (2012), 367 (19), 1783-1791CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)BACKGROUND: Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate incorporating the human epidermal growth factor receptor 2 (HER2)-targeted antitumor properties of trastuzumab with the cytotoxic activity of the microtubule-inhibitory agent DM1. The antibody and the cytotoxic agent are conjugated by means of a stable linker. METHODS: We randomly assigned patients with HER2-pos. advanced breast cancer, who had previously been treated with trastuzumab and a taxane, to T-DM1 or lapatinib plus capecitabine. The primary end points were progression-free survival (as assessed by independent review), overall survival, and safety. Secondary end points included progression-free survival (investigator-assessed), the objective response rate, and the time to symptom progression. Two interim analyses of overall survival were conducted. RESULTS: Among 991 randomly assigned patients, median progression-free survival as assessed by independent review was 9.6 mo with T-DM1 vs. 6.4 mo with lapatinib plus capecitabine (hazard ratio for progression or death from any cause, 0.65; 95% confidence interval [CI], 0.55 to 0.77; P<0.001), and median overall survival at the second interim anal. crossed the stopping boundary for efficacy (30.9 mo vs. 25.1 mo; hazard ratio for death from any cause, 0.68; 95% CI, 0.55 to 0.85; P<0.001). The objective response rate was higher with T-DM1 (43.6%, vs. 30.8% with lapatinib plus capecitabine; P<0.001); results for all addnl. secondary end points favored T-DM1. Rates of grade 3 or above were higher with lapatinib plus capecitabine than with T-DM1 (57% vs. 41%). The incidences of thrombocytopenia and increased serum aminotransferase levels were higher with T-DM1, whereas the incidences of diarrhea, nausea, vomiting, and palmar-plantar erythrodysesthesia were higher with lapatinib plus capecitabine. CONCLUSIONS: T-DM1 significantly prolonged progression-free and overall survival with less toxicity than lapatinib plus capecitabine in patients with HER2-pos. advanced breast cancer previously treated with trastuzumab and a taxane.
- 39Krop, 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– 699Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXntl2gs7o%253D&md5=975dd9e786d96360b163c2583aefc4b8Trastuzumab emtansine versus treatment of physician's choice for pretreated HER2-positive advanced breast cancer (TH3RESA): a randomised, open-label, phase 3 trialKrop, Ian E.; Kim, Sung-Bae; Gonzalez-Martin, Antonio; LoRusso, Patricia M.; Ferrero, Jean-Marc; Smitt, Melanie; Yu, Ron; Leung, Abraham C. F.; Wildiers, HansLancet Oncology (2014), 15 (7), 689-699CODEN: LOANBN; ISSN:1470-2045. (Elsevier Ltd.)Patients with progressive disease after two or more HER2-directed regimens for recurrent or metastatic breast cancer have few effective therapeutic options. We aimed to compare trastuzumab emtansine, an antibody-drug conjugate comprising the cytotoxic agent DM1 linked to trastuzumab, with treatment of physician's choice in this population of patients.This randomised, open-label, phase 3 trial took place in medical centers in 22 countries across Europe, North America, South America, and Asia-Pacific. Eligible patients (≥18 years, left ventricular ejection fraction ≥50%, Eastern Cooperative Oncol. Group performance status 0-2) with progressive HER2-pos. advanced breast cancer who had received two or more HER2-directed regimens in the advanced setting, including trastuzumab and lapatinib, and previous taxane therapy in any setting, were randomly assigned (in a 2:1 ratio) to trastuzumab emtansine (3·6 mg/kg i.v. every 21 days) or physician's choice using a permuted block randomization scheme by an interactive voice and web response system. Patients were stratified according to world region (USA vs western Europe vs other), no. of previous regimens (excluding single-agent hormonal therapy) for the treatment of advanced disease (two to three vs more than three), and presence of visceral disease (any vs none). Coprimary endpoints were investigator-assessed progression-free survival (PFS) and overall survival in the intention-to-treat population. We report the final PFS anal. and the first interim overall survival anal. This study is registered with ClinicalTrials.gov, no. NCT01419197.From Sept 14, 2011, to Nov 19, 2012, 602 patients were randomly assigned (404 to trastuzumab emtansine and 198 to physician's choice). At data cutoff (Feb 11, 2013), 44 patients assigned to physician's choice had crossed over to trastuzumab emtansine. After a median follow-up of 7·2 mo (IQR 5·0-10·1 mo) in the trastuzumab emtansine group and 6·5 mo (IQR 4·1-9·7) in the physician's choice group, 219 (54%) patients in the trastuzumab emtansine group and 129 (65%) of patients in the physician's choice group had PFS events. PFS was significantly improved with trastuzumab emtansine compared with physician's choice (median 6·2 mo [95% CI 5·59-6·87] vs 3·3 mo [2·89-4·14]; stratified hazard ratio [HR] 0·528 [0·422-0·661]; p<0·0001). Interim overall survival anal. showed a trend favoring trastuzumab emtansine (stratified HR 0·552 [95% CI 0·369-0·826]; p=0·0034), but the stopping boundary was not crossed. A lower incidence of grade 3 or worse adverse events was reported with trastuzumab emtansine than with physician's choice (130 events [32%] in 403 patients vs 80 events [43%] in 184 patients). Neutropenia (ten [2%] vs 29 [16%]), diarrhoea (three [<1%] vs eight [4%]), and febrile neutropenia (one [<1%] vs seven [4%]) were grade 3 or worse adverse events that were more common in the physician's choice group than in the trastuzumab emtansine group. Thrombocytopenia (19 [5%] vs three [2%]) was the grade 3 or worse adverse event that was more common in the trastuzumab emtansine group. 74 (18%) patients in the trastuzumab emtansine group and 38 (21%) in the physician's choice group reported a serious adverse event. Trastuzumab emtansine should be considered as a new std. for patients with HER2-pos. advanced breast cancer who have previously received trastuzumab and lapatinib.Genentech.
- 40Baselga, 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– 119Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFSlt7Y%253D&md5=8f8a05c82e4722049923dd069f1863b4Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancerBaselga, Jose; Cortes, Javier; Kim, Sung-Bae; Im, Seock-Ah; Hegg, Roberto; Im, Young-Hyuck; Roman, Laslo; Pedrini, Jose Luiz; Pienkowski, Tadeusz; Knott, Adam; Clark, Emma; Benyunes, Mark C.; Ross, Graham; Swain, Sandra M.New England Journal of Medicine (2012), 366 (2), 109-119CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)BACKGROUND The anti-human epidermal growth factor receptor 2 (HER2) humanized monoclonal antibody trastuzumab improves the outcome in patients with HER2-pos. metastatic breast cancer. However, most cases of advanced disease eventually progress. Pertuzumab, an anti-HER2 humanized monoclonal antibody that inhibits receptor dimerization, has a mechanism of action that is complementary to that of trastuzumab, and combination therapy with the two antibodies has shown promising activity and an acceptable safety profile in phase 2 studies involving patients with HER2-pos. breast cancer. METHODS We randomly assigned 808 patients with HER2-pos. metastatic breast cancer to receive placebo plus trastuzumab plus docetaxel (control group) or pertuzumab plus trastuzumab plus docetaxel (pertuzumab group) as first-line treatment until the time of disease progression or the development of toxic effects that could not be effectively managed. The primary end point was independently assessed progression-free survival. Secondary end points included overall survival, progression-free survival as assessed by the investigator, the objective response rate, and safety. RESULTS The median progression-free survival was 12.4 mo in the control group, as com pared with 18.5 mo in the pertuzumab group (hazard ratio for progression or death, 0.62; 95% confidence interval, 0.51 to 0.75; P < 0.001). The interim anal. of overall survival showed a strong trend in favor of pertuzumab plus trastuzumab plus docetaxel. The safety profile was generally similar in the two groups, with no increase in left ventricular systolic dysfunction; the rates of febrile neutropenia and diarrhea of grade 3 or above were higher in the pertuzumab group than in the control group. CONCLUSIONS The combination of pertuzumab plus trastuzumab plus docetaxel, as compared with placebo plus trastuzumab plus docetaxel, when used as first-line treatment for HER2-pos. metastatic breast cancer, significantly prolonged progression-free survival, with no increase in cardiac toxic effects.
- 41Phillips, 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– 468Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtFKrs7c%253D&md5=71f2be1c5fccaee1d613d09c0eaf48b5Dual Targeting of HER2-Positive Cancer with Trastuzumab Emtansine and Pertuzumab: Critical Role for Neuregulin Blockade in Antitumor Response to Combination TherapyPhillips, Gail D. Lewis; Fields, Carter T.; Li, Guangmin; Dowbenko, Donald; Schaefer, Gabriele; Miller, Kathy; Andre, Fabrice; Burris, Howard A.; Albain, Kathy S.; Harbeck, Nadia; Dieras, Veronique; Crivellari, Diana; Fang, Liang; Guardino, Ellie; Olsen, Steven R.; Crocker, Lisa M.; Sliwkowski, Mark X.Clinical Cancer Research (2014), 20 (2), 456-468CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Purpose: Targeting HER2 with multiple HER2-directed therapies represents a promising area of treatment for HER2-pos. cancers. We investigated combining the HER2-directed antibody-drug conjugate trastuzumab emtansine (T-DM1) with the HER2 dimerization inhibitor pertuzumab (Perjeta). Exptl. Design: Drug combination studies with T-DM1 and pertuzumab were performed on cultured tumor cells and in mouse xenograft models of HER2-amplified cancer. In patients with HER2-pos. locally advanced or metastatic breast cancer (mBC), T-DM1 was dose-escalated with a fixed std. pertuzumab dose in a 3+3 phase Ib/II study design. Results: Treatment of HER2-overexpressing tumor cells in vitro with T-DM1 plus pertuzumab resulted in synergistic inhibition of cell proliferation and induction of apoptotic cell death. The presence of the HER3 ligand, heregulin (NRG-1β), reduced the cytotoxic activity of T-DM1 in a subset of breast cancer lines; this effect was reversed by the addn. of pertuzumab. Results from mouse xenograft models showed enhanced antitumor efficacy with T-DM1 and pertuzumab resulting from the unique antitumor activities of each agent. In patients with mBC previously treated with trastuzumab, lapatinib, and chemotherapy, T-DM1 could be dosed at the max. tolerated dose (MTD; 3.6 mg/kg every 3 wk) with std. dose pertuzumab. Adverse events were mostly grade 1 and 2, with indications of clin. activity. Conclusions: Dual targeting of HER2 with the combination of T-DM1 and pertuzumab in cell culture and mouse xenograft models resulted in enhanced antitumor activity. In patients, this combination showed an encouraging safety and tolerability profile with preliminary evidence of efficacy. Clin Cancer Res; 20(2); 456-68. ©2013 AACR.
- 42Lu, 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– 922Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1OlurrO&md5=ee2756eb89baee7a08a95ff9d43eb736Drug interaction potential of trastuzumab emtansine (T-DM1) combined with pertuzumab in patients with HER2-positive metastatic breast cancerLu, Dan; Burris, Howard A., III; Wang, Bei; Dees, E. Claire; Cortes, Javier; Joshi, Amita; Gupta, Manish; Yi, Joo-Hee; Chu, Yu-Waye; Shih, Ted; Fang, Liang; Girish, SandhyaCurrent Drug Metabolism (2012), 13 (7), 911-922CODEN: CDMUBU; ISSN:1389-2002. (Bentham Science Publishers Ltd.)Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate comprised of trastuzumab and the cytotoxic agent DM1 (deriv. of maytansine) linked by a stable linker N-succinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate (SMCC). T-DM1 targets an epitope located at subdomain IV of human epidermal growth factor receptor 2 (HER2). Pertuzumab is a monoclonal antibody that targets an epitope located at subdomain II of HER2, distinct from the epitope recognized by T-DM1. The pharmacokinetics (PK), safety, and efficacy of T-DM1 combined with pertuzumab were studied in a phase 1b/2 trial in 67 patients with HER2-pos., locally advanced or metastatic breast cancer (MBC). The therapeutic protein-drug interaction (TP-DI) potential of T-DM1 plus pertuzumab was evaluated. The PK of T-DM1-related analytes and pertuzumab were compared with historical PK data. The results show that the exposure of T-DM1 and DM1, as estd. by noncompartmental analyses, was comparable with that reported by historical single-agent studies in patients with HER2-pos. MBC. T-DM1 clearance and vol. of distribution in the central compartment, as estd. by population PK anal., were also comparable between this study and historical single-agent studies in patients with HER2-pos. MBC. Summary statistics of pertuzumab trough and maximal exposure (concns. at predose and 15-30 min after the end of infusion at cycle 1 and at steady state) were similar with those obsd. in a representative historical single-agent study with the same dosing regimen. The visual predictive check plot by population simulation further confirmed that T-DM1 did not alter pertuzumab PK. Based on these data and the PK and pharmacodynamic properties of T-DM1 and pertuzumab, the risk of TP-DI appears to be low when T-DM1 and pertuzumab are given together.
- 43Bartsch, R.; Berghoff, A. S.; Preusser, M. Breast cancer brain metastases responding to primary systemic therapy with T-DM1 J. Neuro-Oncol. 2014, 116, 205– 206Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2c%252Fht1SlsA%253D%253D&md5=cb5391468373267301aadd8c16fc8e06Breast cancer brain metastases responding to primary systemic therapy with T-DM1Bartsch Rupert; Berghoff Anna S; Preusser MatthiasJournal of neuro-oncology (2014), 116 (1), 205-6 ISSN:.There is no expanded citation for this reference.
- 44Girish, 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– 1240Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xmt1Kru70%253D&md5=f4b2a4ef4aaa79a96adbb1810a0c44d9Clinical pharmacology of trastuzumab emtansine (T-DM1): an antibody-drug conjugate in development for the treatment of HER2-positive cancerGirish, Sandhya; Gupta, Manish; Wang, Bei; Lu, Dan; Krop, Ian E.; Vogel, Charles L.; Burris, Howard A., III; LoRusso, Patricia M.; Yi, Joo-Hee; Saad, Ola; Tong, Barbara; Chu, Yu-Waye; Holden, Scott; Joshi, AmitaCancer Chemotherapy and Pharmacology (2012), 69 (5), 1229-1240CODEN: CCPHDZ; ISSN:0344-5704. (Springer)Purpose: Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate comprising trastuzumab and DM1, a microtubule polymn. inhibitor, covalently bound via a stable thioether linker. To characterize the pharmacokinetics (PK) of T-DM1 in patients with human epidermal growth factor receptor 2 (HER2)-pos. metastatic breast cancer, data from four studies (TDM3569g, TDM4258g, TDM4374g, and TDM4688g) of single-agent T-DM1 administered at 3.6 mg/kg every 3 wk (q3w) were assessed in aggregate. Methods: Multiple analytes-T-DM1, total trastuzumab (TT), DM1, and key metabolites-were quantified using enzyme-linked immunosorbent assays or liq. chromatog. tandem mass spectrometry. PK parameters of T-DM1, TT, and DM1 exposure were calcd. using std. noncompartmental approaches and correlated to efficacy (objective response rate) and safety (platelet counts, hepatic transaminase concns.). Immunogenicity was evaluated by measuring anti-therapeutic antibodies (ATA) to T-DM1 after repeated dosing using validated bridging antibody electrochemiluminescence or enzyme-linked immunosorbent assays. Results: PK parameters for T-DM1, TT, and DM1 were consistent across studies at cycle 1 and steady state. T-DM1 PK was not affected by residual trastuzumab from prior therapy or circulating extracellular domain of HER2. No significant correlations were obsd. between T-DM1 exposure and efficacy, thrombocytopenia, or increased concns. of transaminases. Across the studies, ATA formation was detected in 4.5% (13/286) of evaluable patients receiving T-DM1 q3w. Conclusions The PK profile of single-agent T-DM1 (3.6 mg/kg q3w) is predictable, well characterized, and unaffected by circulating levels of HER2 extracellular domain or residual trastuzumab. T-DM1 exposure does not correlate with clin. responses or key adverse events.
- 45Lu, 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– 672Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvVSnu7jI&md5=b5b591684a66774da1b9161926217f32An 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 CancerLu, Dan; Joshi, Amita; Wang, Bei; Olsen, Steve; Yi, Joo-Hee; Krop, Ian E.; Burris, Howard A.; Girish, SandhyaClinical Pharmacokinetics (2013), 52 (8), 657-672CODEN: CPKNDH; ISSN:0312-5963. (Springer International Publishing AG)Background and Objective: Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate recently approved by the US Food and Drug Administration for the treatment of human epidermal growth factor receptor 2 (HER2)-pos. metastatic breast cancer previously treated with trastuzumab and taxane chemotherapy. It comprises the microtubule inhibitory cytotoxic agent DM1 conjugated to the HER2-targeted humanized monoclonal antibody trastuzumab via a stable linker. To characterize the pharmacokinetics of T-DM1 in patients with metastatic breast cancer, concns. of multiple analytes were quantified, including serum concns. of T-DM1 conjugate and total trastuzumab (the sum of conjugated and unconjugated trastuzumab), as well as plasma concns. of DM1. The clearance of T-DM1 conjugate is approx. 2 to 3 times faster than its parent antibody, trastuzumab. However, the clearance pathways accounting for this faster clearance rate are unclear. An integrated population pharmacokinetic model that simultaneously fits the pharmacokinetics of T-DM1 conjugate and total trastuzumab can help to elucidate the clearance pathways of T-DM1. The model can also be used to predict total trastuzumab pharmacokinetic profiles based on T-DM1 conjugate pharmacokinetic data and sparse total trastuzumab pharmacokinetic data, thereby reducing the frequency of pharmacokinetic sampling. Methods: T-DM1 conjugate and total trastuzumab serum concn. data, including baseline trastuzumab concns. prior to T-DM1 treatment, from phase I and II studies were used to develop this integrated population pharmacokinetic model. Based on a hypothetical T-DM1 catabolism scheme, two-compartment models for T-DM1 conjugate and trastuzumab were integrated by assuming a one-step deconjugation clearance from T-DM1 conjugate to trastuzumab. The ability of the model to predict the total trastuzumab pharmacokinetic profile based on T-DM1 conjugate pharmacokinetics and various sampling schemes of total trastuzumab pharmacokinetics was assessed to evaluate total trastuzumab sampling schemes. Results: The final model reflects a simplified catabolism scheme of T-DM1, suggesting that T-DM1 clearance pathways include both deconjugation and proteolytic degrdn. The model fits T-DM1 conjugate and total trastuzumab pharmacokinetic data simultaneously. The deconjugation clearance of T-DM1 was estd. to be ∼0.4 L/day. Proteolytic degrdn. clearances for T-DM1 and trastuzumab were similar (∼0.3 L/day). This model accurately predicts total trastuzumab pharmacokinetic profiles based on T-DM1 conjugate pharmacokinetic data and sparse total trastuzumab pharmacokinetic data sampled at preinfusion and end of infusion in cycle 1, and in one addnl. steady state cycle. Conclusions: This semi-mechanistic integrated model links T-DM1 conjugate and total trastuzumab pharmacokinetic data, and supports the inclusion of both proteolytic degrdn. and deconjugation as clearance pathways in the hypothetical T-DM1 catabolism scheme. The model attributes a faster T-DM1 conjugate clearance vs. that of trastuzumab to the presence of a deconjugation process and suggests a similar proteolytic clearance of T-DM1 and trastuzumab. Based on the model and T-DM1 conjugate pharmacokinetic data, a sparse pharmacokinetic sampling scheme for total trastuzumab provides an entire pharmacokinetic profile with similar predictive accuracy to that of a dense pharmacokinetic sampling scheme.
- 46Chudasama, 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– 527Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhtl2iurfP&md5=1d04057bdead10453770c704e5ed28f4Semi-mechanistic Population Pharmacokinetic Model of Multivalent Trastuzumab Emtansine in Patients with Metastatic Breast CancerChudasama, V. L.; Schaedeli Stark, F.; Harrold, J. M.; Tibbitts, J.; Girish, S. R.; Gupta, M.; Frey, N.; Mager, D. E.Clinical Pharmacology & Therapeutics (New York, NY, United States) (2012), 92 (4), 520-527CODEN: CLPTAT; ISSN:0009-9236. (Nature Publishing Group)Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate (ADC) composed of multiple mols. of the antimicrotubule agent DM1 linked to trastuzumab, a humanized anti-human epidermal growth factor receptor 2 (HER2) monoclonal antibody. Pharmacokinetics data from phase I (n = 52) and phase II (n = 111) studies in HER2-pos. metastatic breast cancer patients show a shorter terminal half-life for T-DM1 than for total trastuzumab (TTmAb). In this work, we translated prior preclin. modeling in monkeys to develop a semi-mechanistic population pharmacokinetics model to characterize T-DM1 and TTmAb concn. profiles. A series of transit compartments with the same disposition parameters was used to describe the deconjugation process from higher to lower drug-to-antibody ratios (DARs). The structure could explain the shorter terminal half-life of T-DM1 relative to TTmab. The final model integrates prior knowledge of T-DM1 DARs from preclin. studies and could provide a platform for understanding and characterizing the pharmacokinetics of other ADC systems. Clin. Pharmacol. & Therapeutics (2012); 92 4, 520-527. doi:10.1038/clpt.2012.153.
- 47Bender, 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– 601Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsVentbjN&md5=52d01f2d24af314edf8c54d87304af23A population pharmacokinetic/pharmacodynamic model of thrombocytopenia characterizing the effect of trastuzumab emtansine (T-DM1) on platelet counts in patients with HER2-positive metastatic breast cancerBender, Brendan C.; Schaedeli-Stark, Franziska; Koch, Reinhold; Joshi, Amita; Chu, Yu-Waye; Rugo, Hope; Krop, Ian E.; Girish, Sandhya; Friberg, Lena E.; Gupta, ManishCancer Chemotherapy and Pharmacology (2012), 70 (4), 591-601CODEN: CCPHDZ; ISSN:0344-5704. (Springer)Purpose: Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate in the development for the treatment of human epidermal growth factor receptor 2-pos. cancers. Thrombocytopenia (TCP) is the dose-limiting toxicity of T-DM1. A semimechanistic population pharmacokinetic/pharmacodynamic (PK/PD) model was developed to characterize the effect of T-DM1 on patient platelet counts. Methods: A PK/PD model with transit compartments that mimic platelet development and circulation was fit to concn.-platelet-time course data from two T-DM1 single-agent studies (TDM3569g; N = 52 and TDM4258g; N = 112). NONMEM 7 software was used for model development. Data from a sep. phase II study (TDM4374g; N = 110) were used for model evaluation. Patient baseline characteristics were evaluated as covariates of model PD parameters. Results: The model described the platelet data well and predicted the incidence of grade ≥3 TCP. The model predicted that with T-DM1 3.6 mg/kg given every 3 wk (q3w), the lowest platelet nadir would occur after the first dose. Also predicted was a patient subgroup (46 %) having variable degrees of downward drifting platelet-time profiles, which were predicted to stabilize by the eighth treatment cycle to platelet counts above grade 3 TCP. Baseline characteristics were not significant covariates of PD parameters in the model. Conclusions: This semimechanistic PK/PD model accurately captures the cycle 1 platelet nadir, the downward drift noted in some patient platelet-time profiles, and the ∼8 % incidence of grade ≥3 TCP with T-DM1 3.6 mg/kg q3w. This model supports T-DM1 3.6 mg/kg q3w as a well-tolerated dose with minimal dose delays or redns. for TCP.
- 48Shen, 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– 910Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1OlurrN&md5=faf70e5692e4d6a5a7789a0510705837Catabolic fate and pharmacokinetic characterization of trastuzumab emtansine (T-DM1): an emphasis on preclinical and clinical catabolismShen, Ben-Quan; Bumbaca, Daniela; Saad, Ola; Yue, Qin; Pastuskovas, Cinthia V.; Khojasteh, S. Cyrus; Tibbitts, Jay; Kaur, Surinder; Wang, Bei; Chu, Yu-Waye; LoRusso, Patricia M.; Girish, SandhyaCurrent Drug Metabolism (2012), 13 (7), 901-910CODEN: CDMUBU; ISSN:1389-2002. (Bentham Science Publishers Ltd.)Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate in clin. development for the treatment of human epidermal growth factor receptor 2 (HER2)-pos. cancers. Herein, we describe a series of studies to assess T-DM1 absorption, distribution, metab., and excretion (ADME) in rats as well as to assess human exposure to T-DM1 catabolites. Following administration of unlabeled and radiolabeled T-DM1 in female Sprague Dawley rats as a single dose, plasma, urine, bile and feces were assessed for mass balance, profiling and identification of catabolites. In rats, the major circulating species in plasma was T-DM1, while DM1 concns. were low (1.08 to 15.6 ng/mL). The major catabolites found circulating in rat plasma were DM1, [N-maleimidomethyl] cyclohexane-1-carboxylate-DM1 (MCC-DM1), and Lysine-MCC-DM1. These catabolites identified in rats were also detected in plasma samples from patients with HER2-pos. metastatic breast cancer who received single-agent T-DM1 (3.6 mg/kg every 3 wk) in a phase 2 clin. study. There was no evidence of tissue accumulation in rats or catabolite accumulation in human plasma following multiple dosing. In rats, T-DM1 was distributed nonspecifically to the organs without accumulation. The major pathway of DM1-contg. catabolite elimination in rats was the fecal/biliary route, with up to 80% of radioactivity recovered in the feces and 50% in the bile. The rat T-DM1 ADME profile is likely similar to the human profile, although there may be differences since trastuzumab does not bind the rat HER2- like receptor. Further research is necessary to more fully understand the T-DM1 ADME profile in humans.
- 49Thon, 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– 1984Google ScholarThere is no corresponding record for this reference.
- 50Sendur, M. A.; Aksoy, S.; Altundag, K. Cardiotoxicity of novel HER2-targeted therapies Curr. Med. Res. Opin. 2013, 29, 1015– 1024Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFeqsL%252FN&md5=a5a593ec4ce6bbbb95584eb1e1d38d05Cardiotoxicity of novel HER2-targeted therapiesSendur, Mehmet A. N.; Aksoy, Sercan; Altundag, KadriCurrent Medical Research and Opinion (2013), 29 (8), 1015-1024CODEN: CMROCX; ISSN:0300-7995. (Informa Healthcare)A review. Background: Trastuzumab, an anti-HER2 humanized monoclonal antibody, is the std. treatment for both early and metastatic HER2-pos. breast cancer. In addn. to other chemotherapeutic agents, trastuzumab significantly improves response rate and survival in HER2-pos. early and metastatic breast cancer. Although it is well known that trastuzumab therapy is closely assocd. with both symptomatic and asymptomatic cardiotoxicity, less is known about novel HER2-targeted therapies. The aim of this review is to discuss the cardiac safety data from recent studies of novel anti-HER2 drugs other than trastuzumab. Scope: Novel HER2-targeted therapies showed favorable results in HER2 pos. metastatic breast cancer patients. Pubmed database, ASCO and San Antonio Breast Cancer Symposium Meeting abstrs. were searched until Jan. 2013 using the following search keywords; trastuzumab, trastuzumab cardiotoxicity, HER-2 targeted therapies, lapatinib, pertuzumab, trastuzumab emtansine, afatinib and neratinib; papers which were considered relevant for the aim of this review were selected by the authors. Lapatinib, pertuzumab, T-DM1, neratinib and afatinib mols. are evaluated in the study. Findings: In a comprehensive anal., 3689 lapatinib treated patients enrolled in 49 trials; asymptomatic cardiac events were reported in 53 patients (1.4%) and symptomatic grade III and IV systolic dysfunction was obsd. only in 7 patients (0.2%) treated with lapatinib. In phase I-III trials of pertuzumab, cardiac dysfunction was seen in 4.5-14.5% of patients with pertuzumab treatment and cardiac dysfunction was usually grade I and II. Cardiotoxicity of pertuzumab was usually reported with the trastuzumab combination and no additive cardiotoxicity was reported with addn. of pertuzumab to trastuzumab. T-DM1 had a better safety profile compared to trastuzumab, no significant cardiotoxicity was obsd. with T-DM1 in heavily pre-treated patients. In the EMILIA study, only in 1.7% of patients in the T-DM1 group experienced redn. of left ventricular ejection fraction (LVEF) and grade III LVEF redn. developed only in one patient (0.2%) in the T-DM1 group compared to the lapatinib plus capecitabine group. In phase I-II trials with neratinib no cardiotoxicity was reported whereas cardiotoxicity was seen between 0-5.3% with afatinib treatment. Conclusion: Although cardiac toxicity has been reported as an adverse event for novel HER2-targeted therapies, cardiac dysfunction rate of the novel HER2-targeted therapies is significantly lower than the trastuzumab and combination of these agents with trastuzumab did not significantly increase the cardiac adverse events.
Cited By
This article is cited by 394 publications.
- 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.
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- Mohsin Khan. Nanoparticle-Mediated Cancer Chemotherapy. 2024, 183-216. https://doi.org/10.1007/978-981-97-3545-7_9
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
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.
Recommended Articles
References
This article references 50 other publications.
- 1Colomer, R.; Shamon, L. A.; Tsai, M. S.; Lupu, R. Herceptin: from the bench to the clinic Cancer Invest. 2001, 19, 49– 56There is no corresponding record for this reference.
- 2Pegram, 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– 7492https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXkt1Sqsbo%253D&md5=1fb831bd3fbb62277de025c4a99f383dRational Combinations of Trastuzumab With Chemotherapeutic Drugs Used in the Treatment of Breast CancerPegram, Mark D.; Konecny, Gottfried E.; O'Callaghan, Carminda; Beryt, Malgorzata; Pietras, Richard; Slamon, Dennis J.Journal of the National Cancer Institute (2004), 96 (10), 739-749CODEN: JNCIEQ; ISSN:0027-8874. (Oxford University Press)Trastuzumab, a humanized anti-HER2 antibody, increases the clin. benefit of first-line chemotherapy in patients with metastatic breast cancers that overexpress HER2. We characterized interactions between trastuzumab and chemotherapeutic agents commonly used in the treatment of breast cancer. Multiple drug effect/combination index isobologram anal. was used to study the efficacy of chemotherapeutic drug plus trastuzumab combinations tested against four HER2-overexpressing breast cancer cell lines (SK-BR-3, BT-474, MDA-MB-361, and MDA-MB-453). Combination index values were derived from parameters of the median effect plots, and statistical tests were used to det. whether the mean combination index values at multiple effect levels were statistically significantly different from a combination index value of 1.0. Values less than 1.0 indicate synergistic interactions, values greater than 1.0 indicate antagonistic interactions, and values equal to 1.0 indicate additive interactions. At a wide range of clin. achievable drug concns., synergistic interactions were obsd. in all four breast cancer cell lines for trastuzumab plus carboplatin (mean combination index values ranged from 0.32 [P<.001] to 0.53 [P<.001]), 4-hydroxycyclophosphamide (mean combination index values ranged from 0.38 [P<.001] to 0.73 [P = .010]), docetaxel (mean combination index values ranged from 0.30 [P<.001] to 0.62 [P<.001]), and vinorelbine (mean combination index values ranged from 0.24 [P<.001] to 0.78 [P<.034]). Additive interactions were obsd. in all four cell lines with trastuzumab plus doxorubicin, epirubicin, and paclitaxel. Interactions between trastuzumab and gemcitabine were synergistic at low concns. of gemcitabine and antagonistic at high concns. A synergistic interaction was obsd. with a three-drug combination of docetaxel plus carboplatin plus trastuzumab in SK-BR-3 cells (mean combination index value = 0.09; P<.001). Consistent synergistic interactions of trastuzumab plus carboplatin, 4-hydroxycyclophosphamide, docetaxel, or vinorelbine across a wide range of clin. relevant concns. in HER2-overexpressing breast cancer cells indicate that these are rational combinations to test in human clin. trials.
- 3Shepard, 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– 1273https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXmsFent7w%253D&md5=8c323a0c6d081fc00a2b3328a8711f75Monoclonal antibody therapy of human cancer: taking the HER2 protooncogene to the clinicShepard, H. Michael; Lewis, Gail D.; Sarup, Jay C.; Fendly, Brian M.; Maneval, Daniel; Mordenti, Joyce; Figari, Irene; Kotts, Claire E.; Palladino, Michael A., Jr.; et al.Journal of Clinical Immunology (1991), 11 (3), 117-27CODEN: JCIMDO; ISSN:0271-9142.A review, with 35 refs., on: the HER2 protoncogene and human cancer; derivation of muMAb 4D5; in vivo preclin. efficacy; and mechanism of action.
- 4Spector, 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– 58474https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtVWitro%253D&md5=d54f228c3b69c973238fb1279ee8572dUnderstanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancerSpector, Neil L.; Blackwell, Kimberly L.Journal of Clinical Oncology (2009), 27 (34), 5838-5847CODEN: JCONDN; ISSN:0732-183X. (American Society of Clinical Oncology)A review. Purpose: Targeted therapy with the humanized monoclonal antibody trastuzumab has become a mainstay for human epidermal growth factor receptor 2 (HER2) -pos. breast cancer (BC). The mechanisms of action of trastuzumab have not been fully elucidated, and data available to date are reviewed here. The impact of the mechanisms of action on clin. benefit also is discussed. Methods: An extensive literature review of trastuzumab and proposed mechanisms of action was performed. Results: At least five potential extracellular and intracellular antitumor mechanisms of trastuzumab have been identified in the preclin. setting. These include activation of antibody-dependent cellular cytotoxicity, inhibition of extracellular domain cleavage, abrogation of intracellular signaling, redn. of angiogenesis, and decreased DNA repair. These effects lead to tumor cell stasis and/or death. Clin. benefit from trastuzumab-based therapy in both early and advanced BC has been demonstrated. The benefit of trastuzumab use beyond progression has also been shown, which indicates the need for continuous suppression of the HER2 pathway. Targeting both HER2, with various approaches, and other pathways may enhance the clin. benefit obsd. with trastuzumab and overcome potential resistance. Novel combinations include pertuzumab (a HER2 dimerization inhibitor), lapatinib (a HER1/HER2 tyrosine kinase inhibitor), bevacizumab (an antiangiogenic agent), tanespimycin (a heat shock protein inhibitor), antiestrogen therapies, and an antibody-drug conjugate (trastuzumab-DM1). Conclusion: Trastuzumab is the foundation of care for patients with HER2-pos. BC. Emerging data from studies of other targeted agents may provide alternative treatment combinations to maximize the clin. benefit from trastuzumab and prevent or delay resistance. The continued development of trastuzumab highlights promising treatment approaches for the future.
- 5Slamon, 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– 7925https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXisVGktrc%253D&md5=81112f11ffc66fc3a898b61e104322b6Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2Slamon, Dennis J.; Leyland-Jones, Brian; Shak, Steven; Fuchs, Hank; Paton, Virginia; Bajamonde, Alex; Fleming, Thomas; Eiermann, Wolfgang; Wolter, Janet; Pegram, Mark; Baselga, Jose; Norton, LarryNew England Journal of Medicine (2001), 344 (11), 783-792CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)The HER2 gene, which encodes the growth factor receptor HER2, is amplified and HER2 is overexpressed in 25 to 30 % of breast cancers, increasing the aggressiveness of the tumor. We evaluated the efficacy and safety of trastuzumab, a recombinant monoclonal antibody against HER2, in women with metastatic breast cancer that overexpressed HER2. We randomly assigned 234 patients to receive std. chemotherapy plus trastuzumab. Patients who had not previously received adjuvant (postoperative) therapy with an anthracycline were treated with doxorubicin (or epirubicin in the case of 36 women) and cyclophosphamide with (143 women) or without trastuzumab (138 women). Patients who had previously received adjuvant anthracycline were treated with paclitaxel alone (96 women) or paclitaxel with trastuzumab (92 women). The addn. of trastuzumab to chemotherapy was assocd. with a longer time to disease progression (median, 7.4 vs. 4.6 mo; P<0.001), a higher rate of objective response (50 % vs. 32 %, P<0.001), a longer duration of response (median, 9.1 vs. 6.1 mo; P<0.001), a lower rate of death at 1 yr (22 % vs. 33 %, P=0.008), longer survival (median survival, 25.1 vs. 20.3 mo; P=0.046), and a 20 % redn. in the risk of death. The most important adverse event was cardiac dysfunction, which occurred in 27 % of the group given an anthracycline, cyclophosphamide, and trastuzumab; 8 % of the group given an anthracycline and cyclophosphamide alone; 13 % of the group given paclitaxel and trastuzumab; and 1 % of the group given paclitaxel alone. Although the cardiotoxicity was potentially severe and, in some cases, life-threatening, the symptoms generally improved with std. medical management. Trastuzumab increases the clin. benefit of first-line chemotherapy in metastatic breast cancer that overexpresses HER2.
- 6Nahta, R.; Esteva, F. J. HER-2-targeted therapy: lessons learned and future directions Clin. Cancer Res. 2003, 9, 5078– 50846https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXovVOgtb4%253D&md5=0c48944fbc257ee35c8197d04b31595fHER-2-Targeted Therapy: Lessons Learned and Future DirectionsNahta, Rita; Esteva, Francisco J.Clinical Cancer Research (2003), 9 (14), 5078-5084CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)A review. HER-2 is overexpressed in 20-25% of invasive breast cancers and is assocd. with an aggressive tumor phenotype and reduced survival rates. The HER-2 status of a tumor is the crit. determinant of response to the HER-2-targeted antibody trastuzumab. Thus, accurate assessment of HER-2 expression levels is essential for identifying breast cancer patients who will benefit from HER-2-targeted therapy. Trastuzumab combined with chemotherapy increases response rates, time to progression, and survival. However, the majority of cancers that initially respond to trastuzumab begin to progress again within 1 yr. This minireview describes HER-2 targeting strategies currently in use or in stages of development for the treatment of breast cancer.
- 7Metzger-Filho, O.; Winer, E. P.; Krop, I. Pertuzumab: optimizing HER2 blockade Clin. Cancer Res. 2013, 19, 5552– 55567https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1ahurzK&md5=a72443fe22216b25ad4b438bd604f44ePertuzumab: Optimizing HER2 BlockadeMetzger-Filho, Otto; Winer, Eric P.; Krop, IanClinical Cancer Research (2013), 19 (20), 5552-5556CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)A review. Pertuzumab has been approved by the U.S. Food and Drug Administration for use in combination with trastuzumab and docetaxel for the first-line treatment of patients with advanced HER2-pos. (HER2+) breast cancer. Pertuzumab is a recombinant humanized monoclonal antibody that binds to the extracellular dimerization domain II of HER2 and inhibits heterodimerization of HER2 with other HER family members, including EGF receptor (EGFR), HER3, and HER4. The HER2-HER3 heterodimer is a robust activator of phosphoinositide 3-kinase (PI3K) pathway signaling and functions as the most transforming and mitogenic of the receptor complexes formed by the HER family of proteins; thus, blockade of HER2-HER3 likely represents the most relevant action of pertuzumab. In the seminal phase III study, patients with HER2+ metastatic breast cancer were randomized to receive trastuzumab and docetaxel, with or without pertuzumab: Addn. of pertuzumab significantly prolonged progression-free survival with an increase of 6.1 mo (12.4 vs. 18.5 mo, resp.). In a subsequent anal. with 30 mo of median follow-up, pertuzumab conferred a 34% redn. in the risk of mortality. Here, we review the mechanism of action of pertuzumab, the rationale for combining it with trastuzumab/pertuzumab, clin. data, and future directions for this work. Clin Cancer Res; 19(20); 5552-6. ©2013 AACR.
- 8Chari, R. V.; Miller, M. L.; Widdison, W. C. Antibody–drug conjugates: an emerging concept in cancer therapy Angew. Chem., Int. Ed. 2014, 53, 3796– 3827There is no corresponding record for this reference.
- 9Blattler, W. A.; Chari, R. V. J. Methods of treatment using anti-ERBB antibody–maytansinoid conjugates. U.S. Patent 8,337,856, 2012.There is no corresponding record for this reference.
- 10Sedlacek, H. H. Antibodies as Carriers of Cytotoxicity; Karger: Basel, Switzerland, 1992; pp 1– 145.There is no corresponding record for this reference.
- 11Chari Ravi, V. J. Targeted cancer therapy: conferring specificity to cytotoxic drugs Acc. Chem. Res. 2008, 41, 98– 107There is no corresponding record for this reference.
- 12Kupchan, 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– 2357There is no corresponding record for this reference.
- 13Widdison, 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– 440813https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XlvFent7o%253D&md5=a06c19d2d5166b0ca806babf9624c48bSemisynthetic Maytansine Analogs for the Targeted Treatment of CancerWiddison, Wayne C.; Wilhelm, Sharon D.; Cavanagh, Emily E.; Whiteman, Kathleen R.; Leece, Barbara A.; Kovtun, Yelena; Goldmacher, Victor S.; Xie, Hongsheng; Steeves, Rita M.; Lutz, Robert J.; Zhao, Robert; Wang, Lintao; Blaettler, Walter A.; Chari, Ravi V. J.Journal of Medicinal Chemistry (2006), 49 (14), 4392-4408CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Maytansine, a highly cytotoxic natural product, failed as an anticancer agent in human clin. trials because of unacceptable systemic toxicity. The potent cell killing ability of maytansine can be used in a targeted delivery approach for the selective destruction of cancer cells. A series of new maytansinoids, bearing a disulfide or thiol substituent were synthesized. The chain length of the ester side chain and the degree of steric hindrance on the carbon atom bearing the thiol substituent were varied. Several of these maytansinoids were found to be even more potent in vitro than maytansine. The targeted delivery of these maytansinoids, using monoclonal antibodies, resulted in a high, specific killing of the targeted cells in vitro and remarkable antitumor activity in vivo.
- 14Issell, B. F.; Crooke, S. T. Maytansine Cancer Treat. Rev. 1978, 5, 199– 20714https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaE1M7gsFektw%253D%253D&md5=73ec733c0581edc2085d87aa26291f26MaytansineIssell B F; Crooke S TCancer treatment reviews (1978), 5 (4), 199-207 ISSN:0305-7372.There is no expanded citation for this reference.
- 15Kupchan, 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– 37There is no corresponding record for this reference.
- 16Chari, 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– 13116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38Xht1Snu7s%253D&md5=33e7613da2a71800fc13e0b2bc6e21f1Immunoconjugates containing novel maytansinoids: promising anticancer drugsChari, Ravi V. J.; Martell, Bridget A.; Gross, Jonathan L.; Cook, Sherrilyn B.; Shah, Sudhir A.; Blattler, Walter A.; McKenzie, Sara J.; Goldmacher, Victor S.Cancer Research (1992), 52 (1), 127-31CODEN: CNREA8; ISSN:0008-5472.The potential of immunoconjugates of cytotoxic drugs for the treatment of cancer has not yet been realized owing to the difficulty of delivering therapeutic concns. of these drugs to the target cells. In an effort to overcome this problem the authors have synthesized maytansinoids that have 100- to 1000-fold higher cytotoxic potency than clin. used anticancer drugs. These maytansinoids are linked to antibodies via disulfide bonds, which ensures the release of fully active drug inside the cells. The conjugates show high antigen-specific cytotoxicity for cultured human cancer cells (50% inhibiting concn., 10 to 40 pM), low systemic toxicity in mice, and good pharmacokinetic behavior.
- 17Kellogg, 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– 72717https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjsFKnsro%253D&md5=ef2969ceaaea23e81da2edf06f62ea7eDisulfide-linked antibody-maytansinoid conjugates: optimization of in vivo activity by varying the steric hindrance at carbon atoms adjacent to the disulfide linkageKellogg, Brenda A.; Garrett, Lisa; Kovtun, Yelena; Lai, Katharine C.; Leece, Barbara; Miller, Michael; Payne, Gillian; Steeves, Rita; Whiteman, Kathleen R.; Widdison, Wayne; Xie, Hongsheng; Singh, Rajeeva; Chari, Ravi V. J.; Lambert, John M.; Lutz, Robert J.Bioconjugate Chemistry (2011), 22 (4), 717-727CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)In this report, we describe the synthesis of a panel of disulfide-linked huC242 (anti-CanAg) antibody maytansinoid conjugates (AMCs), which have varying levels of steric hindrance around the disulfide bond, in order to investigate the relationship between stability to redn. of the disulfide linker and antitumor activity of the conjugate in vivo. The conjugates were first tested for stability to redn. by dithiothreitol in vitro and for plasma stability in CD1 mice. It was found that the conjugates having the more sterically hindered disulfide linkages were more stable to reductive cleavage of the maytansinoid in both settings. When the panel of conjugates was tested for in vivo efficacy in two human colon cancer xenograft models in SCID mice, it was found that the conjugate with intermediate disulfide bond stability having two Me groups on the maytansinoid side of the disulfide bond and no Me groups on the linker side of the disulfide bond (huC242-SPDB-DM4) displayed the best efficacy. The ranking of in vivo efficacies of the conjugates was not predicted by their in vitro potencies, since all conjugates were highly active in vitro, including a huC242-SMCC-DM1 conjugate with a noncleavable linkage which showed only marginal activity in vivo. These data suggest that factors in addn. to intrinsic conjugate potency and conjugate half-life in plasma influence the magnitude of antitumor activity obsd. for an AMC in vivo. We provide evidence that bystander killing of neighboring nontargeted tumor cells by diffusible cytotoxic metabolites produced from target cell processing of disulfide-linked antibody-maytansinoid conjugates may be one addnl. factor contributing to the activity of these conjugates in vivo.
- 18Wakankar, A.; Chen, Y.; Gokarn, Y.; Jacobson, F. S. Analytical methods for physicochemical characterization of antibody drug conjugates mAbs 2011, 3, 161– 172There is no corresponding record for this reference.
- 19Wakankar, 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– 159519https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXpvVyltbk%253D&md5=6c67fb904a35f97adfeb62ec69b7d4bePhysicochemical Stability of the Antibody-Drug Conjugate Trastuzumab-DM1: Changes due to Modification and Conjugation ProcessesWakankar, Aditya A.; Feeney, Maria B.; Rivera, Javier; Chen, Yan; Kim, Michael; Sharma, Vikas K.; Wang, Y. JohnBioconjugate Chemistry (2010), 21 (9), 1588-1595CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)In the manuf. of the antibody-drug conjugate Trastuzumab-DM1 (T-DM1), the lysine residues on the antibody trastuzumab (Tmab) are modified to form the intermediate Tmab-MCC (T-MCC) and then conjugated with the drug DM1. Our goal is to understand the effects of modification and conjugation steps on the physicochem. stability of the antibody. The structural stability of Tmab relative to its modified and conjugated forms was assessed, employing thermally induced stress conditions to formulations contg. Tmab, T-MCC, and T-DM1. DSC, SEC, CE-SDS, and LC-MS were used to study the stability of Tmab, T-MCC, and T-DM1 to thermal stress. The DSC thermograms show a decrease in melting temp. for the CH2 transition, in the order Tmab > T-MCC > T-DM1. As per SEC anal., a significant increase in level of aggregation was detected in T-MCC (∼32%) and T-DM1 (∼5%) after 14 days at 40°. Tmab did not show significant aggregate formation. CE-SDS and LC-MS data demonstrate that the aggregation in the case of T-MCC is largely covalent and involves mechanisms other than formation of intermol. disulfide crosslinks. The aggregation obsd. for T-MCC was significantly inhibited upon addn. of amino acids with nucleophilic side chains contg. thiol (Cys) and hydroxyl moieties (Ser, Tyr). The covalent aggregation obsd. for T-MCC and the ability of nucleophilic amino acids, particularly Cys, to inhibit it indicate that the maleimide moiety in the MCC linker may react to form intermol. covalent crosslinks between T-MCC mols., possibly through a Michael addn. mechanism. In addn., DSC results demonstrate that the conjugation of the drug moiety DM1 to Tmab results in destabilization of the CH2 domain of the antibody.
- 20Lewis 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– 929020https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlOmt7rK&md5=e86e8687d5f8884f8e28514299dcbbf1Targeting HER2-Positive Breast Cancer with Trastuzumab-DM1, an Antibody-Cytotoxic Drug ConjugateLewis Phillips, Gail D.; Li, Guangmin; Dugger, Debra L.; Crocker, Lisa M.; Parsons, Kathryn L.; Mai, Elaine; Blaettler, Walter A.; Lambert, John M.; Chari, Ravi V. J.; Lutz, Robert J.; Wong, Wai Lee T.; Jacobson, Frederic S.; Koeppen, Hartmut; Schwall, Ralph H.; Kenkare-Mitra, Sara R.; Spencer, Susan D.; Sliwkowski, Mark X.Cancer Research (2008), 68 (22), 9280-9290CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)HER2 is a validated target in breast cancer therapy. Two drugs are currently approved for HER2-pos. breast cancer: trastuzumab (Herceptin), introduced in 1998, and lapatinib (Tykerb), in 2007. Despite these advances, some patients progress through therapy and succumb to their disease. A variation on antibody-targeted therapy is utilization of antibodies to deliver cytotoxic agents specifically to antigen-expressing tumors. We detd. in vitro and in vivo efficacy, pharmacokinetics, and toxicity of trastuzumab-maytansinoid (microtubule-depolymg. agents) conjugates using disulfide and thioether linkers. Antiproliferative effects of trastuzumab-maytansinoid conjugates were evaluated on cultured normal and tumor cells. In vivo activity was detd. in mouse breast cancer models, and toxicity was assessed in rats as measured by body wt. loss. Surprisingly, trastuzumab linked to DM1 through a nonreducible thioether linkage (SMCC), displayed superior activity compared with unconjugated trastuzumab or trastuzumab linked to other maytansinoids through disulfide linkers. Serum concns. of trastuzumab-MCC-DM1 remained elevated compared with other conjugates, and toxicity in rats was negligible compared with free DM1 or trastuzumab linked to DM1 through a reducible linker. Potent activity was obsd. on all HER2-overexpressing tumor cells, whereas nontransformed cells and tumor cell lines with normal HER2 expression were unaffected. In addn., trastuzumab-DM1 was active on HER2-overexpressing, trastuzumab-refractory tumors. In summary, trastuzumab-DM1 shows greater activity compared with nonconjugated trastuzumab while maintaining selectivity for HER2-overexpressing tumor cells. Because trastuzumab linked to DM1 through a nonreducible linker offers improved efficacy and pharmacokinetics and reduced toxicity over the reducible disulfide linkers evaluated, trastuzumab-MCC-DM1 was selected for clin. development.
- 21Erickson, 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– 114221https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xms1yrtLs%253D&md5=e7485e8164a5bf3210f6c68f174df112The Effect of Different Linkers on Target Cell Catabolism and Pharmacokinetics/Pharmacodynamics of Trastuzumab Maytansinoid ConjugatesErickson, Hans K.; Lewis Phillips, Gail D.; Leipold, Douglas D.; Provenzano, Carmela A.; Mai, Elaine; Johnson, Holly A.; Gunter, Bert; Audette, Charlene A.; Gupta, Manish; Pinkas, Jan; Tibbitts, JayMolecular Cancer Therapeutics (2012), 11 (5), 1133-1142CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate consisting of the anti-HER2 antibody trastuzumab linked via a nonreducible thioether linker to the maytansinoid antitubulin agent DM1. T-DM1 has shown favorable safety and efficacy in patients with HER2-pos. metastatic breast cancer. In previous animal studies, T-DM1 exhibited better pharmacokinetics (PK) and slightly more efficacy than several disulfide-linked versions. The efficacy findings are unique, as other disulfide-linked antibody-drug conjugates (ADC) have shown greater efficacy than thioether-linked designs. To explore this further, the in vitro and in vivo activity, PK, and target cell activation of T-DM1 and the disulfide-linked T-SPP-DM1 were examd. Both ADCs showed high in vitro potency, with T-DM1 displaying greater potency in two of four breast cancer cell lines. In vitro target cell processing of T-DM1 and T-SPP-DM1 produced lysine-N.vepsiln.-MCC-DM1, and lysine-N.vepsiln.-SPP-DM1 and DM1, resp.; in vivo studies confirmed these results. The in vitro processing rates for the two conjugate to their resp. catabolites were similar. In vivo, the potencies of the conjugates were similar, and T-SPP-DM1 had a faster plasma clearance than T-DM1. Slower T-DM1 clearance translated to higher overall tumor concns. (conjugate plus catabolites), but unexpectedly, similar levels of tumor catabolite. These results indicate that, although the ADC linker can have clear impact on the PK and the chem. nature of the catabolites formed, both linkers seem to offer the same payload delivery to the tumor. Mol Cancer Ther; 11(5); 1133-42.
- 22Junttila, 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– 35622https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXnslehsbo%253D&md5=f3b04d9b9bb099007f338c619dd65f00Trastuzumab-DM1 (T-DM1) retains all the mechanisms of action of trastuzumab and efficiently inhibits growth of lapatinib insensitive breast cancerJunttila, Teemu T.; Li, Guangmin; Parsons, Kathryn; Phillips, Gail Lewis; Sliwkowski, Mark X.Breast Cancer Research and Treatment (2011), 128 (2), 347-356CODEN: BCTRD6; ISSN:0167-6806. (Springer)Trastuzumab (Herceptin) is currently used as a treatment for patients whose breast tumors overexpress HER2/ErbB2. Trastuzumab-DM1 (T-DM1, trastuzumab emtansine) is designed to combine the clin. benefits of trastuzumab with a potent microtubule-disrupting drug, DM1 (a maytansine deriv.). Currently T-DM1 is being tested in multiple clin. trials. The mechanisms of action for trastuzumab include inhibition of PI3K/AKT signaling pathway, inhibition of HER-2 shedding and Fcγ receptor mediated engagement of immune cells, which may result in antibody-dependent cellular cytotoxicity (ADCC). Here we report that T-DM1 retains the mechanisms of action of unconjugated trastuzumab and is active against lapatinib resistant cell lines and tumors.
- 23Barok, 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– 17923https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXlsV2nt7k%253D&md5=b3c549e901da81d69ccdf85af4808310Trastuzumab-DM1 is highly effective in preclinical models of HER2-positive gastric cancerBarok, Mark; Tanner, Minna; Koeninki, Katri; Isola, JormaCancer Letters (New York, NY, United States) (2011), 306 (2), 171-179CODEN: CALEDQ; ISSN:0304-3835. (Elsevier)Background: A novel antibody-drug conjugate (trastuzumab-DM1, T-DM1) is currently in clin. trials for patients with trastuzumab resistant HER2-pos. breast cancer. Since no clin. data is available from gastric cancer, we studied T-DM1 on HER2-pos. human gastric cancer cells and xenograft tumors. Methods: Effects of T-DM1 were studied in four HER2-pos. gastric cancer cell lines (N-87, OE-19, SNU-216 and MKN-7) in vitro. Xenograft tumors from N-87 and OE-19 were studied to det. the effect of T-DM1 in vivo. Results: T-DM1 was found more effective than trastuzumab in N-87 and OE-19, and moderately effective in MKN-7 cells. On SNU-216 cells both trastuzumab and T-DM1 showed limited efficacy. In xenograft tumor expts., complete pathol. response was obsd. in all OE-19 xenografted mice and in half of the N-87 xenografted mice. The results were equally good irresp. of the tumor burden at therapy initiation, or preceding trastuzumab treatment. T-DM1 treatment showed direct effects (apoptotic cell death and aberrant mitosis) as well as it mediated antibody-dependent cellular cytotoxcity (ADCC). Conclusions: T-DM1 showed a promising anti-tumor effect in HER2-pos. gastric cancer cell lines in vitro and in vivo, even in tumors which had developed resistance to trastuzumab. T-DM1 therapy may warrant clin. trials for HER2-pos. gastric cancer patients.
- 24Kovtun, 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– 2537There is no corresponding record for this reference.
- 25Guo, 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 618There is no corresponding record for this reference.
- 26Jumbe, 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– 24226https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnslyktLw%253D&md5=28bac6e680530a3aeba7b44607669c8cModeling the efficacy of trastuzumab-DM1, an antibody drug conjugate, in miceJumbe, Nelson L.; Xin, Yan; Leipold, Douglas D.; Crocker, Lisa; Dugger, Debra; Mai, Elaine; Sliwkowski, Mark X.; Fielder, Paul J.; Tibbitts, JayJournal of Pharmacokinetics and Pharmacodynamics (2010), 37 (3), 221-242CODEN: JPPOAH; ISSN:1567-567X. (Springer)Trastuzumab-DM1 (T-DM1) is a novel antibody-drug conjugate under investigation for the treatment of human epidermal growth factor receptor 2 (HER2)-pos. metastatic breast cancer. One challenge in oncol. drug development is detg. the optimal dose and treatment schedule. A novel dose regimen-finding strategy was developed for T-DM1 using exptl. data and pharmacokinetic/pharmacodynamic modeling. To characterize the disposition of T-DM1, pharmacokinetic studies were conducted in athymic nude and beige nude mice. The pharmacokinetics of T-DM1 were described well by a two-compartment model. Tumor response data were obtained from single-dose, multiple-dose and time-dose-fractionation studies of T-DM1 in animal models of HER2-pos. breast cancer, specifically engineered to be insensitive to trastuzumab. A sequential population-based pharmacokinetic/pharmacodynamic modeling approach was developed to describe the anti-tumor activity of T-DM1. A cell-cycle-phase nonspecific tumor cell kill model incorporating transit compartments captured well the features of tumor growth and the activity of T-DM1. Key findings of the model were that tumor cell growth rate played a significant role in the sensitivity of tumors to T-DM1; anti-tumor activity was schedule independent; and tumor response was linked to the ratio of exposure to a concn. required for tumor stasis.
- 27Kaur, 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– 226There is no corresponding record for this reference.
- 28Poon, 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– 31328https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslGlsbbO&md5=f2ae6f6fc865d8ad027752e1301be3ccPreclinical safety profile of trastuzumab emtansine (T-DM1): Mechanism of action of its cytotoxic component retained with improved tolerabilityPoon, Kirsten Achilles; Flagella, Kelly; Beyer, Joseph; Tibbitts, Jay; Kaur, Surinder; Saad, Ola; Yi, Joo-Hee; Girish, Sandhya; Dybdal, Noel; Reynolds, TheresaToxicology and Applied Pharmacology (2013), 273 (2), 298-313CODEN: TXAPA9; ISSN:0041-008X. (Elsevier Inc.)Trastuzumab emtansine (T-DM1) is the first antibody-drug conjugate (ADC) approved for patients with human epidermal growth factor receptor 2 (HER2)-pos. metastatic breast cancer. The therapeutic premise of ADCs is based on the hypothesis that targeted delivery of potent cytotoxic drugs to tumors will provide better tolerability and efficacy compared with non-targeted delivery, where poor tolerability can limit efficacious doses. Here, we present results from preclin. studies characterizing the toxicity profile of T-DM1, including limited assessment of unconjugated DM1. T-DM1 binds primate ErbB2 and human HER2 but not the rodent homolog c-neu. Therefore, antigen-dependent and non-antigen-dependent toxicity was evaluated in monkeys and rats, resp., in both single- and repeat-dose studies; toxicity of DM1 was assessed in rats only. T-DM1 was well tolerated at doses up to 40 mg/kg (~ 4400 μg DM1/m2) and 30 mg/kg (~ 6000 μg DM1/m2) in rats and monkeys, resp. In contrast, DM1 was only tolerated up to 0.2 mg/kg (1600 μg DM1/m2). This suggests that at least two-fold higher doses of the cytotoxic agent are tolerated in T-DM1, supporting the premise of ADCs to improve the therapeutic index. In addn., T-DM1 and DM1 safety profiles were similar and consistent with the mechanism of action of DM1 (i.e., microtubule disruption). Findings included hepatic, bone marrow/hematol. (primarily platelet), lymphoid organ, and neuronal toxicities, and increased nos. of cells of epithelial and phagocytic origin in metaphase arrest. These adverse effects did not worsen with chronic dosing in monkeys and are consistent with those reported in T-DM1-treated patients to date.
- 29Sun, 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– 73529https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXivFyitL4%253D&md5=042dafa46e287f5a8a76af8714c124ceDesign of Antibody-Maytansinoid Conjugates Allows for Efficient Detoxification via Liver MetabolismSun, Xiuxia; Widdison, Wayne; Mayo, Michele; Wilhelm, Sharon; Leece, Barbara; Chari, Ravi; Singh, Rajeeva; Erickson, HansBioconjugate Chemistry (2011), 22 (4), 728-735CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Antibody-maytansinoid conjugates (AMCs) are targeted chemotherapeutic agents consisting of a potent microtubule-depolymg. maytansinoid (DM1 or DM4) attached to lysine residues of a monoclonal antibody (mAb) using an uncleavable thioether linker or a stable disulfide linker. Most of the administered dose of an antibody-based therapeutic is slowly catabolized by the liver and other tissues of the reticuloendothelial system. Maytansinoids released from an AMC during this catabolic process could potentially be a source of toxicity. To investigate this, we isolated and identified liver metabolites in mice for three different [3H]AMCs with structures similar to those currently undergoing evaluation in the clinic. We then synthesized each metabolite to confirm the identification and assessed their cytotoxic potencies when added extracellularly. We found that the uncleavable mAb-SMCC-[3H]DM1 conjugate was degraded to a single major maytansinoid metabolite, lysine-SMCC-[3H]DM1, that was nearly 50-fold less cytotoxic than maytansine. The two disulfide-linked conjugates, mAb-SPP-[3H]DM1 and mAb-SPDB-[3H]DM4, were also catabolized to the analogous lysine-linked maytansinoid metabolites. However, subsequent redn., S-methylation, and NADPH-dependent oxidn. steps in the liver yielded the corresponding S-Me sulfoxide and S-Me sulfone derivs. The cytotoxic potencies of the oxidized maytansinoids toward several human carcinoma cell lines were 5- to 50-fold less potent than maytansine. Our results suggest that liver plays an important role in the detoxification of both cleavable and uncleavable AMCs.
- 30Fishkin, 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– 10754There is no corresponding record for this reference.
- 31Krop, 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– 270431https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXoslykt7c%253D&md5=98ba29a289f4ac0ea38c37a9b6bd0c5dPhase I study of trastuzumab-DM1, an HER2 antibody-drug conjugate, given every 3 weeks to patients with HER2-positive metastatic breast cancerKrop, Ian E.; Beeram, Muralidhar; Modi, Shanu; Jones, Suzanne F.; Holden, Scott N.; Yu, Wei; Girish, Sandhy; Tibbitts, Jay; Yi, Joo-Hee; Sliwkowski, Mark X.; Jacobson, Fred; Lutzker, Stuart G.; Burris, Howard A.Journal of Clinical Oncology (2010), 28 (16), 2698-2704CODEN: JCONDN; ISSN:0732-183X. (American Society of Clinical Oncology)Purpose: Trastuzumab-DM1 (T-DM1) is an antibody-drug conjugate that uses trastuzumab to specifically deliver the maytansinoid antimicrotubule agent DM1 to HER2-pos. cells. This first-in-human study of T-DM1 evaluated safety, pharmacokinetics, and preliminary activity of T-DM1 in patients with advanced HER2-pos. breast cancer. Patients and Methods: Successive cohorts of patients who had progressed on trastuzumab-based therapy received escalating doses of T-DM1. Outcomes were assessed by std. solid-tumor phase I methods. Results: Twenty-four patients who had received a median of four prior chemotherapeutic agents for metastatic disease received T-DM1 at 0.3 mg/kg to 4.8 mg/kg on an every-3-wk schedule. Transient thrombocytopenia was dose-limiting at 4.8 mg/kg; the max.-tolerated dose (MTD) was 3.6 mg/kg. The half-life of T-DM1 at the MTD was 3.5 days, with peak DM1 levels < 10 ng/mL. Clearance at doses < 1.2 mg/kg was faster than at higher doses. Common drug-related adverse events (AEs) included grade ≤ 2 thrombocytopenia, elevated transaminases, fatigue, nausea, and anemia. No grade > 1 nausea, vomiting, alopecia, or neuropathy events and no cardiac effects requiring dose modification were reported. The clin. benefit rate (objective response plus stable disease at 6 mo) among 15 patients treated at the MTD was 73%, including five objective responses. The confirmed response rate in patients with measurable disease at the MTD (n = 9) was 44%. Conclusion: At the MTD of 3.6 mg/kg every 3 wk, T-DM1 was assocd. with mild, reversible toxicity and substantial clin. activity in a heavily pretreated population. Phase II and III trials in patients with advanced HER2-pos. breast cancer are under way.
- 32Baselga, J. Phase I and II clinical trials of trastuzumab Ann. Oncol. 2001, 12 (Suppl. 1) S49– S55There is no corresponding record for this reference.
- 33Beeram, 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– 5740There is no corresponding record for this reference.
- 34Burris, 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– 40534https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXivFWmsrc%253D&md5=94f0c939a12a7875590fc02f3f056d65Phase 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 therapyBurris, Howard A., III; Rugo, Hope S.; Vukelja, Svetislava J.; Vogel, Charles L.; Borson, Rachel A.; Limentani, Steven; Tan-Chiu, Elizabeth; Krop, Ian E.; Michaelson, Richard A.; Girish, Sandhya; Amler, Lukas; Zheng, Maoxia; Chu, Yu-Waye; Klencke, Barbara; O'Shaughnessy, Joyce A.Journal of Clinical Oncology (2011), 29 (4), 398-405CODEN: JCONDN; ISSN:0732-183X. (American Society of Clinical Oncology)The antibody-drug conjugate trastuzumab-DM1 (T-DM1) combines the biol. activity of trastuzumab with targeted delivery of a potent antimicrotubule agent, DM1, to human epidermal growth factor receptor 2 (HER2) -overexpressing cancer cells. Based on results from a phase I study that showed T-DM1 was well tolerated at the max.-tolerated dose of 3.6 mg/kg every 3 wk, with evidence of efficacy, in patients with HER2-pos. metastatic breast cancer (MBC) who were previously treated with trastuzumab, we conducted a phase II study to further define the safety and efficacy of T-DM1 in this patient population. This report describes a single-arm phase II study (TDM4258g) that assessed efficacy and safety of i.v. T-DM1 (3.6 mg/kg every 3 wk) in patients with HER2-pos. MBC who had tumor progression after prior treatment with HER2-directed therapy and who had received prior chemotherapy. With a follow-up of ≥ 12 mo among 112 treated patients, the objective response rate by independent assessment was 25.9% (95% CI, 18.4% to 34.4%). Median duration of response was not reached as a result of insufficient events (lower limit of 95% CI, 6.2 mo), and median progression-free survival time was 4.6 mo (95% CI, 3.9 to 8.6 mo). The response rates were higher among patients with confirmed HER2-pos. tumors (immunohistochem. 3+ or fluorescent in situ hybridization pos.) by retrospective central testing (n = 74). Higher response rates were also obsd. in patients whose tumors expressed ≥ median HER2 levels by quant. reverse transcriptase polymerase chain reaction for HER2 expression, compared with patients who had less than median HER2 levels. T-DM1 was well tolerated with no dose-limiting cardiotoxicity. Most adverse events (AEs) were grade 1 or 2; the most frequent grade ≥ 3 AEs were hypokalemia (8.9%), thrombocytopenia (8.0%), and fatigue (4.5%). T-DM1 has robust single-agent activity in patients with heavily pretreated, HER2-pos. MBC and is well tolerated at the recommended phase II dose.
- 35Krop, 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– 324135https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsVKrsL%252FJ&md5=e3c54c63b8a2c6a585818e35c0e4aab9A 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 capecitabineKrop, Ian E.; LoRusso, Patricia; Miller, Kathy D.; Modi, Shanu; Yardley, Denise; Rodriguez, Gladys; Guardino, Ellie; Lu, Michael; Zheng, Maoxia; Girish, Sandhya; Amler, Lukas; Winer, Eric P.; Rugo, Hope S.; Krop, Ian E.; Winer, Eric P.Journal of Clinical Oncology (2012), 30 (26), 3234-3241CODEN: JCONDN; ISSN:0732-183X. (American Society of Clinical Oncology)Purpose. To det. whether the antibody-drug conjugate trastuzumab emtansine (T-DM1), which combines human epidermal growth factor receptor 2 (HER2) -targeted delivery of the potent antimicrotubule agent DM1 with the antitumor activity of trastuzumab, is effective in patients with HER2-pos. metastatic breast cancer (MBC) who have previously received all std. HER2-directed therapies. Patients and Methods. In this single-arm phase II study, T-DM1 3.6 mg/kg was administered i.v. every 3 wk to patients with HER2-pos. MBC who had prior treatment with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine. The primary objectives were overall response rate (ORR) by independent review and safety. Results. Among 110 pretreated patients (median, seven prior agents for MBC; median follow-up, 17.4 mo), the ORR was 34.5% (95% CI, 26.1% to 43.9%), clin. benefit rate was 48.2% (95% CI, 38.8% to 57.9%), median progression-free survival (PFS) was 6.9 mo (95% CI, 4.2 to 8.4 mo), and median duration of response was 7.2 mo (95% CI, 4.6 mo to not estimable). In patients with confirmed HER2-pos. tumors (n = 80 by retrospective central testing), the response rate was 41.3% (95% CI, 30.4% to 52.8%), and median PFS was 7.3 mo (95% CI, 4.6 to 12.3 mo). Most adverse events were grades 1 to 2; the most frequent grade ≥ 3 events were thrombocytopenia (9.1%), fatigue (4.5%), and cellulitis (3.6%). Conclusion. T-DM1 is well tolerated and has single-agent activity in patients with HER2-pos. MBC who have previously received both approved HER2-directed therapies and multiple chemotherapy agents. T-DM1 may be an effective new treatment for this patient population.
- 36Hurvitz, 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– 116336https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXntVyhtbs%253D&md5=95cd6eb05ce016e971b2af745d1b587fPhase II randomized study of trastuzumab emtansine versus trastuzumab plus docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancerHurvitz, Sara A.; Dirix, Luc; Kocsis, Judit; Bianchi, Giulia V.; Lu, Janice; Vinholes, Jeferson; Guardino, Ellie; Song, Chunyan; Tong, Barbara; Ng, Vivian; Chu, Yu-Waye; Perez, Edith A.Journal of Clinical Oncology (2013), 31 (9), 1157-1163CODEN: JCONDN; ISSN:0732-183X. (American Society of Clinical Oncology)Purpose: Trastuzumab emtansine (T-DM1), an antibody-drug conjugate composed of the cytotoxic agent DM1 conjugated to trastuzumab via a stable thioether linker, has shown clin. activity in single-arm studies enrolling patients with human epidermal growth factor receptor 2 (HER2)-pos. metastatic breast cancer (MBC) whose disease had progressed on HER2-targeted therapy in the metastatic setting. Patients and Methods: Patients (N = 137) with HER2-pos. MBC or recurrent locally advanced breast cancer were randomly assigned to trastuzumab plus docetaxel (HT; n = 70) or T-DM1 (n = 67) as first-line treatment until disease progression or unacceptable toxicity. Primary end points were investigator-assessed progression-free survival (PFS) and safety. Key secondary end points included overall survival (OS), objective response rate (ORR), duration of objective response, clin. benefit rate, and quality of life. Results: Median PFS was 9.2 mo with HT and 14.2 mo with T-DM1 (hazard ratio, 0.59; 95% CI, 0.36 to 0.97); median follow-up was approx. 14 mo in both arms. ORR was 58.0% (95% CI, 45.5% to 69.2%) with HT and 64.2% (95% CI, 51.8% to 74.8%) with T-DM1. T-DM1 had a favorable safety profile vs. HT, with fewer grade ≥ 3 adverse events (AEs; 46.4% v 90.9%), AEs leading to treatment discontinuations (7.2% v 34.8%), and serious AEs (20.3% v 25.8%). Preliminary OS results were similar between treatment arms; median follow-up was approx. 23 mo in both arms. Conclusion: In this randomized phase II study, first-line treatment with T-DM1 for patients with HER2-pos. MBC provided a significant improvement in PFS, with a favorable safety profile, vs. HT.
- 37Suter, 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– 386537https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtFanu7zE&md5=9aa4ecb50b0566677b2552e7c645b2f2Trastuzumab-associated cardiac adverse effects in the herceptin adjuvant trialSuter, Thomas M.; Procter, Marion; van Veldhuisen, Dirk J.; Muscholl, Michael; Bergh, Jonas; Carlomagno, Chiara; Perren, Timothy; Passalacqua, Rodolfo; Bighin, Claudia; Klijn, Jan G. M.; Ageev, Fail T.; Hitre, Erika; Greetz, Juergen; Iwata, Hiroji; Knap, Malgorzata; Gnant, Michael; Muehlbauer, Susanne; Spence, Alison; Gelber, Richard D.; Piccart-Gebhart, Martine J.Journal of Clinical Oncology (2007), 25 (25), 3859-3865CODEN: JCONDN; ISSN:0732-183X. (American Society of Clinical Oncology)Purpose: The purpose of this anal. was to investigate trastuzumab-assocd. cardiac adverse effects in breast cancer patients after completion of (neo)adjuvant chemotherapy with or without radiotherapy. Patients and Methods: The Herceptin Adjuvant (HERA) trial is a three-group, multicenter, open-label randomized trial that compared 1 or 2 years of trastuzumab given once every 3 wk with observation in patients with HER-2-pos. breast cancer. Only patients who after completion of (neo)adjuvant chemotherapy with or without radiotherapy had normal left ventricular ejection fraction (LVEF ≥ 55%) were eligible. A repeat LVEF assessment was performed in case of cardiac dysfunction. Results: Data were available for 1,693 patients randomly assigned to 1 yr trastuzumab and 1,693 patients randomly assigned to observation. The incidence of trastuzumab discontinuation due to cardiac disorders was low (4.3%). The incidence of cardiac end points was higher in the trastuzumab group compared with observation (severe congestive heart failure [CHF], 0.60% v 0.00%; symptomatic CHF, 2.15% v 0.12%; confirmed significant LVEF drops, 3.04% v 0.53%). Most patients with cardiac dysfunction recovered in fewer than 6 mo. Patients with trastuzumab-assocd. cardiac dysfunction were treated with higher cumulative doses of doxorubicin (287 mg/m2 v 257 mg/m2) or epirubicin (480 mg/m2 v 422 mg/m2) and had a lower screening LVEF and a higher body mass index. Conclusion: Given the clear benefit in disease-free survival, the low incidence of cardiac adverse events, and the suggestion that cardiac dysfunction might be reversible, adjuvant trastuzumab should be considered for treatment of breast cancer patients who fulfill the HERA trial eligibility criteria.
- 38Verma, 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– 179138https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhs1ekt73M&md5=3be5bac27a81b530442dc8a6b4a1f82eTrastuzumab emtansine for HER2-positive advanced breast cancerVerma, Sunil; Miles, David; Gianni, Luca; Krop, Ian E.; Welslau, Manfred; Baselga, Jose; Pegram, Mark; Oh, Do-Youn; Dieras, Veronique; Guardino, Ellie; Fang, Liang; Lu, Michael W.; Olsen, Steven; Blackwell, KimNew England Journal of Medicine (2012), 367 (19), 1783-1791CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)BACKGROUND: Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate incorporating the human epidermal growth factor receptor 2 (HER2)-targeted antitumor properties of trastuzumab with the cytotoxic activity of the microtubule-inhibitory agent DM1. The antibody and the cytotoxic agent are conjugated by means of a stable linker. METHODS: We randomly assigned patients with HER2-pos. advanced breast cancer, who had previously been treated with trastuzumab and a taxane, to T-DM1 or lapatinib plus capecitabine. The primary end points were progression-free survival (as assessed by independent review), overall survival, and safety. Secondary end points included progression-free survival (investigator-assessed), the objective response rate, and the time to symptom progression. Two interim analyses of overall survival were conducted. RESULTS: Among 991 randomly assigned patients, median progression-free survival as assessed by independent review was 9.6 mo with T-DM1 vs. 6.4 mo with lapatinib plus capecitabine (hazard ratio for progression or death from any cause, 0.65; 95% confidence interval [CI], 0.55 to 0.77; P<0.001), and median overall survival at the second interim anal. crossed the stopping boundary for efficacy (30.9 mo vs. 25.1 mo; hazard ratio for death from any cause, 0.68; 95% CI, 0.55 to 0.85; P<0.001). The objective response rate was higher with T-DM1 (43.6%, vs. 30.8% with lapatinib plus capecitabine; P<0.001); results for all addnl. secondary end points favored T-DM1. Rates of grade 3 or above were higher with lapatinib plus capecitabine than with T-DM1 (57% vs. 41%). The incidences of thrombocytopenia and increased serum aminotransferase levels were higher with T-DM1, whereas the incidences of diarrhea, nausea, vomiting, and palmar-plantar erythrodysesthesia were higher with lapatinib plus capecitabine. CONCLUSIONS: T-DM1 significantly prolonged progression-free and overall survival with less toxicity than lapatinib plus capecitabine in patients with HER2-pos. advanced breast cancer previously treated with trastuzumab and a taxane.
- 39Krop, 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– 69939https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXntl2gs7o%253D&md5=975dd9e786d96360b163c2583aefc4b8Trastuzumab emtansine versus treatment of physician's choice for pretreated HER2-positive advanced breast cancer (TH3RESA): a randomised, open-label, phase 3 trialKrop, Ian E.; Kim, Sung-Bae; Gonzalez-Martin, Antonio; LoRusso, Patricia M.; Ferrero, Jean-Marc; Smitt, Melanie; Yu, Ron; Leung, Abraham C. F.; Wildiers, HansLancet Oncology (2014), 15 (7), 689-699CODEN: LOANBN; ISSN:1470-2045. (Elsevier Ltd.)Patients with progressive disease after two or more HER2-directed regimens for recurrent or metastatic breast cancer have few effective therapeutic options. We aimed to compare trastuzumab emtansine, an antibody-drug conjugate comprising the cytotoxic agent DM1 linked to trastuzumab, with treatment of physician's choice in this population of patients.This randomised, open-label, phase 3 trial took place in medical centers in 22 countries across Europe, North America, South America, and Asia-Pacific. Eligible patients (≥18 years, left ventricular ejection fraction ≥50%, Eastern Cooperative Oncol. Group performance status 0-2) with progressive HER2-pos. advanced breast cancer who had received two or more HER2-directed regimens in the advanced setting, including trastuzumab and lapatinib, and previous taxane therapy in any setting, were randomly assigned (in a 2:1 ratio) to trastuzumab emtansine (3·6 mg/kg i.v. every 21 days) or physician's choice using a permuted block randomization scheme by an interactive voice and web response system. Patients were stratified according to world region (USA vs western Europe vs other), no. of previous regimens (excluding single-agent hormonal therapy) for the treatment of advanced disease (two to three vs more than three), and presence of visceral disease (any vs none). Coprimary endpoints were investigator-assessed progression-free survival (PFS) and overall survival in the intention-to-treat population. We report the final PFS anal. and the first interim overall survival anal. This study is registered with ClinicalTrials.gov, no. NCT01419197.From Sept 14, 2011, to Nov 19, 2012, 602 patients were randomly assigned (404 to trastuzumab emtansine and 198 to physician's choice). At data cutoff (Feb 11, 2013), 44 patients assigned to physician's choice had crossed over to trastuzumab emtansine. After a median follow-up of 7·2 mo (IQR 5·0-10·1 mo) in the trastuzumab emtansine group and 6·5 mo (IQR 4·1-9·7) in the physician's choice group, 219 (54%) patients in the trastuzumab emtansine group and 129 (65%) of patients in the physician's choice group had PFS events. PFS was significantly improved with trastuzumab emtansine compared with physician's choice (median 6·2 mo [95% CI 5·59-6·87] vs 3·3 mo [2·89-4·14]; stratified hazard ratio [HR] 0·528 [0·422-0·661]; p<0·0001). Interim overall survival anal. showed a trend favoring trastuzumab emtansine (stratified HR 0·552 [95% CI 0·369-0·826]; p=0·0034), but the stopping boundary was not crossed. A lower incidence of grade 3 or worse adverse events was reported with trastuzumab emtansine than with physician's choice (130 events [32%] in 403 patients vs 80 events [43%] in 184 patients). Neutropenia (ten [2%] vs 29 [16%]), diarrhoea (three [<1%] vs eight [4%]), and febrile neutropenia (one [<1%] vs seven [4%]) were grade 3 or worse adverse events that were more common in the physician's choice group than in the trastuzumab emtansine group. Thrombocytopenia (19 [5%] vs three [2%]) was the grade 3 or worse adverse event that was more common in the trastuzumab emtansine group. 74 (18%) patients in the trastuzumab emtansine group and 38 (21%) in the physician's choice group reported a serious adverse event. Trastuzumab emtansine should be considered as a new std. for patients with HER2-pos. advanced breast cancer who have previously received trastuzumab and lapatinib.Genentech.
- 40Baselga, 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– 11940https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFSlt7Y%253D&md5=8f8a05c82e4722049923dd069f1863b4Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancerBaselga, Jose; Cortes, Javier; Kim, Sung-Bae; Im, Seock-Ah; Hegg, Roberto; Im, Young-Hyuck; Roman, Laslo; Pedrini, Jose Luiz; Pienkowski, Tadeusz; Knott, Adam; Clark, Emma; Benyunes, Mark C.; Ross, Graham; Swain, Sandra M.New England Journal of Medicine (2012), 366 (2), 109-119CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)BACKGROUND The anti-human epidermal growth factor receptor 2 (HER2) humanized monoclonal antibody trastuzumab improves the outcome in patients with HER2-pos. metastatic breast cancer. However, most cases of advanced disease eventually progress. Pertuzumab, an anti-HER2 humanized monoclonal antibody that inhibits receptor dimerization, has a mechanism of action that is complementary to that of trastuzumab, and combination therapy with the two antibodies has shown promising activity and an acceptable safety profile in phase 2 studies involving patients with HER2-pos. breast cancer. METHODS We randomly assigned 808 patients with HER2-pos. metastatic breast cancer to receive placebo plus trastuzumab plus docetaxel (control group) or pertuzumab plus trastuzumab plus docetaxel (pertuzumab group) as first-line treatment until the time of disease progression or the development of toxic effects that could not be effectively managed. The primary end point was independently assessed progression-free survival. Secondary end points included overall survival, progression-free survival as assessed by the investigator, the objective response rate, and safety. RESULTS The median progression-free survival was 12.4 mo in the control group, as com pared with 18.5 mo in the pertuzumab group (hazard ratio for progression or death, 0.62; 95% confidence interval, 0.51 to 0.75; P < 0.001). The interim anal. of overall survival showed a strong trend in favor of pertuzumab plus trastuzumab plus docetaxel. The safety profile was generally similar in the two groups, with no increase in left ventricular systolic dysfunction; the rates of febrile neutropenia and diarrhea of grade 3 or above were higher in the pertuzumab group than in the control group. CONCLUSIONS The combination of pertuzumab plus trastuzumab plus docetaxel, as compared with placebo plus trastuzumab plus docetaxel, when used as first-line treatment for HER2-pos. metastatic breast cancer, significantly prolonged progression-free survival, with no increase in cardiac toxic effects.
- 41Phillips, 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– 46841https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtFKrs7c%253D&md5=71f2be1c5fccaee1d613d09c0eaf48b5Dual Targeting of HER2-Positive Cancer with Trastuzumab Emtansine and Pertuzumab: Critical Role for Neuregulin Blockade in Antitumor Response to Combination TherapyPhillips, Gail D. Lewis; Fields, Carter T.; Li, Guangmin; Dowbenko, Donald; Schaefer, Gabriele; Miller, Kathy; Andre, Fabrice; Burris, Howard A.; Albain, Kathy S.; Harbeck, Nadia; Dieras, Veronique; Crivellari, Diana; Fang, Liang; Guardino, Ellie; Olsen, Steven R.; Crocker, Lisa M.; Sliwkowski, Mark X.Clinical Cancer Research (2014), 20 (2), 456-468CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Purpose: Targeting HER2 with multiple HER2-directed therapies represents a promising area of treatment for HER2-pos. cancers. We investigated combining the HER2-directed antibody-drug conjugate trastuzumab emtansine (T-DM1) with the HER2 dimerization inhibitor pertuzumab (Perjeta). Exptl. Design: Drug combination studies with T-DM1 and pertuzumab were performed on cultured tumor cells and in mouse xenograft models of HER2-amplified cancer. In patients with HER2-pos. locally advanced or metastatic breast cancer (mBC), T-DM1 was dose-escalated with a fixed std. pertuzumab dose in a 3+3 phase Ib/II study design. Results: Treatment of HER2-overexpressing tumor cells in vitro with T-DM1 plus pertuzumab resulted in synergistic inhibition of cell proliferation and induction of apoptotic cell death. The presence of the HER3 ligand, heregulin (NRG-1β), reduced the cytotoxic activity of T-DM1 in a subset of breast cancer lines; this effect was reversed by the addn. of pertuzumab. Results from mouse xenograft models showed enhanced antitumor efficacy with T-DM1 and pertuzumab resulting from the unique antitumor activities of each agent. In patients with mBC previously treated with trastuzumab, lapatinib, and chemotherapy, T-DM1 could be dosed at the max. tolerated dose (MTD; 3.6 mg/kg every 3 wk) with std. dose pertuzumab. Adverse events were mostly grade 1 and 2, with indications of clin. activity. Conclusions: Dual targeting of HER2 with the combination of T-DM1 and pertuzumab in cell culture and mouse xenograft models resulted in enhanced antitumor activity. In patients, this combination showed an encouraging safety and tolerability profile with preliminary evidence of efficacy. Clin Cancer Res; 20(2); 456-68. ©2013 AACR.
- 42Lu, 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– 92242https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1OlurrO&md5=ee2756eb89baee7a08a95ff9d43eb736Drug interaction potential of trastuzumab emtansine (T-DM1) combined with pertuzumab in patients with HER2-positive metastatic breast cancerLu, Dan; Burris, Howard A., III; Wang, Bei; Dees, E. Claire; Cortes, Javier; Joshi, Amita; Gupta, Manish; Yi, Joo-Hee; Chu, Yu-Waye; Shih, Ted; Fang, Liang; Girish, SandhyaCurrent Drug Metabolism (2012), 13 (7), 911-922CODEN: CDMUBU; ISSN:1389-2002. (Bentham Science Publishers Ltd.)Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate comprised of trastuzumab and the cytotoxic agent DM1 (deriv. of maytansine) linked by a stable linker N-succinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate (SMCC). T-DM1 targets an epitope located at subdomain IV of human epidermal growth factor receptor 2 (HER2). Pertuzumab is a monoclonal antibody that targets an epitope located at subdomain II of HER2, distinct from the epitope recognized by T-DM1. The pharmacokinetics (PK), safety, and efficacy of T-DM1 combined with pertuzumab were studied in a phase 1b/2 trial in 67 patients with HER2-pos., locally advanced or metastatic breast cancer (MBC). The therapeutic protein-drug interaction (TP-DI) potential of T-DM1 plus pertuzumab was evaluated. The PK of T-DM1-related analytes and pertuzumab were compared with historical PK data. The results show that the exposure of T-DM1 and DM1, as estd. by noncompartmental analyses, was comparable with that reported by historical single-agent studies in patients with HER2-pos. MBC. T-DM1 clearance and vol. of distribution in the central compartment, as estd. by population PK anal., were also comparable between this study and historical single-agent studies in patients with HER2-pos. MBC. Summary statistics of pertuzumab trough and maximal exposure (concns. at predose and 15-30 min after the end of infusion at cycle 1 and at steady state) were similar with those obsd. in a representative historical single-agent study with the same dosing regimen. The visual predictive check plot by population simulation further confirmed that T-DM1 did not alter pertuzumab PK. Based on these data and the PK and pharmacodynamic properties of T-DM1 and pertuzumab, the risk of TP-DI appears to be low when T-DM1 and pertuzumab are given together.
- 43Bartsch, R.; Berghoff, A. S.; Preusser, M. Breast cancer brain metastases responding to primary systemic therapy with T-DM1 J. Neuro-Oncol. 2014, 116, 205– 20643https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2c%252Fht1SlsA%253D%253D&md5=cb5391468373267301aadd8c16fc8e06Breast cancer brain metastases responding to primary systemic therapy with T-DM1Bartsch Rupert; Berghoff Anna S; Preusser MatthiasJournal of neuro-oncology (2014), 116 (1), 205-6 ISSN:.There is no expanded citation for this reference.
- 44Girish, 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– 124044https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xmt1Kru70%253D&md5=f4b2a4ef4aaa79a96adbb1810a0c44d9Clinical pharmacology of trastuzumab emtansine (T-DM1): an antibody-drug conjugate in development for the treatment of HER2-positive cancerGirish, Sandhya; Gupta, Manish; Wang, Bei; Lu, Dan; Krop, Ian E.; Vogel, Charles L.; Burris, Howard A., III; LoRusso, Patricia M.; Yi, Joo-Hee; Saad, Ola; Tong, Barbara; Chu, Yu-Waye; Holden, Scott; Joshi, AmitaCancer Chemotherapy and Pharmacology (2012), 69 (5), 1229-1240CODEN: CCPHDZ; ISSN:0344-5704. (Springer)Purpose: Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate comprising trastuzumab and DM1, a microtubule polymn. inhibitor, covalently bound via a stable thioether linker. To characterize the pharmacokinetics (PK) of T-DM1 in patients with human epidermal growth factor receptor 2 (HER2)-pos. metastatic breast cancer, data from four studies (TDM3569g, TDM4258g, TDM4374g, and TDM4688g) of single-agent T-DM1 administered at 3.6 mg/kg every 3 wk (q3w) were assessed in aggregate. Methods: Multiple analytes-T-DM1, total trastuzumab (TT), DM1, and key metabolites-were quantified using enzyme-linked immunosorbent assays or liq. chromatog. tandem mass spectrometry. PK parameters of T-DM1, TT, and DM1 exposure were calcd. using std. noncompartmental approaches and correlated to efficacy (objective response rate) and safety (platelet counts, hepatic transaminase concns.). Immunogenicity was evaluated by measuring anti-therapeutic antibodies (ATA) to T-DM1 after repeated dosing using validated bridging antibody electrochemiluminescence or enzyme-linked immunosorbent assays. Results: PK parameters for T-DM1, TT, and DM1 were consistent across studies at cycle 1 and steady state. T-DM1 PK was not affected by residual trastuzumab from prior therapy or circulating extracellular domain of HER2. No significant correlations were obsd. between T-DM1 exposure and efficacy, thrombocytopenia, or increased concns. of transaminases. Across the studies, ATA formation was detected in 4.5% (13/286) of evaluable patients receiving T-DM1 q3w. Conclusions The PK profile of single-agent T-DM1 (3.6 mg/kg q3w) is predictable, well characterized, and unaffected by circulating levels of HER2 extracellular domain or residual trastuzumab. T-DM1 exposure does not correlate with clin. responses or key adverse events.
- 45Lu, 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– 67245https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvVSnu7jI&md5=b5b591684a66774da1b9161926217f32An 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 CancerLu, Dan; Joshi, Amita; Wang, Bei; Olsen, Steve; Yi, Joo-Hee; Krop, Ian E.; Burris, Howard A.; Girish, SandhyaClinical Pharmacokinetics (2013), 52 (8), 657-672CODEN: CPKNDH; ISSN:0312-5963. (Springer International Publishing AG)Background and Objective: Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate recently approved by the US Food and Drug Administration for the treatment of human epidermal growth factor receptor 2 (HER2)-pos. metastatic breast cancer previously treated with trastuzumab and taxane chemotherapy. It comprises the microtubule inhibitory cytotoxic agent DM1 conjugated to the HER2-targeted humanized monoclonal antibody trastuzumab via a stable linker. To characterize the pharmacokinetics of T-DM1 in patients with metastatic breast cancer, concns. of multiple analytes were quantified, including serum concns. of T-DM1 conjugate and total trastuzumab (the sum of conjugated and unconjugated trastuzumab), as well as plasma concns. of DM1. The clearance of T-DM1 conjugate is approx. 2 to 3 times faster than its parent antibody, trastuzumab. However, the clearance pathways accounting for this faster clearance rate are unclear. An integrated population pharmacokinetic model that simultaneously fits the pharmacokinetics of T-DM1 conjugate and total trastuzumab can help to elucidate the clearance pathways of T-DM1. The model can also be used to predict total trastuzumab pharmacokinetic profiles based on T-DM1 conjugate pharmacokinetic data and sparse total trastuzumab pharmacokinetic data, thereby reducing the frequency of pharmacokinetic sampling. Methods: T-DM1 conjugate and total trastuzumab serum concn. data, including baseline trastuzumab concns. prior to T-DM1 treatment, from phase I and II studies were used to develop this integrated population pharmacokinetic model. Based on a hypothetical T-DM1 catabolism scheme, two-compartment models for T-DM1 conjugate and trastuzumab were integrated by assuming a one-step deconjugation clearance from T-DM1 conjugate to trastuzumab. The ability of the model to predict the total trastuzumab pharmacokinetic profile based on T-DM1 conjugate pharmacokinetics and various sampling schemes of total trastuzumab pharmacokinetics was assessed to evaluate total trastuzumab sampling schemes. Results: The final model reflects a simplified catabolism scheme of T-DM1, suggesting that T-DM1 clearance pathways include both deconjugation and proteolytic degrdn. The model fits T-DM1 conjugate and total trastuzumab pharmacokinetic data simultaneously. The deconjugation clearance of T-DM1 was estd. to be ∼0.4 L/day. Proteolytic degrdn. clearances for T-DM1 and trastuzumab were similar (∼0.3 L/day). This model accurately predicts total trastuzumab pharmacokinetic profiles based on T-DM1 conjugate pharmacokinetic data and sparse total trastuzumab pharmacokinetic data sampled at preinfusion and end of infusion in cycle 1, and in one addnl. steady state cycle. Conclusions: This semi-mechanistic integrated model links T-DM1 conjugate and total trastuzumab pharmacokinetic data, and supports the inclusion of both proteolytic degrdn. and deconjugation as clearance pathways in the hypothetical T-DM1 catabolism scheme. The model attributes a faster T-DM1 conjugate clearance vs. that of trastuzumab to the presence of a deconjugation process and suggests a similar proteolytic clearance of T-DM1 and trastuzumab. Based on the model and T-DM1 conjugate pharmacokinetic data, a sparse pharmacokinetic sampling scheme for total trastuzumab provides an entire pharmacokinetic profile with similar predictive accuracy to that of a dense pharmacokinetic sampling scheme.
- 46Chudasama, 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– 52746https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhtl2iurfP&md5=1d04057bdead10453770c704e5ed28f4Semi-mechanistic Population Pharmacokinetic Model of Multivalent Trastuzumab Emtansine in Patients with Metastatic Breast CancerChudasama, V. L.; Schaedeli Stark, F.; Harrold, J. M.; Tibbitts, J.; Girish, S. R.; Gupta, M.; Frey, N.; Mager, D. E.Clinical Pharmacology & Therapeutics (New York, NY, United States) (2012), 92 (4), 520-527CODEN: CLPTAT; ISSN:0009-9236. (Nature Publishing Group)Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate (ADC) composed of multiple mols. of the antimicrotubule agent DM1 linked to trastuzumab, a humanized anti-human epidermal growth factor receptor 2 (HER2) monoclonal antibody. Pharmacokinetics data from phase I (n = 52) and phase II (n = 111) studies in HER2-pos. metastatic breast cancer patients show a shorter terminal half-life for T-DM1 than for total trastuzumab (TTmAb). In this work, we translated prior preclin. modeling in monkeys to develop a semi-mechanistic population pharmacokinetics model to characterize T-DM1 and TTmAb concn. profiles. A series of transit compartments with the same disposition parameters was used to describe the deconjugation process from higher to lower drug-to-antibody ratios (DARs). The structure could explain the shorter terminal half-life of T-DM1 relative to TTmab. The final model integrates prior knowledge of T-DM1 DARs from preclin. studies and could provide a platform for understanding and characterizing the pharmacokinetics of other ADC systems. Clin. Pharmacol. & Therapeutics (2012); 92 4, 520-527. doi:10.1038/clpt.2012.153.
- 47Bender, 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– 60147https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsVentbjN&md5=52d01f2d24af314edf8c54d87304af23A population pharmacokinetic/pharmacodynamic model of thrombocytopenia characterizing the effect of trastuzumab emtansine (T-DM1) on platelet counts in patients with HER2-positive metastatic breast cancerBender, Brendan C.; Schaedeli-Stark, Franziska; Koch, Reinhold; Joshi, Amita; Chu, Yu-Waye; Rugo, Hope; Krop, Ian E.; Girish, Sandhya; Friberg, Lena E.; Gupta, ManishCancer Chemotherapy and Pharmacology (2012), 70 (4), 591-601CODEN: CCPHDZ; ISSN:0344-5704. (Springer)Purpose: Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate in the development for the treatment of human epidermal growth factor receptor 2-pos. cancers. Thrombocytopenia (TCP) is the dose-limiting toxicity of T-DM1. A semimechanistic population pharmacokinetic/pharmacodynamic (PK/PD) model was developed to characterize the effect of T-DM1 on patient platelet counts. Methods: A PK/PD model with transit compartments that mimic platelet development and circulation was fit to concn.-platelet-time course data from two T-DM1 single-agent studies (TDM3569g; N = 52 and TDM4258g; N = 112). NONMEM 7 software was used for model development. Data from a sep. phase II study (TDM4374g; N = 110) were used for model evaluation. Patient baseline characteristics were evaluated as covariates of model PD parameters. Results: The model described the platelet data well and predicted the incidence of grade ≥3 TCP. The model predicted that with T-DM1 3.6 mg/kg given every 3 wk (q3w), the lowest platelet nadir would occur after the first dose. Also predicted was a patient subgroup (46 %) having variable degrees of downward drifting platelet-time profiles, which were predicted to stabilize by the eighth treatment cycle to platelet counts above grade 3 TCP. Baseline characteristics were not significant covariates of PD parameters in the model. Conclusions: This semimechanistic PK/PD model accurately captures the cycle 1 platelet nadir, the downward drift noted in some patient platelet-time profiles, and the ∼8 % incidence of grade ≥3 TCP with T-DM1 3.6 mg/kg q3w. This model supports T-DM1 3.6 mg/kg q3w as a well-tolerated dose with minimal dose delays or redns. for TCP.
- 48Shen, 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– 91048https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xht1OlurrN&md5=faf70e5692e4d6a5a7789a0510705837Catabolic fate and pharmacokinetic characterization of trastuzumab emtansine (T-DM1): an emphasis on preclinical and clinical catabolismShen, Ben-Quan; Bumbaca, Daniela; Saad, Ola; Yue, Qin; Pastuskovas, Cinthia V.; Khojasteh, S. Cyrus; Tibbitts, Jay; Kaur, Surinder; Wang, Bei; Chu, Yu-Waye; LoRusso, Patricia M.; Girish, SandhyaCurrent Drug Metabolism (2012), 13 (7), 901-910CODEN: CDMUBU; ISSN:1389-2002. (Bentham Science Publishers Ltd.)Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate in clin. development for the treatment of human epidermal growth factor receptor 2 (HER2)-pos. cancers. Herein, we describe a series of studies to assess T-DM1 absorption, distribution, metab., and excretion (ADME) in rats as well as to assess human exposure to T-DM1 catabolites. Following administration of unlabeled and radiolabeled T-DM1 in female Sprague Dawley rats as a single dose, plasma, urine, bile and feces were assessed for mass balance, profiling and identification of catabolites. In rats, the major circulating species in plasma was T-DM1, while DM1 concns. were low (1.08 to 15.6 ng/mL). The major catabolites found circulating in rat plasma were DM1, [N-maleimidomethyl] cyclohexane-1-carboxylate-DM1 (MCC-DM1), and Lysine-MCC-DM1. These catabolites identified in rats were also detected in plasma samples from patients with HER2-pos. metastatic breast cancer who received single-agent T-DM1 (3.6 mg/kg every 3 wk) in a phase 2 clin. study. There was no evidence of tissue accumulation in rats or catabolite accumulation in human plasma following multiple dosing. In rats, T-DM1 was distributed nonspecifically to the organs without accumulation. The major pathway of DM1-contg. catabolite elimination in rats was the fecal/biliary route, with up to 80% of radioactivity recovered in the feces and 50% in the bile. The rat T-DM1 ADME profile is likely similar to the human profile, although there may be differences since trastuzumab does not bind the rat HER2- like receptor. Further research is necessary to more fully understand the T-DM1 ADME profile in humans.
- 49Thon, 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– 1984There is no corresponding record for this reference.
- 50Sendur, M. A.; Aksoy, S.; Altundag, K. Cardiotoxicity of novel HER2-targeted therapies Curr. Med. Res. Opin. 2013, 29, 1015– 102450https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFeqsL%252FN&md5=a5a593ec4ce6bbbb95584eb1e1d38d05Cardiotoxicity of novel HER2-targeted therapiesSendur, Mehmet A. N.; Aksoy, Sercan; Altundag, KadriCurrent Medical Research and Opinion (2013), 29 (8), 1015-1024CODEN: CMROCX; ISSN:0300-7995. (Informa Healthcare)A review. Background: Trastuzumab, an anti-HER2 humanized monoclonal antibody, is the std. treatment for both early and metastatic HER2-pos. breast cancer. In addn. to other chemotherapeutic agents, trastuzumab significantly improves response rate and survival in HER2-pos. early and metastatic breast cancer. Although it is well known that trastuzumab therapy is closely assocd. with both symptomatic and asymptomatic cardiotoxicity, less is known about novel HER2-targeted therapies. The aim of this review is to discuss the cardiac safety data from recent studies of novel anti-HER2 drugs other than trastuzumab. Scope: Novel HER2-targeted therapies showed favorable results in HER2 pos. metastatic breast cancer patients. Pubmed database, ASCO and San Antonio Breast Cancer Symposium Meeting abstrs. were searched until Jan. 2013 using the following search keywords; trastuzumab, trastuzumab cardiotoxicity, HER-2 targeted therapies, lapatinib, pertuzumab, trastuzumab emtansine, afatinib and neratinib; papers which were considered relevant for the aim of this review were selected by the authors. Lapatinib, pertuzumab, T-DM1, neratinib and afatinib mols. are evaluated in the study. Findings: In a comprehensive anal., 3689 lapatinib treated patients enrolled in 49 trials; asymptomatic cardiac events were reported in 53 patients (1.4%) and symptomatic grade III and IV systolic dysfunction was obsd. only in 7 patients (0.2%) treated with lapatinib. In phase I-III trials of pertuzumab, cardiac dysfunction was seen in 4.5-14.5% of patients with pertuzumab treatment and cardiac dysfunction was usually grade I and II. Cardiotoxicity of pertuzumab was usually reported with the trastuzumab combination and no additive cardiotoxicity was reported with addn. of pertuzumab to trastuzumab. T-DM1 had a better safety profile compared to trastuzumab, no significant cardiotoxicity was obsd. with T-DM1 in heavily pre-treated patients. In the EMILIA study, only in 1.7% of patients in the T-DM1 group experienced redn. of left ventricular ejection fraction (LVEF) and grade III LVEF redn. developed only in one patient (0.2%) in the T-DM1 group compared to the lapatinib plus capecitabine group. In phase I-II trials with neratinib no cardiotoxicity was reported whereas cardiotoxicity was seen between 0-5.3% with afatinib treatment. Conclusion: Although cardiac toxicity has been reported as an adverse event for novel HER2-targeted therapies, cardiac dysfunction rate of the novel HER2-targeted therapies is significantly lower than the trastuzumab and combination of these agents with trastuzumab did not significantly increase the cardiac adverse events.