Site-Specific Conjugation of the Indolinobenzodiazepine DGN549 to Antibodies Affords Antibody–Drug Conjugates with an Improved Therapeutic Index as Compared with Lysine ConjugationClick to copy article linkArticle link copied!
- Chen BaiChen BaiScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Chen Bai
- Emily E. ReidEmily E. ReidScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Emily E. Reid
- Alan WilhelmAlan WilhelmScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Alan Wilhelm
- Manami ShizukaManami ShizukaScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Manami Shizuka
- Erin K. MaloneyErin K. MaloneyScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Erin K. Maloney
- Rassol LaleauRassol LaleauScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Rassol Laleau
- Lauren HarveyLauren HarveyScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Lauren Harvey
- Katie E. ArcherKatie E. ArcherScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Katie E. Archer
- Dilrukshi VitharanaDilrukshi VitharanaScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Dilrukshi Vitharana
- Sharlene AdamsSharlene AdamsScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Sharlene Adams
- Yelena KovtunYelena KovtunScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Yelena Kovtun
- Michael L. MillerMichael L. MillerScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Michael L. Miller
- Ravi ChariRavi ChariScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Ravi Chari
- Thomas A. KeatingThomas A. KeatingScience, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Thomas A. Keating
- Nicholas C. Yoder*Nicholas C. Yoder*E-mail: [email protected]Science, Technology, and Translation, ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United StatesMore by Nicholas C. Yoder
Abstract
Antibody–drug conjugates have elicited great interest recently as targeted chemotherapies for cancer. Recent preclinical and clinical data have continued to raise questions about optimizing the design of these complex therapeutics. Biochemical methods for site-specific antibody conjugation have been a design feature of recent clinical ADCs, and preclinical reports suggest that site-specifically conjugated ADCs generically offer improved therapeutic indices (i.e., the fold difference between efficacious and maximum tolerated doses). Here we present the results of a systematic preclinical comparison of ADCs embodying the DNA-alkylating linker-payload DGN549 generated with both heterogeneous lysine-directed and site-specific cysteine-directed conjugation chemistries. Importantly, the catabolites generated by each ADC are the same regardless of the conjugation format. In two different model systems evaluated, the site-specific ADC showed a therapeutic index benefit. However, the therapeutic index benefit is different in each case: both show evidence of improved tolerability, though with different magnitudes, and in one case significant efficacy improvement is also observed. These results support our contention that conjugation chemistry of ADCs is best evaluated in the context of a particular antibody, target, and linker-payload, and ideally across multiple disease models.
Introduction
Results and Discussion
Chemistry
Antibody Conjugation
ADC Characterization
HIC
In Vitro Catabolite Identification
In Vitro Binding Cytotoxicity
IC50 (M ADC) | IC50 (M DGN549) | ||||
---|---|---|---|---|---|
Cell Line | ADC | – Block | + Block | – Block | + Block |
KB | 2a | 5 × 10–12 | 9 × 10–10 | 1 × 10–11 | 2 × 10–9 |
2b | 5 × 10–12 | 2 × 10–9 | 7 × 10–12 | 5 × 10–9 | |
2c | 2 × 10–12 | 1 × 10–9 | 5 × 10–12 | 5 × 10–9 | |
T47D | 2a | 2 × 10–11 | 9 × 10–9 | 6 × 10–11 | 3 × 10–8 |
2b | 1 × 10–11 | 1 × 10–8 | 3 × 10–11 | 2 × 10–8 | |
NCI-H2110 | 2a | 4 × 10–11 | 2 × 10–9 | 1 × 10–10 | 5 × 10–9 |
2b | 4 × 10–11 | 2 × 10–8 | 9 × 10–11 | 4 × 10–8 |
Cell Line | ADC | IC50 (M ADC) | IC50 (M DGN549) |
---|---|---|---|
EOL-1 | 3a | 2 × 10–12 | 6 × 10–12 |
3b | 2 × 10–12 | 4 × 10–12 | |
Non-Targeting Lysine-Linked ADC | 8 × 10–10 | 2 × 10–9 | |
Non-Targeting CYSMAB ADC | 2 × 10–9 | 4 × 10–9 | |
HNT-34 | 3a | 8 × 10–13 | 2 × 10–12 |
3b | 1 × 10–12 | 2 × 10–12 | |
Non-Targeting Lysine-Linked ADC | 7 × 10–10 | 2 × 10–9 | |
Non-Targeting CYSMAB ADC | 1 × 10–9 | 2 × 10–9 | |
MV4-11 | 3a | 5 × 10–13 | 2 × 10–12 |
3b | 8 × 10–13 | 2 × 10–12 | |
Non-Targeting Lysine-Linked ADC | 3 × 10–11 | 6 × 10–11 | |
Non-Targeting CYSMAB ADC | 2 × 10–11 | 4 × 10–11 |
In Vivo Efficacy
Dose | Regressions | ||||
---|---|---|---|---|---|
Treatment | μg DGN549/kg | μg ADC/kg | T/C Day 22 (%) | Partial | Complete |
Vehicle | - | - | - | 0/6 | 0/6 |
ADC 2a | 3 | 160 | 32 | 0/6 | 0/6 |
9 | 510 | 1 | 6/6 | 6/6 | |
ADC 2b | 3 | 230 | 39 | 0/6 | 0/6 |
9 | 680 | 2 | 6/6 | 6/6 |
Dose | |||||
---|---|---|---|---|---|
Treatment | μg DGN549/kg | μg ADC/kg | Median Survival (Days) | Tumor Growth Delay | Increased Life Span (%) |
Vehicle | - | 28 | - | - | |
ADC 3a | 0.1 | 5.8 | 44.5 | 16.5 | 59 |
0.3 | 17.4 | >152 | >124 | >443 | |
1.0 | 58 | >152 | >124 | >443 | |
Non-Targeting Lysine-Linked ADC | 1.0 | 54 | 31 | 3 | 11 |
ADC 3b | 0.1 | 7.6 | 101.5 | 73.5 | 262.5 |
0.3 | 22.8 | >152 | >124 | >443 | |
1.0 | 76 | >152 | >124 | >443 | |
Non-Targeting CYSMAB ADC | 1.0 | 6.8 | 26.5 | - | - |
Mouse Tolerability
Mouse Pharmacokinetics
ADC | Cmax (μg/mL) | AUC0-∞ (h·μg/mL) | Cl (mL/h/kg) | t1/2 (h/days) | t1/2 (days) | Vss (mL/kg) | |
---|---|---|---|---|---|---|---|
2a | Total Antibody | 40.3 | 4655 | 0.54 | 221 | 9.2 | 163 |
2a | Conjugated Antibody | 45.3 | 5578 | 0.45 | 232 | 9.7 | 142 |
2b | Total Antibody | 52.1 | 10795 | 0.23 | 383 | 15.6 | 124 |
2b | Conjugated Antibody | 65.4 | 9159 | 0.27 | 322 | 13.4 | 121 |
Conclusions
Materials and Methods
Preparation and Characterization of ADCs
Antibodies
DGN549 Payloads
Synthesis of Lysine-Linked ADCs
Synthesis of CYSMAB ADCs
Characterization of ADCs
In Vitro Experiments
Binding
Cytotoxicity
Bystander Cytotoxicity Assay
Identification of Catabolites from mAb1-DGN549 CYSMAB ADC
In Vivo Studies
Efficacy of mAb1 ADCs in NCI-H2110 Xenografts
Efficacy of mAb2 ADCs in Molm-13 Xenografts
Tolerability
Pharmacokinetics of mAb1 ADCs
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.bioconjchem.9b00777.
Detailed experimental procedures for synthesis of compound 1b, supplementary characterization data for ADCs, and supplementary methods (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
References
This article references 55 other publications.
- 1Chari, R. V. J., Miller, M. L., and Widdison, W. C. (2014) Antibody–Drug Conjugates: An Emerging Concept in Cancer Therapy. Angew. Chem., Int. Ed. 53 (15), 3796– 3827, DOI: 10.1002/anie.201307628Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXjtVGmsr0%253D&md5=fbfb2f843f516654ea7c0eb98949604cAntibody-Drug Conjugates: An Emerging Concept in Cancer TherapyChari, Ravi V. J.; Miller, Michael L.; Widdison, Wayne C.Angewandte Chemie, International Edition (2014), 53 (15), 3796-3827CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Traditional cancer chemotherapy is often accompanied by systemic toxicity to the patient. Monoclonal antibodies against antigens on cancer cells offer an alternative tumor-selective treatment approach. However, most monoclonal antibodies are not sufficiently potent to be therapeutically active on their own. Antibody-drug conjugates (ADCs) use antibodies to deliver a potent cytotoxic compd. selectively to tumor cells, thus improving the therapeutic index of chemotherapeutic agents. The recent approval of two ADCs, brentuximab vedotin and ado-trastuzumab emtansine, for cancer treatment has spurred tremendous research interest in this field. This Review touches upon the early efforts in the field, and describes how the lessons learned from the first-generation ADCs have led to improvements in every aspect of this technol., i.e., the antibody, the cytotoxic compd., and the linker connecting them, leading to the current successes. The design of ADCs currently in clin. development, and results from mechanistic studies and preclin. and clin. evaluation are discussed. Emerging technologies that seek to further advance this exciting area of research are also discussed.
- 2Lambert, J. M. and Chari, R. V. (2014) Ado-trastuzumab Emtansine (T-DM1): an antibody-drug conjugate (ADC) for HER2-positive breast cancer. J. Med. Chem. 57 (16), 6949– 6964, DOI: 10.1021/jm500766wGoogle Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVGlu7rE&md5=3ddee50cc227097126dd47e7683a6ac7Ado-trastuzumab Emtansine (T-DM1): An Antibody-Drug Conjugate (ADC) for HER2-Positive Breast CancerLambert, John M.; Chari, Ravi V. J.Journal of Medicinal Chemistry (2014), 57 (16), 6949-6964CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. 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 deriv. of DM1 is subsequently released by proteolytic degrdn. of the antibody moiety within the lysosome. Initial clin. evaluation led to a phase III trial in advanced HER2-pos. 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-pos. metastatic breast cancer who had previously received trastuzumab and a taxane, sep. or in combination, the first ADC to receive full approval based on a randomized study.
- 3Senter, P. D. and Sievers, E. L. (2012) The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nat. Biotechnol. 30 (7), 631– 637, DOI: 10.1038/nbt.2289Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XpvFCqtbw%253D&md5=6940eb8178e8c8c150a7b21361e23234The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphomaSenter, Peter D.; Sievers, Eric L.Nature Biotechnology (2012), 30 (7), 631-637CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)A review. Progress was made recently in developing antibody-drug conjugates (ADCs) that can selectively deliver cancer drugs to tumor cells. In principle, the idea is simple: by attaching drugs to tumor-seeking antibodies, target cells will be killed and nontarget cells will be spared. In practice, many parameters needed to be addressed to develop safe and effective ADCs, including the expression profiles of tumor vs. normal tissues, the potency of the drug, the linker attaching the drug and placement of the drug on the antibody, and the pharmacokinetic and stability profiles of the resulting ADC. All these issues had been taken into account in developing brentuximab vedotin (Adcetris), an ADC that recently received accelerated approval by the US Food and Drug Administration for the treatment of relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma (ALCL). Research is under way to extend the applications of brentuximab vedotin and to advance the field by developing other ADCs with new linker and conjugation strategies.
- 4Appelbaum, F. R. and Bernstein, I. D. (2017) Gemtuzumab ozogamicin for acute myeloid leukemia. Blood 130 (22), 2373– 2376, DOI: 10.1182/blood-2017-09-797712Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhs1erurfM&md5=970680b53cd9f2971e811d044ef9b6fdGemtuzumab ozogamicin for acute myeloid leukemiaAppelbaum, Frederick R.; Bernstein, Irwin D.Blood (2017), 130 (22), 2373-2376CODEN: BLOOAW; ISSN:1528-0020. (American Society of Hematology)On 1 Sept. 2017, the US Food and Drug Administration (FDA) approved gemtuzumab ozogamicin (GO) for the treatment of adults with newly diagnosed CD33+ acute myeloid leukemia and for patients aged ≥2 years with CD33+ acute myeloid leukemia who have experienced a relapse or who have not responded to initial treatment. This signals a new chapter in the long and unusual story of GO, which was the first antibody-drug conjugate approved for human use by the FDA.
- 5Yurkiewicz, I. R., Muffly, L., and Liedtke, M. (2018) Inotuzumab ozogamicin: a CD22 mAb-drug conjugate for adult relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Drug Des., Dev. Ther. 12, 2293– 2300, DOI: 10.2147/DDDT.S150317Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1OnsrrM&md5=de9ba79b0c4d5a13606416647011fe8cInotuzumab ozogamicin: a CD22 mAb-drug conjugate for adult relapsed or refractory B-cell precursor acute lymphoblastic leukemiaYurkiewicz, Ilana R.; Muffly, Lori; Liedtke, MichaelaDrug Design, Development and Therapy (2018), 12 (), 2293-2300CODEN: DDDTAQ; ISSN:1177-8881. (Dove Medical Press Ltd.)Despite improved rates of remission and cure in newly diagnosed adult acute lymphoblastic leukemia (ALL), the prognosis for patients with relapsed or refractory disease remains poor and the 5-yr overall survival rate after relapse is under 10%. A recent paradigm shift has focused on the promise of targeted immunotherapy rather than std. chemotherapy, as ALL blast cells express a variety of antigens, and monoclonal antibodies may be developed to identify and destroy the leukemic cells. Inotuzumab ozogamicin is a CD22 monoclonal antibody conjugated to the cytotoxic antibiotic calicheamicin. CD22 expression is detected on leukemic blasts in over 90% of patients with ALL. Based on promising results from preclin. studies, inotuzumab ozogamicin was tested in Phase 1/2 and Phase 3 clin. trials and it demonstrated improved complete remission rates, progression-free survival and overall survival in relapsed or refractory adult ALL compared to std. therapy. Ongoing studies are evaluating the value of inotuzumab ozogamicin when given in combination with chemotherapy as part of upfront treatment. This review discusses the drug's biochem. properties and mechanism of action, preclin. research outcomes, clin. trial results, adverse events and toxicities, drug approval and ongoing investigations.
- 6Dornan, D., Bennett, F., Chen, Y., Dennis, M., Eaton, D., Elkins, K., French, D., Go, M. A., Jack, A., Junutula, J. R. (2009) Therapeutic potential of an anti-CD79b antibody-drug conjugate, anti-CD79b-vc-MMAE, for the treatment of non-Hodgkin lymphoma. Blood 114 (13), 2721– 2729, DOI: 10.1182/blood-2009-02-205500Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1eit7jO&md5=02b943d887958c47dd2e5d1a3d490371Therapeutic potential of an anti-CD79b antibody-drug conjugate, anti-CD79b-vc-MMAE, for the treatment of non-Hodgkin lymphomaDornan, David; Bennett, Fiona; Chen, Yvonne; Dennis, Mark; Eaton, Dan; Elkins, Kristi; French, Dorothy; Go, Mary Ann T.; Jack, Andrew; Junutula, Jagath R.; Koeppen, Hartmut; Lau, Jeffrey; McBride, Jacqueline; Rawstron, Andy; Shi, Xiaoyan; Yu, Nancy; Yu, Shang-Fan; Yue, Peng; Zheng, Bing; Ebens, Allen; Polson, Andrew G.Blood (2009), 114 (13), 2721-2729CODEN: BLOOAW; ISSN:0006-4971. (American Society of Hematology)Here we describe the generation of an antibody-drug conjugate (ADC) consisting of a humanized anti-CD79b antibody that is conjugated to monomethylauristatin E (MMAE) through engineered cysteines (THIOMABs) by a protease cleavable linker. By using flow cytometry, we detected the surface expression of CD79b in almost all non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia patients, suggesting that anti-CD79b-vcMMAE could be widely used in these malignancies. By using NHL cell lines to simulate a patient population we discovered that a minimal cell-surface expression level of CD79b was required for in vitro activity. Within the subpopulation of cell lines above this minimal threshold, we found that sensitivity to free MMAE, mutation of cancer genes, and cell doubling time were poorly correlated with in vitro activity; however, the expression level of BCL-XL was correlated with reduced sensitivity to anti-CD79b-vcMMAE. This observation was supported by in vivo data showing that a Bcl-2 family inhibitor, ABT-263, strikingly enhanced the activity of anti-CD79b-vcMMAE. Furthermore, anti-CD79b-vcMMAE was significantly more effective than a std.-of-care regimen, R-CHOP (ie, rituximab with a single i.v. injection of 30 mg/kg cyclophosphamide, 2.475 mg/kg doxorubicin, 0.375 mg/kg vincristine, and oral dosing of 0.15 mg/kg prednisone once a day for 5 days), in 3 xenograft models of NHL. Together, these data suggest that anti-CD79b-vcMMAE could be broadly efficacious for the treatment of NHL.
- 7Donaghy, H. (2016) Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugates. MAbs 8 (4), 659– 671, DOI: 10.1080/19420862.2016.1156829Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmtFant7k%253D&md5=b2ba3e43e8362d8a3f7c368a5af5f94eEffects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugatesDonaghy, HeathermAbs (2016), 8 (4), 659-671CODEN: MABSCP; ISSN:1942-0870. (Taylor & Francis, Inc.)Antibody-drug conjugates (ADCs) represent a new class of cancer therapeutics. Their design involves a tumor-specific antibody, a linker and a cytotoxic payload. They were designed to allow specific targeting of highly potent cytotoxic agents to tumor cells while sparing normal cells. Frequent toxicities that may be driven by any of the components of an ADC have been reported. There are currently more than 50 ADCs in active clin. development, and a further ∼20 that have been discontinued. For this review, the reported toxicities of ADCs were analyzed, and the mechanisms for their effects are explored in detail. Methods to reduce toxicities, including dosing strategies and drug design, are discussed. The toxicities reported for active and discontinued drugs are important to drive the rational design and improve the therapeutic index of ADCs of the future.
- 8Chari, R. V. J. (2016) Expanding the Reach of Antibody-Drug Conjugates. ACS Med. Chem. Lett. 7 (11), 974– 976, DOI: 10.1021/acsmedchemlett.6b00312Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFWntrnN&md5=365b2432f4bc622eb92d01d5cb9c78a8Expanding the Reach of Antibody-Drug ConjugatesChari, Ravi V. J.ACS Medicinal Chemistry Letters (2016), 7 (11), 974-976CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Antibody-drug conjugates (ADCs) represent an emerging new paradigm in cancer therapy. The approval of two ADCs has spurred considerable interest in this area of research, and over 55 ADCs are currently in clin. testing. In order to improve the clin. success rate of ADC therapy, all three components of the ADC: the antibody, linker, and payload have to be optimized. While considerable improvements have been made in antibody properties and target selection, medicinal chem. efforts have lagged behind, and there is a significant need for innovation in linker design and payloads.
- 9Behrens, C. R. and Liu, B. (2014) Methods for site-specific drug conjugation to antibodies. MAbs 6 (1), 46– 53, DOI: 10.4161/mabs.26632Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2c%252FntlShtQ%253D%253D&md5=b23aea418d3a37ab7f5b94c3780a8e8cMethods for site-specific drug conjugation to antibodiesBehrens Christopher R; Liu BinmAbs (2014), 6 (1), 46-53 ISSN:.Antibody drug conjugates (ADCs) are an emerging class of targeted therapeutics with the potential to improve therapeutic index over traditional chemotherapy. Drugs and linkers have been the current focus of ADC development, in addition to antibody and target selection. Recently, however,the importance of conjugate homogeneity has been realized. The current methods for drug attachment lead to a heterogeneous mixture, and some populations of that mixture have poor in vivo performance. New methods for site-specific drug attachment lead to more homogeneous conjugates and allow control of the site of drug attachment. These subtle improvements can have profound effects on in vivo efficacy and therapeutic index. This review examines current methods for site-specific drug conjugation to antibodies, and compares in vivo results with their non-specifically conjugated counterparts. The apparent improvement in pharmacokinetics and the reduced off target toxicity warrant further development of this site-specific modification approach for future ADC development.
- 10Agarwal, P. and Bertozzi, C. R. (2015) Site-specific antibody-drug conjugates: the nexus of bioorthogonal chemistry, protein engineering, and drug development. Bioconjugate Chem. 26 (2), 176– 192, DOI: 10.1021/bc5004982Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFCrsrfK&md5=c3d822b0e53e5cdb074e5b63b8060ccfSite-Specific Antibody-Drug Conjugates: The Nexus of Bioorthogonal Chemistry, Protein Engineering, and Drug DevelopmentAgarwal, Paresh; Bertozzi, Carolyn R.Bioconjugate Chemistry (2015), 26 (2), 176-192CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)A review. Antibody-drug conjugates (ADCs) combine the specificity of antibodies with the potency of small mols. to create targeted drugs. Despite the simplicity of this concept, generation of clin. successful ADCs has been very difficult. Over the past several decades, scientists have learned a great deal about the constraints on antibodies, linkers, and drugs as they relate to successful construction of ADCs. Once these components are in hand, most ADCs are prepd. by nonspecific modification of antibody lysine or cysteine residues with drug-linker reagents, which results in heterogeneous product mixts. that cannot be further purified. With advances in the fields of bioorthogonal chem. and protein engineering, there is growing interest in producing ADCs by site-specific conjugation to the antibody, yielding more homogeneous products that have demonstrated benefits over their heterogeneous counterparts in vivo. Here, we chronicle the development of a multitude of site-specific conjugation strategies for assembly of ADCs and provide a comprehensive account of key advances and their roots in the fields of bioorthogonal chem. and protein engineering.
- 11Panowski, S., Bhakta, S., Raab, H., Polakis, P., and Junutula, J. R. (2014) Site-specific antibody drug conjugates for cancer therapy. MAbs 6 (1), 34– 45, DOI: 10.4161/mabs.27022Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2cvnslaltw%253D%253D&md5=9da883692122c03303f785e61f6c5db7Site-specific antibody drug conjugates for cancer therapyPanowski Siler; Bhakta Sunil; Raab Helga; Polakis Paul; Junutula Jagath RmAbs (2014), 6 (1), 34-45 ISSN:.Antibody therapeutics have revolutionized the treatment of cancer over the past two decades. Antibodies that specifically bind tumor surface antigens can be effective therapeutics; however, many unmodified antibodies lack therapeutic activity. These antibodies can instead be applied successfully as guided missiles to deliver potent cytotoxic drugs in the form of antibody drug conjugates (ADCs). The success of ADCs is dependent on four factors--target antigen, antibody, linker, and payload. The field has made great progress in these areas, marked by the recent approval by the US Food and Drug Administration of two ADCs, brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla). However, the therapeutic window for many ADCs that are currently in pre-clinical or clinical development remains narrow and further improvements may be required to enhance the therapeutic potential of these ADCs. Production of ADCs is an area where improvement is needed because current methods yield heterogeneous mixtures that may include 0-8 drug species per antibody molecule. Site-specific conjugation has been recently shown to eliminate heterogeneity, improve conjugate stability, and increase the therapeutic window. Here, we review and describe various site-specific conjugation strategies that are currently used for the production of ADCs, including use of engineered cysteine residues, unnatural amino acids, and enzymatic conjugation through glycotransferases and transglutaminases. In addition, we also summarize differences among these methods and highlight critical considerations when building next-generation ADC therapeutics.
- 12Junutula, J. R., Raab, H., Clark, S., Bhakta, S., Leipold, D. D., Weir, S., Chen, Y., Simpson, M., Tsai, S. P., Dennis, M. S. (2008) Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat. Biotechnol. 26 (8), 925– 932, DOI: 10.1038/nbt.1480Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXps1Wmu7s%253D&md5=d55f59594fb6349d0337dfed2783e83cSite-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic indexJunutula, Jagath R.; Raab, Helga; Clark, Suzanna; Bhakta, Sunil; Leipold, Douglas D.; Weir, Sylvia; Chen, Yvonne; Simpson, Michelle; Tsai, Siao Ping; Dennis, Mark S.; Lu, Yanmei; Meng, Y. Gloria; Ng, Carl; Yang, Jihong; Lee, Chien C.; Duenas, Eileen; Gorrell, Jeffrey; Katta, Viswanatham; Kim, Amy; McDorman, Kevin; Flagella, Kelly; Venook, Rayna; Ross, Sarajane; Spencer, Susan D.; Wong, Wai Lee; Lowman, Henry B.; Vandlen, Richard; Sliwkowski, Mark X.; Scheller, Richard H.; Polakis, Paul; Mallet, WilliamNature Biotechnology (2008), 26 (8), 925-932CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Antibody-drug conjugates enhance the antitumor effects of antibodies and reduce adverse systemic effects of potent cytotoxic drugs. However, conventional drug conjugation strategies yield heterogeneous conjugates with relatively narrow therapeutic index (max. tolerated dose/curative dose). Using leads from the authors' previously described phage display-based method to predict suitable conjugation sites, the authors engineered cysteine substitutions at positions on light and heavy chains that provide reactive thiol groups and do not perturb Ig folding and assembly, or alter antigen binding. When conjugated to monomethyl auristatin E, an antibody against the ovarian cancer antigen MUC16 is as efficacious as a conventional conjugate in mouse xenograft models. Moreover, it is tolerated at higher doses in rats and cynomolgus monkeys than the same conjugate prepd. by conventional approaches. The favorable in vivo properties of the near-homogeneous compn. of this conjugate suggest that this strategy offers a general approach to retaining the antitumor efficacy of antibody-drug conjugates, while minimizing their systemic toxicity.
- 13Junutula, J. R., Flagella, K. M., Graham, R. A., Parsons, K. L., Ha, E., Raab, H., Bhakta, S., Nguyen, T., Dugger, D. L., Li, G. (2010) Engineered Thio-Trastuzumab-DM1 Conjugate with an Improved Therapeutic Index to Target Human Epidermal Growth Factor Receptor 2–Positive Breast Cancer. Clin. Cancer Res. 16 (19), 4769– 4778, DOI: 10.1158/1078-0432.CCR-10-0987Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXht1aktbjK&md5=888bda7f06d1ed864111b607dae5f969Engineered Thio-Trastuzumab-DM1 Conjugate with an Improved Therapeutic Index to Target Human Epidermal Growth Factor Receptor 2-Positive Breast CancerJunutula, Jagath R.; Flagella, Kelly M.; Graham, Richard A.; Parsons, Kathryn L.; Ha, Edward; Raab, Helga; Bhakta, Sunil; Nguyen, Trung; Dugger, Debra L.; Li, Guangmin; Mai, Elaine; Lewis Phillips, Gail D.; Hiraragi, Hajime; Fuji, Reina N.; Tibbitts, Jay; Vandlen, Richard; Spencer, Susan D.; Scheller, Richard H.; Polakis, Paul; Sliwkowski, Mark X.Clinical Cancer Research (2010), 16 (19), 4769-4778CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Antibody drug conjugates (ADCs) combine the ideal properties of both antibodies and cytotoxic drugs by targeting potent drugs to the antigen-expressing tumor cells, thereby enhancing their antitumor activity. Successful ADC development for a given target antigen depends on optimization of antibody selection, linker stability, cytotoxic drug potency, and mode of linker-drug conjugation to the antibody. Here, we systematically examd. the in vitro potency as well as in vivo preclin. efficacy and safety profiles of a heterogeneous prepn. of conventional trastuzumab-mcc-DM1 (TMAb-mcc-DM1) ADC with that of a homogeneous engineered thio-trastuzumab-mpeo-DM1 (thioTMAb-mpeo-DM1) conjugate. To generate thioTMAb-mpeo-DM1, one drug maytansinoid 1 (DM1) mol. was conjugated to an engineered cysteine residue at Ala114 (Kabat numbering) on each trastuzumab-heavy chain, resulting in two DM1 mols. per antibody. ThioTMAb-mpeo-DM1 retained similar in vitro anti-cell proliferation activity and human epidermal growth factor receptor 2 (HER2) binding properties to that of the conventional ADC. Furthermore, it showed improved efficacy over the conventional ADC at DM1-equiv doses (μg/m2) and retained efficacy at equiv. antibody doses (mg/kg). An improved safety profile of >2-fold was obsd. in a short-term target-independent rat safety study. In cynomolgus monkey safety studies, thioTMAb-mpeo-DM1 was tolerated at higher antibody doses (up to 48 mg/kg or 6000 μg DM1/m2) compared with the conventional ADC that had dose-limiting toxicity at 30 mg/kg (6000 μg DM1/m2). The engineered thioTMAb-mpeo-DM1 with broadened therapeutic index represents a promising antibody drug conjugate for future clin. development of HER2-pos. targeted breast cancer therapies.
- 14Shen, B. Q., Xu, K., Liu, L., Raab, H., Bhakta, S., Kenrick, M., Parsons-Reponte, K. L., Tien, J., Yu, S. F., Mai, E. (2012) Conjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugates. Nat. Biotechnol. 30 (2), 184– 189, DOI: 10.1038/nbt.2108Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVGqtLg%253D&md5=c647fc5bdfe93dfdabb15ca40934837fConjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugatesShen, Ben-Quan; Xu, Keyang; Liu, Luna; Raab, Helga; Bhakta, Sunil; Kenrick, Margaret; Parsons-Reponte, Kathryn L.; Tien, Janet; Yu, Shang-Fan; Mai, Elaine; Li, Dongwei; Tibbitts, Jay; Baudys, Jakub; Saad, Ola M.; Scales, Suzie J.; McDonald, Paul J.; Hass, Philip E.; Eigenbrot, Charles; Nguyen, Trung; Solis, Willy A.; Fuji, Reina N.; Flagella, Kelly M.; Patel, Darshana; Spencer, Susan D.; Khawli, Leslie A.; Ebens, Allen; Wong, Wai Lee; Vandlen, Richard; Kaur, Surinder; Sliwkowski, Mark X.; Scheller, Richard H.; Polakis, Paul; Junutula, Jagath R.Nature Biotechnology (2012), 30 (2), 184-189CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)The reactive thiol in cysteine is used for coupling maleimide linkers in the generation of antibody conjugates. To assess the impact of the conjugation site, we engineered cysteines into a therapeutic HER2/neu antibody at three sites differing in solvent accessibility and local charge. The highly solvent-accessible site rapidly lost conjugated thiol-reactive linkers in plasma owing to maleimide exchange with reactive thiols in albumin, free cysteine or glutathione. In contrast, a partially accessible site with a pos. charged environment promoted hydrolysis of the succinimide ring in the linker, thereby preventing this exchange reaction. The site with partial solvent-accessibility and neutral charge displayed both properties. In a mouse mammary tumor model, the stability and therapeutic activity of the antibody conjugate were affected pos. by succinimide ring hydrolysis and neg. by maleimide exchange with thiol-reactive constituents in plasma. Thus, the chem. and structural dynamics of the conjugation site can influence antibody conjugate performance by modulating the stability of the antibody-linker interface.
- 15Pillow, T. H., Tien, J., Parsons-Reponte, K. L., Bhakta, S., Li, H., Staben, L. R., Li, G., Chuh, J., Fourie-O’Donohue, A., Darwish, M. (2014) Site-specific trastuzumab maytansinoid antibody-drug conjugates with improved therapeutic activity through linker and antibody engineering. J. Med. Chem. 57 (19), 7890– 7899, DOI: 10.1021/jm500552cGoogle Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsV2htbbN&md5=084727e56c1770103910d993a71f95b1Site-Specific Trastuzumab Maytansinoid Antibody-Drug Conjugates with Improved Therapeutic Activity through Linker and Antibody EngineeringPillow, Thomas H.; Tien, Janet; Parsons-Reponte, Kathryn L.; Bhakta, Sunil; Li, Hao; Staben, Leanna R.; Li, Guangmin; Chuh, Josefa; Fourie-O'Donohue, Aimee; Darwish, Martine; Yip, Victor; Liu, Luna; Leipold, Douglas D.; Su, Dian; Wu, Elmer; Spencer, Susan D.; Shen, Ben-Quan; Xu, Keyang; Kozak, Katherine R.; Raab, Helga; Vandlen, Richard; Lewis Phillips, Gail D.; Scheller, Richard H.; Polakis, Paul; Sliwkowski, Mark X.; Flygare, John A.; Junutula, Jagath R.Journal of Medicinal Chemistry (2014), 57 (19), 7890-7899CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Antibody-drug conjugates (ADCs) have a significant impact toward the treatment of cancer, as evidenced by the clin. activity of the recently approved ADCs, brentuximab vedotin for Hodgkin lymphoma and ado-trastuzumab emtansine (trastuzumab-MCC-DM1) for metastatic HER2+ breast cancer. DM1 is an analog of the natural product maytansine, a microtubule inhibitor that by itself has limited clin. activity and high systemic toxicity. However, by conjugation of DM1 to trastuzumab, the safety was improved and clin. activity was demonstrated. Here, we report that through chem. modification of the linker-drug and antibody engineering, the therapeutic activity of trastuzumab maytansinoid ADCs can be further improved. These improvements include eliminating DM1 release in the plasma and increasing the drug load by engineering four cysteine residues into the antibody. The chem. synthesis of highly stable linker-drugs and the modification of cysteine residues of engineered site-specific antibodies resulted in a homogeneous ADC with increased therapeutic activity compared to the clin. approved ADC, trastuzumab-MCC-DM1.
- 16Tian, F., Lu, Y., Manibusan, A., Sellers, A., Tran, H., Sun, Y., Phuong, T., Barnett, R., Hehli, B., Song, F. (2014) A general approach to site-specific antibody drug conjugates. Proc. Natl. Acad. Sci. U. S. A. 111 (5), 1766– 1771, DOI: 10.1073/pnas.1321237111Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFyjs7g%253D&md5=293401bcfaa0d74ce03daca26e9ad18eA general approach to site-specific antibody drug conjugatesTian, Feng; Lu, Yingchun; Manibusan, Anthony; Sellers, Aaron; Tran, Hon; Sun, Ying; Phuong, Trung; Barnett, Richard; Hehli, Brad; Song, Frank; De Guzman, Michael J.; Ensari, Semsi; Pinkstaff, Jason K.; Sullivan, Lorraine M.; Biroc, Sandra L.; Cho, Ho; Schultz, Peter G.; Di Joseph, John; Dougher, Maureen; Ma, Dangshe; Dushin, Russell; Leal, Mauricio; Tchistiakova, Lioudmila; Feyfant, Eric; Gerber, Hans-Peter; Sapra, PujaProceedings of the National Academy of Sciences of the United States of America (2014), 111 (5), 1766-1771CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Using an expanded genetic code, antibodies with site-specifically incorporated nonnative amino acids were produced in stable cell lines derived from a CHO cell line with titers over 1 g/L. Using anti-5T4 and anti-Her2 antibodies as model systems, site-specific antibody drug conjugates (NDCs) were produced, via oxime bond formation between ketones on the side chain of the incorporated nonnative amino acid and hydroxylamine functionalized monomethyl auristatin D with either protease-cleavable or noncleavable linkers. When noncleavable linkers were used, these conjugates were highly stable and displayed improved in vitro efficacy as well as in vivo efficacy and pharmacokinetic stability in rodent models relative to conventional antibody drug conjugates conjugated through either engineered surface-exposed or reduced interchain disulfide bond cysteine residues. The advantages of the oxime-bonded, site-specific NDCs were even more apparent when low-antigen-expressing (2+) target cell lines were used in the comparative studies. NDCs generated with protease-cleavable linkers demonstrated that the site of conjugation had a significant impact on the stability of these rationally designed prodrug linkers. In a single-dose rat toxicol. study, a site-specific anti-Her2 NDC was well tolerated at dose levels up to 90 mg/kg. These expts. support the notion that chem. defined antibody conjugates can be synthesized in com. relevant yields and can lead to antibody drug conjugates with improved properties relative to the heterogeneous conjugates formed by nonspecific chem. modification.
- 17Jackson, D., Atkinson, J., Guevara, C. I., Zhang, C., Kery, V., Moon, S. J., Virata, C., Yang, P., Lowe, C., Pinkstaff, J. (2014) In vitro and in vivo evaluation of cysteine and site specific conjugated herceptin antibody-drug conjugates. PLoS One 9 (1), e83865 DOI: 10.1371/journal.pone.0083865Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXivVOit7g%253D&md5=02721a9b2d6982d9915c76d604cebac3In vitro and in vivo evaluation of cysteine and site specific conjugated herceptin antibody-drug conjugatesJackson, Dowdy; Atkinson, John; Guevara, Claudia I.; Zhang, Chunying; Kery, Vladimir; Moon, Sung-Ju; Virata, Cyrus; Yang, Peng; Lowe, Christine; Pinkstaff, Jason; Cho, Ho; Knudsen, Nick; Manibusan, Anthony; Tian, Feng; Sun, Ying; Lu, Yingchun; Sellers, Aaron; Jia, Xiao-Chi; Joseph, Ingrid; Anand, Banmeet; Morrison, Kendall; Pereira, Daniel S.; Stover, DavidPLoS One (2014), 9 (1), e83865/1-e83865/14, 14 pp.CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Antibody-drug conjugates (ADCs) are monoclonal antibodies designed to deliver a cytotoxic drug selectively to antigen expressing cells. Several components of an ADC including the selection of the antibody, the linker, the cytotoxic drug payload and the site of attachment used to attach the drug to the antibody are crit. to the activity and development of the ADC. The cytotoxic drugs or payloads used to make ADCs are typically conjugated to the antibody through cysteine or lysine residues. This results in ADCs that have a heterogeneous no. of drugs per antibody. The no. of drugs per antibody commonly referred to as the drug to antibody ratio (DAR), can vary between 0 and 8 drugs for a IgG1 antibody. Antibodies with 0 drugs are ineffective and compete with the ADC for binding to the antigen expressing cells. Antibodies with 8 drugs per antibody have reduced in vivo stability, which may contribute to non target related toxicities. In these studies we incorporated a non-natural amino acid, para acetyl phenylalanine, at two unique sites within an antibody against Her2/neu. We covalently attached a cytotoxic drug to these sites to form an ADC which contains two drugs per antibody. We report the results from the first direct preclin. comparison of a site specific non-natural amino acid anti-Her2 ADC and a cysteine conjugated anti-Her2 ADC. We report that the site specific non-natural amino acid anti-Her2 ADCs have superior in vitro serum stability and preclin. toxicol. profile in rats as compared to the cysteine conjugated anti-Her2 ADCs. We also demonstrate that the site specific non-natural amino acid anti-Her2 ADCs maintain their in vitro potency and in vivo efficacy against Her2 expressing human tumor cell lines. Our data suggests that site specific non-natural amino acid ADCs may have a superior therapeutic window than cysteine conjugated ADCs.
- 18Strop, P., Delaria, K., Foletti, D., Witt, J. M., Hasa-Moreno, A., Poulsen, K., Casas, M. G., Dorywalska, M., Farias, S., Pios, A. (2015) Site-specific conjugation improves therapeutic index of antibody drug conjugates with high drug loading. Nat. Biotechnol. 33 (7), 694– 696, DOI: 10.1038/nbt.3274Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1ensLbK&md5=43b6c9f702e02a91887c3f27a5ab7353Site-specific conjugation improves therapeutic index of antibody drug conjugates with high drug loadingStrop, Pavel; Delaria, Kathy; Foletti, Davide; Witt, Jody Melton; Hasa-Moreno, Adela; Poulsen, Kris; Casas, Meritxell Galindo; Dorywalska, Magdalena; Farias, Santiago; Pios, Ariel; Lui, Victor; Dushin, Russell; Zhou, Dahui; Navaratnam, Thayalan; Tran, Thomas-Toan; Sutton, Janette; Lindquist, Kevin C.; Han, Bora; Liu, Shu-Hui; Shelton, David L.; Pons, Jaume; Rajpal, ArvindNature Biotechnology (2015), 33 (7), 694-696CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)There is no expanded citation for this reference.
- 19Behrens, C. R., Ha, E. H., Chinn, L. L., Bowers, S., Probst, G., Fitch-Bruhns, M., Monteon, J., Valdiosera, A., Bermudez, A., Liao-Chan, S. (2015) Antibody-Drug Conjugates (ADCs) Derived from Interchain Cysteine Cross-Linking Demonstrate Improved Homogeneity and Other Pharmacological Properties over Conventional Heterogeneous ADCs. Mol. Pharmaceutics 12 (11), 3986– 3998, DOI: 10.1021/acs.molpharmaceut.5b00432Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFejtLjE&md5=d3657c76c415b621877a90b0d5a82025Antibody-Drug Conjugates (ADCs) Derived from Interchain Cysteine Cross-Linking Demonstrate Improved Homogeneity and Other Pharmacological Properties over Conventional Heterogeneous ADCsBehrens, Christopher R.; Ha, Edward H.; Chinn, Lawrence L.; Bowers, Simeon; Probst, Gary; Fitch-Bruhns, Maureen; Monteon, Jorge; Valdiosera, Amanda; Bermudez, Abel; Liao-Chan, Sindy; Wong, Tiffany; Melnick, Jonathan; Theunissen, Jan-Willem; Flory, Mark R.; Houser, Derrick; Venstrom, Kristy; Levashova, Zoia; Sauer, Paul; Migone, Thi-Sau; van der Horst, Edward H.; Halcomb, Randall L.; Jackson, David Y.Molecular Pharmaceutics (2015), 12 (11), 3986-3998CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Conventional antibody-drug conjugates (ADCs) are heterogeneous mixts. of chem. distinct mols. that vary in both drugs/antibody (DAR) and conjugation sites. Suboptimal properties of heterogeneous ADCs have led to new site-specific conjugation methods for improving ADC homogeneity. Most site-specific methods require extensive antibody engineering to identify optimal conjugation sites and introduce unique functional groups for conjugation with appropriately modified linkers. Alternative nonrecombinant methods have emerged in which bifunctional linkers are utilized to cross-link antibody interchain cysteines and afford ADCs contg. four drugs/antibody. Although these methods have been shown to improve ADC homogeneity and stability in vitro, their effect on the pharmacol. properties of ADCs in vivo is unknown. In order to det. the relative impact of interchain cysteine crosslinking on the therapeutic window and other properties of ADCs in vivo, we synthesized a deriv. of the known ADC payload, MC-MMAF, that contains a bifunctional dibromomaleimide (DBM) linker instead of a conventional maleimide (MC) linker. The DBM-MMAF deriv. was conjugated to trastuzumab and a novel anti-CD98 antibody to afford ADCs contg. predominantly four drugs/antibody. The pharmacol. properties of the resulting cross-linked ADCs were compared with analogous heterogeneous ADCs derived from conventional linkers. The results demonstrate that DBM linkers can be applied directly to native antibodies, without antibody engineering, to yield highly homogeneous ADCs via cysteine crosslinking. The resulting ADCs demonstrate improved pharmacokinetics, superior efficacy, and reduced toxicity in vivo compared to analogous conventional heterogeneous ADCs.
- 20Hamblett, K. J., Le, T., Rock, B. M., Rock, D. A., Siu, S., Huard, J. N., Conner, K. P., Milburn, R. R., O’Neill, J. W., Tometsko, M. E. (2016) Altering Antibody–Drug Conjugate Binding to the Neonatal Fc Receptor Impacts Efficacy and Tolerability. Mol. Pharmaceutics 13 (7), 2387– 2396, DOI: 10.1021/acs.molpharmaceut.6b00153Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XovVylsbg%253D&md5=8883777d4731222b197f1d0109415f5bAltering Antibody-Drug Conjugate Binding to the Neonatal Fc Receptor Impacts Efficacy and TolerabilityHamblett, Kevin J.; Le, Tiep; Rock, Brooke M.; Rock, Dan A.; Siu, Sophia; Huard, Justin N.; Conner, Kip P.; Milburn, Robert R.; O'Neill, Jason W.; Tometsko, Mark E.; Fanslow, William C.Molecular Pharmaceutics (2016), 13 (7), 2387-2396CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Antibody-drug conjugates (ADC) rely on the target-binding specificity of an antibody to selectively deliver potent drugs to cancer cells. IgG antibody half-life is regulated by neonatal Fc receptor (FcRn) binding. Histidine 435 of human IgG was mutated to alanine (H435A) to explore the effect of FcRn binding on the pharmacokinetics, efficacy, and tolerability of two sep. maytansine-based ADC pairs with noncleavable linkers, (c-DM1 and c-H435A-DM1) and (7v-Cys-may and 7v-H435A-Cys-may). The in vitro cell-killing potency of each pair of ADCs was similar, demonstrating that H435A showed no measurable impact on ADC bioactivity. The H435A mutant antibodies showed no detectable binding to human or mouse FcRn in vitro, whereas their counterpart wild-type IgG ADCs were found to bind to FcRn at pH = 6.0. In xenograft bearing SCID mice expressing mouse FcRn, the AUC of 7v-Cys-may was 1.6-fold higher than that of 7v-H435A-may, yet the obsd. efficacy was similar. More severe thrombocytopenia was obsd. with 7v-H435A-Cys-may as compared to 7v-Cys-may at multiple dose levels. The AUC of c-DM1 was approx. 3-fold higher than that of c-H435A-DM1 in 786-0 xenograft bearing SCID mice, which led to a 3-fold difference in efficacy by dose. Murine FcRn knockout, human FcRn transgenic line 32 SCID animals bearing 786-0 xenografts showed an amplified exposure difference between c-DM1 and c-H435A-DM1 as compared to murine FcRn expressing SCID mice, leading to a 10-fold higher dose required for efficacy despite a 6-fold higher AUC of the c-H435A-DM1. The accelerated clearance obsd. for the noncleavable maytansine ADCs with the H435A FcRn mutation led to reduced efficacy at equiv. doses and exacerbation of clin. pathol. parameters (decreased tolerability) at equiv. doses. The results show that reduced ADC clearance mediated by FcRn modulation can improve therapeutic index.
- 21Lhospice, F., Bregeon, D., Belmant, C., Dennler, P., Chiotellis, A., Fischer, E., Gauthier, L., Boedec, A., Rispaud, H., Savard-Chambard, S. (2015) Site-Specific Conjugation of Monomethyl Auristatin E to Anti-CD30 Antibodies Improves Their Pharmacokinetics and Therapeutic Index in Rodent Models. Mol. Pharmaceutics 12 (6), 1863– 1871, DOI: 10.1021/mp500666jGoogle Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsV2gsrs%253D&md5=ef6ab4dab9fa40d15d03c98a58917cd0Site-Specific Conjugation of Monomethyl Auristatin E to Anti-CD30 Antibodies Improves Their Pharmacokinetics and Therapeutic Index in Rodent ModelsLhospice, F.; Bregeon, D.; Belmant, C.; Dennler, P.; Chiotellis, A.; Fischer, E.; Gauthier, L.; Boedec, A.; Rispaud, H.; Savard-Chambard, S.; Represa, A.; Schneider, N.; Paturel, C.; Sapet, M.; Delcambre, C.; Ingoure, S.; Viaud, N.; Bonnafous, C.; Schibli, R.; Romagne, F.Molecular Pharmaceutics (2015), 12 (6), 1863-1871CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Antibody-drug conjugates (ADCs) have demonstrated clin. benefits that have led to the recent FDA approval of KADCYLA and ADCETRIS. Most ADCs that are currently in clin. use or development, including ADCETRIS, are produced by chem. conjugation of a toxin via either lysine or cysteine residues, inevitably leading to heterogeneous products with variable drug-to-antibody ratios (DARs). Here, the authors describe the in vitro and in vivo characterization of four novel ADCs that are based on the anti-CD30 antibody cAC10, which has the same polypeptide backbone as ADCETRIS, and compare the results with the latter. Bacterial transglutaminase (BTG) was exploited to site-specifically conjugate derivs. of monomethyl auristatin E (all comprising a cleavable linker) to the glutamine at positions 295 and 297 of cAC10, thereby yielding homogeneous ADCs with a DAR of 4. In vitro cell toxicity expts. using two different CD30-pos. cell lines (Karpas 299 and Raji-CD30+) revealed comparable EC50 values for ADCETRIS (1.8±0.4 and 3.6±0.6 ng/mL, resp.) and the four cAC10-based ADCs (2.0±0.4 to 4.9±1.0 ng/mL). Quant. time-dependent in vivo biodistribution studies (3-96 h p.i.) in normal and xenografted (Karpas 299 cells) SCID mice were performed with a selected 125I-radioiodinated cAC10 ADC and compared with that of 125I-ADCETRIS. The chemo-enzymically conjugated, radioiodinated ADC showed higher tumor uptake (17.84±2.2% ID/g 24 h p.i.) than 125I-ADCETRIS (10.5±1.8% ID/g 24 h p.i.). Moreover, 125I-ADCETRIS exhibited higher nontargeted liver and spleen uptake. In line with these results, the max. tolerated dose of the BTG-coupled ADC (>60 mg/kg) was significantly higher than that of ADCETRIS (18 mg/kg) in rats. These results suggest that homogeneous ADCs display improved pharmacokinetics and better therapeutic indexes compared to those of chem. modified ADCs with variable DARs.
- 22Yoder, N. C., Bai, C., Tavares, D., Widdison, W. C., Whiteman, K. R., Wilhelm, A., Wilhelm, S. D., McShea, M. A., Maloney, E. K., Ab, O. (2019) A Case Study Comparing Heterogeneous Lysine- and Site-Specific Cysteine-Conjugated Maytansinoid Antibody-Drug Conjugates (ADCs) Illustrates Benefits of Lysine Conjugation. Mol. Pharmaceutics 16 (9), 3926– 3937, DOI: 10.1021/acs.molpharmaceut.9b00529Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlaqsrrL&md5=d88fd3fc17185f75b3ba7ac6afc5bb30A Case Study Comparing Heterogeneous Lysine- and Site-Specific Cysteine-Conjugated Maytansinoid Antibody-Drug Conjugates (ADCs) Illustrates the Benefits of Lysine ConjugationYoder, Nicholas C.; Bai, Chen; Tavares, Daniel; Widdison, Wayne C.; Whiteman, Kathleen R.; Wilhelm, Alan; Wilhelm, Sharon D.; McShea, Molly A.; Maloney, Erin K.; Ab, Olga; Wang, Lintao; Jin, Shan; Erickson, Hans K.; Keating, Thomas A.; Lambert, John M.Molecular Pharmaceutics (2019), 16 (9), 3926-3937CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Antibody-drug conjugates are an emerging class of cancer therapeutics constructed from monoclonal antibodies conjugated with small mol. effectors. First-generation mols. of this class often employed heterogeneous conjugation chem., but many site-specifically conjugated ADCs have been described recently. Here, we undertake a systematic comparison of ADCs made with the same antibody and the same macrocyclic maytansinoid effector but conjugated either heterogeneously at lysine residues or site-specifically at cysteine residues. Characterization of these ADCs in vitro reveals generally similar properties, including a similar catabolite profile, a key element in making a meaningful comparison of conjugation chemistries. In a mouse model of cervical cancer, the lysine-conjugated ADC affords greater efficacy on a molar payload basis. Rather than making general conclusions about ADCs conjugated by a particular chem., we interpret these results as highlighting the complexity of ADCs and the interplay between payload class, linker chem., target antigen, and other variables that det. efficacy in a given setting.
- 23Miller, M. L., Shizuka, M., Wilhelm, A., Salomon, P., Reid, E. E., Lanieri, L., Sikka, S., Maloney, E. K., Harvey, L., Qiu, Q. (2018) A DNA-Interacting Payload Designed to Eliminate Cross-Linking Improves the Therapeutic Index of Antibody-Drug Conjugates (ADCs). Mol. Cancer Ther. 17 (3), 650– 660, DOI: 10.1158/1535-7163.MCT-17-0940Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjslKlu7c%253D&md5=aa83bf47124ecc290bd3e14f56df8b1fA DNA-Interacting Payload Designed to Eliminate Cross-Linking Improves the Therapeutic Index of Antibody-Drug Conjugates (ADCs)Miller, Michael L.; Shizuka, Manami; Wilhelm, Alan; Salomon, Paulin; Reid, Emily E.; Lanieri, Leanne; Sikka, Surina; Maloney, Erin K.; Harvey, Lauren; Qiu, Qifeng; Archer, Katie E.; Bai, Chen; Vitharana, Dilrukshi; Harris, Luke; Singh, Rajeeva; Ponte, Jose F.; Yoder, Nicholas C.; Kovtun, Yelena; Lai, Katharine C.; Ab, Olga; Pinkas, Jan; Keating, Thomas A.; Chari, Ravi V. J.Molecular Cancer Therapeutics (2018), 17 (3), 650-660CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)Tumor-selective delivery of cytotoxic agents in the form of antibody-drug conjugates (ADCs) is now a clin. validated approach for cancer treatment. In an attempt to improve the clin. success rate of ADCs, emphasis has been recently placed on the use of DNA-crosslinking pyrrolobenzodiazepine compds. as the payload. Despite promising early clin. results with this class of ADCs, doses achievable have been low due to systemic toxicity. Here, we describe the development of a new class of potent DNA-interacting agents wherein changing the mechanism of action from a cross-linker to a DNA alkylator improves the tolerability of the ADC. ADCs contg. the DNA alkylator displayed similar in vitro potency, but improved bystander killing and in vivo efficacy, compared with those of the cross-linker. Thus, the improved in vivo tolerability and antitumor activity achieved in rodent models with ADCs of the novel DNA alkylator could provide an efficacious, yet safer option for cancer treatment. Mol Cancer Ther; 17(3); 650-60. ©2018 AACR.
- 24Stimmel, J. B., Merrill, B. M., Kuyper, L. F., Moxham, C. P., Hutchins, J. T., Fling, M. E., and Kull, F. C. (2000) Site-specific conjugation on serine -> cysteine variant monoclonal antibodies. J. Biol. Chem. 275 (39), 30445– 30450, DOI: 10.1074/jbc.M001672200Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXnt1agsbY%253D&md5=0763640e21082823d1b0b83663389f86Site-specific conjugation on serine → cysteine variant monoclonal antibodiesStimmel, Julie B.; Merrill, Barbara M.; Kuyper, Lee F.; Moxham, Cary P.; Hutchins, Jeff T.; Fling, Mary E.; Kull, Frederick C., Jr.Journal of Biological Chemistry (2000), 275 (39), 30445-30450CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)We have engineered a cysteine residue at position 442 (EU/OU numbering) in the third const. domain (CH3) of the heavy chain of several IgGs with different specificities, isoforms, and variants with the intent to introduce a site for chem. conjugation. The variants were expressed in NS0 mouse myeloma cells, where monomeric IgG is the major form and formation of aggregate was minimal. Monomeric IgG contained no free thiol; however, it was discovered that the engineered thiols were reversibly blocked and could be reduced under controlled conditions. Following redn., reactive thiol was conjugated with a cysteine-specific bifunctional chelator, bromoacetyl-TMT to a humanized 323/A3 IgG4 variant. Conjugation had no significant effect on antibody affinity. To prove that the conjugation was site-specific, an antibody-TMT conjugate was labeled with lutetium-177 and subjected to peptide mapping followed by sequence anal. Glu-C digestion demonstrated that 91% of the label was recovered in the COOH-terminal peptide fragment contg. the engineered cysteine.
- 25Cao, M., De Mel, N., Jiao, Y., Howard, J., Parthemore, C., Korman, S., Thompson, C., Wendeler, M., and Liu, D. (2019) Site-specific antibody-drug conjugate heterogeneity characterization and heterogeneity root cause analysis. MAbs 11 (6), 1064– 1076, DOI: 10.1080/19420862.2019.1624127Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFOjsrrN&md5=565ad2567367d9fad04088d85523a876Site-specific antibody-drug conjugate heterogeneity characterization and heterogeneity root cause analysisCao, Mingyan; De Mel, Niluka; Jiao, Yang; Howard, James; Parthemore, Conner; Korman, Samuel; Thompson, Christopher; Wendeler, Michaela; Liu, DengfengmAbs (2019), 11 (6), 1064-1076CODEN: MABSCP; ISSN:1942-0870. (Taylor & Francis, Inc.)Site-specific ADCs are designed to overcome heterogeneity obsd. with first-generation ADCs that use random conjugation to surface-exposed lysine residues or conjugation to interchain disulfide bonds. An elevated level of size variants, such as heavy chain-light chain species, heavy chain-heavy chain-light chain species, and light chain species, is also obsd. with final site-specific ADC product. To understand root cause of heterogeneity generated during ADC conjugation process, we designed time-course studies for each conjugation step, including redn., oxidn., conjugation, and quenching. We developed both non-reduced peptide map and LabChip-based capillary electrophoresis sodium dodecyl sulfate methods for time-course sample anal. On basis of our time-course data, the half ADC and unconjugated antibody were generated during oxidn. as a result of alternative disulfide bond arrangements. During oxidn., two hinge cysteines formed intra-chain disulfide bond in the half ADC, and three inter-chain hinge disulfide bonds were formed in unconjugated antibody. Time-course data also showed that elevated level of size variants, esp. heavy chain-heavy chain-light chain species and light chain species, resulted from quenching step, where quenching reagent engaged in disulfide bond exchange reaction with ADC and broke disulfide bonds connecting heavy chain and light chain. Underconjugated and overconjugated species arose from equil. established during conjugation reaction.
- 26Vollmar, B. S., Wei, B., Ohri, R., Zhou, J., He, J., Yu, S. F., Leipold, D., Cosino, E., Yee, S., Fourie-O’Donohue, A. (2017) Attachment Site Cysteine Thiol pKa Is a Key Driver for Site-Dependent Stability of THIOMAB Antibody-Drug Conjugates. Bioconjugate Chem. 28 (10), 2538– 2548, DOI: 10.1021/acs.bioconjchem.7b00365Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsVGks7vL&md5=ba1805c493f468d4ffbb198dbf8b3d02Attachment Site Cysteine Thiol pKa Is a Key Driver for Site-Dependent Stability of THIOMAB Antibody-Drug ConjugatesVollmar, Breanna S.; Wei, Binqing; Ohri, Rachana; Zhou, Jianhui; He, Jintang; Yu, Shang-Fan; Leipold, Douglas; Cosino, Ely; Yee, Sharon; Fourie-O'Donohue, Aimee; Li, Guangmin; Phillips, Gail L.; Kozak, Katherine R.; Kamath, Amrita; Xu, Keyang; Lee, Genee; Lazar, Greg A.; Erickson, Hans K.Bioconjugate Chemistry (2017), 28 (10), 2538-2548CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The incorporation of cysteines into antibodies by mutagenesis allows for the direct conjugation of small mols. to specific sites on the antibody via disulfide bonds. The stability of the disulfide bond linkage between the small mol. and the antibody is highly dependent on the location of the engineered cysteine in either the heavy chain (HC) or the light chain (LC) of the antibody. Here, we explore the basis for this site-dependent stability. We evaluated the in vivo efficacy and pharmacokinetics of five different cysteine mutants of trastuzumab conjugated to a pyrrolobenzodiazepine (PBD) via disulfide bonds. A significant correlation was obsd. between disulfide stability and efficacy for the conjugates. We hypothesized that the obsd. site-dependent stability of the disulfide-linked conjugates could be due to differences in the attachment site cysteine thiol pKa. We measured the cysteine thiol pKa using isothermal titrn. calorimetry (ITC) and found that the variants with the highest thiol pKa (LC K149C and HC A140C) were found to yield the conjugates with the greatest in vivo stability. Guided by homol. modeling, we identified several mutations adjacent to LC K149C that reduced the cysteine thiol pKa and, thus, decreased the in vivo stability of the disulfide-linked PBD conjugated to LC K149C. We also present results suggesting that the high thiol pKa of LC K149C is responsible for the sustained circulation stability of LC K149C TDCs utilizing a maleimide-based linker. Taken together, our results provide evidence that the site-dependent stability of cys-engineered antibody-drug conjugates may be explained by interactions between the engineered cysteine and the local protein environment that serves to modulate the side-chain thiol pKa. The influence of cysteine thiol pKa on stability and efficacy offers a new parameter for the optimization of ADCs that utilize cysteine engineering.
- 27Jeffrey, S. C., Burke, P. J., Lyon, R. P., Meyer, D. W., Sussman, D., Anderson, M., Hunter, J. H., Leiske, C. I., Miyamoto, J. B., Nicholas, N. D. (2013) A potent anti-CD70 antibody-drug conjugate combining a dimeric pyrrolobenzodiazepine drug with site-specific conjugation technology. Bioconjugate Chem. 24 (7), 1256– 1263, DOI: 10.1021/bc400217gGoogle Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptFKjs7c%253D&md5=31841b0267e41ae861eaf05240c97b15A Potent Anti-CD70 Antibody-Drug Conjugate Combining a Dimeric Pyrrolobenzodiazepine Drug with Site-Specific Conjugation TechnologyJeffrey, Scott C.; Burke, Patrick J.; Lyon, Robert P.; Meyer, David W.; Sussman, Django; Anderson, Martha; Hunter, Joshua H.; Leiske, Chris I.; Miyamoto, Jamie B.; Nicholas, Nicole D.; Okeley, Nicole M.; Sanderson, Russell J.; Stone, Ivan J.; Zeng, Weiping; Gregson, Stephen J.; Masterson, Luke; Tiberghien, Arnaud C.; Howard, Philip W.; Thurston, David E.; Law, Che-Leung; Senter, Peter D.Bioconjugate Chemistry (2013), 24 (7), 1256-1263CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)A highly cytotoxic DNA crosslinking pyrrolobenzodiazepine (PBD) dimer with a valine-alanine dipeptide linker was conjugated to the anti-CD70 h1F6 mAb either through endogenous interchain cysteines or, site-specifically, through engineered cysteines at position 239 of the heavy chains. The h1F6239C-PBD conjugation strategy proved to be superior to interchain cysteine conjugation, affording an antibody-drug conjugate (ADC) with high uniformity in drug-loading and low levels of aggregation. In vitro cytotoxicity expts. demonstrated that the h1F6239C-PBD was potent and immunol. specific on CD70-pos. renal cell carcinoma (RCC) and non-Hodgkin lymphoma (NHL) cell lines. The conjugate was resistant to drug loss in plasma and in circulation, and had a pharmacokinetic profile closely matching that of the parental h1F6239C antibody capped with N-ethylmaleimide (NEM). Evaluation in CD70-pos. RCC and NHL mouse xenograft models showed pronounced antitumor activities at single or weekly doses as low as 0.1 mg/kg of ADC. The ADC was tolerated at 2.5 mg/kg. These results demonstrate that PBDs can be effectively used for antibody-targeted therapy.
- 28Hamblett, K. J., Senter, P. D., Chace, D. F., Sun, M. M. C., Lenox, J., Cerveny, C. G., Kissler, K. M., Bernhardt, S. X., Kopcha, A. K., Zabinski, R. F. (2004) Effects of Drug Loading on the Antitumor Activity of a Monoclonal Antibody Drug Conjugate. Clin. Cancer Res. 10 (20), 7063– 7070, DOI: 10.1158/1078-0432.CCR-04-0789Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXpslGlu78%253D&md5=2d2dd7a498d1181a57dcd3ab8b931e65Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugateHamblett, Kevin J.; Senter, Peter D.; Chace, Dana F.; Sun, Michael M. C.; Lenox, Joel; Cerveny, Charles G.; Kissler, Kim M.; Bernhardt, Starr X.; Kopcha, Anastasia K.; Zabinski, Roger F.; Meyer, Damon L.; Francisco, Joseph A.Clinical Cancer Research (2004), 10 (20), 7063-7070CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)An antibody-drug conjugate consisting of monomethyl auristatin E (MMAE) conjugated to the anti-CD30 monoclonal antibody (mAb) cAC10, with eight drug moieties per mAb, was previously shown to have potent cytotoxic activity against CD30+ malignant cells. To det. the effect of drug loading on antibody-drug conjugate therapeutic potential, we assessed cAC10 antibody-drug conjugates contg. different drug-mAb ratios in vitro and in vivo. Coupling MMAE to the cysteines that comprise the interchain disulfides of cAC10 created an antibody-drug conjugate population, which was purified using hydrophobic interaction chromatog. to yield antibody-drug conjugates with two, four, and eight drugs per antibody (E2, E4, and E8, resp.). Antibody-drug conjugate potency was tested in vitro against CD30+ lines followed by in vivo xenograft models. The max.-tolerated dose and pharmacokinetic profiles of the antibody-drug conjugates were investigated in mice. Although antibody-drug conjugate potency in vitro was directly dependent on drug loading (IC50 values E8<E4<E2), the in vivo antitumor activity of E4 was comparable with E8 at equal mAb doses, although the E4 contained half the amt. of MMAE per mAb. E2 was also an active antitumor agent but required higher doses. The max.-tolerated dose of E2 in mice was at least double that of E4, which in turn was twice that of E8. MMAE loading affected plasma clearance, as E8 cleared 3-fold faster than E4 and 5-fold faster than E2. By decreasing drug loading per antibody, the therapeutic index was increased demonstrating that drug loading is a key design parameter for antibody-drug conjugates.
- 29Sun, X., Ponte, J. F., Yoder, N. C., Laleau, R., Coccia, J., Lanieri, L., Qiu, Q., Wu, R., Hong, E., Bogalhas, M. (2017) Effects of Drug-Antibody Ratio on Pharmacokinetics, Biodistribution, Efficacy, and Tolerability of Antibody-Maytansinoid Conjugates. Bioconjugate Chem. 28 (5), 1371– 1381, DOI: 10.1021/acs.bioconjchem.7b00062Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlslCjurg%253D&md5=60e21df4b6e3189d87a43b0d650f51b4Effects of Drug-Antibody Ratio on Pharmacokinetics, Biodistribution, Efficacy, and Tolerability of Antibody-Maytansinoid ConjugatesSun, Xiuxia; Ponte, Jose F.; Yoder, Nicholas C.; Laleau, Rassol; Coccia, Jennifer; Lanieri, Leanne; Qiu, Qifeng; Wu, Rui; Hong, Erica; Bogalhas, Megan; Wang, Lintao; Dong, Ling; Setiady, Yulius; Maloney, Erin K.; Ab, Olga; Zhang, Xiaoyan; Pinkas, Jan; Keating, Thomas A.; Chari, Ravi; Erickson, Hans K.; Lambert, John M.Bioconjugate Chemistry (2017), 28 (5), 1371-1381CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Antibody-drug conjugates (ADCs) are being actively pursued as a treatment option for cancer following the regulatory approval of brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla). ADCs consist of a cytotoxic agent conjugated to a targeting antibody through a linker. The two approved ADCs (and most ADCs now in the clinic that use a microtubule disrupting agent as the payload) are heterogeneous conjugates with an av. drug to antibody ratio (DAR) of 3-4 (potentially ranging from 0-8 for individual species). Ado-trastuzumab emtansine employs DM1, a semisynthetic cytotoxic payload of the maytansinoid class, which is conjugated via lysine residues of the antibody to an av. DAR of 3.5. To understand the effect of DAR on the preclin. properties of ADCs using maytansinoid cytotoxic agents, we prepd. a series of conjugates with a cleavable linker (M9346A-sulfo-SPDB-DM4 targeting folate receptor alpha (FR)) or an uncleavable linker (J2898A-SMCC-DM1 targeting the epidermal growth factor receptor (EGFR)) with varying DAR, and evaluated their biochem. characteristics, in vivo stability, efficacy, and tolerability. For both formats, a series of ADCs with DARs ranging from low (av. ∼2, range 0-4) to very high (av. 10, range 7-14) were prepd. in good yield with high monomer content and low levels of free cytotoxic agent. The in vitro potency consistently increased with increasing DAR, at const. antibody concn. We then characterized the in vivo disposition of these ADCs. Pharmacokinetic anal. showed that conjugates with an av. DAR below ∼6 had comparable clearance rates, but for those with an av. DAR around 9-10, rapid clearance was obsd. Biodistribution studies in mice showed that these 9-10 DAR ADCs rapidly accumulate in the liver, with max. localization for this organ at 24-28% percentage injected dose per g (%ID/g) compared with 7-10% for lower DAR conjugates (all at 2-6 h post-injection). Our preclin. findings on tolerability and efficacy suggest that maytansinoid conjugates with DAR ranging from 2-6 have a better therapeutic index than conjugates with very high DAR (∼9-10). These very high DAR ADCs suffer from decreased efficacy likely due to faster clearance. These results support the use of DAR 3-4 for maytansinoid ADCs, but suggest the exploration of lower or higher DAR may be warranted depending on the biol. of the target antigen.
- 30Thompson, P., Fleming, R., Bezabeh, B., Huang, F., Mao, S., Chen, C., Harper, J., Zhong, H., Gao, X., Yu, X.-Q. (2016) Rational design, biophysical and biological characterization of site-specific antibody-tubulysin conjugates with improved stability, efficacy and pharmacokinetics. J. Controlled Release 236, 100– 116, DOI: 10.1016/j.jconrel.2016.06.025Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtFehtrbF&md5=61154de48ed3a8ab3586e2815ae99459Rational design, biophysical and biological characterization of site-specific antibody-tubulysin conjugates with improved stability, efficacy and pharmacokineticsThompson, Pamela; Fleming, Ryan; Bezabeh, Binyam; Huang, Fengying; Mao, Shenlan; Chen, Cui; Harper, Jay; Zhong, Haihong; Gao, Xizhe; Yu, Xiang-Qing; Hinrichs, Mary Jane; Reed, Molly; Kamal, Adeela; Strout, Patrick; Cho, Song; Woods, Rob; Hollingsworth, Robert E.; Dixit, Rakesh; Wu, Herren; Gao, Changshou; Dimasi, NazzarenoJournal of Controlled Release (2016), 236 (), 100-116CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Antibody-drug conjugates (ADCs) are among the most promising empowered biologics for cancer treatment. ADCs are commonly prepd. by chem. conjugation of small mol. cytotoxic anti-cancer drugs to antibodies through either lysine side chains or cysteine thiols generated by the redn. of interchain disulfide bonds. Both methods yield heterogeneous conjugates with complex biophys. properties and suboptimal serum stability, efficacy, and pharmacokinetics. To limit the complexity of cysteine-based ADCs, we have engineered and characterized in vitro and in vivo antibody cysteine variants that allow precise control of both site of conjugation and drug load per antibody mol. We demonstrate that the chem.-defined cysteine-engineered antibody-tubulysin conjugates have improved ex vivo and in vivo stability, efficacy, and pharmacokinetics when compared to conventional cysteine-based ADCs with similar drug-to-antibody ratios. In addn., to limit the non-target FcγRs mediated uptake of the ADCs by cells of the innate immune system, which may result in off-target toxicities, the ADCs have been engineered to lack Fc-receptor binding. The strategies described herein are broadly applicable to any full-length IgG or Fc-based ADC and have been incorporated into an ADC that is in phase I clin. development.
- 31Ouyang, J. (2013) Drug-to-antibody ratio (DAR) and drug load distribution by hydrophobic interaction chromatography and reversed phase high-performance liquid chromatography. Methods Mol. Biol. (N. Y., NY, U. S.) 1045, 275– 283, DOI: 10.1007/978-1-62703-541-5_17Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3sfms1Oqtg%253D%253D&md5=933437e336d82a976a50610aa79b4d0bDrug-to-antibody ratio (DAR) and drug load distribution by hydrophobic interaction chromatography and reversed phase high-performance liquid chromatographyOuyang JunMethods in molecular biology (Clifton, N.J.) (2013), 1045 (), 275-83 ISSN:.Hydrophobic interaction chromatography (HIC) is the method of choice for determination of the drug-to-antibody ratio (DAR) and drug load distribution for cysteine (Cys)-linked antibody-drug conjugates (ADCs). The drug-loaded species are resolved based on the increasing hydrophobicity with the least hydrophobic, unconjugated form eluting first and the most hydrophobic, 8-drug form eluting last. The area percentage of a peak represents the relative distribution of the particular drug-loaded ADC species. The weighted average DAR is then calculated using the percentage peak area information and the drug load numbers. Reversed phase high-performance liquid chromatography (RP-HPLC) offers an orthogonal method to obtain DAR for Cys-linked ADCs. The method involves, first, a reduction reaction to completely dissociate the heavy and light chains of the ADC, then separation of the light and heavy chains and their corresponding drug-loaded forms on an RP column. The percentage peak area from integration of the light chain and heavy chain peaks, combined with the assigned drug load for each peak, is used to calculate the weighted average DAR.
- 32Fekete, S., Veuthey, J. L., Beck, A., and Guillarme, D. (2016) Hydrophobic interaction chromatography for the characterization of monoclonal antibodies and related products. J. Pharm. Biomed. Anal. 130, 3– 18, DOI: 10.1016/j.jpba.2016.04.004Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmtVSgtb0%253D&md5=2e13c1de1d790e013013ee462660f69eHydrophobic interaction chromatography for the characterization of monoclonal antibodies and related productsFekete, Szabolcs; Veuthey, Jean-Luc; Beck, Alain; Guillarme, DavyJournal of Pharmaceutical and Biomedical Analysis (2016), 130 (), 3-18CODEN: JPBADA; ISSN:0731-7085. (Elsevier B.V.)A review. Hydrophobic interaction chromatog. (HIC) is a historical strategy used for the anal. purifn. and characterization of proteins. Similarly to what can be done in reversed-phase liq. chromatog. (RPLC), HIC is able to sep. protein species based on their hydrophobicity, but using different conditions. Compared to RPLC, the main benefit of HIC is its ability to perform sepns. under non denaturing conditions (i.e. physiol. pH conditions, ambient mobile phase temp. and no need for org. solvents) and so an orthogonal method. The goal of this review is to provide a general overview of theor. and practical aspects of modern HIC applied for the characterization of therapeutic protein biopharmaceuticals including monoclonal antibodies (mAbs), antibody drug conjugates (ADCs) and bispecific antibodies (bsAbs). Therefore, method development approaches, state-of-the-art column technol., applications and future perspectives are described and critically discussed.
- 33Strop, P., Liu, S. H., Dorywalska, M., Delaria, K., Dushin, R. G., Tran, T. T., Ho, W. H., Farias, S., Casas, M. G., Abdiche, Y. (2013) Location matters: site of conjugation modulates stability and pharmacokinetics of antibody drug conjugates. Chem. Biol. 20 (2), 161– 167, DOI: 10.1016/j.chembiol.2013.01.010Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXjtFWht74%253D&md5=0b419848b5fd4dfa4863a29de0555cf4Location Matters: Site of Conjugation Modulates Stability and Pharmacokinetics of Antibody Drug ConjugatesStrop, Pavel; Liu, Shu-Hui; Dorywalska, Magdalena; Delaria, Kathy; Dushin, Russell G.; Tran, Thomas-Toan; Ho, Wei-Hsien; Farias, Santiago; Casas, Meritxell Galindo; Abdiche, Yasmina; Zhou, Dahui; Chandrasekaran, Ramalakshmi; Samain, Caroline; Loo, Carole; Rossi, Andrea; Rickert, Mathias; Krimm, Stellanie; Wong, Teresa; Chin, Sherman Michael; Yu, Jessica; Dilley, Jeanette; Chaparro-Riggers, Javier; Filzen, Gary F.; O'Donnell, Christopher J.; Wang, Fang; Myers, Jeremy S.; Pons, Jaume; Shelton, David L.; Rajpal, ArvindChemistry & Biology (Oxford, United Kingdom) (2013), 20 (2), 161-167CODEN: CBOLE2; ISSN:1074-5521. (Elsevier Ltd.)Antibody drug conjugates (ADCs) are a therapeutic class offering promise for cancer therapy. The attachment of cytotoxic drugs to antibodies can result in an effective therapy with better safety potential than nontargeted cytotoxics. To understand the role of conjugation site, we developed an enzymic method for site-specific antibody drug conjugation using microbial transglutaminase. This allowed us to attach diverse compds. at multiple positions and investigate how the site influences stability, toxicity, and efficacy. We show that the conjugation site has significant impact on ADC stability and pharmacokinetics in a species-dependent manner. These differences can be directly attributed to the position of the linkage rather than the chem. instability, as was obsd. with a maleimide linkage. With this method, it is possible to produce homogeneous ADCs and tune their properties to maximize the therapeutic window.
- 34Lyon, R. P., Bovee, T. D., Doronina, S. O., Burke, P. J., Hunter, J. H., Neff-LaFord, H. D., Jonas, M., Anderson, M. E., Setter, J. R., and Senter, P. D. (2015) Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic index. Nat. Biotechnol. 33 (7), 733– 735, DOI: 10.1038/nbt.3212Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFaitrfF&md5=cccafa4e97ac463525fd2c898bb62f27Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic indexLyon, Robert P.; Bovee, Tim D.; Doronina, Svetlana O.; Burke, Patrick J.; Hunter, Joshua H.; Neff-LaFord, Haley D.; Jonas, Mechthild; Anderson, Martha E.; Setter, Jocelyn R.; Senter, Peter D.Nature Biotechnology (2015), 33 (7), 733-735CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)The in vitro potency of antibody-drug conjugates (ADCs) increases with the drug-to-antibody ratio (DAR); however, ADC plasma clearance also increases with DAR, reducing exposure and in vivo efficacy. Here we show that accelerated clearance arises from ADC hydrophobicity, which can be modulated through drug-linker design. We exemplify this using hydrophilic auristatin drug linkers and PEGylated ADCs that yield uniform, high-DAR ADCs with superior in vivo performance.
- 35Tumey, L. N., Li, F., Rago, B., Han, X., Loganzo, F., Musto, S., Graziani, E. I., Puthenveetil, S., Casavant, J., Marquette, K. (2017) Site Selection: a Case Study in the Identification of Optimal Cysteine Engineered Antibody Drug Conjugates. AAPS J. 19, 1– 13, DOI: 10.1208/s12248-017-0083-7Google ScholarThere is no corresponding record for this reference.
- 36Burke, P. J., Hamilton, J. Z., Jeffrey, S. C., Hunter, J. H., Doronina, S. O., Okeley, N. M., Miyamoto, J. B., Anderson, M. E., Stone, I. J., Ulrich, M. L. (2017) Optimization of a PEGylated Glucuronide-Monomethylauristatin E Linker for Antibody-Drug Conjugates. Mol. Cancer Ther. 16 (1), 116– 123, DOI: 10.1158/1535-7163.MCT-16-0343Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjsF2hug%253D%253D&md5=94f2bf9dba796252569277f466e9be33Optimization of a PEGylated Glucuronide-Monomethylauristatin E Linker for Antibody-Drug ConjugatesBurke, Patrick J.; Hamilton, Joseph Z.; Jeffrey, Scott C.; Hunter, Joshua H.; Doronina, Svetlana O.; Okeley, Nicole M.; Miyamoto, Jamie B.; Anderson, Martha E.; Stone, Ivan J.; Ulrich, Michelle L.; Simmons, Jessica K.; McKinney, Erica E.; Senter, Peter D.; Lyon, Robert P.Molecular Cancer Therapeutics (2017), 16 (1), 116-123CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)The emergence of antibody-drug conjugates (ADC), such as brentuximab vedotin and ado-trastuzumab emtansine, has led to increased efforts to identify new payloads and develop improved drug-linker technologies. Most antibody payloads impart significant hydrophobicity to the ADC, resulting in accelerated plasma clearance and suboptimal in vivo activity, particularly for conjugates with high drug-to-antibody ratios (DAR). We recently reported on the incorporation of a discrete PEG24 polymer as a side chain in a β-glucuronidase-cleavable monomethylauristatin E (MMAE) linker to provide homogeneous DAR 8 conjugates with decreased plasma clearance and increased antitumor activity in xenograft models relative to a non-PEGylated control. In this work, we optimized the drug-linker by minimizing the size of the PEG side chain and incorporating a self-stabilizing maleimide to prevent payload de-conjugation in vivo. Multiple PEG-glucuronide-MMAE linkers were prepd. with PEG size up to 24 ethylene oxide units, and homogeneous DAR 8 ADCs were evaluated. A clear relationship was obsd. between PEG length and conjugate pharmacol. when tested in vivo. Longer PEG chains resulted in slower clearance, with a threshold length of PEG8 beyond which clearance was not impacted. Conjugates bearing PEG of sufficient length to minimize plasma clearance provided a wider therapeutic window relative to faster clearing conjugates bearing shorter PEGs. A lead PEGylated glucuronide-MMAE linker was identified incorporating a self-stabilizing maleimide and a PEG12 side chain emerged from these efforts, enabling highly potent, homogeneous DAR 8 conjugates and is under consideration for future ADC programs.
- 37Lyon, R. P., Meyer, D. L., Setter, J. R., and Senter, P. D. (2012) Conjugation of anticancer drugs through endogenous monoclonal antibody cysteine residues. Methods Enzymol. 502, 123– 138, DOI: 10.1016/B978-0-12-416039-2.00006-9Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xkt1Cjtbw%253D&md5=32b9da481a24d156f347a2cd9faf3a20Conjugation of anticancer drugs through endogenous monoclonal antibody cysteine residuesLyon, Robert P.; Meyer, David L.; Setter, Jocelyn R.; Senter, Peter D.Methods in Enzymology (2012), 502 (Protein Engineering for Therapeutics, Part A), 123-138CODEN: MENZAU; ISSN:0076-6879. (Elsevier Inc.)A review. Many methods have been described for the conjugation of drugs to monoclonal antibodies. The presence of a discrete no. of readily reducible disulfides in the common IgG subtypes presents a convenient opportunity for conjugation to cysteine residues with thiol-reactive drug-linkers. Such conjugates can be prepd. by a straightforward two-step reaction scheme involving the redn. of the antibody disulfides to the desired no. of av. thiols per antibody, followed by addn. of the drug-linker, ideally with a maleimido functionality for rapid, selective reaction. In a discovery setting, this basic method can be scaled down to produce microgram quantities of conjugate for early screening, and in a manufg. setting can be scaled up to produce grams or kilograms of conjugate for clin. trials and commercialization. The resulting conjugates are readily characterized using common HPLC methods.
- 38Bhakta, S., Raab, H., and Junutula, J. R. (2013) Engineering THIOMABs for site-specific conjugation of thiol-reactive linkers. Methods Mol. Biol. 1045, 189– 203, DOI: 10.1007/978-1-62703-541-5_11Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3sfms1Orug%253D%253D&md5=83bb25f39770bd0156d24ac0e97fe4caEngineering THIOMABs for site-specific conjugation of thiol-reactive linkersBhakta Sunil; Raab Helga; Junutula Jagath RMethods in molecular biology (Clifton, N.J.) (2013), 1045 (), 189-203 ISSN:.Antibody conjugates are used in many therapeutic and research applications and are generated by chemically linking a cysteine or lysine residue to potent chemotherapeutic drugs or other functional groups through a flexible linker. Recently, we have engineered THIOMABs (antibodies with engineered reactive cysteine residues) for site-specific conjugation and showed that these antibody conjugates display homogeneous labeling with optimal in vitro and in vivo characteristics. Here, we describe protocols for engineering, selection, and site-specific conjugation of THIOMABs with thiol-reactive linkers.
- 39Garcia-Echeverria, C. (2014) Developing second generation antibody-drug conjugates: the quest for new technologies. J. Med. Chem. 57 (19), 7888– 7889, DOI: 10.1021/jm501298kGoogle Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsFCjtr7N&md5=681e6c579a64daca1509fb950a1a386dDeveloping Second Generation Antibody-Drug Conjugates: The Quest for New TechnologiesGarcia-Echeverria, CarlosJournal of Medicinal Chemistry (2014), 57 (19), 7888-7889CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. The field of antibody-drug conjugates (ADCs) has gained significant momentum after the recent regulatory approval of two ADCs, and significant research efforts are directed to identify more effective payloads and simplify current manufg. challenges.
- 40Drake, P. M., Albers, A. E., Baker, J., Banas, S., Barfield, R. M., Bhat, A. S., de Hart, G. W., Garofalo, A. W., Holder, P., Jones, L. C. (2014) Aldehyde Tag Coupled with HIPS Chemistry Enables the Production of ADCs Conjugated Site-Specifically to Different Antibody Regions with Distinct in Vivo Efficacy and PK Outcomes. Bioconjugate Chem. 25 (7), 1331– 1341, DOI: 10.1021/bc500189zGoogle Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXps1Knurs%253D&md5=fb08b4277ea6d941861f1cca293b1385Aldehyde Tag Coupled with HIPS Chemistry Enables the Production of ADCs Conjugated Site-Specifically to Different Antibody Regions with Distinct in Vivo Efficacy and PK OutcomesDrake, Penelope M.; Albers, Aaron E.; Baker, Jeanne; Banas, Stefanie; Barfield, Robyn M.; Bhat, Abhijit S.; de Hart, Gregory W.; Garofalo, Albert W.; Holder, Patrick; Jones, Lesley C.; Kudirka, Romas; McFarland, Jesse; Zmolek, Wes; Rabuka, DavidBioconjugate Chemistry (2014), 25 (7), 1331-1341CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)It is becoming increasingly clear that site-specific conjugation offers significant advantages over conventional conjugation chemistries used to make antibody-drug conjugates (ADCs). Site-specific payload placement allows for control over both the drug-to-antibody ratio (DAR) and the conjugation site, both of which play an important role in governing the pharmacokinetics (PK), disposition, and efficacy of the ADC. In addn. to the DAR and site of conjugation, linker compn. also plays an important role in the properties of an ADC. The authors have previously reported a novel site-specific conjugation platform comprising linker payloads designed to selectively react with site-specifically engineered aldehyde tags on an antibody backbone. This chem. results in a stable C-C bond between the antibody and the cytotoxin payload, providing a uniquely stable connection with respect to the other linker chemistries used to generate ADCs. The flexibility and versatility of the aldehyde tag conjugation platform has enabled the authors to undertake a systematic evaluation of the impact of conjugation site and linker compn. on ADC properties. Here, the authors describe the prodn. and characterization of a panel of ADCs bearing the aldehyde tag at different locations on an IgG1 backbone conjugated using Hydrazino-iso-Pictet-Spengler (HIPS) chem. The authors demonstrate that in a panel of ADCs with aldehyde tags at different locations, the site of conjugation has a dramatic impact on in vivo efficacy and pharmacokinetic behavior in rodents; this advantage translates to an improved safety profile in rats as compared to a conventional lysine conjugate.
- 41Catcott, K. C., McShea, M. A., Bialucha, C. U., Miller, K. L., Hicks, S. W., Saxena, P., Gesner, T. G., Woldegiorgis, M., Lewis, M. E., Bai, C. (2016) Microscale screening of antibody libraries as maytansinoid antibody-drug conjugates. MAbs 8 (3), 513– 523, DOI: 10.1080/19420862.2015.1134408Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XislSmtLs%253D&md5=f353875078f05b2b0e43ca26a6d61908Microscale screening of antibody libraries as maytansinoid antibody-drug conjugatesCatcott, Kalli C.; McShea, Molly A.; Bialucha, Carl Uli; Miller, Kathy L.; Hicks, Stuart W.; Saxena, Parmita; Gesner, Thomas G.; Woldegiorgis, Mikias; Lewis, Megan E.; Bai, Chen; Fleming, Michael S.; Ettenberg, Seth A.; Erickson, Hans K.; Yoder, Nicholas C.mAbs (2016), 8 (3), 513-523CODEN: MABSCP; ISSN:1942-0870. (Taylor & Francis, Inc.)Antibody-drug conjugates (ADCs) are of great interest as targeted cancer therapeutics. Prepn. of ADCs for early stage screening is constrained by purifn. and biochem. anal. techniques that necessitate burdensome quantities of antibody. Here we describe a method, developed for the maytansinoid class of ADCs, enabling parallel conjugation of antibodies in 96-well format. The method utilizes ∼100 μg of antibody per well and requires <5 μg of ADC for characterization. We demonstrate the capabilities of this system using model antibodies. We also provide multiple examples applying this method to early-stage screening of maytansinoid ADCs. The method can greatly increase the throughput with which candidate ADCs can be screened in cell-based assays, and may be more generally applicable to high-throughput prepn. and screening of different types of protein conjugates.
- 42Cilliers, C., Guo, H., Liao, J., Christodolu, N., and Thurber, G. M. (2016) Multiscale Modeling of Antibody-Drug Conjugates: Connecting Tissue and Cellular Distribution to Whole Animal Pharmacokinetics and Potential Implications for Efficacy. AAPS J. 18 (5), 1117– 1130, DOI: 10.1208/s12248-016-9940-zGoogle Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XpvV2nsbY%253D&md5=f7df584c11f8a3126ee369933a2f237bMultiscale Modeling of Antibody-Drug Conjugates: Connecting Tissue and Cellular Distribution to Whole Animal Pharmacokinetics and Potential Implications for EfficacyCilliers, Cornelius; Guo, Hans; Liao, Jianshan; Christodolu, Nikolas; Thurber, Greg M.AAPS Journal (2016), 18 (5), 1117-1130CODEN: AJAOB6; ISSN:1550-7416. (Springer)Antibody-drug conjugates exhibit complex pharmacokinetics due to their combination of macromol. and small mol. properties. These issues range from systemic concerns, such as deconjugation of the small mol. drug during the long antibody circulation time or rapid clearance from nonspecific interactions, to local tumor tissue heterogeneity, cell bystander effects, and endosomal escape. Math. models can be used to study the impact of these processes on overall distribution in an efficient manner, and several types of models have been used to analyze varying aspects of antibody distribution including physiol. based pharmacokinetic (PBPK) models and tissue-level simulations. However, these processes are quant. in nature and cannot be handled qual. in isolation. For example, free antibody from deconjugation of the small mol. will impact the distribution of conjugated antibodies within the tumor. To incorporate these effects into a unified framework, we have coupled the systemic and organ-level distribution of a PBPK model with the tissue-level detail of a distributed parameter tumor model. We used this math. model to analyze new exptl. results on the distribution of the clin. antibody-drug conjugate Kadcyla in HER2-pos. mouse xenografts. This model is able to capture the impact of the drug-antibody ratio (DAR) on tumor penetration, the net result of drug deconjugation, and the effect of using unconjugated antibody to drive ADC penetration deeper into the tumor tissue. This modeling approach will provide quant. and mechanistic support to exptl. studies trying to parse the impact of multiple mechanisms of action for these complex drugs.
- 43Bumbaca, D., Boswell, C. A., Fielder, P. J., and Khawli, L. A. (2012) Physiochemical and biochemical factors influencing the pharmacokinetics of antibody therapeutics. AAPS J. 14 (3), 554– 558, DOI: 10.1208/s12248-012-9369-yGoogle Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XptFWit70%253D&md5=acaabca0a83de3a589e334024a5d6f00Physiochemical and Biochemical Factors Influencing the Pharmacokinetics of Antibody TherapeuticsBumbaca, Daniela; Boswell, C. Andrew; Fielder, Paul J.; Khawli, Leslie A.AAPS Journal (2012), 14 (3), 554-558CODEN: AJAOB6; ISSN:1550-7416. (Springer)A review. Monoclonal antibodies are increasingly being developed to treat multiple disease areas, including those related to oncol., immunol., neurol., and ophthalmol. There are multiple factors, such as charge, size, neonatal Fc receptor (FcRn) binding affinity, target affinity and biol., IgG (IgG) subclass, degree and type of glycosylation, injection route, and injection site, that could affect the pharmacokinetics (PK) of these large macromol. therapeutics, which in turn could have ramifications on their efficacy and safety. This mini-review examines how characteristics of the antibodies could be altered to change their PK profiles. For example, it was obsd. that a net charge modification of at least a 1-unit shift in isoelec. point altered antibody clearance. Antibodies with enhanced affinity for FcRn at pH 6.0 display longer serum half-lives and slower clearances than wild type. Antibody fragments have different clearance rates and tissue distribution profiles than full length antibodies. Fc glycosylation is perceived to have a minimal effect on PK while that of terminal high mannose remains unclear. More investigation is warranted to det. if injection route and/or site impacts PK. Nonetheless, a better understanding of the effects of all these variations may allow for the better design of antibody therapeutics.
- 44Bohnert, T. and Gan, L. S. (2013) Plasma protein binding: from discovery to development. J. Pharm. Sci. 102 (9), 2953– 2994, DOI: 10.1002/jps.23614Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXpvVymt7c%253D&md5=d676f7490c6e0f09c8acf7cce5183b17Plasma protein binding: From discovery to developmentBohnert, Tonika; Gan, Liang-ShangJournal of Pharmaceutical Sciences (2013), 102 (9), 2953-2994CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)A review. The importance of plasma protein binding (PPB) in modulating the effective drug concn. at pharmacol. target sites has been the topic of significant discussion and debate amongst drug development groups over the past few decades. Free drug theory, which states that in absence of energy-dependent processes, after steady state equil. has been attained, free drug concn. in plasma is equal to free drug concn. at the pharmacol. target receptor(s) in tissues, has been used to explain pharmacokinetics/pharmacodynamics relationships in a large no. of cases. Any sudden increase in free concn. of a drug could potentially cause toxicity and may need dose adjustment. Free drug concn. is also helpful to est. the effective concn. of drugs that potentially can ppt. metab. (or transporter)-related drug-drug interactions. Disease models are extensively validated in animals to progress a compd. into development. Unbound drug concn., and therefore PPB information across species is very informative in establishing safety margins and guiding selection of First in Human (FIH) dose and human efficacious dose. The scope of this review is to give an overview of reported role of PPB in several therapeutic areas, highlight cases where PPB changes are clin. relevant, and provide drug metab. and pharmacokinetics recommendations in discovery and development settings. © 2013 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 45Alley, S. C., Benjamin, D. R., Jeffrey, S. C., Okeley, N. M., Meyer, D. L., Sanderson, R. J., and Senter, P. D. (2008) Contribution of linker stability to the activities of anticancer immunoconjugates. Bioconjugate Chem. 19 (3), 759– 765, DOI: 10.1021/bc7004329Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXislemurk%253D&md5=62abdec871dda99c160a9b989c12f6f0Contribution of Linker Stability to the Activities of Anticancer ImmunoconjugatesAlley, Stephen C.; Benjamin, Dennis R.; Jeffrey, Scott C.; Okeley, Nicole M.; Meyer, Damon L.; Sanderson, Russell J.; Senter, Peter D.Bioconjugate Chemistry (2008), 19 (3), 759-765CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The linker component of antibody-drug conjugates (ADC) is a key feature in developing optimized therapeutic agents that are highly active at well tolerated doses. For maximal intratumoral drug delivery, linkers are required that are highly stable in the systemic circulation, yet allow for efficient drug release at the target site. In this respect, amide bond-based technologies constitute a technol. advancement, since the linker half-lives in circulation (t1/2 ∼ 7 days) are much longer than earlier generation linkers that break down within 1-2 days. The amide linkers, some of which contain peptides, are appended to the mAb carriers through thioether/maleimide adducts. Here, we describe that use of a bromoacetamidecaproyl (bac) in place of the maleimidocaproyl (mc) increases the plasma stability of resulting thioether ADCs. One such ADC, 1F6-C4v2-bac-MMAF, which is directed against the CD70 antigen on lymphomas and renal cell carcinoma, was prepd. contg. a bac thioether spacer between the drug (MMAF) and the mAb carrier (1F6-C4v2). There was no measurable systemic drug release from this ADC for 2 wk postadministration in mice. In order to assess the impact of improving linker stability beyond mc contg. ADCs, a series of mc and bac-linked 1F6-MMAF conjugates were compared for tolerability, intratumoral drug delivery, and therapeutic efficacy in nude mice with renal cell carcinoma xenografts. There were no statistically significant efficacy differences between sets of mc and bac contg. ADCs, although the bac linker technol. led to 25% higher intratumoral drug exposure over a 7 day period compared to the corresponding mc linker. The mechanism of drug release from maleimide-adducts likely involves a retro-Michael reaction that takes place in plasma, based on in vitro studies demonstrating that some of the released drug-maleimide deriv. became covalently bound to cysteine-34 of serum albumin. In summary, the data indicate that new linkers can be obtained with improved in vivo stability by replacing the maleimide with an acetamide, but the resulting ADCs had similar tolerability and activity profiles.
- 46Ohri, R., Bhakta, S., Fourie-O’Donohue, A., Dela Cruz-Chuh, J., Tsai, S. P., Cook, R., Wei, B., Ng, C., Wong, A. W., Bos, A. B. (2018) High-Throughput Cysteine Scanning To Identify Stable Antibody Conjugation Sites for Maleimide- and Disulfide-Based Linkers. Bioconjugate Chem. 29 (2), 473– 485, DOI: 10.1021/acs.bioconjchem.7b00791Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisVKku7g%253D&md5=318146c1baea8094b671e99b5c429627High-Throughput Cysteine Scanning To Identify Stable Antibody Conjugation Sites for Maleimide- and Disulfide-Based LinkersOhri, Rachana; Bhakta, Sunil; Fourie-O'Donohue, Aimee; dela Cruz-Chuh, Josefa; Tsai, Siao Ping; Cook, Ryan; Wei, Binqing; Ng, Carl; Wong, Athena W.; Bos, Aaron B.; Farahi, Farzam; Bhakta, Jiten; Pillow, Thomas H.; Raab, Helga; Vandlen, Richard; Polakis, Paul; Liu, Yichin; Erickson, Hans; Junutula, Jagath R.; Kozak, Katherine R.Bioconjugate Chemistry (2018), 29 (2), 473-485CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)THIOMAB antibody technol. utilizes cysteine residues engineered onto an antibody to allow for site-specific conjugation. The technol. has enabled the exploration of different attachment sites on the antibody in combination with small mols., peptides, or proteins to yield antibody conjugates with unique properties. As reported previously, the specific location of the site of conjugation on an antibody can impact the stability of the linkage to the engineered cysteine for both thio-succinimide and disulfide bonds. High stability of the linkage is usually desired to maximize the delivery of the cargo to the intended target. In the current study, cysteines were individually substituted into every position of the anti-HER2 antibody (trastuzumab), and the stabilities of drug conjugations at those sites were evaluated. We screened a total of 648 THIOMAB antibody-drug conjugates, each generated from a trastuzamab prepd. by sequentially mutating non-cysteine amino acids in the light and heavy chains to cysteine. Each THIOMAB antibody variant was conjugated to either maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl-monomethyl auristatin E (MC-vc-PAB-MMAE) or pyridyl disulfide monomethyl auristatin E (PDS-MMAE) using a high-throughput, on-bead conjugation and purifn. method. Greater than 50% of the THIOMAB antibody variants were successfully conjugated to both MMAE derivs. with a drug to antibody ratio (DAR) of >0.5 and <50% aggregation. The relative in vitro plasma stabilities for approx. 750 conjugates were assessed using enzyme-linked immunosorbent assays, and stable sites were confirmed with affinity-capture LC/MS-based detection methods. Highly stable conjugation sites for the two types of MMAE derivs. were identified on both the heavy and light chains. Although the stabilities of maleimide conjugates were shown to be greater than those of the disulfide conjugates, many sites were identified that were stable for both. Furthermore, in vitro stabilities of selected stable sites translated across different cytotoxic payloads and different target antibodies as well as to in vivo stability.
- 47Lyon, R. (2018) Drawing lessons from the clinical development of antibody-drug conjugates. Drug Discovery Today: Technol. 30, 105– 109, DOI: 10.1016/j.ddtec.2018.10.001Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cnisVSlsA%253D%253D&md5=dfdb2af91ce423b8912dc33f9187b35fDrawing lessons from the clinical development of antibody-drug conjugatesLyon RobertDrug discovery today. Technologies (2018), 30 (), 105-109 ISSN:.The antibody-drug conjugate (ADC) field has seen a remarkable expansion in the number of entrants in clinical studies. Many of these agents employ newer conjugation technologies that have been developed over the last decade that confer various attributes to the ADCs prepared with them, including stability, potency, and homogeneity. In many cases, these new ADCs appear demonstrably superior to earlier technologies in preclinical models of activity and toxicology, but the degree to which these improvements will translate to the clinic is only starting to be seen. Many of these technologies are now competing head-to-head by targeting the same antigen in similar patient populations, allowing for a direct comparison of their clinical performance properties. As lessons from these experiences feed back into discovery research, future iterations of ADC design may be expected to bring improved therapeutics into the clinic.
- 48Khera, E., Cilliers, C. M., Bhatnagar, S., and Thurber, G. M. (2018) Computation transport analysis of antibody-drug conjugate bystander effects and payload tumoral distribution: implications for therapy. Mol. Syst. Des. Eng. 3, 73– 88, DOI: 10.1039/C7ME00093FGoogle Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1yjs73J&md5=3b39670c741923617c57a9ec3e715806Computational transport analysis of antibody-drug conjugate bystander effects and payload tumoral distribution: implications for therapyKhera, Eshita; Cilliers, Cornelius; Bhatnagar, Sumit; Thurber, Greg M.Molecular Systems Design & Engineering (2018), 3 (1), 73-88CODEN: MSDEBG; ISSN:2058-9689. (Royal Society of Chemistry)Antibody drug conjugates (ADCs) have a proven clin. record with four FDA approved drugs and dozens more in clin. trials. However, a better understanding of the relationship between delivery and efficacy of ADCs is needed to improve the rate of successful clin. development. Recent evidence indicates that heterogeneous distribution can play a large role in the efficacy of these drugs. However, the impact of the drug payload, particularly the ability of the payload to diffuse outside of the original targeted cell into adjacent cells (the bystander effect), is not completely understood. Given the challenges in directly measuring the payload distribution within tumors, we developed a predictive computational model to study payload distribution as a function of antibody dose, payload dose, and payload properties. The computational results indicate that: 1. the heterogeneous tumoral distribution of ADCs impacts efficacy, and increasing the antibody dose improves penetration and efficacy. 2. The increased penetration of payloads with bystander effects can partially compensate for poor antibody penetration, but larger antibody doses still result in further improvement. This occurs because of the higher efficiency of direct cell killing than bystander killing. 3. Bystander effects are important for killing antigen neg. cells, and an optimum in physicochem. properties exists. Payloads with a balance in cellular uptake vs. tissue diffusion enter cells fast enough to avoid tumor washout but slow enough to reach distant cells. Therefore, optimizing the antibody dose, payload dose, and payload physicochem. properties results in ideal delivery to the site of action and max. efficacy.
- 49Cilliers, C., Menezes, B., Nessler, I., Linderman, J., and Thurber, G. M. (2018) Improved Tumor Penetration and Single-Cell Targeting of Antibody-Drug Conjugates Increases Anticancer Efficacy and Host Survival. Cancer Res. 78 (3), 758– 768, DOI: 10.1158/0008-5472.CAN-17-1638Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitVWntr8%253D&md5=9c10da29fd94ccad470329f2dcc48566Improved Tumor Penetration and Single-Cell Targeting of Antibody-Drug Conjugates Increases Anticancer Efficacy and Host SurvivalCilliers, Cornelius; Menezes, Bruna; Nessler, Ian; Linderman, Jennifer; Thurber, Greg M.Cancer Research (2018), 78 (3), 758-768CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Current ADC have made advances in engineering the antibody, linker, conjugation site, small-mol. payload, and drug-to-antibody ratio (DAR). However, the relationship between heterogeneous intratumoral distribution and efficacy of ADCs is poorly understood. Here, we compared trastuzumab and ado-trastuzumab emtansine (T-DM1) to study the impact of ADC tumor distribution on efficacy. In a mouse xenograft model insensitive to trastuzumab, coadministration of trastuzumab with a fixed dose of T-DM1 at 3:1 and 8:1 ratios dramatically improved ADC tumor penetration and resulted in twice the improvement in median survival compared with T-DM1 alone. In this setting, the effective DAR was lowered, decreasing the amt. of payload delivered to each targeted cell but increasing the no. of cells that received payload. This result is counterintuitive because trastuzumab acts as an antagonist in vitro and has no single-agent efficacy in vivo, yet improves the effectiveness of T-DM1 in vivo. Novel dual-channel fluorescence ratios quantified single-cell ADC uptake and metab. and confirmed that the in vivo cellular dose of T-DM1 alone exceeded the min. required for efficacy in this model. In addn., this technique characterized cellular pharmacokinetics with heterogeneous delivery after 1 day, degrdn. and payload release by 2 days, and in vitro cell killing and in vivo tumor shrinkage 2 to 3 days later. This work demonstrates that the intratumoral distribution of ADC, independent of payload dose or plasma clearance, plays a major role in ADC efficacy. This study shows how lowering the drug-to-antibody ratio during treatment can improve the intratumoral distribution of a antibody-drug conjugate, with implications for improving the efficacy of this class of cancer drugs.
- 50Sahota, S. and Vahdat, L. T. (2017) Sacituzumab govitecan: an antibody-drug conjugate. Expert Opin. Biol. Ther. 17 (8), 1027– 1031, DOI: 10.1080/14712598.2017.1331214Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXosVags7s%253D&md5=e2a9aeb979d52eca095bd455aa73667bSacituzumab govitecan: an antibody-drug conjugateSahota, Sheena; Vahdat, Linda T.Expert Opinion on Biological Therapy (2017), 17 (8), 1027-1031CODEN: EOBTA2; ISSN:1471-2598. (Taylor & Francis Ltd.): Despite advances in the diagnosis and treatment of patients with cancer, patients with metastatic cancer have limited therapeutic options after initial lines of therapy. Understanding tumor biol. has translated into the identification of actionable targets that resulted in therapeutics. Antibody-drug conjugates (ADC) are capitalizing on this explosion of scientific information. ADCs allow an antibody to a unique target to be conjugated via an innovative linker, to a highly toxic drug which is delivered to its target. Sacituzumab govitecan is an ADC that combines the active mol. in irinotecan, SN-38, to an antibody targeting trop2.: In this review, the authors introduce the reader to the ADC sacituzumab govitecan providing the reader with details about its pharmacokinetics, pharmacodynamics, efficacy and safety. The authors also give their expert anal. about its potential future use.: Sacituzumab govitecan is a novel and well-tolerated therapeutic showing promising results in difficult to treat cancers. Further studies are underway to optimize the group of patients that would benefit from it. Given its excellent performance, we are cautiously optimistic it will be approved by the FDA.
- 51Nakada, T., Sugihara, K., Jikoh, T., Abe, Y., and Agatsuma, T. (2019) The Latest Research and Development into the Antibody-Drug Conjugate, [fam-] Trastuzumab Deruxtecan (DS-8201a), for HER2 Cancer Therapy. Chem. Pharm. Bull. 67 (3), 173– 185, DOI: 10.1248/cpb.c18-00744Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhsVejurjJ&md5=ab7628fdc4af3c8e1d73e00553c9534eThe latest research and development into the antibody-drug conjugate, [fam-] trastuzumab deruxtecan (DS-8201a), for HER2 cancer therapyNakada, Takashi; Sugihara, Kiyoshi; Jikoh, Takahiro; Abe, Yuki; Agatsuma, ToshinoriChemical & Pharmaceutical Bulletin (2019), 67 (3), 173-185CODEN: CPBTAL; ISSN:0009-2363. (Pharmaceutical Society of Japan)A review. A major limitation of traditional chemotherapy for cancer is dose-limiting toxicity, caused by the exposure of non-tumor cells to cytotoxic agents. Use of mol. targeted drugs, such as specific kinase inhibitors and monoclonal antibodies, is a possible soln. to overcome this limitation and has achieved clin. success so far. Use of an antibody-drug conjugate (ADC) is a rational strategy for improving efficacy and reducing systemic adverse events. ADCs use antibodies selectively to deliver a potent cytotoxic agent to tumor cells, thus drastically improving the therapeutic index of chemotherapeutic agents. Lessons learned from clin. failure of early ADCs during the 1980s to 90s have recently led to improvements in ADC technol., and resulted in the approval of four novel ADCs. Nonetheless, further advances in ADC technol. are still required to streamline their clin. efficacy and reduce toxicity. [fam-] Trastuzumab deruxtecan (DS-8201a) is a next-generation ADC that satisfies these requirements based on currently available evidence. DS-8201a has several innovative features; a highly potent novel payload with a high drug-to-antibody ratio, good homogeneity, a tumor-selective cleavable linker, stable linker-payload in circulation, and a short systemic half-life cytotoxic agent in vivo; the released cytotoxic payload could exert a bystander effect. With respect to its preclin. profiles, DS-8201a could provide a valuable therapy with a great potential against HER2-expressing cancers in clin. settings. In a phase I trial, DS-8201a showed acceptable safety profiles with potential therapeutic efficacy, with the wide therapeutic index.
- 52Reid, E. E., Archer, K. E., Shizuka, M., Wilhelm, A., Yoder, N. C., Bai, C., Fishkin, N. E., Harris, L., Maloney, E. K., E, H. (2019) Effect of linker stereochemistry on the antitumor activity of anti-body-drug conjugates (ADCs) containing indolinobenzodiazepine payloads. ACS Med. Chem. Lett. 10 (8), 1193– 1197, DOI: 10.1021/acsmedchemlett.9b00240Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlGjs7fP&md5=72540be91ff42af39669c697c9b0f162Effect of Linker Stereochemistry on the Activity of Indolinobenzodiazepine Containing Antibody-Drug Conjugates (ADCs)Reid, Emily E.; Archer, Katie E.; Shizuka, Manami; Wilhelm, Alan; Yoder, Nicholas C.; Bai, Chen; Fishkin, Nathan E.; Harris, Luke; Maloney, Erin K.; Salomon, Paulin; Hong, Erica; Wu, Rui; Ab, Olga; Jin, Shan; Lai, Katharine C.; Sikka, Surina; Chari, Ravi V. J.; Miller, Michael L.ACS Medicinal Chemistry Letters (2019), 10 (8), 1193-1197CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Antibody-drug conjugates (ADCs) that incorporate potent indolinobenzodiazepine DNA alkylators as the payload component are currently undergoing clin. evaluation. In one ADC design, the payload mols. are linked to the antibody through a peptidase-labile L-Ala-L-Ala linker. In order to det. the role of amino acid stereochem. on antitumor activity and tolerability, we incorporated L- and D-alanyl groups in the dipeptide, synthesized all four diastereomers, and prepd. and tested the corresponding ADCs. Results of our preclin. evaluation showed that the L-Ala-L-Ala configuration provided the ADC with the highest therapeutic index (antitumor activity vs toxicity).
- 53Lazar, A. C., Wang, L., Blättler, W. A., Amphlett, G., Lambert, J. M., and Zhang, W. (2005) Analysis of the composition of immunoconjugates using size-exclusion chromatography coupled to mass spectrometry. Rapid Commun. Mass Spectrom. 19 (13), 1806– 1814, DOI: 10.1002/rcm.1987Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXmtVegt70%253D&md5=678eee93e086e099fa72f4d2d79e6646Analysis of the composition of immunoconjugates using size-exclusion chromatography coupled to mass spectrometryLazar, Alexandru C.; Wang, Lintao; Blaettler, Walter A.; Amphlett, Godfrey; Lambert, John M.; Zhang, WeiRapid Communications in Mass Spectrometry (2005), 19 (13), 1806-1814CODEN: RCMSEF; ISSN:0951-4198. (John Wiley & Sons Ltd.)Recombinant monoclonal antibody drug products play an increasingly important role in the treatment of various diseases. Antibodies are large, multi-chain proteins and antibody prepns. often contain several mol. variants, which renders them heterogeneous. The heterogeneity is further increased in immunoconjugates prepd. by covalently linking several drug mols. per antibody mol. As part of the product characterization, the mol. wts. of the antibodies or their drug conjugates need to be measured. Electrospray ionization mass spectrometry (ESI-MS) is well suited for the anal. of recombinant antibodies and immunoconjugates. Sample prepn. is an important element of ESI-MS anal., in particular samples need to be freed of interfering charged species, such as salts and buffer components. In this paper, Amicon centrifugal filters, reversed-phase high-performance liq. chromatog. (HPLC), and size-exclusion HPLC were evaluated for sample desalting. Size-exclusion HPLC, using aq. acetonitrile as the mobile phase, directly coupled to ESI-MS provided the best performance and was optimized for the study of immunoconjugates. The results showed that antibodies carrying covalently linked maytansinoid mols. generated charge envelope profiles that differ from those of the non-conjugated antibody. For the detn. of the distribution of the various conjugate species in an immunoconjugate sample prepd. by randomly linking in the av. 3.6 drug mols. per antibody mol., the exptl. conditions needed to be carefully selected to allow acquisition of the whole spectrum contg. the charge envelopes of all species.
- 54Kovtun, Y. V., Audette, C. A., Mayo, M. F., Jones, G. E., Doherty, H., Maloney, E. K., Erickson, H. K., Sun, X., Wilhelm, S., Ab, O. (2010) Antibody-maytansinoid conjugates designed to bypass multidrug resistance. Cancer Res. 70 (6), 2528– 2537, DOI: 10.1158/0008-5472.CAN-09-3546Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjtFygur4%253D&md5=17a2600cc85203b3bace74e31d346daaAntibody-Maytansinoid Conjugates Designed to Bypass Multidrug ResistanceKovtun, Yelena V.; Audette, Charlene A.; Mayo, Michele F.; Jones, Gregory E.; Doherty, Heather; Maloney, Erin K.; Erickson, Hans K.; Sun, Xiuxia; Wilhelm, Sharon; Ab, Olga; Lai, Katharine C.; Widdison, Wayne C.; Kellogg, Brenda; Johnson, Holly; Pinkas, Jan; Lutz, Robert J.; Singh, Rajeeva; Goldmacher, Victor S.; Chari, Ravi V. J.Cancer Research (2010), 70 (6), 2528-2537CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Conjugation of cytotoxic compds. to antibodies that bind to cancer-specific antigens makes these drugs selective in killing cancer cells. However, many of the compds. used in such antibody-drug conjugates (ADC) are substrates for the multidrug transporter MDR1. To evade the MDR1-mediated resistance, we conjugated the highly cytotoxic maytansinoid DM1 to antibodies via the maleimidyl-based hydrophilic linker PEG4Mal. Following uptake into target cells, conjugates made with the PEG4Mal linker were processed to a cytotoxic metabolite that was retained by MDR1-expressing cells better than a metabolite of similar conjugates prepd. with the nonpolar linker N-succinimidyl-4-(maleimidomethyl)cyclohexane-1-carboxylate (SMCC). In accord, PEG4Mal-linked conjugates were more potent in killing MDR1-expressing cells in culture. In addn., PEG4Mal-linked conjugates were markedly more effective in eradicating MDR1-expressing human xenograft tumors than SMCC-linked conjugates while being tolerated similarly, thus showing an improved therapeutic index. This study points the way to the development of ADCs that bypass multidrug resistance. Cancer Res; 70(6); 2528-37.
- 55Ab, O., Whiteman, K. R., Bartle, L. M., Sun, X., Singh, R., Tavares, D., LaBelle, A., Payne, G., Lutz, R. J., Pinkas, J. (2015) IMGN853, a Folate Receptor-α (FRα)-Targeting Antibody-Drug Conjugate, Exhibits Potent Targeted Antitumor Activity against FRα-Expressing Tumors. Mol. Cancer Ther. 14 (7), 1605– 1613, DOI: 10.1158/1535-7163.MCT-14-1095Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFejtLzO&md5=194d678451d25646466be718537d3980IMGN853, a Folate Receptor-α (FRα)-Targeting Antibody-Drug Conjugate, Exhibits Potent Targeted Antitumor Activity against FRα-Expressing TumorsAb, Olga; Whiteman, Kathleen R.; Bartle, Laura M.; Sun, Xiuxia; Singh, Rajeeva; Tavares, Daniel; LaBelle, Alyssa; Payne, Gillian; Lutz, Robert J.; Pinkas, Jan; Goldmacher, Victor S.; Chittenden, Thomas; Lambert, John M.Molecular Cancer Therapeutics (2015), 14 (7), 1605-1613CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)A majority of ovarian and non-small cell lung adenocarcinoma cancers overexpress folate receptor α (FRα). Here, we report the development of an anti-FRα antibody-drug conjugate (ADC), consisting of a FRα-binding antibody attached to a highly potent maytansinoid that induces cell-cycle arrest and cell death by targeting microtubules. From screening a large panel of anti-FRα monoclonal antibodies, we selected the humanized antibody M9346A as the best antibody for targeted delivery of a maytansinoid payload into FRα-pos. cells. We compared M9346A conjugates with various linker/maytansinoid combinations, and found that a conjugate, now denoted as IMGN853, with the N-succinimidyl 4-(2-pyridyldithio)-2-sulfobutanoate (sulfo-SPDB) linker and N2'-deacetyl-N2'-(4-mercapto-4-methyl-1-oxopentyl)-maytansine (DM4) exhibited the most potent antitumor activity in several FRα-expressing xenograft tumor models. The level of expression of FRα on the surface of cells was a major determinant in the sensitivity of tumor cells to the cytotoxic effect of the conjugate. Efficacy studies of IMGN853 in xenografts of ovarian cancer and non-small cell lung cancer cell lines and of a patient tumor-derived xenograft model demonstrated that the ADC was highly active against tumors that expressed FRα at levels similar to those found on a large fraction of ovarian and non-small cell lung cancer patient tumors, as assessed by immunohistochem. IMGN853 displayed cytotoxic activity against FRα-neg. cells situated near FRα-pos. cells (bystander cytotoxic activity), indicating its ability to eradicate tumors with heterogeneous expression of FRα. Together, these findings support the clin. development of IMGN853 as a novel targeted therapy for patients with FRα-expressing tumors. Mol Cancer Ther; 14(7); 1605-13. ©2015 AACR.
Cited By
This article is cited by 21 publications.
- Amissi Sadiki, Shanshan Liu, Shefali R. Vaidya, Eric M. Kercher, Ryan T. Lang, James McIsaac, Bryan Q. Spring, Jared R. Auclair, Zhaohui Sunny Zhou. Site-Specific Conjugation of Native Antibody: Transglutaminase-Mediated Modification of a Conserved Glutamine While Maintaining the Primary Sequence and Core Fc Glycan via Trimming with an Endoglycosidase. Bioconjugate Chemistry 2024, 35
(4)
, 465-471. https://doi.org/10.1021/acs.bioconjchem.4c00013
- Thomas T. C. Yue, Ying Ge, Francesco A. Aprile, Michelle T. Ma, Truc T. Pham, Nicholas J. Long. Site-Specific 68Ga Radiolabeling of Trastuzumab Fab via Methionine for ImmunoPET Imaging. Bioconjugate Chemistry 2023, 34
(10)
, 1802-1810. https://doi.org/10.1021/acs.bioconjchem.3c00344
- Samantha M. Sarrett, Cindy Rodriguez, Grzegorz Rymarczyk, Meena M. Hosny, Outi Keinänen, Samantha Delaney, Sarah Thau, Benjamin A. Krantz, Brian M. Zeglis. Lysine-Directed Site-Selective Bioconjugation for the Creation of Radioimmunoconjugates. Bioconjugate Chemistry 2022, 33
(9)
, 1750-1760. https://doi.org/10.1021/acs.bioconjchem.2c00354
- Shin Hye Ahn, Brett A. Vaughn, Willy A. Solis, Mark L. Lupher, Jr., Trevor J. Hallam, Eszter Boros. Site-Specific 89Zr- and 111In-Radiolabeling and In Vivo Evaluation of Glycan-free Antibodies by Azide–Alkyne Cycloaddition with a Non-natural Amino Acid. Bioconjugate Chemistry 2020, 31
(4)
, 1177-1187. https://doi.org/10.1021/acs.bioconjchem.0c00100
- Qi Ma, Puro Durga, Frederick X.C. Wang, Hang-Ping Yao, Ming-Hai Wang. Pharmaceutical innovation and advanced biotechnology in the biotech-pharmaceutical industry for antibody–drug conjugate development. Drug Discovery Today 2024, 29
(8)
, 104057. https://doi.org/10.1016/j.drudis.2024.104057
- Yi-Ching Lin, Chao-Wei Yang, Shih-Chuan Tsai, Shiou-Shiow Farn, Fang-Yu Ou Yang, Wei-Lin Lo, Liang-Cheng Chen, Kuo-Ting Chen, Mao-Chi Weng, Jui-Yin Kung, Xin-Yu Qiu, Ching-Chun Lu, Feng-Yun J. Huang. Random and site-specific radiolabeling of [89Zr]Zr-DFO-anti-PD-L1-mAb iPET tracer. Journal of Radioanalytical and Nuclear Chemistry 2024, 11 https://doi.org/10.1007/s10967-024-09401-0
- Viktoriia Postupalenko, Léo Marx, Mathilde Pantin, David Viertl, Nadège Gsponer, Gaëlle Giudice, Natalia Gasilova, Margret Schottelius, Frédéric Lévy, Patrick Garrouste, Jean-Manuel Segura, Origène Nyanguile. Site-selective template-directed synthesis of antibody Fc conjugates with concomitant ligand release. Chemical Science 2024, 15
(4)
, 1324-1337. https://doi.org/10.1039/D3SC04324J
- Karoline Bechtold-Peters, Andrea Ruggiero, Nienke Vriezen, Nathan Ihle, Armin Klein, Charles Morgan, Daniel Schweizer, Dengfeng Liu, Fred Jacobson, Jakob Buecheler, Mark Panek, Naomi Duggan, Padma Malyala, Philippe Dupraz, Priyanka Desai, Shufang Niu, Yiqing Feng, Xiangyang Wang. CMC Regulatory Considerations for Antibody-Drug Conjugates. Journal of Pharmaceutical Sciences 2023, 112
(12)
, 2965-2980. https://doi.org/10.1016/j.xphs.2023.09.007
- Ryan J. Pakula, Peter J. H. Scott. Applications of radiolabeled antibodies in neuroscience and neuro‐oncology. Journal of Labelled Compounds and Radiopharmaceuticals 2023, 66
(9)
, 269-285. https://doi.org/10.1002/jlcr.4049
- Cindy Rodriguez, Samantha Delaney, Joni Sebastiano, Samantha M. Sarrett, Mike A. Cornejo, Sarah Thau, Meena M. Hosny, Brian M. Zeglis. Site-selective radiolabeling using mushroom tyrosinase and the strain-promoted oxidation-controlled 1,2-quinone cycloaddition. RSC Advances 2023, 13
(26)
, 17705-17709. https://doi.org/10.1039/D3RA03486K
- Rita Khoury, Khalil Saleh, Nadine Khalife, Mohamad Saleh, Claude Chahine, Rebecca Ibrahim, Axel Lecesne. Mechanisms of Resistance to Antibody-Drug Conjugates. International Journal of Molecular Sciences 2023, 24
(11)
, 9674. https://doi.org/10.3390/ijms24119674
- Arnaud C. Tiberghien, Balakumar Vijayakrishnan, Arman Esfandiari, Mahammad Ahmed, Raul Pardo, John Bingham, Lauren Adams, Kathleen Santos, Gyoung-Dong Kang, Kathryn M. Pugh, Shameen Afif-Rider, Kapil Vashisht, Kemal Haque, Ravinder Tammali, Edward Rosfjord, Adriana Savoca, John A. Hartley, Philip W. Howard. Comparison of Pyrrolobenzodiazepine Dimer Bis-imine versus Mono-imine: DNA Interstrand Cross-linking, Cytotoxicity, Antibody–Drug Conjugate Efficacy and Toxicity. Molecular Cancer Therapeutics 2023, 22
(2)
, 254-263. https://doi.org/10.1158/1535-7163.MCT-21-0693
- Juniper A. Scribner, Stuart W. Hicks, Kerstin W. Sinkevicius, Nicholas C. Yoder, Gundo Diedrich, Jennifer G. Brown, Jacquelynn Lucas, Megan E. Fuller, Thomas Son, Anahita Dastur, Jeff Hooley, Christopher Espelin, Marian Themeles, Francine Z. Chen, Ying Li, Michael Chiechi, Jenny Lee, Bhaswati Barat, Lusiana Widjaja, Sergey Gorlatov, James Tamura, Valentina Ciccarone, Olga Ab, Kerry A. McEachem, Scott Koenig, Eric H. Westin, Paul A. Moore, Thomas Chittenden, Richard J. Gregory, Ezio Bonvini, Deryk Loo. Preclinical Evaluation of IMGC936, a Next-Generation Maytansinoid-based Antibody–drug Conjugate Targeting ADAM9-expressing Tumors. Molecular Cancer Therapeutics 2022, 21
(7)
, 1047-1059. https://doi.org/10.1158/1535-7163.MCT-21-0915
- Ning Zou, Amy Han. Application of Pyrrolobenzodiazepines in Antibody Drug Conjugates. 2022, 293-339. https://doi.org/10.1002/9781119627784.ch13
- Charlotte Sornay, Valentine Vaur, Alain Wagner, Guilhem Chaubet. An overview of chemo- and site-selectivity aspects in the chemical conjugation of proteins. Royal Society Open Science 2022, 9
(1)
https://doi.org/10.1098/rsos.211563
- M. W. Giese, R. H. Woodman, G. T. Hermanson, P. D. Davis. The Use of Uniform PEG Compounds in the Design of ADCs. 2021, 286-376. https://doi.org/10.1039/9781839165153-00286
- Anna Kaempffe, Stephan Dickgiesser, Nicolas Rasche, Andrea Paoletti, Elisa Bertotti, Ilse De Salve, Federico Riccardi Sirtori, Roland Kellner, Doreen Könning, Stefan Hecht, Jan Anderl, Harald Kolmar, Christian Schröter. Effect of Conjugation Site and Technique on the Stability and Pharmacokinetics of Antibody-Drug Conjugates. Journal of Pharmaceutical Sciences 2021, 110
(12)
, 3776-3785. https://doi.org/10.1016/j.xphs.2021.08.002
- Jeffrey Y.-K. Wong, Raja Mukherjee, Jiayuan Miao, Olena Bilyk, Vivian Triana, Mark Miskolzie, Antoine Henninot, John J. Dwyer, Serhii Kharchenko, Anna Iampolska, Dmitriy M. Volochnyuk, Yu-Shan Lin, Lynne-Marie Postovit, Ratmir Derda. Genetically-encoded discovery of proteolytically stable bicyclic inhibitors for morphogen NODAL. Chemical Science 2021, 12
(28)
, 9694-9703. https://doi.org/10.1039/D1SC01916C
- Ming-Chen Yang, Chi-Sheng Shia, Wan-Fen Li, Chun-Chung Wang, I-Ju Chen, Teng-Yi Huang, Yu-Jung Chen, Hui-Wen Chang, Chi-Huan Lu, Yueh-Chin Wu, Nan-Hsuan Wang, Jiann-Shiun Lai, Cheng-Der Yu, Ming-Tain Lai. Preclinical Studies of OBI-999: A Novel Globo H–Targeting Antibody–Drug Conjugate. Molecular Cancer Therapeutics 2021, 20
(6)
, 1121-1132. https://doi.org/10.1158/1535-7163.MCT-20-0763
- Jose F. Ponte, Leanne Lanieri, Eshita Khera, Rassol Laleau, Olga Ab, Christopher Espelin, Neeraj Kohli, Bahar Matin, Yulius Setiady, Michael L. Miller, Thomas A. Keating, Ravi Chari, Jan Pinkas, Richard Gregory, Greg M. Thurber. Antibody Co-Administration Can Improve Systemic and Local Distribution of Antibody–Drug Conjugates to Increase
In Vivo
Efficacy. Molecular Cancer Therapeutics 2021, 20
(1)
, 203-212. https://doi.org/10.1158/1535-7163.MCT-20-0451
- Amissi Sadiki, Shefali R Vaidya, Mina Abdollahi, Gunjan Bhardwaj, Michael E Dolan, Harpreet Turna, Varnika Arora, Athul Sanjeev, Timothy D Robinson, Andrea Koid, Aashka Amin, Zhaohui Sunny Zhou. Site-specific conjugation of native antibody. Antibody Therapeutics 2020, 3
(4)
, 271-284. https://doi.org/10.1093/abt/tbaa027
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 55 other publications.
- 1Chari, R. V. J., Miller, M. L., and Widdison, W. C. (2014) Antibody–Drug Conjugates: An Emerging Concept in Cancer Therapy. Angew. Chem., Int. Ed. 53 (15), 3796– 3827, DOI: 10.1002/anie.2013076281https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXjtVGmsr0%253D&md5=fbfb2f843f516654ea7c0eb98949604cAntibody-Drug Conjugates: An Emerging Concept in Cancer TherapyChari, Ravi V. J.; Miller, Michael L.; Widdison, Wayne C.Angewandte Chemie, International Edition (2014), 53 (15), 3796-3827CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Traditional cancer chemotherapy is often accompanied by systemic toxicity to the patient. Monoclonal antibodies against antigens on cancer cells offer an alternative tumor-selective treatment approach. However, most monoclonal antibodies are not sufficiently potent to be therapeutically active on their own. Antibody-drug conjugates (ADCs) use antibodies to deliver a potent cytotoxic compd. selectively to tumor cells, thus improving the therapeutic index of chemotherapeutic agents. The recent approval of two ADCs, brentuximab vedotin and ado-trastuzumab emtansine, for cancer treatment has spurred tremendous research interest in this field. This Review touches upon the early efforts in the field, and describes how the lessons learned from the first-generation ADCs have led to improvements in every aspect of this technol., i.e., the antibody, the cytotoxic compd., and the linker connecting them, leading to the current successes. The design of ADCs currently in clin. development, and results from mechanistic studies and preclin. and clin. evaluation are discussed. Emerging technologies that seek to further advance this exciting area of research are also discussed.
- 2Lambert, J. M. and Chari, R. V. (2014) Ado-trastuzumab Emtansine (T-DM1): an antibody-drug conjugate (ADC) for HER2-positive breast cancer. J. Med. Chem. 57 (16), 6949– 6964, DOI: 10.1021/jm500766w2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVGlu7rE&md5=3ddee50cc227097126dd47e7683a6ac7Ado-trastuzumab Emtansine (T-DM1): An Antibody-Drug Conjugate (ADC) for HER2-Positive Breast CancerLambert, John M.; Chari, Ravi V. J.Journal of Medicinal Chemistry (2014), 57 (16), 6949-6964CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. 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 deriv. of DM1 is subsequently released by proteolytic degrdn. of the antibody moiety within the lysosome. Initial clin. evaluation led to a phase III trial in advanced HER2-pos. 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-pos. metastatic breast cancer who had previously received trastuzumab and a taxane, sep. or in combination, the first ADC to receive full approval based on a randomized study.
- 3Senter, P. D. and Sievers, E. L. (2012) The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nat. Biotechnol. 30 (7), 631– 637, DOI: 10.1038/nbt.22893https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XpvFCqtbw%253D&md5=6940eb8178e8c8c150a7b21361e23234The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphomaSenter, Peter D.; Sievers, Eric L.Nature Biotechnology (2012), 30 (7), 631-637CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)A review. Progress was made recently in developing antibody-drug conjugates (ADCs) that can selectively deliver cancer drugs to tumor cells. In principle, the idea is simple: by attaching drugs to tumor-seeking antibodies, target cells will be killed and nontarget cells will be spared. In practice, many parameters needed to be addressed to develop safe and effective ADCs, including the expression profiles of tumor vs. normal tissues, the potency of the drug, the linker attaching the drug and placement of the drug on the antibody, and the pharmacokinetic and stability profiles of the resulting ADC. All these issues had been taken into account in developing brentuximab vedotin (Adcetris), an ADC that recently received accelerated approval by the US Food and Drug Administration for the treatment of relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma (ALCL). Research is under way to extend the applications of brentuximab vedotin and to advance the field by developing other ADCs with new linker and conjugation strategies.
- 4Appelbaum, F. R. and Bernstein, I. D. (2017) Gemtuzumab ozogamicin for acute myeloid leukemia. Blood 130 (22), 2373– 2376, DOI: 10.1182/blood-2017-09-7977124https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhs1erurfM&md5=970680b53cd9f2971e811d044ef9b6fdGemtuzumab ozogamicin for acute myeloid leukemiaAppelbaum, Frederick R.; Bernstein, Irwin D.Blood (2017), 130 (22), 2373-2376CODEN: BLOOAW; ISSN:1528-0020. (American Society of Hematology)On 1 Sept. 2017, the US Food and Drug Administration (FDA) approved gemtuzumab ozogamicin (GO) for the treatment of adults with newly diagnosed CD33+ acute myeloid leukemia and for patients aged ≥2 years with CD33+ acute myeloid leukemia who have experienced a relapse or who have not responded to initial treatment. This signals a new chapter in the long and unusual story of GO, which was the first antibody-drug conjugate approved for human use by the FDA.
- 5Yurkiewicz, I. R., Muffly, L., and Liedtke, M. (2018) Inotuzumab ozogamicin: a CD22 mAb-drug conjugate for adult relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Drug Des., Dev. Ther. 12, 2293– 2300, DOI: 10.2147/DDDT.S1503175https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1OnsrrM&md5=de9ba79b0c4d5a13606416647011fe8cInotuzumab ozogamicin: a CD22 mAb-drug conjugate for adult relapsed or refractory B-cell precursor acute lymphoblastic leukemiaYurkiewicz, Ilana R.; Muffly, Lori; Liedtke, MichaelaDrug Design, Development and Therapy (2018), 12 (), 2293-2300CODEN: DDDTAQ; ISSN:1177-8881. (Dove Medical Press Ltd.)Despite improved rates of remission and cure in newly diagnosed adult acute lymphoblastic leukemia (ALL), the prognosis for patients with relapsed or refractory disease remains poor and the 5-yr overall survival rate after relapse is under 10%. A recent paradigm shift has focused on the promise of targeted immunotherapy rather than std. chemotherapy, as ALL blast cells express a variety of antigens, and monoclonal antibodies may be developed to identify and destroy the leukemic cells. Inotuzumab ozogamicin is a CD22 monoclonal antibody conjugated to the cytotoxic antibiotic calicheamicin. CD22 expression is detected on leukemic blasts in over 90% of patients with ALL. Based on promising results from preclin. studies, inotuzumab ozogamicin was tested in Phase 1/2 and Phase 3 clin. trials and it demonstrated improved complete remission rates, progression-free survival and overall survival in relapsed or refractory adult ALL compared to std. therapy. Ongoing studies are evaluating the value of inotuzumab ozogamicin when given in combination with chemotherapy as part of upfront treatment. This review discusses the drug's biochem. properties and mechanism of action, preclin. research outcomes, clin. trial results, adverse events and toxicities, drug approval and ongoing investigations.
- 6Dornan, D., Bennett, F., Chen, Y., Dennis, M., Eaton, D., Elkins, K., French, D., Go, M. A., Jack, A., Junutula, J. R. (2009) Therapeutic potential of an anti-CD79b antibody-drug conjugate, anti-CD79b-vc-MMAE, for the treatment of non-Hodgkin lymphoma. Blood 114 (13), 2721– 2729, DOI: 10.1182/blood-2009-02-2055006https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1eit7jO&md5=02b943d887958c47dd2e5d1a3d490371Therapeutic potential of an anti-CD79b antibody-drug conjugate, anti-CD79b-vc-MMAE, for the treatment of non-Hodgkin lymphomaDornan, David; Bennett, Fiona; Chen, Yvonne; Dennis, Mark; Eaton, Dan; Elkins, Kristi; French, Dorothy; Go, Mary Ann T.; Jack, Andrew; Junutula, Jagath R.; Koeppen, Hartmut; Lau, Jeffrey; McBride, Jacqueline; Rawstron, Andy; Shi, Xiaoyan; Yu, Nancy; Yu, Shang-Fan; Yue, Peng; Zheng, Bing; Ebens, Allen; Polson, Andrew G.Blood (2009), 114 (13), 2721-2729CODEN: BLOOAW; ISSN:0006-4971. (American Society of Hematology)Here we describe the generation of an antibody-drug conjugate (ADC) consisting of a humanized anti-CD79b antibody that is conjugated to monomethylauristatin E (MMAE) through engineered cysteines (THIOMABs) by a protease cleavable linker. By using flow cytometry, we detected the surface expression of CD79b in almost all non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia patients, suggesting that anti-CD79b-vcMMAE could be widely used in these malignancies. By using NHL cell lines to simulate a patient population we discovered that a minimal cell-surface expression level of CD79b was required for in vitro activity. Within the subpopulation of cell lines above this minimal threshold, we found that sensitivity to free MMAE, mutation of cancer genes, and cell doubling time were poorly correlated with in vitro activity; however, the expression level of BCL-XL was correlated with reduced sensitivity to anti-CD79b-vcMMAE. This observation was supported by in vivo data showing that a Bcl-2 family inhibitor, ABT-263, strikingly enhanced the activity of anti-CD79b-vcMMAE. Furthermore, anti-CD79b-vcMMAE was significantly more effective than a std.-of-care regimen, R-CHOP (ie, rituximab with a single i.v. injection of 30 mg/kg cyclophosphamide, 2.475 mg/kg doxorubicin, 0.375 mg/kg vincristine, and oral dosing of 0.15 mg/kg prednisone once a day for 5 days), in 3 xenograft models of NHL. Together, these data suggest that anti-CD79b-vcMMAE could be broadly efficacious for the treatment of NHL.
- 7Donaghy, H. (2016) Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugates. MAbs 8 (4), 659– 671, DOI: 10.1080/19420862.2016.11568297https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmtFant7k%253D&md5=b2ba3e43e8362d8a3f7c368a5af5f94eEffects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugatesDonaghy, HeathermAbs (2016), 8 (4), 659-671CODEN: MABSCP; ISSN:1942-0870. (Taylor & Francis, Inc.)Antibody-drug conjugates (ADCs) represent a new class of cancer therapeutics. Their design involves a tumor-specific antibody, a linker and a cytotoxic payload. They were designed to allow specific targeting of highly potent cytotoxic agents to tumor cells while sparing normal cells. Frequent toxicities that may be driven by any of the components of an ADC have been reported. There are currently more than 50 ADCs in active clin. development, and a further ∼20 that have been discontinued. For this review, the reported toxicities of ADCs were analyzed, and the mechanisms for their effects are explored in detail. Methods to reduce toxicities, including dosing strategies and drug design, are discussed. The toxicities reported for active and discontinued drugs are important to drive the rational design and improve the therapeutic index of ADCs of the future.
- 8Chari, R. V. J. (2016) Expanding the Reach of Antibody-Drug Conjugates. ACS Med. Chem. Lett. 7 (11), 974– 976, DOI: 10.1021/acsmedchemlett.6b003128https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFWntrnN&md5=365b2432f4bc622eb92d01d5cb9c78a8Expanding the Reach of Antibody-Drug ConjugatesChari, Ravi V. J.ACS Medicinal Chemistry Letters (2016), 7 (11), 974-976CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Antibody-drug conjugates (ADCs) represent an emerging new paradigm in cancer therapy. The approval of two ADCs has spurred considerable interest in this area of research, and over 55 ADCs are currently in clin. testing. In order to improve the clin. success rate of ADC therapy, all three components of the ADC: the antibody, linker, and payload have to be optimized. While considerable improvements have been made in antibody properties and target selection, medicinal chem. efforts have lagged behind, and there is a significant need for innovation in linker design and payloads.
- 9Behrens, C. R. and Liu, B. (2014) Methods for site-specific drug conjugation to antibodies. MAbs 6 (1), 46– 53, DOI: 10.4161/mabs.266329https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2c%252FntlShtQ%253D%253D&md5=b23aea418d3a37ab7f5b94c3780a8e8cMethods for site-specific drug conjugation to antibodiesBehrens Christopher R; Liu BinmAbs (2014), 6 (1), 46-53 ISSN:.Antibody drug conjugates (ADCs) are an emerging class of targeted therapeutics with the potential to improve therapeutic index over traditional chemotherapy. Drugs and linkers have been the current focus of ADC development, in addition to antibody and target selection. Recently, however,the importance of conjugate homogeneity has been realized. The current methods for drug attachment lead to a heterogeneous mixture, and some populations of that mixture have poor in vivo performance. New methods for site-specific drug attachment lead to more homogeneous conjugates and allow control of the site of drug attachment. These subtle improvements can have profound effects on in vivo efficacy and therapeutic index. This review examines current methods for site-specific drug conjugation to antibodies, and compares in vivo results with their non-specifically conjugated counterparts. The apparent improvement in pharmacokinetics and the reduced off target toxicity warrant further development of this site-specific modification approach for future ADC development.
- 10Agarwal, P. and Bertozzi, C. R. (2015) Site-specific antibody-drug conjugates: the nexus of bioorthogonal chemistry, protein engineering, and drug development. Bioconjugate Chem. 26 (2), 176– 192, DOI: 10.1021/bc500498210https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFCrsrfK&md5=c3d822b0e53e5cdb074e5b63b8060ccfSite-Specific Antibody-Drug Conjugates: The Nexus of Bioorthogonal Chemistry, Protein Engineering, and Drug DevelopmentAgarwal, Paresh; Bertozzi, Carolyn R.Bioconjugate Chemistry (2015), 26 (2), 176-192CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)A review. Antibody-drug conjugates (ADCs) combine the specificity of antibodies with the potency of small mols. to create targeted drugs. Despite the simplicity of this concept, generation of clin. successful ADCs has been very difficult. Over the past several decades, scientists have learned a great deal about the constraints on antibodies, linkers, and drugs as they relate to successful construction of ADCs. Once these components are in hand, most ADCs are prepd. by nonspecific modification of antibody lysine or cysteine residues with drug-linker reagents, which results in heterogeneous product mixts. that cannot be further purified. With advances in the fields of bioorthogonal chem. and protein engineering, there is growing interest in producing ADCs by site-specific conjugation to the antibody, yielding more homogeneous products that have demonstrated benefits over their heterogeneous counterparts in vivo. Here, we chronicle the development of a multitude of site-specific conjugation strategies for assembly of ADCs and provide a comprehensive account of key advances and their roots in the fields of bioorthogonal chem. and protein engineering.
- 11Panowski, S., Bhakta, S., Raab, H., Polakis, P., and Junutula, J. R. (2014) Site-specific antibody drug conjugates for cancer therapy. MAbs 6 (1), 34– 45, DOI: 10.4161/mabs.2702211https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2cvnslaltw%253D%253D&md5=9da883692122c03303f785e61f6c5db7Site-specific antibody drug conjugates for cancer therapyPanowski Siler; Bhakta Sunil; Raab Helga; Polakis Paul; Junutula Jagath RmAbs (2014), 6 (1), 34-45 ISSN:.Antibody therapeutics have revolutionized the treatment of cancer over the past two decades. Antibodies that specifically bind tumor surface antigens can be effective therapeutics; however, many unmodified antibodies lack therapeutic activity. These antibodies can instead be applied successfully as guided missiles to deliver potent cytotoxic drugs in the form of antibody drug conjugates (ADCs). The success of ADCs is dependent on four factors--target antigen, antibody, linker, and payload. The field has made great progress in these areas, marked by the recent approval by the US Food and Drug Administration of two ADCs, brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla). However, the therapeutic window for many ADCs that are currently in pre-clinical or clinical development remains narrow and further improvements may be required to enhance the therapeutic potential of these ADCs. Production of ADCs is an area where improvement is needed because current methods yield heterogeneous mixtures that may include 0-8 drug species per antibody molecule. Site-specific conjugation has been recently shown to eliminate heterogeneity, improve conjugate stability, and increase the therapeutic window. Here, we review and describe various site-specific conjugation strategies that are currently used for the production of ADCs, including use of engineered cysteine residues, unnatural amino acids, and enzymatic conjugation through glycotransferases and transglutaminases. In addition, we also summarize differences among these methods and highlight critical considerations when building next-generation ADC therapeutics.
- 12Junutula, J. R., Raab, H., Clark, S., Bhakta, S., Leipold, D. D., Weir, S., Chen, Y., Simpson, M., Tsai, S. P., Dennis, M. S. (2008) Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat. Biotechnol. 26 (8), 925– 932, DOI: 10.1038/nbt.148012https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXps1Wmu7s%253D&md5=d55f59594fb6349d0337dfed2783e83cSite-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic indexJunutula, Jagath R.; Raab, Helga; Clark, Suzanna; Bhakta, Sunil; Leipold, Douglas D.; Weir, Sylvia; Chen, Yvonne; Simpson, Michelle; Tsai, Siao Ping; Dennis, Mark S.; Lu, Yanmei; Meng, Y. Gloria; Ng, Carl; Yang, Jihong; Lee, Chien C.; Duenas, Eileen; Gorrell, Jeffrey; Katta, Viswanatham; Kim, Amy; McDorman, Kevin; Flagella, Kelly; Venook, Rayna; Ross, Sarajane; Spencer, Susan D.; Wong, Wai Lee; Lowman, Henry B.; Vandlen, Richard; Sliwkowski, Mark X.; Scheller, Richard H.; Polakis, Paul; Mallet, WilliamNature Biotechnology (2008), 26 (8), 925-932CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Antibody-drug conjugates enhance the antitumor effects of antibodies and reduce adverse systemic effects of potent cytotoxic drugs. However, conventional drug conjugation strategies yield heterogeneous conjugates with relatively narrow therapeutic index (max. tolerated dose/curative dose). Using leads from the authors' previously described phage display-based method to predict suitable conjugation sites, the authors engineered cysteine substitutions at positions on light and heavy chains that provide reactive thiol groups and do not perturb Ig folding and assembly, or alter antigen binding. When conjugated to monomethyl auristatin E, an antibody against the ovarian cancer antigen MUC16 is as efficacious as a conventional conjugate in mouse xenograft models. Moreover, it is tolerated at higher doses in rats and cynomolgus monkeys than the same conjugate prepd. by conventional approaches. The favorable in vivo properties of the near-homogeneous compn. of this conjugate suggest that this strategy offers a general approach to retaining the antitumor efficacy of antibody-drug conjugates, while minimizing their systemic toxicity.
- 13Junutula, J. R., Flagella, K. M., Graham, R. A., Parsons, K. L., Ha, E., Raab, H., Bhakta, S., Nguyen, T., Dugger, D. L., Li, G. (2010) Engineered Thio-Trastuzumab-DM1 Conjugate with an Improved Therapeutic Index to Target Human Epidermal Growth Factor Receptor 2–Positive Breast Cancer. Clin. Cancer Res. 16 (19), 4769– 4778, DOI: 10.1158/1078-0432.CCR-10-098713https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXht1aktbjK&md5=888bda7f06d1ed864111b607dae5f969Engineered Thio-Trastuzumab-DM1 Conjugate with an Improved Therapeutic Index to Target Human Epidermal Growth Factor Receptor 2-Positive Breast CancerJunutula, Jagath R.; Flagella, Kelly M.; Graham, Richard A.; Parsons, Kathryn L.; Ha, Edward; Raab, Helga; Bhakta, Sunil; Nguyen, Trung; Dugger, Debra L.; Li, Guangmin; Mai, Elaine; Lewis Phillips, Gail D.; Hiraragi, Hajime; Fuji, Reina N.; Tibbitts, Jay; Vandlen, Richard; Spencer, Susan D.; Scheller, Richard H.; Polakis, Paul; Sliwkowski, Mark X.Clinical Cancer Research (2010), 16 (19), 4769-4778CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Antibody drug conjugates (ADCs) combine the ideal properties of both antibodies and cytotoxic drugs by targeting potent drugs to the antigen-expressing tumor cells, thereby enhancing their antitumor activity. Successful ADC development for a given target antigen depends on optimization of antibody selection, linker stability, cytotoxic drug potency, and mode of linker-drug conjugation to the antibody. Here, we systematically examd. the in vitro potency as well as in vivo preclin. efficacy and safety profiles of a heterogeneous prepn. of conventional trastuzumab-mcc-DM1 (TMAb-mcc-DM1) ADC with that of a homogeneous engineered thio-trastuzumab-mpeo-DM1 (thioTMAb-mpeo-DM1) conjugate. To generate thioTMAb-mpeo-DM1, one drug maytansinoid 1 (DM1) mol. was conjugated to an engineered cysteine residue at Ala114 (Kabat numbering) on each trastuzumab-heavy chain, resulting in two DM1 mols. per antibody. ThioTMAb-mpeo-DM1 retained similar in vitro anti-cell proliferation activity and human epidermal growth factor receptor 2 (HER2) binding properties to that of the conventional ADC. Furthermore, it showed improved efficacy over the conventional ADC at DM1-equiv doses (μg/m2) and retained efficacy at equiv. antibody doses (mg/kg). An improved safety profile of >2-fold was obsd. in a short-term target-independent rat safety study. In cynomolgus monkey safety studies, thioTMAb-mpeo-DM1 was tolerated at higher antibody doses (up to 48 mg/kg or 6000 μg DM1/m2) compared with the conventional ADC that had dose-limiting toxicity at 30 mg/kg (6000 μg DM1/m2). The engineered thioTMAb-mpeo-DM1 with broadened therapeutic index represents a promising antibody drug conjugate for future clin. development of HER2-pos. targeted breast cancer therapies.
- 14Shen, B. Q., Xu, K., Liu, L., Raab, H., Bhakta, S., Kenrick, M., Parsons-Reponte, K. L., Tien, J., Yu, S. F., Mai, E. (2012) Conjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugates. Nat. Biotechnol. 30 (2), 184– 189, DOI: 10.1038/nbt.210814https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtVGqtLg%253D&md5=c647fc5bdfe93dfdabb15ca40934837fConjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugatesShen, Ben-Quan; Xu, Keyang; Liu, Luna; Raab, Helga; Bhakta, Sunil; Kenrick, Margaret; Parsons-Reponte, Kathryn L.; Tien, Janet; Yu, Shang-Fan; Mai, Elaine; Li, Dongwei; Tibbitts, Jay; Baudys, Jakub; Saad, Ola M.; Scales, Suzie J.; McDonald, Paul J.; Hass, Philip E.; Eigenbrot, Charles; Nguyen, Trung; Solis, Willy A.; Fuji, Reina N.; Flagella, Kelly M.; Patel, Darshana; Spencer, Susan D.; Khawli, Leslie A.; Ebens, Allen; Wong, Wai Lee; Vandlen, Richard; Kaur, Surinder; Sliwkowski, Mark X.; Scheller, Richard H.; Polakis, Paul; Junutula, Jagath R.Nature Biotechnology (2012), 30 (2), 184-189CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)The reactive thiol in cysteine is used for coupling maleimide linkers in the generation of antibody conjugates. To assess the impact of the conjugation site, we engineered cysteines into a therapeutic HER2/neu antibody at three sites differing in solvent accessibility and local charge. The highly solvent-accessible site rapidly lost conjugated thiol-reactive linkers in plasma owing to maleimide exchange with reactive thiols in albumin, free cysteine or glutathione. In contrast, a partially accessible site with a pos. charged environment promoted hydrolysis of the succinimide ring in the linker, thereby preventing this exchange reaction. The site with partial solvent-accessibility and neutral charge displayed both properties. In a mouse mammary tumor model, the stability and therapeutic activity of the antibody conjugate were affected pos. by succinimide ring hydrolysis and neg. by maleimide exchange with thiol-reactive constituents in plasma. Thus, the chem. and structural dynamics of the conjugation site can influence antibody conjugate performance by modulating the stability of the antibody-linker interface.
- 15Pillow, T. H., Tien, J., Parsons-Reponte, K. L., Bhakta, S., Li, H., Staben, L. R., Li, G., Chuh, J., Fourie-O’Donohue, A., Darwish, M. (2014) Site-specific trastuzumab maytansinoid antibody-drug conjugates with improved therapeutic activity through linker and antibody engineering. J. Med. Chem. 57 (19), 7890– 7899, DOI: 10.1021/jm500552c15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsV2htbbN&md5=084727e56c1770103910d993a71f95b1Site-Specific Trastuzumab Maytansinoid Antibody-Drug Conjugates with Improved Therapeutic Activity through Linker and Antibody EngineeringPillow, Thomas H.; Tien, Janet; Parsons-Reponte, Kathryn L.; Bhakta, Sunil; Li, Hao; Staben, Leanna R.; Li, Guangmin; Chuh, Josefa; Fourie-O'Donohue, Aimee; Darwish, Martine; Yip, Victor; Liu, Luna; Leipold, Douglas D.; Su, Dian; Wu, Elmer; Spencer, Susan D.; Shen, Ben-Quan; Xu, Keyang; Kozak, Katherine R.; Raab, Helga; Vandlen, Richard; Lewis Phillips, Gail D.; Scheller, Richard H.; Polakis, Paul; Sliwkowski, Mark X.; Flygare, John A.; Junutula, Jagath R.Journal of Medicinal Chemistry (2014), 57 (19), 7890-7899CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Antibody-drug conjugates (ADCs) have a significant impact toward the treatment of cancer, as evidenced by the clin. activity of the recently approved ADCs, brentuximab vedotin for Hodgkin lymphoma and ado-trastuzumab emtansine (trastuzumab-MCC-DM1) for metastatic HER2+ breast cancer. DM1 is an analog of the natural product maytansine, a microtubule inhibitor that by itself has limited clin. activity and high systemic toxicity. However, by conjugation of DM1 to trastuzumab, the safety was improved and clin. activity was demonstrated. Here, we report that through chem. modification of the linker-drug and antibody engineering, the therapeutic activity of trastuzumab maytansinoid ADCs can be further improved. These improvements include eliminating DM1 release in the plasma and increasing the drug load by engineering four cysteine residues into the antibody. The chem. synthesis of highly stable linker-drugs and the modification of cysteine residues of engineered site-specific antibodies resulted in a homogeneous ADC with increased therapeutic activity compared to the clin. approved ADC, trastuzumab-MCC-DM1.
- 16Tian, F., Lu, Y., Manibusan, A., Sellers, A., Tran, H., Sun, Y., Phuong, T., Barnett, R., Hehli, B., Song, F. (2014) A general approach to site-specific antibody drug conjugates. Proc. Natl. Acad. Sci. U. S. A. 111 (5), 1766– 1771, DOI: 10.1073/pnas.132123711116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFyjs7g%253D&md5=293401bcfaa0d74ce03daca26e9ad18eA general approach to site-specific antibody drug conjugatesTian, Feng; Lu, Yingchun; Manibusan, Anthony; Sellers, Aaron; Tran, Hon; Sun, Ying; Phuong, Trung; Barnett, Richard; Hehli, Brad; Song, Frank; De Guzman, Michael J.; Ensari, Semsi; Pinkstaff, Jason K.; Sullivan, Lorraine M.; Biroc, Sandra L.; Cho, Ho; Schultz, Peter G.; Di Joseph, John; Dougher, Maureen; Ma, Dangshe; Dushin, Russell; Leal, Mauricio; Tchistiakova, Lioudmila; Feyfant, Eric; Gerber, Hans-Peter; Sapra, PujaProceedings of the National Academy of Sciences of the United States of America (2014), 111 (5), 1766-1771CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Using an expanded genetic code, antibodies with site-specifically incorporated nonnative amino acids were produced in stable cell lines derived from a CHO cell line with titers over 1 g/L. Using anti-5T4 and anti-Her2 antibodies as model systems, site-specific antibody drug conjugates (NDCs) were produced, via oxime bond formation between ketones on the side chain of the incorporated nonnative amino acid and hydroxylamine functionalized monomethyl auristatin D with either protease-cleavable or noncleavable linkers. When noncleavable linkers were used, these conjugates were highly stable and displayed improved in vitro efficacy as well as in vivo efficacy and pharmacokinetic stability in rodent models relative to conventional antibody drug conjugates conjugated through either engineered surface-exposed or reduced interchain disulfide bond cysteine residues. The advantages of the oxime-bonded, site-specific NDCs were even more apparent when low-antigen-expressing (2+) target cell lines were used in the comparative studies. NDCs generated with protease-cleavable linkers demonstrated that the site of conjugation had a significant impact on the stability of these rationally designed prodrug linkers. In a single-dose rat toxicol. study, a site-specific anti-Her2 NDC was well tolerated at dose levels up to 90 mg/kg. These expts. support the notion that chem. defined antibody conjugates can be synthesized in com. relevant yields and can lead to antibody drug conjugates with improved properties relative to the heterogeneous conjugates formed by nonspecific chem. modification.
- 17Jackson, D., Atkinson, J., Guevara, C. I., Zhang, C., Kery, V., Moon, S. J., Virata, C., Yang, P., Lowe, C., Pinkstaff, J. (2014) In vitro and in vivo evaluation of cysteine and site specific conjugated herceptin antibody-drug conjugates. PLoS One 9 (1), e83865 DOI: 10.1371/journal.pone.008386517https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXivVOit7g%253D&md5=02721a9b2d6982d9915c76d604cebac3In vitro and in vivo evaluation of cysteine and site specific conjugated herceptin antibody-drug conjugatesJackson, Dowdy; Atkinson, John; Guevara, Claudia I.; Zhang, Chunying; Kery, Vladimir; Moon, Sung-Ju; Virata, Cyrus; Yang, Peng; Lowe, Christine; Pinkstaff, Jason; Cho, Ho; Knudsen, Nick; Manibusan, Anthony; Tian, Feng; Sun, Ying; Lu, Yingchun; Sellers, Aaron; Jia, Xiao-Chi; Joseph, Ingrid; Anand, Banmeet; Morrison, Kendall; Pereira, Daniel S.; Stover, DavidPLoS One (2014), 9 (1), e83865/1-e83865/14, 14 pp.CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Antibody-drug conjugates (ADCs) are monoclonal antibodies designed to deliver a cytotoxic drug selectively to antigen expressing cells. Several components of an ADC including the selection of the antibody, the linker, the cytotoxic drug payload and the site of attachment used to attach the drug to the antibody are crit. to the activity and development of the ADC. The cytotoxic drugs or payloads used to make ADCs are typically conjugated to the antibody through cysteine or lysine residues. This results in ADCs that have a heterogeneous no. of drugs per antibody. The no. of drugs per antibody commonly referred to as the drug to antibody ratio (DAR), can vary between 0 and 8 drugs for a IgG1 antibody. Antibodies with 0 drugs are ineffective and compete with the ADC for binding to the antigen expressing cells. Antibodies with 8 drugs per antibody have reduced in vivo stability, which may contribute to non target related toxicities. In these studies we incorporated a non-natural amino acid, para acetyl phenylalanine, at two unique sites within an antibody against Her2/neu. We covalently attached a cytotoxic drug to these sites to form an ADC which contains two drugs per antibody. We report the results from the first direct preclin. comparison of a site specific non-natural amino acid anti-Her2 ADC and a cysteine conjugated anti-Her2 ADC. We report that the site specific non-natural amino acid anti-Her2 ADCs have superior in vitro serum stability and preclin. toxicol. profile in rats as compared to the cysteine conjugated anti-Her2 ADCs. We also demonstrate that the site specific non-natural amino acid anti-Her2 ADCs maintain their in vitro potency and in vivo efficacy against Her2 expressing human tumor cell lines. Our data suggests that site specific non-natural amino acid ADCs may have a superior therapeutic window than cysteine conjugated ADCs.
- 18Strop, P., Delaria, K., Foletti, D., Witt, J. M., Hasa-Moreno, A., Poulsen, K., Casas, M. G., Dorywalska, M., Farias, S., Pios, A. (2015) Site-specific conjugation improves therapeutic index of antibody drug conjugates with high drug loading. Nat. Biotechnol. 33 (7), 694– 696, DOI: 10.1038/nbt.327418https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1ensLbK&md5=43b6c9f702e02a91887c3f27a5ab7353Site-specific conjugation improves therapeutic index of antibody drug conjugates with high drug loadingStrop, Pavel; Delaria, Kathy; Foletti, Davide; Witt, Jody Melton; Hasa-Moreno, Adela; Poulsen, Kris; Casas, Meritxell Galindo; Dorywalska, Magdalena; Farias, Santiago; Pios, Ariel; Lui, Victor; Dushin, Russell; Zhou, Dahui; Navaratnam, Thayalan; Tran, Thomas-Toan; Sutton, Janette; Lindquist, Kevin C.; Han, Bora; Liu, Shu-Hui; Shelton, David L.; Pons, Jaume; Rajpal, ArvindNature Biotechnology (2015), 33 (7), 694-696CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)There is no expanded citation for this reference.
- 19Behrens, C. R., Ha, E. H., Chinn, L. L., Bowers, S., Probst, G., Fitch-Bruhns, M., Monteon, J., Valdiosera, A., Bermudez, A., Liao-Chan, S. (2015) Antibody-Drug Conjugates (ADCs) Derived from Interchain Cysteine Cross-Linking Demonstrate Improved Homogeneity and Other Pharmacological Properties over Conventional Heterogeneous ADCs. Mol. Pharmaceutics 12 (11), 3986– 3998, DOI: 10.1021/acs.molpharmaceut.5b0043219https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFejtLjE&md5=d3657c76c415b621877a90b0d5a82025Antibody-Drug Conjugates (ADCs) Derived from Interchain Cysteine Cross-Linking Demonstrate Improved Homogeneity and Other Pharmacological Properties over Conventional Heterogeneous ADCsBehrens, Christopher R.; Ha, Edward H.; Chinn, Lawrence L.; Bowers, Simeon; Probst, Gary; Fitch-Bruhns, Maureen; Monteon, Jorge; Valdiosera, Amanda; Bermudez, Abel; Liao-Chan, Sindy; Wong, Tiffany; Melnick, Jonathan; Theunissen, Jan-Willem; Flory, Mark R.; Houser, Derrick; Venstrom, Kristy; Levashova, Zoia; Sauer, Paul; Migone, Thi-Sau; van der Horst, Edward H.; Halcomb, Randall L.; Jackson, David Y.Molecular Pharmaceutics (2015), 12 (11), 3986-3998CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Conventional antibody-drug conjugates (ADCs) are heterogeneous mixts. of chem. distinct mols. that vary in both drugs/antibody (DAR) and conjugation sites. Suboptimal properties of heterogeneous ADCs have led to new site-specific conjugation methods for improving ADC homogeneity. Most site-specific methods require extensive antibody engineering to identify optimal conjugation sites and introduce unique functional groups for conjugation with appropriately modified linkers. Alternative nonrecombinant methods have emerged in which bifunctional linkers are utilized to cross-link antibody interchain cysteines and afford ADCs contg. four drugs/antibody. Although these methods have been shown to improve ADC homogeneity and stability in vitro, their effect on the pharmacol. properties of ADCs in vivo is unknown. In order to det. the relative impact of interchain cysteine crosslinking on the therapeutic window and other properties of ADCs in vivo, we synthesized a deriv. of the known ADC payload, MC-MMAF, that contains a bifunctional dibromomaleimide (DBM) linker instead of a conventional maleimide (MC) linker. The DBM-MMAF deriv. was conjugated to trastuzumab and a novel anti-CD98 antibody to afford ADCs contg. predominantly four drugs/antibody. The pharmacol. properties of the resulting cross-linked ADCs were compared with analogous heterogeneous ADCs derived from conventional linkers. The results demonstrate that DBM linkers can be applied directly to native antibodies, without antibody engineering, to yield highly homogeneous ADCs via cysteine crosslinking. The resulting ADCs demonstrate improved pharmacokinetics, superior efficacy, and reduced toxicity in vivo compared to analogous conventional heterogeneous ADCs.
- 20Hamblett, K. J., Le, T., Rock, B. M., Rock, D. A., Siu, S., Huard, J. N., Conner, K. P., Milburn, R. R., O’Neill, J. W., Tometsko, M. E. (2016) Altering Antibody–Drug Conjugate Binding to the Neonatal Fc Receptor Impacts Efficacy and Tolerability. Mol. Pharmaceutics 13 (7), 2387– 2396, DOI: 10.1021/acs.molpharmaceut.6b0015320https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XovVylsbg%253D&md5=8883777d4731222b197f1d0109415f5bAltering Antibody-Drug Conjugate Binding to the Neonatal Fc Receptor Impacts Efficacy and TolerabilityHamblett, Kevin J.; Le, Tiep; Rock, Brooke M.; Rock, Dan A.; Siu, Sophia; Huard, Justin N.; Conner, Kip P.; Milburn, Robert R.; O'Neill, Jason W.; Tometsko, Mark E.; Fanslow, William C.Molecular Pharmaceutics (2016), 13 (7), 2387-2396CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Antibody-drug conjugates (ADC) rely on the target-binding specificity of an antibody to selectively deliver potent drugs to cancer cells. IgG antibody half-life is regulated by neonatal Fc receptor (FcRn) binding. Histidine 435 of human IgG was mutated to alanine (H435A) to explore the effect of FcRn binding on the pharmacokinetics, efficacy, and tolerability of two sep. maytansine-based ADC pairs with noncleavable linkers, (c-DM1 and c-H435A-DM1) and (7v-Cys-may and 7v-H435A-Cys-may). The in vitro cell-killing potency of each pair of ADCs was similar, demonstrating that H435A showed no measurable impact on ADC bioactivity. The H435A mutant antibodies showed no detectable binding to human or mouse FcRn in vitro, whereas their counterpart wild-type IgG ADCs were found to bind to FcRn at pH = 6.0. In xenograft bearing SCID mice expressing mouse FcRn, the AUC of 7v-Cys-may was 1.6-fold higher than that of 7v-H435A-may, yet the obsd. efficacy was similar. More severe thrombocytopenia was obsd. with 7v-H435A-Cys-may as compared to 7v-Cys-may at multiple dose levels. The AUC of c-DM1 was approx. 3-fold higher than that of c-H435A-DM1 in 786-0 xenograft bearing SCID mice, which led to a 3-fold difference in efficacy by dose. Murine FcRn knockout, human FcRn transgenic line 32 SCID animals bearing 786-0 xenografts showed an amplified exposure difference between c-DM1 and c-H435A-DM1 as compared to murine FcRn expressing SCID mice, leading to a 10-fold higher dose required for efficacy despite a 6-fold higher AUC of the c-H435A-DM1. The accelerated clearance obsd. for the noncleavable maytansine ADCs with the H435A FcRn mutation led to reduced efficacy at equiv. doses and exacerbation of clin. pathol. parameters (decreased tolerability) at equiv. doses. The results show that reduced ADC clearance mediated by FcRn modulation can improve therapeutic index.
- 21Lhospice, F., Bregeon, D., Belmant, C., Dennler, P., Chiotellis, A., Fischer, E., Gauthier, L., Boedec, A., Rispaud, H., Savard-Chambard, S. (2015) Site-Specific Conjugation of Monomethyl Auristatin E to Anti-CD30 Antibodies Improves Their Pharmacokinetics and Therapeutic Index in Rodent Models. Mol. Pharmaceutics 12 (6), 1863– 1871, DOI: 10.1021/mp500666j21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsV2gsrs%253D&md5=ef6ab4dab9fa40d15d03c98a58917cd0Site-Specific Conjugation of Monomethyl Auristatin E to Anti-CD30 Antibodies Improves Their Pharmacokinetics and Therapeutic Index in Rodent ModelsLhospice, F.; Bregeon, D.; Belmant, C.; Dennler, P.; Chiotellis, A.; Fischer, E.; Gauthier, L.; Boedec, A.; Rispaud, H.; Savard-Chambard, S.; Represa, A.; Schneider, N.; Paturel, C.; Sapet, M.; Delcambre, C.; Ingoure, S.; Viaud, N.; Bonnafous, C.; Schibli, R.; Romagne, F.Molecular Pharmaceutics (2015), 12 (6), 1863-1871CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Antibody-drug conjugates (ADCs) have demonstrated clin. benefits that have led to the recent FDA approval of KADCYLA and ADCETRIS. Most ADCs that are currently in clin. use or development, including ADCETRIS, are produced by chem. conjugation of a toxin via either lysine or cysteine residues, inevitably leading to heterogeneous products with variable drug-to-antibody ratios (DARs). Here, the authors describe the in vitro and in vivo characterization of four novel ADCs that are based on the anti-CD30 antibody cAC10, which has the same polypeptide backbone as ADCETRIS, and compare the results with the latter. Bacterial transglutaminase (BTG) was exploited to site-specifically conjugate derivs. of monomethyl auristatin E (all comprising a cleavable linker) to the glutamine at positions 295 and 297 of cAC10, thereby yielding homogeneous ADCs with a DAR of 4. In vitro cell toxicity expts. using two different CD30-pos. cell lines (Karpas 299 and Raji-CD30+) revealed comparable EC50 values for ADCETRIS (1.8±0.4 and 3.6±0.6 ng/mL, resp.) and the four cAC10-based ADCs (2.0±0.4 to 4.9±1.0 ng/mL). Quant. time-dependent in vivo biodistribution studies (3-96 h p.i.) in normal and xenografted (Karpas 299 cells) SCID mice were performed with a selected 125I-radioiodinated cAC10 ADC and compared with that of 125I-ADCETRIS. The chemo-enzymically conjugated, radioiodinated ADC showed higher tumor uptake (17.84±2.2% ID/g 24 h p.i.) than 125I-ADCETRIS (10.5±1.8% ID/g 24 h p.i.). Moreover, 125I-ADCETRIS exhibited higher nontargeted liver and spleen uptake. In line with these results, the max. tolerated dose of the BTG-coupled ADC (>60 mg/kg) was significantly higher than that of ADCETRIS (18 mg/kg) in rats. These results suggest that homogeneous ADCs display improved pharmacokinetics and better therapeutic indexes compared to those of chem. modified ADCs with variable DARs.
- 22Yoder, N. C., Bai, C., Tavares, D., Widdison, W. C., Whiteman, K. R., Wilhelm, A., Wilhelm, S. D., McShea, M. A., Maloney, E. K., Ab, O. (2019) A Case Study Comparing Heterogeneous Lysine- and Site-Specific Cysteine-Conjugated Maytansinoid Antibody-Drug Conjugates (ADCs) Illustrates Benefits of Lysine Conjugation. Mol. Pharmaceutics 16 (9), 3926– 3937, DOI: 10.1021/acs.molpharmaceut.9b0052922https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlaqsrrL&md5=d88fd3fc17185f75b3ba7ac6afc5bb30A Case Study Comparing Heterogeneous Lysine- and Site-Specific Cysteine-Conjugated Maytansinoid Antibody-Drug Conjugates (ADCs) Illustrates the Benefits of Lysine ConjugationYoder, Nicholas C.; Bai, Chen; Tavares, Daniel; Widdison, Wayne C.; Whiteman, Kathleen R.; Wilhelm, Alan; Wilhelm, Sharon D.; McShea, Molly A.; Maloney, Erin K.; Ab, Olga; Wang, Lintao; Jin, Shan; Erickson, Hans K.; Keating, Thomas A.; Lambert, John M.Molecular Pharmaceutics (2019), 16 (9), 3926-3937CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Antibody-drug conjugates are an emerging class of cancer therapeutics constructed from monoclonal antibodies conjugated with small mol. effectors. First-generation mols. of this class often employed heterogeneous conjugation chem., but many site-specifically conjugated ADCs have been described recently. Here, we undertake a systematic comparison of ADCs made with the same antibody and the same macrocyclic maytansinoid effector but conjugated either heterogeneously at lysine residues or site-specifically at cysteine residues. Characterization of these ADCs in vitro reveals generally similar properties, including a similar catabolite profile, a key element in making a meaningful comparison of conjugation chemistries. In a mouse model of cervical cancer, the lysine-conjugated ADC affords greater efficacy on a molar payload basis. Rather than making general conclusions about ADCs conjugated by a particular chem., we interpret these results as highlighting the complexity of ADCs and the interplay between payload class, linker chem., target antigen, and other variables that det. efficacy in a given setting.
- 23Miller, M. L., Shizuka, M., Wilhelm, A., Salomon, P., Reid, E. E., Lanieri, L., Sikka, S., Maloney, E. K., Harvey, L., Qiu, Q. (2018) A DNA-Interacting Payload Designed to Eliminate Cross-Linking Improves the Therapeutic Index of Antibody-Drug Conjugates (ADCs). Mol. Cancer Ther. 17 (3), 650– 660, DOI: 10.1158/1535-7163.MCT-17-094023https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjslKlu7c%253D&md5=aa83bf47124ecc290bd3e14f56df8b1fA DNA-Interacting Payload Designed to Eliminate Cross-Linking Improves the Therapeutic Index of Antibody-Drug Conjugates (ADCs)Miller, Michael L.; Shizuka, Manami; Wilhelm, Alan; Salomon, Paulin; Reid, Emily E.; Lanieri, Leanne; Sikka, Surina; Maloney, Erin K.; Harvey, Lauren; Qiu, Qifeng; Archer, Katie E.; Bai, Chen; Vitharana, Dilrukshi; Harris, Luke; Singh, Rajeeva; Ponte, Jose F.; Yoder, Nicholas C.; Kovtun, Yelena; Lai, Katharine C.; Ab, Olga; Pinkas, Jan; Keating, Thomas A.; Chari, Ravi V. J.Molecular Cancer Therapeutics (2018), 17 (3), 650-660CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)Tumor-selective delivery of cytotoxic agents in the form of antibody-drug conjugates (ADCs) is now a clin. validated approach for cancer treatment. In an attempt to improve the clin. success rate of ADCs, emphasis has been recently placed on the use of DNA-crosslinking pyrrolobenzodiazepine compds. as the payload. Despite promising early clin. results with this class of ADCs, doses achievable have been low due to systemic toxicity. Here, we describe the development of a new class of potent DNA-interacting agents wherein changing the mechanism of action from a cross-linker to a DNA alkylator improves the tolerability of the ADC. ADCs contg. the DNA alkylator displayed similar in vitro potency, but improved bystander killing and in vivo efficacy, compared with those of the cross-linker. Thus, the improved in vivo tolerability and antitumor activity achieved in rodent models with ADCs of the novel DNA alkylator could provide an efficacious, yet safer option for cancer treatment. Mol Cancer Ther; 17(3); 650-60. ©2018 AACR.
- 24Stimmel, J. B., Merrill, B. M., Kuyper, L. F., Moxham, C. P., Hutchins, J. T., Fling, M. E., and Kull, F. C. (2000) Site-specific conjugation on serine -> cysteine variant monoclonal antibodies. J. Biol. Chem. 275 (39), 30445– 30450, DOI: 10.1074/jbc.M00167220024https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXnt1agsbY%253D&md5=0763640e21082823d1b0b83663389f86Site-specific conjugation on serine → cysteine variant monoclonal antibodiesStimmel, Julie B.; Merrill, Barbara M.; Kuyper, Lee F.; Moxham, Cary P.; Hutchins, Jeff T.; Fling, Mary E.; Kull, Frederick C., Jr.Journal of Biological Chemistry (2000), 275 (39), 30445-30450CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)We have engineered a cysteine residue at position 442 (EU/OU numbering) in the third const. domain (CH3) of the heavy chain of several IgGs with different specificities, isoforms, and variants with the intent to introduce a site for chem. conjugation. The variants were expressed in NS0 mouse myeloma cells, where monomeric IgG is the major form and formation of aggregate was minimal. Monomeric IgG contained no free thiol; however, it was discovered that the engineered thiols were reversibly blocked and could be reduced under controlled conditions. Following redn., reactive thiol was conjugated with a cysteine-specific bifunctional chelator, bromoacetyl-TMT to a humanized 323/A3 IgG4 variant. Conjugation had no significant effect on antibody affinity. To prove that the conjugation was site-specific, an antibody-TMT conjugate was labeled with lutetium-177 and subjected to peptide mapping followed by sequence anal. Glu-C digestion demonstrated that 91% of the label was recovered in the COOH-terminal peptide fragment contg. the engineered cysteine.
- 25Cao, M., De Mel, N., Jiao, Y., Howard, J., Parthemore, C., Korman, S., Thompson, C., Wendeler, M., and Liu, D. (2019) Site-specific antibody-drug conjugate heterogeneity characterization and heterogeneity root cause analysis. MAbs 11 (6), 1064– 1076, DOI: 10.1080/19420862.2019.162412725https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFOjsrrN&md5=565ad2567367d9fad04088d85523a876Site-specific antibody-drug conjugate heterogeneity characterization and heterogeneity root cause analysisCao, Mingyan; De Mel, Niluka; Jiao, Yang; Howard, James; Parthemore, Conner; Korman, Samuel; Thompson, Christopher; Wendeler, Michaela; Liu, DengfengmAbs (2019), 11 (6), 1064-1076CODEN: MABSCP; ISSN:1942-0870. (Taylor & Francis, Inc.)Site-specific ADCs are designed to overcome heterogeneity obsd. with first-generation ADCs that use random conjugation to surface-exposed lysine residues or conjugation to interchain disulfide bonds. An elevated level of size variants, such as heavy chain-light chain species, heavy chain-heavy chain-light chain species, and light chain species, is also obsd. with final site-specific ADC product. To understand root cause of heterogeneity generated during ADC conjugation process, we designed time-course studies for each conjugation step, including redn., oxidn., conjugation, and quenching. We developed both non-reduced peptide map and LabChip-based capillary electrophoresis sodium dodecyl sulfate methods for time-course sample anal. On basis of our time-course data, the half ADC and unconjugated antibody were generated during oxidn. as a result of alternative disulfide bond arrangements. During oxidn., two hinge cysteines formed intra-chain disulfide bond in the half ADC, and three inter-chain hinge disulfide bonds were formed in unconjugated antibody. Time-course data also showed that elevated level of size variants, esp. heavy chain-heavy chain-light chain species and light chain species, resulted from quenching step, where quenching reagent engaged in disulfide bond exchange reaction with ADC and broke disulfide bonds connecting heavy chain and light chain. Underconjugated and overconjugated species arose from equil. established during conjugation reaction.
- 26Vollmar, B. S., Wei, B., Ohri, R., Zhou, J., He, J., Yu, S. F., Leipold, D., Cosino, E., Yee, S., Fourie-O’Donohue, A. (2017) Attachment Site Cysteine Thiol pKa Is a Key Driver for Site-Dependent Stability of THIOMAB Antibody-Drug Conjugates. Bioconjugate Chem. 28 (10), 2538– 2548, DOI: 10.1021/acs.bioconjchem.7b0036526https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsVGks7vL&md5=ba1805c493f468d4ffbb198dbf8b3d02Attachment Site Cysteine Thiol pKa Is a Key Driver for Site-Dependent Stability of THIOMAB Antibody-Drug ConjugatesVollmar, Breanna S.; Wei, Binqing; Ohri, Rachana; Zhou, Jianhui; He, Jintang; Yu, Shang-Fan; Leipold, Douglas; Cosino, Ely; Yee, Sharon; Fourie-O'Donohue, Aimee; Li, Guangmin; Phillips, Gail L.; Kozak, Katherine R.; Kamath, Amrita; Xu, Keyang; Lee, Genee; Lazar, Greg A.; Erickson, Hans K.Bioconjugate Chemistry (2017), 28 (10), 2538-2548CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The incorporation of cysteines into antibodies by mutagenesis allows for the direct conjugation of small mols. to specific sites on the antibody via disulfide bonds. The stability of the disulfide bond linkage between the small mol. and the antibody is highly dependent on the location of the engineered cysteine in either the heavy chain (HC) or the light chain (LC) of the antibody. Here, we explore the basis for this site-dependent stability. We evaluated the in vivo efficacy and pharmacokinetics of five different cysteine mutants of trastuzumab conjugated to a pyrrolobenzodiazepine (PBD) via disulfide bonds. A significant correlation was obsd. between disulfide stability and efficacy for the conjugates. We hypothesized that the obsd. site-dependent stability of the disulfide-linked conjugates could be due to differences in the attachment site cysteine thiol pKa. We measured the cysteine thiol pKa using isothermal titrn. calorimetry (ITC) and found that the variants with the highest thiol pKa (LC K149C and HC A140C) were found to yield the conjugates with the greatest in vivo stability. Guided by homol. modeling, we identified several mutations adjacent to LC K149C that reduced the cysteine thiol pKa and, thus, decreased the in vivo stability of the disulfide-linked PBD conjugated to LC K149C. We also present results suggesting that the high thiol pKa of LC K149C is responsible for the sustained circulation stability of LC K149C TDCs utilizing a maleimide-based linker. Taken together, our results provide evidence that the site-dependent stability of cys-engineered antibody-drug conjugates may be explained by interactions between the engineered cysteine and the local protein environment that serves to modulate the side-chain thiol pKa. The influence of cysteine thiol pKa on stability and efficacy offers a new parameter for the optimization of ADCs that utilize cysteine engineering.
- 27Jeffrey, S. C., Burke, P. J., Lyon, R. P., Meyer, D. W., Sussman, D., Anderson, M., Hunter, J. H., Leiske, C. I., Miyamoto, J. B., Nicholas, N. D. (2013) A potent anti-CD70 antibody-drug conjugate combining a dimeric pyrrolobenzodiazepine drug with site-specific conjugation technology. Bioconjugate Chem. 24 (7), 1256– 1263, DOI: 10.1021/bc400217g27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptFKjs7c%253D&md5=31841b0267e41ae861eaf05240c97b15A Potent Anti-CD70 Antibody-Drug Conjugate Combining a Dimeric Pyrrolobenzodiazepine Drug with Site-Specific Conjugation TechnologyJeffrey, Scott C.; Burke, Patrick J.; Lyon, Robert P.; Meyer, David W.; Sussman, Django; Anderson, Martha; Hunter, Joshua H.; Leiske, Chris I.; Miyamoto, Jamie B.; Nicholas, Nicole D.; Okeley, Nicole M.; Sanderson, Russell J.; Stone, Ivan J.; Zeng, Weiping; Gregson, Stephen J.; Masterson, Luke; Tiberghien, Arnaud C.; Howard, Philip W.; Thurston, David E.; Law, Che-Leung; Senter, Peter D.Bioconjugate Chemistry (2013), 24 (7), 1256-1263CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)A highly cytotoxic DNA crosslinking pyrrolobenzodiazepine (PBD) dimer with a valine-alanine dipeptide linker was conjugated to the anti-CD70 h1F6 mAb either through endogenous interchain cysteines or, site-specifically, through engineered cysteines at position 239 of the heavy chains. The h1F6239C-PBD conjugation strategy proved to be superior to interchain cysteine conjugation, affording an antibody-drug conjugate (ADC) with high uniformity in drug-loading and low levels of aggregation. In vitro cytotoxicity expts. demonstrated that the h1F6239C-PBD was potent and immunol. specific on CD70-pos. renal cell carcinoma (RCC) and non-Hodgkin lymphoma (NHL) cell lines. The conjugate was resistant to drug loss in plasma and in circulation, and had a pharmacokinetic profile closely matching that of the parental h1F6239C antibody capped with N-ethylmaleimide (NEM). Evaluation in CD70-pos. RCC and NHL mouse xenograft models showed pronounced antitumor activities at single or weekly doses as low as 0.1 mg/kg of ADC. The ADC was tolerated at 2.5 mg/kg. These results demonstrate that PBDs can be effectively used for antibody-targeted therapy.
- 28Hamblett, K. J., Senter, P. D., Chace, D. F., Sun, M. M. C., Lenox, J., Cerveny, C. G., Kissler, K. M., Bernhardt, S. X., Kopcha, A. K., Zabinski, R. F. (2004) Effects of Drug Loading on the Antitumor Activity of a Monoclonal Antibody Drug Conjugate. Clin. Cancer Res. 10 (20), 7063– 7070, DOI: 10.1158/1078-0432.CCR-04-078928https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXpslGlu78%253D&md5=2d2dd7a498d1181a57dcd3ab8b931e65Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugateHamblett, Kevin J.; Senter, Peter D.; Chace, Dana F.; Sun, Michael M. C.; Lenox, Joel; Cerveny, Charles G.; Kissler, Kim M.; Bernhardt, Starr X.; Kopcha, Anastasia K.; Zabinski, Roger F.; Meyer, Damon L.; Francisco, Joseph A.Clinical Cancer Research (2004), 10 (20), 7063-7070CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)An antibody-drug conjugate consisting of monomethyl auristatin E (MMAE) conjugated to the anti-CD30 monoclonal antibody (mAb) cAC10, with eight drug moieties per mAb, was previously shown to have potent cytotoxic activity against CD30+ malignant cells. To det. the effect of drug loading on antibody-drug conjugate therapeutic potential, we assessed cAC10 antibody-drug conjugates contg. different drug-mAb ratios in vitro and in vivo. Coupling MMAE to the cysteines that comprise the interchain disulfides of cAC10 created an antibody-drug conjugate population, which was purified using hydrophobic interaction chromatog. to yield antibody-drug conjugates with two, four, and eight drugs per antibody (E2, E4, and E8, resp.). Antibody-drug conjugate potency was tested in vitro against CD30+ lines followed by in vivo xenograft models. The max.-tolerated dose and pharmacokinetic profiles of the antibody-drug conjugates were investigated in mice. Although antibody-drug conjugate potency in vitro was directly dependent on drug loading (IC50 values E8<E4<E2), the in vivo antitumor activity of E4 was comparable with E8 at equal mAb doses, although the E4 contained half the amt. of MMAE per mAb. E2 was also an active antitumor agent but required higher doses. The max.-tolerated dose of E2 in mice was at least double that of E4, which in turn was twice that of E8. MMAE loading affected plasma clearance, as E8 cleared 3-fold faster than E4 and 5-fold faster than E2. By decreasing drug loading per antibody, the therapeutic index was increased demonstrating that drug loading is a key design parameter for antibody-drug conjugates.
- 29Sun, X., Ponte, J. F., Yoder, N. C., Laleau, R., Coccia, J., Lanieri, L., Qiu, Q., Wu, R., Hong, E., Bogalhas, M. (2017) Effects of Drug-Antibody Ratio on Pharmacokinetics, Biodistribution, Efficacy, and Tolerability of Antibody-Maytansinoid Conjugates. Bioconjugate Chem. 28 (5), 1371– 1381, DOI: 10.1021/acs.bioconjchem.7b0006229https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlslCjurg%253D&md5=60e21df4b6e3189d87a43b0d650f51b4Effects of Drug-Antibody Ratio on Pharmacokinetics, Biodistribution, Efficacy, and Tolerability of Antibody-Maytansinoid ConjugatesSun, Xiuxia; Ponte, Jose F.; Yoder, Nicholas C.; Laleau, Rassol; Coccia, Jennifer; Lanieri, Leanne; Qiu, Qifeng; Wu, Rui; Hong, Erica; Bogalhas, Megan; Wang, Lintao; Dong, Ling; Setiady, Yulius; Maloney, Erin K.; Ab, Olga; Zhang, Xiaoyan; Pinkas, Jan; Keating, Thomas A.; Chari, Ravi; Erickson, Hans K.; Lambert, John M.Bioconjugate Chemistry (2017), 28 (5), 1371-1381CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Antibody-drug conjugates (ADCs) are being actively pursued as a treatment option for cancer following the regulatory approval of brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla). ADCs consist of a cytotoxic agent conjugated to a targeting antibody through a linker. The two approved ADCs (and most ADCs now in the clinic that use a microtubule disrupting agent as the payload) are heterogeneous conjugates with an av. drug to antibody ratio (DAR) of 3-4 (potentially ranging from 0-8 for individual species). Ado-trastuzumab emtansine employs DM1, a semisynthetic cytotoxic payload of the maytansinoid class, which is conjugated via lysine residues of the antibody to an av. DAR of 3.5. To understand the effect of DAR on the preclin. properties of ADCs using maytansinoid cytotoxic agents, we prepd. a series of conjugates with a cleavable linker (M9346A-sulfo-SPDB-DM4 targeting folate receptor alpha (FR)) or an uncleavable linker (J2898A-SMCC-DM1 targeting the epidermal growth factor receptor (EGFR)) with varying DAR, and evaluated their biochem. characteristics, in vivo stability, efficacy, and tolerability. For both formats, a series of ADCs with DARs ranging from low (av. ∼2, range 0-4) to very high (av. 10, range 7-14) were prepd. in good yield with high monomer content and low levels of free cytotoxic agent. The in vitro potency consistently increased with increasing DAR, at const. antibody concn. We then characterized the in vivo disposition of these ADCs. Pharmacokinetic anal. showed that conjugates with an av. DAR below ∼6 had comparable clearance rates, but for those with an av. DAR around 9-10, rapid clearance was obsd. Biodistribution studies in mice showed that these 9-10 DAR ADCs rapidly accumulate in the liver, with max. localization for this organ at 24-28% percentage injected dose per g (%ID/g) compared with 7-10% for lower DAR conjugates (all at 2-6 h post-injection). Our preclin. findings on tolerability and efficacy suggest that maytansinoid conjugates with DAR ranging from 2-6 have a better therapeutic index than conjugates with very high DAR (∼9-10). These very high DAR ADCs suffer from decreased efficacy likely due to faster clearance. These results support the use of DAR 3-4 for maytansinoid ADCs, but suggest the exploration of lower or higher DAR may be warranted depending on the biol. of the target antigen.
- 30Thompson, P., Fleming, R., Bezabeh, B., Huang, F., Mao, S., Chen, C., Harper, J., Zhong, H., Gao, X., Yu, X.-Q. (2016) Rational design, biophysical and biological characterization of site-specific antibody-tubulysin conjugates with improved stability, efficacy and pharmacokinetics. J. Controlled Release 236, 100– 116, DOI: 10.1016/j.jconrel.2016.06.02530https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtFehtrbF&md5=61154de48ed3a8ab3586e2815ae99459Rational design, biophysical and biological characterization of site-specific antibody-tubulysin conjugates with improved stability, efficacy and pharmacokineticsThompson, Pamela; Fleming, Ryan; Bezabeh, Binyam; Huang, Fengying; Mao, Shenlan; Chen, Cui; Harper, Jay; Zhong, Haihong; Gao, Xizhe; Yu, Xiang-Qing; Hinrichs, Mary Jane; Reed, Molly; Kamal, Adeela; Strout, Patrick; Cho, Song; Woods, Rob; Hollingsworth, Robert E.; Dixit, Rakesh; Wu, Herren; Gao, Changshou; Dimasi, NazzarenoJournal of Controlled Release (2016), 236 (), 100-116CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)Antibody-drug conjugates (ADCs) are among the most promising empowered biologics for cancer treatment. ADCs are commonly prepd. by chem. conjugation of small mol. cytotoxic anti-cancer drugs to antibodies through either lysine side chains or cysteine thiols generated by the redn. of interchain disulfide bonds. Both methods yield heterogeneous conjugates with complex biophys. properties and suboptimal serum stability, efficacy, and pharmacokinetics. To limit the complexity of cysteine-based ADCs, we have engineered and characterized in vitro and in vivo antibody cysteine variants that allow precise control of both site of conjugation and drug load per antibody mol. We demonstrate that the chem.-defined cysteine-engineered antibody-tubulysin conjugates have improved ex vivo and in vivo stability, efficacy, and pharmacokinetics when compared to conventional cysteine-based ADCs with similar drug-to-antibody ratios. In addn., to limit the non-target FcγRs mediated uptake of the ADCs by cells of the innate immune system, which may result in off-target toxicities, the ADCs have been engineered to lack Fc-receptor binding. The strategies described herein are broadly applicable to any full-length IgG or Fc-based ADC and have been incorporated into an ADC that is in phase I clin. development.
- 31Ouyang, J. (2013) Drug-to-antibody ratio (DAR) and drug load distribution by hydrophobic interaction chromatography and reversed phase high-performance liquid chromatography. Methods Mol. Biol. (N. Y., NY, U. S.) 1045, 275– 283, DOI: 10.1007/978-1-62703-541-5_1731https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3sfms1Oqtg%253D%253D&md5=933437e336d82a976a50610aa79b4d0bDrug-to-antibody ratio (DAR) and drug load distribution by hydrophobic interaction chromatography and reversed phase high-performance liquid chromatographyOuyang JunMethods in molecular biology (Clifton, N.J.) (2013), 1045 (), 275-83 ISSN:.Hydrophobic interaction chromatography (HIC) is the method of choice for determination of the drug-to-antibody ratio (DAR) and drug load distribution for cysteine (Cys)-linked antibody-drug conjugates (ADCs). The drug-loaded species are resolved based on the increasing hydrophobicity with the least hydrophobic, unconjugated form eluting first and the most hydrophobic, 8-drug form eluting last. The area percentage of a peak represents the relative distribution of the particular drug-loaded ADC species. The weighted average DAR is then calculated using the percentage peak area information and the drug load numbers. Reversed phase high-performance liquid chromatography (RP-HPLC) offers an orthogonal method to obtain DAR for Cys-linked ADCs. The method involves, first, a reduction reaction to completely dissociate the heavy and light chains of the ADC, then separation of the light and heavy chains and their corresponding drug-loaded forms on an RP column. The percentage peak area from integration of the light chain and heavy chain peaks, combined with the assigned drug load for each peak, is used to calculate the weighted average DAR.
- 32Fekete, S., Veuthey, J. L., Beck, A., and Guillarme, D. (2016) Hydrophobic interaction chromatography for the characterization of monoclonal antibodies and related products. J. Pharm. Biomed. Anal. 130, 3– 18, DOI: 10.1016/j.jpba.2016.04.00432https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmtVSgtb0%253D&md5=2e13c1de1d790e013013ee462660f69eHydrophobic interaction chromatography for the characterization of monoclonal antibodies and related productsFekete, Szabolcs; Veuthey, Jean-Luc; Beck, Alain; Guillarme, DavyJournal of Pharmaceutical and Biomedical Analysis (2016), 130 (), 3-18CODEN: JPBADA; ISSN:0731-7085. (Elsevier B.V.)A review. Hydrophobic interaction chromatog. (HIC) is a historical strategy used for the anal. purifn. and characterization of proteins. Similarly to what can be done in reversed-phase liq. chromatog. (RPLC), HIC is able to sep. protein species based on their hydrophobicity, but using different conditions. Compared to RPLC, the main benefit of HIC is its ability to perform sepns. under non denaturing conditions (i.e. physiol. pH conditions, ambient mobile phase temp. and no need for org. solvents) and so an orthogonal method. The goal of this review is to provide a general overview of theor. and practical aspects of modern HIC applied for the characterization of therapeutic protein biopharmaceuticals including monoclonal antibodies (mAbs), antibody drug conjugates (ADCs) and bispecific antibodies (bsAbs). Therefore, method development approaches, state-of-the-art column technol., applications and future perspectives are described and critically discussed.
- 33Strop, P., Liu, S. H., Dorywalska, M., Delaria, K., Dushin, R. G., Tran, T. T., Ho, W. H., Farias, S., Casas, M. G., Abdiche, Y. (2013) Location matters: site of conjugation modulates stability and pharmacokinetics of antibody drug conjugates. Chem. Biol. 20 (2), 161– 167, DOI: 10.1016/j.chembiol.2013.01.01033https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXjtFWht74%253D&md5=0b419848b5fd4dfa4863a29de0555cf4Location Matters: Site of Conjugation Modulates Stability and Pharmacokinetics of Antibody Drug ConjugatesStrop, Pavel; Liu, Shu-Hui; Dorywalska, Magdalena; Delaria, Kathy; Dushin, Russell G.; Tran, Thomas-Toan; Ho, Wei-Hsien; Farias, Santiago; Casas, Meritxell Galindo; Abdiche, Yasmina; Zhou, Dahui; Chandrasekaran, Ramalakshmi; Samain, Caroline; Loo, Carole; Rossi, Andrea; Rickert, Mathias; Krimm, Stellanie; Wong, Teresa; Chin, Sherman Michael; Yu, Jessica; Dilley, Jeanette; Chaparro-Riggers, Javier; Filzen, Gary F.; O'Donnell, Christopher J.; Wang, Fang; Myers, Jeremy S.; Pons, Jaume; Shelton, David L.; Rajpal, ArvindChemistry & Biology (Oxford, United Kingdom) (2013), 20 (2), 161-167CODEN: CBOLE2; ISSN:1074-5521. (Elsevier Ltd.)Antibody drug conjugates (ADCs) are a therapeutic class offering promise for cancer therapy. The attachment of cytotoxic drugs to antibodies can result in an effective therapy with better safety potential than nontargeted cytotoxics. To understand the role of conjugation site, we developed an enzymic method for site-specific antibody drug conjugation using microbial transglutaminase. This allowed us to attach diverse compds. at multiple positions and investigate how the site influences stability, toxicity, and efficacy. We show that the conjugation site has significant impact on ADC stability and pharmacokinetics in a species-dependent manner. These differences can be directly attributed to the position of the linkage rather than the chem. instability, as was obsd. with a maleimide linkage. With this method, it is possible to produce homogeneous ADCs and tune their properties to maximize the therapeutic window.
- 34Lyon, R. P., Bovee, T. D., Doronina, S. O., Burke, P. J., Hunter, J. H., Neff-LaFord, H. D., Jonas, M., Anderson, M. E., Setter, J. R., and Senter, P. D. (2015) Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic index. Nat. Biotechnol. 33 (7), 733– 735, DOI: 10.1038/nbt.321234https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFaitrfF&md5=cccafa4e97ac463525fd2c898bb62f27Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic indexLyon, Robert P.; Bovee, Tim D.; Doronina, Svetlana O.; Burke, Patrick J.; Hunter, Joshua H.; Neff-LaFord, Haley D.; Jonas, Mechthild; Anderson, Martha E.; Setter, Jocelyn R.; Senter, Peter D.Nature Biotechnology (2015), 33 (7), 733-735CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)The in vitro potency of antibody-drug conjugates (ADCs) increases with the drug-to-antibody ratio (DAR); however, ADC plasma clearance also increases with DAR, reducing exposure and in vivo efficacy. Here we show that accelerated clearance arises from ADC hydrophobicity, which can be modulated through drug-linker design. We exemplify this using hydrophilic auristatin drug linkers and PEGylated ADCs that yield uniform, high-DAR ADCs with superior in vivo performance.
- 35Tumey, L. N., Li, F., Rago, B., Han, X., Loganzo, F., Musto, S., Graziani, E. I., Puthenveetil, S., Casavant, J., Marquette, K. (2017) Site Selection: a Case Study in the Identification of Optimal Cysteine Engineered Antibody Drug Conjugates. AAPS J. 19, 1– 13, DOI: 10.1208/s12248-017-0083-7There is no corresponding record for this reference.
- 36Burke, P. J., Hamilton, J. Z., Jeffrey, S. C., Hunter, J. H., Doronina, S. O., Okeley, N. M., Miyamoto, J. B., Anderson, M. E., Stone, I. J., Ulrich, M. L. (2017) Optimization of a PEGylated Glucuronide-Monomethylauristatin E Linker for Antibody-Drug Conjugates. Mol. Cancer Ther. 16 (1), 116– 123, DOI: 10.1158/1535-7163.MCT-16-034336https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjsF2hug%253D%253D&md5=94f2bf9dba796252569277f466e9be33Optimization of a PEGylated Glucuronide-Monomethylauristatin E Linker for Antibody-Drug ConjugatesBurke, Patrick J.; Hamilton, Joseph Z.; Jeffrey, Scott C.; Hunter, Joshua H.; Doronina, Svetlana O.; Okeley, Nicole M.; Miyamoto, Jamie B.; Anderson, Martha E.; Stone, Ivan J.; Ulrich, Michelle L.; Simmons, Jessica K.; McKinney, Erica E.; Senter, Peter D.; Lyon, Robert P.Molecular Cancer Therapeutics (2017), 16 (1), 116-123CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)The emergence of antibody-drug conjugates (ADC), such as brentuximab vedotin and ado-trastuzumab emtansine, has led to increased efforts to identify new payloads and develop improved drug-linker technologies. Most antibody payloads impart significant hydrophobicity to the ADC, resulting in accelerated plasma clearance and suboptimal in vivo activity, particularly for conjugates with high drug-to-antibody ratios (DAR). We recently reported on the incorporation of a discrete PEG24 polymer as a side chain in a β-glucuronidase-cleavable monomethylauristatin E (MMAE) linker to provide homogeneous DAR 8 conjugates with decreased plasma clearance and increased antitumor activity in xenograft models relative to a non-PEGylated control. In this work, we optimized the drug-linker by minimizing the size of the PEG side chain and incorporating a self-stabilizing maleimide to prevent payload de-conjugation in vivo. Multiple PEG-glucuronide-MMAE linkers were prepd. with PEG size up to 24 ethylene oxide units, and homogeneous DAR 8 ADCs were evaluated. A clear relationship was obsd. between PEG length and conjugate pharmacol. when tested in vivo. Longer PEG chains resulted in slower clearance, with a threshold length of PEG8 beyond which clearance was not impacted. Conjugates bearing PEG of sufficient length to minimize plasma clearance provided a wider therapeutic window relative to faster clearing conjugates bearing shorter PEGs. A lead PEGylated glucuronide-MMAE linker was identified incorporating a self-stabilizing maleimide and a PEG12 side chain emerged from these efforts, enabling highly potent, homogeneous DAR 8 conjugates and is under consideration for future ADC programs.
- 37Lyon, R. P., Meyer, D. L., Setter, J. R., and Senter, P. D. (2012) Conjugation of anticancer drugs through endogenous monoclonal antibody cysteine residues. Methods Enzymol. 502, 123– 138, DOI: 10.1016/B978-0-12-416039-2.00006-937https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xkt1Cjtbw%253D&md5=32b9da481a24d156f347a2cd9faf3a20Conjugation of anticancer drugs through endogenous monoclonal antibody cysteine residuesLyon, Robert P.; Meyer, David L.; Setter, Jocelyn R.; Senter, Peter D.Methods in Enzymology (2012), 502 (Protein Engineering for Therapeutics, Part A), 123-138CODEN: MENZAU; ISSN:0076-6879. (Elsevier Inc.)A review. Many methods have been described for the conjugation of drugs to monoclonal antibodies. The presence of a discrete no. of readily reducible disulfides in the common IgG subtypes presents a convenient opportunity for conjugation to cysteine residues with thiol-reactive drug-linkers. Such conjugates can be prepd. by a straightforward two-step reaction scheme involving the redn. of the antibody disulfides to the desired no. of av. thiols per antibody, followed by addn. of the drug-linker, ideally with a maleimido functionality for rapid, selective reaction. In a discovery setting, this basic method can be scaled down to produce microgram quantities of conjugate for early screening, and in a manufg. setting can be scaled up to produce grams or kilograms of conjugate for clin. trials and commercialization. The resulting conjugates are readily characterized using common HPLC methods.
- 38Bhakta, S., Raab, H., and Junutula, J. R. (2013) Engineering THIOMABs for site-specific conjugation of thiol-reactive linkers. Methods Mol. Biol. 1045, 189– 203, DOI: 10.1007/978-1-62703-541-5_1138https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3sfms1Orug%253D%253D&md5=83bb25f39770bd0156d24ac0e97fe4caEngineering THIOMABs for site-specific conjugation of thiol-reactive linkersBhakta Sunil; Raab Helga; Junutula Jagath RMethods in molecular biology (Clifton, N.J.) (2013), 1045 (), 189-203 ISSN:.Antibody conjugates are used in many therapeutic and research applications and are generated by chemically linking a cysteine or lysine residue to potent chemotherapeutic drugs or other functional groups through a flexible linker. Recently, we have engineered THIOMABs (antibodies with engineered reactive cysteine residues) for site-specific conjugation and showed that these antibody conjugates display homogeneous labeling with optimal in vitro and in vivo characteristics. Here, we describe protocols for engineering, selection, and site-specific conjugation of THIOMABs with thiol-reactive linkers.
- 39Garcia-Echeverria, C. (2014) Developing second generation antibody-drug conjugates: the quest for new technologies. J. Med. Chem. 57 (19), 7888– 7889, DOI: 10.1021/jm501298k39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsFCjtr7N&md5=681e6c579a64daca1509fb950a1a386dDeveloping Second Generation Antibody-Drug Conjugates: The Quest for New TechnologiesGarcia-Echeverria, CarlosJournal of Medicinal Chemistry (2014), 57 (19), 7888-7889CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. The field of antibody-drug conjugates (ADCs) has gained significant momentum after the recent regulatory approval of two ADCs, and significant research efforts are directed to identify more effective payloads and simplify current manufg. challenges.
- 40Drake, P. M., Albers, A. E., Baker, J., Banas, S., Barfield, R. M., Bhat, A. S., de Hart, G. W., Garofalo, A. W., Holder, P., Jones, L. C. (2014) Aldehyde Tag Coupled with HIPS Chemistry Enables the Production of ADCs Conjugated Site-Specifically to Different Antibody Regions with Distinct in Vivo Efficacy and PK Outcomes. Bioconjugate Chem. 25 (7), 1331– 1341, DOI: 10.1021/bc500189z40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXps1Knurs%253D&md5=fb08b4277ea6d941861f1cca293b1385Aldehyde Tag Coupled with HIPS Chemistry Enables the Production of ADCs Conjugated Site-Specifically to Different Antibody Regions with Distinct in Vivo Efficacy and PK OutcomesDrake, Penelope M.; Albers, Aaron E.; Baker, Jeanne; Banas, Stefanie; Barfield, Robyn M.; Bhat, Abhijit S.; de Hart, Gregory W.; Garofalo, Albert W.; Holder, Patrick; Jones, Lesley C.; Kudirka, Romas; McFarland, Jesse; Zmolek, Wes; Rabuka, DavidBioconjugate Chemistry (2014), 25 (7), 1331-1341CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)It is becoming increasingly clear that site-specific conjugation offers significant advantages over conventional conjugation chemistries used to make antibody-drug conjugates (ADCs). Site-specific payload placement allows for control over both the drug-to-antibody ratio (DAR) and the conjugation site, both of which play an important role in governing the pharmacokinetics (PK), disposition, and efficacy of the ADC. In addn. to the DAR and site of conjugation, linker compn. also plays an important role in the properties of an ADC. The authors have previously reported a novel site-specific conjugation platform comprising linker payloads designed to selectively react with site-specifically engineered aldehyde tags on an antibody backbone. This chem. results in a stable C-C bond between the antibody and the cytotoxin payload, providing a uniquely stable connection with respect to the other linker chemistries used to generate ADCs. The flexibility and versatility of the aldehyde tag conjugation platform has enabled the authors to undertake a systematic evaluation of the impact of conjugation site and linker compn. on ADC properties. Here, the authors describe the prodn. and characterization of a panel of ADCs bearing the aldehyde tag at different locations on an IgG1 backbone conjugated using Hydrazino-iso-Pictet-Spengler (HIPS) chem. The authors demonstrate that in a panel of ADCs with aldehyde tags at different locations, the site of conjugation has a dramatic impact on in vivo efficacy and pharmacokinetic behavior in rodents; this advantage translates to an improved safety profile in rats as compared to a conventional lysine conjugate.
- 41Catcott, K. C., McShea, M. A., Bialucha, C. U., Miller, K. L., Hicks, S. W., Saxena, P., Gesner, T. G., Woldegiorgis, M., Lewis, M. E., Bai, C. (2016) Microscale screening of antibody libraries as maytansinoid antibody-drug conjugates. MAbs 8 (3), 513– 523, DOI: 10.1080/19420862.2015.113440841https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XislSmtLs%253D&md5=f353875078f05b2b0e43ca26a6d61908Microscale screening of antibody libraries as maytansinoid antibody-drug conjugatesCatcott, Kalli C.; McShea, Molly A.; Bialucha, Carl Uli; Miller, Kathy L.; Hicks, Stuart W.; Saxena, Parmita; Gesner, Thomas G.; Woldegiorgis, Mikias; Lewis, Megan E.; Bai, Chen; Fleming, Michael S.; Ettenberg, Seth A.; Erickson, Hans K.; Yoder, Nicholas C.mAbs (2016), 8 (3), 513-523CODEN: MABSCP; ISSN:1942-0870. (Taylor & Francis, Inc.)Antibody-drug conjugates (ADCs) are of great interest as targeted cancer therapeutics. Prepn. of ADCs for early stage screening is constrained by purifn. and biochem. anal. techniques that necessitate burdensome quantities of antibody. Here we describe a method, developed for the maytansinoid class of ADCs, enabling parallel conjugation of antibodies in 96-well format. The method utilizes ∼100 μg of antibody per well and requires <5 μg of ADC for characterization. We demonstrate the capabilities of this system using model antibodies. We also provide multiple examples applying this method to early-stage screening of maytansinoid ADCs. The method can greatly increase the throughput with which candidate ADCs can be screened in cell-based assays, and may be more generally applicable to high-throughput prepn. and screening of different types of protein conjugates.
- 42Cilliers, C., Guo, H., Liao, J., Christodolu, N., and Thurber, G. M. (2016) Multiscale Modeling of Antibody-Drug Conjugates: Connecting Tissue and Cellular Distribution to Whole Animal Pharmacokinetics and Potential Implications for Efficacy. AAPS J. 18 (5), 1117– 1130, DOI: 10.1208/s12248-016-9940-z42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XpvV2nsbY%253D&md5=f7df584c11f8a3126ee369933a2f237bMultiscale Modeling of Antibody-Drug Conjugates: Connecting Tissue and Cellular Distribution to Whole Animal Pharmacokinetics and Potential Implications for EfficacyCilliers, Cornelius; Guo, Hans; Liao, Jianshan; Christodolu, Nikolas; Thurber, Greg M.AAPS Journal (2016), 18 (5), 1117-1130CODEN: AJAOB6; ISSN:1550-7416. (Springer)Antibody-drug conjugates exhibit complex pharmacokinetics due to their combination of macromol. and small mol. properties. These issues range from systemic concerns, such as deconjugation of the small mol. drug during the long antibody circulation time or rapid clearance from nonspecific interactions, to local tumor tissue heterogeneity, cell bystander effects, and endosomal escape. Math. models can be used to study the impact of these processes on overall distribution in an efficient manner, and several types of models have been used to analyze varying aspects of antibody distribution including physiol. based pharmacokinetic (PBPK) models and tissue-level simulations. However, these processes are quant. in nature and cannot be handled qual. in isolation. For example, free antibody from deconjugation of the small mol. will impact the distribution of conjugated antibodies within the tumor. To incorporate these effects into a unified framework, we have coupled the systemic and organ-level distribution of a PBPK model with the tissue-level detail of a distributed parameter tumor model. We used this math. model to analyze new exptl. results on the distribution of the clin. antibody-drug conjugate Kadcyla in HER2-pos. mouse xenografts. This model is able to capture the impact of the drug-antibody ratio (DAR) on tumor penetration, the net result of drug deconjugation, and the effect of using unconjugated antibody to drive ADC penetration deeper into the tumor tissue. This modeling approach will provide quant. and mechanistic support to exptl. studies trying to parse the impact of multiple mechanisms of action for these complex drugs.
- 43Bumbaca, D., Boswell, C. A., Fielder, P. J., and Khawli, L. A. (2012) Physiochemical and biochemical factors influencing the pharmacokinetics of antibody therapeutics. AAPS J. 14 (3), 554– 558, DOI: 10.1208/s12248-012-9369-y43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XptFWit70%253D&md5=acaabca0a83de3a589e334024a5d6f00Physiochemical and Biochemical Factors Influencing the Pharmacokinetics of Antibody TherapeuticsBumbaca, Daniela; Boswell, C. Andrew; Fielder, Paul J.; Khawli, Leslie A.AAPS Journal (2012), 14 (3), 554-558CODEN: AJAOB6; ISSN:1550-7416. (Springer)A review. Monoclonal antibodies are increasingly being developed to treat multiple disease areas, including those related to oncol., immunol., neurol., and ophthalmol. There are multiple factors, such as charge, size, neonatal Fc receptor (FcRn) binding affinity, target affinity and biol., IgG (IgG) subclass, degree and type of glycosylation, injection route, and injection site, that could affect the pharmacokinetics (PK) of these large macromol. therapeutics, which in turn could have ramifications on their efficacy and safety. This mini-review examines how characteristics of the antibodies could be altered to change their PK profiles. For example, it was obsd. that a net charge modification of at least a 1-unit shift in isoelec. point altered antibody clearance. Antibodies with enhanced affinity for FcRn at pH 6.0 display longer serum half-lives and slower clearances than wild type. Antibody fragments have different clearance rates and tissue distribution profiles than full length antibodies. Fc glycosylation is perceived to have a minimal effect on PK while that of terminal high mannose remains unclear. More investigation is warranted to det. if injection route and/or site impacts PK. Nonetheless, a better understanding of the effects of all these variations may allow for the better design of antibody therapeutics.
- 44Bohnert, T. and Gan, L. S. (2013) Plasma protein binding: from discovery to development. J. Pharm. Sci. 102 (9), 2953– 2994, DOI: 10.1002/jps.2361444https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXpvVymt7c%253D&md5=d676f7490c6e0f09c8acf7cce5183b17Plasma protein binding: From discovery to developmentBohnert, Tonika; Gan, Liang-ShangJournal of Pharmaceutical Sciences (2013), 102 (9), 2953-2994CODEN: JPMSAE; ISSN:0022-3549. (John Wiley & Sons, Inc.)A review. The importance of plasma protein binding (PPB) in modulating the effective drug concn. at pharmacol. target sites has been the topic of significant discussion and debate amongst drug development groups over the past few decades. Free drug theory, which states that in absence of energy-dependent processes, after steady state equil. has been attained, free drug concn. in plasma is equal to free drug concn. at the pharmacol. target receptor(s) in tissues, has been used to explain pharmacokinetics/pharmacodynamics relationships in a large no. of cases. Any sudden increase in free concn. of a drug could potentially cause toxicity and may need dose adjustment. Free drug concn. is also helpful to est. the effective concn. of drugs that potentially can ppt. metab. (or transporter)-related drug-drug interactions. Disease models are extensively validated in animals to progress a compd. into development. Unbound drug concn., and therefore PPB information across species is very informative in establishing safety margins and guiding selection of First in Human (FIH) dose and human efficacious dose. The scope of this review is to give an overview of reported role of PPB in several therapeutic areas, highlight cases where PPB changes are clin. relevant, and provide drug metab. and pharmacokinetics recommendations in discovery and development settings. © 2013 Wiley Periodicals, Inc. and the American Pharmacists Assocn. J Pharm Sci.
- 45Alley, S. C., Benjamin, D. R., Jeffrey, S. C., Okeley, N. M., Meyer, D. L., Sanderson, R. J., and Senter, P. D. (2008) Contribution of linker stability to the activities of anticancer immunoconjugates. Bioconjugate Chem. 19 (3), 759– 765, DOI: 10.1021/bc700432945https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXislemurk%253D&md5=62abdec871dda99c160a9b989c12f6f0Contribution of Linker Stability to the Activities of Anticancer ImmunoconjugatesAlley, Stephen C.; Benjamin, Dennis R.; Jeffrey, Scott C.; Okeley, Nicole M.; Meyer, Damon L.; Sanderson, Russell J.; Senter, Peter D.Bioconjugate Chemistry (2008), 19 (3), 759-765CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The linker component of antibody-drug conjugates (ADC) is a key feature in developing optimized therapeutic agents that are highly active at well tolerated doses. For maximal intratumoral drug delivery, linkers are required that are highly stable in the systemic circulation, yet allow for efficient drug release at the target site. In this respect, amide bond-based technologies constitute a technol. advancement, since the linker half-lives in circulation (t1/2 ∼ 7 days) are much longer than earlier generation linkers that break down within 1-2 days. The amide linkers, some of which contain peptides, are appended to the mAb carriers through thioether/maleimide adducts. Here, we describe that use of a bromoacetamidecaproyl (bac) in place of the maleimidocaproyl (mc) increases the plasma stability of resulting thioether ADCs. One such ADC, 1F6-C4v2-bac-MMAF, which is directed against the CD70 antigen on lymphomas and renal cell carcinoma, was prepd. contg. a bac thioether spacer between the drug (MMAF) and the mAb carrier (1F6-C4v2). There was no measurable systemic drug release from this ADC for 2 wk postadministration in mice. In order to assess the impact of improving linker stability beyond mc contg. ADCs, a series of mc and bac-linked 1F6-MMAF conjugates were compared for tolerability, intratumoral drug delivery, and therapeutic efficacy in nude mice with renal cell carcinoma xenografts. There were no statistically significant efficacy differences between sets of mc and bac contg. ADCs, although the bac linker technol. led to 25% higher intratumoral drug exposure over a 7 day period compared to the corresponding mc linker. The mechanism of drug release from maleimide-adducts likely involves a retro-Michael reaction that takes place in plasma, based on in vitro studies demonstrating that some of the released drug-maleimide deriv. became covalently bound to cysteine-34 of serum albumin. In summary, the data indicate that new linkers can be obtained with improved in vivo stability by replacing the maleimide with an acetamide, but the resulting ADCs had similar tolerability and activity profiles.
- 46Ohri, R., Bhakta, S., Fourie-O’Donohue, A., Dela Cruz-Chuh, J., Tsai, S. P., Cook, R., Wei, B., Ng, C., Wong, A. W., Bos, A. B. (2018) High-Throughput Cysteine Scanning To Identify Stable Antibody Conjugation Sites for Maleimide- and Disulfide-Based Linkers. Bioconjugate Chem. 29 (2), 473– 485, DOI: 10.1021/acs.bioconjchem.7b0079146https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisVKku7g%253D&md5=318146c1baea8094b671e99b5c429627High-Throughput Cysteine Scanning To Identify Stable Antibody Conjugation Sites for Maleimide- and Disulfide-Based LinkersOhri, Rachana; Bhakta, Sunil; Fourie-O'Donohue, Aimee; dela Cruz-Chuh, Josefa; Tsai, Siao Ping; Cook, Ryan; Wei, Binqing; Ng, Carl; Wong, Athena W.; Bos, Aaron B.; Farahi, Farzam; Bhakta, Jiten; Pillow, Thomas H.; Raab, Helga; Vandlen, Richard; Polakis, Paul; Liu, Yichin; Erickson, Hans; Junutula, Jagath R.; Kozak, Katherine R.Bioconjugate Chemistry (2018), 29 (2), 473-485CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)THIOMAB antibody technol. utilizes cysteine residues engineered onto an antibody to allow for site-specific conjugation. The technol. has enabled the exploration of different attachment sites on the antibody in combination with small mols., peptides, or proteins to yield antibody conjugates with unique properties. As reported previously, the specific location of the site of conjugation on an antibody can impact the stability of the linkage to the engineered cysteine for both thio-succinimide and disulfide bonds. High stability of the linkage is usually desired to maximize the delivery of the cargo to the intended target. In the current study, cysteines were individually substituted into every position of the anti-HER2 antibody (trastuzumab), and the stabilities of drug conjugations at those sites were evaluated. We screened a total of 648 THIOMAB antibody-drug conjugates, each generated from a trastuzamab prepd. by sequentially mutating non-cysteine amino acids in the light and heavy chains to cysteine. Each THIOMAB antibody variant was conjugated to either maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl-monomethyl auristatin E (MC-vc-PAB-MMAE) or pyridyl disulfide monomethyl auristatin E (PDS-MMAE) using a high-throughput, on-bead conjugation and purifn. method. Greater than 50% of the THIOMAB antibody variants were successfully conjugated to both MMAE derivs. with a drug to antibody ratio (DAR) of >0.5 and <50% aggregation. The relative in vitro plasma stabilities for approx. 750 conjugates were assessed using enzyme-linked immunosorbent assays, and stable sites were confirmed with affinity-capture LC/MS-based detection methods. Highly stable conjugation sites for the two types of MMAE derivs. were identified on both the heavy and light chains. Although the stabilities of maleimide conjugates were shown to be greater than those of the disulfide conjugates, many sites were identified that were stable for both. Furthermore, in vitro stabilities of selected stable sites translated across different cytotoxic payloads and different target antibodies as well as to in vivo stability.
- 47Lyon, R. (2018) Drawing lessons from the clinical development of antibody-drug conjugates. Drug Discovery Today: Technol. 30, 105– 109, DOI: 10.1016/j.ddtec.2018.10.00147https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cnisVSlsA%253D%253D&md5=dfdb2af91ce423b8912dc33f9187b35fDrawing lessons from the clinical development of antibody-drug conjugatesLyon RobertDrug discovery today. Technologies (2018), 30 (), 105-109 ISSN:.The antibody-drug conjugate (ADC) field has seen a remarkable expansion in the number of entrants in clinical studies. Many of these agents employ newer conjugation technologies that have been developed over the last decade that confer various attributes to the ADCs prepared with them, including stability, potency, and homogeneity. In many cases, these new ADCs appear demonstrably superior to earlier technologies in preclinical models of activity and toxicology, but the degree to which these improvements will translate to the clinic is only starting to be seen. Many of these technologies are now competing head-to-head by targeting the same antigen in similar patient populations, allowing for a direct comparison of their clinical performance properties. As lessons from these experiences feed back into discovery research, future iterations of ADC design may be expected to bring improved therapeutics into the clinic.
- 48Khera, E., Cilliers, C. M., Bhatnagar, S., and Thurber, G. M. (2018) Computation transport analysis of antibody-drug conjugate bystander effects and payload tumoral distribution: implications for therapy. Mol. Syst. Des. Eng. 3, 73– 88, DOI: 10.1039/C7ME00093F48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1yjs73J&md5=3b39670c741923617c57a9ec3e715806Computational transport analysis of antibody-drug conjugate bystander effects and payload tumoral distribution: implications for therapyKhera, Eshita; Cilliers, Cornelius; Bhatnagar, Sumit; Thurber, Greg M.Molecular Systems Design & Engineering (2018), 3 (1), 73-88CODEN: MSDEBG; ISSN:2058-9689. (Royal Society of Chemistry)Antibody drug conjugates (ADCs) have a proven clin. record with four FDA approved drugs and dozens more in clin. trials. However, a better understanding of the relationship between delivery and efficacy of ADCs is needed to improve the rate of successful clin. development. Recent evidence indicates that heterogeneous distribution can play a large role in the efficacy of these drugs. However, the impact of the drug payload, particularly the ability of the payload to diffuse outside of the original targeted cell into adjacent cells (the bystander effect), is not completely understood. Given the challenges in directly measuring the payload distribution within tumors, we developed a predictive computational model to study payload distribution as a function of antibody dose, payload dose, and payload properties. The computational results indicate that: 1. the heterogeneous tumoral distribution of ADCs impacts efficacy, and increasing the antibody dose improves penetration and efficacy. 2. The increased penetration of payloads with bystander effects can partially compensate for poor antibody penetration, but larger antibody doses still result in further improvement. This occurs because of the higher efficiency of direct cell killing than bystander killing. 3. Bystander effects are important for killing antigen neg. cells, and an optimum in physicochem. properties exists. Payloads with a balance in cellular uptake vs. tissue diffusion enter cells fast enough to avoid tumor washout but slow enough to reach distant cells. Therefore, optimizing the antibody dose, payload dose, and payload physicochem. properties results in ideal delivery to the site of action and max. efficacy.
- 49Cilliers, C., Menezes, B., Nessler, I., Linderman, J., and Thurber, G. M. (2018) Improved Tumor Penetration and Single-Cell Targeting of Antibody-Drug Conjugates Increases Anticancer Efficacy and Host Survival. Cancer Res. 78 (3), 758– 768, DOI: 10.1158/0008-5472.CAN-17-163849https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitVWntr8%253D&md5=9c10da29fd94ccad470329f2dcc48566Improved Tumor Penetration and Single-Cell Targeting of Antibody-Drug Conjugates Increases Anticancer Efficacy and Host SurvivalCilliers, Cornelius; Menezes, Bruna; Nessler, Ian; Linderman, Jennifer; Thurber, Greg M.Cancer Research (2018), 78 (3), 758-768CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Current ADC have made advances in engineering the antibody, linker, conjugation site, small-mol. payload, and drug-to-antibody ratio (DAR). However, the relationship between heterogeneous intratumoral distribution and efficacy of ADCs is poorly understood. Here, we compared trastuzumab and ado-trastuzumab emtansine (T-DM1) to study the impact of ADC tumor distribution on efficacy. In a mouse xenograft model insensitive to trastuzumab, coadministration of trastuzumab with a fixed dose of T-DM1 at 3:1 and 8:1 ratios dramatically improved ADC tumor penetration and resulted in twice the improvement in median survival compared with T-DM1 alone. In this setting, the effective DAR was lowered, decreasing the amt. of payload delivered to each targeted cell but increasing the no. of cells that received payload. This result is counterintuitive because trastuzumab acts as an antagonist in vitro and has no single-agent efficacy in vivo, yet improves the effectiveness of T-DM1 in vivo. Novel dual-channel fluorescence ratios quantified single-cell ADC uptake and metab. and confirmed that the in vivo cellular dose of T-DM1 alone exceeded the min. required for efficacy in this model. In addn., this technique characterized cellular pharmacokinetics with heterogeneous delivery after 1 day, degrdn. and payload release by 2 days, and in vitro cell killing and in vivo tumor shrinkage 2 to 3 days later. This work demonstrates that the intratumoral distribution of ADC, independent of payload dose or plasma clearance, plays a major role in ADC efficacy. This study shows how lowering the drug-to-antibody ratio during treatment can improve the intratumoral distribution of a antibody-drug conjugate, with implications for improving the efficacy of this class of cancer drugs.
- 50Sahota, S. and Vahdat, L. T. (2017) Sacituzumab govitecan: an antibody-drug conjugate. Expert Opin. Biol. Ther. 17 (8), 1027– 1031, DOI: 10.1080/14712598.2017.133121450https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXosVags7s%253D&md5=e2a9aeb979d52eca095bd455aa73667bSacituzumab govitecan: an antibody-drug conjugateSahota, Sheena; Vahdat, Linda T.Expert Opinion on Biological Therapy (2017), 17 (8), 1027-1031CODEN: EOBTA2; ISSN:1471-2598. (Taylor & Francis Ltd.): Despite advances in the diagnosis and treatment of patients with cancer, patients with metastatic cancer have limited therapeutic options after initial lines of therapy. Understanding tumor biol. has translated into the identification of actionable targets that resulted in therapeutics. Antibody-drug conjugates (ADC) are capitalizing on this explosion of scientific information. ADCs allow an antibody to a unique target to be conjugated via an innovative linker, to a highly toxic drug which is delivered to its target. Sacituzumab govitecan is an ADC that combines the active mol. in irinotecan, SN-38, to an antibody targeting trop2.: In this review, the authors introduce the reader to the ADC sacituzumab govitecan providing the reader with details about its pharmacokinetics, pharmacodynamics, efficacy and safety. The authors also give their expert anal. about its potential future use.: Sacituzumab govitecan is a novel and well-tolerated therapeutic showing promising results in difficult to treat cancers. Further studies are underway to optimize the group of patients that would benefit from it. Given its excellent performance, we are cautiously optimistic it will be approved by the FDA.
- 51Nakada, T., Sugihara, K., Jikoh, T., Abe, Y., and Agatsuma, T. (2019) The Latest Research and Development into the Antibody-Drug Conjugate, [fam-] Trastuzumab Deruxtecan (DS-8201a), for HER2 Cancer Therapy. Chem. Pharm. Bull. 67 (3), 173– 185, DOI: 10.1248/cpb.c18-0074451https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhsVejurjJ&md5=ab7628fdc4af3c8e1d73e00553c9534eThe latest research and development into the antibody-drug conjugate, [fam-] trastuzumab deruxtecan (DS-8201a), for HER2 cancer therapyNakada, Takashi; Sugihara, Kiyoshi; Jikoh, Takahiro; Abe, Yuki; Agatsuma, ToshinoriChemical & Pharmaceutical Bulletin (2019), 67 (3), 173-185CODEN: CPBTAL; ISSN:0009-2363. (Pharmaceutical Society of Japan)A review. A major limitation of traditional chemotherapy for cancer is dose-limiting toxicity, caused by the exposure of non-tumor cells to cytotoxic agents. Use of mol. targeted drugs, such as specific kinase inhibitors and monoclonal antibodies, is a possible soln. to overcome this limitation and has achieved clin. success so far. Use of an antibody-drug conjugate (ADC) is a rational strategy for improving efficacy and reducing systemic adverse events. ADCs use antibodies selectively to deliver a potent cytotoxic agent to tumor cells, thus drastically improving the therapeutic index of chemotherapeutic agents. Lessons learned from clin. failure of early ADCs during the 1980s to 90s have recently led to improvements in ADC technol., and resulted in the approval of four novel ADCs. Nonetheless, further advances in ADC technol. are still required to streamline their clin. efficacy and reduce toxicity. [fam-] Trastuzumab deruxtecan (DS-8201a) is a next-generation ADC that satisfies these requirements based on currently available evidence. DS-8201a has several innovative features; a highly potent novel payload with a high drug-to-antibody ratio, good homogeneity, a tumor-selective cleavable linker, stable linker-payload in circulation, and a short systemic half-life cytotoxic agent in vivo; the released cytotoxic payload could exert a bystander effect. With respect to its preclin. profiles, DS-8201a could provide a valuable therapy with a great potential against HER2-expressing cancers in clin. settings. In a phase I trial, DS-8201a showed acceptable safety profiles with potential therapeutic efficacy, with the wide therapeutic index.
- 52Reid, E. E., Archer, K. E., Shizuka, M., Wilhelm, A., Yoder, N. C., Bai, C., Fishkin, N. E., Harris, L., Maloney, E. K., E, H. (2019) Effect of linker stereochemistry on the antitumor activity of anti-body-drug conjugates (ADCs) containing indolinobenzodiazepine payloads. ACS Med. Chem. Lett. 10 (8), 1193– 1197, DOI: 10.1021/acsmedchemlett.9b0024052https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlGjs7fP&md5=72540be91ff42af39669c697c9b0f162Effect of Linker Stereochemistry on the Activity of Indolinobenzodiazepine Containing Antibody-Drug Conjugates (ADCs)Reid, Emily E.; Archer, Katie E.; Shizuka, Manami; Wilhelm, Alan; Yoder, Nicholas C.; Bai, Chen; Fishkin, Nathan E.; Harris, Luke; Maloney, Erin K.; Salomon, Paulin; Hong, Erica; Wu, Rui; Ab, Olga; Jin, Shan; Lai, Katharine C.; Sikka, Surina; Chari, Ravi V. J.; Miller, Michael L.ACS Medicinal Chemistry Letters (2019), 10 (8), 1193-1197CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Antibody-drug conjugates (ADCs) that incorporate potent indolinobenzodiazepine DNA alkylators as the payload component are currently undergoing clin. evaluation. In one ADC design, the payload mols. are linked to the antibody through a peptidase-labile L-Ala-L-Ala linker. In order to det. the role of amino acid stereochem. on antitumor activity and tolerability, we incorporated L- and D-alanyl groups in the dipeptide, synthesized all four diastereomers, and prepd. and tested the corresponding ADCs. Results of our preclin. evaluation showed that the L-Ala-L-Ala configuration provided the ADC with the highest therapeutic index (antitumor activity vs toxicity).
- 53Lazar, A. C., Wang, L., Blättler, W. A., Amphlett, G., Lambert, J. M., and Zhang, W. (2005) Analysis of the composition of immunoconjugates using size-exclusion chromatography coupled to mass spectrometry. Rapid Commun. Mass Spectrom. 19 (13), 1806– 1814, DOI: 10.1002/rcm.198753https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXmtVegt70%253D&md5=678eee93e086e099fa72f4d2d79e6646Analysis of the composition of immunoconjugates using size-exclusion chromatography coupled to mass spectrometryLazar, Alexandru C.; Wang, Lintao; Blaettler, Walter A.; Amphlett, Godfrey; Lambert, John M.; Zhang, WeiRapid Communications in Mass Spectrometry (2005), 19 (13), 1806-1814CODEN: RCMSEF; ISSN:0951-4198. (John Wiley & Sons Ltd.)Recombinant monoclonal antibody drug products play an increasingly important role in the treatment of various diseases. Antibodies are large, multi-chain proteins and antibody prepns. often contain several mol. variants, which renders them heterogeneous. The heterogeneity is further increased in immunoconjugates prepd. by covalently linking several drug mols. per antibody mol. As part of the product characterization, the mol. wts. of the antibodies or their drug conjugates need to be measured. Electrospray ionization mass spectrometry (ESI-MS) is well suited for the anal. of recombinant antibodies and immunoconjugates. Sample prepn. is an important element of ESI-MS anal., in particular samples need to be freed of interfering charged species, such as salts and buffer components. In this paper, Amicon centrifugal filters, reversed-phase high-performance liq. chromatog. (HPLC), and size-exclusion HPLC were evaluated for sample desalting. Size-exclusion HPLC, using aq. acetonitrile as the mobile phase, directly coupled to ESI-MS provided the best performance and was optimized for the study of immunoconjugates. The results showed that antibodies carrying covalently linked maytansinoid mols. generated charge envelope profiles that differ from those of the non-conjugated antibody. For the detn. of the distribution of the various conjugate species in an immunoconjugate sample prepd. by randomly linking in the av. 3.6 drug mols. per antibody mol., the exptl. conditions needed to be carefully selected to allow acquisition of the whole spectrum contg. the charge envelopes of all species.
- 54Kovtun, Y. V., Audette, C. A., Mayo, M. F., Jones, G. E., Doherty, H., Maloney, E. K., Erickson, H. K., Sun, X., Wilhelm, S., Ab, O. (2010) Antibody-maytansinoid conjugates designed to bypass multidrug resistance. Cancer Res. 70 (6), 2528– 2537, DOI: 10.1158/0008-5472.CAN-09-354654https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjtFygur4%253D&md5=17a2600cc85203b3bace74e31d346daaAntibody-Maytansinoid Conjugates Designed to Bypass Multidrug ResistanceKovtun, Yelena V.; Audette, Charlene A.; Mayo, Michele F.; Jones, Gregory E.; Doherty, Heather; Maloney, Erin K.; Erickson, Hans K.; Sun, Xiuxia; Wilhelm, Sharon; Ab, Olga; Lai, Katharine C.; Widdison, Wayne C.; Kellogg, Brenda; Johnson, Holly; Pinkas, Jan; Lutz, Robert J.; Singh, Rajeeva; Goldmacher, Victor S.; Chari, Ravi V. J.Cancer Research (2010), 70 (6), 2528-2537CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Conjugation of cytotoxic compds. to antibodies that bind to cancer-specific antigens makes these drugs selective in killing cancer cells. However, many of the compds. used in such antibody-drug conjugates (ADC) are substrates for the multidrug transporter MDR1. To evade the MDR1-mediated resistance, we conjugated the highly cytotoxic maytansinoid DM1 to antibodies via the maleimidyl-based hydrophilic linker PEG4Mal. Following uptake into target cells, conjugates made with the PEG4Mal linker were processed to a cytotoxic metabolite that was retained by MDR1-expressing cells better than a metabolite of similar conjugates prepd. with the nonpolar linker N-succinimidyl-4-(maleimidomethyl)cyclohexane-1-carboxylate (SMCC). In accord, PEG4Mal-linked conjugates were more potent in killing MDR1-expressing cells in culture. In addn., PEG4Mal-linked conjugates were markedly more effective in eradicating MDR1-expressing human xenograft tumors than SMCC-linked conjugates while being tolerated similarly, thus showing an improved therapeutic index. This study points the way to the development of ADCs that bypass multidrug resistance. Cancer Res; 70(6); 2528-37.
- 55Ab, O., Whiteman, K. R., Bartle, L. M., Sun, X., Singh, R., Tavares, D., LaBelle, A., Payne, G., Lutz, R. J., Pinkas, J. (2015) IMGN853, a Folate Receptor-α (FRα)-Targeting Antibody-Drug Conjugate, Exhibits Potent Targeted Antitumor Activity against FRα-Expressing Tumors. Mol. Cancer Ther. 14 (7), 1605– 1613, DOI: 10.1158/1535-7163.MCT-14-109555https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFejtLzO&md5=194d678451d25646466be718537d3980IMGN853, a Folate Receptor-α (FRα)-Targeting Antibody-Drug Conjugate, Exhibits Potent Targeted Antitumor Activity against FRα-Expressing TumorsAb, Olga; Whiteman, Kathleen R.; Bartle, Laura M.; Sun, Xiuxia; Singh, Rajeeva; Tavares, Daniel; LaBelle, Alyssa; Payne, Gillian; Lutz, Robert J.; Pinkas, Jan; Goldmacher, Victor S.; Chittenden, Thomas; Lambert, John M.Molecular Cancer Therapeutics (2015), 14 (7), 1605-1613CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)A majority of ovarian and non-small cell lung adenocarcinoma cancers overexpress folate receptor α (FRα). Here, we report the development of an anti-FRα antibody-drug conjugate (ADC), consisting of a FRα-binding antibody attached to a highly potent maytansinoid that induces cell-cycle arrest and cell death by targeting microtubules. From screening a large panel of anti-FRα monoclonal antibodies, we selected the humanized antibody M9346A as the best antibody for targeted delivery of a maytansinoid payload into FRα-pos. cells. We compared M9346A conjugates with various linker/maytansinoid combinations, and found that a conjugate, now denoted as IMGN853, with the N-succinimidyl 4-(2-pyridyldithio)-2-sulfobutanoate (sulfo-SPDB) linker and N2'-deacetyl-N2'-(4-mercapto-4-methyl-1-oxopentyl)-maytansine (DM4) exhibited the most potent antitumor activity in several FRα-expressing xenograft tumor models. The level of expression of FRα on the surface of cells was a major determinant in the sensitivity of tumor cells to the cytotoxic effect of the conjugate. Efficacy studies of IMGN853 in xenografts of ovarian cancer and non-small cell lung cancer cell lines and of a patient tumor-derived xenograft model demonstrated that the ADC was highly active against tumors that expressed FRα at levels similar to those found on a large fraction of ovarian and non-small cell lung cancer patient tumors, as assessed by immunohistochem. IMGN853 displayed cytotoxic activity against FRα-neg. cells situated near FRα-pos. cells (bystander cytotoxic activity), indicating its ability to eradicate tumors with heterogeneous expression of FRα. Together, these findings support the clin. development of IMGN853 as a novel targeted therapy for patients with FRα-expressing tumors. Mol Cancer Ther; 14(7); 1605-13. ©2015 AACR.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.bioconjchem.9b00777.
Detailed experimental procedures for synthesis of compound 1b, supplementary characterization data for ADCs, and supplementary methods (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.