Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Angew Chem Int Ed Engl ; 63(18): e202319578, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38442302

RESUMO

The development of inert, biocompatible chelation methods is required to harness the emerging positron emitting radionuclide 45Ti for radiopharmaceutical applications. Herein, we evaluate the Ti(IV)-coordination chemistry of four catechol-based, hexacoordinate chelators using synthetic, structural, computational, and radiochemical approaches. The siderophore enterobactin (Ent) and its synthetic mimic TREN-CAM readily form mononuclear Ti(IV) species in aqueous solution at neutral pH. Radiolabeling studies reveal that Ent and TREN-CAM form mononuclear complexes with the short-lived, positron-emitting radionuclide 45Ti(IV), and do not transchelate to plasma proteins in vitro and exhibit rapid renal clearance in naïve mice. These features guide efforts to target the 45Ti isotope to prostate cancer tissue through the design, synthesis, and evaluation of Ent-DUPA, a small molecule conjugate composed of a prostate specific membrane antigen (PSMA) targeting peptide and a monofunctionalized Ent scaffold. The [45Ti][Ti(Ent-DUPA)]2- complex forms readily at room temperature. In a tumor xenograft model in mice, selective tumor tissue accumulation (8±5 %, n=5), and low off-target uptake in other organs is observed. Overall, this work demonstrates targeted imaging with 45Ti(IV), provides a foundation for advancing the application of 45Ti in nuclear medicine, and reveals that Ent can be repurposed as a 45Ti-complexing cargo for targeted nuclear imaging applications.


Assuntos
Neoplasias da Próstata , Sideróforos , Humanos , Masculino , Animais , Camundongos , Sideróforos/química , Enterobactina/metabolismo , Titânio/química , Uso Off-Label , Neoplasias da Próstata/metabolismo , Radioisótopos
2.
Acc Chem Res ; 57(6): 933-944, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38501206

RESUMO

ConspectusNuclear medicine harnesses radioisotopes for the diagnosis and treatment of disease. While the isotopes 99mTc and 111In have enabled the clinical diagnosis of millions of patients over the past 3 decades, more recent clinical translation of numerous 68Ga/177Lu-based radiopharmaceuticals for diagnostic imaging and therapy underscores the clinical utility of metal-based radiopharmaceuticals in mainstream cancer treatment. In addition to such established radionuclides, advancements in radioisotope production have enabled the production of radionuclides with a broad range of half-lives and emission properties of interest for nuclear medicine. Chemical means to form kinetically inert, in vivo-compatible species that can be modified with disease-targeting vectors is imperative. This presents a challenge for radiosiotopes of elements where the aqueous chemistry is still underdeveloped and poorly understood. Here, we discuss our efforts to date in exploring the aqueous, radioactive coordination chemistry of highly Lewis acidic metal ions and how our discoveries apply to the diagnosis and treatment of cancer in preclinical models of disease. The scope of this Account includes approaches to aqueous coordination of to-date understudied highly Lewis acidic metal ions with radioisotopes of emerging interest and the modulation of well-understood coordination environments of radio-coordination complexes to induce metal-catalyzed reactivity for separation and pro-drug applications.First, we discuss the development of seven-coordinate, small-cavity macrocyclic chelator platform mpatcn/picaga as an exemplary case study, which forms robust complexes with 44Sc/47Sc isotopes. Due to the high chemical hardness and pronounced Lewis acidity of the Sc3+ ion, the displacement of ternary ligand H2O by 18/natF- can be achieved to form an inert Sc-18/natF bond. Corresponding coordination complex natSc-18F is in vivo compatible and forms a theranostic tetrad with corresponding 44Sc/47Sc, 177Lu complexes all exhibiting homologous biodistribution profiles. Another exceptionally hard, highly Lewis acidic ion with underdeveloped aqueous chemistry and emerging interest in nuclear medicine is 45Ti4+. To develop de novo approaches to the mononuclear chelation of this ion under aqueous conditions, we employed a fragment-based bidentate ligand screening approach which identified two leads. The screen successfully predicted the formation of [45Ti][Ti(TREN-CAM)], a Ti-triscatechol complex that exhibits remarkable in vivo stability. Furthermore, the fragment-based screen also identified approaches that enabled solid-phase separation of Ti4+ and Sc3+ of interest in streamlining the isotope production of 45Ti and accessing new ways to separate 44Ti/44Sc for the development of a long-lived generator system. In addition to establishing the inert chelation of Ti4+ and Sc3+, we introduce controlled, metal-induced reactivity of corresponding coordination complexes on macroscopic and radiotracer scales. Metal-mediated autolytic amide bond cleavage (MMAAC) enables the temperature-dependent release of high-molar-activity, ready-to-inject radiopharmaceuticals; cleavage is selectively triggered by coordinated trivalent Lewis acid nat/68Ga3+ or Sc3+. Following the scope of reactivity and mechanistic studies, we validated MMAAC for the synthesis of high-molar-activity radiopharmaceuticals to image molecular targets with low expression and metal-mediated prodrug hydrolysis in vivo.This Account summarizes how developing the aqueous coordination chemistry and tuning the chemical reactivity of metal ions with high Lewis acidity at the macroscopic and tracer scales directly apply to the radiopharmaceutical synthesis with clinical potential.


Assuntos
Complexos de Coordenação , Medicina Nuclear , Humanos , Compostos Radiofarmacêuticos/química , Ácidos de Lewis , Complexos de Coordenação/química , Ligantes , Distribuição Tecidual , Radioisótopos/química , Quelantes/química , Metais , Íons
3.
J Am Chem Soc ; 145(44): 24358-24366, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37869897

RESUMO

Discrete luminescent lanthanide complexes represent a potential alternative to organic chromophores due to their tunability of optical properties, insensitivity to photobleaching, and large pseudo-Stokes shifts. Previously, we demonstrated that the lack of depth penetration of UV excitation required to sensitize discrete terbium and europium complexes can be overcome using Cherenkov radiation emitted by clinically employed radioisotopes in situ. Here, we show that the second-generation europium complexes [Eu(III)(pcta-PEPA2)] and [Eu(III)(tacn-pic-PEPA2)] (Φ = 57% and 76%, respectively) lower the limit of detection (LoD) to 1 nmol in the presence of 10 µCi of Cherenkov emitting isotopes, 18F and 68Ga. Bifunctionalization provides access to cysteine-linked peptide conjugates with comparable brightness and LoD. The conjugate, [Eu(tacn-(pic-PSMA)-PEPA2)], displays high binding affinity to prostate-specific membrane antigen (PSMA)-expressing PC-3 prostate cancer cells in vitro and can be visualized in the membrane-bound state using confocal microscopy. Biodistribution studies with the [86Y][Y(III)(tacn-(pic-PSMA)-PEPA2)] analogue in a mouse xenograft model were employed to study pharmacokinetics. Systemic administration of the targeted Cherenkov emitter, [68Ga][Ga(III)(PSMA-617)], followed by intratumoral injection or topical application of 20 or 10 nmol [Eu(III)(tacn-(pic-PSMA)-PEPA2)], respectively, in live mice resulted in statistically significant signal enhancement using conventional small animal imaging (620 nm bandpass filter). Optical imaging informed successful tumor resection. Ex vivo imaging of the fixed tumor tissue with 1 and 2 photon excitation further reveals the accumulation of the administered Eu(III) complex in target tissues. This work represents a significant step toward the application of luminescent lanthanide complexes for optical imaging in a clinical setting.


Assuntos
Elementos da Série dos Lantanídeos , Neoplasias , Masculino , Humanos , Animais , Camundongos , Európio/química , Luminescência , Distribuição Tecidual , Radioisótopos de Gálio , Elementos da Série dos Lantanídeos/química , Neoplasias/diagnóstico por imagem , Neoplasias/cirurgia , Microscopia Confocal
4.
J Am Chem Soc ; 145(29): 16261-16270, 2023 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-37434328

RESUMO

Activation of metalloprodrugs or prodrug activation using transition metal catalysts represents emerging strategies for drug development; however, they are frequently hampered by poor spatiotemporal control and limited catalytic turnover. Here, we demonstrate that metal complex-mediated, autolytic release of active metallodrugs can be successfully employed to prepare clinical grade (radio-)pharmaceuticals. Optimization of the Lewis-acidic metal ion, chelate, amino acid linker, and biological targeting vector provides means to release peptide-based (radio-)metallopharmaceuticals in solution and from the solid phase using metal-mediated, autolytic amide bond cleavage (MMAAC). Our findings indicate that coordinative polarization of an amide bond by strong, trivalent Lewis acids such as Ga3+ and Sc3+ adjacent to serine results in the N, O acyl shift and hydrolysis of the corresponding ester without dissociation of the corresponding metal complex. Compound [68Ga]Ga-10, incorporating a cleavable and noncleavable functionalization, was used to demonstrate that only the amide bond-adjacent serine effectively triggered hydrolysis in solution and from the solid phase. The corresponding solid-phase released compound [68Ga]Ga-8 demonstrated superior in vivo performance in a mouse tumor model compared to [68Ga]Ga-8 produced using conventional, solution-phase radiolabeling. A second proof-of-concept system, [67Ga]Ga-17A (serine-linked) and [67Ga]Ga-17B (glycine-linked) binding to serum albumin via the incorporated ibuprofen moiety, was also synthesized. These constructs demonstrated that complete hydrolysis of the corresponding [68Ga]Ga-NOTA complex from [67Ga]Ga-17A can be achieved in naïve mice within 12 h, as traceable in urine and blood metabolites. The glycine-linked control [68Ga]Ga-17B remained intact. Conclusively, MMAAC provides an attractive tool for selective, thermal, and metal ion-mediated control of metallodrug activation compatible with biological conditions.


Assuntos
Amidas , Complexos de Coordenação , Camundongos , Animais , Radioisótopos de Gálio/química , Preparações de Ação Retardada , Metais/química , Complexos de Coordenação/química , Catálise
5.
RSC Chem Biol ; 4(6): 414-421, 2023 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-37292057

RESUMO

Short-lived, radioactive lanthanides comprise an emerging class of radioisotopes attractive for biomedical imaging and therapy applications. To deliver such isotopes to target tissues, they must be appended to entities that target antigens overexpressed on the target cell's surface. However, the thermally sensitive nature of biomolecule-derived targeting vectors requires the incorporation of these isotopes without the use of denaturing temperatures or extreme pH conditions; chelating systems that can capture large radioisotopes under mild conditions are therefore highly desirable. Herein, we demonstrate the successful radiolabeling of the lanthanide-binding protein, lanmodulin (LanM), with medicinally relevant radioisotopes: 177Lu, 132/135La and 89Zr. Radiolabeling of the endogenous metal-binding sites of LanM, as well exogenous labeling of a protein-appended chelator, was successfully conducted at 25 °C and pH 7 with radiochemical yields ranging from 20-82%. The corresponding radiolabeled constructs possess good formulation stability in pH 7 MOPS buffer over 24 hours (>98%) in the presence of 2 equivalents of natLa carrier. In vivo experiments with [177Lu]-LanM, [132/135La]-LanM, and a prostate cancer targeting-vector linked conjugate, [132/135La]-LanM-PSMA, reveal that endogenously labeled constructs produce bone uptake in vivo. Exogenous, chelator-tag mediated radiolabeling to produce [89Zr]-DFO-LanM enables further study of the protein's in vivo behavior, demonstrating low bone and liver uptake, and renal clearance of the protein itself. While these results indicate that additional stabilization of LanM is required, this study establishes precedence for the radiochemical labeling of LanM with medically relevant lanthanide radioisotopes.

6.
Chem Sci ; 14(19): 5038-5050, 2023 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-37206398

RESUMO

The efficient, large-scale synthesis of radiometallated radiopharmaceuticals represents an emerging clinical need which, to date, is inherently limited by time consuming, sequential procedures to conduct isotope separation, radiochemical labeling and purification prior to formulation for injection into the patient. In this work, we demonstrate that a solid-phase based, concerted separation and radiosynthesis strategy followed by photochemical release of radiotracer in biocompatible solvents can be employed to prepare ready-to-inject, clinical grade radiopharmaceuticals. Optimization of resin base, resin loading, and radiochemical labeling capacity are demonstrated with 67Ga and 64Cu radioisotopes using a short model peptide sequence and further validated using two peptide-based radiopharmaceuticals with clinical relevance, targeting the gastrin-releasing peptide and the prostate specific membrane antigen. We also demonstrate that the solid-phase approach enables separation of non-radioactive carrier ions Zn2+ and Ni2+ present at 105-fold excess over 67Ga and 64Cu by taking advantage of the superior Ga3+ and Cu2+ binding affinity of the solid-phase appended, chelator-functionalized peptide. Finally, a proof of concept radiolabeling and subsequent preclinical PET-CT study with the clinically employed positron emitter 68Ga successfully exemplifies that Solid Phase Radiometallation Photorelease (SPRP) allows the streamlined preparation of radiometallated radiopharmaceuticals by concerted, selective radiometal ion capture, radiolabeling and photorelease.

7.
Eur J Inorg Chem ; 26(35)2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-38495596

RESUMO

To harness radiometals in clinical settings, a chelator forming a stable complex with the metal of interest and targets the desired pathological site is needed. Toward this goal, we previously reported a unique set of chelators that can stably bind to both large and small metal ions, via a conformational switch. Within this chelator class, py-macrodipa is particularly promising based on its ability to stably bind several medicinally valuable radiometals including large 132/135La3+, 213Bi3+, and small 44Sc3+. Here, we report a 10-step organic synthesis of its bifunctional analogue py-macrodipa-NCS, which contains an amine-reactive -NCS group that is amenable for bioconjugation reactions to targeting vectors. The hydrolytic stability of py-macordipa-NCS was assessed, revealing a half-life of 6.0 d in pH 9.0 aqueous buffer. This bifunctional chelator was then conjugated to a prostate-specific membrane antigen (PSMA)-binding moiety, yielding the bioconjugate py-macrodipa-PSMA, which was subsequently radiolabeled with large 132/135La3+ and small 47Sc3+, revealing efficient and quantitative complex formation. The resulting radiocomplexes were injected into mice bearing both PSMA-expressing and PSMA-non-expressing tumor xenografts to determine their biodistribution patterns, revealing delivery of both 132/135La3+ and 47Sc3+ to PSMA+ tumor sites. However, partial radiometal dissociation was observed, suggesting that py-macrodipa-PSMA needs further structural optimization.

8.
Chem Commun (Camb) ; 58(99): 13728-13730, 2022 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-36426996

RESUMO

The nine-coordinate aza-macrocycle DO3Apic-NO2 and its kinetically inert rare earth complexes [M(DO3A-pic-NO2)]- (M = La, Tb, Eu, Lu, Y) can be readily bioconjugated to surface accessible thioles on peptides and proteins with a minimal structural footprint. All complexes express thioconjugation rate constants in the same order of magnitude (k = 0.3 h-1) with the exception of Sc (k = 0.89 h-1). Coupling to peptides and biologics with accessible cysteines also enables post-radiochelation bioconjugation at room temperature to afford injection-ready radiopharmaceuticals as demonstrated by formation of [177Lu][Lu(DO3Apic-NO2)]- and [86Y][Y(DO3Apic-NO2)]-, followed by post-labeling conjugation to a cysteine-functionalized peptide targeting the prostate specific membrane antigen. The 86Y-labeled construct efficiently localizes in target tumors with no significant off-target accumulation as evidenced by positron emission tomography, biodistribution studies and metabolite analysis.


Assuntos
Metais Terras Raras , Compostos Radiofarmacêuticos , Masculino , Humanos , Distribuição Tecidual , Compostos Radiofarmacêuticos/química , Tomografia por Emissão de Pósitrons , Peptídeos/metabolismo
9.
Mol Pharm ; 19(9): 3217-3227, 2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-35895995

RESUMO

An immunosuppressive tumor microenvironment and tumor heterogeneity have led to the resilience of metastatic castrate resistant prostate cancer (mCRPC) to current treatments. To address these challenges, we developed and evaluated a new drug paradigm, Radio-IMmunostimulant (RIMS), in a syngeneic model of murine prostate cancer. RIMS-1 was generated using a convergent synthesis employing solid phase peptide and solution chemistries. The prostate-specific membrane antigen (PSMA) inhibitory constant for natLu-RIMS-1 was determined, and radiolabeling with 177Lu generated 177Lu-RIMS-1. The TLR 7/8 agonist payload release from natLu-RIMS-1 was determined using a cathepsin B assay. The biodistribution of 177Lu-RIMS-1 was evaluated in a bilateral xenograft model in NCru nude mice bearing PSMA(+) (PC3-PiP) and PSMA(-) (PC3-Flu) tumors at 2, 24, and 72 h. The therapeutic effect of 177Lu-RIMS-1 was evaluated in C57BL/6J mice bearing RM1-PGLS (PSMA-positive, green fluorescent protein-positive, and luciferase-positive) tumors and compared to that of 177Lu-PSMA-617 at the same total administered radioactivity of 57 MBq and molar activity of 5.18 MBq/nmol. natLu-RIMS-1 and vehicle were evaluated as the controls. Immuno-positron emission tomography (PET) using 89Zr-DFO-anti-CD3 was used to visualize T-cell distribution during treatment. 177Lu-RIMS-1 was quantitatively radiolabeled at >99% radiochemical purity and maintained a high affinity toward PSMA (Ki = 3.77 ± 0.5 nM). Cathepsin B efficiently released the entire immunostimulant payload in 17.6 h. 177Lu-RIMS-1 displayed a sustained uptake in PSMA(+) tumor tissue up to 72 h (2.65 ± 1.03% ID/g) and was not statistically different (P = 0.1936) compared to 177Lu-PSMA-617 (3.65 ± 0.59% ID/g). All animals treated with 177Lu-RIMS-1 displayed tumor growth suppression and provided a median survival of 30 days (P = 0.0007) while 177Lu-PSMA-617 provided a median survival of 15 days, which was not statistically significant (P = 0.3548) compared to the vehicle group (14 days). ImmunoPET analysis revealed 2-fold more tumor infiltrating T-cells in 177Lu-RIMS-1-treated animals compared to 177Lu-PSMA-617-treated animals; 177Lu-RIMS-1 improves therapeutic outcomes in a syngeneic model of mouse prostate cancer and elicits greater T-cell infiltration to the tumor compared to 177Lu-PSMA-617. These results support further investigation of the RIMS paradigm as the first example of a single molecular entity combining radiotherapy and immunostimulation.


Assuntos
Catepsina B , Neoplasias da Próstata , Adjuvantes Imunológicos/uso terapêutico , Animais , Antígenos de Superfície/metabolismo , Linhagem Celular Tumoral , Glutamato Carboxipeptidase II/metabolismo , Humanos , Lutécio/química , Lutécio/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Antígeno Prostático Específico , Neoplasias da Próstata/patologia , Compostos Radiofarmacêuticos/química , Linfócitos T/metabolismo , Distribuição Tecidual , Microambiente Tumoral
10.
Structure ; 29(2): 114-124.e3, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32966763

RESUMO

Bcl-2 proteins orchestrate the mitochondrial pathway of apoptosis, pivotal for cell death. Yet, the structural details of the conformational changes of pro- and antiapoptotic proteins and their interactions remain unclear. Pulse dipolar spectroscopy (double electron-electron resonance [DEER], also known as PELDOR) in combination with spin-labeled apoptotic Bcl-2 proteins unveils conformational changes and interactions of each protein player via detection of intra- and inter-protein distances. Here, we present the synthesis and characterization of pro-apoptotic BimBH3 peptides of different lengths carrying cysteines for labeling with nitroxide or gadolinium spin probes. We show by DEER that the length of the peptides modulates their homo-interactions in the absence of other Bcl-2 proteins and solve by X-ray crystallography the structure of a BimBH3 tetramer, revealing the molecular details of the inter-peptide interactions. Finally, we prove that using orthogonal labels and three-channel DEER we can disentangle the Bim-Bim, Bcl-xL-Bcl-xL, and Bim-Bcl-xL interactions in a simplified interactome.


Assuntos
Proteína 11 Semelhante a Bcl-2/química , Multimerização Proteica , Animais , Apoptose , Proteína 11 Semelhante a Bcl-2/metabolismo , Sítios de Ligação , Humanos , Camundongos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Ratos , Proteína bcl-X/química , Proteína bcl-X/metabolismo
11.
Sci Rep ; 10(1): 21652, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33303899

RESUMO

The plasma-mediated decomposition of volatile organic compounds has previously been investigated in the gas phase with metal oxides as heterogeneous catalysts. While the reactive species in plasma itself are well investigated, very little is known about the influence of metal catalysts in solution. Here, we present initial investigations on the time-dependent plasma-supported oxidation of benzyl alcohol, benzaldehyde and phenol in the presence of molecular iron complexes in solution. Products were identified by HPLC, ESI-MS, FT-IR, and [Formula: see text] spectroscopy. Compared to metal-free oxidation of the substrates, which is caused by reactive oxygen species and leads to a mixture of products, the metal-mediated reactions lead to one product cleanly, and faster than in the metal-free reactions. Most noteworthy, even catalytic amounts of metal complexes induce these clean transformations. The findings described here bear important implications for plasma-supported industrial waste transformations, as well as for plasma-mediated applications in biomedicine, given the fact that iron is the most abundant redox-active metal in the human body.

12.
Materials (Basel) ; 13(23)2020 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-33255252

RESUMO

Herein, we present dicarboxylate platinum(II) complexes of the general formula [Pt(mal)(DMSO)(L)] and [Pt(CBDC)(DMSO)(L)], where L is dbtp 5,7-ditertbutyl-1,2,4-triazolo[1,5-a]pyrimidine) or ibmtp (7-isobutyl-5-methyl-1,2,4- triazolo[1,5-a]pyrimidine), as prospective prodrugs. The platinum(II) complexes were synthesized in a one-pot reaction between cis-[PtCl2(DMSO)2], silver malonate or silver cyclobutane-1,1-dicarboxylate and triazolopyrimidines. All platinum(II) compounds were characterized by FT-IR, and 1H, 13C, 15N and 195Pt NMR; and their square planar geometries with one monodentate N(3)-bonded 5,7-disubstituted-1,2,4-triazolo[1,5-a]pyrimidine, one S-bonded molecule of dimethyl sulfoxide and one O,O-chelating malonato (1, 2) or O,O-chelating cyclobutane-1,1-dicarboxylato (3, 4) was determined. Additionally, [Pt(CBDC)(dbtp)(DMSO)] (3) exhibited (i) substantial in vitro cytotoxicity against the lung adenocarcinoma epithelial cell line (A549) (IC50 = 5.00 µM) and the cisplatin-resistant human ductal breast epithelial tumor cell line (T47D) (IC50 = 6.60 µM); and (ii) definitely exhibited low toxicity against normal murine embryonic fibroblast cells (BALB/3T3).

13.
J Inorg Biochem ; 206: 111041, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32120161

RESUMO

In this work we synthesized a chelating Schiff base by a single condensation of salicylaldehyde with 3,4-diamino benzoic acid (1). This ligand was used further for complexation to CoCl2·6H2O under nitrogen. In the next step, three six-coordinate Co(III) complexes were synthesized by coordinating this complex with imidazole (2), 2-methyimidazole (3) and N-Boc-l-histidine methyl ester (4) (Boc: tert.-butoxycarbonyl) in axial positions with simultaneous oxidation of Co(II) to Co(III) under ambient environment. All Co(III) complexes were characterized by multinuclear NMR spectroscopy (1H, 13C and 59Co NMR), FT-IR, mass spectrometry and HPLC. The Co(III) complexes were conjugated to three different cell penetrating peptides: FFFF (P1), RRRRRRRRRGAL (P2) and FFFFRRRRRRRRRGAL (P3). Standard solid-phase peptide chemistry was used for the synthesis of cell penetrating peptides. Coupling of N-terminal peptides with the cobalt complexes, possessing a carboxylic group on the tetradentate Schiff base ligand, afforded Co(III)-peptide bioconjugates, which were purified by semi-preparative HPLC and characterized by analytical HPLC and mass spectrometry. The antiproliferative activity of the synthesized compounds was studied against different human tumour cell lines: lung cancer A549, liver cancer HepG2 and normal human fibroblasts GM5657T, in comparison with the activity of cisplatin as a reference drug. The bioconjugate 21 containing the Co complex 4 and the combined phenylalanine and polyarginine cell penetrating sequence P3 shows better activity against the liver cancer line HepG2 than the parent Co(III) complex 4.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células , Peptídeos Penetradores de Células/química , Cobalto/química , Complexos de Coordenação/farmacologia , Bases de Schiff/química , Técnicas de Síntese em Fase Sólida/métodos , Células A549 , Antineoplásicos/química , Complexos de Coordenação/química , Células Hep G2 , Humanos , Ligantes , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico
14.
Mol Pharm ; 16(11): 4572-4581, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31596097

RESUMO

Cell-targeting peptides (CTPs) are increasingly used in the field of cancer research due to their high affinity and specificity to cell or tissue targets. In the search for novel metal-based drug candidates, our research group is particularly focused on bioconjugates by utilizing peptides to increase the selectivity of cytotoxic organometallic compounds. Motivated by the relatively high cytotoxic activity of gold complexes, such as Auranofin (approved to treat rheumatoid arthritis), for the treatment of various diseases, we anticipated that gold peptide bioconjugates would present interesting candidates for novel breast cancer therapies. For this, we investigate the use of the natural compound lipoic acid (Lpa) as a bioconjugation handle to link Au complexes in the oxidation state +III to peptides using the dithiol moiety. Using this strategy, we have synthesized Au(III) complex bioconjugates linked to the linear LTVSPWY peptide and two cyclic DfKRG and KTTHWGFTLG tumor-targeting peptides. Solid-phase peptide synthesis (SPPS) was used to prepare the peptides, with lipoic acid introduced N-terminally as a conjugation handle. After peptide cleavage, the metal complex was introduced in solution by first reducing the internal disulfide bond, followed by reaction with Au(ppy)Cl2 (1, ppy: 2-phenyl-pyridine), to yield the Au(III)-Lpa-peptide bioconjugates. The new bioconjugates were successfully synthesized, purified by semi-preparative HPLC, and characterized by ESI-MS. Au(III)-peptide bioconjugates were tested as cytotoxic agents against two different human breast cancer cell lines (MCF-7 and MDA-MB-231) and normal human fibroblasts cells (GM5657T) and compared to cisplatin, the parent Au(III) dichloride complex, and metal-free peptides. These in vitro data show that the Au(III)-peptide bioconjugate 5, possessing the cyclic integrin-targeting RGD-derived peptide sequence in the structure, exhibits improved activity compared to the parent gold(III) compound Au(ppy)Cl2 (1) as well as to cisplatin or the metal-free peptide. Moreover, the excellent targeting properties of 5 are supported by the fact that a Au(III)-peptide conjugate with the exact same peptide sequence, but a linear rather than the cyclic form of 5 exhibits 10 times lower cytotoxic activity.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Cisplatino/química , Cisplatino/farmacologia , Ouro/química , Compostos Organometálicos/química , Peptídeos Cíclicos/química , Ácido Tióctico/química , Antineoplásicos , Auranofina/química , Auranofina/farmacologia , Linhagem Celular Tumoral , Complexos de Coordenação/química , Feminino , Fibroblastos/efeitos dos fármacos , Humanos , Células MCF-7 , Oligopeptídeos/química
15.
ACS Chem Neurosci ; 10(8): 3555-3564, 2019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31141342

RESUMO

Extensive neuronal cell death is among the pathological hallmarks of Alzheimer's disease. While neuron death is coincident with formation of plaques comprising the beta-amyloid (Aß) peptide, a direct causative link between Aß (or other Alzheimer's-associated proteins) and cell toxicity is yet to be found. Here we show that BIM-BH3, the primary proapoptotic domain of BIM, a key protein in varied apoptotic cascades of which elevated levels have been found in brain cells of patients afflicted with Alzheimer's disease, interacts with the 42-residue amyloid isoform Aß42. Remarkably, BIM-BH3 modulated the structure, fibrillation pathway, aggregate morphology, and membrane interactions of Aß42. In particular, BIM-BH3 inhibited Aß42 fibril-formation, while it simultaneously enhanced protofibril assembly. Furthermore, we discovered that BIM-BH3/Aß42 interactions induced cell death in a human neuroblastoma cell model. Overall, our data provide a crucial mechanistic link accounting for neuronal cell death in Alzheimer's disease patients and the participation of both BIM and Aß42 in the neurotoxicity process.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Proteína 11 Semelhante a Bcl-2/metabolismo , Morte Celular/fisiologia , Neurônios/metabolismo , Doença de Alzheimer/patologia , Apoptose/fisiologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Humanos , Neurônios/patologia , Ligação Proteica , Conformação Proteica
16.
Dalton Trans ; 47(43): 15465-15476, 2018 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-30334055

RESUMO

Herein we present platinum(iv) bioconjugates with polyarginine peptides as prospective prodrug delivery systems. Asymmetrical platinum(iv) complexes 3 were obtained via oxidation of parent platinum(ii) complexes 2 with N-bromosuccinimide (NBS) in the presence of succinic anhydride. The combination of these two oxidation reagents furnishes the platinum(iv) environment with two different axial ligands, one of which bears a free carboxylic acid. All platinum(ii) and (iv) compounds were characterized by FT-IR, ESI-MS, HPLC, 1H-, 13C- and 195Pt-NMR. Standard solid-phase peptide chemistry was used for the synthesis of polyarginine (R9) peptides. Coupling of the platinum complexes with peptides N-terminally afforded peptide monoconjugates, which were purified by semi-preparative HPLC and characterized by analytical HPLC and ESI-MS. Platinum(iv)-peptide bioconjugates as well as platinum(ii) and platinum(iv) complexes were tested as cytotoxic agents against two different human cancer cell lines (MCF-7, HepG2) and normal human fibroblasts cell lines (GM5657T). Preliminary in vitro data showed that all platinum(iv) complexes exhibit lower activity than their platinum(ii) precursors towards most cell lines. Interestingly, in the case of HepG2 cells, the Pt(iv)-(R)9-G-A-L bioconjugate (4a) showed even higher activity compared to the non-targeting platinum(iv) parent compound.


Assuntos
Antineoplásicos/química , Antineoplásicos/síntese química , Ácidos Carboxílicos/química , Oligopeptídeos/química , Compostos Organoplatínicos/química , Compostos Organoplatínicos/síntese química , Sequência de Aminoácidos , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Técnicas de Química Sintética , Humanos , Compostos Organoplatínicos/farmacologia
17.
Sci Rep ; 7(1): 13828, 2017 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-29062059

RESUMO

Cold atmospheric pressure plasmas are gaining increased interest in the medical sector and clinical trials to treat skin diseases are underway. Plasmas are capable of producing several reactive oxygen and nitrogen species (RONS). However, there are open questions how plasma-generated RONS interact on a molecular level in a biological environment, e.g. cells or cell components. The redox pair glutathione (GSH) and glutathione disulphide (GSSG) forms the most important redox buffer in organisms responsible for detoxification of intracellular reactive species. We apply Raman spectroscopy, mass spectrometry, and molecular dynamics simulations to identify the time-dependent chemical modifications on GSH and GSSG that are caused by dielectric barrier discharge under ambient conditions. We find GSSG, S-oxidised glutathione species, and S-nitrosoglutathione as oxidation products with the latter two being the final products, while glutathione sulphenic acid, glutathione sulphinic acid, and GSSG are rather reaction intermediates. Experiments using stabilized pH conditions revealed the same main oxidation products as were found in unbuffered solution, indicating that the dominant oxidative or nitrosative reactions are not influenced by acidic pH. For more complex systems these results indicate that too long treatment times can cause difficult-to-handle modifications to the cellular redox buffer which can impair proper cellular function.


Assuntos
Dissulfeto de Glutationa/química , Glutationa/química , Gases em Plasma/farmacologia , Glutationa/análise , Glutationa/efeitos dos fármacos , Dissulfeto de Glutationa/análise , Dissulfeto de Glutationa/efeitos dos fármacos , Humanos , Espectrometria de Massas , Oxirredução , Estresse Oxidativo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA