Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.258
Filter
1.
J Control Release ; 369: 444-457, 2024 May.
Article in English | MEDLINE | ID: mdl-38575076

ABSTRACT

Efficient intratumoral penetration is essential for nanomedicine to eradicate pancreatic tumors. Although nanomedicine can enter the perivascular space of pancreatic tumors, their access to distal tumor cells, aloof from the vessels, remains a formidable challenge. Here, we synthesized an acid-activatable macromolecular prodrug of floxuridine (FUDR)-poly(FUDR-ketal), engineered a micellar nanomedicine of FUDR, and intravenously co-administered the nanomedicine with the tumor-penetrating peptide iRGD for enhanced treatment of pancreatic tumor. A FUDR-derived mono-isopropenyl ether was synthesized and underwent self-addition polymerization to afford the hydrophobic poly(FUDR-ketal), which was subsequently co-assembled with amphiphilic DSPE-mPEG into the micellar nanomedicine with size of 12 nm and drug content of 56.8 wt% using nanoprecipitation technique. The acetone-based ketal-linked poly(FUDR-ketal) was triggered by acid to release FUDR to inhibit cell proliferation. In an orthotopic pancreatic tumor model derived from KPC (KrasLSL-G12D/+; Trp53LSL-R172H/+; Pdx1-Cre) cells that overexpress neuropilin-1 (NRP-1) receptor, iRGD improved penetration of FUDR nanomedicine into tumor parenchyma and potentiated the therapeutic efficacy. Our nanoplatform, along with iRGD, thus appears to be promising for efficient penetration and activation of acid-responsive nanomedicines for enhanced pancreatic cancer therapy.


Subject(s)
Floxuridine , Nanomedicine , Oligopeptides , Pancreatic Neoplasms , Prodrugs , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Animals , Nanomedicine/methods , Cell Line, Tumor , Humans , Floxuridine/administration & dosage , Floxuridine/pharmacokinetics , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Oligopeptides/chemistry , Oligopeptides/administration & dosage , Polyethylene Glycols/chemistry , Polyethylene Glycols/administration & dosage , Micelles , Mice, Nude , Mice, Inbred BALB C , Mice , Female
2.
J Control Release ; 369: 630-641, 2024 May.
Article in English | MEDLINE | ID: mdl-38599548

ABSTRACT

Successful treatment of ulcerative colitis (UC) is highly dependent on several parameters, including dosing regimen and the ability to deliver drugs to the disease site. In this study two strategies for delivering mesalazine (5-aminosalicylic acid, 5-ASA) to the colon were compared in an advanced in vitro model of the human gastrointestinal (GI) tract, the SHIME® system. Herein, a prodrug strategy employing bacteria-mediated drug release (sulfasalazine, Azulfidine®) was evaluated alongside a formulation strategy that utilised pH and bacteria-mediated release (5-ASA, Octasa® 1600 mg). SHIME® experiments were performed simulating both the GI physiology and colonic microbiota under healthy and inflammatory bowel disease (IBD) conditions, to study the impact of the disease state and ileal pH variability on colonic 5-ASA delivery. In addition, the effects of the products on the colonic microbiome were investigated by monitoring bacterial growth and metabolites. Results demonstrated that both the prodrug and formulation approaches resulted in a similar percentage of 5-ASA recovery under healthy conditions. On the contrary, during experiments simulating the GI physiology and microbiome of IBD patients (the target population) the formulation strategy resulted in a higher proportion of 5-ASA delivery to the colonic region as compared to the prodrug approach (P < 0.0001). Interestingly, the two products had distinct effects on the synthesis of key bacterial metabolites, such as lactate and short chain fatty acids, which varied according to disease state and ileal pH variability. Further, both 5-ASA and sulfasalazine significantly reduced the growth of the faecal microbiota sourced from six healthy humans. The findings support that the approach selected for colonic drug delivery could significantly influence the effectiveness of UC treatment, and highlight that drugs licensed for UC may differentially impact the growth and functioning of the colonic microbiota.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Colon , Gastrointestinal Microbiome , Mesalamine , Sulfasalazine , Mesalamine/administration & dosage , Mesalamine/pharmacology , Humans , Colon/microbiology , Colon/metabolism , Colon/drug effects , Gastrointestinal Microbiome/drug effects , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Sulfasalazine/administration & dosage , Prodrugs/administration & dosage , Drug Delivery Systems , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/microbiology , Hydrogen-Ion Concentration , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/microbiology , Drug Liberation
3.
J Control Release ; 369: 622-629, 2024 May.
Article in English | MEDLINE | ID: mdl-38604383

ABSTRACT

Enhancing the delivery and release efficiency of hydroxyl agents, constrained by high pKa values and issues of release rate or unstable linkage, is a critical challenge. To address this, a self-immolative linker, composed of a modifiable p-hydroxybenzyl ether and a fast cyclization adapter (N-(ortho-hydroxyphenyl)-N-methylcarbamate) was strategically designed, for the synthesis of prodrugs. The innovative linker not only provides a side chain modification but also facilitates the rapid release of the active payloads, thereby enabling precise drug delivery. Particularly, five prodrug model compounds (J1, J2, J3, J5 and J6) were synthesized to evaluate the release rates by using ß-glucuronic acid as trigger and five hydroxyl compounds as model payloads. Significantly, all prodrug model compounds could efficiently release the hydroxyl payloads under the action of ß-glucuronidase, validating the robustness of the linker. And then, to assess the drug delivery and release efficiency using endogenous albumin as a transport vehicle, J1148, a SN38 prodrug modified with maleimide side chain was synthesized. Results demonstrated that J1148 covalently bound to plasma albumin through in situ Michael addition, effectively targeting the tumor microenvironment. Activated by ß-glucuronidase, J1148 underwent a classical 1, 6-elimination, followed by rapid cyclization of the adapter, thereby releasing SN38. Impressively, J1148 showed excellent therapeutic efficacy against human colonic cancer xenograft model, leading to a significant reduction or even disappearance of tumors (3/6 of mice cured). These findings underscore the potential of the designed linker in the delivery system of hydroxyl agents, positioning it at the forefront of advancements in drug delivery technology.


Subject(s)
Drug Delivery Systems , Irinotecan , Prodrugs , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Animals , Humans , Irinotecan/administration & dosage , Irinotecan/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Camptothecin/chemistry , Drug Liberation , Mice, Nude , Cell Line, Tumor , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Female , Mice , Albumins/administration & dosage , Albumins/chemistry , Glucuronidase/metabolism , Mice, Inbred BALB C
4.
Science ; 383(6688): eadk6176, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38484056

ABSTRACT

Obeldesivir (ODV, GS-5245) is an orally administered prodrug of the parent nucleoside of remdesivir (RDV) and is presently in phase 3 trials for COVID-19 treatment. In this work, we show that ODV and its circulating parent nucleoside metabolite, GS-441524, have similar in vitro antiviral activity against filoviruses, including Marburg virus, Ebola virus, and Sudan virus (SUDV). We also report that once-daily oral ODV treatment of cynomolgus monkeys for 10 days beginning 24 hours after SUDV exposure confers 100% protection against lethal infection. Transcriptomics data show that ODV treatment delayed the onset of inflammation and correlated with antigen presentation and lymphocyte activation. Our results offer promise for the further development of ODV to control outbreaks of filovirus disease more rapidly.


Subject(s)
Alanine , Antiviral Agents , Ebolavirus , Hemorrhagic Fever, Ebola , Nucleosides , Prodrugs , Animals , Administration, Oral , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/prevention & control , Macaca fascicularis , Nucleosides/administration & dosage , Nucleosides/pharmacology , Adenosine Monophosphate/administration & dosage , Adenosine Monophosphate/pharmacology , Alanine/administration & dosage , Alanine/analogs & derivatives , Alanine/pharmacology , Prodrugs/administration & dosage , Prodrugs/pharmacology , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology
5.
J Control Release ; 369: 63-74, 2024 May.
Article in English | MEDLINE | ID: mdl-38513729

ABSTRACT

Recent studies in colorectal cancer patients (CRC) have shown that increased resistance to thymidylate synthase (TS) inhibitors such as 5-fluorouracil (5-FU), reduce the efficacy of standard of care (SoC) treatment regimens. The nucleotide pool cleanser dUTPase is highly expressed in CRC and is an attractive target for potentiating anticancer activity of chemotherapy. The purpose of the current work was to investigate the activity of P1, P4-di(2',5'-dideoxy-5'-selenouridinyl)-tetraphosphate (P4-SedU2), a selenium-modified symmetrically capped dinucleoside with prodrug capabilities that is specifically activated by dUTPase. Using mechanochemistry, P4-SedU2 and the corresponding selenothymidine analogue P4-SeT2 were prepared with a yield of 19% and 30% respectively. The phosphate functionality facilitated complexation with the amphipathic cell-penetrating peptide RALA to produce nanoparticles (NPs). These NPs were designed to deliver P4-SedU2 intracellularly and thereby maximise in vivo activity. The NPs demonstrated effective anti-cancer activity and selectivity in the HCT116 CRC cell line, a cell line that overexpresses dUTPase; compared to HT29 CRC cells and NCTC-929 fibroblast cells which have reduced levels of dUTPase expression. In vivo studies in BALB/c SCID mice revealed no significant toxicity with respect to weight or organ histology. Pharmacokinetic analysis of blood serum showed that RALA facilitates effective delivery and rapid internalisation into surrounding tissues with NPs eliciting lower plasma Cmax than the equivalent injection of free P4-SedU2, translating the in vitro findings. Tumour growth delay studies have demonstrated significant inhibition of growth dynamics with the tumour doubling time extended by >2weeks. These studies demonstrate the functionality and action of a new pro-drug nucleotide for CRC.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Nanoparticles , Prodrugs , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Prodrugs/chemistry , Prodrugs/pharmacology , Humans , Nanoparticles/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Pyrophosphatases/antagonists & inhibitors , Female , Cell Line, Tumor , Peptides/chemistry , Peptides/administration & dosage , Peptides/pharmacokinetics , Peptides/pharmacology , Mice, Inbred BALB C , Mice , Nucleotides/administration & dosage , Nucleotides/chemistry , Nucleotides/pharmacokinetics , HCT116 Cells
6.
J Control Release ; 369: 351-362, 2024 May.
Article in English | MEDLINE | ID: mdl-38552963

ABSTRACT

Polymeric prodrug nanoparticles have gained increasing attention in the field of anticancer drug delivery because of their dual functions as a drug carrier and a therapeutic agent. Doxorubicin (DOX) is a highly effective chemotherapeutic agent for various cancers but causes cardiotoxicity. In this work, we developed polymeric prodrug (pHU) nanoparticles that serve as both a drug carrier of DOX and a therapeutic agent. The composition of pHU includes antiangiogenic hydroxybenzyl alcohol (HBA) and ursodeoxycholic acid (UDCA), covalently incorporated through hydrogen peroxide (H2O2)-responsive peroxalate. To enhance cancer cell specificity, pHU nanoparticles were surface decorated with taurodeoxycholic acid (TUDCA) to facilitate p-selectin-mediated cancer targeting. TUDCA-coated and DOX-loaded pHU nanoparticles (t-pHUDs) exhibited controlled release of DOX triggered by H2O2, characteristic of the tumor microenvironment. t-pHUDs also effectively suppressed cancer cell migration and vascular endothelial growth factor (VEGF) expression in response to H2O2. In animal studies, t-pHUDs exhibited highly potent anticancer activity. Notably, t-pHUDs, with their ability to accumulate preferentially in tumors due to the p-selectin targeting, surpassed the therapeutic efficacy of equivalent DOX and pHU nanoparticles alone. What is more, t-pHUDs significantly suppressed VEGF expression in tumors and mitigated hepato- and cardiotoxicity of DOX. Given their cancer targeting ability, enhanced therapeutic efficacy and minimized off-target toxicity, t-pHUDs present an innovative and targeted approach with great translational potential as an anticancer therapeutic agent.


Subject(s)
Doxorubicin , Nanoparticles , Prodrugs , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Prodrugs/administration & dosage , Prodrugs/chemistry , Nanoparticles/chemistry , Animals , Humans , Cell Line, Tumor , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/pathology , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Hydrogen Peroxide , Drug Carriers/chemistry , Ursodeoxycholic Acid/administration & dosage , Ursodeoxycholic Acid/chemistry , Drug Liberation , Mice, Nude , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Polymers/chemistry , Vascular Endothelial Growth Factor A/metabolism , Mice , Female , Drug Delivery Systems , Cell Movement/drug effects , Benzyl Alcohols/administration & dosage , Benzyl Alcohols/chemistry
7.
J Control Release ; 369: 376-393, 2024 May.
Article in English | MEDLINE | ID: mdl-38554772

ABSTRACT

Despite their great versatility and ease of functionalization, most polymer-based nanocarriers intended for use in drug delivery often face serious limitations that can prevent their clinical translation, such as uncontrolled drug release and off-target toxicity, which mainly originate from the burst release phenomenon. In addition, residual solvents from the formulation process can induce toxicity, alter the physico-chemical and biological properties and can strongly impair further pharmaceutical development. To address these issues, we report polymer prodrug nanoparticles, which are prepared without organic solvents via an all-aqueous formulation process, and provide sustained drug release. This was achieved by the "drug-initiated" synthesis of well-defined copolymer prodrugs exhibiting a lower critical solution temperature (LCST) and based on the anticancer drug gemcitabine (Gem). After screening for different structural parameters, prodrugs based on amphiphilic diblock copolymers were formulated into stable nanoparticles by all-aqueous nanoprecipitation, with rather narrow particle size distribution and average diameters in the 50-80 nm range. They exhibited sustained Gem release in human serum and acetate buffer, rapid cellular uptake and significant cytotoxicity on A549 and Mia PaCa-2 cancer cells. We also demonstrated the versatility of this approach by formulating Gem-based polymer prodrug nanoparticles loaded with doxorubicin (Dox) for combination therapy. The dual-drug nanoparticles exhibited sustained release of Gem in human serum and acidic release of Dox under accelerated pathophysiological conditions. Importantly, they also induced a synergistic effect on triple-negative breast cancer line MDA-MB-231, which is a relevant cell line to this combination.


Subject(s)
Deoxycytidine , Drug Liberation , Gemcitabine , Nanoparticles , Polymers , Prodrugs , Temperature , Prodrugs/administration & dosage , Prodrugs/chemistry , Humans , Nanoparticles/chemistry , Deoxycytidine/analogs & derivatives , Deoxycytidine/administration & dosage , Deoxycytidine/chemistry , Deoxycytidine/pharmacokinetics , Polymers/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Delayed-Action Preparations , Drug Carriers/chemistry , Chemical Precipitation , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacokinetics
8.
J Clin Anesth ; 95: 111442, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38493706

ABSTRACT

STUDY OBJECTIVE: Fospropofol disodium is a propofol prodrug that is water-soluble and has a reduced risk of bacterial contamination and hypertriglyceridemia compared with propofol. Prior to implementing a large randomized trial, we investigated the feasibility, initial efficacy, and safety of fospropofol disodium compared with propofol in long-term mild-to-moderate sedation in intensive care units (ICUs). DESIGN: Single-centered, prospective, unblind, randomized, parallel-group clinical trial. SETTING: The general ICU of university-affiliated teaching hospital. PATIENTS: Adult patients (n = 60) expected to have mechanical ventilation for >24 h were enrolled and randomly assigned to the fospropofol or propofol group. INTERVENTIONS: The fospropofol group received continuous fospropofol disodium infusions and the propofol group received continuous propofol infusions. The sedation goal was a score of -3 to 0 on the Richmond Agitation and Sedation Scale (RASS). MEASUREMENTS: The primary outcome was the percentage of time spent in the target sedation range without rescue sedation. Safety outcomes were based on adverse events. Blood samples were collected to measure formate concentration in plasma. MAIN RESULTS: The median dose was 4.33 (IQR, 3.08-4.94) mg/kg/h in the fospropofol group and 1.96 (IQR, 1.44-2.94) mg/kg/h in the propofol group. The median percentage of time spent in the target RASS range without rescue sedation was identical in both groups, with 83.33% (IQR, 74.43%-100.00%) in the fospropofol group and 83.33% (IQR, 77.45%-100.00%) in the propofol group (p = 0.887). At least one adverse event was identifed in 23 (76.7%) fospropofol patients and 27 (90.0%) propofol patients. The most common adverse events were tachycardia and hypotension. No paresthesia, catheter-related bloodstream infection or propofol infusion syndrome in both groups was reported. Three patients in the fospropofol group had mild hypertriglyceridemia, and nine patients in propofol group had hypertriglyceridemia (mild in eight patients and moderate in one patient) (10% versus 30%, p = 0.104). The formate concentration in plasma was very low, and no significant difference was identified at any time point between the two groups. CONCLUSIONS: Fospropofol disodium appears to be a feasible, effective and safe sedative for patients receiving invasive mechanical ventilation with long-term sedation.


Subject(s)
Hypnotics and Sedatives , Propofol , Propofol/analogs & derivatives , Respiration, Artificial , Humans , Propofol/administration & dosage , Propofol/adverse effects , Male , Female , Middle Aged , Pilot Projects , Respiration, Artificial/adverse effects , Prospective Studies , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/adverse effects , Aged , Intensive Care Units , Feasibility Studies , Adult , Conscious Sedation/methods , Conscious Sedation/adverse effects , Infusions, Intravenous , Prodrugs/administration & dosage , Prodrugs/adverse effects
9.
Science ; 383(6688): 1181-1182, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38484073
10.
J Control Release ; 369: 179-198, 2024 May.
Article in English | MEDLINE | ID: mdl-38368947

ABSTRACT

Engineering human enzymes for therapeutic applications is attractive but introducing new amino acids may adversely affect enzyme stability and immunogenicity. Here we used a mammalian membrane-tethered screening system (ECSTASY) to evolve human lysosomal beta-glucuronidase (hBG) to hydrolyze a glucuronide metabolite (SN-38G) of the anticancer drug irinotecan (CPT-11). Three human beta-glucuronidase variants (hBG3, hBG10 and hBG19) with 3, 10 and 19 amino acid substitutions were identified that display up to 40-fold enhanced enzymatic activity, higher stability than E. coli beta-glucuronidase in human serum, and similar pharmacokinetics in mice as wild-type hBG. The hBG variants were two to three orders of magnitude less immunogenic than E. coli beta-glucuronidase in hBG transgenic mice. Intravenous administration of an immunoenzyme (hcc49-hBG10) targeting a sialyl-Tn tumor-associated antigen to mice bearing human colon xenografts significantly enhanced the anticancer activity of CPT-11 as measured by tumor suppression and mouse survival. Our results suggest that genetically-modified human enzymes represent a good alternative to microbially-derived enzymes for therapeutic applications.


Subject(s)
Camptothecin , Glucuronidase , Irinotecan , Mice, Transgenic , Prodrugs , Animals , Prodrugs/administration & dosage , Humans , Irinotecan/administration & dosage , Irinotecan/pharmacokinetics , Glucuronidase/genetics , Glucuronidase/metabolism , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/therapeutic use , Protein Engineering , Mice , Cell Line, Tumor , Female , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Xenograft Model Antitumor Assays , Enzyme Stability , Mice, Nude
11.
Brasília; CONITEC; 2024.
Non-conventional in Portuguese | BRISA/RedTESA | ID: biblio-1554034

ABSTRACT

INTRODUÇÃO: O HIV-1 é um vírus que apresenta em seu envelope viral a glicoproteína gp120, capaz de se ligar aos receptores CD4+ dos linfócitos T do hospedeiro, inviabilizando o funcionamento normal ou levando à destruição das células do sistema imune da pessoa vivendo com esse agente infeccioso. No contexto do tratamento contra o vírus, as quasispécies de HIV-1 podem sofrer uma ou mais mutações genéticas que afetam a atividade de um ou mais ARVs que já foram efetivos anteriormente, processo que é denominado resistência. Segundo a Organização Mundial de Saúde (OMS), observa-se em diversos países que a resistência atinge mais de 10% dos indivíduos em início ou reinício do tratamento contra o HIV. PERGUNTA: Fostensavir 600 mg é eficaz, custo-efetivo e seguro no tratamento de pessoas adultas vivendo com HIV-1 multirresistente aos ARVs? EVIDÊNCIAS CLÍNICAS: A partir da pergunta de pesquisa, foi desenvolvida estratégia de busca nas bases de dados MEDLINE via PubMed e EMBASE. A busca realizada resultou em 318 publicações. Foram inicialmente excluídas 72 por serem duplicatas. Posteriormente, foram excluídas outras 220 após triagem. Após leitura dos textos completos, chegou-se ao resultado de cinco publicações elegíveis, todas fruto do ensaio clínico randomizado de fase III, BRIGHTE. Foram relatados os desfechos de média de redução de carga viral, resposta virológica, falha virológica, variação média de linfócitos T CD4+, eventos adversos, morte, qualidade de vida e adesão ao tratamento. Em geral, o nível de certeza das evidências foi classificado como baixo, com risco de viés grave. AVALIAÇÃO ECONÔMICA: Utilizando um modelo de árvore de decisão, foi realizada uma análise para estimar a razão de custo-efetividade incremental (RCEI) do fostensavir 600 mg para pessoas vivendo com HIV-Aids, adultos e com multirresistência a pelo menos quatro classes terapêuticas de antirretrovirais desde que combinado a pelo menos um ARV totalmente ativo para um ano. O modelo comparou o fostensavir à terapia de base otimizada (TBO) apresentada no estudo pivotal BRIGHTE. ANÁLISE DE IMPACTO ORÇAMENTÁRIO: Foram projetados dois cenários de incorporação para a difusão do fostensavir: conservador e moderado. No cenário de difusão conservador (market share 10% ao ano), o impacto da incorporação do fostensavir em cinco anos variou entre R$ 10.975.053,60 e R$ 65.109.874,58, de 2024 a 2028 respectivamente. O impacto orçamentário acumulado em cinco anos no cenário conservador foi R$ 185.241.468,80. No cenário de difusão moderado (market share 20% ao ano), o impacto da incorporação em cinco anos variou entre R$ 10.975.053,60 e R$ 117.197.774,25, de 2024 a 2028. O impacto orçamentário acumulado em cinco anos no cenário de difusão moderado foi R$ 310.435.446,95. Monitoramento do horizonte tecnológico: Foram identificadas duas tecnologias para o tratamento de pessoas adultas convivendo com HIV multirresistente. Lenacapavir, que está registrado nas agências EMA e FDA, e ibalizumabe, com registro na FDA. PERSPECTIVA DO PACIENTE: A chamada pública nº 30/2023 ficou aberta entre 14 e 24 de agosto de 2023. Duas pessoas se inscreveram. O representante titular contou que tem 50 anos e há 25 vive com HIV. Acredita já ter usado todas as classes de medicamentos por conta da multirresistência do HIV aos ARV, chegando a ficar sem opção terapêutica por cinco ou seis anos. Atualmente utiliza a combinação maraviroque, dolutegravir, tenofovir, lamivudina, darunavir e ritonavir, com a qual consegue obter a supressão da carga viral. Ressaltou que quem vive com HIV há muito tempo corre o risco de ficar sem opção de medicamentos e que a incorporação de novas tecnologias pode beneficiar pessoas como ele. RECOMENDAÇÃO PRELIMINAR DA CONITEC: Os membros do Comitê de Medicamentos presentes na 125ª Reunião Ordinária, realizada no dia 6 de dezembro de 2023, deliberaram, por unanimidade, encaminhar o tema para consulta pública com recomendação preliminar favorável à incorporação do fostensavir trometamol 600 mg para o tratamento de pessoas vivendo com HIV-Aids multirresistentes a terapia antirretroviral. Considerou-se a oportunidade de uma opção terapêutica aos indivíduos multirresitentes, a capacidade das Câmaras Técnicas Estaduais e da área técnica do Ministério da Saúde no monitoramento dos benefícios clínicos e dos eventos adversos do fostensavir e a expectativa de uma nova proposta de preço encaminhada pela empresa durante a consulta pública. CONSULTA PÚBLICA: A consulta pública (CP) nº 69/2023 foi realizada entre os dias 29/12/2023 e 17/01/2024. Foram recebidas 11 contribuições, sendo sete pelo formulário para contribuições técnico-científico e quatro pelo formulário pra contribuições sobre experiência ou opinião de pacientes, familiares, amigos ou cuidadores de pacientes, profissionais de saúde ou pessoas interessadas no tema. Sobre as contribuições técnicas, todas foram favoráveis às recomendações preliminares da Conitec e uma possuía referencial teórico para a abordagem técnico-científica. Entretanto, não se identificou nenhuma evidência científica adicional que pudesse modificar o entendimento preliminar da Conitec. Nas contribuições de experiência ou opinião, todos os participantes manifestaram-se favoráveis à recomendação preliminar da tecnologia avaliada. Os argumentos relevantes foram classificados em "Sobrevida do paciente", uma vez que a abordagem foi especificamente quanto às falhas terapêuticas do uso de outros antirretrovirais e a evolução para complicações e óbito. Metade dos respondedores apontaram ter experiência com o fostensavir. No que diz respeito à experiência com outras tecnologias, um dos três respondentes, o paciente, mencionou o uso de medicamentos, como tenofovir, lamivudina, efavirenz e dolutegravir e apontou como evento negativo os seus efeitos colaterais. Além disso, destacaram o valor elevado da aquisição do fostensavir como uma das principais dificuldades para acesso a este tratamento. NOVA PROPOSTA COMERCIAL: Foi submetida ao DGITS/SECTICS/MS o valor de USD 38,67 por comprimido, com uma quantidade mínima de aquisição estabelecida em 360.000 (trezentos e sessenta mil) unidades. Considerando o novo valor proposto, o custo mensal do tratamento será de R$ 11.513,40, representando aproximadamente 65,59% de desconto em relação ao preço CMED PMVG 18% e uma redução de 1,66% em relação à proposta apresentada em 2023. Ainda com base no novo valor proposto, atualizou-se a avaliação econômica e a AIO. Os critérios considerados englobam os termos da recente proposta comercial e a taxa de câmbio do dólar no dia 15 de fevereiro de 2024, fixada em 1 USD = R$ 4,9624. A RCEI foi estimada em R$ 257.370,65, apresentando uma efetividade incremental de 0,54. No cenário conservador da nova AIO, levando em consideração uma participação de mercado de 10% ao ano, o impacto orçamentário incremental em cinco anos foi R$ 100.714.577,75. Já no cenário moderado, com um aumento de 20% após o primeiro ano, o impacto orçamentário acumulado em cinco anos foi R$ 247.300.234,85. RECOMENDAÇÃO FINAL DA CONITEC: Os membros do Comitê de Medicamentos presentes na 127ª Reunião Ordinária, realizada no dia 6 de março de 2024, deliberaram, por unanimidade, recomendar a incorporação do fostensavir trometamol 600 mg para o tratamento de indivíduos adultos vivendo com HIV multirresistentes aos antirretrovirais, conforme Protocolo Clínico do Ministério da Saúde. Considerou-se as expectativas da ampliação das opções terapêuticas e da redução da carga viral aos pacientes multirresistentes. Foi assinado o Registro de Deliberação nº 881/2024. DECISÃO: incorporar, no âmbito do Sistema Único de Saúde - SUS, o fostensavir trometamol 600 mg para o tratamento de indivíduos adultos vivendo com HIV multirresistente aos antirretrovirais, conforme Protocolo Clínico do Ministério da Saúde, publicada no Diário Oficial da União nº 77, seção 1, página 177, em 22 de abril de 2024.


Subject(s)
Humans , Prodrugs/administration & dosage , HIV Envelope Protein gp120/antagonists & inhibitors , HIV Infections/drug therapy , Drug Resistance, Multiple/drug effects , Anti-Retroviral Agents/adverse effects , Health Evaluation/economics , Unified Health System , Brazil , Cost-Benefit Analysis/economics
12.
Int J Mol Sci ; 24(5)2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36902076

ABSTRACT

Triple-negative breast cancer (TNBC) is an extremely aggressive subtype associated with a poor prognosis. At present, the treatment for TNBC mainly relies on surgery and traditional chemotherapy. As a key component in the standard treatment of TNBC, paclitaxel (PTX) effectively inhibits the growth and proliferation of tumor cells. However, the application of PTX in clinical treatment is limited due to its inherent hydrophobicity, weak penetrability, nonspecific accumulation, and side effects. To counter these problems, we constructed a novel PTX conjugate based on the peptide-drug conjugates (PDCs) strategy. In this PTX conjugate, a novel fused peptide TAR consisting of a tumor-targeting peptide, A7R, and a cell-penetrating peptide, TAT, is used to modify PTX. After modification, this conjugate is named PTX-SM-TAR, which is expected to improve the specificity and penetrability of PTX at the tumor site. Depending on hydrophilic TAR peptide and hydrophobic PTX, PTX-SM-TAR can self-assemble into nanoparticles and improve the water solubility of PTX. In terms of linkage, the acid- and esterase-sensitive ester bond was used as the linking bond, with which PTX-SM-TAR NPs could remain stable in the physiological environment, whereas PTX-SM-TAR NPs could be broken and PTX be released at the tumor site. A cell uptake assay showed that PTX-SM-TAR NPs were receptor-targeting and could mediate endocytosis by binding to NRP-1. The vascular barrier, transcellular migration, and tumor spheroids experiments showed that PTX-SM-TAR NPs exhibit great transvascular transport and tumor penetration ability. In vivo experiments, PTX-SM-TAR NPs showed higher antitumor effects than PTX. As a result, PTX-SM-TAR NPs may overcome the shortcomings of PTX and present a new transcytosable and targeted delivery system for PTX in TNBC treatment.


Subject(s)
Nanoparticle Drug Delivery System , Oligopeptides , Paclitaxel , Prodrugs , Triple Negative Breast Neoplasms , Humans , Cell Line, Tumor , Nanoparticles/administration & dosage , Paclitaxel/administration & dosage , Prodrugs/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Nanoparticle Drug Delivery System/administration & dosage , Oligopeptides/administration & dosage
13.
FASEB J ; 36(3): e22213, 2022 03.
Article in English | MEDLINE | ID: mdl-35192728

ABSTRACT

High-risk solid tumors continue to pose a tremendous therapeutic challenge due to multidrug resistance. Biological mechanisms driving chemoresistance in high-risk primary and recurrent disease are distinct: in newly diagnosed patients, non-response to therapy is often associated with a higher level of tumor "stemness" paralleled by overexpression of the ABCG2 drug efflux pump, whereas in tumors relapsing after non-curative therapy, poor drug sensitivity is most commonly linked to the dysfunction of the tumor suppressor protein, p53. In this study, we used preclinical models of aggressive neuroblastoma featuring these characteristic mechanisms of primary and acquired drug resistance to experimentally evaluate a macromolecular prodrug of a structurally enhanced camptothecin analog, SN22, resisting ABCG2-mediated export, and glucuronidation. Together with extended tumor exposure to therapeutically effective drug levels via reversible conjugation to Pluronic F-108 (PF108), these features translated into rapid tumor regression and long-term survival in models of both ABCG2-overexpressing and p53-mutant high-risk neuroblastomas, in contrast to a marginal effect of the clinically used camptothecin derivative, irinotecan. Our results demonstrate that pharmacophore enhancement, increased tumor uptake, and optimally stable carrier-drug association integrated into the design of the hydrolytically activatable PF108-[SN22]2  have the potential to effectively combat multiple mechanisms governing chemoresistance in newly diagnosed (chemo-naïve) and recurrent forms of aggressive malignancies. As a macromolecular carrier-based delivery system exhibiting remarkable efficacy against two particularly challenging forms of high-risk neuroblastoma, PF108-[SN22]2 can pave the way to a robust and clinically viable therapeutic strategy urgently needed for patients with multidrug-resistant disease presently lacking effective treatment options.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm , Neuroblastoma/drug therapy , Prodrugs/therapeutic use , Topoisomerase I Inhibitors/therapeutic use , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Camptothecin/analogs & derivatives , Cell Line, Tumor , Humans , Mice , Mice, Nude , Mice, SCID , Poloxamer/chemistry , Prodrugs/administration & dosage , Prodrugs/chemistry , Topoisomerase I Inhibitors/chemistry
14.
J Immunother Cancer ; 10(1)2022 01.
Article in English | MEDLINE | ID: mdl-35101947

ABSTRACT

BACKGROUND: Interleukin-15 (IL-15) is an important cytokine necessary for proliferation and maintenance of natural killer (NK) and CD8+ T cells, and with great promise as an immuno-oncology therapeutic. However, IL-15 has a very short half-life and a single administration does not provide the sustained exposure required for optimal stimulation of target immune cells. The purpose of this work was to develop a very long-acting prodrug that would maintain IL-15 within a narrow therapeutic window for long periods-similar to a continuous infusion. METHODS: We prepared and characterized hydrogel microspheres (MS) covalently attached to IL-15 (MS~IL-15) by a releasable linker. The pharmacokinetics and pharmacodynamics of MS~IL-15 were determined in C57BL/6J mice. The antitumor activity of MS~IL-15 as a single agent, and in combination with a suitable therapeutic antibody, was tested in a CD8+ T cell-driven bilateral transgenic adenocarcinoma mouse prostate (TRAMP)-C2 model of prostatic cancer and a NK cell-driven mouse xenograft model of human ATL (MET-1) murine model of adult T-cell leukemia. RESULTS: On subcutaneous administration to mice, the cytokine released from the depot maintained a long half-life of about 168 hours over the first 5 days, followed by an abrupt decrease to about ~30 hours in accordance with the development of a cytokine sink. A single injection of MS~IL-15 caused remarkably prolonged expansions of NK and ɣδ T cells for 2 weeks, and CD44hiCD8+ T cells for 4 weeks. In the NK cell-driven MET-1 murine model of adult T-cell leukemia, single-agent MS~IL-1550 µg or anti-CCR4 provided modest increases in survival, but a combination-through antibody-depedent cellular cytotoxicity (ADCC)-significantly extended survival. In a CD8+ T cell-driven bilateral TRAMP-C2 model of prostatic cancer, single agent subcutaneous MS~IL-15 or unilateral intratumoral agonistic anti-CD40 showed modest growth inhibition, but the combination exhibited potent, prolonged bilateral antitumor activity. CONCLUSIONS: Our results show MS~IL-15 provides a very long-acting IL-15 with low Cmax that elicits prolonged expansion of target immune cells and high anticancer activity, especially when administered in combination with a suitable immuno-oncology agent.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Interleukin-15/administration & dosage , Leukemia, T-Cell/drug therapy , Prodrugs/administration & dosage , Prostatic Neoplasms/drug therapy , Animals , CD40 Antigens/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , Drug Delivery Systems , Half-Life , Humans , Immunotherapy , Interleukin-15/pharmacokinetics , Male , Mice, Inbred C57BL , Microspheres , Prodrugs/pharmacokinetics , Receptors, CCR4/antagonists & inhibitors
15.
Carbohydr Polym ; 282: 119087, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35123755

ABSTRACT

The efficient triggering of prodrug release has become a challengeable task for stimuli-responsive nanomedicine utilized in cancer therapy due to the subtle differences between normal and tumor tissues and heterogeneity. In this work, a dual ROS-responsive nanocarriers with the ability to self-regulate the ROS level was constructed, which could gradually respond to the endogenous ROS to achieve effective, hierarchical and specific drug release in cancer cells. In brief, DOX was conjugated with MSNs via thioketal bonds and loaded with ß-Lapachone. TPP modified chitosan was then coated to fabricate nanocarriers for mitochondria-specific delivery. The resultant nanocarriers respond to the endogenous ROS and release Lap specifically in cancer cells. Subsequently, the released Lap self-regulated the ROS level, resulting in the specific DOX release and mitochondrial damage in situ, enhancing synergistic oxidation-chemotherapy. The tumor inhibition Ratio was achieved to 78.49%. The multi-functional platform provides a novel remote drug delivery system in vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Nanoparticles/administration & dosage , Naphthoquinones/administration & dosage , Neoplasms/drug therapy , Oxidative Stress , Prodrugs/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Chitosan/administration & dosage , Chitosan/chemistry , Chitosan/pharmacokinetics , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Liberation , Female , Humans , Mice, Inbred BALB C , Mitochondria/physiology , Nanoparticles/chemistry , Naphthoquinones/chemistry , Naphthoquinones/pharmacokinetics , Neoplasms/metabolism , Neoplasms/pathology , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Oxidation-Reduction , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Reactive Oxygen Species/metabolism , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacokinetics , Tumor Burden/drug effects
16.
J Clin Endocrinol Metab ; 107(1): e372-e385, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34347093

ABSTRACT

CONTEXT: Hypoparathyroidism is characterized by insufficient levels of parathyroid hormone (PTH). TransCon PTH is an investigational long-acting prodrug of PTH(1-34) for the treatment of hypoparathyroidism. OBJECTIVE: This work aimed to investigate the safety, tolerability, and efficacy of daily TransCon PTH in adults with hypoparathyroidism. METHODS: This phase 2, randomized, double-blind, placebo-controlled 4-week trial with open-label extension enrolled 59 individuals with hypoparathyroidism. Interventions included TransCon PTH 15, 18, or 21 µg PTH(1-34)/day or placebo for 4 weeks, followed by a 22-week extension during which TransCon PTH dose was titrated (6-60 µg PTH[1-34]/day). RESULTS: By Week 26, 91% of participants treated with TransCon PTH achieved independence from standard of care (SoC, defined as active vitamin D = 0 µg/day and calcium [Ca] ≤ 500 mg/day). Mean 24-hour urine Ca (uCa) decreased from a baseline mean of 415 mg/24h to 178 mg/24h by Week 26 (n = 44) while normal serum Ca (sCa) was maintained and serum phosphate and serum calcium-phosphate product fell within the normal range. By Week 26, mean scores on the generic 36-Item Short Form Health Survey domains increased from below normal at baseline to within the normal range. The Hypoparathyroidism Patient Experience Scale symptom and impact scores improved through 26 weeks. TransCon PTH was well tolerated with no treatment-related serious or severe adverse events. CONCLUSION: TransCon PTH enabled independence from oral active vitamin D and reduced Ca supplements (≤ 500 mg/day) for most participants, achieving normal sCa, serum phosphate, uCa, serum calcium-phosphate product, and demonstrating improved health-related quality of life. These results support TransCon PTH as a potential hormone replacement therapy for adults with hypoparathyroidism.


Subject(s)
Hormone Replacement Therapy/methods , Hypoparathyroidism/drug therapy , Parathyroid Hormone/administration & dosage , Adult , Aged , Calcium/administration & dosage , Calcium/blood , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/adverse effects , Double-Blind Method , Drug Administration Schedule , Female , Hormone Replacement Therapy/adverse effects , Humans , Hypoparathyroidism/blood , Hypoparathyroidism/complications , Hypoparathyroidism/diagnosis , Male , Middle Aged , Parathyroid Hormone/adverse effects , Parathyroid Hormone/blood , Patient Reported Outcome Measures , Placebos/administration & dosage , Placebos/adverse effects , Prodrugs/administration & dosage , Prodrugs/adverse effects , Quality of Life , Treatment Outcome , Vitamin D/administration & dosage , Vitamin D/blood
17.
ChemMedChem ; 17(1): e202100634, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34870892

ABSTRACT

Because of their large polar surface area, carbohydrates often exhibit insufficient pharmacokinetic properties. Specifically, the carboxylic acid function of the tetrasaccharide sialyl Lewisx , a pharmacophore crucial for the formation of a salt bridge with selectins, prevents oral availability. A common approach is the transfer of carboxylic acid into ester prodrugs. Once the prodrug is either actively or passively absorbed, the active principle is released by hydrolysis. In the present study, ester prodrugs of selectin antagonists with aliphatic promoieties were synthesized and their potential for oral availability was investigated in vitro and in vivo. The addition of lipophilic ester moieties to overcome insufficient lipophilicity improved passive permeation into enterocytes, however at the same time supported efflux back to the small intestines as well as oxidation into non-hydrolysable metabolites. In summary, our examples demonstrate that different modifications of carbohydrates can result in opposing effects and have to be studied in their entirety.


Subject(s)
E-Selectin/antagonists & inhibitors , Esters/pharmacology , Prodrugs/pharmacology , Administration, Oral , Animals , Biological Availability , Caco-2 Cells , Dose-Response Relationship, Drug , E-Selectin/metabolism , Esters/administration & dosage , Esters/chemistry , Female , Humans , Male , Mice , Mice, Inbred BALB C , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Prodrugs/administration & dosage , Prodrugs/chemistry , Rats , Structure-Activity Relationship
18.
ACS Appl Mater Interfaces ; 14(1): 297-306, 2022 Jan 12.
Article in English | MEDLINE | ID: mdl-34958191

ABSTRACT

Aromatized thioketal (ATK) linked the immunoregulatory molecule (budesonide, Bud) and the cytotoxic molecule (gemcitabine, Gem) to construct a ROS-activated Janus-prodrug, termed as BAG. Benefiting from the hydrogen bonding, π-π stacking, and other intermolecular interactions, BAG could self-assemble into nanoaggregates (BAG NA) with a well-defined spherical shape and uniform size distribution. Compared to the carrier-based drug delivery system, BAG NA have ultrahigh drug loading content and ROS concentration-dependent drug release. Colitis-associated colorectal cancer (CAC) is a typical disease in which chronic inflammation transforms into tumors. BAG NA can be internalized by colon cancer C26 cells and then triggered by excessive intracellular ROS to release nearly 100% of the drugs. Based on this, BAG NA showed a stronger pro-apoptotic effect than free Bud combined with free Gem. What is gratifying is that orally administered BAG NA can precisely accumulate in the diseased colon tissues of CAC mice induced by AOM/DSS and simultaneously release Bud and Gem. Bud can regulate the tumor immune microenvironment to restore and enhance the cytotoxicity of Gem. Therefore, BAG NA maximizes the synergistic therapeutic effect through co-delivery of Bud and Gem. This work provided a cutting-edge method for constructing self-delivery Janus-prodrug based on ATK and confirmed its potential application in inflammation-related carcinogenesis.


Subject(s)
Antineoplastic Agents/pharmacology , Biocompatible Materials/pharmacology , Colitis-Associated Neoplasms/drug therapy , Colorectal Neoplasms/drug therapy , Drug Delivery Systems , Prodrugs/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemistry , Budesonide/administration & dosage , Budesonide/chemistry , Budesonide/pharmacology , Cell Line , Colitis-Associated Neoplasms/metabolism , Colitis-Associated Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Materials Testing , Mice , Mice, Inbred BALB C , Molecular Dynamics Simulation , Molecular Structure , Prodrugs/administration & dosage , Prodrugs/chemistry , Gemcitabine
19.
Angew Chem Int Ed Engl ; 61(4): e202114250, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34800083

ABSTRACT

PtIV prodrugs can overcome resistance and side effects of conventional PtII anticancer therapies. By 19 F-labeling of a PtIV prodrug (Pt-FBA, FBA=p-fluorobenzoate), the activation under physiological conditions could be investigated. Unlike single-electron reductants, multi-electron agents can efficiently promote the two electrons reduction of PtIV to PtII . The activation of Pt-FBA in cell lysate is highly dependent upon the type of cancer cells. When administered to E. coli, Pt-FBA is reduced intracellularly and free FBA can shuttle out of the cell. The reduction rate greatly increases by inducing metallothionein overexpression and is lowered by addition of ZnII ions. When injected into mice, Pt-FBA undergoes fast reduction in the bloodstream accompanied by metabolic degradation of FBA; nevertheless, unreduced Pt-FBA can accumulate to detectable levels in liver and kidneys. The 19 F NMR approach has the advantage of avoiding the interference of all background signals.


Subject(s)
Organoplatinum Compounds/metabolism , Prodrugs/metabolism , Animals , Fluorides , Mice , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/chemistry , Prodrugs/administration & dosage , Prodrugs/chemistry
20.
Int J Mol Sci ; 22(23)2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34884910

ABSTRACT

Activation of thyroid hormone receptor ß (THRß) has shown beneficial effects on metabolic alterations, including non-alcoholic fatty liver disease (NAFLD). Here, we investigated the effect of TG68, a novel THRß agonist, on fatty liver accumulation and liver injury in mice fed a high-fat diet (HFD). C57BL/6 mice fed HFD for 17 or 18 weeks, a time when all mice developed massive steatohepatitis, were then given TG68 at a dose of 9.35 or 2.8 mg/kg for 2 or 3 weeks, respectively. As a reference compound, the same treatment was adopted using equimolar doses of MGL-3196, a selective THRß agonist currently in clinical phase III. The results showed that treatment with TG68 led to a reduction in liver weight, hepatic steatosis, serum transaminases, and circulating triglycerides. qRT-PCR analyses demonstrated activation of THRß, as confirmed by increased mRNA levels of Deiodinase-1 and Malic enzyme-1, and changes in lipid metabolism, as revealed by increased expression of Acyl-CoA Oxidase-1 and Carnitine palmitoyltransferase-1. The present results showed that this novel THRß agonist exerts an anti-steatogenic effect coupled with amelioration of liver injury in the absence of extra-hepatic side effects, suggesting that TG68 may represent a useful tool for the treatment of NAFLD.


Subject(s)
Diet, High-Fat/adverse effects , Non-alcoholic Fatty Liver Disease/drug therapy , Prodrugs/administration & dosage , Pyridazines/administration & dosage , Thyroid Hormone Receptors beta/agonists , Uracil/analogs & derivatives , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Lipid Metabolism/drug effects , Male , Mice , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism , Organ Size/drug effects , Prodrugs/pharmacology , Pyridazines/pharmacology , Transaminases/blood , Triglycerides/blood , Uracil/administration & dosage , Uracil/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...