1
|
Jin W, Zhang M, Dong C, Huang L, Luo Q. The multifaceted role of MUC1 in tumor therapy resistance. Clin Exp Med 2023; 23:1441-1474. [PMID: 36564679 DOI: 10.1007/s10238-022-00978-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
Tumor therapeutic resistances are frequently linked to the recurrence and poor prognosis of cancers and have been a key bottleneck in clinical tumor treatment. Mucin1 (MUC1), a heterodimeric transmembrane glycoprotein, exhibits abnormally overexpression in a variety of human tumors and has been confirmed to be related to the formation of therapeutic resistance. In this review, the multifaceted roles of MUC1 in tumor therapy resistance are summarized from aspects of pan-cancer principles shared among therapies and individual mechanisms dependent on different therapies. Concretely, the common mechanisms of therapy resistance across cancers include interfering with gene expression, promoting genome instability, modifying tumor microenvironment, enhancing cancer heterogeneity and stemness, and activating evasion and metastasis. Moreover, the individual mechanisms of therapy resistance in chemotherapy, radiotherapy, and biotherapy are introduced. Last but not least, MUC1-involved therapy resistance in different types of cancers and MUC1-related clinical trials are summarized.
Collapse
Affiliation(s)
- Weiqiu Jin
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China
- Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mengwei Zhang
- Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Changzi Dong
- Department of Bioengineering, School of Engineering and Science, University of Pennsylvania, Philadelphia, 19104, USA
| | - Lei Huang
- Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Innovative Research Team of High-Level Local Universities in Shanghai, Shanghai, China.
| | - Qingquan Luo
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
2
|
Samantasinghar A, Sunildutt NP, Ahmed F, Soomro AM, Salih ARC, Parihar P, Memon FH, Kim KH, Kang IS, Choi KH. A comprehensive review of key factors affecting the efficacy of antibody drug conjugate. Biomed Pharmacother 2023; 161:114408. [PMID: 36841027 DOI: 10.1016/j.biopha.2023.114408] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/27/2023] Open
Abstract
Antibody Drug Conjugate (ADC) is an emerging technology to overcome the limitations of chemotherapy by selectively targeting the cancer cells. ADC binds with an antigen, specifically over expressed on the surface of cancer cells, results decrease in bystander effect and increase in therapeutic index. The potency of an ideal ADC is entirely depending on several physicochemical factors such as site of conjugation, molecular weight, linker length, Steric hinderance, half-life, conjugation method, binding energy and so on. Inspite of the fact that there is more than 100 of ADCs are in clinical trial only 14 ADCs are approved by FDA for clinical use. However, to design an ideal ADC is still challenging and there is much more to be done. Here in this review, we have discussed the key components along with their significant role or contribution towards the efficacy of an ADC. Moreover, we also explained about the recent advancement in the conjugation method. Additionally, we spotlit the mode of action of an ADC, recent challenges, and future perspective regarding ADC. The profound knowledge regarding key components and their properties will help in the synthesis or production of different engineered ADCs. Therefore, contributes to develop an ADC with low safety concern and high therapeutic index. We hope this review will improve the understanding and encourage the practicing of research in anticancer ADCs development.
Collapse
Affiliation(s)
| | | | - Faheem Ahmed
- Department of Mechatronics Engineering, Jeju National University, the Republic of Korea
| | | | | | - Pratibha Parihar
- Department of Mechatronics Engineering, Jeju National University, the Republic of Korea
| | - Fida Hussain Memon
- Department of Mechatronics Engineering, Jeju National University, the Republic of Korea
| | | | - In Suk Kang
- Department of Mechatronics Engineering, Jeju National University, the Republic of Korea
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, the Republic of Korea.
| |
Collapse
|
3
|
Pander G, Uhl P, Kühl N, Haberkorn U, Anderl J, Mier W. Antibody-drug conjugates: What drives their progress? Drug Discov Today 2022; 27:103311. [PMID: 35787480 DOI: 10.1016/j.drudis.2022.06.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/01/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022]
Abstract
Antibody-drug conjugates (ADCs) are on the brink of widespread use for the targeted treatment of cancer. ADCs manage the toxicity of drugs with unacceptable narrow therapeutic windows by guiding highly toxic compounds to the target cells, thereby sparing healthy cells. In this review, we describe approved ADCs and discuss their modes of action, together with medicinal chemical aspects, to evaluate the potential for improvement and to combat tumor-acquired resistance. A recent research focus has centered on the stimulation of immune responses to induce immunogenic cell death and the influence on the tumor microenvironment to enhance bystander effects.
Collapse
Affiliation(s)
- Giulia Pander
- Heidelberg University Hospital, Department of Nuclear Medicine, INF 400, 69120 Heidelberg, Germany
| | - Philipp Uhl
- Heidelberg University Hospital, Department of Nuclear Medicine, INF 400, 69120 Heidelberg, Germany
| | - Nikos Kühl
- Heidelberg University, Institute of Pharmacy and Molecular Biotechnology, INF 364, 69120 Heidelberg, Germany
| | - Uwe Haberkorn
- Heidelberg University Hospital, Department of Nuclear Medicine, INF 400, 69120 Heidelberg, Germany
| | - Jan Anderl
- Merck KGaA, Antibody Drug Conjugates & Targeted NBE Therapeutics, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Walter Mier
- Heidelberg University Hospital, Department of Nuclear Medicine, INF 400, 69120 Heidelberg, Germany.
| |
Collapse
|
4
|
Vollmar BS, Frantz C, Schutten MM, Zhong F, Del Rosario G, Go MAT, Yu SF, Leipold DD, Kamath AV, Ng C, Xu K, Dela Cruz-Chuh J, Kozak KR, Chen J, Xu Z, Wai J, Adhikari P, Erickson HK, Dragovich PS, Polson AG, Pillow TH. Calicheamicin Antibody-Drug Conjugates with Improved Properties. Mol Cancer Ther 2021; 20:1112-1120. [PMID: 33722856 DOI: 10.1158/1535-7163.mct-20-0035] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/02/2020] [Accepted: 02/26/2021] [Indexed: 11/16/2022]
Abstract
Calicheamicin antibody-drug conjugates (ADCs) are effective therapeutics for leukemias with two recently approved in the United States: Mylotarg (gemtuzumab ozogamicin) targeting CD33 for acute myeloid leukemia and Besponsa (inotuzumab ozogamicin) targeting CD22 for acute lymphocytic leukemia. Both of these calicheamicin ADCs are heterogeneous, aggregation-prone, and have a shortened half-life due to the instability of the acid-sensitive hydrazone linker in circulation. We hypothesized that we could improve upon the heterogeneity, aggregation, and circulation stability of calicheamicin ADCs by directly attaching the thiol of a reduced calicheamicin to an engineered cysteine on the antibody via a disulfide bond to generate a linkerless and traceless conjugate. We report herein that the resulting homogeneous conjugates possess minimal aggregation and display high in vivo stability with 50% of the drug remaining conjugated to the antibody after 21 days. Furthermore, these calicheamicin ADCs are highly efficacious in mouse models of both solid tumor (HER2+ breast cancer) and hematologic malignancies (CD22+ non-Hodgkin lymphoma). Safety studies in rats with this novel calicheamicin ADC revealed an increased tolerability compared with that reported for Mylotarg. Overall, we demonstrate that applying novel linker chemistry with site-specific conjugation affords an improved, next-generation calicheamicin ADC.
Collapse
Affiliation(s)
| | - Chris Frantz
- Genentech, Inc., South San Francisco, California
| | | | - Fiona Zhong
- Genentech, Inc., South San Francisco, California
| | | | | | - Shang-Fan Yu
- Genentech, Inc., South San Francisco, California
| | | | | | - Carl Ng
- Genentech, Inc., South San Francisco, California
| | - Keyang Xu
- Genentech, Inc., South San Francisco, California
| | | | | | | | - Zijin Xu
- WuXi AppTec Co., Ltd, Shanghai, China
| | - John Wai
- WuXi AppTec Co., Ltd, Shanghai, China
| | | | | | | | | | | |
Collapse
|
5
|
Amani N, Dorkoosh FA, Mobedi H. ADCs, as Novel Revolutionary Weapons for Providing a Step Forward in Targeted Therapy of Malignancies. Curr Drug Deliv 2020; 17:23-51. [DOI: 10.2174/1567201816666191121145109] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/01/2019] [Accepted: 10/29/2019] [Indexed: 11/22/2022]
Abstract
:Antibody drug conjugates (ADCs), as potent pharmaceutical trojan horses for cancer treatment, provide superior efficacy and specific targeting along with low risk of adverse reactions compared to traditional chemotherapeutics. In fact, the development of these agents combines the selective targeting capability of monoclonal antibody (mAb) with high cytotoxicity of chemotherapeutics for controlling the neoplastic mass growth. Different ADCs (more than 60 ADCs) in preclinical and clinical trials were introduced in this novel pharmaceutical field. Various design-based factors must be taken into account for improving the functionality of ADC technology, including selection of appropriate target antigen and high binding affinity of fragment (miniaturized ADCs) or full mAbs (preferentially use of humanized or fully human antibodies compared to murine and chimeric ones), use of bispecific antibodies for dual targeting effect, linker engineering and conjugation method efficacy to obtain more controlled drug to antibody ratio (DAR). Challenging issues affecting therapeutic efficacy and safety of ADCs, including bystander effect, on- and off-target toxicities, multi drug resistance (MDR) are also addressed. 4 FDA-approved ADCs in the market, including ADCETRIS ®, MYLOTARG®, BESPONSA ®, KADCYLA®. The goal of the current review is to evaluate the key parameters affecting ADCs development.
Collapse
Affiliation(s)
- Nooshafarin Amani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Abedin Dorkoosh
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Mobedi
- Novel Drug Delivery Systems (NDDS) Department, Iran Polymer and Petrochemical Institute, Tehran, Iran
| |
Collapse
|
6
|
Karpov AS, Nieto-Oberhuber CM, Abrams T, Beng-Louka E, Blanco E, Chamoin S, Chene P, Dacquignies I, Daniel D, Dillon MP, Doumampouom-Metoul L, Drosos N, Fedoseev P, Furegati M, Granda B, Grotzfeld RM, Hess Clark S, Joly E, Jones D, Lacaud-Baumlin M, Lagasse-Guerro S, Lorenzana EG, Mallet W, Martyniuk P, Marzinzik AL, Mesrouze Y, Nocito S, Oei Y, Perruccio F, Piizzi G, Richard E, Rudewicz PJ, Schindler P, Velay M, Venstrom K, Wang P, Zurini M, Lafrance M. Discovery of Potent and Selective Antibody-Drug Conjugates with Eg5 Inhibitors through Linker and Payload Optimization. ACS Med Chem Lett 2019; 10:1674-1679. [PMID: 31857845 DOI: 10.1021/acsmedchemlett.9b00468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/03/2019] [Indexed: 12/16/2022] Open
Abstract
Targeted antimitotic agents are a promising class of anticancer therapies. Herein, we describe the development of a potent and selective antimitotic Eg5 inhibitor based antibody-drug conjugate (ADC). Preliminary studies were performed using proprietary Eg5 inhibitors which were conjugated onto a HER2-targeting antibody using maleimido caproyl valine-citrulline para-amino benzocarbamate, or MC-VC-PABC cleavable linker. However, the resulting ADCs lacked antigen-specificity in vivo, probably from premature release of the payload. Second-generation ADCs were then developed, using noncleavable linkers, and the resulting conjugates (ADC-4 and ADC-10) led to in vivo efficacy in an HER-2 expressing (SK-OV-3ip) mouse xenograft model while ADC-11 led to in vivo efficacy in an anti-c-KIT (NCI-H526) mouse xenograft model in a target-dependent manner.
Collapse
Affiliation(s)
- Alexei S. Karpov
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | | | - Tinya Abrams
- Novartis Institutes for BioMedical Research, 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Edwige Beng-Louka
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Enrique Blanco
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Sylvie Chamoin
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Patrick Chene
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Isabelle Dacquignies
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Dylan Daniel
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Michael P. Dillon
- Novartis Institutes for BioMedical Research, 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | | | | | - Pavel Fedoseev
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Markus Furegati
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Brian Granda
- Novartis Institutes for BioMedical Research, 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Robert M. Grotzfeld
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Suzanna Hess Clark
- Novartis Institutes for BioMedical Research, 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Emilie Joly
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Darryl Jones
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Marion Lacaud-Baumlin
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | | | - Edward G. Lorenzana
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - William Mallet
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Piotr Martyniuk
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Andreas L. Marzinzik
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Yannick Mesrouze
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Sandro Nocito
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Yoko Oei
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Francesca Perruccio
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Grazia Piizzi
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Etienne Richard
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Patrick J. Rudewicz
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Patrick Schindler
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Mélanie Velay
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Kristine Venstrom
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Peiyin Wang
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Mauro Zurini
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
| | - Marc Lafrance
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, CH-4056 Basel, Switzerland
- Novartis Institutes for BioMedical Research, 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
7
|
Antibody-drug conjugates (ADCs): Potent biopharmaceuticals to target solid and hematological cancers- an overview. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.08.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
8
|
Nasiri H, Valedkarimi Z, Aghebati‐Maleki L, Majidi J. Antibody‐drug conjugates: Promising and efficient tools for targeted cancer therapy. J Cell Physiol 2018; 233:6441-6457. [DOI: 10.1002/jcp.26435] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 01/05/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Hadi Nasiri
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Zahra Valedkarimi
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Leili Aghebati‐Maleki
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
| | - Jafar Majidi
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyFaculty of MedicineTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
9
|
Lee MTW, Maruani A, Richards DA, Baker JR, Caddick S, Chudasama V. Enabling the controlled assembly of antibody conjugates with a loading of two modules without antibody engineering. Chem Sci 2017; 8:2056-2060. [PMID: 28451324 PMCID: PMC5399535 DOI: 10.1039/c6sc03655d] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/24/2016] [Indexed: 12/12/2022] Open
Abstract
The generation of antibody conjugates with a loading of two modules is desirable for a host of reasons. Whilst certain antibody engineering approaches have been useful in the preparation of such constructs, a reliable method based on a native antibody scaffold without the use of enzymes or harsh oxidative conditions has hitherto not been achieved. The use of native antibodies has several advantages in terms of cost, practicality, accessibility, time and overall efficiency. Herein we present a novel, reliable method of furnishing antibody conjugates with a loading of two modules starting from a native antibody scaffold.
Collapse
Affiliation(s)
- Maximillian T W Lee
- Department of Chemistry , University College London , 20 Gordon Street , London , WC1H 0AJ , UK . ; Tel: +44 (0)207 679 2077
| | - Antoine Maruani
- Department of Chemistry , University College London , 20 Gordon Street , London , WC1H 0AJ , UK . ; Tel: +44 (0)207 679 2077
| | - Daniel A Richards
- Department of Chemistry , University College London , 20 Gordon Street , London , WC1H 0AJ , UK . ; Tel: +44 (0)207 679 2077
| | - James R Baker
- Department of Chemistry , University College London , 20 Gordon Street , London , WC1H 0AJ , UK . ; Tel: +44 (0)207 679 2077
| | - Stephen Caddick
- Department of Chemistry , University College London , 20 Gordon Street , London , WC1H 0AJ , UK . ; Tel: +44 (0)207 679 2077
| | - Vijay Chudasama
- Department of Chemistry , University College London , 20 Gordon Street , London , WC1H 0AJ , UK . ; Tel: +44 (0)207 679 2077
| |
Collapse
|
10
|
Puthenveetil S, Loganzo F, He H, Dirico K, Green M, Teske J, Musto S, Clark T, Rago B, Koehn F, Veneziale R, Falahaptisheh H, Han X, Barletta F, Lucas J, Subramanyam C, O'Donnell CJ, Tumey LN, Sapra P, Gerber HP, Ma D, Graziani EI. Natural Product Splicing Inhibitors: A New Class of Antibody-Drug Conjugate (ADC) Payloads. Bioconjug Chem 2016; 27:1880-8. [PMID: 27412791 DOI: 10.1021/acs.bioconjchem.6b00291] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
There is a considerable ongoing work to identify new cytotoxic payloads that are appropriate for antibody-based delivery, acting via mechanisms beyond DNA damage and microtubule disruption, highlighting their importance to the field of cancer therapeutics. New modes of action will allow a more diverse set of tumor types to be targeted and will allow for possible mechanisms to evade the drug resistance that will invariably develop to existing payloads. Spliceosome inhibitors are known to be potent antiproliferative agents capable of targeting both actively dividing and quiescent cells. A series of thailanstatin-antibody conjugates were prepared in order to evaluate their potential utility in the treatment of cancer. After exploring a variety of linkers, we found that the most potent antibody-drug conjugates (ADCs) were derived from direct conjugation of the carboxylic acid-containing payload to surface lysines of the antibody (a "linker-less" conjugate). Activity of these lysine conjugates was correlated to drug-loading, a feature not typically observed for other payload classes. The thailanstatin-conjugates were potent in high target expressing cells, including multidrug-resistant lines, and inactive in nontarget expressing cells. Moreover, these ADCs were shown to promote altered splicing products in N87 cells in vitro, consistent with their putative mechanism of action. In addition, the exposure of the ADCs was sufficient to result in excellent potency in a gastric cancer xenograft model at doses as low as 1.5 mg/kg that was superior to the clinically approved ADC T-DM1. The results presented herein therefore open the door to further exploring splicing inhibition as a potential new mode-of-action for novel ADCs.
Collapse
Affiliation(s)
| | - Frank Loganzo
- Oncology-Rinat Research & Development, Pfizer , 401 North Middletown Road, Pearl River, New York 10965, United States
| | | | | | | | | | - Sylvia Musto
- Oncology-Rinat Research & Development, Pfizer , 401 North Middletown Road, Pearl River, New York 10965, United States
| | | | | | | | - Robert Veneziale
- Drug Safety Research and Development, Pfizer , 401 North Middletown Road, Pearl River, New York 10977, United States
| | - Hadi Falahaptisheh
- Drug Safety Research and Development, Pfizer , 401 North Middletown Road, Pearl River, New York 10977, United States
| | | | | | - Judy Lucas
- Oncology-Rinat Research & Development, Pfizer , 401 North Middletown Road, Pearl River, New York 10965, United States
| | | | | | | | - Puja Sapra
- Oncology-Rinat Research & Development, Pfizer , 401 North Middletown Road, Pearl River, New York 10965, United States
| | - Hans Peter Gerber
- Oncology-Rinat Research & Development, Pfizer , 401 North Middletown Road, Pearl River, New York 10965, United States
| | - Dangshe Ma
- Oncology-Rinat Research & Development, Pfizer , 401 North Middletown Road, Pearl River, New York 10965, United States
| | | |
Collapse
|
11
|
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: 3.1] [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
|
12
|
Recent advances in the construction of antibody–drug conjugates. Nat Chem 2016; 8:114-9. [DOI: 10.1038/nchem.2415] [Citation(s) in RCA: 228] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 11/10/2015] [Indexed: 01/06/2023]
|
13
|
Patino T, Mahajan U, Palankar R, Medvedev N, Walowski J, Münzenberg M, Mayerle J, Delcea M. Multifunctional gold nanorods for selective plasmonic photothermal therapy in pancreatic cancer cells using ultra-short pulse near-infrared laser irradiation. NANOSCALE 2015; 7:5328-37. [PMID: 25721177 DOI: 10.1039/c5nr00114e] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Gold nanorods (AuNRs) have attracted considerable attention in plasmonic photothermal therapy for cancer treatment by exploiting their selective and localized heating effect due to their unique photophysical properties. Here we describe a strategy to design a novel multifunctional platform based on AuNRs to: (i) specifically target the adenocarcinoma MUC-1 marker through the use of the EPPT-1 peptide, (ii) enhance cellular uptake through a myristoylated polyarginine peptide (MPAP) and (iii) selectively induce cell death by ultra-short near infrared laser pulses. We used a biotin-avidin based approach to conjugate EPPT-1 and MPAP to AuNRs. Dual-peptide (EPPT-1+MPAP) labelled AuNRs showed a significantly higher uptake by pancreatic ductal adenocarcinoma cells when compared to their single peptide or avidin conjugated counterparts. In addition, we selectively induced cell death by ultra-short near infrared laser pulses in small target volumes (∼1 μm3), through the creation of plasmonic nanobubbles that lead to the destruction of a local cell environment. Our approach opens new avenues for conjugation of multiple ligands on AuNRs targeting cancer cells and tumors and it is relevant for plasmonic photothermal therapy.
Collapse
Affiliation(s)
- Tania Patino
- Nanostructure Group, ZIK HIKE - Center for Innovation Competence, Humoral Immune Reactions in Cardiovascular Diseases, Ernst-Moritz-Arndt-University, 17489 Greifswald, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Klute K, Nackos E, Tasaki S, Nguyen DP, Bander NH, Tagawa ST. Microtubule inhibitor-based antibody-drug conjugates for cancer therapy. Onco Targets Ther 2014; 7:2227-36. [PMID: 25506226 PMCID: PMC4259504 DOI: 10.2147/ott.s46887] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The specificity of monoclonal antibodies represents a potential therapeutic advantage, but their use as single agents in oncology has proven limited to date. The development of antibody-drug conjugates (ADCs) takes advantage of the specificity of the monoclonal antibody and potent cytotoxic effect of chemotherapy, leading to enhanced cytotoxicity in target cells and limiting toxicity to normal tissue. Microtubules represent a validated oncologic target in a range of tumor types, with a number of anti-microtubule targeting cytotoxic drugs approved for cancer use. The systemic use of potent microtubule-binding agents is limited by their effects in normal cells, which leads to toxicity including myelosuppression and peripheral neuropathy. Linking these agents to monoclonal antibodies may limit toxicity to normal tissues and increase drug concentration in target tissues, also allowing the use of more potent agents which would be too toxic to administer in their unbound form. Two such ADCs have been approved for clinical use and many others are in development. Here we review the characteristics of each of the ADC components that have led to efficacious therapies and discuss some of the tubulin inhibitor-based ADCs in development for cancer therapy.
Collapse
Affiliation(s)
- Kelsey Klute
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Eleni Nackos
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Shinsuke Tasaki
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Daniel P Nguyen
- Department of Urology, Weill Cornell Medical College, New York, NY, USA
| | - Neil H Bander
- Department of Urology, Weill Cornell Medical College, New York, NY, USA
| | - Scott T Tagawa
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA ; Department of Urology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
15
|
Shefet-Carasso L, Benhar I. Antibody-targeted drugs and drug resistance--challenges and solutions. Drug Resist Updat 2014; 18:36-46. [PMID: 25476546 DOI: 10.1016/j.drup.2014.11.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 11/16/2014] [Accepted: 11/17/2014] [Indexed: 11/24/2022]
Abstract
Antibody-based therapy of various human malignancies has shown efficacy in the past 30 years and is now one of the most successful and leading strategies for targeted treatment of patients harboring hematological malignancies and solid tumors. Antibody-drug conjugates (ADCs) aim to take advantage of the affinity and specificity of monoclonal antibodies (mAbs) to selectively deliver potent cytotoxic drugs to antigen-expressing tumor cells. Key parameters for ADC include choosing the optimal components of the ADC (the antibody, the linker and the cytotoxic drug) and selecting the suitable cell-surface target antigen. Building on the success of recent FDA approval of brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla), ADCs are currently a class of drugs with a robust pipeline with clinical applications that are rapidly expanding. The more ADCs are being evaluated in preclinical models and clinical trials, the clearer are becoming the parameters and the challenges required for their therapeutic success. This rapidly growing knowledge and clinical experience are revealing novel modalities and mechanisms of resistance to ADCs, hence offering plausible solutions to such challenges. Here, we review the key parameters for designing a powerful ADC, focusing on how ADCs are addressing the challenge of multiple drug resistance (MDR) and its rational overcoming.
Collapse
Affiliation(s)
- LeeRon Shefet-Carasso
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, Israel
| | - Itai Benhar
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, Israel.
| |
Collapse
|
16
|
Zhou Q, Stefano JE, Manning C, Kyazike J, Chen B, Gianolio DA, Park A, Busch M, Bird J, Zheng X, Simonds-Mannes H, Kim J, Gregory RC, Miller RJ, Brondyk WH, Dhal PK, Pan CQ. Site-specific antibody-drug conjugation through glycoengineering. Bioconjug Chem 2014; 25:510-20. [PMID: 24533768 DOI: 10.1021/bc400505q] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Antibody-drug conjugates (ADCs) have been proven clinically to be more effective anti-cancer agents than native antibodies. However, the classical conjugation chemistries to prepare ADCs by targeting primary amines or hinge disulfides have a number of shortcomings including heterogeneous product profiles and linkage instability. We have developed a novel site-specific conjugation method by targeting the native glycosylation site on antibodies as an approach to address these limitations. The native glycans on Asn-297 of antibodies were enzymatically remodeled in vitro using galactosyl and sialyltransferases to introduce terminal sialic acids. Periodate oxidation of these sialic acids yielded aldehyde groups which were subsequently used to conjugate aminooxy functionalized cytotoxic agents via oxime ligation. The process has been successfully demonstrated with three antibodies including trastuzumab and two cytotoxic agents. Hydrophobic interaction chromatography and LC-MS analyses revealed the incorporation of ~1.6 cytotoxic agents per antibody molecule, approximating the number of sialic acid residues. These glyco-conjugated ADCs exhibited target-dependent antiproliferative activity toward antigen-positive tumor cells and significantly greater antitumor efficacy than naked antibody in a Her2-positive tumor xenograft model. These findings suggest that enzymatic remodeling combined with oxime ligation of the native glycans of antibodies offers an attractive approach to generate ADCs with well-defined product profiles. The site-specific conjugation approach presented here provides a viable alternative to other methods, which involve a need to either re-engineer the antibody sequence or develop a highly controlled chemical process to ensure reproducible drug loading.
Collapse
Affiliation(s)
- Qun Zhou
- Sanofi-Genzyme R&D Center, Genzyme Corporation, A Sanofi Company , Framingham, Massachusetts 01701, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Chari RVJ, Miller ML, Widdison WC. Antibody-drug conjugates: an emerging concept in cancer therapy. Angew Chem Int Ed Engl 2014; 53:3796-827. [PMID: 24677743 DOI: 10.1002/anie.201307628] [Citation(s) in RCA: 711] [Impact Index Per Article: 71.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Indexed: 01/17/2023]
Abstract
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 compound 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 technology, i.e., the antibody, the cytotoxic compound, and the linker connecting them, leading to the current successes. The design of ADCs currently in clinical development, and results from mechanistic studies and preclinical and clinical evaluation are discussed. Emerging technologies that seek to further advance this exciting area of research are also discussed.
Collapse
Affiliation(s)
- Ravi V J Chari
- ImmunoGen, Inc. 830 Winter St, Waltham, MA 02451 (USA) http://www.immunogen.com.
| | | | | |
Collapse
|
18
|
Chari RVJ, Miller ML, Widdison WC. Antikörper-Wirkstoff-Konjugate: ein neues Konzept in der Krebstherapie. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201307628] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
19
|
Cowan AJ, Laszlo GS, Estey EH, Walter RB. Antibody-based therapy of acute myeloid leukemia with gemtuzumab ozogamicin. Front Biosci (Landmark Ed) 2013; 18:1311-34. [PMID: 23747885 DOI: 10.2741/4181] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Antibodies have created high expectations for effective yet tolerated therapeutics in acute myeloid leukemia (AML). Hitherto the most exploited target is CD33, a myeloid differentiation antigen found on AML blasts in most patients and, perhaps, leukemic stem cells in some. Treatment efforts have focused on conjugated antibodies, particularly gemtuzumab ozogamicin (GO), an anti-CD33 antibody carrying a toxic calicheamicin-g 1 derivative that, after intracellular hydrolytic release, induces DNA strand breaks, apoptosis, and cell death. Serving as paradigm for this strategy, GO was the first anti-cancer immunoconjugate to obtain regulatory approval in the U.S. While efficacious as monotherapy in acute promyelocytic leukemia (APL), GO alone induces remissions in less than 25-35% of non-APL AML patients. However, emerging data from well controlled trials now indicate that GO improves survival for many non-APL AML patients, supporting the conclusion that CD33 is a clinically relevant target for some disease subsets. It is thus unfortunate that GO has become unavailable in many parts of the world, and the drug's usefulness should be reconsidered and selected patients granted access to this immunoconjugate.
Collapse
Affiliation(s)
- Andrew J Cowan
- Hematology/Oncology Fellowship Program, University of Washington, Seattle, WA, USA
| | | | | | | |
Collapse
|
20
|
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
|
21
|
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]
|
22
|
Antibody-drug conjugates: using monoclonal antibodies for delivery of cytotoxic payloads to cancer cells. Ther Deliv 2012; 2:397-416. [PMID: 22834009 DOI: 10.4155/tde.10.98] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
One approach to improving activity of anticancer drugs is to conjugate them to antibodies that recognize tumor-associated, cell-surface antigens. The antibody-drug conjugate concept evolved following major advances, first, in the development of humanized and fully human antibodies; second, in the discoveries of highly cytotoxic compounds ('drugs) linkable to antibodies; and finally, in the optimization of linkers that couple the drug to the antibody and provide sufficient stability of the antibody-drug conjugate in the circulation, optimal activation of the drug in the tumor, and the ability of the activated drug to overcome multidrug resistance. In this article, we will review the considerations for selecting a target antigen, the design of the conjugate, and the pre-clinical and clinical experiences with the current generation of antibody-drug conjugates.
Collapse
|
23
|
Lee MH, Kim JY, Han JH, Bhuniya S, Sessler JL, Kang C, Kim JS. Direct Fluorescence Monitoring of the Delivery and Cellular Uptake of a Cancer-Targeted RGD Peptide-Appended Naphthalimide Theragnostic Prodrug. J Am Chem Soc 2012; 134:12668-74. [DOI: 10.1021/ja303998y] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Min Hee Lee
- Department of Chemistry, Korea University, Seoul, 136-701, Korea
| | - Jin Young Kim
- The School of East-West Medical
Science, Kyung Hee University, Yongin,
446-701, Korea
| | - Ji Hye Han
- The School of East-West Medical
Science, Kyung Hee University, Yongin,
446-701, Korea
| | | | - Jonathan L. Sessler
- Department
of Chemistry and Biochemistry, The University of Texas at Austin, Austin, Texas 78712-0165,
United States
- Department
of Chemistry, Yonsei University, 262 Seonsanno
Sinchon-dong, Seodaemun-gu,
Seoul 120-749, Korea
| | - Chulhun Kang
- The School of East-West Medical
Science, Kyung Hee University, Yongin,
446-701, Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 136-701, Korea
| |
Collapse
|
24
|
Antibody–Drug Conjugates for Targeted Cancer Therapy. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2012. [DOI: 10.1016/b978-0-12-396492-2.00023-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
25
|
Mahato R, Tai W, Cheng K. Prodrugs for improving tumor targetability and efficiency. Adv Drug Deliv Rev 2011; 63:659-70. [PMID: 21333700 PMCID: PMC3132824 DOI: 10.1016/j.addr.2011.02.002] [Citation(s) in RCA: 256] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 01/14/2011] [Accepted: 02/03/2011] [Indexed: 12/14/2022]
Abstract
As the mainstay in the treatment of various cancers for several decades, chemotherapy is successful but still faces challenges including non-selectivity and high toxicity. Improving the selectivity is therefore a critical step to improve the therapeutic efficacy of chemotherapy. Prodrug is one of the most promising approaches to increase the selectivity and efficacy of a chemotherapy drug. The classical prodrug approach is to improve the pharmaceutical properties (solubility, stability, permeability, irritation, distribution, etc.) via a simple chemical modification. This review will focus on various targeted prodrug designs that have been developed to increase the selectivity of chemotherapy drugs. Various tumor-targeting ligands, transporter-associated ligands, and polymers can be incorporated in a prodrug to enhance the tumor uptake. Prodrugs can also be activated by enzymes that are specifically expressed at a higher level in tumors, leading to a selective anti-tumor effect. This can be achieved by conjugating the enzyme to a tumor-specific antibody, or delivering a vector expressing the enzyme into tumor cells.
Collapse
Affiliation(s)
- Rubi Mahato
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108
| | - Wanyi Tai
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108
| | - Kun Cheng
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108
| |
Collapse
|
26
|
Synthesis and Evaluation of Hydrophilic Linkers for Antibody–Maytansinoid Conjugates. J Med Chem 2011; 54:3606-23. [DOI: 10.1021/jm2002958] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Affiliation(s)
- Jonas J. Forsman
- Laboratory of Organic Chemistry, Åbo Akademi University, FI-20500 Åbo, Finland
| | - Reko Leino
- Laboratory of Organic Chemistry, Åbo Akademi University, FI-20500 Åbo, Finland
| |
Collapse
|
28
|
Bildstein L, Dubernet C, Couvreur P. Prodrug-based intracellular delivery of anticancer agents. Adv Drug Deliv Rev 2011; 63:3-23. [PMID: 21237228 DOI: 10.1016/j.addr.2010.12.005] [Citation(s) in RCA: 218] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 12/15/2010] [Accepted: 12/21/2010] [Indexed: 01/08/2023]
Abstract
There are numerous anticancer agents based on a prodrug approach. However, no attempt has been made to review the ample available literature with a specific focus on the altered cell uptake pathways enabled by the conjugation and on the intracellular drug-release mechanisms. This article focuses on the cellular interactions of a broad selection of parenterally administered anticancer prodrugs based on synthetic polymers, proteins or lipids. The report also aims to highlight the prodrug design issues, which are key points to obtain an efficient intracellular drug delivery. The chemical basis of these molecular concepts is put into perspective with the uptake and intracellular activation mechanisms, the in vitro and in vivo proofs of concepts and the clinical results. Several active targeting strategies and stimuli-responsive architectures are discussed throughout the article.
Collapse
Affiliation(s)
- L Bildstein
- UMR CNRS 8612, IFR 141-ITFM, Faculté de Pharmacie, University Paris-Sud, Châtenay-Malabry 92296, France
| | | | | |
Collapse
|
29
|
Wu A, Xu Y, Qian X. Novel naphthalimide–indomethacin hybrids as potential antitumor agents: effects of linkers on hypoxic/oxic cytotoxicity and apoptosis-inducing activity. MONATSHEFTE FUR CHEMIE 2010. [DOI: 10.1007/s00706-010-0337-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
30
|
Miyano T, Wijagkanalan W, Kawakami S, Yamashita F, Hashida M. Anionic Amino Acid Dendrimer−Trastuzumab Conjugates for Specific Internalization in HER2-Positive Cancer Cells. Mol Pharm 2010; 7:1318-27. [DOI: 10.1021/mp100105c] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Takuya Miyano
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan, Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo 102-8471, Japan, and Institute of Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Wassana Wijagkanalan
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan, Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo 102-8471, Japan, and Institute of Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Shigeru Kawakami
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan, Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo 102-8471, Japan, and Institute of Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Fumiyoshi Yamashita
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan, Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo 102-8471, Japan, and Institute of Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Mitsuru Hashida
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan, Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo 102-8471, Japan, and Institute of Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
31
|
Kovtun YV, Audette CA, Mayo MF, Jones GE, Doherty H, Maloney EK, Erickson HK, Sun X, Wilhelm S, Ab O, Lai KC, Widdison WC, Kellogg B, Johnson H, Pinkas J, Lutz RJ, Singh R, Goldmacher VS, Chari RV. Antibody-Maytansinoid Conjugates Designed to Bypass Multidrug Resistance. Cancer Res 2010; 70:2528-37. [DOI: 10.1158/0008-5472.can-09-3546] [Citation(s) in RCA: 189] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
32
|
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: 18.2] [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
|
33
|
Lee SH. Disulfide and multisulfide antitumor agents and their modes of action. Arch Pharm Res 2009; 32:299-315. [DOI: 10.1007/s12272-009-1300-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2008] [Revised: 12/20/2008] [Accepted: 01/11/2009] [Indexed: 11/25/2022]
|
34
|
Rubinstein DB, Karmely M, Pichinuk E, Ziv R, Benhar I, Feng N, Smorodinsky NI, Wreschner DH. The MUC1 oncoprotein as a functional target: Immunotoxin binding to α/β junction mediates cell killing. Int J Cancer 2009; 124:46-54. [DOI: 10.1002/ijc.23910] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
35
|
|
36
|
Abstract
Colloidal drug delivery systems have been providing alternative formulation approaches for problematic drug candidates, and improved delivery for existing compounds for decades. Colloidal systems for drug delivery have all evolved down a similar pathway, almost irrespective of the delivery system, from conception, to the use of safer excipients, PEGylation for passive targeting and attachment of ligands for active targeting. The recent emergence of truly biologically interactive systems represents the latest step forward in colloidal delivery systems. In this article, the maturation pathway and recent advances for the major classes of colloidal delivery systems are reviewed, and the paper poses the question of whether the nanotechnology boom will create a revolution in colloidal delivery, or just the next natural stage in evolution.
Collapse
Affiliation(s)
- Ben J Boyd
- Department of Pharmaceutics, Victorian College of Pharmacy - Monash University, 381 Royal Pde, Parkville, VIC, 3052, Australia.
| |
Collapse
|
37
|
Alley SC, Benjamin DR, Jeffrey SC, Okeley NM, Meyer DL, Sanderson RJ, Senter PD. Contribution of Linker Stability to the Activities of Anticancer Immunoconjugates. Bioconjug Chem 2008; 19:759-65. [DOI: 10.1021/bc7004329] [Citation(s) in RCA: 351] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Stephen C. Alley
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021
| | | | - Scott C. Jeffrey
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021
| | - Nicole M. Okeley
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021
| | - Damon L. Meyer
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021
| | | | - Peter D. Senter
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021
| |
Collapse
|
38
|
Kratz F, Müller I, Ryppa C, Warnecke A. Prodrug Strategies in Anticancer Chemotherapy. ChemMedChem 2008; 3:20-53. [DOI: 10.1002/cmdc.200700159] [Citation(s) in RCA: 374] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
39
|
Shen Y, Tang H, Radosz M, Van Kirk E, Murdoch WJ. pH-responsive nanoparticles for cancer drug delivery. Methods Mol Biol 2008; 437:183-216. [PMID: 18369970 DOI: 10.1007/978-1-59745-210-6_10] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Solid tumors have an acidic extracellular environment and an altered pH gradient across their cell compartments. Nanoparticles responsive to the pH gradients are promising for cancer drug delivery. Such pH-responsive nanoparticles consist of a corona and a core, one or both of which respond to the external pH to change their soluble/insoluble or charge states. Nanoparticles whose coronas become positively charged or become soluble to make their targeting groups available for binding at the tumor extracellular pH have been developed for promoting cellular targeting and internalization. Nanoparticles whose cores become soluble or change their structures to release the carried drugs at the tumor extracellular pH or lysosomal pH have been developed for fast drug release into the extracellular fluid or cytosol. Such pH-responsive nanoparticles have therapeutic advantages over the conventional pH-insensitive counterparts.
Collapse
Affiliation(s)
- Youqing Shen
- Soft Materials Laboratory, Department of Chemical and Petroleum Engineering, University of Wyoming, Laramie, WY, USA
| | | | | | | | | |
Collapse
|
40
|
Wang N, Dong A, Tang H, Van Kirk EA, Johnson PA, Murdoch WJ, Radosz M, Shen Y. Synthesis of Degradable Functional Poly(ethylene glycol) Analogs as Versatile Drug Delivery Carriers. Macromol Biosci 2007; 7:1187-98. [PMID: 17665412 DOI: 10.1002/mabi.200700065] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Poly(ethylene glycol) (PEG) is widely used as a water soluble carrier for polymer-drug conjugates. Herein, we report degradable linear PEG analogs (DPEGs) carrying multifunctional groups. The DPEGs were synthesized by a Michael addition based condensation polymerization of dithiols and PEG diacrylates (PEGDA) or dimethacrylates (PEGDMA). They were stable at pH 7.4 but quickly degraded at pH 6.0 and 5.0. Thus, DPEGs could be used as drug carriers without concern for their retention in the body. DPEGs could be made to carry such functional groups as terminal thiol or (meth)acrylate and pendant hydroxyl groups. The functional groups were used for conjugation of drugs and targeting groups. This new type of PEG analog will be useful for drug delivery and the PEGylation of biomolecules and colloidal particles.
Collapse
Affiliation(s)
- Na Wang
- Soft Materials Laboratory and Department of Chemical and Petroleum Engineering, University of Wyoming, Laramie, Wyoming, 82071, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Hollander I, Kunz A, Hamann PR. Selection of Reaction Additives Used in the Preparation of Monomeric Antibody−Calicheamicin Conjugates. Bioconjug Chem 2007; 19:358-61. [DOI: 10.1021/bc700321z] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Irwin Hollander
- Wyeth Research, 401 N. Middletown Road, Pearl River, New York 10965
| | - Arthur Kunz
- Wyeth Research, 401 N. Middletown Road, Pearl River, New York 10965
| | | |
Collapse
|
42
|
Kovtun YV, Goldmacher VS. Cell killing by antibody-drug conjugates. Cancer Lett 2007; 255:232-40. [PMID: 17553616 DOI: 10.1016/j.canlet.2007.04.010] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Revised: 04/10/2007] [Accepted: 04/25/2007] [Indexed: 11/22/2022]
Abstract
Antibody-drug conjugates (ADCs) are designed to specifically bind to and kill cells expressing their target antigens. In addition to the obvious requirement of the presence of the target antigen on the cell surface, several other factors contribute to the sensitivity of target cells to the action of ADCs. These include (i) the rate of internalization of the ADC, (ii) its proteolytic degradation in late endosomes and lysosomes and the subsequent release of cytotoxic drug, and (iii) the intracellular concentration of the released drug. In addition to killing antigen-expressing cells, some ADCs were found to kill bystander cells irrespective of their antigen expression. This review summarizes the current knowledge of the mechanisms of killing of antigen-expressing and bystander cells by antibody-drug conjugates.
Collapse
Affiliation(s)
- Yelena V Kovtun
- ImmunoGen Inc., 128 Sidney Street, Cambridge, MA 02139-4239, USA.
| | | |
Collapse
|
43
|
Mazor Y, Barnea I, Keydar I, Benhar I. Antibody internalization studied using a novel IgG binding toxin fusion. J Immunol Methods 2007; 321:41-59. [PMID: 17336321 DOI: 10.1016/j.jim.2007.01.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2006] [Revised: 11/29/2006] [Accepted: 01/07/2007] [Indexed: 10/23/2022]
Abstract
Targeted therapy encompasses a wide variety of different strategies, which can be divided into direct or indirect approaches. Direct approaches target tumor-associated antigens by monoclonal antibodies (mAbs) binding to the relevant antigens or by small-molecule drugs that interfere with these proteins. Indirect approaches rely on tumor-associated antigens expressed on the cell surface with antibody-drug conjugates or antibody-based fusion proteins containing different kinds of effector molecules. To deliver a lethal cargo into tumor cells, the targeting antibodies should efficiently internalize into the cells. Similarly, to qualify as targets for such drugs newly-discovered cell-surface molecules should facilitate the internalization of antibodies that bind to them. Internalization can be studied be several biochemical and microscopy approaches. An undisputed proof of internalization can be provided by the ability of an antibody to specifically deliver a drug into the target cells and kill it. We present a novel IgG binding toxin fusion, ZZ-PE38, in which the Fc-binding ZZ domain, derived from Streptococcal protein A, is linked to a truncated Pseudomonas exotoxin A, the preparation of complexes between ZZ-PE38 and IgGs that bind tumor cells and the specific cytotoxicity of such immunocomplexes is reported. Our results suggest that ZZ-PE38 could prove to be an invaluable tool for the evaluation of the suitability potential of antibodies and their cognate cell-surface antigens to be targeted by immunotherapeutics based on armed antibodies that require internalization.
Collapse
Affiliation(s)
- Yariv Mazor
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Green Building, Room 202, Tel-Aviv University, Ramat Aviv 69978, Israel
| | | | | | | |
Collapse
|
44
|
Filpula D. Antibody engineering and modification technologies. ACTA ACUST UNITED AC 2007; 24:201-15. [PMID: 17466589 DOI: 10.1016/j.bioeng.2007.03.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Accepted: 03/20/2007] [Indexed: 10/23/2022]
Abstract
Antibody engineering has become a well-developed discipline, encompassing discovery methods, production strategies, and modification techniques that have brought forth clinically investigated and marketed therapeutics. The realization of the long-standing goal of production of fully human monoclonal antibodies has focused intensive research on the clinical employment of this potent drug category. However, antibodies are large macromolecules that pose numerous challenges in formulation, optimal pharmacokinetics, manufacturing, stability, and process development. While further improvements in discovery technologies, such as phage display, ribosome display, and transgenic animals continue to advance our capacity to rapidly screen and refine optimal binding molecules, antibody engineers have recently focused more of their efforts on improving protein production and stability, as well as engineering improved biological properties in the effector domains of monoclonal antibodies. A second long-standing goal of antibody engineering, the development of targeted drugs, has not been wholly realized, but this obvious application for antibodies is currently undergoing increasing exploration. Minimal binding proteins, such as Fab, scFv, and single variable domains are the preferred targeting elements for some investigational drugs, whereas non-immunoglobulin scaffold proteins have been explored as binding proteins in other designs. The necessity to utilize non-protein components in targeted drugs, such as polymers, linkers, and cytotoxics, has brought a convergence of the fields of bioconjugate chemistry and protein engineering in experimental antibody therapeutics.
Collapse
Affiliation(s)
- David Filpula
- Enzon Pharmaceuticals, Piscataway, NJ 08854-3969, USA.
| |
Collapse
|
45
|
|
46
|
Davies J, Wang H, Taylor T, Warabi K, Huang XH, Andersen RJ. Uncialamycin, A New Enediyne Antibiotic. Org Lett 2005; 7:5233-6. [PMID: 16268546 DOI: 10.1021/ol052081f] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
[structure: see text] Laboratory cultures of an undescribed streptomycete obtained from the surface of a British Columbia lichen produce uncialamycin (1), a new enediyne antibiotic. The structure of uncialamycin (1) has been elucidated by analysis of spectroscopic data. Uncialamycin (1) exhibits potent in vitro antibacterial activity against gram-positive and gram-negative human pathogens, including Burkholderia cepacia, a major cause of morbidity and mortality in patients with cystic fibrosis.
Collapse
Affiliation(s)
- Julian Davies
- Life Sciences Institute, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z1
| | | | | | | | | | | |
Collapse
|
47
|
Chen J, Jaracz S, Zhao X, Chen S, Ojima I. Antibody–cytotoxic agent conjugates for cancer therapy. Expert Opin Drug Deliv 2005; 2:873-90. [PMID: 16296784 DOI: 10.1517/17425247.2.5.873] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Antibody-based delivery of cytotoxic agents, including toxins, to tumours can dramatically reduce systemic toxicity and increase therapeutic efficacy. The advantage of a monoclonal antibody (mAb) is superior selectivity towards antigens expressed on the surface of cancer cells. Recent advances in biotechnology accelerated progress in the pharmaceutical applications of mAbs. A cytotoxic warhead is attached to a mAb in an immunoconjugate via a linker, which is stable in circulation but efficiently cleaved in the tumour tissue. The warhead, mAb and linker play important roles in the successful design of potent and efficient immunoconjugates. To date, one mAb-cytotoxic agent conjugate has been approved by the FDA and several other candidates are in various stages of clinical trials. This review describes the recent progress in the design and development of mAb-based immunoconjugates of cytotoxic agents, and summarises the criteria for the critical choices of a suitable mAb, linker and cytotoxic agent to design an efficacious immunoconjugate.
Collapse
Affiliation(s)
- Jin Chen
- Institute of Chemical Biology & Drug Discovery, State University of New York, Stony Brook, 11794-3400, USA
| | | | | | | | | |
Collapse
|
48
|
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
|