Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(8)2024 Apr 14.
Article in English | MEDLINE | ID: mdl-38673926

ABSTRACT

Acute myeloid leukemia (AML) is a hematological malignancy that is characterized by an expansion of immature myeloid precursors. Despite therapeutic advances, the prognosis of AML patients remains poor and there is a need for the evaluation of promising therapeutic candidates to treat the disease. The objective of this study was to evaluate the efficacy of duocarmycin Stable A (DSA) in AML cells in vitro. We hypothesized that DSA would induce DNA damage in the form of DNA double-strand breaks (DSBs) and exert cytotoxic effects on AML cells within the picomolar range. Human AML cell lines Molm-14 and HL-60 were used to perform 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT), DNA DSBs, cell cycle, 5-ethynyl-2-deoxyuridine (EdU), colony formation unit (CFU), Annexin V, RNA sequencing and other assays described in this study. Our results showed that DSA induced DNA DSBs, induced cell cycle arrest at the G2M phase, reduced proliferation and increased apoptosis in AML cells. Additionally, RNA sequencing results showed that DSA regulates genes that are associated with cellular processes such as DNA repair, G2M checkpoint and apoptosis. These results suggest that DSA is efficacious in AML cells and is therefore a promising potential therapeutic candidate that can be further evaluated for the treatment of AML.


Subject(s)
Apoptosis , Cell Proliferation , Duocarmycins , Leukemia, Myeloid, Acute , Humans , Apoptosis/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/metabolism , Cell Proliferation/drug effects , Duocarmycins/pharmacology , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , HL-60 Cells , Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , DNA Damage/drug effects
2.
Molecules ; 28(12)2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37375372

ABSTRACT

The construction of duocarmycin-like compounds is often associated with lengthy synthetic routes. Presented herein is the development of a short and convenient synthesis of a type of duocarmycin prodrug. The 1,2,3,6-tetrahydropyrrolo[3,2-e]indole-containing core is here constructed from commercially available Boc-5-bromoindole in four steps and 23% overall yield, utilizing a Buchwald-Hartwig amination followed by a sodium hydride-induced regioselective bromination. In addition, protocols for selective mono- and di-halogenations of positions 3 and 4 were also developed, which could be useful for further exploration of this scaffold.


Subject(s)
Prodrugs , Duocarmycins , Amination
3.
Trends Pharmacol Sci ; 43(4): 266-268, 2022 04.
Article in English | MEDLINE | ID: mdl-35058076

ABSTRACT

A recent antimalarial screen by Alder and colleagues has uncovered a natural product, PDE-I2, with DNA-binding and schizonticidal activity against Plasmodium falciparum. Parasite specificity is likely conferred by the extremely high A+T content of the P. falciparum genome. We discuss here this character as a potential target for future drugs.


Subject(s)
Antimalarials , Malaria, Falciparum , Parasites , Animals , Antimalarials/pharmacology , DNA/therapeutic use , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Parasites/genetics , Plasmodium falciparum/genetics
4.
Curr Cancer Drug Targets ; 22(4): 312-327, 2022.
Article in English | MEDLINE | ID: mdl-34951367

ABSTRACT

BACKGROUND: Aberrant expression of the MET receptor tyrosine kinase is an oncogenic determinant and a drug target for cancer therapy. Currently, antibody-based biotherapeutics targeting MET are under clinical trials. OBJECTIVE: Here, we report the preclinical and therapeutic evaluation of a novel anti-MET antibody- drug conjugate PCMC1D3-duocarmycin SA (PCMC1D3-DCM) for targeted cancer therapy. METHODS: The monoclonal antibody PCMC1D3 (IgG1a/κ), generated by a hybridoma technique and specific to one of the MET extracellular domains, was selected based on its high specificity to human MET with a binding affinity of 1.60 nM. PCMC1D3 was conjugated to DCM via a cleavable valine-citrulline dipeptide linker to form an antibody-drug conjugate with a drug-to-antibody ratio of 3.6:1. PCMC1D3-DCM in vitro rapidly induced MET internalization with an internalization efficacy ranging from 6.5 to 17.2h dependent on individual cell lines. RESULTS: Studies using different types of cancer cell lines showed that PCMC1D3-DCM disrupted the cell cycle, reduced cell viability, and caused massive cell death within 96h after treatment initiation. The calculated IC50 values for cell viability reduction were 1.5 to 15.3 nM. Results from mouse xenograft tumor models demonstrated that PCMC1D3-DCM in a single dose injection at 10 mg/kg body weight effectively delayed xenograft tumor growth up to two weeks without signs of tumor regrowth. The calculated tumoristatic concentration, a minimal dose required to balance tumor growth and inhibition, was around 2 mg/kg body weight. Taken together, PCMC1D3-DCM was effective in targeting the inhibition of tumor growth in xenograft models. CONCLUSION: This work provides the basis for the development of humanized PCMC1D3-DCM for MET-targeted cancer therapy in the future.


Subject(s)
Immunoconjugates , Neoplasms , Animals , Body Weight , Cell Line, Tumor , Duocarmycins , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Mice , Neoplasms/drug therapy , Proto-Oncogene Proteins c-met , Xenograft Model Antitumor Assays
5.
Molecules ; 26(12)2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34207832

ABSTRACT

Triple-negative breast cancer (TNBC) is a heterogeneous subtype of tumors that tests negative for estrogen receptors, progesterone receptors, and excess HER2 protein. The mainstay of treatment remains chemotherapy, but the therapeutic outcome remains inadequate. This paper investigates the potential of a duocarmycin derivative, tafuramycin A (TFA), as a new and more effective chemotherapy agent in TNBC treatment. To this extent, we optimized the chemical synthesis of TFA, and we encapsulated TFA in a micellar system to reduce side effects and increase tumor accumulation. In vitro and in vivo studies suggest that both TFA and SMA-TFA possess high anticancer effects in TNBC models. Finally, the encapsulation of TFA offered a preferential avenue to tumor accumulation by increasing its concentration at the tumor tissues by around four times in comparison with the free drug. Overall, the results provide a new potential strategy useful for TNBC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Indole Alkaloids/pharmacology , Nanoparticles/chemistry , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Female , Humans , Indole Alkaloids/chemistry , Maleates/chemistry , Maleates/pharmacology , Mice , Mice, Inbred BALB C , Micelles , Polystyrenes/chemistry , Polystyrenes/pharmacology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
6.
Cancer Chemother Pharmacol ; 88(4): 673-687, 2021 10.
Article in English | MEDLINE | ID: mdl-34245333

ABSTRACT

PURPOSE: Hypoxia-activated prodrugs (HAPs) have the potential for eliminating chemo- and radiation-resistant hypoxic tumour cells, but their activity is often compromised by limited penetration into hypoxic zones. Nitrochloromethylbenzindoline (nitroCBI) HAPs are reduced in hypoxic cells to highly cytotoxic DNA minor groove alkylating aminoCBI metabolites. In this study, we investigate whether a lead nitroCBI, SN30548, generates a significant bystander effect through the diffusion of its aminoCBI metabolite and whether this compensates for any diffusion limitations of the prodrug in tumour tissue. METHODS: Metabolism and uptake of the nitroCBI in oxic and anoxic cells, and diffusion through multicellular layer cultures, was characterised by LC-MS/MS. To quantify bystander effects, clonogenic cell killing of HCT116 cells was assessed in multicellular spheroid co-cultures comprising cells transfected with cytochrome P450 oxidoreductase (POR) or E. coli nitroreductase NfsA. Spatially-resolved pharmacokinetic/pharmacodynamic (PK/PD) models, parameterised by the above measurements, were developed for spheroids and tumours using agent-based and Green's function modelling, respectively. RESULTS: NitroCBI was reduced to aminoCBI by POR under anoxia and by NfsA under oxia, and was the only significant cytotoxic metabolite in both cases. In spheroid co-cultures comprising 30% NfsA-expressing cells, non-metabolising cells were as sensitive as the NfsA cells, demonstrating a marked bystander effect. Agent-based PK/PD models provided good prediction of cytotoxicity in spheroids, while use of the same parameters in a Green's function model for a tumour microregion demonstrated that local diffusion of aminoCBI overcomes the penetration limitation of the prodrug. CONCLUSIONS: The nitroCBI HAP SN30548 generates a highly efficient bystander effect through local diffusion of its active metabolite in tumour tissue.


Subject(s)
Bystander Effect/drug effects , Cell Hypoxia , Indoles/pharmacology , Models, Biological , Chromatography, Liquid , Coculture Techniques , Escherichia coli Proteins/genetics , HCT116 Cells , Humans , Indoles/pharmacokinetics , NADPH-Ferrihemoprotein Reductase/genetics , Nitroreductases/genetics , Prodrugs , Spheroids, Cellular/cytology , Tandem Mass Spectrometry
7.
Bioorg Med Chem ; 40: 116167, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33932713

ABSTRACT

The duocarmycins belong to a class of agent which has great potential for use in cancer therapy. Their exquisite potency means they are too toxic for systemic use, and targeted approaches are required to unlock their clinical potential. In this study, we have explored seco-OH-chloromethylindoline (CI) duocarmycin-based bioprecursors for their potential for cytochrome P450 (CYP)-mediated cancer cell kill. We report on synthetic and biological explorations of racemic seco-CI-MI, where MI is a 5-methoxy indole motif, and dehydroxylated analogues. We show up to a 10-fold bioactivation of de-OH CI-MI and a fluoro bioprecursor analogue in CYP1A1-transfected cells. Using CYP bactosomes, we also demonstrate that CYP1A2 but not CYP1B1 or CYP3A4 has propensity for potentiating these compounds, indicating preference for CYP1A bioactivation.


Subject(s)
Antineoplastic Agents/pharmacology , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Duocarmycins/pharmacology , Indoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cytochrome P-450 Enzyme Inhibitors/chemical synthesis , Cytochrome P-450 Enzyme Inhibitors/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Duocarmycins/chemical synthesis , Duocarmycins/chemistry , Humans , Indoles/chemical synthesis , Indoles/chemistry , Molecular Structure , Structure-Activity Relationship
8.
Biomedicines ; 9(3)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33809117

ABSTRACT

Despite significant advances in treatment strategies over the past decade, selective treatment of breast cancer with limited side-effects still remains a great challenge. The cytochrome P450 (CYP) family of enzymes contribute to cancer cell proliferation, cell signaling and drug metabolism with implications for treatment outcomes. A clearer understanding of CYP expression is important in the pathogenesis of breast cancer as several isoforms play critical roles in metabolising steroid hormones and xenobiotics that contribute to the genesis of breast cancer. The purpose of this review is to provide an update on how the presence of CYPs impacts on standard of care (SoC) drugs used to treat breast cancer as well as discuss opportunities to exploit CYP expression for therapeutic intervention. Finally, we provide our thoughts on future work in CYP research with the aim of supporting ongoing efforts to develop drugs with improved therapeutic index for patient benefit.

9.
Pharmaceutics ; 13(4)2021 Apr 02.
Article in English | MEDLINE | ID: mdl-33918158

ABSTRACT

HER-3 is becoming an attractive target for antibody-drug conjugate (ADC)-based therapy. Indeed, this receptor and its ligands are found to be overexpressed in several malignancies, and re-activation of its downstream signaling axis is known to play a critical role in modulating the sensitivity of targeted therapeutics in different tumors. In this study, we generated a novel ADC named EV20/NMS-P945 by coupling the anti-HER-3 antibody EV20 with a duocarmycin-like derivative, the thienoindole (TEI) NMS-P528, a DNA minor groove alkylating agent through a peptidic cleavable linker. This ADC showed target-dependent cytotoxic activity in vitro on several tumor cell lines and therapeutic activity in mouse xenograft tumor models, including those originating from pancreatic, prostatic, head and neck, gastric and ovarian cancer cells and melanoma. Pharmacokinetics and toxicological studies in monkeys demonstrated that this ADC possesses a favorable terminal half-life and stability and it is well tolerated. These data support further EV20/NMS-P945 clinical development as a therapeutic agent against HER-3-expressing malignancies.

10.
Aging Cell ; 19(4): e13133, 2020 04.
Article in English | MEDLINE | ID: mdl-32175667

ABSTRACT

Senescence is a stable growth arrest that impairs the replication of damaged, old or preneoplastic cells, therefore contributing to tissue homeostasis. Senescent cells accumulate during ageing and are associated with cancer, fibrosis and many age-related pathologies. Recent evidence suggests that the selective elimination of senescent cells can be effective on the treatment of many of these senescence-associated diseases. A universal characteristic of senescent cells is that they display elevated activity of the lysosomal ß-galactosidase, and this has been exploited as a marker for senescence (senescence-associated ß-galactosidase activity). Consequently, we hypothesized that galactose-modified cytotoxic prodrugs will be preferentially processed by senescent cells, resulting in their selective killing. Here, we show that different galactose-modified duocarmycin (GMD) derivatives preferentially kill senescent cells. GMD prodrugs induce selective apoptosis of senescent cells in a lysosomal ß-galactosidase (GLB1)-dependent manner. GMD prodrugs can eliminate a broad range of senescent cells in culture, and treatment with a GMD prodrug enhances the elimination of bystander senescent cells that accumulate upon whole-body irradiation treatment of mice. Moreover, taking advantage of a mouse model of adamantinomatous craniopharyngioma (ACP), we show that treatment with a GMD prodrug selectively reduced the number of ß-catenin-positive preneoplastic senescent cells. In summary, the above results make a case for testing the potential of galactose-modified duocarmycin prodrugs to treat senescence-related pathologies.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cellular Senescence/drug effects , Craniopharyngioma/drug therapy , Duocarmycins/pharmacology , Galactose/pharmacology , Prodrugs/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Coculture Techniques , Craniopharyngioma/metabolism , Craniopharyngioma/pathology , Humans , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , beta-Galactosidase/metabolism
11.
Methods ; 154: 125-135, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30292795

ABSTRACT

Antibody selection for antibody-drug conjugates (ADCs) has traditionally depended on its internalization into the target cell, although ADC efficacy also relies on recycling of the receptor-ADC complex, endo-lysosomal trafficking, and subsequent linker/antibody proteolysis. In this study, we observed that a bispecific anti-murine platelet-derived growth factor receptor beta (mPDGFRß) x cotinine single-chain variable fragment (scFv)-kappa constant region (Cκ)-scFv fusion protein and cotinine-duocarmycin can form an ADC-like complex to induce cytotoxicity against mPDGFRß expressing cells. Multiple anti-mPDGFRß antibody candidates can be produced in this bispecific scFv-Cκ-scFv fusion protein format and tested for their ability to deliver cotinine-conjugated cytotoxic drugs, thus providing an improved approach for antibody selection in ADC development.


Subject(s)
Antibodies, Bispecific/therapeutic use , Immunoconjugates/therapeutic use , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Animals , Antibodies, Bispecific/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cotinine , Humans , Immunoconjugates/pharmacology , Mice , Receptor, Platelet-Derived Growth Factor beta/immunology
12.
Toxicol Pathol ; 46(7): 746-752, 2018 10.
Article in English | MEDLINE | ID: mdl-30295169

ABSTRACT

Antibody-drug conjugates (ADCs) are an emerging class of anticancer therapeutics, delivering highly cytotoxic molecules directly to cancer cells. ADCs are composed of an antibody, a small molecule drug, and a linker attaching one to another. Antibodies are directed to a large variety of antigens overexpressed on tumor cells, tumor vasculature, or tumor-supporting stroma. After internalization, the ADC is transferred to lysosomes where the cytotoxic component is released, finally killing the target cell. All ADCs are administered via intravenous injection. Once in the circulation, linker stability in plasma is of high importance. In vivo studies in animals address the release of payload over time and typically measure total antibody, conjugated ADC, and free drug. ADC development is driven by ICH (International Council for Harmonisation) guidelines S6(R1) and S9. Dose-limiting toxicities of current ADCs are mainly associated with the payload and correlate well between clinical trials and nonclinical studies in rodents and nonrodents. This mini review is intended to provide general information about ADCs in oncology and shall assist the toxicologic pathologist in correctly interpreting morphological findings acquired in toxicity studies with this entity.


Subject(s)
Antineoplastic Agents/pharmacology , Drug-Related Side Effects and Adverse Reactions/etiology , Drug-Related Side Effects and Adverse Reactions/pathology , Immunoconjugates/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Female , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/pharmacokinetics , Immunoconjugates/toxicity , Injections, Intravenous , Liver/drug effects , Liver/pathology , Male , Species Specificity , Tissue Distribution , Toxicity Tests
13.
J Nucl Med ; 59(3): 494-501, 2018 03.
Article in English | MEDLINE | ID: mdl-29146698

ABSTRACT

Prostate cancer (PCa) is the most common cancer in men worldwide. In general, PCa responds poorly to chemotherapy. Therefore, antibody-drug conjugates (ADCs) have been developed to specifically deliver highly cytotoxic drugs to the tumor. Because the prostate-specific membrane antigen (PSMA) is overexpressed in PCa, it represents a promising target for ADC-based therapies. The aim of this study was to evaluate the therapeutic efficacy of site-specifically conjugated duocarmycin- and monomethyl auristatin E (MMAE)-based anti-PSMA ADCs with drug-to-antibody ratios (DARs) of 2 and 4. Methods: The glycan group of the anti-PSMA antibody D2B was chemoenzymatically conjugated with duocarmycin or MMAE. Preservation of the immunoreactivity of the antibody on site-specific conjugation was investigated in vitro. Biodistribution and small-animal SPECT/CT imaging (18.5 ± 2.6 MBq) with 25 µg of 111In-labeled ADCs were performed on BALB/c nude mice with subcutaneous PSMA-positive LS174T-PSMA xenografts. Finally, the therapeutic efficacy of the 4 different ADCs was assessed in mice with LS174T-PSMA tumors. Results: The immunoreactivity of the anti-PSMA antibody was preserved on site-specific conjugation. Biodistribution revealed high tumor uptake of all agents. The highest tumor uptake was observed in mice administered with 111In-D2B-DAR2-MMAE, reaching 119.7 ± 37.4 percentage injected dose per gram at 3 d after injection. Tumors of mice injected with 111In-D2B, 111In-D2B-DAR2-duocarmycin, 111In-D2B-DAR4-duocarmycin, 111In-D2B-DAR2-MMAE, and 111In-D2B-DAR4-MMAE could clearly be visualized with small-animal SPECT/CT. In contrast to unconjugated D2B or vehicle, treatment with either of the MMAE-based ADCs, but not with a duocarmycin-based ADC, significantly impaired tumor growth and prolonged median survival from 13 d (phosphate-buffered saline) to 20 and 29 d for DAR2 and DAR4 ADC, respectively. Tumor-doubling time increased from 3.5 ± 0.5 d to 5.2 ± 1.8 and 9.2 ± 2.1 d after treatment with D2B-DAR2-MMAE and D2B-DAR4-MMAE, respectively. Conclusion: The site-specifically conjugated anti-PSMA ADCs D2B-DAR2-MMAE and D2B-DAR4-MMAE efficiently targeted PSMA-expressing xenografts, effectively inhibited tumor growth of PSMA-expressing tumors, and significantly prolonged survival of mice.


Subject(s)
Gene Expression Regulation, Neoplastic , Glutamate Carboxypeptidase II/metabolism , Immunoconjugates/chemistry , Immunoconjugates/therapeutic use , Indoles/chemistry , Oligopeptides/chemistry , Prostatic Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Duocarmycins , Immunoconjugates/pharmacokinetics , Male , Mice , Mice, Nude , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Pyrrolidinones/chemistry , Single Photon Emission Computed Tomography Computed Tomography , Tissue Distribution
14.
Oncotarget ; 8(36): 60368-60377, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28947977

ABSTRACT

The endosialin/CD248/TEM1 receptor is expressed on the cell surface of tumor-associated stroma cells as well as in sarcoma and neuroblastoma cells. This receptor is emerging as an attractive molecule in diagnostics and therapeutics because of its expression across the stroma of many human tumors, the low to absent expression in normal tissues and accessibility from the vascular circulation. In this study, we present evidence of the preclinical efficacy of a novel Antibody-Drug Conjugate (ENDOS/ADC). It consists of a humanized endosialin monoclonal antibody, named hMP-E-8.3, conjugated to a potent duocarmycin derivative. In endosialin expressing cancer cell lines, this ENDOS/ADC showed a powerful, specific and target-dependent killing activity. High expression levels of endosialin in cells correlated with efficient internalization and cytotoxic effects in vitro. Efficacy studies demonstrated that ENDOS/ADC treatment led to a long-lasting tumor growth inhibition of a cell line-based model of human osteosarcoma. Taken together, our results demonstrate that endosialin is an attractive target in sarcoma and suggest that ENDOS/ADC has the potential to be developed into a bio-therapeutic agent for these malignancies.

15.
MAbs ; 9(4): 603-614, 2017.
Article in English | MEDLINE | ID: mdl-28273004

ABSTRACT

The currently marketed antibody-drug conjugates (ADC) destabilize microtubule assembly in cancer cells and initiate apoptosis in patients. However, few tumor antigens (TA) are expressed at high densities on cancer lesions, potentially minimizing the therapeutic index of current ADC regimens. The peptide/human leukocyte antigen (HLA) complex can be specifically targeted by therapeutic antibodies (designated T cell receptor [TCR]-like antibodies) and adequately distinguish malignant cells, but has not been the focus of ADC development. We analyzed the killing potential of TCR-like ADCs when cross-linked to the DNA alkylating compound duocarmycin. Our data comprise proof-of-principle results that TCR-like ADCs mediate potent tumor cytotoxicity, particularly under common scenarios of low TA/HLA density, and support their continued development alongside agents that disrupt DNA replication. Additionally, TCR-like antibody ligand binding appears to play an important role in ADC functionality and should be addressed during therapy development to avoid binding patterns that negate ADC killing efficacy.


Subject(s)
Antibodies, Neoplasm/pharmacology , Drug Delivery Systems/methods , HLA Antigens/immunology , Indoles/pharmacology , Neoplasms/drug therapy , Peptides/immunology , Receptors, Antigen, T-Cell , Animals , Cell Line, Tumor , Duocarmycins , Humans , Mice , Neoplasms/immunology , Neoplasms/pathology , Pyrrolidinones/pharmacology
16.
Bioorg Med Chem Lett ; 27(1): 102-108, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27889454

ABSTRACT

The pyrrolobenzodiazepine (PBD) and duocarmycin families are DNA-interactive agents that covalently bond to guanine (G) and adenine (A) bases, respectively, and that have been joined together to create synthetic dimers capable of cross-linking G-G, A-A, and G-A bases. Three G-A alkylating dimers have been reported in publications to date, with defined DNA-binding sites proposed for two of them. In this study we have used molecular dynamics simulations to elucidate preferred DNA-binding sites for the three published molecular types. For the PBD-CPI dimer UTA-6026 (1), our simulations correctly predicted its favoured binding site (i.e., 5'-C(G)AATTA-3') as identified by DNA cleavage studies. However, for the PBD-CI molecule ('Compound 11', 3), we were unable to reconcile the results of our simulations with the reported preferred cross-linking sequence (5'-ATTTTCC(G)-3'). We found that the molecule is too short to span the five base pairs between the A and G bases as claimed, but should target instead a sequence such as 5'-ATTTC(G)-3' with two less base pairs between the reacting G and A residues. Our simulation results for this hybrid dimer are also in accord with the very low interstrand cross-linking and in vitro cytotoxicity activities reported for it. Although a preferred cross-linking sequence was not reported for the third hybrid dimer ('27eS', 2), our simulations predict that it should span two base pairs between covalently reacting G and A bases (e.g., 5'-GTAT(A)-3').


Subject(s)
Benzodiazepines/pharmacology , Cross-Linking Reagents/pharmacology , DNA/chemistry , Indoles/pharmacology , Pyrroles/pharmacology , Base Sequence , Benzodiazepines/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cross-Linking Reagents/chemistry , DNA/drug effects , Dimerization , Dose-Response Relationship, Drug , Duocarmycins , Humans , Indoles/chemistry , Ligands , Molecular Dynamics Simulation , Molecular Structure , Pyrroles/chemistry , Pyrrolidinones/chemistry , Pyrrolidinones/pharmacology , Structure-Activity Relationship
17.
Bioorg Med Chem ; 24(22): 6075-6081, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27745990

ABSTRACT

A Pd-catalysed amination method is used to convert seco-CBI, a synthetic analogue of the alkylating subunit of the duocarmycin natural products, from the phenol to amino form. This allows efficient enantioselective access to the more potent S enantiomer of aminoCBI and its incorporation into analogues of DNA minor groove cross-linking agents. Evaluation in a panel of nine human tumour cell lines shows that the bifunctional agents containing aminoCBI are generally less cytotoxic than their phenolCBI analogues and more susceptible to P-glycoprotein-mediated resistance. However, all bifunctional agents are potent cytotoxins, some in the sub-pM IC50 range, with in vitro properties that compare favourably with established microtubule-targeted ADC payloads.


Subject(s)
Antibodies/pharmacology , Antineoplastic Agents/pharmacology , Cross-Linking Reagents/pharmacology , Indoles/pharmacology , Antibodies/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cross-Linking Reagents/chemical synthesis , Cross-Linking Reagents/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Duocarmycins , Humans , Indoles/chemistry , Molecular Structure , Pyrrolidinones/chemistry , Pyrrolidinones/pharmacology , Structure-Activity Relationship
18.
Bioorg Med Chem ; 24(20): 4779-4786, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27221071

ABSTRACT

The design, synthesis, and evaluation of methyl 1,2,8,8a-tetrahydrocyclopropa[c]imidazolo[4,5-e]indol-4-one-6-carboxylate (CImI) derivatives are detailed representing analogs of duocarmycin SA and yatakemycin containing an imidazole replacement for the fused pyrrole found in the DNA alkylation subunit.


Subject(s)
Imidazoles/pharmacology , Indoles/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Duocarmycins , Imidazoles/chemistry , Indoles/chemical synthesis , Indoles/chemistry , Mice , Molecular Structure , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrroles/pharmacology , Stereoisomerism
19.
Chemistry ; 22(26): 8756-8, 2016 Jun 20.
Article in English | MEDLINE | ID: mdl-27123891

ABSTRACT

To evaluate the reactivity of antitumor agents in a nucleosome architecture, we conducted in vitro studies to assess the alkylation level of duocarmycin B2 on nucleosomes with core and linker DNA using sequencing gel electrophoresis. Our results suggested that the alkylating efficiencies of duocarmycin B2 were significantly decreased in core DNA and increased at the histone-free linker DNA sites when compared with naked DNA conditions. Our finding that nucleosome assembly alters the accessibility of duocarmycin B2 to duplex DNA could advance its design as an antitumor agent.


Subject(s)
Antineoplastic Agents/chemistry , DNA/chemistry , Indoles/chemistry , Alkylation , Antineoplastic Agents/metabolism , Base Sequence , DNA/metabolism , Duocarmycins , Indoles/metabolism , Nucleosomes/metabolism , Pyrrolidinones/chemistry , Pyrrolidinones/metabolism
20.
Angew Chem Int Ed Engl ; 54(46): 13550-4, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26373694

ABSTRACT

Analogues of the natural product duocarmycin bearing an indole moiety were shown to bind aldehyde dehydrogenase 1A1 (ALDH1A1) in addition to DNA, while derivatives without the indole solely addressed the ALDH1A1 protein. The molecular mechanism of selective ALDH1A1 inhibition by duocarmycin analogues was unraveled through cocrystallization, mutational studies, and molecular dynamics simulations. The structure of the complex shows the compound embedded in a hydrophobic pocket, where it is stabilized by several crucial π-stacking and van der Waals interactions. This binding mode positions the cyclopropyl electrophile for nucleophilic attack by the noncatalytic residue Cys302, thereby resulting in covalent attachment, steric occlusion of the active site, and inhibition of catalysis. The selectivity of duocarmycin analogues for ALDH1A1 is unique, since only minor alterations in the sequence of closely related protein isoforms restrict compound accessibility.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Antibiotics, Antineoplastic/pharmacology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Antibiotics, Antineoplastic/chemistry , Crystallography, X-Ray , Duocarmycins , Enzyme Inhibitors/chemistry , Humans , Indoles/chemistry , Molecular Dynamics Simulation , Molecular Structure , Pyrrolidinones/chemistry , Pyrrolidinones/pharmacology , Retinal Dehydrogenase , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL