1
|
Liang Y, Lin W, Chen Y, Yang W, Zhou X, Ai S, Qiu L, Cao R, Wang J. Synthesis and in vitro and in vivo evaluation of novel bivalent quinolines as antitumor agents via targeting autophagy in cervical cancer. Eur J Med Chem 2025; 288:117421. [PMID: 39987834 DOI: 10.1016/j.ejmech.2025.117421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
A series of novel bivalent quinolines with a spacer of four to six methylene units between the phenoxy group in the position-7 and various substituents in the position-4 of quinoline skeleton, respectively, were synthesized and evaluated as anticancer agents. The data showed that the majority of the compounds had significant antiproliferative activity with IC50 values less than 50 μM against human cancer cell lines. Among them, compound 4b exhibited the strongest antiproliferative activity against HCT116, A549, BGC823, HeLa and MCF-7 cell lines with an IC50 values of 0.26, 2.75, 4.06, 3.71 and 3.08 μM, respectively. Further studies on the anticancer effects in mice of compound 4b showed its capacity to inhibit tumor growth and markedly reduce tumor size of cervical cancer. Moreover investigation on the underlying mechanism of action indicated that compound 4b didn't trigger apoptotic processes in cervical cancer cell lines, but inhibit cervical cancer growth through inducing autophagy via the ATG5/ATG7 pathway.
Collapse
Affiliation(s)
- Yuexiu Liang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, 510275, PR China; Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, PR China
| | - Wenxian Lin
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Yuzhen Chen
- Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, PR China; Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Baise, 533000, PR China
| | - Weijie Yang
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Xiaoyu Zhou
- Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, PR China; Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Baise, 533000, PR China
| | - Shishen Ai
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Liqin Qiu
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Rihui Cao
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China.
| | - Junli Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, 510275, PR China; Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, PR China.
| |
Collapse
|
2
|
Chang TY, Lin CJ, Wen SN, Wu YC, Wei CY, Huang JY, Tsao YH, Chen YJ, Tang WC, Wu YC, Lee WH, Huang TY, Kuo TM, Li WF, Lai MT. Preclinical evaluation of a novel antibody-drug conjugate OBI-992 for Cancer therapy. Sci Rep 2025; 15:8735. [PMID: 40082588 PMCID: PMC11906863 DOI: 10.1038/s41598-025-92697-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
Trophoblast cell surface antigen 2 (TROP2), a transmembrane glycoprotein highly expressed in a variety of epithelial cancers, has been considered as a primary therapeutic target for the development of antibody-drug conjugates (ADCs). OBI-992, an investigational TROP2-targeted ADC, is composed of a novel TROP2 antibody (R4702) conjugated to the topoisomerase I (TOP1) inhibitor exatecan through a hydrophilic enzyme-cleavable linker. This study aimed to characterize R4702 and OBI-992 in vitro. TROP2-targeted antibodies sacituzumab and datopotamab were employed as the comparators for R4702. ADCs sacituzumab govitecan (SG) and datopotamab deruxtecan (Dato-DXd) were used as benchmarks for OBI-992. Results revealed that R4702 binds to an epitope that is distinct from sacituzumab and datopotamab. The cytotoxicity of the OBI-992, SG, and Dato-DXd against different cancer cells is comparable despite they have different internalization profile. Upregulation of breast cancer resistance protein (BCRP) was observed in SG-resistant and Dato-DXd-resistant cells, but not in OBI-992-resistant cells. In addition, significant downregulation of TROP2 expression was detected with Dato-DXd-resistant cells and only slightly downregulation with SG- and OBI-992-resistant cells was observed. Moreover, substantial enhancement of cytotoxicity and DNA damage was found in the combination of OBI-992 with a poly (ADP-ribose) polymerase (PARP) inhibitor (talazoparib). Taken together, the findings in this study support further clinical development of OBI-992.
Collapse
Affiliation(s)
- Ting-Yu Chang
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Chun-Jung Lin
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Shih-Ni Wen
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Yi-Chen Wu
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Cheng-Yen Wei
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Jye-Yu Huang
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Yu-Hsuan Tsao
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Yu-Jung Chen
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Wei-Chien Tang
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Yuen-Chin Wu
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Wei-Han Lee
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Teng-Yi Huang
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Tzer-Min Kuo
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Wan-Fen Li
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan
| | - Ming-Tain Lai
- OBI Pharma, Inc., 6F, No. 508, Section 7 Zhongxiao East Road, Nangang District, Taipei, Taiwan.
| |
Collapse
|
3
|
Chandrasekhar B, Gor R, Ramalingam S, Thiagarajan A, Sohn H, Madhavan T. Repurposing FDA-approved compounds to target JAK2 for colon cancer treatment. Discov Oncol 2024; 15:226. [PMID: 38869738 DOI: 10.1007/s12672-024-01050-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 05/20/2024] [Indexed: 06/14/2024] Open
Abstract
Colorectal cancer is one of the common cancers worldwide and the second leading cause of cancer-related death. The current treatment has the inherent drawbacks and there is a need of developing a new treatment. Interleukin-6 a pleiotropic cytokine involved in immune regulation and activation of JAK2/STAT3 pathway in colorectal cancer. JAK2/STAT3 signaling pathway functions as a critical regulator of cell growth, differentiation, and immune expression. The abnormality in the JAK2/STAT3 pathway is involved in the tumorigenesis of colon cancer including apoptosis. In this study, we identified novel inhibitors for JAK2 protein by performing virtual screening against FDA-approved compounds. To address the selectivity issue, we implemented cross-docking method followed by DFT calculations to understand the chemical reactivity of the identified compounds. Additionally, molecular dynamics (MD) simulations were performed for the top FDA compounds against JAK2 to understand the molecular interactions and structural stability of the complex over a period of 200 ns. Our results indicated that ergotamine, entrectinib, exatecan, dihydroergotamine, and paritaprevir can be used as alternative drugs for colon cancer. In addition, ergotamine was found to efficiently lower the cell viability with IC50 values of 100 µM on colon cancer cell lines. The long-term inhibitory effect of the ergotamine led to a decrease in colony size, and the toxicity properties were studied using hemolysis assay. Our study shows the potential of targeting JAK2 as a novel approach to colon cancer treatment, and demonstrate that ergotamine as a promising effects as an anti-cancer drug.
Collapse
Affiliation(s)
- Bavya Chandrasekhar
- Computational Biology Laboratory, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Potheri, Chengalpattu District, Kattankulathur, 603203, Tamilnadu, India
| | - Ravi Gor
- Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Potheri, Chengalpattu District, Kattankulathur, 603203, Tamilnadu, India
| | - Satish Ramalingam
- Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Potheri, Chengalpattu District, Kattankulathur, 603203, Tamilnadu, India
| | - Anuradha Thiagarajan
- Deparment of Physics with Computer Application, Agurchand Manmull Jain College, Meenambakam, Chennai, Tamilnadu, India
| | - Honglae Sohn
- Department of Chemistry and Department of Carbon Materials, Chosun University, Gwangju, South Korea.
| | - Thirumurthy Madhavan
- Computational Biology Laboratory, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Potheri, Chengalpattu District, Kattankulathur, 603203, Tamilnadu, India.
| |
Collapse
|
4
|
Chen Z, Liu M, Wang N, Xiao W, Shi J. Unleashing the Potential of Camptothecin: Exploring Innovative Strategies for Structural Modification and Therapeutic Advancements. J Med Chem 2024; 67:3244-3273. [PMID: 38421819 DOI: 10.1021/acs.jmedchem.3c02115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Camptothecin (CPT) is a potent anti-cancer agent targeting topoisomerase I (TOP1). However, CPT has poor pharmacokinetic properties, causes toxicities, and leads to drug resistance, which limit its clinical use. In this paper, to review the current state of CPT research. We first briefly explain CPT's TOP1 inhibition mechanism and the key hurdles in CPT drug development. Then we examine strategies to overcome CPT's limitations through structural modifications and advanced delivery systems. Though modifications alone seem insufficient to fully enhance CPT's therapeutic potential, structure-activity relationship analysis provides insights to guide optimization of CPT analogs. In comparison, advanced delivery systems integrating controlled release, imaging capabilities, and combination therapies via stimulus-responsive linkers and targeting moieties show great promise for improving CPT's pharmacological profile. Looking forward, multifaceted approaches combining selective CPT derivatives with advanced delivery systems, informed by emerging biological insights, hold promise for fully unleashing CPT's anti-cancer potential.
Collapse
Affiliation(s)
- Zheng Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Maoyu Liu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Ningyu Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenjing Xiao
- Department of Pharmacy, The General Hospital of Western Theater Command of PLA, Chengdu 610083, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
5
|
Schöffski P, Wang CC, Schöffski MP, Wozniak A. Current Role of Topoisomerase I Inhibitors for the Treatment of Mesenchymal Malignancies and Their Potential Future Use as Payload of Sarcoma-Specific Antibody-Drug Conjugates. Oncol Res Treat 2023; 47:18-41. [PMID: 38016427 PMCID: PMC10860894 DOI: 10.1159/000535491] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/19/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Topoisomerase I is an enzyme that plays a crucial part in DNA replication and transcription by the relaxation of supercoiled double-stranded DNA. Topoisomerase I inhibitors bind to the topoisomerase I cleavage complex, thereby stabilizing it and preventing the religation of the DNA strands, leading to DNA damage, cell cycle arrest, and apoptosis. Various topoisomerase I inhibitors have been evaluated in solid tumors, and irinotecan and topotecan have been approved for the treatment of epithelial malignancies. None of them have been approved for sarcoma, a diverse group of rare solid tumors with an unmet need for effective treatments. SUMMARY Topoisomerase I inhibitors have been evaluated in preclinical studies as single agents or in combination in solid tumors, some of which have included sarcomas where activity was observed. Clinical trials evaluating topoisomerase I inhibitors for the treatment of sarcoma have shown limited efficacy as monotherapy. In combination with other cytotoxic agents, topoisomerase I inhibitors have become part of clinical routine in selected sarcoma subtypes. Regimens such as irinotecan/vincristine/temozolomide are used in relapsed rhabdomyosarcoma, irinotecan/temozolomide and vincristine/topotecan/cyclophosphamide are commonly given in refractory Ewing sarcoma, and topotecan/carboplatin showed some activity in advanced soft tissue sarcoma. This review provides an overview of key studies with topoisomerase I inhibitors for the treatment of sarcoma. Topoisomerase I inhibitors are currently also being assessed as "payloads" for antibody-drug conjugates (ADCs), allowing for the targeting of specific antigen-expressing tumor cells and the delivery of the inhibitor directly to the tumor cells with the potential of enhancing therapeutic efficacy while minimizing systemic toxicity. Here, we also provide a brief overview on topoisomerase I-ADCs. KEY MESSAGE Topoisomerase I inhibitors are an important component of some systemic therapies for selected sarcomas and have potent cytotoxic properties and pharmacological characteristics that make them relevant candidates as payloads for the development of sarcoma-specific ADCs. ADCs are antibody-based targeted agents allowing for efficient and specific delivery of a given drug to the tumor cell. Topoisomerase I-ADCs are a novel targeted delivery approach which may have the potential to improve the therapeutic index of topoisomerase I inhibitors in the treatment of sarcoma and warrants investigation in a broad variety of mesenchymal malignancies.
Collapse
Affiliation(s)
- Patrick Schöffski
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium
| | - Chao-Chi Wang
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium
| | | | - Agnieszka Wozniak
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Balamkundu S, Liu CF. Lysosomal-Cleavable Peptide Linkers in Antibody-Drug Conjugates. Biomedicines 2023; 11:3080. [PMID: 38002080 PMCID: PMC10669454 DOI: 10.3390/biomedicines11113080] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Antibody-drug Conjugates (ADCs) are a powerful therapeutic modality for cancer treatment. ADCs are multi-functional biologics in which a disease-targeting antibody is conjugated to an effector payload molecule via a linker. The success of currently used ADCs has been largely attributed to the development of linker systems, which allow for the targeted release of cytocidal payload drugs inside cancer cells. Many lysosomal proteases are over expressed in human cancers. They can effectively cleave a variety of peptide sequences, which can be exploited for the design of ADC linker systems. As a well-established linker, valine-citrulline-p-aminobenzyl carbamate (ValCitPABC) is used in many ADCs that are already approved or under preclinical and clinical development. Although ValCitPABC and related linkers are readily cleaved by cathepsins in the lysosome while remaining reasonably stable in human plasma, many studies have shown that they are susceptible to carboxylesterase 1C (Ces1C) in mouse and rat plasma, which hinders the preclinical evaluation of ADCs. Furthermore, neutropenia and thrombocytopenia, two of the most commonly observed dose-limiting adverse effects of ADCs, are believed to result from the premature hydrolysis of ValCitPABC by human neutrophil elastase. In addition to ValCitPABC, the GGFG tetrapeptidyl-aminomethoxy linker is also cathepsin-cleavable and is used in the highly successful ADC drug, DS8201a. In addition to cathepsin-cleavable linkers, there is also growing interest in legumain-sensitive linkers for ADC development. Increasing plasma stability while maintaining lysosomal cleavability of ADC linkers is an objective of intensive current research. This review reports recent advances in the design and structure-activity relationship studies of various peptide/peptidomimetic linkers in this field.
Collapse
Affiliation(s)
| | - Chuan-Fa Liu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
| |
Collapse
|
7
|
Fontaine SD, Carreras CW, Reid RR, Ashley GW, Santi DV. A Very Long-acting Exatecan and Its Synergism with DNA Damage Response Inhibitors. CANCER RESEARCH COMMUNICATIONS 2023; 3:908-916. [PMID: 37377899 PMCID: PMC10208276 DOI: 10.1158/2767-9764.crc-22-0517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/03/2023] [Accepted: 04/18/2023] [Indexed: 06/29/2023]
Abstract
Exatecan (Exa) is a very potent inhibitor of topoisomerase I and anticancer agent. It has been intensively studied as a single agent, a large macromolecular conjugate and as the payload component of antigen-dependent antibody-drug conjugates. The current work describes an antigen-independent conjugate of Exa with polyethylene glycol (PEG) that slowly releases free Exa. Exa was conjugated to a 4-arm 40 kDa PEG through a β-eliminative cleavable linker. Pharmacokinetic studies in mice showed that the conjugate has an apparent circulating half-life of 12 hours, which reflects a composite of both the rate of renal elimination (half-life ∼18 hours) and release of Exa (half-life ∼40 hours). Remarkably, a single low dose of 10 μmol/kg PEG-Exa-only approximately 0.2 μmol/mouse-caused complete suppression of tumor growth of BRCA1-deficient MX-1 xenografts lasting over 40 days. A single low dose of 2.5 μmol/kg PEG-Exa administered with low but efficacious doses of the PARP inhibitor talazoparib showed strong synergy and caused significant tumor regression. Furthermore, the same low, single dose of PEG-Exa administered with the ATR inhibitor VX970 at doses of the DNA damage response inhibitor that do not affect tumor growth show high tumor regression, strong synergy, and synthetic lethality. Significance A circulating conjugate that slowly releases Exa is described. It is efficacious after a single dose and synergistic with ATR and PARP inhibitors.
Collapse
|
8
|
Strzelecka K, Piotrowska U, Sobczak M, Oledzka E. The Advancement of Biodegradable Polyesters as Delivery Systems for Camptothecin and Its Analogues-A Status Report. Int J Mol Sci 2023; 24:ijms24021053. [PMID: 36674567 PMCID: PMC9866533 DOI: 10.3390/ijms24021053] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Camptothecin (CPT) has demonstrated antitumor activity in lung, ovarian, breast, pancreas, and stomach cancers. However, this drug, like many other potent anticancer agents, is extremely water-insoluble. Furthermore, pharmacology studies have revealed that prolonged schedules must be administered continuously. For these reasons, several of its water-soluble analogues, prodrugs, and macromolecular conjugates have been synthesized, and various formulation approaches have been investigated. Biodegradable polyesters have gained popularity in cancer treatment in recent years. A number of biodegradable polymeric drug delivery systems (DDSs), designed for localized and systemic administration of therapeutic agents, as well as tumor-targeting macromolecules, have entered clinical trials, demonstrating the importance of biodegradable polyesters in cancer therapy. Biodegradable polyester-based DDSs have the potential to deliver the payload to the target while also increasing drug availability at intended site. The systemic toxicity and serious side-effects associated with conventional cancer therapies can be significantly reduced with targeted polymeric systems. This review elaborates on the use of biodegradable polyesters in the delivery of CPT and its analogues. The design of various DDSs based on biodegradable polyesters has been described, with the drug either adsorbed on the polymer's surface or encapsulated within its macrostructure, as well as those in which a hydrolyzed chemical bond is formed between the active substance and the polymer chain. The data related to the type of DDSs, the kind of linkage, and the details of in vitro and in vivo studies are included.
Collapse
Affiliation(s)
- Katarzyna Strzelecka
- Department of Analytical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Urszula Piotrowska
- Department of Analytical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Marcin Sobczak
- Department of Analytical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
- Military Institute of Hygiene and Epidemiology, 4 Kozielska Str., 01-163 Warsaw, Poland
| | - Ewa Oledzka
- Department of Analytical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-572-07-55
| |
Collapse
|
9
|
Synthesis of novel 4,7-disubstituted quinoline derivatives as autophagy inducing agents via targeting stabilization of ATG5. Bioorg Chem 2022; 127:105998. [DOI: 10.1016/j.bioorg.2022.105998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 06/03/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022]
|
10
|
Jo U, Murai Y, Agama KK, Sun Y, Saha LK, Yang X, Arakawa Y, Gayle S, Jones K, Paralkar V, Sundaram RK, Doorn JV, Vasquez JC, Bindra RS, Choi WS, Pommier Y. TOP1-DNA Trapping by Exatecan and Combination Therapy with ATR Inhibitor. Mol Cancer Ther 2022; 21:1090-1102. [PMID: 35439320 PMCID: PMC9256811 DOI: 10.1158/1535-7163.mct-21-1000] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/18/2022] [Accepted: 04/12/2022] [Indexed: 01/07/2023]
Abstract
Exatecan and deruxtecan are antineoplastic camptothecin derivatives in development as tumor-targeted-delivery warheads in various formulations including peptides, liposomes, polyethylene glycol nanoparticles, and antibody-drug conjugates. Here, we report the molecular pharmacology of exatecan compared with the clinically approved topoisomerase I (TOP1) inhibitors and preclinical models for validating biomarkers and the combination of exatecan with ataxia telangiectasia and Rad3-related kinase (ATR) inhibitors. Modeling exatecan binding at the interface of a TOP1 cleavage complex suggests two novel molecular interactions with the flanking DNA base and the TOP1 residue N352, in addition to the three known interactions of camptothecins with the TOP1 residues R364, D533, and N722. Accordingly, exatecan showed much stronger TOP1 trapping, higher DNA damage, and apoptotic cell death than the classical TOP1 inhibitors used clinically. We demonstrate the value of SLFN11 expression and homologous recombination (HR) deficiency (HRD) as predictive biomarkers of response to exatecan. We also show that exatecan kills cancer cells synergistically with the clinical ATR inhibitor ceralasertib (AZD6738). To establish the translational potential of this combination, we tested CBX-12, a clinically developed pH-sensitive peptide-exatecan conjugate that selectively targets cancer cells and is currently in clinical trials. The combination of CBX-12 with ceralasertib significantly suppressed tumor growth in mouse xenografts. Collectively, our results demonstrate the potency of exatecan as a TOP1 inhibitor and its clinical potential in combination with ATR inhibitors, using SLFN11 and HRD as predictive biomarkers.
Collapse
Affiliation(s)
- Ukhyun Jo
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yasuhisa Murai
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Keli K. Agama
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yilun Sun
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Liton Kumar Saha
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Xi Yang
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yasuhiro Arakawa
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Kelli Jones
- Cybrexa Therapeutics, New Haven, CT 06511, USA
| | | | - Ranjini K. Sundaram
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Jinny Van Doorn
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Juan C. Vasquez
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06511, USA
| | - Ranjit S. Bindra
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Woo Suk Choi
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
11
|
Chia CSB. A Patent Review on FDA-Approved Antibody-Drug Conjugates, Their Linkers and Drug Payloads. ChemMedChem 2022; 17:e202200032. [PMID: 35384350 DOI: 10.1002/cmdc.202200032] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/22/2022] [Indexed: 12/30/2022]
Abstract
Antibody-drug conjugates (ADCs) have emerged as a promising class of biologics since the first approval of Gemtuzumab ozogamicin in 2000. Compared to small molecule drugs, ADCs are structurally much more complex as they comprise of an antibody conjugated to cytotoxic payloads by specially-designed linkers. Correspondingly, the ADC patent landscape is also much more complex. This review collates and discusses the patents protecting ADCs approved by the FDA up to 31 December 2021, with particular emphasis on their linker and cytotoxin payload technologies.
Collapse
Affiliation(s)
- C S Brian Chia
- Experimental Drug Development Centre, 10 Biopolis Road, Chromos #08-01, 138670, Singapore, Singapore
| |
Collapse
|
12
|
Martín-Encinas E, Selas A, Palacios F, Alonso C. The design and discovery of topoisomerase I inhibitors as anticancer therapies. Expert Opin Drug Discov 2022; 17:581-601. [PMID: 35321631 DOI: 10.1080/17460441.2022.2055545] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Cancer has been identified as one of the leading causes of death worldwide. The biological target of some anticancer agents is topoisomerase I, an enzyme involved in the relaxation of supercoiled DNA. The synthesis of new compounds with antiproliferative effect and behaving as topoisomerase I inhibitors has become an active field of research. Depending on their mechanism of inhibition, they can be classified as catalytic inhibitors or poisons. AREAS COVERED This review article summarizes the state of the art for the development of selective topoisomerase I inhibitors. Collected compounds showed inhibition of the enzyme, highlighting those approved for clinical use, the combination therapies developed, as well as related drawbacks and future focus. EXPERT OPINION Research related to topoisomerase I inhibitors in cancer therapy started with camptothecin (CPT). This compound was first selected as a good anticancer agent and then topoisomerase I was identified as its therapeutic target. Derivatives of CPT irinotecan, topotecan, and belotecan are the only clinically approved inhibitors. Currently, their limitations are being addressed by different stretegies. Future studies should focus not only on developing other active molecules but also on improving the bioavailability and pharmacokinetics of potent synthetic derivatives.
Collapse
Affiliation(s)
- Endika Martín-Encinas
- Departamento de Química Orgánica I - Centro de Investigación Lascaray, Facultad de Farmacia, Universidad del País Vasco, Paseo de la Universidad 7, 01006 Vitoria, Spain
| | - Asier Selas
- Departamento de Química Orgánica I - Centro de Investigación Lascaray, Facultad de Farmacia, Universidad del País Vasco, Paseo de la Universidad 7, 01006 Vitoria, Spain
| | - Francisco Palacios
- Departamento de Química Orgánica I - Centro de Investigación Lascaray, Facultad de Farmacia, Universidad del País Vasco, Paseo de la Universidad 7, 01006 Vitoria, Spain
| | - Concepción Alonso
- Departamento de Química Orgánica I - Centro de Investigación Lascaray, Facultad de Farmacia, Universidad del País Vasco, Paseo de la Universidad 7, 01006 Vitoria, Spain
| |
Collapse
|
13
|
Reddy DS, Novitskiy IM, Kutateladze AG. Maximizing Step‐Normalized Increases in Molecular Complexity: Formal [4+2+2+2] Photoinduced Cyclization Cascade to Access Polyheterocycles Possessing Privileged Substructures. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202112573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- D. Sai Reddy
- Department of Chemistry and Biochemistry University of Denver 2190 E. Iliff Ave. Denver CO 80208 USA
| | - Ivan M. Novitskiy
- Department of Chemistry and Biochemistry University of Denver 2190 E. Iliff Ave. Denver CO 80208 USA
| | - Andrei G. Kutateladze
- Department of Chemistry and Biochemistry University of Denver 2190 E. Iliff Ave. Denver CO 80208 USA
| |
Collapse
|
14
|
Zhang X, Huang AC, Chen F, Chen H, Li L, Kong N, Luo W, Fang J. Novel development strategies and challenges for anti-Her2 antibody-drug conjugates. Antib Ther 2022; 5:18-29. [PMID: 35146330 PMCID: PMC8826051 DOI: 10.1093/abt/tbac001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 12/16/2021] [Accepted: 01/02/2022] [Indexed: 11/24/2022] Open
Abstract
Antibody-drug conjugates (ADCs) combining potent cytotoxicity of small-molecule drugs with the selectivity and excellent pharmacokinetic profile of monoclonal antibody (mAb) are promising therapeutic modalities for a diverse range of cancers. Owing to overexpression in a wide range of tumors, human epidermal growth factor receptor 2 (Her2) is one of the most utilized targeting antigens for ADCs to treat Her2-positive cancers. Owing to the high density of Her2 antigens on the tumor cells and high affinity and high internalization capacity of corresponding antibodies, 56 anti-Her2 ADCs which applied >10 different types of novel payloads had entered preclinical or clinical trials. Seven of 12 Food and Drug Administration (FDA)-approved ADCs including Polivy (2019), Padcev (2019), EnHertu (2019), Trodelvy (2020), Blenrep (2020), Zynlonta (2021), and Tivdak) (2021) have been approved by FDA in the past three years alone, indicating that the maturing of ADC technology brings more productive clinical outcomes. This review, focusing on the anti-Her2 ADCs in clinical trials or on the market, discusses the strategies to select antibody formats, the linkages between linker and mAb, and effective payloads with particular release and action mechanisms for a good clinical outcome.
Collapse
Affiliation(s)
- Xinling Zhang
- ADC R&D Department, RemeGen Co., Ltd, 58 Middle Beijing Road, Yantai, ShanDong 264006, China
| | - Andrew C Huang
- Innovation Research Center, MabPlex International Ltd, 60 Middle Beijing Road, Yantai, ShanDong 264006, China
| | - Fahai Chen
- CEO Office, RemeGen Co., Ltd, 58 Middle Beijing Road, Yantai, ShanDong 264006, China
| | - Hu Chen
- ADC R&D Department, RemeGen Co., Ltd, 58 Middle Beijing Road, Yantai, ShanDong 264006, China
| | - Lele Li
- Innovation Research Center, MabPlex International Ltd, 60 Middle Beijing Road, Yantai, ShanDong 264006, China
| | - Nana Kong
- Innovation Research Center, MabPlex International Ltd, 60 Middle Beijing Road, Yantai, ShanDong 264006, China
| | - Wenting Luo
- ADC R&D Department, RemeGen Co., Ltd, 58 Middle Beijing Road, Yantai, ShanDong 264006, China
| | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| |
Collapse
|
15
|
Díaz-Rodríguez E, Gandullo-Sánchez L, Ocaña A, Pandiella A. Novel ADCs and Strategies to Overcome Resistance to Anti-HER2 ADCs. Cancers (Basel) 2021; 14:154. [PMID: 35008318 PMCID: PMC8750930 DOI: 10.3390/cancers14010154] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/27/2021] [Accepted: 12/27/2021] [Indexed: 12/23/2022] Open
Abstract
During recent years, a number of new compounds against HER2 have reached clinics, improving the prognosis and quality of life of HER2-positive breast cancer patients. Nonetheless, resistance to standard-of-care drugs has motivated the development of novel agents, such as new antibody-drug conjugates (ADCs). The latter are a group of drugs that benefit from the potency of cytotoxic agents whose action is specifically guided to the tumor by the target-specific antibody. Two anti-HER2 ADCs have reached the clinic: trastuzumab-emtansine and, more recently, trastuzumab-deruxtecan. In addition, several other HER2-targeted ADCs are in preclinical or clinical development, some of them with promising signs of activity. In the present review, the structure, mechanism of action, and potential resistance to all these ADCs will be described. Specific attention will be given to discussing novel strategies to circumvent resistance to ADCs.
Collapse
Affiliation(s)
- Elena Díaz-Rodríguez
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-IBSAL and CIBERONC, 37007 Salamanca, Spain; (E.D.-R.); (L.G.-S.)
- Departamento de Bioquímica y Biología Molecular, University of Salamanca, 37007 Salamanca, Spain
| | - Lucía Gandullo-Sánchez
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-IBSAL and CIBERONC, 37007 Salamanca, Spain; (E.D.-R.); (L.G.-S.)
| | - Alberto Ocaña
- Hospital Clínico San Carlos, Centro de Investigación Biomédica en Red de Oncología (CIBERONC), 28040 Madrid, Spain;
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-IBSAL and CIBERONC, 37007 Salamanca, Spain; (E.D.-R.); (L.G.-S.)
| |
Collapse
|
16
|
Reddy DS, Novitskiy IM, Kutateladze AG. Maximizing Step-Normalized Increases in Molecular Complexity: Formal [4+2+2+2] Photoinduced Cyclization Cascade to Access Polyheterocycles Possessing Privileged Substructures. Angew Chem Int Ed Engl 2021; 61:e202112573. [PMID: 34850525 DOI: 10.1002/anie.202112573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Indexed: 11/10/2022]
Abstract
A new complexity building photoinduced cascade which amounts to an unprecedented formal [4+2+2+2] cycloaddition topology is developed to access complex nitrogen polyheterocycles. This photocascade is initiated by the excited state intramolecular proton transfer (ESIPT) in aromatic amino ketones with tethered dual unsaturated pendants, i.e. pyrrole and alkenic moieties, resulting in the formation of four σ-bonds and setting six new stereogenic centers in a single experimentally simple photochemical step with up to 220 mcbit complexity increases.
Collapse
Affiliation(s)
- D Sai Reddy
- Department of Chemistry and Biochemistry, University of Denver, 2190 E. Iliff Ave., Denver, CO 80208, USA
| | - Ivan M Novitskiy
- Department of Chemistry and Biochemistry, University of Denver, 2190 E. Iliff Ave., Denver, CO 80208, USA
| | - Andrei G Kutateladze
- Department of Chemistry and Biochemistry, University of Denver, 2190 E. Iliff Ave., Denver, CO 80208, USA
| |
Collapse
|
17
|
Gayle S, Aiello R, Leelatian N, Beckta JM, Bechtold J, Bourassa P, Csengery J, Maguire RJ, Marshall D, Sundaram RK, Van Doorn J, Jones K, Moore H, Lopresti-Morrow L, Paradis T, Tylaska L, Zhang Q, Visca H, Reshetnyak YK, Andreev OA, Engelman DM, Glazer PM, Bindra RS, Paralkar VM. Tumor-selective, antigen-independent delivery of a pH sensitive peptide-topoisomerase inhibitor conjugate suppresses tumor growth without systemic toxicity. NAR Cancer 2021; 3:zcab021. [PMID: 34316708 PMCID: PMC8210154 DOI: 10.1093/narcan/zcab021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 05/03/2021] [Accepted: 05/19/2021] [Indexed: 02/04/2023] Open
Abstract
Topoisomerase inhibitors are potent DNA damaging agents which are widely used in oncology, and they demonstrate robust synergistic tumor cell killing in combination with DNA repair inhibitors, including poly(ADP)-ribose polymerase (PARP) inhibitors. However, their use has been severely limited by the inability to achieve a favorable therapeutic index due to severe systemic toxicities. Antibody-drug conjugates address this issue via antigen-dependent targeting and delivery of their payloads, but this approach requires specific antigens and yet still suffers from off-target toxicities. There is a high unmet need for a more universal tumor targeting technology to broaden the application of cytotoxic payloads. Acidification of the extracellular milieu arises from metabolic adaptions associated with the Warburg effect in cancer. Here we report the development of a pH-sensitive peptide-drug conjugate to deliver the topoisomerase inhibitor, exatecan, selectively to tumors in an antigen-independent manner. Using this approach, we demonstrate potent in vivo cytotoxicity, complete suppression of tumor growth across multiple human tumor models, and synergistic interactions with a PARP inhibitor. These data highlight the identification of a peptide-topoisomerase inhibitor conjugate for cancer therapy that provides a high therapeutic index, and is applicable to all types of human solid tumors in an antigen-independent manner.
Collapse
Affiliation(s)
| | | | - Nalin Leelatian
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jason M Beckta
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | - Ranjini K Sundaram
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jinny Van Doorn
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Kelli Jones
- Cybrexa Therapeutics, New Haven, CT 06511, USA
| | | | | | | | | | - Qing Zhang
- Cybrexa Therapeutics, New Haven, CT 06511, USA
| | - Hannah Visca
- Physics Department, University of Rhode Island, Kingston, RI 02881, USA
| | - Yana K Reshetnyak
- Physics Department, University of Rhode Island, Kingston, RI 02881, USA
| | - Oleg A Andreev
- Physics Department, University of Rhode Island, Kingston, RI 02881, USA
| | - Donald M Engelman
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Peter M Glazer
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ranjit S Bindra
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|
18
|
Lüftner D, Peipp M. New Therapeutic Strategies in Advanced Nonoperable or Metastatic HER2-positive Breast Cancer. Geburtshilfe Frauenheilkd 2021; 81:666-678. [PMID: 34168380 PMCID: PMC8216785 DOI: 10.1055/a-1471-4063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 03/27/2021] [Indexed: 11/24/2022] Open
Abstract
Despite therapeutic gains in the treatment of HER2-positive (HER2: human epidermal growth factor receptor 2) advanced/metastatic breast cancer, there remains an urgent need for more effective treatment options. At present, there is no definitive approved standard therapy beyond second-line treatment. One of the major challenges is overcoming treatment resistance. Depending on the underlying resistance mechanism, different strategies are being pursued for new innovative treatment concepts in HER2-positive breast cancer. Specifically designed antibodies for targeted therapy are one important focus to successfully meet these challenges. Trastuzumab deruxtecan (T-DXd, DS-8201a), an optimised antibody drug conjugate (ADC) is in clinical trials, showing promising outcomes in patients with advanced, nonoperable or metastatic HER2-positive breast cancer who had already undergone intensive prior treatment. Based on this data, T-DXd has already been approved in the US and Japan for HER2-positive advanced nonoperable and metastatic breast cancer - in the US after at least two prior anti-HER2 targeted treatment lines and in Japan after prior chemotherapy. T-DXd represents successful "antibody engineering". Since the beginning of the year, T-DXd has also been approved in Europe as monotherapy for inoperable or metastatic HER2-positive breast cancer in patients who are pretreated with at least two anti-HER2 directed therapies. This paper presents strategies for improving treatment options in advanced nonoperable and metastatic HER2-positive breast cancer, with the development of T-DXd as an example.
Collapse
Affiliation(s)
- Diana Lüftner
- Medizinische Klinik mit Schwerpunkt Hämatologie, Onkologie und Tumorimmunologie, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Peipp
- Sektion für Stammzelltransplantation und Immuntherapie, Dr. Mildred-Scheel-Haus, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
19
|
Exatecan Antibody Drug Conjugates Based on a Hydrophilic Polysarcosine Drug-Linker Platform. Pharmaceuticals (Basel) 2021; 14:ph14030247. [PMID: 33803327 PMCID: PMC8000490 DOI: 10.3390/ph14030247] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 12/20/2022] Open
Abstract
We herein report the development and evaluation of a novel HER2-targeting antibody-drug conjugate (ADC) based on the topoisomerase I inhibitor payload exatecan, using our hydrophilic monodisperse polysarcosine (PSAR) drug-linker platform (PSARlink). In vitro and in vivo experiments were conducted in breast and gastric cancer models to characterize this original ADC and gain insight about the drug-linker structure-activity relationship. The inclusion of the PSAR hydrophobicity masking entity efficiently reduced the overall hydrophobicity of the conjugate and yielded an ADC sharing the same pharmacokinetic profile as the unconjugated antibody despite the high drug-load of the camptothecin-derived payload (drug-antibody ratio of 8). Tra-Exa-PSAR10 demonstrated strong anti-tumor activity at 1 mg/kg in an NCI-N87 xenograft model, outperforming the FDA-approved ADC DS-8201a (Enhertu), while being well tolerated in mice at a dose of 100 mg/kg. In vitro experiments showed that this exatecan-based ADC demonstrated higher bystander killing effect than DS-8201a and overcame resistance to T-DM1 (Kadcyla) in preclinical HER2+ breast and esophageal models, suggesting potential activity in heterogeneous and resistant tumors. In summary, the polysarcosine-based hydrophobicity masking approach allowsfor the generation of highly conjugated exatecan-based ADCs having excellent physicochemical properties, an improved pharmacokinetic profile, and potent in vivo anti-tumor activity.
Collapse
|
20
|
Nguyen X, Hooper M, Borlagdan JP, Palumbo A. A Review of Fam-Trastuzumab Deruxtecan-nxki in HER2-Positive Breast Cancer. Ann Pharmacother 2021; 55:1410-1418. [PMID: 33629601 DOI: 10.1177/1060028021998320] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE To review the pharmacology, efficacy, and safety of antibody-drug conjugate fam-trastuzumab deruxtecan-nxki in the treatment of advanced, unresectable, or metastatic breast cancer. DATA SOURCES Relevant information was identified through a MEDLINE/PubMed (January 2015 to December 2020) literature search. The new drug application, prescribing information, clinical practice guideline, and abstracts from scientific meetings were also reviewed. STUDY SELECTION AND DATA EXTRACTION The literature search was limited to human studies published in the English language. All studies evaluating the pharmacology, efficacy, or safety of fam-trastuzumab deruxtecan-nxki for breast cancer were included. DATA SYNTHESIS Fam-trastuzumab deruxtecan-nxki is composed of an anti-human epidermal growth factor receptor 2 (HER2) antibody and topoisomerase I inhibitor (DXd), which causes DNA damage and apoptotic cell death. Major phase I and phase II clinical trials established the efficacy and safety of fam-trastuzumab deruxtecan-nxki for treatment of HER2-positive advanced, unresectable, or metastatic breast cancers that are refractory or intolerant to standard treatment. In these trials, the response rate was 60.9% (95% CI = 53.4-68.0) Common adverse effects included fatigue, nausea, vomiting, decreased appetite, constipation, diarrhea, alopecia, neutropenia, anemia, and thrombocytopenia. Serious adverse effects included interstitial lung disease or pneumonia, febrile neutropenia, left ventricular dysfunction, and embryo-fetal toxicity. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE Fam-trastuzumab deruxtecan-nxki is an option for HER2-positive breast cancer following 2 previous lines of HER2-targeted therapy. CONCLUSIONS Fam-trastuzumab deruxtecan-nxki is an effective treatment for HER2-positive breast cancer in the metastatic setting, but randomized controlled trials are needed.
Collapse
|
21
|
Wu ZC, Boger DL. The quest for supernatural products: the impact of total synthesis in complex natural products medicinal chemistry. Nat Prod Rep 2020; 37:1511-1531. [PMID: 33169762 PMCID: PMC7678878 DOI: 10.1039/d0np00060d] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Covering: 2000 up to 2020This review presents select recent advances in the medicinal chemistry of complex natural products that are prepared by total synthesis. The underlying studies highlight enabling divergent synthetic strategies and methods that permit the systematic medicinal chemistry studies of key analogues bearing deep-seated structural changes not readily accessible by semisynthetic or biosynthetic means. Select and recent examples are detailed where the key structural changes are designed to improve defined properties or to overcome an intrinsic limitation of the natural product itself. In the examples presented, the synthetic efforts provided supernatural products, a term first introduced by our colleague Ryan Shenvi (Synlett, 2016, 27, 1145-1164), with properties superseding the parent natural product. The design principles and approaches for creating the supernatural products are highlighted with an emphasis on the properties addressed that include those that improve activity or potency, increase selectivity, enhance durability, broaden the spectrum of activity, improve chemical or metabolic stability, overcome limiting physical properties, add mechanisms of action, enhance PK properties, overcome drug resistance, and/or improve in vivo efficacy. Some such improvements may be regarded by some as iterative enhancements whereas others, we believe, truly live up to their characterization as supernatural products. Most such efforts are also accompanied by advances in synthetic organic chemistry, inspiring the development of new synthetic methodology and providing supernatural products with improved synthetic accessibility.
Collapse
Affiliation(s)
- Zhi-Chen Wu
- Department of Chemistry, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| | | |
Collapse
|
22
|
Andrikopoulou A, Zografos E, Liontos M, Koutsoukos K, Dimopoulos MA, Zagouri F. Trastuzumab Deruxtecan (DS-8201a): The Latest Research and Advances in Breast Cancer. Clin Breast Cancer 2020; 21:e212-e219. [PMID: 32917537 DOI: 10.1016/j.clbc.2020.08.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/21/2020] [Accepted: 08/13/2020] [Indexed: 01/05/2023]
Abstract
The development of antibody-drug conjugates composed of a cytotoxic agent and a monoclonal antibody carrier offers an important alternative to classic chemotherapy strategies. Trastuzumab deruxtecan (DS-8201a) is a next-generation antibody-drug conjugate composed of a monoclonal anti-HER2 antibody and a topoisomerase I inhibitor, an exatecan derivative (DX-8951f). DS-8201a resulted in favorable outcomes in HER2-positive heavily pretreated breast cancer patients and also had a promising efficacy in patients with HER2-negative/low-expressing disease, whose options are limited. Interestingly, a recently published phase 2 trial (NCT03248492) reported 60% overall response and 97% disease control in patients with HER2-positive disease previously treated with multiple regimens, including trastuzumab emtansine. On the basis of recent clinical trials, the US Food and Drug Administration granted accelerated approval to DS-8201a in advanced or unresectable HER2-positive breast cancer pretreated with at least two HER2-targeting treatment lines. We review all preclinical and clinical data of DS-8201a regarding breast cancer.
Collapse
Affiliation(s)
| | - Eleni Zografos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, Athens, Greece
| | - Michalis Liontos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, Athens, Greece
| | | | | | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, Athens, Greece.
| |
Collapse
|
23
|
Bispecific Antibodies and Antibody-Drug Conjugates for Cancer Therapy: Technological Considerations. Biomolecules 2020; 10:biom10030360. [PMID: 32111076 PMCID: PMC7175114 DOI: 10.3390/biom10030360] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/20/2020] [Accepted: 02/23/2020] [Indexed: 01/07/2023] Open
Abstract
The ability of monoclonal antibodies to specifically bind a target antigen and neutralize or stimulate its activity is the basis for the rapid growth and development of the therapeutic antibody field. In recent years, traditional immunoglobulin antibodies have been further engineered for better efficacy and safety, and technological developments in the field enabled the design and production of engineered antibodies capable of mediating therapeutic functions hitherto unattainable by conventional antibody formats. Representative of this newer generation of therapeutic antibody formats are bispecific antibodies and antibody–drug conjugates, each with several approved drugs and dozens more in the clinical development phase. In this review, the technological principles and challenges of bispecific antibodies and antibody–drug conjugates are discussed, with emphasis on clinically validated formats but also including recent developments in the fields, many of which are expected to significantly augment the current therapeutic arsenal against cancer and other diseases with unmet medical needs.
Collapse
|
24
|
Birrer MJ, Moore KN, Betella I, Bates RC. Antibody-Drug Conjugate-Based Therapeutics: State of the Science. J Natl Cancer Inst 2020; 111:538-549. [PMID: 30859213 DOI: 10.1093/jnci/djz035] [Citation(s) in RCA: 286] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/21/2019] [Accepted: 03/08/2019] [Indexed: 02/06/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are complex engineered therapeutics consisting of monoclonal antibodies, directed toward tumor-associated antigens, to which highly potent cytotoxic agents are attached using chemical linkers. This targeted drug delivery strategy couples the precision of the antibody targeting moiety with the cytocidal activity of the payload, which is generally too toxic on its own to be systemically administered. In this manner, ADCs confer a means to reduce off-target toxicities in patients by limiting the exposure of normal tissues to the payload, thus broadening the potential therapeutic window compared with traditional chemotherapy. The pace of ADC development is accelerating, with the number of investigational agents in human trials having more than tripled over the past 5 years, underscoring the enthusiasm for this transformative approach to cancer treatment. Here, we review the key structural elements of ADC design (antibody, linker, and payload), highlighting critical aspects and technological advances that have affected the clinical effectiveness of this class of biopharmaceuticals. The ADC field continues to evolve, including ongoing efforts aimed at improving target selection, developing payloads with varied mechanisms of action and increased potency, designing innovative bioconjugation strategies, as well as maximizing efficacy and tolerability in patients. An overview of the current clinical trial landscape is provided, with emphasis on the clinical experience of the four ADCs to have received regulatory approval to date, as well as additional promising candidates currently in late-stage clinical development in both solid tumor and hematological malignancies.
Collapse
Affiliation(s)
- Michael J Birrer
- Division of Hematology-Oncology, University of Alabama at Birmingham Comprehensive Cancer Center, Birmingham, AL
| | - Kathleen N Moore
- Stephenson Department of Obstetrics and Gynecology, Oklahoma Cancer Center at the University of Oklahoma Health Sciences Center, Oklahoma City, OK.,Sarah Cannon Research Institute, Nashville, TN
| | - Ilaria Betella
- Division of Hematology-Oncology, University of Alabama at Birmingham Comprehensive Cancer Center, Birmingham, AL
| | | |
Collapse
|
25
|
Agarwal G, Carcache PJB, Addo EM, Kinghorn AD. Current status and contemporary approaches to the discovery of antitumor agents from higher plants. Biotechnol Adv 2020; 38:107337. [PMID: 30633954 PMCID: PMC6614024 DOI: 10.1016/j.biotechadv.2019.01.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 01/03/2019] [Accepted: 01/07/2019] [Indexed: 12/13/2022]
Abstract
Higher plant constituents have afforded clinically available anticancer drugs. These include both chemically unmodified small molecules and their synthetic derivatives currently used or those in clinical trials as antineoplastic agents, and an updated summary is provided. In addition, botanical dietary supplements, exemplified by mangosteen and noni constituents, are also covered as potential cancer chemotherapeutic agents. Approaches to metabolite purification, rapid dereplication, and biological evaluation including analytical hyphenated techniques, molecular networking, and advanced cellular and animal models are discussed. Further, enhanced and targeted drug delivery systems for phytochemicals, including micelles, nanoparticles and antibody drug conjugates (ADCs) are described herein.
Collapse
Affiliation(s)
- Garima Agarwal
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Peter J Blanco Carcache
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Ermias Mekuria Addo
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
26
|
Xu Z, Guo D, Jiang Z, Tong R, Jiang P, Bai L, Chen L, Zhu Y, Guo C, Shi J, Yu D. Novel HER2-Targeting Antibody-Drug Conjugates of Trastuzumab Beyond T-DM1 in Breast Cancer: Trastuzumab Deruxtecan(DS-8201a) and (Vic-)Trastuzumab Duocarmazine (SYD985). Eur J Med Chem 2019; 183:111682. [PMID: 31563805 DOI: 10.1016/j.ejmech.2019.111682] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/21/2022]
Abstract
Targeted drug delivery has improved cancer treatment significantly in recent years, although it is difficult to achieve. Different approaches have been developed to apply targeted drug delivery. Among which, antibody-drug conjugate (ADC) provides a potentially ideal solution to such a challenge. ADC is an innovative drug treatment model with three key components: payload, monoclonal antibody, and linker. The monoclonal antibody targets the antigen-expressing tumor cells and internalizes the payload linked by the linker to the target cells to reduce the side effects of the traditional chemotherapy drugs. The off-target effect has an excellent therapeutic prospect. Among them, ado-trastuzumab emtansine (T-DM1) is a successful example of targeting human epidermal growth factor receptor-2 (HER2). Its antibody (trastuzumab) is derived from Herceptin with annual sales of more than $6 billion. It has excellent targeting and specific anti-tumor activity against HER2. Its linker is not cleavable and releases the Lys-linker-payload to kill the cells. The two ADCs described here use the same antibody as T-DM1, but the cleavable linker and the more toxic payload allow them to have the not only targeting of T-DM1, but also the reduce T-DM1 resistance and improve efficacy in heterogeneous tumors. This paper describes the mechanism of action and the biochemical characteristics of different parts and preclinical and clinical progress of trastuzumab deruxtecan(DS-8201a) and (vic-)trastuzumab duocarmazine (SYD985).
Collapse
Affiliation(s)
- Zhuyu Xu
- Department of Pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dandan Guo
- Department of Pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | | | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Peidu Jiang
- Department of Pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lan Bai
- Department of Pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Chen
- Department of Pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuxuan Zhu
- Department of Pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chun Guo
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, China.
| | - Jianyou Shi
- Department of Pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Dongke Yu
- Department of Pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
27
|
Nakada T, Sugihara K, Jikoh T, Abe Y, Agatsuma T. The Latest Research and Development into the Antibody-Drug Conjugate, [fam-] Trastuzumab Deruxtecan (DS-8201a), for HER2 Cancer Therapy. Chem Pharm Bull (Tokyo) 2019; 67:173-185. [PMID: 30827997 DOI: 10.1248/cpb.c18-00744] [Citation(s) in RCA: 259] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A major limitation of traditional chemotherapy for cancer is dose-limiting toxicity, caused by the exposure of non-tumor cells to cytotoxic agents. Use of molecular targeted drugs, such as specific kinase inhibitors and monoclonal antibodies, is a possible solution to overcome this limitation and has achieved clinical success so far. Use of an antibody-drug conjugate (ADC) is a rational strategy for improving efficacy and reducing systemic adverse events. ADCs use antibodies selectively to deliver a potent cytotoxic agent to tumor cells, thus drastically improving the therapeutic index of chemotherapeutic agents. Lessons learned from clinical failure of early ADCs during the 1980s to 90s have recently led to improvements in ADC technology, and resulted in the approval of four novel ADCs. Nonetheless, further advances in ADC technology are still required to streamline their clinical efficacy and reduce toxicity. [fam-] Trastuzumab deruxtecan (DS-8201a) is a next-generation ADC that satisfies these requirements based on currently available evidence. DS-8201a has several innovative features; a highly potent novel payload with a high drug-to-antibody ratio, good homogeneity, a tumor-selective cleavable linker, stable linker-payload in circulation, and a short systemic half-life cytotoxic agent in vivo; the released cytotoxic payload could exert a bystander effect. With respect to its preclinical profiles, DS-8201a could provide a valuable therapy with a great potential against HER2-expressing cancers in clinical settings. In a phase I trial, DS-8201a showed acceptable safety profiles with potential therapeutic efficacy, with the wide therapeutic index.
Collapse
Affiliation(s)
- Takashi Nakada
- Research and Development Division, Daiichi Sankyo Co., Ltd
| | | | - Takahiro Jikoh
- Research and Development Division, Daiichi Sankyo Co., Ltd
| | - Yuki Abe
- Research and Development Division, Daiichi Sankyo Co., Ltd
| | | |
Collapse
|
28
|
Kumar V, Kumar V, McGuire T, Coulter DW, Sharp JG, Mahato RI. Challenges and Recent Advances in Medulloblastoma Therapy. Trends Pharmacol Sci 2017; 38:1061-1084. [PMID: 29061299 DOI: 10.1016/j.tips.2017.09.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/19/2017] [Accepted: 09/25/2017] [Indexed: 10/18/2022]
Abstract
Medulloblastoma (MB) is the most common childhood brain tumor, which occurs in the posterior fossa. MB tumors are highly heterogeneous and have diverse genetic make-ups, with differential microRNA (miRNA) expression profiles and variable prognoses. MB can be classified into four subgroups, each with different origins, pathogenesis, and potential therapeutic targets. miRNA and small-molecule targeted therapies have emerged as a potential new therapeutic paradigm in MB treatment. However, the development of chemoresistance due to surviving cancer stem cells and dysregulation of miRNAs remains a challenge. Combination therapies using multiple drugs and miRNAs could be effective approaches. In this review we discuss various MB subtypes, barriers, and novel therapeutic options which may be less toxic than current standard treatments.
Collapse
Affiliation(s)
- Vinod Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Timothy McGuire
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Donald W Coulter
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - John G Sharp
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
29
|
Safety, pharmacokinetics, and antitumour activity of trastuzumab deruxtecan (DS-8201), a HER2-targeting antibody-drug conjugate, in patients with advanced breast and gastric or gastro-oesophageal tumours: a phase 1 dose-escalation study. Lancet Oncol 2017; 18:1512-1522. [PMID: 29037983 DOI: 10.1016/s1470-2045(17)30604-6] [Citation(s) in RCA: 355] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/26/2017] [Accepted: 08/01/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Antibody-drug conjugates have emerged as a powerful strategy in cancer therapy and combine the ability of monoclonal antibodies to specifically target tumour cells with the highly potent killing activity of drugs with payloads too toxic for systemic administration. Trastuzumab deruxtecan (also known as DS-8201) is an antibody-drug conjugate comprised of a humanised antibody against HER2, a novel enzyme-cleavable linker, and a topoisomerase I inhibitor payload. We assessed its safety and tolerability in patients with advanced breast and gastric or gastro-oesophageal tumours. METHODS This was an open-label, dose-escalation phase 1 trial done at two study sites in Japan. Eligible patients were at least 20 years old with breast or gastric or gastro-oesophageal carcinomas refractory to standard therapy regardless of HER2 status. Participants received initial intravenous doses of trastuzumab deruxtecan from 0·8 to 8·0 mg/kg and dose-limiting toxicities were assessed over a 21-day cycle; thereafter, dose reductions were implemented as needed and patients were treated once every 3 weeks until they had unacceptable toxic effects or their disease progressed. Primary endpoints included identification of safety and the maximum tolerated dose or recommended phase 2 dosing and were analysed in all participants who received at least one dose of study drug. The dose-escalation study is the first part of a two-part study with the second dose-expansion part ongoing and enrolling patients as of July 8, 2017, in Japan and the USA. This trial is registered at ClinicalTrials.gov, number NCT02564900. FINDINGS Between Aug 28, 2015, and Aug 26, 2016, 24 patients were enrolled and received trastuzumab deruxtecan (n=3 for each of 0·8, 1·6, 3·2, and 8·0 mg/kg doses; n=6 for each of 5·4 and 6·4 mg/kg). Up to the study cutoff date of Feb 1, 2017, no dose-limiting toxic effects, substantial cardiovascular toxic effects, or deaths occurred. One patient was removed from the activity analysis because they had insufficient target lesions for analysis. The most common grade 3 adverse events were decreased lymphocyte (n=3) and decreased neutrophil count (n=2); and grade 4 anaemia was reported by one patient. Three serious adverse events-febrile neutropenia, intestinal perforation, and cholangitis-were reported by one patient each. Overall, in 23 evaluable patients, including six patients with low HER2-expressing tumours, ten patients achieved an objective response (43%, 95% CI 23·2-65·5). Disease control was achieved in 21 (91%; 95% CI 72·0-98·9) of 23 patients. Median follow-up time was 6·7 months (IQR 4·4-10·2), with nine (90%) of ten responses seen at doses of 5·4 mg/kg or greater. INTERPRETATION The maximum tolerated dose of trastuzumab deruxtecan was not reached. In this small, heavily pretreated study population, trastuzumab deruxtecan showed antitumour activity, even in low HER2-expressing tumours. Based on safety and activity, the most likely recommended phase 2 dosing is 5·4 or 6·4 mg/kg. FUNDING Daiichi Sankyo Co, Ltd.
Collapse
|
30
|
Du Y, Zhang W, He R, Ismail M, Ling L, Yao C, Fu Z, Li X. Dual 7-ethyl-10-hydroxycamptothecin conjugated phospholipid prodrug assembled liposomes with in vitro anticancer effects. Bioorg Med Chem 2017; 25:3247-3258. [PMID: 28465086 DOI: 10.1016/j.bmc.2017.04.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 04/09/2017] [Indexed: 11/20/2022]
Abstract
7-Ethyl-10-hydroxycamptothecin (SN38), as a highly active topoisomerase I inhibitor, is 200-2000-fold more cytotoxic than irinotecan (CPT-11) commercially available as Camptosar®. However, poor solubility and low stability extensively restricted its clinical utility. In this report, dual SN38 phospholipid conjugate (Di-SN38-PC) prodrug based liposomes were developed in order to compact these drawbacks. Di-SN38-PC prodrug was first synthesized by inhomogeneous conjugation of two SN38-20-O-succinic acid molecules with L-α-glycerophosphorylcholine (GPC). The assembly of the prodrug was carried out without any excipient by using thin film method. Dynamic light scattering (DLS), transmission electron microscope (TEM) and cryogenic transmission electron microscopy (cyro-TEM) characterization indicated that Di-SN38-PC can form spherical liposomes with narrow particle size (<200nm) and negatively charged surface (-21.6±3.5mV). The loading efficiency of SN38 is 65.2 wt.% after a simple calculation. In vitro release test was further performed in detail. The results demonstrated that Di-SN38-PC liposomes were stable in neutral environment but degraded in a weakly acidic condition thereby released parent drug SN38 effectively. Cellular uptake studies reflected that the liposomes could be internalized into cells more significantly than SN38. In vitro antitumor activities were finally evaluated by MTT assay, colony formation assay, flow cytometry, RT-PCR analysis and Western Blot. The results showed that Di-SN38-PC liposomes had a comparable cytotoxicity with SN38 against MCF-7 and HBL-100, and a selective promotion of apoptosis of tumor cells. Furthermore, a pharmacokinetics test showed that Di-SN38-PC liposomes had a longer circulating time in blood compared with the parent drug. All the results indicate that Di-SN38-PC liposomes are an effective delivery system of SN38.
Collapse
Affiliation(s)
- Yawei Du
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Wei Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Ruiyu He
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Muhammad Ismail
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Longbing Ling
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Chen Yao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Zhenglin Fu
- National Center for Protein Science, Shanghai 201210, PR China
| | - Xinsong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|
31
|
Pan X, Yang C, Cleveland JL, Bannister TD. Synthesis and Cytoxicity of Sempervirine and Analogues. J Org Chem 2016; 81:2194-200. [PMID: 26828413 DOI: 10.1021/acs.joc.6b00022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Sempervirine and analogues were synthesized using a route featuring Sonogashira and Larock Pd-catalyzed reactions. Structure-activity relationships were investigated using three human cancer cell lines. 10-Fluorosempervirine is the most potently cytotoxic member of the family yet described.
Collapse
Affiliation(s)
- Xiaohong Pan
- Department of Chemistry, The Scripps Research Institute , Jupiter, Florida 33458, United States
| | - Chunying Yang
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute , Tampa, Florida 33612, United States
| | - John L Cleveland
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute , Tampa, Florida 33612, United States
| | - Thomas D Bannister
- Department of Chemistry, The Scripps Research Institute , Jupiter, Florida 33458, United States
| |
Collapse
|
32
|
Nakada T, Masuda T, Naito H, Yoshida M, Ashida S, Morita K, Miyazaki H, Kasuya Y, Ogitani Y, Yamaguchi J, Abe Y, Honda T. Novel antibody drug conjugates containing exatecan derivative-based cytotoxic payloads. Bioorg Med Chem Lett 2016; 26:1542-1545. [PMID: 26898815 DOI: 10.1016/j.bmcl.2016.02.020] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 02/04/2016] [Accepted: 02/08/2016] [Indexed: 11/28/2022]
Abstract
Trastuzumab conjugates consisting of exatecan derivatives were prepared and their biological activities and physicochemical properties were evaluated. The ADCs showed strong efficacy and a low aggregation rate. The exatecan derivatives were covalently connected via a peptidyl spacer (Gly-Gly-Phe-Gly), which is assumed to be stable in circulation, and were cleaved by lysosomal enzymes following ADC internalization into tumor tissue. These anti-HER2 ADCs exhibited a high potency, specifically against HER2-positive cancer cell lines in vitro. The ADCs, bearing exatecan derivatives which have more than two methylene chains, exhibited superior cytotoxicity. It was speculated that steric hindrance of the cleavable amide moiety could be involved in the drug release. The adequate alkyl lengths of exatecan derivatives (13, 14, 15) were from two to four in terms of aggregation rate. The ADC having a hydrophilic moiety showed good efficacy in a HER2-positive and Trastuzumab-resistant breast carcinoma cell model in mice.
Collapse
Affiliation(s)
- 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
| | - Hiroyuki Naito
- Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Masao Yoshida
- Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Shinji Ashida
- 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
| | - Hideki Miyazaki
- Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yuji Kasuya
- Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yusuke Ogitani
- 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
| | - Yuki Abe
- Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Takeshi Honda
- Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| |
Collapse
|
33
|
McInnes SJP, Michl TD, Delalat B, Al-Bataineh SA, Coad BR, Vasilev K, Griesser HJ, Voelcker NH. "Thunderstruck": Plasma-Polymer-Coated Porous Silicon Microparticles As a Controlled Drug Delivery System. ACS APPLIED MATERIALS & INTERFACES 2016; 8:4467-4476. [PMID: 26836366 DOI: 10.1021/acsami.5b12433] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Controlling the release kinetics from a drug carrier is crucial to maintain a drug's therapeutic window. We report the use of biodegradable porous silicon microparticles (pSi MPs) loaded with the anticancer drug camphothecin, followed by a plasma polymer overcoating using a loudspeaker plasma reactor. Homogenous "Teflon-like" coatings were achieved by tumbling the particles by playing AC/DC's song "Thunderstruck". The overcoating resulted in a markedly slower release of the cytotoxic drug, and this effect correlated positively with the plasma polymer coating times, ranging from 2-fold up to more than 100-fold. Ultimately, upon characterizing and verifying pSi MP production, loading, and coating with analytical methods such as time-of-flight secondary ion mass spectrometry, scanning electron microscopy, thermal gravimetry, water contact angle measurements, and fluorescence microscopy, human neuroblastoma cells were challenged with pSi MPs in an in vitro assay, revealing a significant time delay in cell death onset.
Collapse
Affiliation(s)
- Steven J P McInnes
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Thomas D Michl
- Future Industries Institute, University of South Australia , Mawson Lakes, South Australia 5095, Australia
| | - Bahman Delalat
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia , Adelaide, South Australia 5001, Australia
| | - Sameer A Al-Bataineh
- Future Industries Institute, University of South Australia , Mawson Lakes, South Australia 5095, Australia
| | - Bryan R Coad
- Future Industries Institute, University of South Australia , Mawson Lakes, South Australia 5095, Australia
| | - Krasimir Vasilev
- Future Industries Institute, University of South Australia , Mawson Lakes, South Australia 5095, Australia
| | - Hans J Griesser
- Future Industries Institute, University of South Australia , Mawson Lakes, South Australia 5095, Australia
| | - Nicolas H Voelcker
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia , Adelaide, South Australia 5001, Australia
| |
Collapse
|
34
|
Xiao Z, Morris-Natschke SL, Lee KH. Strategies for the Optimization of Natural Leads to Anticancer Drugs or Drug Candidates. Med Res Rev 2016; 36:32-91. [PMID: 26359649 PMCID: PMC4679534 DOI: 10.1002/med.21377] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Natural products have made significant contribution to cancer chemotherapy over the past decades and remain an indispensable source of molecular and mechanistic diversity for anticancer drug discovery. More often than not, natural products may serve as leads for further drug development rather than as effective anticancer drugs by themselves. Generally, optimization of natural leads into anticancer drugs or drug candidates should not only address drug efficacy, but also improve absorption, distribution, metabolism, excretion, and toxicity (ADMET) profiles and chemical accessibility associated with the natural leads. Optimization strategies involve direct chemical manipulation of functional groups, structure-activity relationship directed optimization and pharmacophore-oriented molecular design based on the natural templates. Both fundamental medicinal chemistry principles (e.g., bioisosterism) and state-of-the-art computer-aided drug design techniques (e.g., structure-based design) can be applied to facilitate optimization efforts. In this review, the strategies to optimize natural leads to anticancer drugs or drug candidates are illustrated with examples and described according to their purposes. Furthermore, successful case studies on lead optimization of bioactive compounds performed in the Natural Products Research Laboratories at UNC are highlighted.
Collapse
Affiliation(s)
- Zhiyan Xiao
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Susan L. Morris-Natschke
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599-7568, USA
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599-7568, USA
- Chinese Medicine Research and Development Center, China Medical University and Hospital, Taichung, Taiwan
| |
Collapse
|
35
|
Serrano-Olvera A, Cetina L, Coronel J, Dueñas-González A. Emerging drugs for the treatment of cervical cancer. Expert Opin Emerg Drugs 2015; 20:165-82. [PMID: 25578210 DOI: 10.1517/14728214.2015.1002768] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Worldwide, most cervical cancer (CC) patients require the use of drug therapy either adjuvant, concurrent with radiation or palliative. AREAS COVERED This review briefly discusses the current achievements in treating CC with an emphasis in emerging agents. EXPERT OPINION Concurrent cisplatin with radiation and lately, gemcitabine-cisplatin chemoradiation has resulted in small but significant improvements in the treatment of locally advanced and high-risk early-stage patients. So far, only antiangiogenic therapy with bevacizumab added to cisplatin chemoradiation has demonstrated safety and encouraging results in a Phase II study. In advanced disease, cisplatin doublets yield median survival rates not exceeding 14 months. The first Phase III study of bevacizumab, added to standard chemotherapy cisplatin- or non-cisplatin-containing doublet, has shown significant increase in both overall survival and progression-free survival. Further studies are needed before bevacizumab plus chemotherapy can be considered the standard of care for advanced disease. The characterization of the mutational landscape of CC and developments of novel targeted therapies may result in more effective and individualized treatments for CC. The potential efficacy of knocking down the key alterations in CC, E6 and E7 human papilloma virus oncoproteins must not be overlooked.
Collapse
|
36
|
Matsunaga S, Kanai M, Yamamoto S. Cobalt-Catalyzed C5-Selective C-H Functionalization of 4-Me-Quinolines with Styrenes: An Approach to 5,6-Dihydro-4H-benzo[de]quinolines. HETEROCYCLES 2015. [DOI: 10.3987/com-14-s(k)7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
37
|
Stolle C, Storp J, Ivanova BB, Spiteller M. SYNTHESIS, ISOLATION, STRUCTURAL AND SPECTROSCOPIC STUDY OF THIOCAMPTOTHECINS AND THEIR SILVER(I) COMPLEXES—THEORETICAL AND EXPERIMENTAL ELUCIDATION. PHOSPHORUS SULFUR 2013. [DOI: 10.1080/10426507.2013.765423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Cornelia Stolle
- a Institut für Umweltforschung, Universität Dortmund , Otto-Hahn-Strasse 6, 44221 , Dortmund , Germany
| | - Jürgen Storp
- a Institut für Umweltforschung, Universität Dortmund , Otto-Hahn-Strasse 6, 44221 , Dortmund , Germany
| | - Bojidarka B. Ivanova
- a Institut für Umweltforschung, Universität Dortmund , Otto-Hahn-Strasse 6, 44221 , Dortmund , Germany
| | - Michael Spiteller
- a Institut für Umweltforschung, Universität Dortmund , Otto-Hahn-Strasse 6, 44221 , Dortmund , Germany
| |
Collapse
|
38
|
Koshkaryev A, Sawant R, Deshpande M, Torchilin V. Immunoconjugates and long circulating systems: origins, current state of the art and future directions. Adv Drug Deliv Rev 2013; 65:24-35. [PMID: 22964425 DOI: 10.1016/j.addr.2012.08.009] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/15/2012] [Accepted: 08/22/2012] [Indexed: 12/31/2022]
Abstract
Significant progress has been made recently in the area of immunoconjugated drugs and drug delivery systems (DDS). The immuno-modification of either the drug or DDS has proven to be a very promising approach that has significantly improved the targeted accumulation in pathological sites while decreasing its undesirable side effects in healthy tissues. The arrangement for both prolonged life in the circulation and specific target recognition represents another potent strategy in the development of immuno-targeted systems. The longevity of immuno-targeted DDS such as immunoliposomes and immunomicelles improves their targetability even in the presence of the additional passive accumulation in areas with a compromised vasculature. The added use of the immuno-targeted systems takes advantage of the specific microenvironment of pathological sites including lowered pH, increased temperature, and variation in the enzymatic activity. "Smart" stimulus-responsive systems combine different valuable functionalities including PEG-protection, targeting antibody, cell-penetration, and stimulus-sensitive functions. In this review we examined the evolution, current status and future directions in the area of therapeutical immunoconjugates and long-circulating immuno-targeted DDS.
Collapse
Affiliation(s)
- Alexander Koshkaryev
- Center for Pharmaceutical Biotechnology & Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
39
|
Dueñas-Gonzalez A, Cetina L, Coronel J, Cervantes-Madrid D. Emerging drugs for cervical cancer. Expert Opin Emerg Drugs 2012; 17:203-18. [PMID: 22530838 DOI: 10.1517/14728214.2012.683409] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION There is a shortage of therapeutical agents for invasive cervical cancer in late stages of development; however, a number of promising molecules are currently in early phases of development. AREAS COVERED This review briefly discusses the current achievements in treating cervical cancer with an emphasis in emerging agents based on a literature search on pubmed and related sites for cervical cancer information. This is not a systematic review. EXPERT OPINION In advanced disease, modest survival gains have been achieved with cisplatin doublets. Contrariwise, chemoradiation has increased survival rates in locally advanced disease, but there is still room for improvement. Anti-vascular endothelial growth factor and anti-epidermal growth factor receptor monoclonal antibodies are promising molecules that are at present in late-phase development, but a high number of miscellaneous agents are in early development. Strong experimental bases support that the 'Achilles' heel' of cervical cancer are the HPV-E6/E7 oncogenes. Unfortunately, agents aimed at targeting these cervical cancer-driven players are found in very early development; hence, major research efforts must be focused on developing technological strategies for their effective targeting using nucleic acid-based vehicles for safe and effective delivery to cancer cells as well as accelerating the search for small-molecule inhibitors of E6/E7 themselves or their interacting cellular proteins.
Collapse
Affiliation(s)
- Alfonso Dueñas-Gonzalez
- Division of Clinical Research, Instituto Nacional de Cancerología-México (INCan), Mexico City, Mexico.
| | | | | | | |
Collapse
|
40
|
Venditto VJ, Simanek EE. Cancer therapies utilizing the camptothecins: a review of the in vivo literature. Mol Pharm 2010; 7:307-49. [PMID: 20108971 DOI: 10.1021/mp900243b] [Citation(s) in RCA: 304] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review summarizes the in vivo assessment-preliminary, preclinical, and clinical-of chemotherapeutics derived from camptothecin or a derivative. Camptothecin is a naturally occurring, pentacyclic quinoline alkaloid that possesses high cytotoxic activity in a variety of cell lines. Major limitations of the drug, including poor solubility and hydrolysis under physiological conditions, prevent full clinical utilization. Camptothecin remains at equilibrium in an active lactone form and inactive hydrolyzed carboxylate form. The active lactone binds to DNA topoisomerase I cleavage complex, believed to be the single site of activity. Binding inhibits DNA religation, resulting in apoptosis. A series of small molecule camptothecin derivatives have been developed that increase solubility, lactone stability and bioavailability to varying levels of success. A number of macromolecular agents have also been described wherein camptothecin(s) are covalently appended or noncovalently associated with the goal of improving solubility and lactone stability, while taking advantage of the tumor physiology to deliver larger doses of drug to the tumor with lower systemic toxicity. With the increasing interest in drug delivery and polymer therapeutics, additional constructs are anticipated. The goal of this review is to summarize the relevant literature for others interested in the field of camptothecin-based therapeutics, specifically in the context of biodistribution, dosing regimens, and pharmacokinetics with the desire of providing a useful source of comparative data. To this end, only constructs where in vivo data is available are reported. The review includes published reports in English through mid-2009.
Collapse
Affiliation(s)
- Vincent J Venditto
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, USA
| | | |
Collapse
|
41
|
Deshmukh M, Chao P, Kutscher HL, Gao D, Sinko PJ. A series of alpha-amino acid ester prodrugs of camptothecin: in vitro hydrolysis and A549 human lung carcinoma cell cytotoxicity. J Med Chem 2010; 53:1038-47. [PMID: 20063889 DOI: 10.1021/jm901029n] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The objective of the present study was to identify a camptothecin (CPT) prodrug with optimal release and cytotoxicity properties for immobilization on a passively targeted microparticle delivery system. A series of alpha-amino acid ester prodrugs of CPT were synthesized, characterized, and evaluated. Four CPT prodrugs were synthesized with increasing aliphatic chain length (glycine (Gly) (2a), alanine (Ala) (2b), aminobutyric acid (Abu) (2c), and norvaline (Nva) (2d)). Prodrug reconversion was studied at pH 6.6, 7.0, and 7.4 corresponding to tumor, lung, and extracellular/physiological pH, respectively. Cytotoxicity was evaluated in A549 human lung carcinoma cells using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The hydrolytic reconversion rate to parent CPT increased with decreasing side chain length as well as increasing pH. The Hill slope of 2d was significantly less than CPT and the other prodrugs tested, indicating a higher cell death rate at lower concentrations. These results suggest that 2d is the best candidate for a passively targeted sustained release lung delivery system.
Collapse
Affiliation(s)
- Manjeet Deshmukh
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, USA
| | | | | | | | | |
Collapse
|
42
|
Basili S, Moro S. Novel camptothecin derivatives as topoisomerase I inhibitors. Expert Opin Ther Pat 2009; 19:555-74. [DOI: 10.1517/13543770902773437] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Ohwada J, Ozawa S, Kohchi M, Fukuda H, Murasaki C, Suda H, Murata T, Niizuma S, Tsukazaki M, Ori K, Yoshinari K, Itezono Y, Endo M, Ura M, Tanimura H, Miyazaki Y, Kawashima A, Nagao S, Namba E, Ogawa K, Kobayashi K, Okabe H, Umeda I, Shimma N. Synthesis and biological activities of a pH-dependently activated water-soluble prodrug of a novel hexacyclic camptothecin analog. Bioorg Med Chem Lett 2009; 19:2772-6. [DOI: 10.1016/j.bmcl.2009.03.123] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 03/12/2009] [Accepted: 03/25/2009] [Indexed: 10/21/2022]
|
44
|
Niizuma S, Tsukazaki M, Suda H, Murata T, Ohwada J, Ozawa S, Fukuda H, Murasaki C, Kohchi M, Morikami K, Yoshinari K, Endo M, Ura M, Tanimura H, Miyazaki Y, Takasuka T, Kawashima A, Nanba E, Nakano K, Ogawa K, Kobayashi K, Okabe H, Umeda I, Shimma N. Synthesis of new camptothecin analogs with improved antitumor activities. Bioorg Med Chem Lett 2009; 19:2018-21. [DOI: 10.1016/j.bmcl.2009.02.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Revised: 02/04/2009] [Accepted: 02/07/2009] [Indexed: 11/16/2022]
|
45
|
Discovery of a small-molecule inhibitor of {beta}-1,6-glucan synthesis. Antimicrob Agents Chemother 2008; 53:670-7. [PMID: 19015325 DOI: 10.1128/aac.00844-08] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It is possible that antifungal drugs with novel modes of action will provide favorable options to treat fungal infections. In the course of our screening for antifungal compounds acting on the cell wall, a pyridobenzimidazole derivative with unique activities, named D75-4590, was discovered. During treatment of Saccharomyces cerevisiae with D75-4590, (i) incorporation of [(14)C]glucose into the beta-1,6-glucan component was selectively reduced, (ii) proteins released from the cell had lost the beta-1,6-glucan moiety, and (iii) cells tended to clump, resulting in impaired cell growth. Genetic analysis of a D75-4590-resistant mutant of S. cerevisiae indicated that its primary target was Kre6p, which is considered to be one of the beta-1,6-glucan synthases. These results strongly suggest that D75-4590 is a specific inhibitor of beta-1,6-glucan synthesis. D75-4590 showed potent activities against various Candida species. It inhibited hyphal elongation of C. albicans as well. KRE6 is conserved in various fungi, but no homologue has been found in mammalian cells. These lines of evidence indicate that D75-4590 is a promising lead compound for novel antifungal drugs. To our knowledge, this is the first report of a beta-1,6-glucan inhibitor.
Collapse
|
46
|
Zhang J, Cheng C, He CL, Zhou YJ, Cao Y. The expression of Bcl-XL, Bcl-XS and p27Kip1 in topotecan-induced apoptosis in hepatoblastoma HepG2 cell line. Cancer Invest 2008; 26:456-63. [PMID: 18568767 DOI: 10.1080/07357900701683968] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND To assess the efficacy of topotecan, a topoisomerase I specific inhibitor in S-phase, the reagent-induced apoptosis and cytotoxicity as well as related proteins expression, had been preliminarily investigated in human hepatoblastoma HepG2 cells. METHODS Microculture tetrazolium assay (MTT), HE staining, transmission electron microscopy (TEM), flow cytometry (FCM), quantitative immunocytochemistry (QI), gene tranfection and RNAi technology were employed to carry out the exploration. RESULTS Topotecan could potently kill HepG2 cells via inducing apoptosis and demonstrated strong cytotoxicity in a time, dose-dependent manner with IC50 of about 95 mu g/L. According to morphologic observation and FCM analyses, it was confirmed that the drug treatment, causing significant S-phase arrest, could trigger a typical interphase apoptosis, the main traits of which were identified as chromatin pycnosis and cytoplasm condensation. It was shown that the expression of Bcl-XL was simultaneously down-regulated with the up-regulation of Bcl-XS in cytoplasm, which was possibly a key downstream event following the topotecan-induced DNA damage in nucleus. The expression level of p27Kip1, a negative regulator in cell cycle at G1/S transient, was also elevated. Transfection of pcDNA 3.1-p27Kip1 into HepG2 cells could abrogate the cytotoxicity in a degree while silence of p27Kip1 with siRNA in drug treatments could significantly increased the chemosensitivity, strongly indicating that the up-regulation of p27Kip1 was not an apoptosis-promoting, but a self-rescue response against drug by moderate G0/G1 arrest. CONCLUSION Topotecan had potent cytotoxicity against HepG2 cells by triggering an interphase apoptosis possibly mediated by increasing the ratio of Bcl-XS/Bcl-XL. Up-regulation of p27Kip1in TPT treatments could be a protective response for self-rescue and silence of the gene markedly augmented TPT cytotoxicity. Therefore, the experiment in vitro could provide a new idea for the clinical chemotherapy based on the combination of traditional drugs with the specific-siRNA targeted on the protective response gene.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Cell Biology and Genetics, Chongqing University of Medical Sciences, Chongqing, People's Republic of China.
| | | | | | | | | |
Collapse
|
47
|
Hartmann JT, Lipp HP. Camptothecin and podophyllotoxin derivatives: inhibitors of topoisomerase I and II - mechanisms of action, pharmacokinetics and toxicity profile. Drug Saf 2007; 29:209-30. [PMID: 16524321 DOI: 10.2165/00002018-200629030-00005] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Camptothecins represent an established class of effective agents that selectively target topoisomerase I by trapping the catalytic intermediate of the topoisomerase I-DNA reaction, the cleavage complex. The water-soluble salt camptothecin-sodium - introduced in early trials in the 1960s - was highly toxic in animals, whereas the semisynthetic derivatives irinotecan and topotecan did not cause haemorrhagic cystitis because of their higher physicochemical stability and solubility at lower pH values. Myelosuppression, neutropenia and, to a lesser extent, thrombocytopenia are dose-limiting toxic effects of topotecan. In contrast to the structurally-related topotecan, irinotecan is a prodrug which has to be converted to SN-38, its active form. SN-38 is inactivated by conjugation, thus patients with Gilbert's syndrome and other forms of genetic glucuronidation deficiency are at an increased risk of irinotecan-induced adverse effects, such as neutropenia and diarrhoea. The cytotoxic mechanism of podophyllotoxin is the inhibition of topoisomerase II. Common adverse effects of etoposide include dose-limiting myelosuppression. Hypersensitivity reactions are more common with etoposide and teniposide than with etoposide phosphate because the formulations of the former contain sensitising solubilisers. Leukopenia and thrombocytopenia occur in 65% and 80%, respectively, of patients after administration of conventional doses of teniposide. Anorexia, vomiting and diarrhoea are generally of mild severity after administration of conventional doses of topoisomerase II inhibitors. Clinical pharmacokinetic studies have revealed substantial interindividual variabilities regarding the area under the concentration-time curve values and steady-state concentrations for all drugs reviewed in this article. Irinotecan, etoposide and teniposide are degraded via complex metabolic pathways. In contrast, topotecan primarily undergoes renal excretion. Regarding etoposide and teniposide, the extent of catechol formation over time during drug metabolism may be associated with a higher risk for secondary malignancies.
Collapse
Affiliation(s)
- Jörg T Hartmann
- Department of Oncology/Hematology/Immunology/Pneumology/Rheumatology, Eberhard Karls University Tübingen, UKT - Medical Center II, Tübingen, Germany.
| | | |
Collapse
|
48
|
Shiose Y, Kuga H, Yamashita F, Hashida M. Quantitative acid hydrolysis of DE-310, a macromolecular carrier system for the camptothecin analog DX-8951f. J Pharm Biomed Anal 2006; 43:1290-6. [PMID: 17127025 DOI: 10.1016/j.jpba.2006.10.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2006] [Revised: 10/17/2006] [Accepted: 10/20/2006] [Indexed: 11/25/2022]
Abstract
DE-310 is a novel macromolecular prodrug of the topoisomerase-I inhibitor DX-8951. DX-8951 is covalently linked to carboxymethyl dextran polyalcohol (CM-Dex-PA) via a Gly-Gly-Phe-Gly (GGFG) tetrapeptide spacer. The present study was conducted to identify the portions of DX-8951 linked to DE-310, as well as to quantify the number of DX-8951 molecules associated with DE-310. Two different structures terminated with either glycolaldehyde (CM-GA-GGFG-DX-8951) or glycerol (CM-Glr-GGFG-DX-8951) are obtained when the polymer backbone is fragmented with 1 M HCl. The two products, i.e., CM-GA-GGFG-DX-8951 and CM-Glr-GGFG-DX-8951, indicate linkage of GGFG-DX-8951 with carboxymethyl (CM) group at C-2 and C-4 position of the glucose units, respectively. In the present study, CM-GA-GGFG-DX-8951 was reduced to CM-ethyleneglycol (EG)-GGFG-DX-8951 in order to improve stability prior to HPLC analysis. Hydrolysis results revealed that the amount of CM-GA-GGFG-DX-8951 liberated was 84.7 nmol/mg DE-310 and the amount of CM-Glr-GGFG-DX-8951 was 71.8 nmol/mg DE-310. Considering the ratio of generation between CM-GA-GGFG-DX8951 and CM-Glr-GGFG-DX8951, it suggested that slightly larger amount of GGFG-DX-8951 was linked to carboxymethyl groups at the C-2 position of glucose units in DE-310. The sum of the amounts of CM-GA-GGFG-DX-8951 and CM-Glr-GGFG-DX-8951 agrees well with the amount of G-DX-8951 produced from DE-310 by alpha-chymotrypsin treatment (157.5 nmol/mg DE-310). The data indicate that the established hydrolysis give a quantitative evaluation of the DX-8951 linked to DE-310.
Collapse
Affiliation(s)
- Yoshinobu Shiose
- New Product Research Laboratories III, Daiichi Pharmaceutical Co. Ltd., Tokyo R&D Center, Kita-Kasai 1-16-13, Edogawa-Ku, Tokyo 134-8630, Japan.
| | | | | | | |
Collapse
|
49
|
Abou-Alfa GK, Letourneau R, Harker G, Modiano M, Hurwitz H, Tchekmedyian NS, Feit K, Ackerman J, De Jager RL, Eckhardt SG, O'Reilly EM. Randomized phase III study of exatecan and gemcitabine compared with gemcitabine alone in untreated advanced pancreatic cancer. J Clin Oncol 2006; 24:4441-7. [PMID: 16983112 DOI: 10.1200/jco.2006.07.0201] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Exatecan mesylate is a hexacyclic, water-soluble, topoisomerase-1 inhibitor. Exatecan has single-agent and combination activity with gemcitabine in advanced pancreatic cancer. A multicenter, randomized, phase III trial comparing exatecan plus gemcitabine versus gemcitabine alone in advanced pancreatic cancer was conducted. PATIENTS AND METHODS Eligibility criteria included Karnofsky performance status > or = 60%, locally advanced or metastatic pancreatic adenocarcinoma, and no prior chemotherapy. Radiation alone for locally advanced disease was permitted. Patients were randomly assigned on a 1:1 basis. For the exatecan plus gemcitabine arm, exatecan 2.0 mg/m2 and gemcitabine 1,000 mg/m2 were administered on days 1 and 8, every 3 weeks. Gemcitabine alone was dosed at 1,000 mg/m2 up to 7 weeks in the first cycle, then once a week for the first 3 weeks of a 4-week cycle. Tumor assessment was performed every 6 weeks. The primary end point was overall survival. An intent-to-treat analysis was used. RESULTS From August 2001 to January 2003, 349 patients were randomly assigned, 175 to exatecan plus gemcitabine and 174 to gemcitabine alone. Twenty-four patients (6.9%) were not treated. The median survival time was 6.7 months for exatecan plus gemcitabine and 6.2 months for gemcitabine alone (P = .52). One complete response (CR; < 1%) and 11 partial responses (PRs; 6.3%) were observed in the exatecan plus gemcitabine treatment group, and one CR (< 1%) and eight PRs (4.6%) were observed in the gemcitabine-alone group. Grade 3 and 4 toxicities were higher for the exatecan plus gemcitabine arm versus the gemcitabine alone arm; neutropenia (30% v 15%) and thrombocytopenia (15% v 4%). CONCLUSION Exatecan plus gemcitabine was not superior to gemcitabine alone with respect to overall survival in the first-line treatment of advanced pancreatic cancer.
Collapse
|
50
|
|