1
|
Ito K, Tatsumi T, Takahashi K, Shimizu Y, Yamatsugu K, Kanai M. A Stable and Cleavable O-Linked Spacer for Drug Delivery Systems. Chem Pharm Bull (Tokyo) 2020; 68:212-215. [PMID: 31189762 DOI: 10.1248/cpb.c19-00376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Anti-cancer chemotherapy with good efficacy and fewer side effects is highly desirable. A drug delivery system comprising a cancer-targeting module and a cytotoxic agent connected with a cleavable linker is promising for reducing side effects. The development of a cleavable linker satisfying the requirements of both stability and cleavability, however, is difficult, especially when a carbonate moiety is used for conjugating the linker to a hydroxy group in a drug of interest. We herein report a new stable linker comprising carbamate and ester spacers, which can be introduced on a hydroxy group of a drug. This linker is more stable in aqueous neutral buffer than a corresponding carbonate-type linker, and releases a payload anti-cancer drug, SN-38, through a two-step sequence upon cathepsin B treatment. This linker may have potential use in other drug delivery systems to lower side effects by selectively transporting cytotoxic drugs to tumor cells.
Collapse
Affiliation(s)
- Kei Ito
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | | | - Kazuki Takahashi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Yohei Shimizu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Kenzo Yamatsugu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Motomu Kanai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| |
Collapse
|
2
|
Sengee M, Eksteen JJ, Nergård SL, Vasskog T, Sydnes LK. Preparation and Assessment of Self-Immolative Linkers for Therapeutic Bioconjugates with Amino- and Hydroxyl-Containing Cargoes. Bioconjug Chem 2019; 30:1489-1499. [DOI: 10.1021/acs.bioconjchem.9b00214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Myagmarsuren Sengee
- NORCE Norwegian Research Centre AS, P.O. Box
6434, Tromsø Science Park, NO-9294 Tromsø, Norway
| | - J. Johannes Eksteen
- NORCE Norwegian Research Centre AS, P.O. Box
6434, Tromsø Science Park, NO-9294 Tromsø, Norway
| | - Silje Lillemark Nergård
- Department of Pharmacy, UiT The Arctic University of Norway, P.O. Box 6050, Langnes, NO-9037 Tromsø, Norway
| | - Terje Vasskog
- NORCE Norwegian Research Centre AS, P.O. Box
6434, Tromsø Science Park, NO-9294 Tromsø, Norway
- Department of Pharmacy, UiT The Arctic University of Norway, P.O. Box 6050, Langnes, NO-9037 Tromsø, Norway
| | - Leiv K. Sydnes
- NORCE Norwegian Research Centre AS, P.O. Box
6434, Tromsø Science Park, NO-9294 Tromsø, Norway
- Department of Chemistry, University of Bergen, P.O. Box 7800, NO-5020 Bergen, Norway
| |
Collapse
|
3
|
Lau UY, Benoit LT, Stevens NS, Emmerton KK, Zaval M, Cochran JH, Senter PD. Lactone Stabilization is Not a Necessary Feature for Antibody Conjugates of Camptothecins. Mol Pharm 2018; 15:4063-4072. [DOI: 10.1021/acs.molpharmaceut.8b00477] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Uland Y. Lau
- Seattle Genetics, Inc, 21823 30th Drive SE, Bothell, Washington 98021, United States
| | - Lauren T. Benoit
- Seattle Genetics, Inc, 21823 30th Drive SE, Bothell, Washington 98021, United States
| | - Nicole S. Stevens
- Seattle Genetics, Inc, 21823 30th Drive SE, Bothell, Washington 98021, United States
| | - Kim K. Emmerton
- Seattle Genetics, Inc, 21823 30th Drive SE, Bothell, Washington 98021, United States
| | - Margo Zaval
- Seattle Genetics, Inc, 21823 30th Drive SE, Bothell, Washington 98021, United States
| | - Julia H. Cochran
- Seattle Genetics, Inc, 21823 30th Drive SE, Bothell, Washington 98021, United States
| | - Peter D. Senter
- Seattle Genetics, Inc, 21823 30th Drive SE, Bothell, Washington 98021, United States
| |
Collapse
|
4
|
Glucuronide-Linked Antibody–Tubulysin Conjugates Display Activity in MDR+ and Heterogeneous Tumor Models. Mol Cancer Ther 2018; 17:1752-1760. [DOI: 10.1158/1535-7163.mct-18-0073] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/06/2018] [Accepted: 05/18/2018] [Indexed: 11/16/2022]
|
5
|
Ogitani Y, Abe Y, Iguchi T, Yamaguchi J, Terauchi T, Kitamura M, Goto K, Goto M, Oitate M, Yukinaga H, Yabe Y, Nakada T, Masuda T, Morita K, Agatsuma T. Wide application of a novel topoisomerase I inhibitor-based drug conjugation technology. Bioorg Med Chem Lett 2016; 26:5069-5072. [PMID: 27599744 DOI: 10.1016/j.bmcl.2016.08.082] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/09/2016] [Accepted: 08/26/2016] [Indexed: 11/26/2022]
Abstract
To establish a novel and widely applicable payload-linker technology for antibody-drug conjugates (ADCs), we have focused our research on applying exatecan mesylate (DX-8951f), a potent topoisomerase I inhibitor, which exhibits extensive antitumor activity as well as significant myelotoxicity, as the payload part. Through this study, we discovered a promising exatecan derivative (DX-8951 derivative, DXd), that has the characteristics of low membrane permeability and shows considerably less myelotoxicity than that shown by exatecan mesylate in an in vitro human colony forming unit-granulocyte macrophage assay. DXd was further used for drug conjugation by using commercially or clinically useful monoclonal antibodies to evaluate the potency of the ADC. The result revealed that the DXd-ADCs targeting CD30, CD33, and CD70 were effective against each of their respective target-expressing tumor cell lines. Moreover, a novel DXd-ADC targeting B7-H3, which is a new target for ADCs, also showed potent antitumor efficacy both in vitro and in vivo. In conclusion, this study showed that this novel topoisomerase I inhibitor-based ADC technology is widely applicable to a diverse number of antibodies and is expected to mitigate myelotoxicity, thereby possibly resulting in better safety profiles than that of existing ADC technologies.
Collapse
Affiliation(s)
- Yusuke Ogitani
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yuki Abe
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Takuma Iguchi
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Junko Yamaguchi
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Tomoko Terauchi
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Michiko Kitamura
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Koichi Goto
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Mayumi Goto
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Masataka Oitate
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Hideo Yukinaga
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yoshiyuki Yabe
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Takashi Nakada
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Takeshi Masuda
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Koji Morita
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Toshinori Agatsuma
- Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| |
Collapse
|
6
|
Govindan SV, Griffiths GL, Hansen HJ, Horak ID, Goldenberg DM. Cancer Therapy with Radiolabeled and Drug/Toxin-conjugated Antibodies. Technol Cancer Res Treat 2016; 4:375-91. [PMID: 16029057 DOI: 10.1177/153303460500400406] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Radioimmunotherapy and antibody-directed chemotherapy have emerged as cancer treatment modalities with the regulatory approval of products for non-Hodgkin's lymphoma and acute myeloid leukemia. Antibody-toxin therapy is likewise on the verge of clinical fruition. Accumulating evidence suggests that radioimmunotherapy may have the best impact in minimal-disease and adjuvant settings, especially with radioresistant solid tumors. For the latter, ongoing efforts in ‘pretargeting’ to increase deliverable tumor radiation dose, combination therapies, and locoregional applications are also of importance. Antibody-drug conjugates have the potential to increase the therapeutic index of chemotherapy by minimizing systemic toxicity and improving tumor targeting. The design of optimal drug conjugates in this regard is predicated upon the proper choice of the target antigen, the cleavable-linker, and the drug. In respect of antibody-toxin conjugates, considerable progress has been made in chemical and recombinant immunotoxin designs, and in the advancement of many products to clinical trials. Continued development of antibody-directed therapies should expand the options available for the management of cancer.
Collapse
|
7
|
Kolakowski RV, Haelsig KT, Emmerton KK, Leiske CI, Miyamoto JB, Cochran JH, Lyon RP, Senter PD, Jeffrey SC. The Methylene Alkoxy Carbamate Self-Immolative Unit: Utilization for the Targeted Delivery of Alcohol-Containing Payloads with Antibody-Drug Conjugates. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201601506] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
8
|
Kolakowski RV, Haelsig KT, Emmerton KK, Leiske CI, Miyamoto JB, Cochran JH, Lyon RP, Senter PD, Jeffrey SC. The Methylene Alkoxy Carbamate Self-Immolative Unit: Utilization for the Targeted Delivery of Alcohol-Containing Payloads with Antibody-Drug Conjugates. Angew Chem Int Ed Engl 2016; 55:7948-51. [PMID: 27198854 DOI: 10.1002/anie.201601506] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/04/2016] [Indexed: 11/09/2022]
Abstract
A strategy for the conjugation of alcohol-containing payloads to antibodies has been developed and involves the methylene alkoxy carbamate (MAC) self-immolative unit. A series of MAC β-glucuronide model constructs were prepared to evaluate stability and enzymatic release, and the results demonstrated high stability at physiological pH in a substitution-dependent manner. All the MAC model compounds efficiently released alcohol drug surrogates under the action of β-glucuronidase. To assess the MAC technology for ADCs, the potent microtubule-disrupting agent auristatin E (AE) was incorporated through the norephedrine alcohol. Conjugation of the MAC β-glucuronide AE drug linker to the anti-CD30 antibody cAC10, and an IgG control antibody, gave potent and immunologically specific activities in vitro and in vivo. These studies validate the MAC self-immolative unit for alcohol-containing payloads within ADCs, a class that has not been widely exploited.
Collapse
Affiliation(s)
| | - Karl T Haelsig
- Seattle Genetics, 21823 30th Dr SE, Bothell, WA, 98021, USA
| | - Kim K Emmerton
- Seattle Genetics, 21823 30th Dr SE, Bothell, WA, 98021, USA
| | - Chris I Leiske
- Seattle Genetics, 21823 30th Dr SE, Bothell, WA, 98021, USA
| | | | | | - Robert P Lyon
- Seattle Genetics, 21823 30th Dr SE, Bothell, WA, 98021, USA
| | - Peter D Senter
- Seattle Genetics, 21823 30th Dr SE, Bothell, WA, 98021, USA
| | - Scott C Jeffrey
- Seattle Genetics, 21823 30th Dr SE, Bothell, WA, 98021, USA.
| |
Collapse
|
9
|
Yao H, Jiang F, Lu A, Zhang G. Methods to Design and Synthesize Antibody-Drug Conjugates (ADCs). Int J Mol Sci 2016; 17:E194. [PMID: 26848651 PMCID: PMC4783928 DOI: 10.3390/ijms17020194] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 01/28/2016] [Accepted: 01/28/2016] [Indexed: 11/16/2022] Open
Abstract
Antibody-drug conjugates (ADCs) have become a promising targeted therapy strategy that combines the specificity, favorable pharmacokinetics and biodistributions of antibodies with the destructive potential of highly potent drugs. One of the biggest challenges in the development of ADCs is the application of suitable linkers for conjugating drugs to antibodies. Recently, the design and synthesis of linkers are making great progress. In this review, we present the methods that are currently used to synthesize antibody-drug conjugates by using thiols, amines, alcohols, aldehydes and azides.
Collapse
Affiliation(s)
- Houzong Yao
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Feng Jiang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Faculty of Materials Science and Chemical Engineering, the State Key Laboratory Base of Novel Functional Materials and Preparation Science, Ningbo University, Ningbo 315211, Zhejiang, China.
| | - Aiping Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Ge Zhang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
10
|
Akash MSH, Rehman K, Parveen A, Ibrahim M. Antibody-drug conjugates as drug carrier systems for bioactive agents. INT J POLYM MATER PO 2015. [DOI: 10.1080/00914037.2015.1038818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
11
|
Abstract
Antibody-drug conjugates (ADCs), which combine the specificity, favorable pharmacokinetics, and biodistribution of a monoclonal antibody (mAb) with the cytotoxic potency of a drug, are promising new therapies for cancer. Along with the development of monoclonal antibodies (mAbs) and cytotoxic drugs, the design of the linker is of essential importance, because it impacts the efficacy and tolerability of ADCs. The linker needs to provide sufficient stability during systemic circulation but allow for the rapid and efficient release of the cytotoxic drug in an active form inside the tumor cells. This review provides an overview of linker technologies currently used for ADCs and advances that have resulted in linkers with improved properties. Also provided is a brief summary of some considerations for the conjugation of antibody and drug linker such as drug-to-antibody ratio and site of conjugation.
Collapse
Affiliation(s)
- Birte Nolting
- Biotherapeutics Research and Development, Pfizer, Pearl River, NY, USA
| |
Collapse
|
12
|
Drake PM, Rabuka D. Antibody-Drug Conjugates: Can Coupling Cytotoxicity and Specificity Overcome Therapeutic Resistance? RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2013. [DOI: 10.1007/978-1-4614-7654-2_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
13
|
Redy O, Shabat D. Modular theranostic prodrug based on a FRET-activated self-immolative linker. J Control Release 2012; 164:276-82. [DOI: 10.1016/j.jconrel.2012.05.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 05/02/2012] [Accepted: 05/03/2012] [Indexed: 11/24/2022]
|
14
|
Zhu L, Cheng P, Lei N, Yao J, Sheng C, Zhuang C, Guo W, Liu W, Zhang Y, Dong G, Wang S, Miao Z, Zhang W. Synthesis and Biological Evaluation of Novel Homocamptothecins Conjugating with Dihydropyrimidine Derivatives as Potent Topoisomerase I Inhibitors. Arch Pharm (Weinheim) 2011; 344:726-34. [DOI: 10.1002/ardp.201000402] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 02/28/2011] [Accepted: 03/02/2011] [Indexed: 11/07/2022]
|
15
|
Challenges in developing bioanalytical assays for characterization of antibody–drug conjugates. Bioanalysis 2011; 3:677-700. [DOI: 10.4155/bio.11.30] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
With more than 34 targets being investigated and nearly 20 clinical trials at various phases of development, antibody–drug conjugates (ADCs) hold a lot of promise for improving oncological malignancy therapy. This therapeutic strategy designed to specifically or preferentially deliver a cytotoxic agent to tumor cells through conjugation to a monoclonal antibody is not new. Although this approach is relatively simple conceptually, the history of ADCs clearly attests to the high degree of complexity in their development. Each component of an ADC is important to achieve efficacy with minimal toxicity, and the ability to monitor this multicomponent therapeutic entity is deemed to be critical for their successful optimization. In this article we review the different bioanalytical strategies that have been implemented to characterize various ADCs and discuss the challenges and issues associated with these approaches.
Collapse
|
16
|
Weinstain R, Segal E, Satchi-Fainaro R, Shabat D. Real-time monitoring of drug release. Chem Commun (Camb) 2010; 46:553-5. [DOI: 10.1039/b919329d] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
17
|
Burke PJ, Senter PD, Meyer DW, Miyamoto JB, Anderson M, Toki BE, Manikumar G, Wani MC, Kroll DJ, Jeffrey SC. Design, Synthesis, and Biological Evaluation of Antibody−Drug Conjugates Comprised of Potent Camptothecin Analogues. Bioconjug Chem 2009; 20:1242-50. [DOI: 10.1021/bc9001097] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Patrick J. Burke
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021, Research Triangle Institute, Natural Products Laboratory, Research Triangle Park, North Carolina 27709, and North Carolina Central University, Pharmaceutical Sciences, BRITE, Durham, North Carolina 27707
| | - Peter D. Senter
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021, Research Triangle Institute, Natural Products Laboratory, Research Triangle Park, North Carolina 27709, and North Carolina Central University, Pharmaceutical Sciences, BRITE, Durham, North Carolina 27707
| | - David W. Meyer
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021, Research Triangle Institute, Natural Products Laboratory, Research Triangle Park, North Carolina 27709, and North Carolina Central University, Pharmaceutical Sciences, BRITE, Durham, North Carolina 27707
| | - Jamie B. Miyamoto
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021, Research Triangle Institute, Natural Products Laboratory, Research Triangle Park, North Carolina 27709, and North Carolina Central University, Pharmaceutical Sciences, BRITE, Durham, North Carolina 27707
| | - Martha Anderson
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021, Research Triangle Institute, Natural Products Laboratory, Research Triangle Park, North Carolina 27709, and North Carolina Central University, Pharmaceutical Sciences, BRITE, Durham, North Carolina 27707
| | - Brian E. Toki
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021, Research Triangle Institute, Natural Products Laboratory, Research Triangle Park, North Carolina 27709, and North Carolina Central University, Pharmaceutical Sciences, BRITE, Durham, North Carolina 27707
| | - Govindarajan Manikumar
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021, Research Triangle Institute, Natural Products Laboratory, Research Triangle Park, North Carolina 27709, and North Carolina Central University, Pharmaceutical Sciences, BRITE, Durham, North Carolina 27707
| | - Mansukh C. Wani
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021, Research Triangle Institute, Natural Products Laboratory, Research Triangle Park, North Carolina 27709, and North Carolina Central University, Pharmaceutical Sciences, BRITE, Durham, North Carolina 27707
| | - David J. Kroll
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021, Research Triangle Institute, Natural Products Laboratory, Research Triangle Park, North Carolina 27709, and North Carolina Central University, Pharmaceutical Sciences, BRITE, Durham, North Carolina 27707
| | - Scott C. Jeffrey
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021, Research Triangle Institute, Natural Products Laboratory, Research Triangle Park, North Carolina 27709, and North Carolina Central University, Pharmaceutical Sciences, BRITE, Durham, North Carolina 27707
| |
Collapse
|
18
|
Senter PD. Potent antibody drug conjugates for cancer therapy. Curr Opin Chem Biol 2009; 13:235-44. [PMID: 19414278 DOI: 10.1016/j.cbpa.2009.03.023] [Citation(s) in RCA: 273] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Accepted: 03/30/2009] [Indexed: 11/19/2022]
Abstract
Significant progress has been made in the past few years in the area of antibody drug conjugates (ADCs) for the selective delivery of cytotoxic drugs to tumors. Early work in this field incorporated clinically approved drugs and mouse monoclonal antibodies (mAbs), which had modest activities, and were generally immunogenic. The results of these studies prompted investigation that led to the identity of several key parameters that influenced activity and tolerability. These included the antigen target, the use of non-immunogenic mAb carriers, the incorporation of highly potent drugs and novel conditionally stable linker technologies, and the specific methods used to attach drugs to mAbs. As a result of these investigations, new agents with pronounced clinical activities have been developed. These include SGN-35, an ADC directed against the CD30-positive malignancies such as Hodgkin's disease and anaplastic large cell lymphoma, and trastuzumab-DM1 which has shown activity in metastatic breast carcinoma. This review details many of the technological advancements, and provides examples of promising ADCs that are currently in clinical trials.
Collapse
Affiliation(s)
- Peter D Senter
- Seattle Genetics, Inc., 21823 30th Dr. SE, Bothell, WA 98021, United States.
| |
Collapse
|
19
|
Abu Ajaj K, Graeser R, Fichtner I, Kratz F. In vitro and in vivo study of an albumin-binding prodrug of doxorubicin that is cleaved by cathepsin B. Cancer Chemother Pharmacol 2009; 64:413-8. [DOI: 10.1007/s00280-009-0942-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Accepted: 01/16/2009] [Indexed: 10/21/2022]
|
20
|
Moon SJ, Govindan SV, Cardillo TM, D'Souza CA, Hansen HJ, Goldenberg DM. Antibody conjugates of 7-ethyl-10-hydroxycamptothecin (SN-38) for targeted cancer chemotherapy. J Med Chem 2008; 51:6916-26. [PMID: 18939816 DOI: 10.1021/jm800719t] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
CPT-11 is a clinically used cancer drug, and it is a prodrug of the potent topoisomerase I inhibitor, SN-38 (7-ethyl-10-hydroxycamptothecin). To bypass the need for the in vivo conversion of CPT-11 and increase the therapeutic index, bifunctional derivatives of SN-38 were prepared for use in antibody-based targeted therapy of cancer. The general synthetic scheme incorporated an acetylene-azide click cycloaddition step in the design, a short polyethylene glycol spacer for aqueous solubility, and a maleimide group for conjugation. Conjugates of a humanized anti-CEACAM5 monoclonal antibody, hMN-14, prepared using these SN-38 derivatives were evaluated in vitro for stability in buffer and human serum and for antigen-binding and cytotoxicity in a human colon adenocarcinoma cell line. Conjugates of hMN-14 and SN-38 derivatives 16 and 17 were found promising for further development.
Collapse
Affiliation(s)
- Sung-Ju Moon
- Immunomedics, Inc., Morris Plains, New Jersey 07950, USA
| | | | | | | | | | | |
Collapse
|
21
|
|
22
|
Jeffrey SC, Nguyen MT, Moser RF, Meyer DL, Miyamoto JB, Senter PD. Minor groove binder antibody conjugates employing a water soluble beta-glucuronide linker. Bioorg Med Chem Lett 2007; 17:2278-80. [PMID: 17293111 DOI: 10.1016/j.bmcl.2007.01.071] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Revised: 01/16/2007] [Accepted: 01/18/2007] [Indexed: 11/21/2022]
Abstract
The minor groove binder beta-glucuronide drug-linker 3 was constructed from amino CBI 1 and determined to be a substrate for Escherichia coli beta-glucuronidase (EC 3.2.1.31), resulting in facile drug release. Compound 3 was conjugated to mAbs cAC10 (anti-CD30) and h1F6 (anti-CD70) to give antibody-drug conjugates (ADCs) with potencies comparable to that of free drug 1. The ADCs were largely monomeric at intermediate loading levels (4-5drug/mAb), in contrast to highly aggregated p-aminobenzylcarbamate dipeptide-based ADCs of 1 previously reported. Significant levels of immunologic specificity were observed with cAC10-3 by comparing antigen positive versus negative cell lines and binding versus non-binding control ADCs. The water soluble beta-glucuronide linker is stable in plasma and effectively delivers drugs to target cells leading to potent cytotoxic activities.
Collapse
Affiliation(s)
- Scott C Jeffrey
- Seattle Genetics Inc., 21823 30th Drive S.E., Bothell, WA 98021, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Henne WA, Doorneweerd DD, Hilgenbrink AR, Kularatne SA, Low PS. Synthesis and activity of a folate peptide camptothecin prodrug. Bioorg Med Chem Lett 2006; 16:5350-5. [PMID: 16901694 DOI: 10.1016/j.bmcl.2006.07.076] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Accepted: 07/24/2006] [Indexed: 10/24/2022]
Abstract
A folate receptor targeted camptothecin prodrug was synthesized using a hydrophilic peptide spacer linked to folate via a releasable disulfide carbonate linker. The conjugate was found to possess high affinity for folate receptor-expressing cells and inhibited cell proliferation in human KB cells with an IC(50) of 10nM. Activity of the prodrug was completely blocked by excess folic acid, demonstrating receptor-mediated uptake.
Collapse
Affiliation(s)
- Walter A Henne
- Department of Chemistry and Purdue Cancer Center, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | | | |
Collapse
|
24
|
Le Sann C. Maleimide spacers as versatile linkers in the synthesis of bioconjugates of anthracyclines. Nat Prod Rep 2006; 23:357-67. [PMID: 16741584 DOI: 10.1039/b600666n] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Christine Le Sann
- School of Chemistry and Chemical Engineering, David Keir Building, Queen's University Belfast, Northern Ireland.
| |
Collapse
|
25
|
|
26
|
Sriram D, Yogeeswari P, Thirumurugan R, Bal TR. Camptothecin and its analogues: a review on their chemotherapeutic potential. Nat Prod Res 2005; 19:393-412. [PMID: 15938148 DOI: 10.1080/14786410412331299005] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Topoisomerase I (Topo-I) is a major target for anticancer drug discovery and design. As a result, Topo-I inhibitors constitute an important class of the current anticancer drugs. To date, all of the Topo-I inhibitors that have been clinically evaluated are analogues of camptothecin (CPT), an extract of the Chinese tree Camptotheca acuminata. CPT has shown significant antitumor activity to lung, ovarian, breast, pancreas and stomach cancers. In this article the, phytochemical aspect, and various structural modifications are comprehensively reviewed as in rings A, B, C, D and E. Biological activity of camptothecin, other than anticancer, reported till the year 2003 has also been discussed.
Collapse
Affiliation(s)
- Dharmarajan Sriram
- Medicinal Chemistry Research Laboratory, Pharmacy Group, Birla Institute of Technology and Science, Pilani 333 031, India.
| | | | | | | |
Collapse
|
27
|
Abstract
Immunoconjugates--monoclonal antibodies (mAbs) coupled to highly toxic agents, including radioisotopes and toxic drugs (ineffective when administered systemically alone)--are becoming a significant component of anticancer treatments. By combining the exquisite targeting specificity of mAbs with the enhanced tumor-killing power of toxic effector molecules, immunoconjugates permit sensitive discrimination between target and normal tissue, resulting in fewer toxic side effects than most conventional chemotherapeutic drugs. Two radioimmunoconjugates, ibritumomab tiuxetan (Zevalin) and tositumomab-131I (Bexxar), and one drug conjugate, gemtuzumab ozogamicin (Mylotarg), are now on the market. For the next generation of immunoconjugates, advances in protein engineering will permit greater control of mAb targeting, clearance and pharmacokinetics, resulting in significantly improved delivery to tumors of radioisotopes and potent anticancer drugs. Pre-targeting strategies, which separate the two functions of antibody-based localization and delivery or generation of the toxic agent into two steps, also promise to afford superior tumor targeting and therapeutic efficacy. Several challenges in optimizing immunoconjugates remain, however, including poor intratumoral mAb uptake, normal tissue conjugate exposure and issues surrounding drug potency and conditional release from mAb carriers. Nonetheless, highly promising results from preclinical models will continue to drive the clinical development of this therapeutic class.
Collapse
Affiliation(s)
- Anna M Wu
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Box 951770, 700 Westwood Plaza, Los Angeles, California 90095, USA.
| | | |
Collapse
|
28
|
Thomas CJ, Rahier NJ, Hecht SM. Camptothecin: current perspectives. Bioorg Med Chem 2004; 12:1585-604. [PMID: 15028252 DOI: 10.1016/j.bmc.2003.11.036] [Citation(s) in RCA: 321] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2003] [Accepted: 11/28/2003] [Indexed: 01/20/2023]
Abstract
This review provides a detailed discussion of recent advances in the medicinal chemistry of camptothecin, a potent antitumor antibiotic. Two camptothecin analogues are presently approved for use in the clinic as antitumor agents and several others are in clinical trials. Camptothecin possesses a novel mechanism of action involving the inhibition of DNA relaxation by DNA topoisomerase I, and more specifically the stabilization of a covalent binary complex formed between topoisomerase I and DNA. This review summarizes the current status of studies of the mechanism of action of camptothecin, including topoisomerase I inhibition and additional cellular responses. Modern synthetic approaches to camptothecin and several of the semi-synthetic methods are also discussed. Finally, a systematic evaluation of novel and important analogues of camptothecin and their contribution to the current structure-activity profile are considered.
Collapse
Affiliation(s)
- Craig J Thomas
- Departments of Chemistry and Biology, University of Virginia, Charlottesville, VA 22901, USA
| | | | | |
Collapse
|
29
|
Walker MA, King HD, Dalterio RA, Trail P, Firestone R, Dubowchik GM. Monoclonal antibody mediated intracellular targeting of tallysomycin S10b. Bioorg Med Chem Lett 2004; 14:4323-7. [PMID: 15261295 DOI: 10.1016/j.bmcl.2004.05.089] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2003] [Revised: 05/26/2004] [Accepted: 05/27/2004] [Indexed: 11/22/2022]
Abstract
The potency of tallysomycin S(10b) (TLM S(10b)) an analogue bleomycin was enhanced by up to 875-fold when it was conjugated to the internalizing antibody BR96. Attachment to the antibody is achieved via a Cathepsin B cleavable linker. The enhancement in potency is believed to be a result of cellular uptake of the conjugate upon antigen binding followed by rapid release of the drug inside the lysosome. This method provides a novel approach for increasing the potency and therapeutic index of nominally moderately-active cytotoxic agents.
Collapse
Affiliation(s)
- Michael A Walker
- Bristol-Myers Squibb Pharmaceutical Research Institute, Medicinal Chemistry, PO Box 5100, 5 Research Parkway, Wallingford, CT 06492, USA.
| | | | | | | | | | | |
Collapse
|