Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.335
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Alkaloids Chem Biol ; 92: 1-45, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39384253

RESUMO

Indolactam alkaloids are a family of aromatic toxins produced by various actinobacteria and the cyanobacterium, Moorena producens. The best characterized examples include the teleocidins, lyngbyatoxins, olivoretins, blastmycetins, and pendolmycins, which share a nine-membered lactam core, comprised from l-tryptophanol and l-valine. Contact with indolactam alkaloids has been linked to severe dermatitis (swimmers itch), while accidental ingestion may lead to illness and fatalities. Indolactam alkaloids are also potent tumor promotors, due to their activation of protein kinase C isozymes. This chapter reviews the current literature on indolactam alkaloids, from their discovery in the early 1960s up to 2024. Topics covered include the isolation, structural elucidation, biosynthesis, bioactivity, and total synthesis of the indolactam alkaloid core.


Assuntos
Alcaloides , Lactamas , Lactamas/química , Lactamas/metabolismo , Lactamas/farmacologia , Alcaloides/química , Alcaloides/biossíntese , Alcaloides/farmacologia , Alcaloides/metabolismo , Humanos , Animais , Estrutura Molecular , Cianobactérias/metabolismo , Cianobactérias/química , Actinobacteria/metabolismo , Actinobacteria/química , Alcaloides Indólicos/química , Alcaloides Indólicos/metabolismo , Alcaloides Indólicos/farmacologia
2.
J Med Chem ; 67(20): 18098-18123, 2024 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-39361251

RESUMO

The development of therapeutic resistance in the majority of patients limits the long-term benefit of ROS1 inhibitor treatment. On-target mutations of the ROS1 kinase domain confer resistance to crizotinib and lorlatinib in more than one-third of acquired resistance cases with no current effective treatment option. As an alternative to stoichiometric inhibition, proteolytic degradation of ROS1 could provide an effective tool to combat resistance generated by these mutations. Our study has identified a potent, orally active ROS1 degrader with an excellent pharmacokinetics profile. The degrader can effectively inhibit ROS1-dependent cell proliferation and tumor growth by degrading the ROS1 kinase, thereby eliminating the active phospho-ROS1. More importantly, the degradation-based therapeutic modality can overcome on-target mutation resistance to tyrosine kinase inhibitors by efficient degradation of the mutated kinase to achieve greater potency than inhibition.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Mutação , Inibidores de Proteínas Quinases , Proteínas Tirosina Quinases , Proteínas Proto-Oncogênicas , Humanos , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacocinética , Animais , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Administração Oral , Proliferação de Células/efeitos dos fármacos , Linhagem Celular Tumoral , Antineoplásicos/farmacologia , Antineoplásicos/química , Camundongos , Proteólise/efeitos dos fármacos , Lactamas/farmacologia , Lactamas/química , Crizotinibe/farmacologia , Crizotinibe/química , Descoberta de Drogas , Pirazóis/farmacologia , Pirazóis/química , Pirazóis/farmacocinética , Relação Estrutura-Atividade , Ratos , Aminopiridinas
3.
Lung Cancer ; 196: 107954, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39303401

RESUMO

OBJECTIVES: Lorlatinib, a third-generation anaplastic lymphoma kinase (ALK) inhibitor, causes distinct adverse events (AEs), including hyperlipidemia and central nervous system (CNS) disorders. Although dose modifications are recommended to manage these AEs, whether dose modifications can achieve optimal blood lorlatinib concentrations and reduce the incidence of lorlatinib-induced AEs remains unclear. Therefore, we investigated the association between lorlatinib exposure and AEs in each patient. MATERIALS AND METHODS: We retrospectively reviewed patients with advanced ALK-rearranged non-small cell lung cancer treated with lorlatinib between November 2018 and July 2022. Serum lorlatinib concentrations were assessed using high-performance liquid chromatography-tandem mass spectrometry. All AEs were evaluated using the Common Terminology Criteria for Adverse Events version 5.0. RESULTS: The median age of the 55 eligible patients was 59 years (range: 23-79 years). All patients were administered lorlatinib after first line ALK-tyrosine kinase inhibitor failure. Grade ≥ 3 AEs occurred in 25 patients (25/55, 45 %), including hyperlipidemia in 17 (17/55, 31 %), CNS disorders in 7 (7/55, 13 %), and edema in 6 (6/55, 11 %). Dose modification was required in 23 patients (23/55, 42 %). Among the 36 patients with available data on serum lorlatinib levels at day 28 (±14) and no drug dose modifications, lorlatinib serum concentrations were significantly higher in patients with grade ≥ 3 AEs than in those without AEs (median: 462 ng/mL vs. 177 ng/mL, p < 0.01). In eight patients with data on serial lorlatinib serum concentrations following dose modifications, lorlatinib serum concentrations were effectively reduced, facilitating the ongoing administration of lorlatinib. Additionally, no significant difference was observed in the landmark analysis of progression-free survival between patients with dose modification within the first 16 weeks and those without (median: 24.8 months vs. 10.1 months, p = 0.46). CONCLUSION: Dose modification of lorlatinib was associated with successful management of AEs and decreased serum concentration of lorlatinib.


Assuntos
Aminopiridinas , Carcinoma Pulmonar de Células não Pequenas , Lactamas , Neoplasias Pulmonares , Pirazóis , Humanos , Masculino , Pessoa de Meia-Idade , Feminino , Idoso , Adulto , Estudos Retrospectivos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Aminopiridinas/efeitos adversos , Aminopiridinas/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Lactamas/efeitos adversos , Adulto Jovem , Pirazóis/efeitos adversos , Pirazóis/administração & dosagem , Pirazóis/farmacocinética , Pirazóis/uso terapêutico , Lactamas Macrocíclicas/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/uso terapêutico , Quinase do Linfoma Anaplásico/antagonistas & inibidores , Quinase do Linfoma Anaplásico/genética
5.
Nat Commun ; 15(1): 7999, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39294134

RESUMO

We investigated the impact of antiviral treatment on the emergence of SARS-CoV-2 resistance during persistent infections in immunocompromised patients (n = 15). All patients received remdesivir and some also received nirmatrelvir-ritonavir (n = 3) or therapeutic monoclonal antibodies (n = 4). Sequence analysis showed that nine patients carried viruses with mutations in the nsp12 (RNA dependent RNA polymerase), while four had viruses with nsp5 (3C protease) mutations. Infectious SARS-CoV-2 with a double mutation in nsp5 (T169I) and nsp12 (V792I) was recovered from respiratory secretions 77 days after initial COVID-19 diagnosis from a patient sequentially treated with nirmatrelvir-ritonavir and remdesivir. In vitro characterization confirmed its decreased sensitivity to remdesivir and nirmatrelvir, which was overcome by combined antiviral treatment. Studies in golden Syrian hamsters demonstrated efficient transmission to contact animals. This study documents the isolation of SARS-CoV-2 carrying resistance mutations to both nirmatrelvir and remdesivir from a patient and demonstrates its transmissibility in vivo.


Assuntos
Monofosfato de Adenosina , Alanina , Antivirais , Tratamento Farmacológico da COVID-19 , COVID-19 , Farmacorresistência Viral , Hospedeiro Imunocomprometido , Mutação , Ritonavir , SARS-CoV-2 , SARS-CoV-2/genética , SARS-CoV-2/efeitos dos fármacos , Animais , Alanina/análogos & derivados , Alanina/uso terapêutico , Alanina/farmacologia , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/uso terapêutico , Monofosfato de Adenosina/farmacologia , Antivirais/uso terapêutico , Antivirais/farmacologia , Humanos , COVID-19/virologia , Feminino , Farmacorresistência Viral/genética , Masculino , Pessoa de Meia-Idade , Ritonavir/uso terapêutico , Ritonavir/farmacologia , Idoso , Mesocricetus , Adulto , Cricetinae , Leucina , Lactamas , Prolina , Nitrilas , RNA-Polimerase RNA-Dependente de Coronavírus
6.
Antimicrob Agents Chemother ; 68(10): e0056224, 2024 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-39225484

RESUMO

We have synthesized a novel and highly selective severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease peptide mimetic inhibitor mimicking the replicase 1ab recognition sequence -Val-Leu-Gln- and utilizing a cysteine selective acyloxymethyl ketone as the electrophilic warhead to target the active site Cys145. Utilizing a constrained cyclic peptide that locks the conformation between the P3 (Val) and P2 (Leu) residues, we identified a highly selective inhibitor that fills the P2 pocket occupied by the leucine residue sidechain of PF-00835231 and the dimethyl-3-azabicyclo-hexane motif in nirmatrelvir (PF-07321332). This strategy resulted in potent and highly selective Mpro inhibitors without inhibiting essential host cathepsin cysteine or serine proteases. The lead prototype compound 1 (MPro IC50 = 230 ± 18 nM) also inhibits the replication of multiple SARS-CoV-2 variants in vitro, including SARS-CoV-2 variants of concern, and can synergize at lower concentrations with the viral RNA polymerase inhibitor, remdesivir, to inhibit replication. It also reduces SARS-CoV-2 replication in SARS-CoV-2 Omicron-infected Syrian golden hamsters without obvious toxicities, demonstrating in vivo efficacy. This novel lead structure provides the basis for optimization of improved agents targeting evolving SARS-CoV-2 drug resistance that can selectively act on Mpro versus host proteases and are less likely to have off-target effects due to non-specific targeting. Developing inhibitors against the active site of the main protease (Mpro), which is highly conserved across coronaviruses, is expected to impart a higher genetic barrier to evolving SARS-CoV-2 drug resistance. Drugs that selectively inhibit the viral Mpro are less likely to have off-target effects warranting efforts to improve this therapy.


Assuntos
Monofosfato de Adenosina , Antivirais , Proteases 3C de Coronavírus , Inibidores de Proteases , SARS-CoV-2 , Replicação Viral , SARS-CoV-2/efeitos dos fármacos , Animais , Antivirais/farmacologia , Antivirais/química , Inibidores de Proteases/farmacologia , Inibidores de Proteases/química , Replicação Viral/efeitos dos fármacos , Proteases 3C de Coronavírus/antagonistas & inibidores , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacologia , Humanos , COVID-19/virologia , Desenho de Fármacos , Alanina/análogos & derivados , Alanina/farmacologia , Chlorocebus aethiops , Betacoronavirus/efeitos dos fármacos , Células Vero , Tratamento Farmacológico da COVID-19 , Cricetinae , Mesocricetus , Peptídeos Cíclicos/farmacologia , Peptídeos Cíclicos/química , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/metabolismo , Domínio Catalítico , Lactamas , Leucina , Nitrilas , Prolina
8.
Cancer Res Commun ; 4(9): 2553-2564, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39177282

RESUMO

Patients with anaplastic lymphoma kinase (ALK)-driven neuroblastoma may respond to tyrosine kinase inhibitors, but resistance to treatment occurs and methods currently used for detection of residual disease have limited sensitivity. Here, we present a national unselected cohort of five patients with relapsed or refractory ALK-driven neuroblastoma treated with lorlatinib as monotherapy and test the potential of targeted circulating tumor DNA (ctDNA) analysis as a guide for treatment decisions in these patients. We developed a sequencing panel for ultrasensitive detection of ALK mutations associated with neuroblastoma or resistance to tyrosine kinase inhibitors and used it for ctDNA analysis in 83 plasma samples collected longitudinally from the four patients who harbored somatic ALK mutations. All four patients with ALK p.R1275Q experienced major responses and were alive 35 to 61 months after starting lorlatinib. A fifth patient with ALK p.F1174L initially had a partial response but relapsed after 10 months of treatment. In all cases, ctDNA was detected at the start of lorlatinib single-agent treatment and declined gradually, correlating with clinical responses. In the two patients exhibiting relapse, ctDNA increased 9 and 3 months, respectively, before clinical detection of disease progression. In one patient harboring HRAS p.Q61L in the relapsed tumor, retrospective ctDNA analysis showed that the mutation appeared de novo after 8 months of lorlatinib treatment. We conclude that some patients with relapsed or refractory high-risk neuroblastoma show durable responses to lorlatinib as monotherapy, and targeted ctDNA analysis is effective for evaluation of treatment and early detection of relapse in ALK-driven neuroblastoma. SIGNIFICANCE: We present five patients with ALK-driven relapsed or refractory neuroblastoma treated with lorlatinib as monotherapy. All patients responded to treatment, and four of them were alive after 3 to 5 years of follow-up. We performed longitudinal ctDNA analysis with ultra-deep sequencing of the ALK tyrosine kinase domain. We conclude that ctDNA analysis may guide treatment decisions in ALK-driven neuroblastoma, also when the disease is undetectable using standard clinical methods.


Assuntos
Aminopiridinas , Quinase do Linfoma Anaplásico , DNA Tumoral Circulante , Lactamas Macrocíclicas , Lactamas , Neuroblastoma , Pirazóis , Humanos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/sangue , Neuroblastoma/genética , Quinase do Linfoma Anaplásico/genética , DNA Tumoral Circulante/sangue , DNA Tumoral Circulante/genética , Aminopiridinas/uso terapêutico , Feminino , Lactamas Macrocíclicas/uso terapêutico , Masculino , Pirazóis/uso terapêutico , Pré-Escolar , Resistencia a Medicamentos Antineoplásicos/genética , Criança , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/sangue , Mutação , Inibidores de Proteínas Quinases/uso terapêutico , Lactente , Resultado do Tratamento
9.
Phytochemistry ; 228: 114250, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39168424

RESUMO

Sixteen undescribed pyrrololactam alkaloids, including five 2-bromopyrrole-ε-lactam (1a, 1b, 4a, 4b and 5), two 3-bromopyrrole-ε-lactam (9 and 10), eight pyrrole-ε-lactam (2a-3 and 6a-8), and one pyrrole-δ-lactam alkaloids (11), along with three previously reported compounds (12-14) were isolated from the marine sponge Phakellia fusca collected in the South China Sea. The planar structures were determined by NMR and MS analyses, while the absolute configurations were clearly elucidated by comparing the experimental and calculated ECD spectra. Compounds 2a, 2b, 4a-7b, 10, 12 and 13 exhibited anti-inflammatory activity in inhibiting the production of inflammatory cytokines IL-6 in LPS-induced RAW264.7 macrophages.


Assuntos
Alcaloides , Interleucina-6 , Lactamas , Poríferos , Pirróis , Animais , Camundongos , Alcaloides/farmacologia , Alcaloides/química , Alcaloides/isolamento & purificação , Poríferos/química , Interleucina-6/antagonistas & inibidores , Interleucina-6/metabolismo , Células RAW 264.7 , Lactamas/química , Lactamas/farmacologia , Lactamas/isolamento & purificação , Pirróis/farmacologia , Pirróis/química , Pirróis/isolamento & purificação , China , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/antagonistas & inibidores , Estrutura Molecular , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/química , Anti-Inflamatórios/isolamento & purificação , Relação Estrutura-Atividade , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Relação Dose-Resposta a Droga
10.
Cancer Rep (Hoboken) ; 7(8): e2164, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39188081

RESUMO

BACKGROUND: Anaplastic lymphoma kinase (ALK) rearrangements are rare in non-myofibroblastic sarcoma and there is limited data on the efficacy of ALK tyrosine kinase inhibitors (TKIs) and mechanisms of resistance in these patients. CASE: A 58 year-old man with metastatic non-myofibroblastic sarcoma was found to have an EML4-ALK fusion on molecular sequencing. After progression on first line systemic therapy with doxorubicin, the patient received alectinib, a second generation ALK inhibitor, and had a marked clinical and radiological response. He progressed after 5 months of treatment. Repeat lung biopsy identified the emergence of an ALK I1171N resistance mutation. He was then treated with lorlatinib, again with rapid clinical improvement and significant partial radiological response. He progressed after 4 months, at which time a repeat lung biopsy identified a new ALK kinase domain mutation G1202R. The patient was subsequently treated with chemotherapy, though unfortunately died shortly after due to rapidly progressive disease. CONCLUSION: This case report adds to a body of evidence demonstrating the potential transformative response to targeted therapy in non-lung solid organ tumours harbouring ALK fusions. This is the first description tracking the development of resistance mutations in a patient with non-myofibroblastic sarcoma and questions the utility of the presence of G1202R mutation as a marker of lorlatinib sensitivity in non-lung ALK rearranged tumours, contrary to experience in lung cancer.


Assuntos
Quinase do Linfoma Anaplásico , Resistencia a Medicamentos Antineoplásicos , Lactamas Macrocíclicas , Lactamas , Mutação , Proteínas de Fusão Oncogênica , Inibidores de Proteínas Quinases , Sarcoma , Humanos , Masculino , Proteínas de Fusão Oncogênica/genética , Pessoa de Meia-Idade , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores de Proteínas Quinases/uso terapêutico , Quinase do Linfoma Anaplásico/genética , Quinase do Linfoma Anaplásico/antagonistas & inibidores , Sarcoma/genética , Sarcoma/tratamento farmacológico , Sarcoma/patologia , Lactamas Macrocíclicas/uso terapêutico , Pirazóis/uso terapêutico , Evolução Fatal , Aminopiridinas/uso terapêutico , Carbazóis/uso terapêutico , Carbazóis/administração & dosagem , Piperidinas/uso terapêutico
11.
Anticancer Drugs ; 35(9): 867-871, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39008537

RESUMO

Metastatic spread to the central nervous system (CNS) is frequent in anaplastic lymphoma kinase ( ALK )-rearranged non-small cell lung cancer (NSCLC) and has an important impact on patient prognosis and quality of life. Leptomeningeal involvement may occur in up to 10% of cases of ALK-positive NSCLC. Lorlatinib is a third-generation ALK inhibitor that has excellent CNS penetrability and demonstrated its efficacy both in pretreated and treatment-naive patients. Herein, we present the case of a 34-year-old patient diagnosed with stage IV ALK-rearranged NSCLC who received two lines of ALK inhibitors (crizotinib followed by alectinib) and several courses of brain stereotactic ablative radiotherapy until leptomeningeal involvement was detected. Third-line lorlatinib was then administered, and 2 months later encephalic MRI documented complete regression of the leptomeningeal involvement that is still maintained after 36 months while treatment with lorlatinib is still ongoing with good tolerability. To the best of our knowledge, this is the longer intracranial response reported in the literature, underlining the importance of the most appropriate choice of systemic treatments and their integration with loco-regional approaches to improve outcomes.


Assuntos
Aminopiridinas , Carcinoma Pulmonar de Células não Pequenas , Lactamas , Neoplasias Pulmonares , Pirazóis , Humanos , Lactamas/uso terapêutico , Adulto , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Aminopiridinas/uso terapêutico , Aminopiridinas/administração & dosagem , Pirazóis/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Lactamas Macrocíclicas/uso terapêutico , Masculino , Neoplasias Meníngeas/tratamento farmacológico , Neoplasias Meníngeas/patologia , Inibidores de Proteínas Quinases/uso terapêutico , Quinase do Linfoma Anaplásico/antagonistas & inibidores , Quinase do Linfoma Anaplásico/genética
12.
Antiviral Res ; 230: 105970, 2024 10.
Artigo em Inglês | MEDLINE | ID: mdl-39067667

RESUMO

Variants of SARS-CoV-2 pose significant challenges in public health due to their increased transmissibility and ability to evade natural immunity, vaccine protection, and monoclonal antibody therapeutics. The emergence of the highly transmissible Omicron variant and subsequent subvariants, characterized by an extensive array of over 32 mutations within the spike protein, intensifies concerns regarding vaccine evasion. In response, multiple antiviral therapeutics have received FDA emergency use approval, targeting the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp) and main protease (Mpro) regions, known to have relatively fewer mutations across novel variants. In this study, we evaluated the efficacy of nirmatrelvir (PF-07321332) and other clinically significant SARS-CoV-2 antivirals against a diverse panel of SARS-CoV-2 variants, encompassing the newly identified Omicron subvariants XBB1.5 and JN.1, using live-virus antiviral assays. Our findings demonstrate that while the last Omicron subvariants exhibited heightened pathogenicity in our animal model, nirmatrelvir and other clinically relevant antivirals consistently maintained their efficacy against all tested variants, including the XBB1.5 subvariant.


Assuntos
Antivirais , Tratamento Farmacológico da COVID-19 , Hidroxilaminas , SARS-CoV-2 , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/genética , Antivirais/farmacologia , Antivirais/uso terapêutico , Animais , Hidroxilaminas/farmacologia , Hidroxilaminas/uso terapêutico , Camundongos , Humanos , Células Vero , Chlorocebus aethiops , COVID-19/virologia , Citidina/análogos & derivados , Citidina/farmacologia , Citidina/uso terapêutico , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacologia , Monofosfato de Adenosina/uso terapêutico , Glicoproteína da Espícula de Coronavírus/genética , Mutação , Alanina/farmacologia , Alanina/análogos & derivados , Lactamas , Leucina , Nitrilas , Prolina
13.
Nat Commun ; 15(1): 5779, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38987535

RESUMO

To the best of our knowledge, enzymes that catalyse intramolecular Diels-Alder ([4+2] cycloaddition) reactions are frequently reported in natural product biosynthesis; however, no native enzymes utilising Lewis acid catalysis have been reported. Verticilactam is a representative member of polycyclic macrolactams, presumably produced by spontaneous cycloaddition. We report that the intramolecular [4+2] cycloadditions can be significantly accelerated by ferredoxins (Fds), a class of small iron-sulphur (Fe-S) proteins. Through iron atom substitution by Lewis acidic gallium (Ga) iron and computational calculations, we confirm that the ubiquitous Fe-S cluster efficiently functions as Lewis acid to accelerate the tandem [4+2] cycloaddition and Michael addition reactions by lowering free energy barriers. Our work highlights Nature's ingenious strategy to generate complex molecule structures using the ubiquitous Fe-S protein. Furthermore, our study sheds light on the future design of Fd as a versatile Lewis acid catalyst for [4+2] cycloaddition reactions.


Assuntos
Produtos Biológicos , Reação de Cicloadição , Proteínas Ferro-Enxofre , Ácidos de Lewis , Produtos Biológicos/metabolismo , Produtos Biológicos/química , Proteínas Ferro-Enxofre/metabolismo , Proteínas Ferro-Enxofre/química , Ácidos de Lewis/química , Ácidos de Lewis/metabolismo , Catálise , Ferro/química , Ferro/metabolismo , Lactamas/metabolismo , Lactamas/química , Biocatálise
14.
Lancet Infect Dis ; 24(11): 1213-1224, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39025098

RESUMO

BACKGROUND: Remdesivir (Veklury, Gilead Sciences, Foster City, CA, USA) and nirmatrelvir-ritonavir (Paxlovid, Pfizer, New York, NY, USA) were reported to improve the outcome of patients with mild-to-moderate COVID-19 symptoms. Preclinical data suggest that nirmatrelvir-ritonavir might be more effective than remdesivir alone or in combination with nirmatrelvir-ritonavir for people at high risk of severe COVID-19. We aimed to assess the safety and effectiveness of combining remdesivir and nirmatrelvir-ritonavir compared with using each drug alone for adults hospitalised with COVID-19. METHODS: In this target trial emulation study, we used electronic health records of patients aged 18 years or older who received either combination treatment of nirmatrelvir-ritonavir and remdesivir or monotherapy of either drug between March 16 and Dec 31, 2022, within 5 days of hospitalisation for COVID-19 in Hong Kong. Inverse probability of treatment weighting was applied to balance baseline patient characteristics across the treatment groups. The primary outcome was all-cause mortality. Cox proportional hazards regression adjusting weighting was used to compare the risk of all-cause mortality, intensive care unit (ICU) admission, or ventilatory support for 90 days of follow-up between groups. FINDINGS: Between March 16 and Dec 31, 2022, 18 196 participants were identified from electronic health records and assigned to receive remdesivir (n=4232), nirmatrelvir-ritonavir (n=13 656), or nirmatrelvir-ritonavir and remdesivir (n=308). By applying an inverse probability of treatment weighting, a weighted sample composed of 18 410 recipients of nirmatrelvir-ritonavir and remdesivir combination treatment, 18 178 recipients of remdesivir monotherapy, and 18 287 recipients of nirmatrelvir-ritonavir monotherapy was obtained. After a median follow-up of 84 days (IQR 45-90), risk of mortality was lower in patients who received nirmatrelvir-ritonavir monotherapy (hazard ratio [HR] 0·18 [95% CI 0·15 to 0·20]; absolute risk reduction [ARR] -16·33% [95% CI -16·98 to -15·68]) or remdesivir and nirmatrelvir-ritonavir combination therapy (HR 0·66 [95% CI 0·49 to 0·89]; ARR -6·52% [95% CI -7·29 to -5·74]) than in patients who received remdesivir monotherapy. Similar results were observed for ICU admission or ventilatory support (nirmatrelvir-ritonavir monotherapy: HR 0·09 [95% CI 0·07 to 0·11]; ARR -10·04% [95% CI -10·53 to -9·56]; combination therapy: HR 0·68 [95% CI 0·42 to 1·12]; ARR -3·24% [95% CI -3·84 to -2·64]). Compared with combination therapy, nirmatrelvir-ritonavir monotherapy was associated with lower risk of mortality (HR 0·27 [95% CI 0·20 to 0·37]; ARR -9·81% [95% CI -10·39 to -9·24]) and ICU admission or ventilatory support (HR 0·13 [95% CI 0·08 to 0·22]; ARR -6·80% [95% CI -7·22 to -6·39]). INTERPRETATION: Our study highlighted the potential for reduced risk of mortality, ICU admission, or the need for ventilatory support in patients hospitalised with COVID-19 treated with nirmatrelvir-ritonavir as a monotherapy compared with treatment regimens based on nirmatrelvir-ritonavir and remdesivir combination therapy or remdesivir monotherapy. Further randomised controlled trials are needed to support the validity of the current results. FUNDING: The Health and Medical Research Fund Commissioned Research on COVID-19. TRANSLATION: For the Chinese translation of the abstract see Supplementary Materials section.


Assuntos
Monofosfato de Adenosina , Alanina , Antivirais , Tratamento Farmacológico da COVID-19 , Quimioterapia Combinada , Hospitalização , Ritonavir , SARS-CoV-2 , Humanos , Alanina/análogos & derivados , Alanina/uso terapêutico , Ritonavir/uso terapêutico , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/uso terapêutico , Monofosfato de Adenosina/administração & dosagem , Masculino , Feminino , Pessoa de Meia-Idade , Antivirais/uso terapêutico , Antivirais/administração & dosagem , Idoso , Hospitalização/estatística & dados numéricos , Resultado do Tratamento , Adulto , Leucina/análogos & derivados , Leucina/uso terapêutico , COVID-19/mortalidade , COVID-19/virologia , Indóis/uso terapêutico , Indóis/administração & dosagem , Lactamas , Nitrilas , Prolina
15.
Cells ; 13(14)2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-39056757

RESUMO

Lorlatinib is a pharmaceutical ALK kinase inhibitor used to treat ALK driven non-small cell lung cancers. This paper analyses the intersection of past published data on the physiological consequences of two unrelated drugs from general medical practice-itraconazole and cilostazol-with the pathophysiology of ALK positive non-small cell lung cancer. A conclusion from that data analysis is that adding itraconazole and cilostazol may make lorlatinib more effective. Itraconazole, although marketed worldwide as a generic antifungal drug, also inhibits Hedgehog signaling, Wnt signaling, hepatic CYP3A4, and the p-gp efflux pump. Cilostazol, marketed worldwide as a generic thrombosis preventative drug, acts by inhibiting phosphodiesterase 3, and, by so doing, lowers platelets' adhesion, thereby partially depriving malignant cells of the many tumor trophic growth factors supplied by platelets. Itraconazole may enhance lorlatinib effectiveness by (i) reducing or stopping a Hedgehog-ALK amplifying feedback loop, by (ii) increasing lorlatinib's brain levels by p-gp inhibition, and by (iii) inhibiting growth drive from Wnt signaling. Cilostazol, surprisingly, carries minimal bleeding risk, lower than that of aspirin. Risk/benefit assessment of the combination of metastatic ALK positive lung cancer being a low-survival disease with the predicted safety of itraconazole-cilostazol augmentation of lorlatinib favors a trial of this drug trio in ALK positive lung cancer.


Assuntos
Aminopiridinas , Cilostazol , Resistencia a Medicamentos Antineoplásicos , Itraconazol , Humanos , Itraconazol/farmacologia , Itraconazol/uso terapêutico , Cilostazol/farmacologia , Cilostazol/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Aminopiridinas/farmacologia , Aminopiridinas/uso terapêutico , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Reposicionamento de Medicamentos , Lactamas/farmacologia , Lactamas/uso terapêutico , Quinase do Linfoma Anaplásico/metabolismo , Quinase do Linfoma Anaplásico/antagonistas & inibidores , Quinase do Linfoma Anaplásico/genética , Inibidores de Proteínas Quinases/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia
16.
Eur J Med Chem ; 276: 116669, 2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-39053189

RESUMO

The present study describes a small library of peptides derived from a potent and selective CXCR4 antagonist (3), wherein the native disulfide bond is replaced using a side-chain to tail macrolactamization technique to vary ring size and amino acid composition. The peptides were preliminary assessed for their ability to interfere with the interaction between the receptor and anti-CXCR4 PE-conjugated antibody clone 12G5. Two promising candidates (13 and 17) were identified and further evaluated in a125I-CXCL12 competition binding assay, exhibiting IC50 in the low-nanomolar range. Furthermore, both candidates displayed high selectivity towards CXCR4 with respect to the cognate receptor CXCR7, ability to block CXCL12-dependent cancer cell migration, and receptor internalization, albeit at a higher concentration compared to 3. Molecular modeling studies on 13 and 17 produced a theoretical model that may serve as a guide for future modifications, aiding in the development of analogs with improved affinity. Finally, the study provides valuable insights into developing therapeutic agents targeting CXCR4-mediated processes, demonstrating the adaptability of our lead peptide 3 to alternative cyclization approaches and offering prospects for comprehensive investigations into the receptor region's interaction with its C-terminal region.


Assuntos
Dissulfetos , Peptídeos , Receptores CXCR4 , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/metabolismo , Humanos , Sítios de Ligação/efeitos dos fármacos , Peptídeos/química , Peptídeos/farmacologia , Peptídeos/síntese química , Dissulfetos/química , Dissulfetos/farmacologia , Relação Estrutura-Atividade , Estrutura Molecular , Relação Dose-Resposta a Droga , Lactamas/química , Lactamas/farmacologia , Lactamas/síntese química , Movimento Celular/efeitos dos fármacos , Modelos Moleculares , Linhagem Celular Tumoral
17.
J Agric Food Chem ; 72(31): 17431-17443, 2024 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-39021257

RESUMO

The fungus Talaromyces hainanensis, isolated from the mangrove soil, was characterized as a novel species by morphology observation and phylogenetic analyses. Four new γ-lactam alkaloids talaroilactams A-D (1-4) and two reported compounds harzianic acid (5) and isoharzianic acid (6) were identified from the fungus T. hainanensis WHUF0341, assisted by OSMAC along with molecular networking approaches. Their structures were determined through ECD calculations and spectroscopic analyses. Moreover, the biosynthetic route of 1-4 was also proposed. Compound 1 displayed potent cytotoxicity against HepG2 cell lines, with an IC50 value of 10.75 ± 1.11 µM. In addition, network pharmacology was employed to dissect the probable mechanisms contributing to the antihepatocellular carcinoma effects of compound 1, revealing that cytotoxicity was mainly associated with proteolysis, negative regulation of autophagy, inflammatory response, and the renin-angiotensin system. These results not only expanded the chemical space of natural products from the mangrove associated fungi but also afforded promising lead compounds for developing the antihepatocellular carcinoma agents.


Assuntos
Alcaloides , Antineoplásicos , Lactamas , Talaromyces , Talaromyces/química , Talaromyces/metabolismo , Humanos , Alcaloides/farmacologia , Alcaloides/química , Células Hep G2 , Antineoplásicos/farmacologia , Antineoplásicos/química , Lactamas/química , Lactamas/farmacologia , Filogenia , Estrutura Molecular , Áreas Alagadas , Rhizophoraceae/microbiologia , Rhizophoraceae/química
18.
Clin Drug Investig ; 44(8): 553-576, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39085682

RESUMO

The use of anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKIs), such as lorlatinib, for the treatment of patients with ALK gene rearrangement (or ALK-positive) non-small cell lung cancer (NSCLC) has been shown to improve the overall survival and quality of life of these patients. However, lorlatinib is not exempt from potential adverse events. Adequate monitoring and management of these adverse events are critical for increasing patient adherence to lorlatinib, thereby maximizing the benefits of treatment and minimizing the risks associated with treatment discontinuation. Considering that the adverse events of lorlatinib can affect different organs and systems, the participation of a multidisciplinary team, including cardiologists, neurologists, internal medicine specialists, and oncology pharmacists, is needed. This article presents specific and pragmatic strategies for identifying and treating the most relevant adverse events associated with lorlatinib in patients with advanced ALK-positive NSCLC based on the clinical experience of a multidisciplinary panel of experts.


Assuntos
Aminopiridinas , Quinase do Linfoma Anaplásico , Carcinoma Pulmonar de Células não Pequenas , Lactamas Macrocíclicas , Lactamas , Neoplasias Pulmonares , Pirazóis , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Quinase do Linfoma Anaplásico/antagonistas & inibidores , Quinase do Linfoma Anaplásico/genética , Aminopiridinas/efeitos adversos , Pirazóis/efeitos adversos , Pirazóis/uso terapêutico , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/uso terapêutico , Consenso , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico
19.
Thorac Cancer ; 15(23): 1772-1775, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38924375

RESUMO

We present the case of a 34-year-old Japanese man with anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer and brain metastases. After central nervous system (CNS) disease progression with alecintib and brigatinib, treatment with lorlatinib resulted in a good intracranial response. In this case, we investigated brain penetration ratio of brigatinib using cerebrospinal fluid and paired serum samples, and the ratio was 0.012. Further, we investigated resistance mechanisms via next-generation sequencing (NGS) using lung biopsy at lung cancer diagnosis and brain biopsy sample at progressive disease of brigatinib. No apparent resistance mechanism of known ALK resistance, such as ALK mutations, amplifications, epithelial-mesenchymal transition (EMT) and bypass pathway activation were detected. Taken together, we speculate that the low CNS penetration rate of brigatinib confers CNS progression. Further studies are warranted to reveal the resistance mechanism and propose a treatment strategy for CNS progression in ALK-positive patients.


Assuntos
Adenocarcinoma de Pulmão , Aminopiridinas , Quinase do Linfoma Anaplásico , Carbazóis , Resistencia a Medicamentos Antineoplásicos , Lactamas Macrocíclicas , Lactamas , Neoplasias Pulmonares , Compostos Organofosforados , Piperidinas , Pirazóis , Pirimidinas , Humanos , Masculino , Compostos Organofosforados/uso terapêutico , Compostos Organofosforados/farmacologia , Compostos Organofosforados/administração & dosagem , Lactamas/uso terapêutico , Adulto , Aminopiridinas/uso terapêutico , Aminopiridinas/farmacologia , Quinase do Linfoma Anaplásico/genética , Pirimidinas/uso terapêutico , Pirimidinas/farmacologia , Pirimidinas/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Lactamas Macrocíclicas/uso terapêutico , Lactamas Macrocíclicas/farmacologia , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/patologia , Adenocarcinoma de Pulmão/genética , Pirazóis/uso terapêutico , Pirazóis/farmacologia , Carbazóis/uso terapêutico , Carbazóis/farmacologia , Carbazóis/administração & dosagem , Piperidinas/uso terapêutico , Piperidinas/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/secundário
20.
Cell Death Differ ; 31(7): 910-923, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38858548

RESUMO

Solute Carrier Family 3, Member 2 (SLC3A2 or 4F2hc) is a multifunctional glycoprotein that mediates integrin-dependent signaling, acts as a trafficking chaperone for amino acid transporters, and is involved in polyamine transportation. We identified SLC3A2 as a potential Anaplastic Lymphoma Kinase (ALK) interacting partner in a BioID-proximity labeling screen in neuroblastoma (NB) cells. In this work we show that endogenous SLC3A2 and ALK interact in NB cells and that this SLC3A2:ALK interaction was abrogated upon treatment with the ALK inhibitor lorlatinib. We show here that loss of ALK activity leads to decreased SLC3A2 expression and reduced SLC3A2 protein stability in a panel of NB cell lines, while stimulation of ALK with ALKAL2 ligand resulted in increased SLC3A2 protein levels. We further identified MARCH11, an E3 ligase, as a regulator of SLC3A2 ubiquitination downstream of ALK. Further, knockdown of SLC3A2 resulted in inhibition of NB cell growth. To investigate the therapeutic potential of SLC3A2 targeting, we performed monotreatment of NB cells with AMXT-1501 (a polyamine transport inhibitor), which showed only moderate effects in NB cells. In contrast, a combination lorlatinib/AMXT-1501 treatment resulted in synergistic inhibition of cell growth in ALK-driven NB cell lines. Taken together, our results identify a novel role for the ALK receptor tyrosine kinase (RTK), working in concert with the MARCH11 E3 ligase, in regulating SLC3A2 protein stability and function in NB cells. The synergistic effect of combined ALK and polyamine transport inhibition shows that ALK/MARCH11/SLC3A2 regulation of amino acid transport is important for oncogenic growth and survival in NB cells.


Assuntos
Quinase do Linfoma Anaplásico , Proliferação de Células , Cadeia Pesada da Proteína-1 Reguladora de Fusão , Neuroblastoma , Transdução de Sinais , Humanos , Quinase do Linfoma Anaplásico/metabolismo , Quinase do Linfoma Anaplásico/antagonistas & inibidores , Quinase do Linfoma Anaplásico/genética , Neuroblastoma/patologia , Neuroblastoma/metabolismo , Cadeia Pesada da Proteína-1 Reguladora de Fusão/metabolismo , Cadeia Pesada da Proteína-1 Reguladora de Fusão/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Lactamas/farmacologia , Aminopiridinas/farmacologia , Ubiquitinação/efeitos dos fármacos , Pirazóis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA