87 results on '"Kowol CR"'
Search Results
2. Metal drugs and the anticancer immune response
- Author
-
Walter Berger, Bernhard Englinger, Petra Heffeter, Alessio Terenzi, Bernhard K. Keppler, Christian R. Kowol, Christine Pirker, Englinger, B, Pirker, C, Heffeter, P, Terenzi, A, Kowol, CR, Keppler, BK, and Berger, W
- Subjects
Metal Drugs, Immune Response, Anticancer, cisplatin ,animal diseases ,medicine.medical_treatment ,Evasion (network security) ,chemical and pharmacologic phenomena ,Antineoplastic Agents ,010402 general chemistry ,01 natural sciences ,Malignant transformation ,Immune system ,Immunity ,Coordination Complexes ,Neoplasms ,medicine ,Humans ,Lymphocytes ,Tumor microenvironment ,010405 organic chemistry ,Chemistry ,General Chemistry ,Immunotherapy ,biochemical phenomena, metabolism, and nutrition ,Acquired immune system ,Immunity, Innate ,0104 chemical sciences ,Gastrointestinal Microbiome ,Metals ,Settore CHIM/03 - Chimica Generale E Inorganica ,Cancer cell ,bacteria ,Nanoparticles ,Neuroscience - Abstract
The immune system deploys a multitude of innate and adaptive mechanisms not only to ward off pathogens but also to prevent malignant transformation ("immune surveillance"). Hence, a clinically apparent tumor already reflects selection for those malignant cell clones capable of evading immune recognition ("immune evasion"). Metal drugs, besides their well-investigated cytotoxic anticancer effects, massively interact with the cancer-immune interface and can reverse important aspects of immune evasion. This topic has recently gained intense attention based on combination approaches with anticancer immunotherapy (e.g., immune checkpoint inhibitors), a strategy recently delivering first exciting results in clinical settings. This review summarizes the promising but still extremely fragmentary knowledge on the interplay of metal drugs with the fidelity of anticancer immune responses but also their role in adverse effects. It highlights that, at least in some cases, metal drugs can induce long-lasting anticancer immune responses. Important steps in this process comprise altered visibility and susceptibility of cancer cells toward innate and adaptive immunity, as well as direct impacts on immune cell populations and the tumor microenvironment. On the basis of the gathered information, we suggest initiating joint multidisciplinary programs to implement comprehensive immune analyses into strategies to develop novel and smart anticancer metal compounds.
- Published
- 2019
3. The tyrosine kinase inhibitor Nintedanib induces lysosomal dysfunctionality: Role of protonation-dependent crystallization processes.
- Author
-
Mosca E, Federa A, Pirker C, Schosserer M, Liendl L, Eckhard M, Sombke A, Dömötör O, Kirchhofer D, Timelthaler G, Baier D, Gurschka P, Gabler L, Reithofer M, Chin JM, Elsayad K, Englinger B, Tahir A, Kowol CR, and Berger W
- Subjects
- Animals, Mice, Hydrogen-Ion Concentration, Humans, Protons, Tyrosine Kinase Inhibitors, Lysosomes metabolism, Lysosomes drug effects, Indoles chemistry, Indoles pharmacology, Crystallization, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors chemistry
- Abstract
Nintedanib (NIN), a multi-tyrosine kinase inhibitor clinically approved for idiopathic pulmonary fibrosis and lung cancer, is characterized by protonation-dependent lysosomotropic behavior and appearance of lysosome-specific fluorescence emission properties. Here we investigate whether spontaneous formation of a so far unknown NIN matter within the acidic cell compartment is underlying these unexpected emissive properties and investigate the consequences on lysosome functionality. Lysosomes of cells treated with NIN, but not non-protonatable NIN derivatives, exhibited lysosome-associated birefringence signals co-localizing with the NIN-derived fluorescence emission. Sensitivity of both parameters towards vATPase inhibitors confirmed pH-dependent, spontaneous adoption of novel crystalline NIN structures in lysosomes. Accordingly, NIN crystallization from buffer solutions resulted in formation of multiple crystal polymorphs with pH-dependent fluorescence properties. Cell-free crystals grown at lysosomal-like pH conditions resembled NIN-treated cell lysosomes concerning fluorescence pattern, photobleaching dynamics, and Raman spectra. However, differences in birefringence intensity and FAIM-determined anisotropy, as well as predominant association with (intra)lysosomal membrane structures, suggested formation of a semi-solid NIN crystalline matter in acidic lysosomes. Despite comparable target kinase inhibition, NIN, but not its non-protonatable derivatives, impaired lysosomal functionality, mediated massive cell vacuolization, enhanced autophagy, deregulated lipid metabolism, and induced atypical phospholipidosis. Moreover, NIN exerted distinct phototoxicity, strictly dependent on lysosomal microcrystallization events. The spontaneous formation of NIN crystalline structures was also observable in the gut mucosa of orally NIN-treated mice. Summarizing, the here-described kinase inhibition-independent impact of NIN on lysosomal functionality mediates several of its cell biological activities and might contribute to NIN adverse effects., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: All authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. Paraptotic Cell Death as an Unprecedented Mode of Action Observed for New Bipyridine-Silver(I) Compounds Bearing Phosphane Coligands.
- Author
-
Teixeira RG, Stefanelli A, Pilon A, Warmers R, Fontrodona X, Romero I, Costa PJ, Villa de Brito MJ, Hudec X, Pirker C, Türck S, Antunes AMM, Kowol CR, Ott I, Brozovic A, Sombke A, Eckhard M, Tomaz AI, Heffeter P, and Valente A
- Subjects
- Humans, Cell Line, Tumor, Coordination Complexes pharmacology, Coordination Complexes chemistry, Coordination Complexes chemical synthesis, Apoptosis drug effects, Crystallography, X-Ray, Ligands, Cell Death drug effects, Drug Screening Assays, Antitumor, Structure-Activity Relationship, Drug Resistance, Neoplasm drug effects, Phosphines chemistry, Phosphines pharmacology, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry, Antineoplastic Agents chemical synthesis, Silver chemistry, Silver pharmacology, 2,2'-Dipyridyl chemistry, 2,2'-Dipyridyl pharmacology
- Abstract
In this work, we investigated the anticancer activity of several novel silver(I) 2,2'-bipyridine complexes containing either triphenylphosphane (PPh
3 ) or 1,2-bis(diphenylphosphino)ethane (dppe) ligands. All compounds were characterized by diverse analytical methods including ESI-MS spectrometry; NMR, UV-vis, and FTIR spectroscopies; and elemental analysis. Moreover, several compounds were also studied by X-ray single-crystal diffraction. Subsequently, the compounds were investigated for their anticancer activity against drug-resistant and -sensitive cancer cells. Noteworthily, neither carboplatin and oxaliplatin resistance nor p53 deletion impacted on their anticancer efficacy. MES - OV cells displayed exceptional hypersensitivity to the dppe-containing drugs. This effect was not based on thioredoxin reductase inhibition, enhanced drug uptake, or apoptosis induction. In contrast, dppe silver drugs induced paraptosis, a novel recently described form of programmed cell death. Together with the good tumor specificity of this compound's class, this work suggests that dppe-containing silver complexes could be interesting drug candidates for the treatment of resistant ovarian cancer.- Published
- 2024
- Full Text
- View/download PDF
5. Schiff bases and their metal complexes to target and overcome (multidrug) resistance in cancer.
- Author
-
Podolski-Renić A, Čipak Gašparović A, Valente A, López Ó, Bormio Nunes JH, Kowol CR, Heffeter P, and Filipović NR
- Subjects
- Schiff Bases pharmacology, Schiff Bases chemistry, Drug Resistance, Multiple, Coordination Complexes pharmacology, Coordination Complexes chemistry, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry, Neoplasms drug therapy
- Abstract
Overcoming multidrug resistance (MDR) is one of the major challenges in cancer therapy. In this respect, Schiff base-related compounds (bearing a R
1 R2 CNR3 bond) gained high interest during the past decades. Schiff bases are considered privileged ligands for various reasons, including the easiness of their preparation and the possibility to form complexes with almost all transition metal ions. Schiff bases and their metal complexes exhibit many types of biological activities and are used for the treatment and diagnosis of various diseases. Until now, 13 Schiff bases have been investigated in clinical trials for cancer treatment and hypoxia imaging. This review represents the first collection of Schiff bases and their complexes which demonstrated MDR-reversal activity. The areas of drug resistance covered in this article involve: 1) Modulation of ABC transporter function, 2) Targeting lysosomal ABCB1 overexpression, 3) Circumvention of ABC transporter-mediated drug efflux by alternative routes of drug uptake, 4) Selective activity against MDR cancer models (collateral sensitivity), 5) Targeting GSH-detoxifying systems, 6) Overcoming apoptosis resistance by inducing necrosis and paraptosis, 7) Reactivation of mutated p53, 8) Restoration of sensitivity to DNA-damaging anticancer therapy, and 9) Overcoming drug resistance through modulation of the immune system. Through this approach, we would like to draw attention to Schiff bases and their metal complexes representing highly interesting anticancer drug candidates with the ability to overcome MDR., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier Masson SAS.)- Published
- 2024
- Full Text
- View/download PDF
6. A Comparative Study on the Complexation of the Anticancer Iron Chelator VLX600 with Essential Metal Ions.
- Author
-
Pósa V, Federa A, Cseh K, Wenisch D, Spengler G, May NV, Lihi N, Samu GF, Jakupec MA, Keppler BK, Kowol CR, and Enyedy ÉA
- Subjects
- Humans, Copper pharmacology, Copper chemistry, Metals chemistry, Iron chemistry, Ions, Iron Chelating Agents pharmacology, Ferrous Compounds, Ferric Compounds, Coordination Complexes pharmacology, Coordination Complexes chemistry, Hydrazones, Triazoles
- Abstract
As cancer cells exhibit an increased uptake of iron, targeting the interaction with iron has become a straightforward strategy in the fight against cancer. This work comprehensively characterizes the chemical properties of 6-methyl-3-{(2 E )-2-[1-(2-pyridinyl)ethylidene]hydrazino}-5 H -[1,2,4]triazino[5,6- b ]indole (VLX600), a clinically investigated iron chelator, in solution. Its protonation processes, lipophilicity, and membrane permeability as well as its complexation with essential metal ions were investigated using UV-visible, electron paramagnetic resonance, and NMR spectroscopic and computational methods. Formation constants revealed the following order of metal binding affinity at pH 7.4: Cu(II) > Fe(II) > Zn(II). The structures of VLX600 (denoted as HL) and the coordination modes in its metal complexes [Cu(II)(LH)Cl
2 ], [Cu(II)(L)(CH3 OH)Cl], [Zn(II)(LH)Cl2 ], and [Fe(II)(LH)2 ](NO3 )2 were elucidated by single-crystal X-ray diffraction. Redox properties of the iron complexes characterized by cyclic voltammetry showed strong preference of VLX600 toward Fe(II) over Fe(III). In vitro cytotoxicity of VLX600 was determined in six different human cancer cell lines, with IC50 values ranging from 0.039 to 0.51 μM. Premixing VLX600 with Fe(III), Zn(II), and Cu(II) salts in stoichiometric ratios had a rather little effect overall, thus neither potentiating nor abolishing cytotoxicity. Together, although clinically investigated as an iron chelator, this is the first comprehensive solution study of VLX600 and its interaction with physiologically essential metal ions.- Published
- 2024
- Full Text
- View/download PDF
7. In vitro biodistribution studies on clinically approved FGFR inhibitors ponatinib, nintedanib, erlotinib and the investigational inhibitor KP2692.
- Author
-
Dömötör O, Mathuber M, and Kowol CR
- Subjects
- Humans, Erlotinib Hydrochloride pharmacology, Tissue Distribution, Protein Binding, Imidazoles pharmacology, Serum Albumin, Human
- Abstract
Binding towards human serum albumin (HSA) and α1-acid glycoprotein (AGP) of three approved fibroblast growth factor receptor (FGFR) inhibitors ponatinib (PON), nintedanib (NIN) and erdafitinib (ERD), as well as the experimental drug KP2692 was studied by means of spectrofluorometric and UV-visible spectrophotometric methods. Additionally, proton dissociation processes, lipophilicity, and fluorescence properties of these four molecules were investigated in detail. The FGFR inhibitors were predominantly presented in their single protonated form (HL
+ ) at pH 7.4 (at blood pH). At gastric pH (pH 1-2) the protonated forms (+1 - +3) are present, which provide relatively good aqueous solubility of the drugs. All of the four inhibitors are highly or extremely lipophilic at pH 7.4 (logD7.4 ≥ 2.7). At acidic pH 2.0 PON and ERD are rather lipophilic, NIN is amphiphilic, while KP2692 is highly hydrophilic. All four compounds bind to HSA and AGP. Moderate binding of PON, KP2692 and NIN was found towards albumin (logK' = 4.5-4.7), while their affinity for AGP was about one order of magnitude higher (logK' = 5.2-5.7). ERD shows a larger affinity for both proteins (logK'HSA ≈ 5.2, logK'AGP ≈ 7.0). The computed constants were used to model the distribution of the FGFR inhibitors in blood plasma under physiological and pathological (acute phase) conditions. The changing levels of the two proteins under pathological conditions compensate each other for PON and NIN, so that the free drug fractions do not change considerably. In the case of ERD the higher AGP levels distinctly reduce the free available fraction of the drug. Comparison with clinical pharmacokinetic data indicates that the here presented solution distribution studies can very well predict the conditions in cancer patients., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Author(s). Published by Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
8. Stepwise optimization of tumor-targeted dual-action platinum(iv)-gemcitabine prodrugs.
- Author
-
Kastner A, Mendrina T, Babu T, Karmakar S, Poetsch I, Berger W, Keppler BK, Gibson D, Heffeter P, and Kowol CR
- Abstract
While platinum-based chemotherapeutic agents have established themselves as indispensable components of anticancer therapy, they are accompanied by a variety of side effects and the rapid occurrence of drug resistance. A promising strategy to address these challenges is the use of platinum(iv) prodrugs, which remain inert until they reach the tumor tissue, thereby mitigating detrimental effects on healthy cells. Typically, platinum drugs are part of combination therapy settings. Consequently, a very elegant strategy is the development of platinum(iv) prodrugs bearing a second, clinically relevant therapeutic in axial position. In the present study, we focused on gemcitabine as an approved antimetabolite, which is highly synergistic with platinum drugs. In addition, to increase plasma half-life and facilitate tumor-specific accumulation, an albumin-binding maleimide moiety was attached. Our investigations revealed that maleimide-cisplatin(iv)-gemcitabine complexes cannot carry sufficient amounts of gemcitabine to induce a significant effect in vivo . Consequently, we designed a carboplatin(iv) analog, that can be applied at much higher doses. Remarkably, this novel analog demonstrated impressive in vivo results, characterized by significant improvements in overall survival. Notably, these encouraging results could also be transferred to an in vivo xenograft model with acquired gemcitabine resistance, indicating the high potential of this approach., Competing Interests: There are no conflicts to declare., (This journal is © the Partner Organisations.)
- Published
- 2023
- Full Text
- View/download PDF
9. Entinostat Enhances the Efficacy of Chemotherapy in Small Cell Lung Cancer Through S-phase Arrest and Decreased Base Excision Repair.
- Author
-
Solta A, Boettiger K, Kovács I, Lang C, Megyesfalvi Z, Ferk F, Mišík M, Hoetzenecker K, Aigner C, Kowol CR, Knasmueller S, Grusch M, Szeitz B, Rezeli M, Dome B, and Schelch K
- Subjects
- Humans, Cisplatin, Proteomics, Apoptosis, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylase Inhibitors therapeutic use, DNA Repair, Cell Line, Tumor, Small Cell Lung Carcinoma drug therapy, Small Cell Lung Carcinoma genetics, Small Cell Lung Carcinoma pathology, Lung Neoplasms pathology
- Abstract
Purpose: Acquired chemoresistance is a frequent event in small cell lung cancer (SCLC), one of the deadliest human malignancies. Histone deacetylase inhibitors (HDACi) have been shown to synergize with different chemotherapeutic agents including cisplatin. Accordingly, we aimed to investigate the dual targeting of HDAC inhibition and chemotherapy in SCLC., Experimental Design: The efficacy of HDACi and chemotherapy in SCLC was investigated both in vitro and in vivo. Synergistic drug interactions were calculated based on the HSA model (Combenefit software). Results from the proteomic analysis were confirmed via ICP-MS, cell-cycle analysis, and comet assays., Results: Single entinostat- or chemotherapy significantly reduced cell viability in human neuroendocrine SCLC cells. The combination of entinostat with either cisplatin, carboplatin, irinotecan, epirubicin, or etoposide led to strong synergy in a subset of resistant SCLC cells. Combination treatment with entinostat and cisplatin significantly decreased tumor growth in vivo. Proteomic analysis comparing the groups of SCLC cell lines with synergistic and additive response patterns indicated alterations in cell-cycle regulation and DNA damage repair. Cell-cycle analysis revealed that cells exhibiting synergistic drug responses displayed a shift from G1 to S-phase compared with cells showing additive features upon dual treatment. Comet assays demonstrated more DNA damage and decreased base excision repair in SCLC cells more responsive to combination therapy., Conclusions: In this study, we decipher the molecular processes behind synergistic interactions between chemotherapy and HDAC inhibition. Moreover, we report novel mechanisms to overcome drug resistance in SCLC, which may be relevant to increasing therapeutic success., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
10. Einbau von (Bioaktiven) Äquatorialen Liganden in Platin(IV)-Komplexe.
- Author
-
Kastner A, Schueffl H, Yassemipour PA, Keppler BK, Heffeter P, and Kowol CR
- Abstract
Competing Interests: Die Autoren erklären, dass keine Interessenkonflikte vorliegen.
- Published
- 2023
- Full Text
- View/download PDF
11. Insertion of (Bioactive) Equatorial Ligands into Platinum(IV) Complexes.
- Author
-
Kastner A, Schueffl H, Yassemipour PA, Keppler BK, Heffeter P, and Kowol CR
- Subjects
- Humans, Platinum, Oxaliplatin, Organoplatinum Compounds, Ligands, Cell Line, Tumor, Prodrugs, Neoplasms, Antineoplastic Agents
- Abstract
Platinum(IV) prodrugs are highly interesting alternatives to platinum(II) anticancer therapeutics due to their increased tumor selectivity and reduced side effects. In contrast to the established theory, we recently observed that the equatorial ligand(s) of e.g. oxaliplatin(IV) complexes can be hydrolyzed with formation of [(DACH)Pt(OH
eq )2 (OAcax )2 ]. In the work presented here, we investigated the reactivity and synthetic usability of this complex to be exploited as a precursor for the development of novel platinum(IV) complexes, not able to be synthesized by conventional protocols. Indeed, we could substitute the equatorial hydroxido ligand(s) e.g. by one or two monodentate biotin ligands (which would be oxidized under standard methods). The formed complexes turned out to be very stable with slow ligand release after reduction, ideal for long-circulating tumor-targeting strategies. Therefore, two platinum(IV) complexes with equatorial maleimides, capable of exploiting serum albumin as a natural nanocarrier, were synthesized as well. The complexes showed massively prolonged plasma half-life and distinctly improved anticancer activity in vivo compared to oxaliplatin. Taken together, the newly developed synthetic platform allows the simple and specific insertion of equatorial ligands into platinum(IV) complexes. This will enable the attachment of three different (bioactive) moieties generating targeted triple-action platinum(IV) prodrugs within one single platinum complex., (© 2023 The Authors. Angewandte Chemie International Edition published by Wiley-VCH GmbH.)- Published
- 2023
- Full Text
- View/download PDF
12. Multi-action platinum(IV) prodrugs conjugated with COX-inhibiting NSAIDs.
- Author
-
Liu X, Wenisch D, Dahlke P, Jordan PM, Jakupec MA, Kowol CR, Liebing P, Werz O, Keppler BK, and Weigand W
- Subjects
- Humans, Platinum pharmacology, Prostaglandin-Endoperoxide Synthases, Cell Line, Tumor, Anti-Inflammatory Agents, Non-Steroidal pharmacology, Cisplatin pharmacology, Prodrugs pharmacology
- Abstract
In the last decades, inflammation has been recognized as being closely connected to cancer, and joint strategies encompassing chemotherapeutic and anti-inflammatory agents have been extensively studied. In this work, a series of novel cisplatin and oxaliplatin-based Pt(IV) complexes comprising non-steroidal anti-inflammatory drugs (NSAIDs) and their carboxyl ester analogues as axial moieties were synthesized. Several of the cisplatin-based Pt(IV) complexes 22-30 showed increased cytotoxicity in the human cancer cell lines CH1/PA-1, SW480 and A549 compared to the Pt(II) drug. For the most potent complex 26, comprising two aceclofenac (AFC) moieties, the formation of Pt(II)-9-methylguanine (9-MeG) adducts after activation with ascorbic acid (AsA) was proven. Additionally, a significant inhibition of cyclooxygenase (COX) activity and prostaglandin E
2 (PGE2 ) production was observed, as well as increased cellular accumulation, depolarization of mitochondrial membranes, and strong proapoptotic potencies in SW480 cells. Overall, these systematic effects shown in vitro confer 26 as a potential anticancer agent combined with anti-inflammatory properties., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier Masson SAS.. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
13. Human serum albumin as a copper source for anticancer thiosemicarbazones.
- Author
-
Schaier M, Falcone E, Prstek T, Vileno B, Hager S, Keppler BK, Heffeter P, Koellensperger G, Faller P, and Kowol CR
- Subjects
- Humans, Serum Albumin, Human, Copper chemistry, Chelating Agents chemistry, Thiosemicarbazones pharmacology, Thiosemicarbazones chemistry, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry
- Abstract
Thiosemicarbazones (TSCs) are a class of biologically active compounds with promising anticancer activity. Their typical mechanism, especially of the clinically far developed representative Triapine, is chelation of iron (Fe), with the Fe-containing enzyme ribonucleotide reductase as primary intracellular target. However, for the subclass of terminally disubstituted, nanomolar-active derivatives like Dp44mT and Me2NNMe2, recent findings suggest that the chelation, stability, and reduction properties of the copper(II) (Cu) complexes are essential for their modes of action. Consequently, it is important to elucidate whether blood serum Cu(II) is a potential metal source for these TSCs. To gain more insights, the interaction of Triapine, Dp44mT or Me2NNMe2 with purified human serum albumin (HSA) as the main pool of labile Cu(II) was investigated by UV-vis and electron paramagnetic resonance measurements. Subsequently, a size-exclusion chromatography inductively coupled plasma mass spectrometry method for the differentiation of Cu species in serum was developed, especially separating the non-labile Cu enzyme ceruloplasmin from HSA. The results indicate that the TSCs specifically chelate copper from the N-terminal Cu-binding site of HSA. Furthermore, the Cu(II)-TSC complexes were shown to form ternary HSA conjugates, most likely via histidine. Noteworthy, Fe-chelation from transferrin was not overserved, even not for Triapine. In summary, the labile Cu pool of HSA is a potential source for Cu-TSC complex formation and, consequently, distinctly influences the anticancer activity and pharmacological behavior of TSCs., (© The Author(s) 2023. Published by Oxford University Press.)
- Published
- 2023
- Full Text
- View/download PDF
14. A novel EGFR inhibitor acts as potent tool for hypoxia-activated prodrug systems and exerts strong synergistic activity with VEGFR inhibition in vitro and in vivo.
- Author
-
Caban M, Koblmueller B, Groza D, Schueffl HH, Terenzi A, Tolios A, Mohr T, Mathuber M, Kryeziu K, Jaunecker C, Pirker C, Keppler BK, Berger W, Kowol CR, and Heffeter P
- Subjects
- Humans, ErbB Receptors metabolism, Protein Kinase Inhibitors therapeutic use, Erlotinib Hydrochloride pharmacology, Cell Proliferation, Hypoxia metabolism, Cell Line, Tumor, Prodrugs pharmacology, Prodrugs therapeutic use, Lung Neoplasms metabolism, Antineoplastic Agents therapeutic use
- Abstract
Small-molecule EGFR inhibitors have distinctly improved the overall survival especially in EGFR-mutated lung cancer. However, their use is often limited by severe adverse effects and rapid resistance development. To overcome these limitations, a hypoxia-activatable Co(III)-based prodrug (KP2334) was recently synthesized releasing the new EGFR inhibitor KP2187 in a highly tumor-specific manner only in hypoxic areas of the tumor. However, the chemical modifications in KP2187 necessary for cobalt chelation could potentially interfere with its EGFR-binding ability. Consequently, in this study, the biological activity and EGFR inhibition potential of KP2187 was compared to clinically approved EGFR inhibitors. In general, the activity as well as EGFR binding (shown in docking studies) was very similar to erlotinib and gefitinib (while other EGFR-inhibitory drugs behaved different) indicating no interference of the chelating moiety with the EGFR binding. Moreover, KP2187 significantly inhibited cancer cell proliferation as well as EGFR pathway activation in vitro and in vivo. Finally, KP2187 proved to be highly synergistic with VEGFR inhibitors such as sunitinib. This indicates that KP2187-releasing hypoxia-activated prodrug systems are promising candidates to overcome the clinically observed enhanced toxicity of EGFR-VEGFR inhibitor combination therapies., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
15. Tumor-targeted dual-action NSAID-platinum(iv) anticancer prodrugs.
- Author
-
Kastner A, Mendrina T, Bachmann F, Berger W, Keppler BK, Heffeter P, and Kowol CR
- Abstract
Platinum(iv) prodrugs are a promising class of anticancer agents designed to overcome the limitations of conventional platinum(ii) therapeutics. In this work, we present oxaliplatin(iv)-based complexes, which upon reduction, release acetylsalicylic acid (aspirin), known for its antitumor activity against colon cancer and currently investigated in combination with oxaliplatin in a phase III clinical study. Comparison with a recently reported cisplatin analog (asplatin) revealed a massive increase in reduction stability for the oxaliplatin complex in mouse serum. This was in line with the cell culture data indicating the desired prodrug properties for the newly synthesized complex. For in vivo studies, a new derivative containing an albumin-binding maleimide unit was synthesized. Indeed, distinctly longer plasma half-life as well as higher tumor accumulation in comparison to asplatin and oxaliplatin were observed, also leading to significantly higher antitumor activity and overall survival of CT26 tumor-bearing mice., Competing Interests: There are no conflicts to declare., (This journal is © the Partner Organisations.)
- Published
- 2023
- Full Text
- View/download PDF
16. Degradable Bottlebrush Polypeptides and the Impact of their Architecture on Cell Uptake, Pharmacokinetics, and Biodistribution In Vivo.
- Author
-
Strasser P, Montsch B, Weiss S, Sami H, Kugler C, Hager S, Schueffl H, Mader R, Brüggemann O, Kowol CR, Ogris M, Heffeter P, and Teasdale I
- Subjects
- Tissue Distribution, Macromolecular Substances, Peptides, Polymers chemistry, Water chemistry
- Abstract
Bottlebrush polymers are highly promising as unimolecular nanomedicines due to their unique control over the critical parameters of size, shape and chemical function. However, since they are prepared from biopersistent carbon backbones, most known bottlebrush polymers are non-degradable and thus unsuitable for systemic therapeutic administration. Herein, we report the design and synthesis of novel poly(organo)phosphazene-g-poly(α-glutamate) (PPz-g-PGA) bottlebrush polymers with exceptional control over their structure and molecular dimensions (Dh ≈ 15-50 nm). These single macromolecules show outstanding aqueous solubility, ultra-high multivalency and biodegradability, making them ideal as nanomedicines. While well-established in polymer therapeutics, it has hitherto not been possible to prepare defined single macromolecules of PGA in these nanosized dimensions. A direct correlation was observed between the macromolecular dimensions of the bottlebrush polymers and their intracellular uptake in CT26 colon cancer cells. Furthermore, the bottlebrush macromolecular structure visibly enhanced the pharmacokinetics by reducing renal clearance and extending plasma half-lives. Real-time analysis of the biodistribution dynamics showed architecture-driven organ distribution and enhanced tumor accumulation. This work, therefore, introduces a robust, controlled synthesis route to bottlebrush polypeptides, overcoming limitations of current polymer-based nanomedicines and, in doing so, offers valuable insights into the influence of architecture on the in vivo performance of nanomedicines., (© 2023 The Authors. Small published by Wiley-VCH GmbH.)
- Published
- 2023
- Full Text
- View/download PDF
17. Influence of the Fatty Acid Metabolism on the Mode of Action of a Cisplatin(IV) Complex with Phenylbutyrate as Axial Ligands.
- Author
-
Mendrina T, Poetsch I, Schueffl H, Baier D, Pirker C, Ries A, Keppler BK, Kowol CR, Gibson D, Grusch M, Berger W, and Heffeter P
- Abstract
For a variety of cancer types, platinum compounds are still among the best treatment options. However, their application is limited by side effects and drug resistance. Consequently, multi-targeted platinum(IV) prodrugs that target specific traits of the malignant tissue are interesting new candidates. Recently, cisPt(PhB)
2 was synthesized which, upon reduction in the malignant tissue, releases phenylbutyrate (PhB), a metabolically active fatty acid analog, in addition to cisplatin. In this study, we in-depth investigated the anticancer properties of this new complex in cell culture and in mouse allograft experiments. CisPt(PhB)2 showed a distinctly improved anticancer activity compared to cisplatin as well as to PhB alone and was able to overcome various frequently occurring drug resistance mechanisms. Furthermore, we observed that differences in the cellular fatty acid metabolism and mitochondrial activity distinctly impacted the drug's mode of action. Subsequent analyses revealed that "Warburg-like" cells, which are characterized by deficient mitochondrial function and fatty acid catabolism, are less capable of coping with cisPt(PhB)2 leading to rapid induction of a non-apoptotic form of cell death. Summarizing, cisPt(PhB)2 is a new orally applicable platinum(IV) prodrug with promising activity especially against cisplatin-resistant cancer cells with "Warburg-like" properties.- Published
- 2023
- Full Text
- View/download PDF
18. Novel oxaliplatin(IV) complexes conjugated with ligands bearing pendant 1,2-dithiolane/1,2-diselenolane/cyclopentyl motifs.
- Author
-
Liu X, Wenisch D, Barth MC, Cseh K, Kowol CR, Jakupec MA, Gibson D, Keppler BK, and Weigand W
- Subjects
- Oxaliplatin pharmacology, Ligands, Reactive Oxygen Species metabolism, Cell Line, Tumor, Antineoplastic Agents chemistry
- Abstract
In this work, biologically active α-lipoic acid (ALA) and its isologous 1,2-diselenolane (SeA) and cyclopentyl (CpA) analogues were investigated for their differences in redox potentials, cytotoxicity and ROS production. In addition, the corresponding Pt(IV) complexes comprising ALA (1-4), SeA (5-8) and CpA (9-12) as axial ligands were synthesized. Those Pt(IV) complexes were characterized by NMR spectroscopy, ESI-mass spectrometry and elemental analysis. The cytotoxicity study showed that 1,2-diselenolane containing Pt(IV) (1, 3 and 4) complexes are more cytotoxic than the 1,2-dithiolane analogues (5, 7, and 8) throughout all tested cell lines, intriguingly, cyclopentyl containing species (9, 11 and 12) are the most effective, in some cases even more potent than the parent drug oxaliplatin. Three representative complexes 2, 6 and 10 were further assessed for their redox potentials, reduction with AsA, lipophilicity, cellular accumulation and ROS production. It turned out that the cytotoxicity profile is an overall result of good lipophilicity, high cellular accumulation, and (partially) enhanced ROS generation.
- Published
- 2022
- Full Text
- View/download PDF
19. Oxoplatin-Based Pt(IV) Lipoate Complexes and Their Biological Activity.
- Author
-
Liu X, Barth MC, Cseh K, Kowol CR, Jakupec MA, Keppler BK, Gibson D, and Weigand W
- Subjects
- Antioxidants, Ascorbic Acid, Cell Line, Tumor, DNA, Ligands, Molecular Structure, Reactive Oxygen Species metabolism, Antineoplastic Agents chemistry, Prodrugs pharmacology, Prodrugs chemistry, Thioctic Acid
- Abstract
α-Lipoic acid, known for its anti-inflammatory and antioxidant activity, represents a promising ligand for Pt(IV) prodrugs. Three new Pt(IV) lipoate complexes were synthesized and characterized by NMR spectroscopy (
1 H,13 C,195 Pt), mass spectrometry and elemental analysis. Due to the low solubility of the complex containing two axial lipoate ligands, further experiments to examine the biological activity were performed with two Pt(IV) complexes containing just one axial lipoate ligand. Both complexes exhibit anticancer activity and produce reactive oxygen species (ROS) in the cell lines tested. Especially, the monosubstituted complex can be reduced by ascorbic acid and forms adducts with 9-methylguanine (9MeG), which is favorable for the formation of DNA-crosslinks in the cells., (© 2022 The Authors. Chemistry & Biodiversity published by Wiley-VHCA AG, Zurich, Switzerland.)- Published
- 2022
- Full Text
- View/download PDF
20. Thiosemicarbazone Derivatives Developed to Overcome COTI-2 Resistance.
- Author
-
Pósa V, Stefanelli A, Nunes JHB, Hager S, Mathuber M, May NV, Berger W, Keppler BK, Kowol CR, Enyedy ÉA, and Heffeter P
- Abstract
COTI-2 is currently being evaluated in a phase I clinical trial for the treatment of gynecological and other solid cancers. As a thiosemicarbazone, this compound contains an N,N,S-chelating moiety and is, therefore, expected to bind endogenous metal ions. However, besides zinc, the metal interaction properties of COTI-2 have not been investigated in detail so far. This is unexpected, as we have recently shown that COTI-2 forms stable ternary complexes with copper and glutathione, which renders this drug a substrate for the resistance efflux transporter ABCC1. Herein, the complex formation of COTI-2, two novel terminal N-disubstituted derivatives (COTI-NMe
2 and COTI-NMeCy), and the non-substituted analogue (COTI-NH2 ) with iron, copper, and zinc ions was characterized in detail. Furthermore, their activities against drug-resistant cancer cells was investigated in comparison to COTI-2 and Triapine. These data revealed that, besides zinc, also iron and copper ions need to be considered to play a role in the mode of action and resistance development of these thiosemicarbazones. Moreover, we identified COTI-NMe2 as an interesting new drug candidate with improved anticancer activity and resistance profile.- Published
- 2022
- Full Text
- View/download PDF
21. Copper-Catalyzed Glutathione Oxidation is Accelerated by the Anticancer Thiosemicarbazone Dp44mT and Further Boosted at Lower pH.
- Author
-
Falcone E, Ritacca AG, Hager S, Schueffl H, Vileno B, El Khoury Y, Hellwig P, Kowol CR, Heffeter P, Sicilia E, and Faller P
- Subjects
- Animals, Catalysis, Glutathione chemistry, Glutathione Disulfide chemistry, Glutathione Disulfide metabolism, Hydrogen-Ion Concentration, Mammals metabolism, Oxidation-Reduction, Sulfhydryl Compounds chemistry, Copper chemistry, Thiosemicarbazones chemistry, Thiosemicarbazones pharmacology
- Abstract
Glutathione (GSH) is the most abundant thiol in mammalian cells and plays a crucial role in maintaining redox cellular homeostasis. The thiols of two GSH molecules can be oxidized to the disulfide GSSG. The cytosolic GSH/GSSG ratio is very high (>100), and its reduction can lead to apoptosis or necrosis, which are of interest in cancer research. Cu
II ions are very efficient oxidants of thiols, but with an excess of GSH, CuI n (GS)m clusters are formed, in which CuI is very slowly reoxidized by O2 at pH 7.4 and even more slowly at lower pH. Here, the aerobic oxidation of GSH by CuII was investigated at different pH values in the presence of the anticancer thiosemicarbazone Dp44mT, which accumulates in lysosomes and induces lysosomal membrane permeabilization in a Cu-dependent manner. The results showed that CuII -Dp44mT catalyzes GSH oxidation faster than CuII alone at pH 7.4 and hence accelerates the production of very reactive hydroxyl radicals. Moreover, GSH oxidation and hydroxyl radical production by CuII -Dp44mT were accelerated at the acidic pH found in lysosomes. To decipher this unusually faster thiol oxidation at lower pH, density functional theory (DFT) calculations, electrochemical and spectroscopic studies were performed. The results suggest that the acceleration is due to the protonation of CuII -Dp44mT on the hydrazinic nitrogen, which favors the rate-limiting reduction step without subsequent dissociation of the CuI intermediate. Furthermore, preliminary biological studies in cell culture using the proton pump inhibitor bafilomycin A1 indicated that the lysosomal pH plays a role in the activity of CuII -Dp44mT.- Published
- 2022
- Full Text
- View/download PDF
22. The coordination modes of (thio)semicarbazone copper(II) complexes strongly modulate the solution chemical properties and mechanism of anticancer activity.
- Author
-
Pósa V, Hajdu B, Tóth G, Dömötör O, Kowol CR, Keppler BK, Spengler G, Gyurcsik B, and Enyedy ÉA
- Subjects
- Copper chemistry, Ferric Compounds, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Coordination Complexes chemistry, Coordination Complexes pharmacology, Semicarbazones pharmacology
- Abstract
Thiosemicarbazones are promising candidates for anticancer therapy and their mechanism of action is often linked to their metal chelating ability. In this study, five (thio)semicarbazones with different donor sets (NNS, NNO, ONS, ONO) were selected and their behaviour in aqueous solution, the stability of their copper(II) complexes in addition to their cytotoxicity, DNA-binding, DNA cleavage ability and inhibition of topoisomerase IIα were investigated and compared. We aimed to reveal relationships between the structural variations, the significantly different physico-chemical properties, solution speciation and biological activity. The cytotoxicity of the ligands did not show correlation with the solubility, lipophilicity and permeability; and the decreased activity of the oxygen donor containing compounds was explained by their stronger preference towards chelation of iron(III) over iron(II). Meanwhile, among the copper complexes the most lipophilic species with the highest stability and membrane permeability exhibited the highest cytotoxicity. The studied copper(II) complexes interact with DNA, and reaction with glutathione led to heavy DNA cleavage in the case of the highly stable complexes which could be reduced in a reversible reaction with moderate rate. All the tested copper complexes inhibited topoisomerase IIα, however, this property of the complexes with low stability is most probably linked to the liberated free copper(II)., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
23. A platinum(IV) prodrug strategy to overcome glutathione-based oxaliplatin resistance.
- Author
-
Fronik P, Gutmann M, Vician P, Stojanovic M, Kastner A, Heffeter P, Pirker C, Keppler BK, Berger W, and Kowol CR
- Abstract
Clinical efficacy of oxaliplatin is frequently limited by severe adverse effects and therapy resistance. Acquired insensitivity to oxaliplatin is, at least in part, associated with elevated levels of glutathione (GSH). In this study we report on an oxaliplatin-based platinum(IV) prodrug, which releases L-buthionine-S,R-sulfoximine (BSO), an inhibitor of glutamate-cysteine ligase, the rate-limiting enzyme in GSH biosynthesis. Two complexes bearing either acetate (BSO-OxOAc) or an albumin-binding maleimide (BSO-OxMal) as second axial ligand were synthesized and characterized. The in vitro anticancer activity of BSO-OxOAc was massively reduced in comparison to oxaliplatin, proving its prodrug nature. Nevertheless, the markedly lower intracellular oxaliplatin uptake in resistant HCT116/OxR cells was widely overcome by BSO-OxOAc resulting in distinctly reduced resistance levels. Platinum accumulation in organs of a colorectal cancer mouse model revealed higher tumor selectivity of BSO-OxMal as compared to oxaliplatin. This corresponded with increased antitumor activity, resulting in significantly enhanced overall survival. BSO-OxMal-treated tumors exhibited reduced GSH levels, proliferative activity and enhanced DNA damage (pH2AX) compared to oxaliplatin. Conversely, pH2AX staining especially in kidney cells was distinctly increased by oxaliplatin but not by BSO-OxMal. Taken together, our data provide compelling evidence for enhanced tumor specificity of the oxaliplatin(IV)/BSO prodrug., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
24. Landomycins as glutathione-depleting agents and natural fluorescent probes for cellular Michael adduct-dependent quinone metabolism.
- Author
-
Terenzi A, La Franca M, van Schoonhoven S, Panchuk R, Martínez Á, Heffeter P, Gober R, Pirker C, Vician P, Kowol CR, Stoika R, Salassa L, Rohr J, and Berger W
- Abstract
Landomycins are angucyclines with promising antineoplastic activity produced by Streptomyces bacteria. The aglycone landomycinone is the distinctive core, while the oligosaccharide chain differs within derivatives. Herein, we report that landomycins spontaneously form Michael adducts with biothiols, including reduced cysteine and glutathione, both cell-free or intracellularly involving the benz[a]anthraquinone moiety of landomycinone. While landomycins generally do not display emissive properties, the respective Michael adducts exerted intense blue fluorescence in a glycosidic chain-dependent manner. This allowed label-free tracking of the short-lived nature of the mono-SH-adduct followed by oxygen-dependent evolution with addition of another SH-group. Accordingly, hypoxia distinctly stabilized the fluorescent mono-adduct. While extracellular adduct formation completely blocked the cytotoxic activity of landomycins, intracellularly it led to massively decreased reduced glutathione levels. Accordingly, landomycin E strongly synergized with glutathione-depleting agents like menadione but exerted reduced activity under hypoxia. Summarizing, landomycins represent natural glutathione-depleting agents and fluorescence probes for intracellular anthraquinone-based angucycline metabolism., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
25. Liposomal formulations of anticancer copper(II) thiosemicarbazone complexes.
- Author
-
Mathuber M, Hager S, Keppler BK, Heffeter P, and Kowol CR
- Subjects
- Animals, Cell Line, Tumor, Cell Survival drug effects, Drug Liberation, Female, Humans, Liposomes, Methemoglobin metabolism, Mice, Inbred BALB C, Mice, Antineoplastic Agents administration & dosage, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacokinetics, Coordination Complexes administration & dosage, Coordination Complexes chemistry, Coordination Complexes pharmacokinetics, Copper administration & dosage, Copper chemistry, Copper pharmacokinetics, Thiosemicarbazones administration & dosage, Thiosemicarbazones chemistry, Thiosemicarbazones pharmacokinetics
- Abstract
α-N-Heterocyclic thiosemicarbazones such as triapine and COTI-2 are currently investigated as anticancer therapeutics in clinical trials. However, triapine was widely inactive against solid tumor types. A likely explanation is the short plasma half-life time and fast metabolism. One promising approach to overcome these drawbacks is the encapsulation of the drug into nanoparticles (passive drug-targeting). In a previous work we showed that it was not possible to stably encapsulate free triapine into liposomes. Hence, in this manuscript we present the successful preparation of liposomal formulations of the copper(II) complexes of triapine and COTI-2. To this end, various drug-loading strategies were examined and the resulting liposomes were physico-chemically characterized. Especially for liposomal Cu-triapine, a decent encapsulation efficacy and a slow drug release behavior could be observed. In contrast, for COTI-2 and its copper(II) complex no stable loading could be achieved. Subsequent in vitro studies in different cell lines with liposomal Cu-triapine showed the expected strongly reduced cytotoxicity and DNA damage induction. Also in vivo distinctly higher copper plasma levels and a continuous release could be observed for the liposomal formulation compared to free Cu-triapine. Taken together, the here presented nanoformulation of Cu-triapine is an important step further to increase the plasma half-life time and tumor targeting properties of anticancer thiosemicarbazones.
- Published
- 2021
- Full Text
- View/download PDF
26. Albumin-targeting of an oxaliplatin-releasing platinum(iv) prodrug results in pronounced anticancer activity due to endocytotic drug uptake in vivo .
- Author
-
Schueffl H, Theiner S, Hermann G, Mayr J, Fronik P, Groza D, van Schonhooven S, Galvez L, Sommerfeld NS, Schintlmeister A, Reipert S, Wagner M, Mader RM, Koellensperger G, Keppler BK, Berger W, Kowol CR, Legin A, and Heffeter P
- Abstract
Oxaliplatin is a very potent platinum(ii) drug which is frequently used in poly-chemotherapy schemes against advanced colorectal cancer. However, its benefit is limited by severe adverse effects as well as resistance development. Based on their higher tolerability, platinum(iv) prodrugs came into focus of interest. However, comparable to their platinum(ii) counterparts they lack tumor specificity and are frequently prematurely activated in the blood circulation. With the aim to exploit the enhanced albumin consumption and accumulation in the malignant tissue, we have recently developed a new albumin-targeted prodrug, which supposed to release oxaliplatin in a highly tumor-specific manner. In more detail, we designed a platinum(iv) complex containing two maleimide moieties in the axial position (KP2156), which allows selective binding to the cysteine 34. In the present study, diverse cell biological and analytical tools such as laser ablation inductively-coupled plasma mass spectrometry (LA-ICP-MS), isotope labeling, and nano-scale secondary ion mass spectrometry (NanoSIMS) were employed to better understand the in vivo distribution and activation process of KP2156 (in comparison to free oxaliplatin and a non-albumin-binding succinimide analogue). KP2156 forms very stable albumin adducts in the bloodstream resulting in a superior pharmacological profile, such as distinctly prolonged terminal excretion half-life and enhanced effective platinum dose (measured by ICP-MS). The albumin-bound drug is accumulating in the malignant tissue, where it enters the cancer cells via clathrin- and caveolin-dependent endocytosis, and is activated by reduction to release oxaliplatin. This results in profound, long-lasting anticancer activity of KP2156 against CT26 colon cancer tumors in vivo based on cell cycle arrest and apoptotic cell death. Summarizing, albumin-binding of platinum(iv) complexes potently enhances the efficacy of oxaliplatin therapy and should be further developed towards clinical phase I trials., Competing Interests: BKK, WB, CRK and PH are co-inventors of a patent on albumin-targeted platinum(iv) drugs and co-founders of the spin-off company P4 Therapeutics. All other authors declare no conflict of interest., (This journal is © The Royal Society of Chemistry.)
- Published
- 2021
- Full Text
- View/download PDF
27. Structure-Activity Relationships of Triple-Action Platinum(IV) Prodrugs with Albumin-Binding Properties and Immunomodulating Ligands.
- Author
-
Fronik P, Poetsch I, Kastner A, Mendrina T, Hager S, Hohenwallner K, Schueffl H, Herndler-Brandstetter D, Koellensperger G, Rampler E, Kopecka J, Riganti C, Berger W, Keppler BK, Heffeter P, and Kowol CR
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents pharmacology, Cell Line, Tumor, Coordination Complexes chemical synthesis, Coordination Complexes pharmacology, Drug Screening Assays, Antitumor, Female, Humans, Immunologic Factors chemical synthesis, Immunologic Factors pharmacology, Indoleamine-Pyrrole 2,3,-Dioxygenase antagonists & inhibitors, Male, Maleimides chemical synthesis, Maleimides pharmacology, Mice, Inbred BALB C, Mice, SCID, Molecular Structure, Platinum chemistry, Prodrugs chemical synthesis, Prodrugs pharmacology, Structure-Activity Relationship, Succinimides chemical synthesis, Succinimides pharmacology, Succinimides therapeutic use, Mice, Antineoplastic Agents therapeutic use, Coordination Complexes therapeutic use, Immunologic Factors therapeutic use, Maleimides therapeutic use, Neoplasms drug therapy, Prodrugs therapeutic use
- Abstract
Chemotherapy with platinum complexes is essential for clinical anticancer therapy. However, due to side effects and drug resistance, further drug improvement is urgently needed. Herein, we report on triple-action platinum(IV) prodrugs, which, in addition to tumor targeting via maleimide-mediated albumin binding, release the immunomodulatory ligand 1-methyl-d-tryptophan (1-MDT). Unexpectedly, structure-activity relationship analysis showed that the mode of 1-MDT conjugation distinctly impacts the reducibility and thus activation of the prodrugs. This in turn affected ligand release, pharmacokinetic properties, efficiency of immunomodulation, and the anticancer activity in vitro and in a mouse model in vivo . Moreover, we could demonstrate that the design of albumin-targeted multi-modal prodrugs using platinum(IV) is a promising strategy to enhance the cellular uptake of bioactive ligands with low cell permeability (1-MDT) and to improve their selective delivery into the malignant tissue. This will allow tumor-specific anticancer therapy supported by a favorably tuned immune microenvironment.
- Published
- 2021
- Full Text
- View/download PDF
28. Development of a cobalt(iii)-based ponatinib prodrug system.
- Author
-
Mathuber M, Gutmann M, La Franca M, Vician P, Laemmerer A, Moser P, Keppler BK, Berger W, and Kowol CR
- Abstract
Receptor tyrosine kinase inhibitors have become a central part of modern targeted cancer therapy. However, their curative potential is distinctly limited by both rapid resistance development and severe adverse effects. Consequently, tumor-specific drug activation based on prodrug designs, exploiting tumor-specific properties such as hypoxic oxygen conditions, is a feasible strategy to widen the therapeutic window. After proof-of-principal molecular docking studies, we have synthesized two cobalt(iii) complexes using a derivative of the clinically approved Abelson (ABL) kinase and fibroblast growth factor receptor (FGFR) inhibitor ponatinib. Acetylacetone (acac) or methylacetylacetone (Meacac) have been used as ancillary ligands to modulate the reduction potential. The ponatinib derivative, characterized by an ethylenediamine moiety instead of the piperazine ring, exhibited comparable cell-free target kinase inhibition potency. Hypoxia-dependent release of the ligand from the cobalt(iii) complexes was proven by changed fluorescence properties, enhanced downstream signaling inhibition and increased in vitro anticancer activity in BCR-ABL- and FGFR-driven cancer models. Respective tumor-inhibiting in vivo effects in the BCR-ABL-driven K-562 leukemia model were restricted to the cobalt(iii) complex with the higher reduction potential and confirmed in a FGFR-driven urothelial carcinoma xenograft model. Summarizing, we here present for the first time hypoxia-activatable prodrugs of the clinically approved tyrosine kinase inhibitor ponatinib and a correlation of the in vivo activity with their reduction potential., Competing Interests: There are no conflicts to declare., (This journal is © the Partner Organisations.)
- Published
- 2021
- Full Text
- View/download PDF
29. Complex formation and cytotoxicity of Triapine derivatives: a comparative solution study on the effect of the chalcogen atom and NH-methylation.
- Author
-
Enyedy ÉA, May NV, Pape VFS, Heffeter P, Szakács G, Keppler BK, and Kowol CR
- Subjects
- Cell Line, Tumor, Copper chemistry, Humans, Iron chemistry, Methylation, Solutions, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Chalcogens chemistry, Coordination Complexes chemistry, Coordination Complexes pharmacology, Pyridines chemistry, Thiosemicarbazones chemistry
- Abstract
α-N-Heterocyclic thiosemicarbazones are an important class of investigational anticancer drugs. The most prominent representative is 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (Triapine), which has shown promising results in clinical trials and is currently evaluated in phase III. In this study, we investigated the influence of a chalcogen atom exchange from S (Triapine) to O (O-Triapine) and Se (Se-Triapine) and the methylation of the hydrazonic NH moiety (Me-Triapine) on their complexation with Fe(ii), Fe(iii) and Cu(ii) ions and their cytotoxicity. The main aim of this study was to characterize and compare the most feasible chemical forms in solution, their stability and redox properties, as well as to reveal the relationships of the solution speciation and kinetic data with cytotoxic activity. The complex equilibria and redox properties of the complexes were characterized by the combined use of pH-potentiometry, UV-visible spectrophotometry, electron paramagnetic resonance spectroscopy, and cyclic voltammetry. These revealed that Se-Triapine forms Cu(ii) complexes with higher, and O-Triapine with lower stability as compared with Triapine. Me-Triapine, which is not able to coordinate via the typical (N,N,S-) donor set, nevertheless coordinates to Cu(ii) with unexpected high stability. The Cu(ii) complexes of Se-Triapine and Me-Triapine can be relatively slowly reduced by glutathione at pH 7.4 (but not by ascorbate), similarly to Cu(ii)-Triapine. In contrast, the Cu(ii)-O-Triapine complex can be reduced by both reducing agents in rapid redox reactions. Se-Triapine and Triapine form high stability complexes with both Fe(ii) and Fe(iii) ions, while O-Triapine has a much stronger preference towards Fe(iii) and Me-Triapine towards Fe(ii). This difference in the iron preference of the ligands seems to have a strong impact on their cytotoxic effects, which was measured in a human uterine sarcoma cell line (MES-SA) and its multidrug-resistant subline (MES-SA/Dx5). The Cu(ii) complexes of these calcogensemicarbazones are moderately toxic, and the highest level of ROS generation was found for the Cu(ii) complex of O-Triapine, which is the most reducible.
- Published
- 2020
- Full Text
- View/download PDF
30. Improving the Stability of EGFR Inhibitor Cobalt(III) Prodrugs.
- Author
-
Mathuber M, Schueffl H, Dömötör O, Karnthaler C, Enyedy ÉA, Heffeter P, Keppler BK, and Kowol CR
- Subjects
- Cobalt chemistry, Coordination Complexes chemical synthesis, Coordination Complexes chemistry, Drug Stability, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism, Humans, Ligands, Molecular Structure, Prodrugs chemical synthesis, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors chemistry, Tumor Cells, Cultured, Cobalt pharmacology, Coordination Complexes pharmacology, Prodrugs chemistry, Prodrugs pharmacology, Protein Kinase Inhibitors pharmacology
- Abstract
Although tyrosine kinase inhibitors (TKIs) have revolutionized cancer therapy in the past two decades, severe drawbacks such as strong adverse effects and drug resistance limit their clinical application. Prodrugs represent a valuable approach to overcoming these disadvantages by administration of an inactive drug with tumor-specific activation. We have recently shown that hypoxic prodrug activation is a promising strategy for a cobalt(III) complex bearing a TKI of the epidermal growth factor receptor (EGFR). The aim of this study was the optimization of the physicochemical properties and enhancement of the stability of this compound class. Therefore, we synthesized a series of novel derivatives to investigate the influence of the electron-donating properties of methyl substituents at the metal-chelating moiety of the EGFR inhibitor and/or the ancillary acetylacetonate (acac) ligand. To understand the effect of the different methylations on the redox properties, the newly synthesized complexes were analyzed by cyclic voltammetry and their behavior was studied in the presence of natural low-molecular weight reducing agents. Furthermore, it was proven that reduction to cobalt(II) resulted in a lower stability of the complexes and subsequent release of the coordinated TKI ligand. Moreover, the stability of the cobalt(III) prodrugs was investigated in blood serum as well as in cell culture by diverse cell and molecular biological methods. These analyses revealed that the complexes bearing the methylated acac ligand are characterized by distinctly enhanced stability. Finally, the cytotoxic activity of all new compounds was tested in cell culture under normoxic and various hypoxic conditions, and their prodrug nature could be correlated convincingly with the stability data. In summary, the performed chemical modifications resulted in new cobalt(III) prodrugs with strongly improved stabilities together with retained hypoxia-activatable properties.
- Published
- 2020
- Full Text
- View/download PDF
31. Improving the Stability of Maleimide-Thiol Conjugation for Drug Targeting.
- Author
-
Lahnsteiner M, Kastner A, Mayr J, Roller A, Keppler BK, and Kowol CR
- Subjects
- Cyclization, Drug Delivery Systems, Drug Stability, Succinimides chemistry, Immunoconjugates chemistry, Maleimides chemistry, Sulfhydryl Compounds chemistry
- Abstract
Maleimides are essential compounds for drug conjugation reactions via thiols to antibodies, peptides and other targeting units. However, one main drawback is the occurrence of thiol exchange reactions with, for example, glutathione resulting in loss of the targeting ability. A new strategy to overcome such retro-Michael exchange processes of maleimide-thiol conjugates by stabilization of the thiosuccinimide via a transcyclization reaction is presented. This reaction enables the straightforward synthesis of stable maleimide-thiol adducts essential in drug-conjugation applications., (© 2020 The Authors. Published by Wiley-VCH GmbH.)
- Published
- 2020
- Full Text
- View/download PDF
32. Cancer Cell Resistance Against the Clinically Investigated Thiosemicarbazone COTI-2 Is Based on Formation of Intracellular Copper Complex Glutathione Adducts and ABCC1-Mediated Efflux.
- Author
-
Bormio Nunes JH, Hager S, Mathuber M, Pósa V, Roller A, Enyedy ÉA, Stefanelli A, Berger W, Keppler BK, Heffeter P, and Kowol CR
- Subjects
- Aminoquinolines chemistry, Aminoquinolines pharmacology, Antineoplastic Agents chemistry, Antineoplastic Agents metabolism, Antineoplastic Agents pharmacology, Cell Line, Tumor, Copper chemistry, Drug Resistance, Neoplasm drug effects, Glutathione chemistry, Humans, Intracellular Fluid drug effects, Thiosemicarbazones chemistry, Thiosemicarbazones pharmacology, X-Ray Diffraction, Aminoquinolines metabolism, Copper metabolism, Drug Resistance, Neoplasm physiology, Glutathione metabolism, Intracellular Fluid metabolism, Multidrug Resistance-Associated Proteins metabolism, Thiosemicarbazones metabolism
- Abstract
COTI-2 is a novel anticancer thiosemicarbazone in phase I clinical trial. However, the effects of metal complexation (a main characteristic of thiosemicarbazones) and acquired resistance mechanisms are widely unknown. Therefore, in this study, the copper and iron complexes of COTI-2 were synthesized and evaluated for their anticancer activity and impact on drug resistance in comparison to metal-free thiosemicarbazones. Investigations using Triapine-resistant SW480/Tria and newly established COTI-2-resistant SW480/Coti cells revealed distinct structure-activity relationships. SW480/Coti cells were found to overexpress ABCC1, and COTI-2 being a substrate for this efflux pump. This was unexpected, as ABCC1 has strong selectivity for glutathione adducts. The recognition by ABCC1 could be explained by the reduction kinetics of a ternary Cu-COTI-2 complex with glutathione. Thus, only thiosemicarbazones forming stable, nonreducible copper(II)-glutathione adducts are recognized and, in turn, effluxed by ABCC1. This reveals a crucial connection between copper complex chemistry, glutathione interaction, and the resistance profile of clinically relevant thiosemicarbazones.
- Published
- 2020
- Full Text
- View/download PDF
33. Lipid droplet-mediated scavenging as novel intrinsic and adaptive resistance factor against the multikinase inhibitor ponatinib.
- Author
-
Englinger B, Laemmerer A, Moser P, Kallus S, Röhrl C, Pirker C, Baier D, Mohr T, Niederstaetter L, Meier-Menches SM, Gerner C, Gabler L, Gojo J, Timelthaler G, Senkiv J, Jäger W, Kowol CR, Heffeter P, and Berger W
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Humans, Imidazoles therapeutic use, Lung Neoplasms genetics, Lung Neoplasms pathology, Mice, Protein Kinase Inhibitors therapeutic use, Pyridazines therapeutic use, Receptor, Fibroblast Growth Factor, Type 1 antagonists & inhibitors, Receptor, Fibroblast Growth Factor, Type 1 genetics, Signal Transduction, Tumor Microenvironment, Xenograft Model Antitumor Assays, Drug Resistance, Neoplasm, Imidazoles pharmacokinetics, Lipid Droplets metabolism, Lung Neoplasms drug therapy, Protein Kinase Inhibitors pharmacokinetics, Pyridazines pharmacokinetics
- Abstract
Ponatinib is a small molecule multi-tyrosine kinase inhibitor clinically approved for anticancer therapy. Molecular mechanisms by which cancer cells develop resistance against ponatinib are currently poorly understood. Likewise, intracellular drug dynamics, as well as potential microenvironmental factors affecting the activity of this compound are unknown. Cell/molecular biological and analytical chemistry methods were applied to investigate uptake kinetics/subcellular distribution, the role of lipid droplets (LDs) and lipoid microenvironment compartments in responsiveness of FGFR1-driven lung cancer cells toward ponatinib. Selection of lung cancer cells for acquired ponatinib resistance resulted in elevated intracellular lipid levels. Uncovering intrinsic ponatinib fluorescence enabled dissection of drug uptake/retention kinetics in vitro as well as in mouse tissue cryosections, and revealed selective drug accumulation in LDs of cancer cells. Pharmacological LD upmodulation or downmodulation indicated that the extent of LD formation and consequent ponatinib incorporation negatively correlated with anticancer drug efficacy. Co-culturing with adipocytes decreased ponatinib levels and fostered survival of cancer cells. Ponatinib-selected cancer cells exhibited increased LD levels and enhanced ponatinib deposition into this organelle. Our findings demonstrate intracellular deposition of the clinically approved anticancer compound ponatinib into LDs. Furthermore, increased LD biogenesis was identified as adaptive cancer cell-defense mechanism via direct drug scavenging. Together, this suggests that LDs represent an underestimated organelle influencing intracellular pharmacokinetics and activity of anticancer tyrosine kinase inhibitors. Targeting LD integrity might constitute a strategy to enhance the activity not only of ponatinib, but also other clinically approved, lipophilic anticancer therapeutics., (© 2020 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)
- Published
- 2020
- Full Text
- View/download PDF
34. High Copper Complex Stability and Slow Reduction Kinetics as Key Parameters for Improved Activity, Paraptosis Induction, and Impact on Drug-Resistant Cells of Anticancer Thiosemicarbazones.
- Author
-
Hager S, Pape VFS, Pósa V, Montsch B, Uhlik L, Szakács G, Tóth S, Jabronka N, Keppler BK, Kowol CR, Enyedy ÉA, and Heffeter P
- Subjects
- Antioxidants chemistry, Antioxidants pharmacology, Cell Line, Tumor, Cell Survival drug effects, Endoplasmic Reticulum metabolism, Humans, Oxidation-Reduction drug effects, Oxidative Stress drug effects, Solubility, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Copper chemistry, Drug Resistance, Neoplasm drug effects, Thiosemicarbazones chemistry, Thiosemicarbazones pharmacology
- Abstract
Aims: Due to their significant biological activity, thiosemicarbazones (TSCs) are promising candidates for anticancer therapy. In part, the efficacy of TSCs is linked to their ability to chelate essential metal ions such as copper and iron. Triapine, the best-studied anticancer TSC, has been tested clinically with promising results in hematological diseases. During the past few years, a novel subclass of TSCs with improved anticancer activity was found to induce paraptosis, a recently characterized form of cell death. The aim of this study was to identify structural and chemical properties associated with anticancer activity and paraptosis induction of TSCs. Results: When testing a panel of structurally related TSCs, compounds with nanomolar anticancer activity and paraptosis-inducing properties showed higher copper(II) complex solution stability and a slower reduction rate, which resulted in reduced redox activity. In contrast, TSCs with lower anticancer activity induced higher levels of superoxide that rapidly stimulated superoxide dismutase expression in treated cells, effectively protecting the cells from drug-induced redox stress. Innovation: Consequently, we hypothesize that in the case of close Triapine derivatives, intracellular reduction leads to rapid dissociation of intracellularly formed copper complexes. In contrast, TSCs characterized by highly stable, slowly reducible copper(II) complexes are able to reach new intracellular targets such as the endoplasmic reticulum-resident protein disulfide isomerase. Conclusion: The additional modes of actions observed with highly active TSC derivatives are based on intracellular formation of stable copper complexes, offering a new approach to combat (drug-resistant) cancer cells.
- Published
- 2020
- Full Text
- View/download PDF
35. Development and biological investigations of hypoxia-sensitive prodrugs of the tyrosine kinase inhibitor crizotinib.
- Author
-
Bielec B, Schueffl H, Terenzi A, Berger W, Heffeter P, Keppler BK, and Kowol CR
- Subjects
- Anaplastic Lymphoma Kinase metabolism, Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Cell Proliferation drug effects, Cells, Cultured, Crizotinib chemical synthesis, Crizotinib chemistry, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Humans, Molecular Structure, Prodrugs chemical synthesis, Prodrugs chemistry, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors chemistry, Proto-Oncogene Proteins c-met metabolism, Structure-Activity Relationship, Anaplastic Lymphoma Kinase antagonists & inhibitors, Antineoplastic Agents pharmacology, Cell Hypoxia drug effects, Crizotinib pharmacology, Drug Development, Prodrugs pharmacology, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-met antagonists & inhibitors
- Abstract
Despite the huge success of tyrosine kinase inhibitors as anticancer agents, severe side effects are a major problem. In order to overcome this drawback, the first hypoxia-activatable 2-nitroimidazole-based prodrugs of the clinically approved ALK and c-MET inhibitor crizotinib were developed. The 2-aminopyridine functionality of crizotinib (essential for target kinase binding) was considered as ideal position for prodrug derivatization. Consequently, two different prodrugs were synthesized with the nitroimidazole unit attached to crizotinib either via carbamoylation (A) or alkylation (B) of the 2-aminopyridine moiety. The successful prodrug design could be proven by docking studies and a dramatically reduced ALK and c-MET kinase-inhibitory potential. Furthermore, the prodrugs showed high stability in serum and release of crizotinib in an enzymatic nitroreductase-based cleavage assay was observed for prodrug A. The in vitro activity of both prodrugs was investigated against ALK- and c-MET-dependent or -overexpressing cells, revealing a distinct hypoxia-dependent activation for prodrug A. Finally, inhibition of c-MET phosphorylation and cell proliferation could also be proven in vivo. In summary of the theoretical, chemical and biological studies, prodrug derivatization of the 2-aminopyridine position can be considered as a promising strategy to reduce the side effects and improve the anticancer activity of crizotinib., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
36. Reactive Oxygen Species (ROS)-Sensitive Prodrugs of the Tyrosine Kinase Inhibitor Crizotinib.
- Author
-
Bielec B, Poetsch I, Ahmed E, Heffeter P, Keppler BK, and Kowol CR
- Subjects
- Boronic Acids chemistry, Cell Line, Tumor, Cell Survival drug effects, Flow Cytometry, Humans, Prodrugs chemistry, Prodrugs metabolism, Protein Kinase Inhibitors pharmacology, Protein Stability, Proto-Oncogene Proteins chemistry, Proto-Oncogene Proteins metabolism, Reactive Oxygen Species chemistry, Reactive Oxygen Species metabolism, Crizotinib chemistry, Protein Kinase Inhibitors chemistry
- Abstract
Tyrosine kinase inhibitors revolutionized cancer therapy but still evoke strong adverse effects that can dramatically reduce patients' quality of life. One possibility to enhance drug safety is the exploitation of prodrug strategies to selectively activate a drug inside the tumor tissue. In this study, we designed a prodrug strategy for the approved c-MET, ALK, and ROS1 tyrosine kinase inhibitor crizotinib. Therefore, a boronic-acid trigger moiety was attached to the 2-aminopyridine group of crizotinib, which is a crucial position for target kinase binding. The influence of the modifications on the c-MET- and ALK-binding ability was investigated by docking studies, and the strongly reduced interactions could be confirmed by cell-free kinase inhibition assay. Furthermore, the newly synthesized compounds were tested for their activation behavior with H
2 O2 and their stability in cell culture medium and serum. Finally, the biological activity of the prodrugs was investigated in three cancer cell lines and revealed a good correlation between activity and intrinsic H2 O2 levels of the cells for prodrug A . Furthermore, the activity of this prodrug was distinctly reduced in a non-malignant, c-MET expressing human lung fibroblast (HLF) cell line.- Published
- 2020
- Full Text
- View/download PDF
37. Synthesis and Cytotoxicity of Water-Soluble Dual- and Triple-Action Satraplatin Derivatives: Replacement of Equatorial Chlorides of Satraplatin by Acetates.
- Author
-
Karmakar S, Poetsch I, Kowol CR, Heffeter P, and Gibson D
- Abstract
Pt(II) complexes, such as cisplatin and oxaliplatin, are in widespread use as anticancer drugs. Their use is limited by the toxic side effects and the ability of tumors to develop resistance to the drugs. A popular approach to overcome these drawbacks is to use their kinetically inert octahedral Pt(IV) derivatives that act as prodrugs. The most successful Pt(IV) complex in clinical trials to date is satraplatin, cct -[Pt(NH
3 )(c-hexylamine)Cl2 (OAc)2 ], that upon cellular reduction releases the cytotoxic cis -[Pt(NH3 )(c-hexylamine)Cl2 ]. In an attempt to obtain water-soluble and more effective cytotoxic Pt(IV) complexes, we prepared a series of dual- and triple-action satraplatin analogues, where the equatorial chlorido ligands were replaced with acetates and the axial ligands include innocent and bioactive ligands. Replacement of the chlorides with acetates enhanced the water solubility of the compounds and, with one exception, all of the compounds were very stable in buffer. In general, compounds with one or two axial hydroxido ligands were reduced by ascorbate significantly more quickly than compounds with two axial carboxylates. While replacement of the chlorides with acetates in satraplatin led to a reduction in cytotoxicity, the dual- and triple-action analogues with equatorial acetates had low- to sub-micromolar IC50 values in a panel of eight cancer cells. The triple-action compound cct -[Pt(NH3 )(c-hexylamine)(OAc)2 (PhB)(DCA)] was active in all cell lines, causing DNA damage that induced cell cycle inhibition and apoptosis. Its good activity against CT26 cells in vitro translated into good in vivo efficacy against the CT26 allograft, an in vivo model with intrinsic satraplatin resistance. This indicates that multiaction Pt(IV) derivatives of diamine dicarboxylates are interesting anticancer drug candidates.- Published
- 2019
- Full Text
- View/download PDF
38. A Dogma in Doubt: Hydrolysis of Equatorial Ligands of Pt IV Complexes under Physiological Conditions.
- Author
-
Kastner A, Poetsch I, Mayr J, Burda JV, Roller A, Heffeter P, Keppler BK, and Kowol CR
- Abstract
Due to their high kinetic inertness and consequently reduced side reactions with biomolecules, Pt
IV complexes are considered to define the future of anticancer platinum drugs. The aqueous stability of a series of biscarboxylato PtIV complexes was studied under physiologically relevant conditions. Unexpectedly and in contrast to the current chemical understanding, especially oxaliplatin and satraplatin complexes underwent fast hydrolysis in equatorial position (even in cell culture medium and serum). Notably, the resulting hydrolysis products strongly differ in their reduction kinetics, a crucial parameter for the activation of PtIV drugs, which also changes the anticancer potential of the compounds in cell culture. The discovery that intact PtIV complexes can hydrolyze at equatorial position contradicts the dogma on the general kinetic inertness of PtIV compounds and needs to be considered in the screening and design for novel platinum-based anticancer drugs., (© 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)- Published
- 2019
- Full Text
- View/download PDF
39. Anticancer Thiosemicarbazones: Chemical Properties, Interaction with Iron Metabolism, and Resistance Development.
- Author
-
Heffeter P, Pape VFS, Enyedy ÉA, Keppler BK, Szakacs G, and Kowol CR
- Subjects
- Aminoquinolines pharmacology, Antineoplastic Agents pharmacology, Copper metabolism, Drug Resistance, Neoplasm, Humans, Neoplasms drug therapy, Pyridines pharmacology, Thiosemicarbazones pharmacology, Antineoplastic Agents chemistry, Iron metabolism, Neoplasms metabolism, Thiosemicarbazones chemistry
- Abstract
Significance: During the past decades, thiosemicarbazones were clinically developed for a variety of diseases, including tuberculosis, viral infections, malaria, and cancer. With regard to malignant diseases, the class of α-N-heterocyclic thiosemicarbazones, and here especially 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (Triapine), was intensively developed in multiple clinical phase I/II trials. Recent Advances: Very recently, two new derivatives, namely COTI-2 and di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) have entered phase I evaluation. Based on the strong metal-chelating/metal-interacting properties of thiosemicarbazones, interference with the cellular iron (and copper) homeostasis is assumed to play an important role in their biological activity., Critical Issues: In this review, we summarize and analyze the data on the interaction of (α-N-heterocyclic) thiosemicarbazones with iron, with the special aim of bridging the current knowledge on their mode of action from chemistry to (cell) biology. In addition, we highlight the difference to classical iron(III) chelators such as desferrioxamine (DFO), which are used for the treatment of iron overload., Future Directions: We want to emphasize that thiosemicarbazones are not solely removing iron from the cells/organism. In contrast, they should be considered as iron-interacting drugs influencing diverse biological pathways in a complex and multi-faceted mode of action. Consequently, in addition to the discussion of physicochemical properties (e.g., complex stability, redox activity), this review contains an overview on the diversity of cellular thiosemicarbazone targets and drug resistance mechanisms.
- Published
- 2019
- Full Text
- View/download PDF
40. Metal Drugs and the Anticancer Immune Response.
- Author
-
Englinger B, Pirker C, Heffeter P, Terenzi A, Kowol CR, Keppler BK, and Berger W
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Coordination Complexes pharmacology, Coordination Complexes therapeutic use, Gastrointestinal Microbiome drug effects, Humans, Immunity, Innate drug effects, Immunotherapy, Lymphocytes drug effects, Lymphocytes immunology, Nanoparticles chemistry, Neoplasms drug therapy, Neoplasms immunology, Antineoplastic Agents chemistry, Coordination Complexes chemistry, Metals chemistry, Neoplasms therapy
- Abstract
The immune system deploys a multitude of innate and adaptive mechanisms not only to ward off pathogens but also to prevent malignant transformation ("immune surveillance"). Hence, a clinically apparent tumor already reflects selection for those malignant cell clones capable of evading immune recognition ("immune evasion"). Metal drugs, besides their well-investigated cytotoxic anticancer effects, massively interact with the cancer-immune interface and can reverse important aspects of immune evasion. This topic has recently gained intense attention based on combination approaches with anticancer immunotherapy (e.g., immune checkpoint inhibitors), a strategy recently delivering first exciting results in clinical settings. This review summarizes the promising but still extremely fragmentary knowledge on the interplay of metal drugs with the fidelity of anticancer immune responses but also their role in adverse effects. It highlights that, at least in some cases, metal drugs can induce long-lasting anticancer immune responses. Important steps in this process comprise altered visibility and susceptibility of cancer cells toward innate and adaptive immunity, as well as direct impacts on immune cell populations and the tumor microenvironment. On the basis of the gathered information, we suggest initiating joint multidisciplinary programs to implement comprehensive immune analyses into strategies to develop novel and smart anticancer metal compounds.
- Published
- 2019
- Full Text
- View/download PDF
41. Synthesis, Characterization and in vitro Studies of a Cathepsin B-Cleavable Prodrug of the VEGFR Inhibitor Sunitinib.
- Author
-
Karnthaler-Benbakka C, Koblmüller B, Mathuber M, Holste K, Berger W, Heffeter P, Kowol CR, and Keppler BK
- Subjects
- Antineoplastic Agents chemical synthesis, Antineoplastic Agents pharmacology, Cell Line, Tumor, Drug Design, Drug Screening Assays, Antitumor, Enzyme Activation, Humans, In Vitro Techniques, Proteolysis, Angiogenesis Inhibitors chemical synthesis, Angiogenesis Inhibitors pharmacology, Cathepsin B metabolism, Prodrugs chemical synthesis, Prodrugs pharmacology, Receptors, Vascular Endothelial Growth Factor antagonists & inhibitors, Sunitinib chemical synthesis, Sunitinib pharmacology
- Abstract
Since several decades, the prodrug concept has raised considerable interest in cancer research due to its potential to overcome common problems associated with chemotherapy. However, for small-molecule tyrosine kinase inhibitors, which also cause severe side effects, hardly any strategies to generate prodrugs for therapeutic improvement have been reported so far. Here, we present the synthesis and biological investigation of a cathepsin B-cleavable prodrug of the VEGFR inhibitor sunitinib. Cell viability assays and Western blot analyses revealed, that, in contrast to the non-cathepsin B-cleavable reference compound, the prodrug shows activity comparable to the original drug sunitinib in the highly cathepsin B-expressing cell lines Caki-1 and RU-MH. Moreover, a cathepsin B cleavage assay confirmed the desired enzymatic activation of the prodrug. Together, the obtained data show that the concept of cathepsin B-cleavable prodrugs can be transferred to the class of targeted therapeutics, allowing the development of optimized tyrosine kinase inhibitors for the treatment of cancer., (© 2019 The Authors. Published by Wiley-VHCA AG, Zurich, Switzerland.)
- Published
- 2019
- Full Text
- View/download PDF
42. Synthesis and biological evaluation of biotin-conjugated anticancer thiosemicarbazones and their iron(III) and copper(II) complexes.
- Author
-
Kallus S, Uhlik L, van Schoonhoven S, Pelivan K, Berger W, Enyedy ÉA, Hofmann T, Heffeter P, Kowol CR, and Keppler BK
- Subjects
- Animals, Cell Line, Tumor, Coordination Complexes chemical synthesis, Humans, Mice, Antineoplastic Agents chemistry, Biotin chemistry, Coordination Complexes chemistry, Coordination Complexes pharmacology, Copper chemistry, Iron chemistry, Thiosemicarbazones chemistry
- Abstract
Triapine, the most prominent anticancer drug candidate from the substance class of thiosemicarbazones, was investigated in >30 clinical phase I and II studies. However, the results were rather disappointing against solid tumors, which can be explained (at least partially) due to inefficient delivery to the tumor site. Hence, we synthesized the first biotin-functionalized thiosemicarbazone derivatives in order to increase tumor specificity and accumulation. Additionally, for Triapine and one biotin conjugate the iron(III) and copper(II) complexes were prepared. Subsequently, the novel compounds were biologically evaluated on a cell line panel with different biotin uptake. The metal-free biotin-conjugated ligands showed comparable activity to the reference compound Triapine. However, astonishingly, the metal complexes of the biotinylated derivative showed strikingly decreased anticancer activity. To further analyze possible differences between the metal complexes, detailed physico- and electrochemical experiments were performed. However, neither lipophilicity or complex solution stability, nor the reduction potential or behavior in the presence of biologically relevant reducing agents showed strong variations between the biotinylated and non-biotinylated derivatives (only some differences in the reduction kinetics were observed). Nonetheless, the metal-free biotin-conjugate of Triapine revealed distinct activity in a colon cancer mouse model upon oral application comparable to Triapine. Therefore, this type of biotin-conjugated thiosemicarbazone is of interest for further synthetic strategies and biological studies., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
43. Lysosomal Sequestration Impairs the Activity of the Preclinical FGFR Inhibitor PD173074.
- Author
-
Englinger B, Kallus S, Senkiv J, Laemmerer A, Moser P, Gabler L, Groza D, Kowol CR, Heffeter P, Grusch M, and Berger W
- Abstract
Knowledge of intracellular pharmacokinetics of anticancer agents is imperative for understanding drug efficacy as well as intrinsic and acquired cellular resistance mechanisms. However, the factors driving subcellular drug distribution are complex and poorly understood. Here, we describe for the first time the intrinsic fluorescence properties of the fibroblast growth factor receptor inhibitor PD1703074 as well as utilization of this physicochemical feature to investigate intracellular accumulation and compartmentalization of this compound in human lung cancer cells. Cell-free PD173074 fluorescence, intracellular accumulation and distribution were investigated using analytical chemistry and molecular biology approaches. Analyses on a subcellular scale revealed selective drug accumulation in lysosomes. Coincubation with inhibitors of lysosomal acidification strongly enhanced PD173074-mediated fibroblast growth factor receptor (FGFR) inhibition and cytotoxicity. In conclusion, intrinsic fluorescence enables analysis of molecular factors influencing intracellular pharmacokinetics of PD173074. Lysosome-alkalinizing agents might represent candidates for rational combination treatment, preventing cancer cell-intrinsic PD173074 resistance based on lysosomal trapping.
- Published
- 2018
- Full Text
- View/download PDF
44. Critical assessment of different methods for quantitative measurement of metallodrug-protein associations.
- Author
-
Galvez L, Theiner S, Grabarics M, Kowol CR, Keppler BK, Hann S, and Koellensperger G
- Subjects
- Antineoplastic Agents analysis, Blood Proteins analysis, Chromatography, Gel methods, Chromatography, High Pressure Liquid methods, Flow Injection Analysis methods, Humans, Mass Spectrometry methods, Metals analysis, Metals metabolism, Organoplatinum Compounds analysis, Protein Binding, Ultrafiltration methods, Antineoplastic Agents metabolism, Blood Proteins metabolism, Organoplatinum Compounds metabolism
- Abstract
Quantitative screening for potential drug-protein binding is an essential step in developing novel metal-based anticancer drugs. ICP-MS approaches are at the core of this task; however, many applications lack in the capability of large-scale high-throughput screenings and proper validation. In this work, we critically discuss the analytical figures of merit and the potential method-based quantitative differences applying four different ICP-MS strategies to ex vivo drug-serum incubations. Two candidate drugs, more specifically, two Pt(IV) complexes with known differences of binding affinity towards serum proteins were selected. The study integrated centrifugal ultrafiltration followed by flow injection analysis, turbulent flow chromatography (TFC), and size exclusion chromatography (SEC), all combined with inductively coupled plasma-mass spectrometry (ICP-MS). As a novelty, for the first time, UHPLC SEC-ICP-MS was implemented to enable rapid protein separation to be performed within a few minutes at > 90% column recovery for protein adducts and small molecules. Graphical abstract Quantitative screening for potential drug-protein binding is an essential step in developingnovel metal-based anticancer drugs.
- Published
- 2018
- Full Text
- View/download PDF
45. Nanoformulations of anticancer FGFR inhibitors with improved therapeutic index.
- Author
-
Kallus S, Englinger B, Senkiv J, Laemmerer A, Heffeter P, Berger W, Kowol CR, and Keppler BK
- Subjects
- Animals, Bone Neoplasms metabolism, Bone Neoplasms pathology, Cell Proliferation drug effects, Humans, Imidazoles pharmacology, Indoles pharmacology, Liposomes chemistry, Lung Neoplasms metabolism, Lung Neoplasms pathology, Mice, Mice, Inbred BALB C, Mice, SCID, Nanoparticles chemistry, Osteosarcoma metabolism, Osteosarcoma pathology, Pyridazines pharmacology, Therapeutic Index, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Bone Neoplasms drug therapy, Enzyme Inhibitors pharmacology, Liposomes administration & dosage, Lung Neoplasms drug therapy, Nanoparticles administration & dosage, Osteosarcoma drug therapy, Receptor, Fibroblast Growth Factor, Type 1 antagonists & inhibitors
- Abstract
Fibroblast growth factor receptor (FGFR) inhibitors like ponatinib and nintedanib are clinically approved for defined cancer patient cohorts but often exert dose-limiting adverse effects. Hence, we encapsulated the FGFR inhibitors ponatinib, PD173074, and nintedanib into polylactic acid nanoparticles and liposomes to enable increased tumor accumulation/specificity and reduce side effects. Different methods of drug loading were tested and the resulting formulations compared regarding average size distribution as well as encapsulation efficiency. Appropriate encapsulation levels were achieved for liposomal preparations only. Nanoencapsulation resulted in significantly decelerated uptake kinetics in vitro with clearly decreased short-term (up to 72 h) cytotoxicity at higher concentrations. However, in long-term clonogenic assays liposomal formations were equally or even more active as compared to the free drugs. Accordingly, in an FGFR inhibitor-sensitive murine osteosarcoma transplantation model (K7M2), only liposomal but not free ponatinib resulted in significant tumor growth inhibition (by 60.4%) at markedly reduced side effects., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
46. The thiosemicarbazone Me 2 NNMe 2 induces paraptosis by disrupting the ER thiol redox homeostasis based on protein disulfide isomerase inhibition.
- Author
-
Hager S, Korbula K, Bielec B, Grusch M, Pirker C, Schosserer M, Liendl L, Lang M, Grillari J, Nowikovsky K, Pape VFS, Mohr T, Szakács G, Keppler BK, Berger W, Kowol CR, and Heffeter P
- Subjects
- Antineoplastic Agents chemical synthesis, Cell Death drug effects, Cell Line, Tumor, Cell Survival drug effects, Coordination Complexes chemistry, Coordination Complexes pharmacology, Copper chemistry, Copper metabolism, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum pathology, Endoplasmic Reticulum Stress drug effects, Epithelial Cells drug effects, Epithelial Cells metabolism, Epithelial Cells pathology, Gene Expression, HCT116 Cells, Humans, MAP Kinase Signaling System genetics, Mitochondria metabolism, Mitochondria pathology, Mitochondrial Swelling drug effects, Oxidation-Reduction drug effects, Protein Disulfide-Isomerases genetics, Protein Disulfide-Isomerases metabolism, Pyridines pharmacology, Sulfhydryl Compounds antagonists & inhibitors, Sulfhydryl Compounds metabolism, Thiosemicarbazones chemical synthesis, Antineoplastic Agents pharmacology, Endoplasmic Reticulum drug effects, MAP Kinase Signaling System drug effects, Mitochondria drug effects, Protein Disulfide-Isomerases antagonists & inhibitors, Thiosemicarbazones pharmacology
- Abstract
Due to their high biological activity, thiosemicarbazones have been developed for treatment of diverse diseases, including cancer, resulting in multiple clinical trials especially of the lead compound Triapine. During the last years, a novel subclass of anticancer thiosemicarbazones has attracted substantial interest based on their enhanced cytotoxic activity. Increasing evidence suggests that the double-dimethylated Triapine derivative Me
2 NNMe2 differs from Triapine not only in its efficacy but also in its mode of action. Here we show that Me2 NNMe2 - (but not Triapine)-treated cancer cells exhibit all hallmarks of paraptotic cell death including, besides the appearance of endoplasmic reticulum (ER)-derived vesicles, also mitochondrial swelling and caspase-independent cell death via the MAPK signaling pathway. Subsequently, we uncover that the copper complex of Me2 NNMe2 (a supposed intracellular metabolite) inhibits the ER-resident protein disulfide isomerase, resulting in a specific form of ER stress based on disruption of the Ca2+ and ER thiol redox homeostasis. Our findings indicate that compounds like Me2 NNMe2 are of interest especially for the treatment of apoptosis-resistant cancer and provide new insights into mechanisms underlying drug-induced paraptosis.- Published
- 2018
- Full Text
- View/download PDF
47. Comparative studies on the human serum albumin binding of the clinically approved EGFR inhibitors gefitinib, erlotinib, afatinib, osimertinib and the investigational inhibitor KP2187.
- Author
-
Dömötör O, Pelivan K, Borics A, Keppler BK, Kowol CR, and Enyedy ÉA
- Subjects
- Acrylamides, Afatinib, Aniline Compounds, Binding Sites, Gefitinib, Humans, Hydrogen Bonding, Molecular Docking Simulation, Protein Binding, Solvents chemistry, Spectrometry, Fluorescence methods, ErbB Receptors metabolism, Erlotinib Hydrochloride pharmacology, Piperazines pharmacology, Protein Kinase Inhibitors pharmacology, Quinazolines pharmacology, Serum Albumin, Human metabolism
- Abstract
Binding interactions between human serum albumin (HSA) and four approved epidermal growth factor receptor (EGFR) inhibitors gefitinib (GEF), erlotinib (ERL), afatinib (AFA), osimertinib (OSI), as well as the experimental drug KP2187, were investigated by means of spectrofluorometric and molecular modelling methods. Steady-state and time resolved spectrofluorometric techniques were carried out, including direct quenching of protein fluorescence and site marker displacement measurements. Proton dissociation processes and solvent dependent fluorescence properties were investigated as well. The EGFR inhibitors were predominantly presented in their single protonated form (HL
+ ) at physiological pH except ERL, which is charge-neutral. Significant solvent dependent fluorescence properties were found for GEF, ERL and KP2187, namely their emission spectra show strong dependence on the polarity and the hydrogen bonding ability of the solvents. The inhibitors proved to be bound at site I of HSA (in subdomain IIA) in a weak-to-moderate fashion (logK' 3.9-4.9) using spectrofluorometry. OSI (logK' 4.3) and KP2187 can additionally bind in site II (in subdomain IIIA), while GEF, ERL and AFA clearly show no interaction here. Docking methods qualitatively confirmed binding site preferences of compounds GEF and KP2187, and indicated that they probably bind to HSA in their neutral forms. Binding constants calculated on the basis of the various experimental data indicate a weak-to-moderate binding on HSA, only OSI exhibits somewhat higher affinity towards this protein. However, model calculations performed at physiological blood concentrations of HSA resulted in high (ca. 90%) bound fractions for the inhibitors, highlighting the importance of plasma protein binding., (Copyright © 2018 Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
48. Structure elucidation and quantification of the reduction products of anticancer Pt(iv) prodrugs by electrochemistry/mass spectrometry (EC-MS).
- Author
-
Frensemeier LM, Mayr J, Koellensperger G, Keppler BK, Kowol CR, and Karst U
- Abstract
Pt(iv) prodrugs are a class of promising anticancer agents, which are activated by reduction to the active Pt(ii) species. Consequently, the reduction process is a crucial parameter. Herein, a new approach using electrochemistry (EC) coupled to liquid chromatography (LC) and electrospray ionization-mass spectrometry (ESI-MS) or inductively coupled plasma (ICP)-MS was applied. This enabled getting insights into the differences in the reduction and ligand release of platinum(iv) complexes with varying equatorial core structures.
- Published
- 2018
- Full Text
- View/download PDF
49. Comparison of metabolic pathways of different α-N-heterocyclic thiosemicarbazones.
- Author
-
Pelivan K, Frensemeier LM, Karst U, Koellensperger G, Heffeter P, Keppler BK, and Kowol CR
- Subjects
- Animals, Humans, Hydroxylation, Kidney metabolism, Liver metabolism, Mice, Inbred BALB C, Microsomes, Liver metabolism, Oxidation-Reduction, Pyridines analysis, Pyridines blood, Pyridines urine, Thiosemicarbazones analysis, Thiosemicarbazones blood, Thiosemicarbazones urine, Metabolic Networks and Pathways, Pyridines metabolism, Thiosemicarbazones metabolism
- Abstract
Clinical failure of novel drugs is often related to their rapid metabolism and excretion. This highlights the importance of elucidation of their pharmacokinetic profile already at the preclinical stage of drug development. Triapine, the most prominent representative of α-N-heterocyclic thiosemicarbazones, was investigated in more than 30 clinical phase I/II trials, but the results against solid tumors were disappointing. Recent investigations from our group suggested that this is, at least partially, based on the fast metabolism and excretion. In order to establish more detailed structure/activity/metabolism relationships, herein a panel of 10 different Triapine derivatives was investigated for their metabolic pathways. From the biological point of view, the panel consists of terminally dimethylated thiosemicarbazones with nanomolar IC
50 values, derivatives with micromolar cytotoxicities comparable to Triapine and a completely inactive representative. To study the oxidative metabolism, a purely instrumental approach based on electrochemistry/mass spectrometry was applied and the results were compared to the data obtained from microsomal incubations. Overall, the investigated thiosemicarbazones underwent the phase I metabolic reactions dehydrogenation, hydroxylation, oxidative desulfuration (to semicarbazone and amidrazone) and demethylation. Notably, dehydrogenation resulted in a ring-closure reaction with formation of thiadiazoles. Although strong differences between the metabolic pathways of the different thiosemicarbazones were observed, they could not be directly correlated to their cytotoxicities. Finally, the metabolic pathways for the most cytotoxic compound were elucidated also in tissues collected from drug-treated mice, confirming the data obtained by electrochemical oxidation and microsomes. In addition, the in vivo experiments revealed a very fast metabolism and excretion of the compound. Graphical abstract Structure/activity/metabolisation relationships for 10 anticancer thiosemicarbazones were established using electrochemical oxidation coupled to mass spectrometry (EC-MS) and human liver microsomes analyzed by LC-MS.- Published
- 2018
- Full Text
- View/download PDF
50. Bacterial ghosts as adjuvant to oxaliplatin chemotherapy in colorectal carcinomatosis.
- Author
-
Groza D, Gehrig S, Kudela P, Holcmann M, Pirker C, Dinhof C, Schueffl HH, Sramko M, Hoebart J, Alioglu F, Grusch M, Ogris M, Lubitz W, Keppler BK, Pashkunova-Martic I, Kowol CR, Sibilia M, Berger W, and Heffeter P
- Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers and a major cause of cancer mortality worldwide. At late stage of the disease CRC often shows (multiple) metastatic lesions in the peritoneal cavity which cannot be efficiently targeted by systemic chemotherapy. This is one major factor contributing to poor prognosis. Oxaliplatin is one of the most commonly used systemic treatment options for advanced CRC. However, drug resistance - often due to insufficient drug delivery - is still hampering successful treatment. The anticancer activity of oxaliplatin includes besides DNA damage also a strong immunogenic component. Consequently, the aim of this study was to investigate the effect of bacterial ghosts (BGs) as adjuvant immunostimulant on oxaliplatin efficacy. BGs are empty envelopes of gram-negative bacteria with a distinct immune-stimulatory potential. Indeed, we were able to show that the combination of BGs with oxaliplatin treatment had strong synergistic anticancer activity against the CT26 allograft, resulting in prolonged survival and even a complete remission in this murine model of CRC carcinomatosis. This synergistic effect was based on an enhanced induction of immunogenic cell death and activation of an efficient T-cell response leading to long-term anti-tumor memory effects. Taken together, co-application of BGs strengthens the immunogenic component of the oxaliplatin anticancer response and thus represents a promising natural immune-adjuvant to chemotherapy in advanced CRC.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.