1
|
Procopiou G, Jackson PJM, Andriollo P, Hasan MM, Veillard N, Rahman KM, Thurston DE. Synthesis of novel pyrrolobenzodiazepine (PBD) C1-substituted monomers and dimers with DNA-binding activity and cytotoxicity. Bioorg Med Chem Lett 2025; 119:130095. [PMID: 39778751 DOI: 10.1016/j.bmcl.2025.130095] [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: 10/08/2024] [Revised: 12/17/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
The pyrrolobenzodiazepines (PBDs) represent a major class of sequence-selective DNA-alkylating molecules, one example of which, in its dimeric DNA-cross-linking form, is employed as the payload in the anticancer Antibody Drug Conjugate (ADC) loncastuximab tesirine-lpyl. To date, PBD analogues have been produced with substituents at every position of the tricyclic skeleton except the C1-position. We report here the first synthesis of a C1-subsitituted PBD monomer and dimer, both of which possess DNA-binding activity and cytotoxicity in a cancer cell line.
Collapse
Affiliation(s)
- George Procopiou
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Paul J M Jackson
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Paolo Andriollo
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Md Mahbub Hasan
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Nicolas Veillard
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Khondaker Miraz Rahman
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - David E Thurston
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| |
Collapse
|
2
|
Akram F, Ali AM, Akhtar MT, Fatima T, Shabbir I, Ul Haq I. The journey of antibody-drug conjugates for revolutionizing cancer therapy: A review. Bioorg Med Chem 2025; 117:118010. [PMID: 39586174 DOI: 10.1016/j.bmc.2024.118010] [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: 09/07/2024] [Revised: 11/09/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Abstract
Antibody-drug conjugates (ADCs) represent a powerful class of targeted cancer therapies that harness the specificity of monoclonal antibodies to deliver cytotoxic payloads directly to tumor cells, minimizing off-target effects. This review explores the advancements in ADC technologies, focusing on advancing next-generation ADCs with novel payloads, conjugation strategies, and enhanced pharmacokinetic profiles. In particular, we highlight innovative payloads, including microtubule inhibitors, spliceosome modulators, and RNA polymerase inhibitors, that offer new mechanisms of cytotoxicity beyond traditional apoptosis induction. Additionally, the introduction of sophisticated conjugation techniques, such as site-specific conjugation using engineered cysteines, enzymatic methods, and integration of non-natural amino acids, has greatly improved the homogeneity, efficacy, and safety of ADCs. Furthermore, the review delves into the mechanistic insights into ADC action, detailing the intracellular pathways that facilitate drug release and cell death, and discussing the significance of bioconjugation methods in optimizing drug-antibody ratios (DARs). The establishment of comprehensive databases like ADCdb, which catalog vital pharmacological and biological data for ADCs, is also explored as a critical resource for advancing ADC research and clinical application. Finally, the clinical landscape of ADCs is examined, with a focus on the evolution of FDA-approved ADCs, such as Gemtuzumab Ozogamicin and Trastuzumab Emtansine, as well as emerging candidates in ongoing trials. As ADCs continue to evolve, their potential to revolutionize cancer therapy remains immense, offering new hope for more effective and personalized treatment options. ADCs also offer a significant advancement in targeted cancer therapy by merging the specificity of monoclonal antibodies with cytotoxic potency of chemotherapeutic agents. Hence, this dual mechanism intensifies tumor selectivity while minimizing systemic toxicity, paving the way for more effective and safer cancer treatments.
Collapse
Affiliation(s)
- Fatima Akram
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan; Department of Biology, Saint Louis University, St. Louis, MO, USA.
| | - Amna Murrawat Ali
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Muhammad Tayyab Akhtar
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Taseer Fatima
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Ifrah Shabbir
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Ikram Ul Haq
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan; Pakistan Academy of Sciences, Islamabad, Pakistan
| |
Collapse
|
3
|
Tarantelli C, Wald D, Munz N, Spriano F, Bruscaggin A, Cannas E, Cascione L, Gaudio E, Arribas AJ, Manjappa S, Golino G, Scalise L, Cacciapuoti MT, Zucca E, Stathis A, Inghirami G, Van Berkel PH, Rossi D, Caimi PF, Zammarchi F, Bertoni F. Targeting CD19-positive lymphomas with the antibodydrug conjugate loncastuximab tesirine: preclinical evidence of activity as a single agent and in combination therapy. Haematologica 2024; 109:3314-3326. [PMID: 38721745 PMCID: PMC11443381 DOI: 10.3324/haematol.2023.284197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Indexed: 10/02/2024] Open
Abstract
Antibody-drug conjugates (ADC) represent one of the most successful therapeutic approaches introduced into clinical practice in the last few years. Loncastuximab tesirine (ADCT-402) is a CD19-targeting ADC in which the antibody is conjugated through a protease cleavable dipeptide linker to a pyrrolobenzodiazepine dimer warhead (SG3199). Based on the results of a phase II study, loncastuximab tesirine was recently approved for adult patients with relapsed/refractory large B-cell lymphoma. We assessed the activity of loncastuximab tesirine using in vitro and in vivo models of lymphomas, correlated its activity with levels of CD19 expression, and identified combination partners providing synergy with the ADC. Loncastuximab tesirine was tested across 60 lymphoma cell lines. It had strong cytotoxic activity in B-cell lymphoma cell lines. The in vitro activity was correlated with the level of CD19 expression and intrinsic sensitivity of cell lines to the ADC's warhead. Loncastuximab tesirine was more potent than other anti-CD19 ADC (coltuximab ravtansine, huB4-DGN462), although the pattern of activity across cell lines was correlated. The activity of loncastuximab tesirine was also largely correlated with cell line sensitivity to R-CHOP. Combinatorial in vitro and in vivo experiments identified the benefit of adding loncastuximab tesirine to other agents, especially BCL2 and PI3K inhibitors. Our data support the further development of loncastuximab tesirine for use as a single agent and in combination for patients affected by mature B-cell neoplasms. The results also highlight the importance of CD19 expression and the existence of lymphoma populations characterized by resistance to multiple therapies.
Collapse
Affiliation(s)
- Chiara Tarantelli
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona.
| | - David Wald
- Case Western Reserve University, Cleveland, OH
| | - Nicolas Munz
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona
| | - Filippo Spriano
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona
| | - Alessio Bruscaggin
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona
| | - Eleonora Cannas
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona
| | - Luciano Cascione
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland; SIB Swiss Institute of Bioinformatics, Lausanne
| | - Eugenio Gaudio
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona
| | - Alberto J Arribas
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland; SIB Swiss Institute of Bioinformatics, Lausanne
| | | | - Gaetanina Golino
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona
| | - Lorenzo Scalise
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona
| | | | - Emanuele Zucca
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland; Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, USI, Lugano
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | | | - Davide Rossi
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland; Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona
| | - Paolo F Caimi
- Cleveland Clinic/Case Comprehensive Cancer Center, Cleveland, OH
| | | | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland; Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona.
| |
Collapse
|
4
|
Lu N, Wu J, Tian M, Zhang S, Li Z, Shi L. Comprehensive review on the elaboration of payloads derived from natural products for antibody-drug conjugates. Eur J Med Chem 2024; 268:116233. [PMID: 38408390 DOI: 10.1016/j.ejmech.2024.116233] [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: 01/29/2024] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 02/28/2024]
Abstract
Antibody-drug conjugates (ADCs) have arisen as a promising class of biotherapeutics for targeted cancer treatment, combining the specificity of monoclonal antibodies with the cytotoxicity of small-molecule drugs. The choice of an appropriate payload is crucial for the success development of ADCs, as it determines the therapeutic efficacy and safety profile. This review focuses on payloads derived from natural products, including cytotoxic agents, DNA-damaging agents, and immunomodulators. These offer several advantages such as diverse chemical structures, unique mechanism of actions, and potential for improved therapeutic index. Challenges and opportunities associated with their development were highlighted. This review underscores the significance of natural product payloads in the elaboration of ADCs, which serves as a valuable resource for researchers involved in developing and optimizing next-generation ADCs for cancer treatment.
Collapse
Affiliation(s)
- Nan Lu
- XDC Analytical Sciences, WuXi XDC Co., Ltd., 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China
| | - Jiaqi Wu
- XDC Analytical Sciences, WuXi XDC Co., Ltd., 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China
| | - Mengwei Tian
- XDC Analytical Sciences, WuXi XDC Co., Ltd., 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China
| | - Shanshan Zhang
- XDC Analytical Sciences, WuXi XDC Co., Ltd., 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China.
| | - Zhiguo Li
- XDC Analytical Sciences, WuXi XDC Co., Ltd., 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China.
| | - Liming Shi
- XDC Analytical Sciences, WuXi XDC Co., Ltd., 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China.
| |
Collapse
|
5
|
Chen LH, Lin MY, Lin HC, Yang FW, Liao HW, Shiau CW, Chiu HC, Su JC. Discovery of new dibenzodiazepine derivatives as antibacterials against intracellular bacteria. RSC Med Chem 2024; 15:283-292. [PMID: 38283231 PMCID: PMC10809566 DOI: 10.1039/d3md00418j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/15/2023] [Indexed: 01/30/2024] Open
Abstract
The emergence and spread of multidrug-resistant bacteria underscore the critical need for novel antibacterial interventions. In our screening of 12 synthesized thienobenzodiazepines, pyridobenzodiazepines, and dibenzodiazepines, we successfully identified a small molecule compound SW33. Notably, SW33 demonstrated potent inhibitory activity against intracellular multidrug-resistant and fluoroquinolone-resistant strains of S. typhimurium in both macrophages and epithelial cells. Furthermore, SW33 was also effective against intramacrophagic Salmonella typhi, Yersinia enterocolitica, and Listeria monocytogenes. These significant findings suggest that SW33 possesses broad-spectrum activity against intracellular bacteria.
Collapse
Affiliation(s)
- Ling-Han Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University Taipei 10048 Taiwan (+886) 22371-1574 (+886) 22312-3456 ext 66902
| | - Man-Yi Lin
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University Taipei 11221 Taiwan
| | - Hsueh-Chun Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University Taipei 10048 Taiwan (+886) 22371-1574 (+886) 22312-3456 ext 66902
| | - Fan-Wei Yang
- Department of Pharmacy, National Yang Ming Chiao Tung University Taipei 11221 Taiwan (+886) 22826-7000 ext 66401
| | - Hsiao-Wei Liao
- Department of Pharmacy, National Yang Ming Chiao Tung University Taipei 11221 Taiwan (+886) 22826-7000 ext 66401
| | - Chung-Wai Shiau
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University Taipei 11221 Taiwan
| | - Hao-Chieh Chiu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University Taipei 10048 Taiwan (+886) 22371-1574 (+886) 22312-3456 ext 66902
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University Taipei 10021 Taiwan
| | - Jung-Chen Su
- Department of Pharmacy, National Yang Ming Chiao Tung University Taipei 11221 Taiwan (+886) 22826-7000 ext 66401
| |
Collapse
|
6
|
Permingeat Squizatto C, Bianchini MA, Delpiccolo CML. Synthesis of 2,3-Dihydropyrroles by Rhodium(II)-Catalyzed Transannulation of N-Sulfonyl-1,2,3-triazoles: Diversity Generation by One-Pot Methodologies. J Org Chem 2023; 88:16091-16103. [PMID: 37910436 DOI: 10.1021/acs.joc.3c01337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
A versatile one-pot strategy for the generation of compounds of synthetic interest has been presented, promoting the development of practical processes. First, the transannulation of N-sulfonyltriazoles through alkenes and rhodium catalysis was described, giving 2,3-dihydropyrroles in 13-76% yield. As contributions of the strategy, the evaluation of alkenes with different properties, and the use of only drops of solvent (0.40 M) was highlighted. In addition, we described a methodology for the modulation of the N-sulfonyltriazoles, to obtain selectively cyclopropyl tosylimines or 2,3-dihydropyrroles. For the latter products, neat conditions were also included. Finally, the potential of the methodology was demonstrated by the synthesis of six structurally different analogues starting from the same substrates and late-stage transformation of bioactive molecules. These compounds were generated in 38-63% yield, after two or more conversion steps carried out in the same reaction vessel.
Collapse
Affiliation(s)
- Caterina Permingeat Squizatto
- Instituto de Química Rosario, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario-CONICET, Suipacha 531, Rosario S2002LRK, Argentina
| | - Maira A Bianchini
- Instituto de Química Rosario, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario-CONICET, Suipacha 531, Rosario S2002LRK, Argentina
| | - Carina M L Delpiccolo
- Instituto de Química Rosario, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario-CONICET, Suipacha 531, Rosario S2002LRK, Argentina
| |
Collapse
|
7
|
Leitis Z, Sakaine G, Brokāne K, Smits G. Concise access to C2-ethylidene pyrrolo[1,4]benzodiazepine natural products. Org Biomol Chem 2023; 21:9061-9064. [PMID: 37937611 DOI: 10.1039/d3ob01430d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
A 3-step route toward pyrrolo[1,4]benzodiazepine (PBD) antitumor antibiotic class members oxo-prothracarcin and boseongazepine B has been developed. This methodology also enables preparing oxo-tomaymycin in only 4 linear steps representing the shortest total synthesis known to date. The synthesis features an olefination with sterically demanding Julia-Kocienski reagents as the key step.
Collapse
Affiliation(s)
- Zigmārs Leitis
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV-1006, Latvia.
| | - Guna Sakaine
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV-1006, Latvia.
| | - Katrīna Brokāne
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV-1006, Latvia.
| | - Gints Smits
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga, LV-1006, Latvia.
| |
Collapse
|
8
|
Jarończyk M, Ostrowski S, Dobrowolski JC. On Integral INICS Aromaticity of Pyridodiazepine Constitutional Isomers and Tautomers. Molecules 2023; 28:5684. [PMID: 37570653 PMCID: PMC10419959 DOI: 10.3390/molecules28155684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
The structure, energetics, and aromaticity of c.a. 100 constitutional isomers and tautomers of pyrido[m,n]diazepines (m = 1, 2; n = 2, 3, 4, 5; m ≠ n) were studied at the B3LYP/cc-pVTZ level. The pyrido[1,3]diazepines appear the most, while pyrido[2,4]diazepines are the least stable (ca. 26 kcal/mol). In the pyrido[1,n]diazepine group (n = 2-5), the [1,5] isomers are higher in energy by ca. 4.5 kcal/mol and the [1,4] ones by ca. 7 kcal/mol, and the pyrido[1,2]diazepines are the least stable (ca. 20 kcal/mol). All the most stable pyrido[1,n]diazepines have N-atoms near the ring's junction bond but on opposite sites. The most stable [2,n]-forms are also those with the pyridine ring N6-atom near the junction bond. Surprisingly, for the [1,2]-, [1,3]-, and [1,4]-isomer condensation types of pyridine and diazepine rings, the same N9 > N7 > N6 > N8 stability pattern obeys. The stability remains similar in a water medium simulated with the Polarizable Continuum Model of the solvent and is conserved when calculated using the CAM-B3LYP or BHandHlyp functionals. The ring's aromaticity in the pyridine[m,n]diazepines was established based on the integral INICS index resulting from the NICSzz-scan curves' integration. The integral INICS index is physically justified through its relation to the ringcurrent as demonstrated by Berger, R.J.F., et al. Phys. Chem. Chem. Phys. 2022, 24, 624. The six-membered pyrido rings have negative INICSZZ indices and can be aromatic only if they are not protonated at the N-atom. All protonated pyrido and seven-membered rings exhibit meaningful positive INICSZZ values and can be assigned as antiaromatic. However, some non-protonated pyrido rings also have substantial positive INICSZZ indices and are antiaromatic. A weak linear correlation (R2 = 0.72) between the INICSZZ values of the pyridine I(6) and diazepine I(7) rings exists and is a consequence of the communication between the π-electron systems of the two rings. The juxtaposition of the INICS descriptor of the six- and seven-membered rings and diverse electron density parameters at the Ring Critical Points (RCP) revealed good correlations only with the Electrostatic Potentials from the electrons and nuclei (ESPe and ESPn). The relationships with other RCP parameters like electron density and its Laplacian, total energy, and the Hamiltonian form of kinetic energy density were split into two parts: one nearly constant for the six-membered rings and one linearly correlating for the seven-membered rings. Thus, most of the electron density parameters at the RCP of the six-membered rings of pyridodiazepines practically do not change with the diazepine type and the labile proton position. In contrast, those of the seven-membered rings display aromaticity changes in the antiaromatic diazepine with its ring structural modifications.
Collapse
Affiliation(s)
| | - Sławomir Ostrowski
- Institute of Chemistry and Nuclear Technology, 16 Dorodna Street, 03-195 Warsaw, Poland;
| | - Jan Cz. Dobrowolski
- National Medicines Institute, 30/34 Chełmska Street, 00-725 Warsaw, Poland
- Institute of Chemistry and Nuclear Technology, 16 Dorodna Street, 03-195 Warsaw, Poland;
| |
Collapse
|
9
|
Saquib M, Ahamad S, Khan MF, Khan MI, Hussain MK. An ultrasound assisted, ionic liquid-molecular iodine synergy driven efficient green synthesis of pyrrolobenzodiazepine-triazole hybrids as potential anticancer agents. Front Pharmacol 2023; 14:1168566. [PMID: 37214464 PMCID: PMC10196072 DOI: 10.3389/fphar.2023.1168566] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/17/2023] [Indexed: 05/24/2023] Open
Abstract
Herein, we report an efficient and eco-friendly, ultrasound assisted synthetic strategy for the construction of diversified pyrrolobenzodiazepine-triazole hybrids, which are potentially pharmaceutically important scaffolds, via a domino reaction involving intermolecular electrophilic substitution followed by intramolecular Huisgen 1,3-dipolar azide-alkyne cycloaddition. The USP of the reported protocol is the use of benign and inexpensive, recyclable molecular iodine-ionic liquid synergistic catalytic system cum reaction media for achieving the synthesis. The other salient features of this method are the use of mild reaction conditions, high yield and atom economy, operational simplicity, broad substrate scope and easy workup and purification. All the synthesized compounds were evaluated for in vitro anti-proliferative activity against various cancer cell lines. From among the synthesized title compounds, 9,9-dimethyl-8-phenyl-9H-benzo [b]pyrrolo [1,2-d][1,2,3]triazolo[5,1-g][1,4]diazepine (7) was found most to be the most active compound exhibiting IC50 value of 6.60, 5.45, 7.85, 11.21, 12.24, 10.12, and 11.32 µM against MCF-7, MDA-MB-231, HeLa, SKOV-3, A549, HCT-116 and DLD-1 cell lines, respectively. Further the compounds were found to be non-toxic against normal human embryonic kidney (HEK-293) cell line.
Collapse
Affiliation(s)
- Mohammad Saquib
- Department of Chemistry, University of Allahabad, Prayagraj, Uttar Pradesh, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Mohammad Faheem Khan
- Department of Biotechnology, Era’s Lucknow Medical College, Era University, Lucknow, Uttar Pradesh, India
| | - Mohammad Imran Khan
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Artificial Intelligence in Precision Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohd Kamil Hussain
- Department of Chemistry, Govt. Raza P.G. College, Rampur, Uttar Pradesh, India
- M.J.P Rohilkahand University, Bareilly, Uttar Pradesh, India
| |
Collapse
|
10
|
Speina E, Wilczek M, Mieczkowski A. Dimeric Benzodiazepines as Peptide Mimetics to Overcome p53-Dependent Drug Resistance of Tumors. Biomolecules 2023; 13:biom13020291. [PMID: 36830660 PMCID: PMC9953746 DOI: 10.3390/biom13020291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Benzodiazepines that consist of one α- and one β-amino acid residues linked together in a seven-membered heterocyclic ring could be treated as small, rigid, cyclic dipeptides capable of exhibiting a wide range of biological activities. During our research on novel analogues of anthramycin, a tricyclic antibiotic benzodiazepine, we developed the synthesis of two benzodiazepine dimers, obtained through the cyclization of appropriate linear tripeptides. The synthesized compounds were tested on a panel of seven cancer and normal cell lines. The developed molecules exhibited promising cytotoxic activity against the lung cancer cell lines A549 and NCI-H1299 and the epidermoid carcinoma cell line A-431. Moreover, they showed significant selectivity compared to the reference cell lines (BJ-human normal skin fibroblasts and MRC-5-human normal lung cell line). When tested on two isogenic cell lines, HCT116 and HCT116p53-/- (colon cancer), contrary to cisplatin being used as a positive control, the obtained compounds showed a cytotoxic effect independent of the p53 protein status. For the above reasons, the obtained compounds can be considered a new group of promising anticancer agents, useful in the fight against p53-dependent drug resistance in cancers. They can also be treated as convenient, leading structures suitable for further optimization and searching for more active and selective molecules.
Collapse
Affiliation(s)
- Elżbieta Speina
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Poland
| | - Marcin Wilczek
- Department of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Adam Mieczkowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Poland
- Correspondence:
| |
Collapse
|
11
|
Barreca M, Lang N, Tarantelli C, Spriano F, Barraja P, Bertoni F. Antibody-drug conjugates for lymphoma patients: preclinical and clinical evidences. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:763-794. [PMID: 36654819 PMCID: PMC9834635 DOI: 10.37349/etat.2022.00112] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/08/2022] [Indexed: 12/28/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a recent, revolutionary approach for malignancies treatment, designed to provide superior efficacy and specific targeting of tumor cells, compared to systemic cytotoxic chemotherapy. Their structure combines highly potent anti-cancer drugs (payloads or warheads) and monoclonal antibodies (Abs), specific for a tumor-associated antigen, via a chemical linker. Because the sensitive targeting capabilities of monoclonal Abs allow the direct delivery of cytotoxic payloads to tumor cells, these agents leave healthy cells unharmed, reducing toxicity. Different ADCs have been approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for the treatment of a wide range of malignant conditions, both as monotherapy and in combination with chemotherapy, including for lymphoma patients. Over 100 ADCs are under preclinical and clinical investigation worldwide. This paper it provides an overview of approved and promising ADCs in clinical development for the treatment of lymphoma. Each component of the ADC design, their mechanism of action, and the highlights of their clinical development progress are discussed.
Collapse
Affiliation(s)
- Marilia Barreca
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90123 Palermo, Italy
| | - Noémie Lang
- Division of Oncology, Department of Oncology, Faculty of Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Chiara Tarantelli
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland
| | - Filippo Spriano
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland
| | - Paola Barraja
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90123 Palermo, Italy
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| |
Collapse
|
12
|
Covalent DNA Binding Is Essential for Gram-Negative Antibacterial Activity of Broad Spectrum Pyrrolobenzodiazepines. Antibiotics (Basel) 2022; 11:antibiotics11121770. [PMID: 36551427 PMCID: PMC9774941 DOI: 10.3390/antibiotics11121770] [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: 11/01/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
It is urgent to find new antibiotic classes against multidrug-resistant bacteria as the rate of discovery of new classes of antibiotics has been very slow in the last 50 years. Recently, pyrrolobenzodiazepines (PBDs) with a C8-linked aliphatic-heterocycle have been identified as a new broad-spectrum antibiotic class with activity against Gram-negative bacteria. The active imine moiety of the reported lead pyrrolobenzodiazepine compounds was replaced with amide to obtain the non-DNA binding and noncytotoxic dilactam analogues to understand the structure-activity relationship further and improve the safety potential of this class. The synthesised compounds were tested against panels of multidrug-resistant Gram-positive and Gram-negative bacteria, including WHO priority pathogens. Minimum inhibitory concentrations for the dilactam analogues ranged from 4 to 32 mg/L for MDR Gram-positive bacteria, compared to 0.03 to 2 mg/L for the corresponding imine analogues. At the same time, they were found to be inactive against MDR Gram-negative bacteria, with a MIC > 32 mg/L, compared to a MIC of 0.5 to 32 mg/L for imine analogues. A molecular modelling study suggests that the lack of imine functionality also affects the interaction of PBDs with DNA gyrase. This study suggests that the presence of N10-C11 imine moiety is crucial for the broad-spectrum activity of pyrrolobenzodiazepines.
Collapse
|
13
|
Leitis Z, Sakaine G, Kine̅ns A, Smits G. Stereoselective Olefination with Sterically Demanding Julia-Kocienski Reagents: Total Synthesis of Oxo-prothracarcin, Oxo-tomaymycin, and Boseongazepine B. ACS OMEGA 2022; 7:30519-30534. [PMID: 36061714 PMCID: PMC9434771 DOI: 10.1021/acsomega.2c03732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/10/2022] [Indexed: 06/15/2023]
Abstract
Total syntheses of three pyrrolo[1,4]benzodiazepine anticancer antibiotic family members oxo-prothracarcin, oxo-tomaymycin, and boseongazepine B are described. The total syntheses feature late-stage stereoselective olefination employing modified Julia-Kocienski reagents that can be conveniently prepared in only two steps and allows for a significant reduction in the number of linear steps. Detailed density functional theory (DFT) studies explain the stereochemical outcome of the key step.
Collapse
Affiliation(s)
- Zigma̅rs Leitis
- Latvian
Institute of Organic Synthesis, Aizkraukles str. 21, Riga LV-1006, Latvia
| | - Guna Sakaine
- Latvian
Institute of Organic Synthesis, Aizkraukles str. 21, Riga LV-1006, Latvia
| | - Artis Kine̅ns
- Latvian
Institute of Organic Synthesis, Aizkraukles str. 21, Riga LV-1006, Latvia
- Department
of Chemistry, University of Latvia, Jelgavas str. 1, Riga LV-1004, Latvia
| | - Gints Smits
- Latvian
Institute of Organic Synthesis, Aizkraukles str. 21, Riga LV-1006, Latvia
| |
Collapse
|
14
|
Lai W, Zhao S, Lai Q, Zhou W, Wu M, Jiang X, Wang X, Peng Y, Wei X, Ouyang L, Gou L, Chen H, Wang Y, Yang J. Design, Synthesis, and Bioevaluation of a Novel Hybrid Molecular Pyrrolobenzodiazepine-Anthracenecarboxyimide as a Payload for Antibody-Drug Conjugate. J Med Chem 2022; 65:11679-11702. [PMID: 35982539 DOI: 10.1021/acs.jmedchem.2c00471] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A novel series of hybrid molecules combining pyrrolobenzodiazepine (PBD) and anthracenecarboxyimide pharmacophores were designed, synthesized, and tested for in vitro cytotoxicity against various cancer cell lines. The most potent compound from this series, 37b3, exhibited a subnanomolar level of cytotoxicity with an IC50 of 0.17-0.94 nM. 37b3 induced DNA damage and led to tumor cell cycle arrest and apoptosis. We employed 37b3 as a payload to conjugate with trastuzumab to obtain the antibody-drug conjugate (ADC) T-PBA. T-PBA maintained its mode of target and internalization ability of trastuzumab. We demonstrated that T-PBA could be degraded through the lysosomal pathway to release the payload 37b3 after internalization. T-PBA showed a powerful killing effect on Her2-positive cancer cells in vitro. Furthermore, T-PBA significantly inhibited tumor growth in gastric and ovarian cancer xenograft mouse models without overt toxicity. Collectively, these studies suggest that T-PBA represents a promising new ADC that deserves further investigation.
Collapse
Affiliation(s)
- Weirong Lai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Shengyan Zhao
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Qinhuai Lai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Wei Zhou
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Mengdan Wu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Xiaohua Jiang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Xin Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Yujia Peng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Xian Wei
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Lantu Gou
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163 Tennessee, United States
| | - Yuxi Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.,Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| |
Collapse
|
15
|
Li QZ, Hou SH, Kang JC, Lian PF, Hao Y, Chen C, Zhou J, Ding TM, Zhang SY. Bioinspired Palladium-Catalyzed Intramolecular C(sp 3 )-H Activation for the Collective Synthesis of Proline Natural Products. Angew Chem Int Ed Engl 2022; 61:e202207088. [PMID: 35751877 DOI: 10.1002/anie.202207088] [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] [Received: 05/23/2022] [Indexed: 12/18/2022]
Abstract
Bioinspired palladium-catalyzed intramolecular cyclization of amino acid derivatives containing a vinyl iodide moiety by C-H activation enabled rapid access to a wide range of functionalized proline derivatives with an exocyclic olefin. To demonstrate the practicality of this methodology, the functionalized prolines were used as intermediates for the synthesis of several natural products: lucentamycin A, oxotomaymycin, oxoprothracarcin, and barmumycin.
Collapse
Affiliation(s)
- Quan-Zhe Li
- Shanghai Key Laboratory for Molecular Engineer of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China.,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Si-Hua Hou
- Shanghai Key Laboratory for Molecular Engineer of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China.,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Jun-Chen Kang
- Shanghai Key Laboratory for Molecular Engineer of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Peng-Fei Lian
- Shanghai Key Laboratory for Molecular Engineer of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yu Hao
- Shanghai Key Laboratory for Molecular Engineer of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Chao Chen
- Shanghai Key Laboratory for Molecular Engineer of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Jia Zhou
- Shanghai Key Laboratory for Molecular Engineer of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Tong-Mei Ding
- Shanghai Key Laboratory for Molecular Engineer of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shu-Yu Zhang
- Shanghai Key Laboratory for Molecular Engineer of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
16
|
Thadem N, Rajesh M, Balaboina H, Das S. Synthesis of bridgehead-azacycles via dual C-N/C-C annulation of α-amino acids, aminals and maleimides. Org Biomol Chem 2022; 20:6368-6383. [PMID: 35861324 DOI: 10.1039/d2ob01117d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The synthesis of various bridged azacyclic adducts has recently become a reemerging topic due to their bioactive and natural product mimic profiles. Accordingly, herein, we report a method for easy access to succinamide-bridged azacyclic derivatives through the metal-free polarization-controlled dual C-N/C-C annulation of readily available α-amino acids, 2-amino benzaldehydes or pyrrole/indole-2-aldehyde and maleimide substrates. This cascade features a rare dipolarophile-induced diastereo-selective amidative annulation, followed by 3 + 2 cycloaddition as key steps.
Collapse
Affiliation(s)
- Nagender Thadem
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Manda Rajesh
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India.
| | - Harikrishna Balaboina
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India.
| | - Saibal Das
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
17
|
Li QZ, Hou SH, Kang JC, Lian PF, Hao Y, Chen C, Zhou J, Ding TM, Zhang SY. Bioinspired Palladium‐Catalyzed Intramolecular C(sp3)−H Activation for the Collective Synthesis of Proline Natural Products. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202207088] [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)
- Quan-Zhe Li
- Shanghai Jiao Tong University Chemistry CHINA
| | - Si-Hua Hou
- SJTU: Shanghai Jiao Tong University CHEMISTRY CHINA
| | | | | | - Yu Hao
- SJTU: Shanghai Jiao Tong University Chemistry CHINA
| | - Chao Chen
- SJTU: Shanghai Jiao Tong University Chemistry CHINA
| | - Jia Zhou
- SJTU: Shanghai Jiao Tong University Chemistry CHINA
| | | | - Shu-Yu Zhang
- Shanghai Jiao Tong University School of Chemistry and Chemical Engineering 800 Dongchuan RoadB329 Chemsitry BuildingShanghai Jiao Tong University 200240 Shanghai CHINA
| |
Collapse
|
18
|
Debnath U, Verma S, Patra J, Mandal SK. A review on recent synthetic routes and computational approaches for antibody drug conjugation developments used in anti-cancer therapy. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
19
|
Ma C, Wang Y, Chen G, Li J, Jiang Y, Zhang X, Fan X. Divergent construction of 3-(indol-2-yl)succinimide/maleimide and fused benzodiazepine skeletons from 2-(1 H-indol-1-yl)anilines and maleimides. Org Chem Front 2022. [DOI: 10.1039/d2qo00779g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Divergent construction of 3-(indol-2-yl)succinimide/maleimide and indoyl/pyrrolyl fused benzodiazepine skeletons from 2-(1H-indol-1-yl)anilines and maleimides is presented.
Collapse
Affiliation(s)
- Chunhua Ma
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Yue Wang
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Guang Chen
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Jingyi Li
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Yuqin Jiang
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Xinying Zhang
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Xuesen Fan
- NMPA Key Laboratory for Research and Evaluation of Innovative Drug, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| |
Collapse
|
20
|
Farooq S, Ngaini Z. Chalcone derived benzoheterodiazepines for medicinal applications:
A Two‐pot
and
one‐pot
synthetic approach. J Heterocycl Chem 2021. [DOI: 10.1002/jhet.4337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Saba Farooq
- Faculty of Resource Science and Technology Universiti Malaysia Sarawak Kota Samarahan Malaysia
| | - Zainab Ngaini
- Faculty of Resource Science and Technology Universiti Malaysia Sarawak Kota Samarahan Malaysia
| |
Collapse
|
21
|
Fang F, Hu S, Li C, Wang Q, Wang R, Han X, Zhou Y, Liu H. Catalytic System‐Controlled Divergent Reaction Strategies for the Construction of Diversified Spiropyrazolone Skeletons from Pyrazolidinones and Diazopyrazolones. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202105857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Feifei Fang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry China Pharmaceutical University 24 Tong Jia Xiang Nanjing Jiangsu 210009 China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zu Chong Zhi Road Shanghai 201203 China
- School of Pharmaceutical Science and Technology Hangzhou Institute for Advanced Study University of Chinese Academy of Sciences Hangzhou 310024 China
| | - Shulei Hu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry China Pharmaceutical University 24 Tong Jia Xiang Nanjing Jiangsu 210009 China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zu Chong Zhi Road Shanghai 201203 China
- School of Pharmaceutical Science and Technology Hangzhou Institute for Advanced Study University of Chinese Academy of Sciences Hangzhou 310024 China
| | - Chunpu Li
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zu Chong Zhi Road Shanghai 201203 China
- School of Pharmaceutical Science and Technology Hangzhou Institute for Advanced Study University of Chinese Academy of Sciences Hangzhou 310024 China
| | - Qian Wang
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zu Chong Zhi Road Shanghai 201203 China
- School of Pharmaceutical Science and Technology Hangzhou Institute for Advanced Study University of Chinese Academy of Sciences Hangzhou 310024 China
| | - Run Wang
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zu Chong Zhi Road Shanghai 201203 China
- School of Pharmaceutical Science and Technology Hangzhou Institute for Advanced Study University of Chinese Academy of Sciences Hangzhou 310024 China
| | - Xu Han
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zu Chong Zhi Road Shanghai 201203 China
- School of Pharmaceutical Science and Technology Hangzhou Institute for Advanced Study University of Chinese Academy of Sciences Hangzhou 310024 China
| | - Yu Zhou
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zu Chong Zhi Road Shanghai 201203 China
- School of Pharmaceutical Science and Technology Hangzhou Institute for Advanced Study University of Chinese Academy of Sciences Hangzhou 310024 China
| | - Hong Liu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry China Pharmaceutical University 24 Tong Jia Xiang Nanjing Jiangsu 210009 China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences 555 Zu Chong Zhi Road Shanghai 201203 China
- School of Pharmaceutical Science and Technology Hangzhou Institute for Advanced Study University of Chinese Academy of Sciences Hangzhou 310024 China
| |
Collapse
|
22
|
Nudelman A. Dimeric Drugs. Curr Med Chem 2021; 29:2751-2845. [PMID: 34375175 DOI: 10.2174/0929867328666210810124159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/18/2021] [Accepted: 06/29/2021] [Indexed: 11/22/2022]
Abstract
This review intends to summarize the structures of an extensive number of symmetrical-dimeric drugs, having two monomers linked via a bridging entity while emphasizing the large versatility of biologically active substances reported to possess dimeric structures. The largest number of classes of these compounds consist of anticancer agents, antibiotics/antimicrobials, and anti-AIDS drugs. Other symmetrical-dimeric drugs include antidiabetics, antidepressants, analgesics, anti-inflammatories, drugs for the treatment of Alzheimer's disease, anticholesterolemics, estrogenics, antioxidants, enzyme inhibitors, anti-Parkisonians, laxatives, antiallergy compounds, cannabinoids, etc. Most of the articles reviewed do not compare the activity/potency of the dimers to that of their corresponding monomers. Only in limited cases, various suggestions have been made to justify unexpected higher activity of the dimers vs. the corresponding monomers. These suggestions include statistical effects, the presence of dimeric receptors, binding of a dimer to two receptors simultaneously, and others. It is virtually impossible to predict which dimers will be preferable to their respective monomers, or which linking bridges will lead to the most active compounds. It is expected that the extensive number of articles summarized, and the large variety of substances mentioned, which display various biological activities, should be of interest to many academic and industrial medicinal chemists.
Collapse
Affiliation(s)
- Abraham Nudelman
- Chemistry Department, Bar Ilan University, Ramat Gan 52900, Israel
| |
Collapse
|
23
|
Vézina-Dawod S, Perreault M, Guay LD, Gerber N, Gobeil S, Biron E. Synthesis and biological evaluation of novel 1,4-benzodiazepin-3-one derivatives as potential antitumor agents against prostate cancer. Bioorg Med Chem 2021; 45:116314. [PMID: 34333393 DOI: 10.1016/j.bmc.2021.116314] [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: 04/13/2021] [Revised: 07/08/2021] [Accepted: 07/15/2021] [Indexed: 12/24/2022]
Abstract
A novel tumor suppressing agent was discovered against PC-3 prostate cancer cells from the screening of a 1,4-benzodiazepin-3-one library. In this study, 96 highly diversified 2,4,5-trisubstituted 1,4-benzodiazepin-3-one derivatives were prepared by a two-step approach using sequential Ugi multicomponent reaction and simultaneous deprotection and cyclization to afford pure compounds bearing a wide variety of substituents. The most promising compound showed a potent and selective antiproliferative activity against prostate cancer cell line PC-3 (GI50 = 10.2 µM), but had no effect on LNCAP, LAPC4 and DU145 cell lines. The compound was initially prepared as a mixture of two diastereomers and after their separation by HPLC, similar antiproliferative activities against PC-3 cells were observed for both diastereomers (2S,5S: GI50 = 10.8 µM and 2S,5R: GI50 = 7.0 µM). Additionally, both diastereomers showed comparable stability profiles after incubation with human liver microsomes. Finally, in vivo evaluation of the hit compound with the chick chorioallantoic membrane xenograft assay revealed a good toxicity profile and significant antitumor activity after intravenous injection.
Collapse
Affiliation(s)
- Simon Vézina-Dawod
- Faculté de pharmacie, Université Laval, Québec, QC G1V 0A6, Canada; Centre de recherche du CHU de Québec-Université Laval, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Martin Perreault
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada; Centre de recherche du CHU de Québec-Université Laval, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Louis-David Guay
- Faculté de pharmacie, Université Laval, Québec, QC G1V 0A6, Canada; Centre de recherche du CHU de Québec-Université Laval, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Nicolas Gerber
- Faculté de pharmacie, Université Laval, Québec, QC G1V 0A6, Canada; Centre de recherche du CHU de Québec-Université Laval, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Stéphane Gobeil
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada; Centre de recherche du CHU de Québec-Université Laval, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Eric Biron
- Faculté de pharmacie, Université Laval, Québec, QC G1V 0A6, Canada; Centre de recherche du CHU de Québec-Université Laval, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada.
| |
Collapse
|
24
|
Fang F, Hu S, Li C, Wang Q, Wang R, Han X, Zhou Y, Liu H. Catalytic System-Controlled Divergent Reaction Strategies for the Construction of Diversified Spiropyrazolone Skeletons from Pyrazolidinones and Diazopyrazolones. Angew Chem Int Ed Engl 2021; 60:21327-21333. [PMID: 34180572 DOI: 10.1002/anie.202105857] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/15/2021] [Indexed: 12/13/2022]
Abstract
A catalytic system-controlled divergent reaction strategy was here reported to construct four types of intriguing spiroheterocyclic skeletons from simple and readily available starting materials via a precise chemical bond activation/[n+1] annulation cascade. The tetraazaspiroheterocyclic and trizazspiroheterocyclic scaffolds could be independently constructed by a selective N-N bond activation/[n+1] annulation cascade, a C(sp2 )-H activation/[4+1] annulation and a novel tandem C(sp2 )-H/C(sp3 )-H bond activation/[4+1] annulation strategy, along with a broad scope of substrates, moderate to excellent yields and valuable transformations. More importantly, in these transformations, we are the first time to capture a N-N bond activation and a C(sp3 )-H bond activation of pyrazolidinones under different catalytic system.
Collapse
Affiliation(s)
- Feifei Fang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, 210009, China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Shulei Hu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, 210009, China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Chunpu Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Qian Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Run Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Xu Han
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Yu Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Hong Liu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, Jiangsu, 210009, China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| |
Collapse
|
25
|
Hadiyal SD, Lalpara JN, Parmar ND, Joshi HS. Microwave Irradiated Targeted Synthesis of Pyrrolobenzodiazepine Embrace 1,2,3-Triazole by Click Chemistry Synthetic Aspect and Evaluation of Anticancer and Antimicrobial Activity. Polycycl Aromat Compd 2021. [DOI: 10.1080/10406638.2021.1913425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Sanjay D. Hadiyal
- Department of Chemistry, Saurashtra University, Rajkot, Gujarat, India
- School of Science, Department of Chemistry, RK University, Rajkot, Gujarat, India
| | - Jaydeep N. Lalpara
- School of Science, Department of Chemistry, RK University, Rajkot, Gujarat, India
| | - Nilesh D. Parmar
- Department of Chemistry, Saurashtra University, Rajkot, Gujarat, India
| | - Hitendra S. Joshi
- Department of Chemistry, Saurashtra University, Rajkot, Gujarat, India
| |
Collapse
|
26
|
Holte D, Rao M, Huters A, Simanis J, Califano JC, Kempema A, Levy JN. Early Development, Scale-Up, and Reverse-Phase Purification of a Highly Potent Pyrrolobenzodiazepine Dimer, SG3259, for Use in Antibody–Drug Conjugates. Org Process Res Dev 2021. [DOI: 10.1021/acs.oprd.0c00497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Dane Holte
- AbbVie Process Research and Development, 995 East Arques Avenue, Sunnyvale, California 94085, United States
| | - Meera Rao
- AbbVie Research & Development, 450 East Jamie Court, South San Francisco, California 94080, United States
| | - Alexander Huters
- AbbVie Process Research and Development, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Justin Simanis
- AbbVie Process Research and Development, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Jean-Christophe Califano
- AbbVie Process Research and Development, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Aaron Kempema
- AbbVie Research & Development, 450 East Jamie Court, South San Francisco, California 94080, United States
| | - Jean-Noel Levy
- AbbVie Research & Development, 450 East Jamie Court, South San Francisco, California 94080, United States
| |
Collapse
|
27
|
Sakaine G, Ture A, Pedroni J, Smits G. Isolation, chemistry, and biology of pyrrolo[1,4]benzodiazepine natural products. Med Res Rev 2021; 42:5-55. [PMID: 33846985 DOI: 10.1002/med.21803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 02/11/2021] [Accepted: 03/23/2021] [Indexed: 11/06/2022]
Abstract
The isolation of the antitumor antibiotic anthramycin in the 1960s prompted extensive research into pyrrolo[1,4]benzodiazepines (PBD) as potential therapeutics for the treatment of cancers. Since then, nearly 60 PBD natural products have been isolated and evaluated with regard to their biological activity. Synthetic studies and total syntheses have enabled access to PBD analogues, culminating in the development of highly potent anticancer agents. This review provides a summary of the occurrence and biological activity of PBD natural products and covers the strategies employed for their total syntheses.
Collapse
Affiliation(s)
- Guna Sakaine
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | | | - Julia Pedroni
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Gints Smits
- Latvian Institute of Organic Synthesis, Riga, Latvia
| |
Collapse
|
28
|
Lewis T, Corcoran DB, Thurston DE, Giles PJ, Ashelford K, Walsby EJ, Fegan CD, Pepper AGS, Miraz Rahman K, Pepper C. Novel pyrrolobenzodiazepine benzofused hybrid molecules inhibit NF-κB activity and synergise with bortezomib and ibrutinib in hematological cancers. Haematologica 2021; 106:958-967. [PMID: 32381576 PMCID: PMC8018133 DOI: 10.3324/haematol.2019.238584] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Indexed: 12/28/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) and multiple myeloma are incurable hematologic malignancies that are pathologically linked with aberrant nuclear factor-kappa B (NF-κB) activation. In this study, we identified a group of novel C8-linked benzofused pyrrolo[2,1- c][1,4]benzodiazepine monomeric hybrids capable of sequence-selective inhibition of NF-κB with low nanomolar LD50 values in CLL (n=46) and multiple myeloma cell lines (n=5). The lead compound, DC-1-192, significantly inhibited NF-κB DNA binding after just 4 h of exposure, demonstrating inhibitory effects on both canonical and non-canonical NF-κB subunits. In primary CLL cells, sensitivity to DC-1-192 was inversely correlated with RelA subunit expression (r2=0.2) and samples with BIRC3 or NOTCH1 mutations showed increased sensitivity (P=0.001). RNAsequencing and gene set enrichment analysis confirmed the over-representation of NF-κB regulated genes in the downregulated gene list. Furthermore, in vivo efficacy studies in NOD/SCID mice, using a systemic RPMI 8226 human multiple myeloma xenograft model, showed that DC- 1-192 significantly prolonged survival (P=0.017). In addition, DC1-192 showed synergy with bortezomib and ibrutinib; synergy with ibrutinib was enhanced when CLL cells were co-cultured on CD40L-expressing fibroblasts in order to mimic the cytoprotective lymph node microenvironment (P=0.01). Given that NF-κB plays a role in both bortezomib and ibrutinib resistance mechanisms, these data provide a strong rationale for the use of DC-1-192 in the treatment of NF-κB-driven cancers, particularly in the context of relapsed/refractory disease.
Collapse
Affiliation(s)
- Thomas Lewis
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - David B Corcoran
- School of Cancer and Pharmaceutical Science, King College London, UK
| | - David E Thurston
- School of Cancer and Pharmaceutical Science, King College London, UK
| | - Peter J Giles
- Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Kevin Ashelford
- Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Elisabeth J Walsby
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Christopher D Fegan
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Andrea G S Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | | | - Chris Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| |
Collapse
|
29
|
Sivaprakasam P, McDonald I, Iwuagwu C, Chowdari NS, Peese KM, Langley DR, Cheng H, Luzung MR, Schmidt MA, Zheng B, Tan Y, Cho P, Rakshit S, Lakshminarasimhan T, Guturi S, Kanagavel K, Kanusu U, Niyogi AG, Sidhar S, Vaidyanathan R, Eastgate MD, Kotapati S, Deshpande M, Pan C, Cardarelli PM, Xie C, Rao C, Holder P, Sarma G, Vite G, Gangwar S. DNA-Model-Based Design and Execution of Some Fused Benzodiazepine Hybrid Payloads for Antibody-Drug Conjugate Modality. ACS Med Chem Lett 2021; 12:404-412. [PMID: 33738068 DOI: 10.1021/acsmedchemlett.0c00578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/27/2021] [Indexed: 11/28/2022] Open
Abstract
A new series with the tetrahydroisoquinoline-fused benzodiazepine (TBD) ring system combined with the surrogates of (1-methyl-1H-pyrrol-3-yl)benzene ("MPB") payloads were designed and executed for conjugation with a monoclonal antibody for anticancer therapeutics. DNA models helped in rationally identifying modifications of the "MPB" binding component and guided structure-activity relationship generation. This hybrid series of payloads exhibited excellent in vitro activity when tested against a panel of various cancer cell lines. One of the payloads was appended with a lysosome-cleavable peptide linker and conjugated with an anti-mesothelin antibody via a site-specific conjugation method mediated by the enzyme bacterial transglutaminase (BTGase). Antibody-drug conjugate (ADC) 50 demonstrated good plasma stability and lysosomal cleavage. A single intravenous dose of ADC 50 (5 or 10 nmol/kg) showed robust efficacy in an N87 gastric cancer xenograft model.
Collapse
Affiliation(s)
- Prasanna Sivaprakasam
- Computer-Aided Drug Design, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Ivar McDonald
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Christiana Iwuagwu
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Naidu S. Chowdari
- Discovery Chemistry, Bristol-Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, United States
| | - Kevin M. Peese
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - David R. Langley
- Computer-Aided Drug Design, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, Connecticut 06492, United States
| | - Heng Cheng
- Discovery Chemistry, Bristol-Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, United States
| | - Michael R. Luzung
- Chemical and Synthetic Development, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Michael A. Schmidt
- Chemical and Synthetic Development, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Bin Zheng
- Chemical and Synthetic Development, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Yichen Tan
- Chemical and Synthetic Development, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Patricia Cho
- Chemical and Synthetic Development, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Souvik Rakshit
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Thirumalai Lakshminarasimhan
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Sivakrishna Guturi
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Kishorekumar Kanagavel
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Umamaheswararao Kanusu
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Ankita G. Niyogi
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Somprabha Sidhar
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Rajappa Vaidyanathan
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Martin D. Eastgate
- Chemical and Synthetic Development, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Srikanth Kotapati
- Discovery Biotherapeutics, Bristol-Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, United States
| | - Madhura Deshpande
- Discovery Biotherapeutics, Bristol-Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, United States
| | - Chin Pan
- Cell Biology and Pharmacology, Bristol-Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, United States
| | - Pina M. Cardarelli
- Cell Biology and Pharmacology, Bristol-Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, United States
| | - Chunshan Xie
- Lead Discovery and Optimization, Bristol-Myers Squibb Research and Development, P.O.
Box 4000, Princeton, New Jersey 08543, United States
| | - Chetana Rao
- Discovery Biotherapeutics, Bristol-Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, United States
| | - Patrick Holder
- Discovery Biotherapeutics, Bristol-Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, United States
| | - Ganapathy Sarma
- Discovery Biotherapeutics, Bristol-Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, United States
| | - Gregory Vite
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Sanjeev Gangwar
- Discovery Chemistry, Bristol-Myers Squibb Research & Development, 700 Bay Road, Redwood City, California 94063, United States
| |
Collapse
|
30
|
Majeed K, Wang L, Liu B, Guo Z, Zhou F, Zhang Q. Metal-Free Tandem Approach for Triazole-Fused Diazepinone Scaffolds via [3 + 2]Cycloaddition/C–N Coupling Reaction. J Org Chem 2020; 86:207-222. [DOI: 10.1021/acs.joc.0c02022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Kashif Majeed
- Xi’an Key Laboratory of Functional Organic Porous Materials, Department of Applied Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi 710072, P. R. China
| | - Lingna Wang
- Xi’an Key Laboratory of Functional Organic Porous Materials, Department of Applied Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi 710072, P. R. China
| | - Bangjie Liu
- Xi’an Key Laboratory of Functional Organic Porous Materials, Department of Applied Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi 710072, P. R. China
| | - Zijian Guo
- Xi’an Key Laboratory of Functional Organic Porous Materials, Department of Applied Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi 710072, P. R. China
| | - Fengtao Zhou
- Xi’an Key Laboratory of Functional Organic Porous Materials, Department of Applied Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi 710072, P. R. China
| | - Qiuyu Zhang
- Xi’an Key Laboratory of Functional Organic Porous Materials, Department of Applied Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi 710072, P. R. China
| |
Collapse
|
31
|
Shirsath SR, Ghotekar GS, Bahadur V, Gonnade RG, Muthukrishnan M. Silver-Catalyzed Cascade Cyclization/1,6-Conjugate Addition of Homopropargyl Sulfonamides to p-Quinone Methides: An Approach to Diverse 3-Diarylmethine Substituted Dihydropyrroles. J Org Chem 2020; 85:15038-15050. [DOI: 10.1021/acs.joc.0c01922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Sachin R. Shirsath
- Division of Organic Chemistry, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ganesh S. Ghotekar
- Division of Organic Chemistry, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vir Bahadur
- Centre for Materials Characterization, CSIR-National Chemical Laboratory, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rajesh G. Gonnade
- Centre for Materials Characterization, CSIR-National Chemical Laboratory, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - M. Muthukrishnan
- Division of Organic Chemistry, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
32
|
Velasco‐Rubio Á, Varela JA, Saá C. Recent Advances in Transition‐Metal‐Catalyzed Oxidative Annulations to Benzazepines and Benzodiazepines. Adv Synth Catal 2020. [DOI: 10.1002/adsc.202000808] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Álvaro Velasco‐Rubio
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS) Departamento de Química Orgánica Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| | - Jesús A. Varela
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS) Departamento de Química Orgánica Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| | - Carlos Saá
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS) Departamento de Química Orgánica Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| |
Collapse
|
33
|
Rogers CM, Simmons Iii RH, Fluhler Thornburg GE, Buehler NJ, Bochman ML. Fanconi anemia-independent DNA inter-strand crosslink repair in eukaryotes. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 158:33-46. [PMID: 32877700 DOI: 10.1016/j.pbiomolbio.2020.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023]
Abstract
DNA inter-strand crosslinks (ICLs) are dangerous lesions that can be caused by a variety of endogenous and exogenous bifunctional compounds. Because covalently linking both strands of the double helix locally disrupts DNA replication and transcription, failure to remove even a single ICL can be fatal to the cell. Thus, multiple ICL repair pathways have evolved, with the best studied being the canonical Fanconi anemia (FA) pathway. However, recent research demonstrates that different types of ICLs (e.g., backbone distorting vs. non-distorting) can be discriminated by the cell, which then mounts a specific repair response using the FA pathway or one of a variety of FA-independent ICL repair pathways. This review focuses on the latter, covering current work on the transcription-coupled, base excision, acetaldehyde-induced, and SNM1A/RecQ4 ICL repair pathways and highlighting unanswered questions in the field. Answering these questions will provide mechanistic insight into the various pathways of ICL repair and enable ICL-inducing agents to be more effectively used as chemotherapeutics.
Collapse
Affiliation(s)
- Cody M Rogers
- Molecular and Cellular Biochemistry Department, Indiana University, 212 S. Hawthorne Dr., Simon Hall MSB1 room 405B, Bloomington, IN, 47405, USA
| | - Robert H Simmons Iii
- Molecular and Cellular Biochemistry Department, Indiana University, 212 S. Hawthorne Dr., Simon Hall MSB1 room 405B, Bloomington, IN, 47405, USA
| | - Gabriella E Fluhler Thornburg
- Molecular and Cellular Biochemistry Department, Indiana University, 212 S. Hawthorne Dr., Simon Hall MSB1 room 405B, Bloomington, IN, 47405, USA
| | - Nicholas J Buehler
- Molecular and Cellular Biochemistry Department, Indiana University, 212 S. Hawthorne Dr., Simon Hall MSB1 room 405B, Bloomington, IN, 47405, USA
| | - Matthew L Bochman
- Molecular and Cellular Biochemistry Department, Indiana University, 212 S. Hawthorne Dr., Simon Hall MSB1 room 405B, Bloomington, IN, 47405, USA.
| |
Collapse
|
34
|
Antibody-Drug Conjugates: The New Frontier of Chemotherapy. Int J Mol Sci 2020; 21:ijms21155510. [PMID: 32752132 PMCID: PMC7432430 DOI: 10.3390/ijms21155510] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/15/2022] Open
Abstract
In recent years, antibody-drug conjugates (ADCs) have become promising antitumor agents to be used as one of the tools in personalized cancer medicine. ADCs are comprised of a drug with cytotoxic activity cross-linked to a monoclonal antibody, targeting antigens expressed at higher levels on tumor cells than on normal cells. By providing a selective targeting mechanism for cytotoxic drugs, ADCs improve the therapeutic index in clinical practice. In this review, the chemistry of ADC linker conjugation together with strategies adopted to improve antibody tolerability (by reducing antigenicity) are examined, with particular attention to ADCs approved by the regulatory agencies (the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA)) for treating cancer patients. Recent developments in engineering Immunoglobulin (Ig) genes and antibody humanization have greatly reduced some of the problems of the first generation of ADCs, beset by problems, such as random coupling of the payload and immunogenicity of the antibody. ADC development and clinical use is a fast, evolving area, and will likely prove an important modality for the treatment of cancer in the near future.
Collapse
|
35
|
Picconi P, Hind CK, Nahar KS, Jamshidi S, Di Maggio L, Saeed N, Evans B, Solomons J, Wand ME, Sutton JM, Rahman KM. New Broad-Spectrum Antibiotics Containing a Pyrrolobenzodiazepine Ring with Activity against Multidrug-Resistant Gram-Negative Bacteria. J Med Chem 2020; 63:6941-6958. [PMID: 32515951 DOI: 10.1021/acs.jmedchem.0c00328] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
It is urgent to find new antibiotic classes with activity against multidrug-resistant (MDR) Gram-negative pathogens as the pipeline of antibiotics is essentially empty. Modified pyrrolobenzodiazepines with a C8-linked aliphatic heterocycle provide a new class of broad-spectrum antibacterial agents with activity against MDR Gram-negative bacteria, including WHO priority pathogens. The structure-activity relationship established that the third ring was particularly important for Gram-negative activity. Minimum inhibitory concentrations for the lead compounds ranged from 0.125 to 2 mg/L for MDR Gram-negative, excluding Pseudomonas aeruginosa, and between 0.03 and 1 mg/L for MDR Gram-positive species. The lead compounds were rapidly bactericidal with >5 log reduction in viable count within 4 h for Acinetobacter baumannii and Klebsiella pneumoniae. The lead compound inhibited DNA gyrase in gel-based assays, with an IC50 of 3.16 ± 1.36 mg/L. This study provides a new chemical scaffold for developing novel broad-spectrum antibiotics which can help replenish the pipeline of antibiotics.
Collapse
Affiliation(s)
- Pietro Picconi
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, U.K
| | - Charlotte K Hind
- National Infections Service, Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, U.K
| | - Kazi S Nahar
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, U.K
| | - Shirin Jamshidi
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, U.K
| | - Lucia Di Maggio
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, U.K
| | - Naima Saeed
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, U.K
| | - Bonnie Evans
- National Infections Service, Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, U.K
| | - Jessica Solomons
- National Infections Service, Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, U.K
| | - Matthew E Wand
- National Infections Service, Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, U.K
| | - J Mark Sutton
- National Infections Service, Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, U.K
| | | |
Collapse
|
36
|
Sakaine G, Smits G, Arsenyan P. Synthetic studies toward novel pyrrolobenzodiazepine–coumarin hybrids. Chem Heterocycl Compd (N Y) 2020. [DOI: 10.1007/s10593-020-02702-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
37
|
Vandera KKA, Picconi P, Valero M, González-Gaitano G, Woods A, Zain NMM, Bruce KD, Clifton LA, Skoda MWA, Rahman KM, Harvey RD, Dreiss CA. Antibiotic-in-Cyclodextrin-in-Liposomes: Formulation Development and Interactions with Model Bacterial Membranes. Mol Pharm 2020; 17:2354-2369. [DOI: 10.1021/acs.molpharmaceut.0c00096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Kalliopi-Kelli A. Vandera
- School of Cancer & Pharmaceutical Science, Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Pietro Picconi
- School of Cancer & Pharmaceutical Science, Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Margarita Valero
- Department of Physical Chemistry, University of Salamanca, ES E-37007 Salamanca, Spain
| | | | - Arcadia Woods
- School of Cancer & Pharmaceutical Science, Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Nur Masirah M. Zain
- School of Cancer & Pharmaceutical Science, Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Kenneth D. Bruce
- School of Cancer & Pharmaceutical Science, Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Luke A. Clifton
- Rutherford Appleton Laboratory, ISIS, 1-27, R3, Harwell Campus, Didcot OX11 0QX, U.K
| | | | - Khondaker Miraz Rahman
- School of Cancer & Pharmaceutical Science, Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Richard D. Harvey
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, Vienna, Austria
| | - Cécile A. Dreiss
- School of Cancer & Pharmaceutical Science, Institute of Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| |
Collapse
|
38
|
A Novel Antibody-Drug Conjugate (ADC) Delivering a DNA Mono-Alkylating Payload to Chondroitin Sulfate Proteoglycan (CSPG4)-Expressing Melanoma. Cancers (Basel) 2020; 12:cancers12041029. [PMID: 32331483 PMCID: PMC7226475 DOI: 10.3390/cancers12041029] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/13/2020] [Accepted: 04/18/2020] [Indexed: 12/18/2022] Open
Abstract
Despite emerging targeted and immunotherapy treatments, no monoclonal antibodies or antibody-drug conjugates (ADCs) directly targeting tumor cells are currently approved for melanoma therapy. The tumor-associated antigen chondroitin sulphate proteoglycan 4 (CSPG4), a neural crest glycoprotein over-expressed on 70% of melanomas, contributes to proliferative signaling pathways, but despite highly tumor-selective expression it has not yet been targeted using ADCs. We developed a novel ADC comprising an anti-CSPG4 antibody linked to a DNA minor groove-binding agent belonging to the novel pyrridinobenzodiazepine (PDD) class. Unlike conventional DNA-interactive pyrrolobenzodiazepine (PBD) dimer payloads that cross-link DNA, PDD-based payloads are mono-alkylating agents but have similar efficacy and substantially enhanced tolerability profiles compared to PBD-based cross-linkers. We investigated the anti-tumor activity and safety of the anti-CSPG4-(PDD) ADC in vitro and in human melanoma xenografts. Anti-CSPG4-(PDD) inhibited CSPG4-expressing melanoma cell growth and colony formation and triggered apoptosis in vitro at low nanomolar to picomolar concentrations without off-target Fab-mediated or Fc-mediated toxicity. Anti-CSPG4-(PDD) restricted xenograft growth in vivo at 2 mg/kg doses. One 5 mg/kg injection triggered tumor regression in the absence of overt toxic effects or of acquired residual tumor cell resistance. This anti-CSPG4-(PDD) can deliver a highly cytotoxic DNA mono-alkylating payload to CSPG4-expressing tumors at doses tolerated in vivo.
Collapse
|
39
|
Vlahov IR, Qi L, Santhapuram HKR, Felten A, Parham GL, Zou N, Wang K, You F, Vaughn JF, Hahn SJ, Klein HF, Kleindl PJ, Reddy J, Reno D, Nicoson J, Leamon CP. Design and synthesis of a folate-receptor targeted diazepine-ring-opened pyrrolobenzodiazepine prodrug conjugate. Bioorg Med Chem Lett 2020; 30:126987. [DOI: 10.1016/j.bmcl.2020.126987] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/13/2020] [Accepted: 01/23/2020] [Indexed: 12/20/2022]
|
40
|
Pertejo P, Carreira-Barral I, Peña-Calleja P, Quesada R, García-Valverde M. Post-Ugi Transformations for the Access to Pyrrolobenzodiazepine Scaffolds with Different Degrees of Unsaturation. J Org Chem 2020; 85:2291-2302. [PMID: 31927929 DOI: 10.1021/acs.joc.9b02995] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The synthesis of three novel families of pyrrolo[2,1-c][1,4]benzodiazepine-5-ones is described. The compounds were prepared according to a three-step sequence, involving an Ugi reaction, building of the pyrrolo nucleus, and reduction-cyclization to the corresponding diazepine. Depending on the amine employed in the synthesis of the Ugi adducts, different unsaturation degrees could be obtained in the pyrrolo ring (saturated or with endo or exo unsaturations), a key feature determining their biological activity, as it affects the affinity of the pyrrolobenzodiazepines toward DNA and thus their cytotoxicity. This synthetic methodology represents a significant improvement with respect to those described in the literature so far, as it uses inexpensive and commercially available starting materials without needing derivatization or the use of protecting groups.
Collapse
Affiliation(s)
- Pablo Pertejo
- Department of Chemistry, Faculty of Science , University of Burgos , 09001 Burgos , Spain
| | - Israel Carreira-Barral
- Department of Chemistry, Faculty of Science , University of Burgos , 09001 Burgos , Spain
| | - Pablo Peña-Calleja
- Department of Chemistry, Faculty of Science , University of Burgos , 09001 Burgos , Spain
| | - Roberto Quesada
- Department of Chemistry, Faculty of Science , University of Burgos , 09001 Burgos , Spain
| | - María García-Valverde
- Department of Chemistry, Faculty of Science , University of Burgos , 09001 Burgos , Spain
| |
Collapse
|
41
|
Benedetti F, Perrin MA, Bosc S, Chouteau F, Champion N, Bigot A. Total Synthesis of (+)-Oxo-tomaymycin. Org Process Res Dev 2020. [DOI: 10.1021/acs.oprd.0c00009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
| | | | - Sebastien Bosc
- Novasep Synthesis, 497 route de Givors, BP 9, 38670 Chasse-Sur-Rhône, France
| | - Franck Chouteau
- Novasep Synthesis, 497 route de Givors, BP 9, 38670 Chasse-Sur-Rhône, France
| | - Nicolas Champion
- Novasep Synthesis, 497 route de Givors, BP 9, 38670 Chasse-Sur-Rhône, France
| | | |
Collapse
|
42
|
Sequential decarboxylative [3+2] cycloaddition and Staudinger/aza-Wittig reactions for diastereoselective synthesis of tetrahydro-pyrroloquinazolines and tetrahedro-pyrrolobenzodiazepines. Tetrahedron Lett 2020. [DOI: 10.1016/j.tetlet.2019.151392] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
43
|
Zhang L, Xu Y, Zhang X, Zhang X, Fan X. Synthesis of pyrazolone fused benzodiazepines via Rh(iii)-catalyzed [4 + 3] annulation of 1-phenylpyrazolidinones with propargyl alcohols. Org Chem Front 2020. [DOI: 10.1039/d0qo00657b] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
An efficient synthesis of pyrazolone fused benzodiazepines through an unprecedented regioselective [4 + 3] annulation of 1-phenylpyrazolidinones with propargyl alcohols via Rh(iii)-catalyzed redox-neutral C–H/N–H/C–O bond activation is presented.
Collapse
Affiliation(s)
- Linghua Zhang
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- School of Chemistry and Chemical Engineering
| | - Yuanshuang Xu
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- School of Chemistry and Chemical Engineering
| | - Xiaopeng Zhang
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- School of Chemistry and Chemical Engineering
| | - Xinying Zhang
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- School of Chemistry and Chemical Engineering
| | - Xuesen Fan
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- School of Chemistry and Chemical Engineering
| |
Collapse
|
44
|
Cui X, Song P, Zhang L. [New Advances in the Treatment for Small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2019; 22:355-362. [PMID: 31196369 PMCID: PMC6580078 DOI: 10.3779/j.issn.1009-3419.2019.06.05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Small cell lung cancer (SCLC) is a refractory cancer with high degree of malignancy, rapid disease progression, poor prognosis and easy recurrence. In the past 30 years, the traditional treatment of SCLC, mainly chemotherapy and radiotherapy, has not changed significantly, and the effective treatment method for clinical needs is extremely urgent. The rapid development of precision medicine has revealed the molecular biological characteristics of SCLC, so its diagnosis and treatment will into a new era. At present, some studies have shown that anti-angiogenic drugs, immunotherapy and so on have improved the efficacy of SCLC treatment to some extent, and there are more studies on the diagnosis and treatment of SCLC, so a new field of SCLC treatment are coming and bringing more survival benefits to patients. New studies on targeted therapy, anti-angiogenesis drugs and immunotherapy of molecular pathology of SCLC are emerging. This paper reviews the new diagnosis and treatment methods of SCLC to provide new guidance for its clinical treatment.
.
Collapse
Affiliation(s)
- Xiaoxia Cui
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Peng Song
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
45
|
A new route to 1,2,3-triazole fused benzooxazepine and benzodiazepine analogues through metal-free intramolecular azide-olefin oxidative cycloaddition. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2019.151164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
46
|
Zhang D, Dragovich PS, Yu SF, Ma Y, Pillow TH, Sadowsky JD, Su D, Wang W, Polson A, Khojasteh SC, Hop CECA. Exposure-Efficacy Analysis of Antibody-Drug Conjugates Delivering an Excessive Level of Payload to Tissues. Drug Metab Dispos 2019; 47:1146-1155. [PMID: 31358513 DOI: 10.1124/dmd.119.087023] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 07/26/2019] [Indexed: 02/13/2025] Open
Abstract
Antibody-drug conjugates (ADCs) contain a disease-receptor antibody and a payload drug connected via a linker. The payload delivery depends on both tumor properties and ADC characteristics. In this study, we used different linkers, attachment sites, and doses to modulate payload delivery of several ADCs bearing maytansinoids (e.g., DM1), auristatins (e.g., MMAE), and DNA alkylating agents [e.g., pyrrolo[2,1-c][1,4]benzodiazepine-dimer (PBD)] as payloads in HER2- or CD22-expressing xenograft models. The tumor growth inhibition and ADC stability and exposure data were collected and analyzed from these dosed animals. The trend analysis suggests that intratumoral payload exposures that directly related the combination of conjugate linker and dose correlate with the corresponding efficacies of three payload types in two antigen-expressing xenograft models. These preliminary correlations also suggest that a minimal threshold concentration of intratumoral payload is required to support sustained efficacy. In addition, an ADC can deliver an excessive level of payload to tumors that does not enhance efficacy ("Plateau" effect). In contrast to tumor payload concentrations, the assessments of systemic exposures of total antibody (Tab) as well as the linker, dose, site of attachment, plasma stability, and drug-to-antibody ratio changes of these ADCs did not consistently rationalize the observed ADC efficacies. The requirement of a threshold payload concentration for efficacy is further supported by dose fractionation studies with DM1-, MMAE-, and PBD-containing ADCs, which demonstrated that single-dose regimens showed better efficacies than fractionated dosing. Overall, this study demonstrates that 1) the linker and dose together determine the tissue payload concentration that correlates with the antitumor efficacy of ADCs and 2) an ADC can deliver an unnecessary level of payload to tumors in xenograft models.
Collapse
Affiliation(s)
- Donglu Zhang
- Drug Metabolism & Pharmacokinetics (D.Z., Y.M., D.S., W.W., S.C.K., C.E.C.A.H.), Discovery Chemistry (P.S.D., T.H.P.), Translational Oncology (S.-F.Y., A.P.), and Protein Chemistry (J.D.S.), Genentech, South San Francisco, California
| | - Peter S Dragovich
- Drug Metabolism & Pharmacokinetics (D.Z., Y.M., D.S., W.W., S.C.K., C.E.C.A.H.), Discovery Chemistry (P.S.D., T.H.P.), Translational Oncology (S.-F.Y., A.P.), and Protein Chemistry (J.D.S.), Genentech, South San Francisco, California
| | - Shang-Fan Yu
- Drug Metabolism & Pharmacokinetics (D.Z., Y.M., D.S., W.W., S.C.K., C.E.C.A.H.), Discovery Chemistry (P.S.D., T.H.P.), Translational Oncology (S.-F.Y., A.P.), and Protein Chemistry (J.D.S.), Genentech, South San Francisco, California
| | - Yong Ma
- Drug Metabolism & Pharmacokinetics (D.Z., Y.M., D.S., W.W., S.C.K., C.E.C.A.H.), Discovery Chemistry (P.S.D., T.H.P.), Translational Oncology (S.-F.Y., A.P.), and Protein Chemistry (J.D.S.), Genentech, South San Francisco, California
| | - Thomas H Pillow
- Drug Metabolism & Pharmacokinetics (D.Z., Y.M., D.S., W.W., S.C.K., C.E.C.A.H.), Discovery Chemistry (P.S.D., T.H.P.), Translational Oncology (S.-F.Y., A.P.), and Protein Chemistry (J.D.S.), Genentech, South San Francisco, California
| | - Jack D Sadowsky
- Drug Metabolism & Pharmacokinetics (D.Z., Y.M., D.S., W.W., S.C.K., C.E.C.A.H.), Discovery Chemistry (P.S.D., T.H.P.), Translational Oncology (S.-F.Y., A.P.), and Protein Chemistry (J.D.S.), Genentech, South San Francisco, California
| | - Dian Su
- Drug Metabolism & Pharmacokinetics (D.Z., Y.M., D.S., W.W., S.C.K., C.E.C.A.H.), Discovery Chemistry (P.S.D., T.H.P.), Translational Oncology (S.-F.Y., A.P.), and Protein Chemistry (J.D.S.), Genentech, South San Francisco, California
| | - Wei Wang
- Drug Metabolism & Pharmacokinetics (D.Z., Y.M., D.S., W.W., S.C.K., C.E.C.A.H.), Discovery Chemistry (P.S.D., T.H.P.), Translational Oncology (S.-F.Y., A.P.), and Protein Chemistry (J.D.S.), Genentech, South San Francisco, California
| | - Andrew Polson
- Drug Metabolism & Pharmacokinetics (D.Z., Y.M., D.S., W.W., S.C.K., C.E.C.A.H.), Discovery Chemistry (P.S.D., T.H.P.), Translational Oncology (S.-F.Y., A.P.), and Protein Chemistry (J.D.S.), Genentech, South San Francisco, California
| | - S Cyrus Khojasteh
- Drug Metabolism & Pharmacokinetics (D.Z., Y.M., D.S., W.W., S.C.K., C.E.C.A.H.), Discovery Chemistry (P.S.D., T.H.P.), Translational Oncology (S.-F.Y., A.P.), and Protein Chemistry (J.D.S.), Genentech, South San Francisco, California
| | - Cornelis E C A Hop
- Drug Metabolism & Pharmacokinetics (D.Z., Y.M., D.S., W.W., S.C.K., C.E.C.A.H.), Discovery Chemistry (P.S.D., T.H.P.), Translational Oncology (S.-F.Y., A.P.), and Protein Chemistry (J.D.S.), Genentech, South San Francisco, California
| |
Collapse
|
47
|
Gour J, Gatadi S, Akunuri R, Yaddanapudi MV, Nengroo MA, Datta D, Chopra S, Nanduri S. Catalyst-free facile synthesis of polycyclic indole/pyrrole substituted-1,2,3-triazoles. Org Biomol Chem 2019; 17:8153-8165. [PMID: 31460554 DOI: 10.1039/c9ob01560d] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A general and catalyst-free access to the fused polycyclic N-heterocycles via an intramolecular azide-alkene cascade reaction under mild reaction conditions has been developed. The reaction is applicable to both indole and pyrrole substrates, and a variety of substituents are tolerated. The entire sequence can be carried out in a one-pot operation. This methodology provides a sustainable and efficient access to a variety of novel polycyclic indole/pyrrole substituted-1,2,3-triazoles.
Collapse
Affiliation(s)
- Jitendra Gour
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| | - Srikanth Gatadi
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| | - Ravikumar Akunuri
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| | | | - Mushtaq Ahmad Nengroo
- Biochemistry Division, CSIR-Central Drug Research Institute (CDRI), Lucknow 226031, India
| | - Dipak Datta
- Biochemistry Division, CSIR-Central Drug Research Institute (CDRI), Lucknow 226031, India
| | - Sidharth Chopra
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow 226031, Uttar Pradesh, India
| | - Srinivas Nanduri
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| |
Collapse
|
48
|
Gour J, Gatadi S, Pooladanda V, Ghouse SM, Malasala S, Madhavi YV, Godugu C, Nanduri S. Facile synthesis of 1,2,3-triazole-fused indolo- and pyrrolo[1,4]diazepines, DNA-binding and evaluation of their anticancer activity. Bioorg Chem 2019; 93:103306. [PMID: 31586710 DOI: 10.1016/j.bioorg.2019.103306] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 12/09/2022]
Abstract
A facile synthetic strategy has been developed for the generation of structurally diverse N-fused heterocycles. The formation of fused 1,2,3-triazole indolo and pyrrolodiazepines proceeds through an initial Knoevenagel condensation followed by intramolecular azide-alkyne cycloaddition reaction at room temperature without recourse to the traditional Cu(I)-catalyzed azide-alkyne cycloadditions. The synthesized compounds were evaluated for their in vitro anti-cancer activity against the NCI 60 cell line panel. Among the tested compounds, 3a and 3h were found to exhibit potent inhibitory activity against many of the cell lines. Cell cycle analysis indicated that the compounds inhibit the cell cycle at sub G1 phase. The DNA- nano drop method, viscosity experiment and docking studies suggested these compounds possess DNA binding affinity.
Collapse
Affiliation(s)
- Jitendra Gour
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Srikanth Gatadi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Venkatesh Pooladanda
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Shaik Mahammad Ghouse
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Satyaveni Malasala
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Y V Madhavi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Chandraiah Godugu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Srinivas Nanduri
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| |
Collapse
|
49
|
Abstract
Heterocycles are very common substructures in a number of pharmaceuticals. Over the past several years, the use of palladium-catalyzed oxidative cyclization for heterocyclic synthesis has become much more prevalent. This review collects recent reports using palladium catalysis to synthesize a wide variety of heterocyclic scaffolds. Many of these reactions use oxygen as the terminal oxidant. Some salient mechanistic features are discussed.
Collapse
Affiliation(s)
- John C. Hershberger
- Department of Chemistry and Physics, Arkansas State University, State University, AR, United States
| |
Collapse
|
50
|
Gregson SJ, Tiberghien AC, Masterson LA, Howard PW. Pyrrolobenzodiazepine Dimers as Antibody–Drug Conjugate (ADC) Payloads. CYTOTOXIC PAYLOADS FOR ANTIBODY – DRUG CONJUGATES 2019. [DOI: 10.1039/9781788012898-00296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The pyrrolobenzodiazepine (PBD) ring system was first discovered in the 1960s and is found in several naturally occurring potent anti-tumour antibiotics. The mode of action of PBDs involves sequence-selective [purine–guanine–purine (PuGPu)] alkylation in the minor groove of DNA through covalent binding from guanine N2 to the PBD C11-position. Dimerization of the PBD ring system gives molecules that can cross-link DNA, which leads to a substantial increase in potency and DNA binding affinity and an extension of sequence-selectivity compared with monomers. PBD dimers feature as the cytotoxic component of numerous ADCs being evaluated in clinical trials. PBD-ADC clinical candidates loncastuximab tesirine, camidanlumab tesirine and rovalpituzumab tesirine employ a PBD N10 linkage while vadastuximab talirine uses a C2-linkage. The PBD dimer scaffold is versatile and offers many opportunities to diversify the ADC platform, with extensive research being performed worldwide to develop the next generation of PBD payload–linker molecules. The search for new PBD payload–linker molecules has mainly focused on changes in payload structure (e.g. PBD C2 modification and macrocyclisation), alternative conjugation strategies (e.g. haloacetamides, ‘click’ approaches and pyridyl disulphides), non-peptide triggers in the linker (e.g. disulphides) and non-cleavable derivatives (i.e. payload release through antibody degradation).
Collapse
|