Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.882
Filter
1.
BMC Cardiovasc Disord ; 24(1): 344, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977993

ABSTRACT

BACKGROUND: The link between diabetes mellitus and chronic hepatitis C infection remains well established. It is estimated that up to one third of chronic hepatitis C patients have type II diabetes mellitus. Hepatitis C virus infection is one of the main global health burdens. Sofosbuvir and Daclatasvir are used as effective antiviral inhibitors of hepatitis C virus. The cardiovascular effects of those drugs are not well studied. We used electrocardiography and echocardiography with global longitudinal strain assessment by speckle tracking to detect their effect on cardiac function. METHODS AND RESULTS: One hundred diabetic patients with hepatitis C infection were included in the study. Abdominal ultrasound and laboratory work up were carried out for all participants. Left ventricular systolic and diastolic function were assessed by 2D-echocardiography and global longitudinal strain, before and 3 months after treatment. Results showed significant decrease in global longitudinal strain 3 months after therapy (-21 ± 4 vs. -18 ± 7; P < 0.001) but other echocardiographic findings showed no significant changes. CONCLUSIONS: Sofosbuvir and Daclatasvir were associated with early left ventricular systolic dysfunction as assessed by global longitudinal strain in diabetic patients. More deterioration in left ventricular systolic function was detected among those with Child-Pough class B. Further long-term follow-up may be required.


Subject(s)
Antiviral Agents , Carbamates , Diabetes Mellitus, Type 2 , Hepatitis C, Chronic , Imidazoles , Pyrrolidines , Sofosbuvir , Valine , Ventricular Dysfunction, Left , Ventricular Function, Left , Humans , Antiviral Agents/therapeutic use , Antiviral Agents/adverse effects , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/complications , Male , Female , Middle Aged , Sofosbuvir/therapeutic use , Sofosbuvir/adverse effects , Valine/analogs & derivatives , Valine/therapeutic use , Pyrrolidines/therapeutic use , Imidazoles/therapeutic use , Treatment Outcome , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/diagnosis , Carbamates/therapeutic use , Ventricular Function, Left/drug effects , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/diagnosis , Time Factors , Aged , Electrocardiography , Adult
2.
Medicina (Kaunas) ; 60(7)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39064466

ABSTRACT

A 76-year-old female patient presented with an iodine-refractory papillary thyroid carcinoma (PTC), diagnosed eight years earlier, with several lymph node recurrences requiring successive surgeries. Fluorodeoxyglucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT) imaging revealed a new unresectable loco-regional recurrence. The patient was diagnosed with a somatic BRAF V600E mutation. Therefore, dabrafenib and trametinib combination therapy was introduced and closely monitored by a dedicated multidisciplinary team, involving pharmaceutical consultations. As early as six weeks after treatment initiation, the patient reported multiple adverse events (AEs) to the clinical pharmacy team, who provided advice on resolving AEs or improving tolerance. Close interprofessional collaboration among healthcare workers involved in the care pathway allowed for the identification of the most opportune times for temporary suspension of treatment (four suspensions over seven months) or dose reduction (two reductions over 3.5 months). This resulted in a total treatment duration (one year) longer than the average times reported in the literature. The patient showed a rapid and excellent response to treatment immediately after initiation, culminating in a complete metabolic response assessed by [18F]FDG PET/CT imaging at nine months. Twenty-five months after treatment discontinuation, the disease remained controlled. Overall, dabrafenib and trametinib combination could offer excellent outcomes in selected patients with refractory BRAF-mutated PTC, with additional clinical pharmacy initiatives allowing for the optimized management of AEs and prolonged treatment periods.


Subject(s)
Imidazoles , Oximes , Pyridones , Pyrimidinones , Thyroid Neoplasms , Humans , Female , Oximes/therapeutic use , Oximes/administration & dosage , Imidazoles/therapeutic use , Imidazoles/administration & dosage , Pyridones/therapeutic use , Pyridones/administration & dosage , Aged , Pyrimidinones/therapeutic use , Pyrimidinones/administration & dosage , Thyroid Neoplasms/drug therapy , Positron Emission Tomography Computed Tomography/methods , Iodine Radioisotopes/therapeutic use , Iodine Radioisotopes/administration & dosage , Thyroid Cancer, Papillary/drug therapy , Treatment Outcome , Proto-Oncogene Proteins B-raf/genetics
3.
Hematology ; 29(1): 2360843, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38828928

ABSTRACT

The outcomes of relapsed Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ALL) resistant to new drugs such as tyrosine kinase inhibitors, inotuzumab ozogamicin (InO) and blinatumomab are dismal. We treated two cases of Ph+ALL resistant to these drugs that achieved long-term survival after treatment with chimeric antigen receptor (CAR)-T cell therapy or a second allogeneic hematopoietic stem cell transplantation (HCT) with a sequential conditioning regimen. Case 1: A 15-year-old boy was diagnosed with Ph+ALL. Despite the second HCT after the treatment of ponatinib and blinatumomab, hematological relapse occurred. InO was ineffective and he was transferred to a CAR-T center. After the CAR-T cell therapy, negative measurable residual disease (MRD) was achieved and maintained for 38 months without maintenance therapy. Case 2: A 21-year-old man was diagnosed with Ph+ALL. Hematological relapse occurred after the first HCT. Despite of the treatment with InO, ponatinib, and blinatumomab, hematological remission was not achieved. The second HCT was performed using a sequential conditioning regimen with clofarabine. Negative MRD was subsequently achieved and maintained for 42 months without maintenance therapy. These strategies are suggestive and helpful to treat Ph+ALL resistant to multiple immunotherapies.


Subject(s)
Hematopoietic Stem Cell Transplantation , Imidazoles , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Pyridazines , Humans , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Pyridazines/therapeutic use , Adolescent , Imidazoles/therapeutic use , Young Adult , Immunotherapy , Recurrence , Transplantation, Homologous
4.
J Cardiovasc Pharmacol Ther ; 29: 10742484241258381, 2024.
Article in English | MEDLINE | ID: mdl-38828542

ABSTRACT

BACKGROUND: Moxonidine, an imidazoline I1 receptor agonist, is an effective antihypertensive drug that was shown to improve insulin sensitivity. RAAS-blockers are recommended as first-line therapy in patients with diabetes, alone or in combination with a calcium-channel antagonist or a diuretic. AIMS: This study compared the effects of moxonidine and ramipril on blood pressure (BP) and glucose metabolism in overweight patients with mild-to-moderate hypertension and impaired fasting glucose or type 2 diabetes. METHODS: Treatment-naïve patients for hypertension and dysglycemia were randomized to 12 weeks of double-blind moxonidine 0.4 mg or ramipril 5 mg once-daily treatment. At 12 weeks, for a further 12 weeks non-responders received combination of mox/ram, while responders continued blinded treatment. RESULTS: Moxonidine and ramipril were equivalent in lowering SiDBP and SiSBP at the end of the first 12 weeks. The responder rate was approximately 50% in both groups, with a mean SiDBP and SiSBP decrease of 10 and 15 mm Hg in the responders, respectively. The normalization rate (SiDBP < 85 mm Hg) was non significantly different between treatments groups. Moxonidine reduced heart rate (HR) (average -3.5 bpm, p = 0.017) during monotherapy, and when added to ramipril. HbA1c decreased significantly at Week 12 in both groups. Neither drug affected glucose or insulin response to the oral glucose tolerance test. In non-responders, moxonidine/ramipril combination further reduced BP without compromising metabolic parameters. CONCLUSION: Moxonidine 0.4 mg and ramipril 5 mg were equally effective on BP lowering and were well tolerated and mostly metabolically neutral either as monotherapies or in combination. HR was lowered on moxonidine treatment.


Subject(s)
Antihypertensive Agents , Blood Glucose , Blood Pressure , Diabetes Mellitus, Type 2 , Drug Therapy, Combination , Heart Rate , Hypertension , Imidazoles , Overweight , Ramipril , Humans , Ramipril/administration & dosage , Ramipril/therapeutic use , Ramipril/pharmacology , Hypertension/drug therapy , Hypertension/physiopathology , Male , Middle Aged , Female , Blood Pressure/drug effects , Heart Rate/drug effects , Double-Blind Method , Imidazoles/pharmacology , Imidazoles/therapeutic use , Imidazoles/administration & dosage , Antihypertensive Agents/therapeutic use , Antihypertensive Agents/pharmacology , Antihypertensive Agents/adverse effects , Blood Glucose/drug effects , Blood Glucose/metabolism , Overweight/drug therapy , Overweight/physiopathology , Overweight/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Aged , Adult , Treatment Outcome , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/adverse effects
5.
Sci Rep ; 14(1): 11840, 2024 05 23.
Article in English | MEDLINE | ID: mdl-38782988

ABSTRACT

The treatment of HCV and its sequelae are used to be predominantly based on Interferon (IFN). However, this was associated with significant adverse events as a result of its immunostimulant capabilities. Since their introduction, the directly acting antiviral drugs (DAAs), have become the standard of care to treat of HCV and its complications including mixed cryoglobulinemic vasculitis (MCV). In spite of achieving sustained viral response (SVR), there appeared many reports describing unwelcome complications such as hepatocellular and hematological malignancies as well as relapses. Prolonged inflammation induced by a multitude of factors, can lead to DNA damage and affects BAFF and APRIL, which serve as markers of B-cell proliferation. We compared, head-to-head, three antiviral protocols for HCV-MCV treatment As regards the treatment response and relapse, levels of BAFF and APRIL among pegylated interferon α-based and free regimens (Sofosbuvir + Ribavirin; SOF-RIBA, Sofosbuvir + Daclatasvir; SOF-DACLA). Regarding clinical response HCV-MCV and SVR; no significant differences could be identified among the 3 different treatment protocols, and this was also independent form using IFN. We found no significant differences between IFN-based and free regimens DNA damage, markers of DNA repair, or levels of BAFF and APRIL. However, individualized drug-to-drug comparisons showed many differences. Those who were treated with IFN-based protocol showed decreased levels of DNA damage, while the other two IFN-free groups showed increased DNA damage, being the worst in SOF-DACLA group. There were increased levels of BAFF through follow-up periods in the 3 protocols being the best in SOF-DACLA group (decreased at 24 weeks). In SOF-RIBA, CGs relapsed significantly during the follow-up period. None of our patients who were treated with IFN-based protocol had significant clinico-laboratory relapse. Those who received IFN-free DAAs showed a statistically significant relapse of constitutional manifestations. Our findings suggest that IFN-based protocols are effective in treating HCV-MCV similar to IFN-free protocols. They showed lower levels of DNA damage and repair. We believe that our findings may offer an explanation for the process of lymphoproliferation, occurrence of malignancies, and relapses by shedding light on such possible mechanisms.


Subject(s)
Antiviral Agents , Cryoglobulinemia , Vasculitis , Humans , Cryoglobulinemia/drug therapy , Cryoglobulinemia/etiology , Antiviral Agents/therapeutic use , Male , Vasculitis/drug therapy , Vasculitis/virology , Middle Aged , Female , Aged , Hepacivirus/drug effects , Ribavirin/therapeutic use , Sofosbuvir/therapeutic use , Imidazoles/therapeutic use , Valine/analogs & derivatives , Valine/therapeutic use , Pyrrolidines/therapeutic use , B-Cell Activating Factor , Interferon-alpha/therapeutic use , Drug Therapy, Combination , Hepatitis C/drug therapy , Hepatitis C/complications , Hepatitis C/virology , Treatment Outcome , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/virology , Carbamates
6.
Pharmacol Res Perspect ; 12(3): e1215, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38807350

ABSTRACT

Cardiovascular disorders are the leading cause of death in the world. Many organ diseases (kidney, heart, and brain) are substantially more prone to develop in people with hypertension. In the treatment of hypertension, first-line medications are recommended, while imidazoline receptor agonists are not first-line antihypertensives. Our goal was to conduct a network meta-analysis to assess the efficacy and safety of imidazoline receptor agonists. The meta-analysis was performed following the PRISMA guidelines using the PICOS format, considering the CONSORT recommendations. Studies were collected from four databases: PubMed, Cochrane Library, Web of Science, and Embase. A total of 5960 articles were found. After filtering, 27 studies remained eligible for network meta-analysis. Moxonidine reduced blood pressure in sitting position statistically significantly after 8 weeks of treatment (SBP MD: 23.80; 95% CI: 17.45-30.15; DBP MD: 10.90; 95% CI: 8.45-13.35) compared to placebo. Moreover, moxonidine reduced blood pressure more effectively than enalapril; however, this difference was not significant (SBP MD: 3.10; 95% CI: -2.60-8.80; DBP MD: 1.30; 95% CI: -1.25-3.85). Dry mouth was experienced as a side effect in the case of all imidazoline receptor agonists. After 8 weeks of treatment, the appearance of dry mouth was highest with clonidine (OR: 9.27 95% CI: 4.70-18.29) and lowest with rilmenidine (OR: 6.46 95% CI: 0.85-49.13) compared to placebo. Somnolence was less frequent with moxonidine compared to rilmenidine (OR: 0.63 95% CI: 0.17-2.31). Imidazoline receptor agonists were nearly as effective as the first-line drugs in the examined studies. However, their utility as antihypertensives is limited due to their side effects. As a result, they are not first-line antihypertensives and should not be used in monotherapy. However, in the case of resistant hypertension, they are a viable option. According to our findings, from the point of view of safety and efficacy, moxonidine appears to be the best choice among imidazoline receptor agonists.


Subject(s)
Antihypertensive Agents , Hypertension , Imidazoles , Imidazoline Receptors , Humans , Imidazoline Receptors/agonists , Antihypertensive Agents/therapeutic use , Antihypertensive Agents/adverse effects , Hypertension/drug therapy , Imidazoles/therapeutic use , Imidazoles/adverse effects , Blood Pressure/drug effects , Network Meta-Analysis , Treatment Outcome
7.
Ups J Med Sci ; 1292024.
Article in English | MEDLINE | ID: mdl-38716076

ABSTRACT

Background: Metastatic neuroendocrine carcinoma (NEC) is associated with short survival. Other than platinum-based chemotherapy, there is no clear standard regimen. Current guidelines suggest that combination treatment with BRAF-inhibitors should be considered for patients with BRAF V600E-mutated NEC. However, since only eight such patients have been reported in the literature, our object was to confirm the validity of this recommendation. Methods: This was a single-center retrospective cohort study conducted at Uppsala University Hospital. The included patients 1) had a histopathologically confirmed diagnosis of NEC, 2) were diagnosed between January 1st, 2018 and December 31st, 2023, 3) had tumor tissue genetically screened by a broad next-generation sequencing (NGS) panel, and 4) showed a tumor mutation for which there is a currently available targeted therapy. Results: We screened 48 patients diagnosed with NEC between January 1st, 2018 and December 31st, 2023. Twelve had been analyzed with a broad NGS-panel, and two had a targetable mutation. Both these patients harbored a BRAF V600E-mutated colon-NEC and were treated with BRAF- and MEK-inhibitors dabrafenib and trametinib in second-line. At first radiological evaluation (RECIST 1.1), both patients had a reduction of tumor size, which decreased by 31 and 40%. Both had short response periods, and their overall survival was 12 and 9 months. Conclusions: BRAF-mutated NEC is sensitive to treatment with BRAF- and MEK-inhibitor combination. These results further support that DNA sequencing should be considered as standard of care in NECs to screen for potential treatment targets.


Subject(s)
Carcinoma, Neuroendocrine , Oximes , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Pyridones , Pyrimidinones , Adult , Aged , Female , Humans , Male , Middle Aged , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/drug therapy , High-Throughput Nucleotide Sequencing , Imidazoles/therapeutic use , Imidazoles/administration & dosage , Mutation , Oximes/therapeutic use , Oximes/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Pyridones/therapeutic use , Pyridones/administration & dosage , Pyrimidinones/therapeutic use , Retrospective Studies , Treatment Outcome
9.
Cancer Immunol Immunother ; 73(7): 119, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38713205

ABSTRACT

BACKGROUND: The programmed death 1 inhibitor toripalimab plus the angio-immuno kinase inhibitor surufatinib showed a tolerable safety profile and preliminary efficacy in patients with advanced solid tumors in a phase I study. METHODS: This open-label, multi-cohort study in China enrolled patients with advanced solid tumors who had failed or were intolerable to standard treatment into tumor-specific cohorts. Patients received surufatinib (250 mg orally, once daily) plus toripalimab (240 mg intravenously, once every three weeks). Results for three cohorts (gastric/gastroesophageal junction [GC/GEJ] adenocarcinoma, esophageal squamous cell carcinoma [ESCC], and biliary tract carcinoma [BTC]) are reported here. The primary endpoint was investigator-assessed objective response rate (ORR) per Response Evaluation criteria in Solid Tumors version 1.1. RESULTS: Between December 17, 2019, and January 29, 2021, 60 patients were enrolled (GC/GEJ, n = 20; ESCC, n = 20; BTC, n = 20). At data cutoff (February 28, 2023), ORRs were 31.6%, 30.0%, and 11.1%, respectively. Median progression-free survival was 4.1, 2.7, and 2.9 months, respectively. Median overall survival was 13.7, 10.4, and 7.0 months, respectively. Overall, grade ≥ 3 treatment-related adverse events occurred in 28 (46.7%) patients. CONCLUSIONS: Surufatinib plus toripalimab showed promising antitumor activity and a tolerable safety profile in immunotherapy-naïve patients with GC/GEJ adenocarcinoma, ESCC, or BTC. These findings warrant further study in larger randomized trials comparing surufatinib plus toripalimab with standard therapies in these tumors. CLINICALTRIALS: gov NCT04169672.


Subject(s)
Adenocarcinoma , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Biliary Tract Neoplasms , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Male , Female , Middle Aged , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Biliary Tract Neoplasms/drug therapy , Biliary Tract Neoplasms/pathology , Biliary Tract Neoplasms/mortality , Adult , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/mortality , Stomach Neoplasms/drug therapy , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Esophagogastric Junction/pathology , Imidazoles/administration & dosage , Imidazoles/therapeutic use , Imidazoles/adverse effects , Aged, 80 and over , Cohort Studies
11.
BMJ Case Rep ; 17(5)2024 May 08.
Article in English | MEDLINE | ID: mdl-38719253

ABSTRACT

The combination therapy of v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) and mitogen-activated protein kinase kinase (MEK) inhibitors is approved for treating patients with BRAF V600E-positive tumours, including melanoma and lung cancer. Several case reports indicated autoimmune side effects associated with the use of BRAF and MEK inhibitors. Still, the effects of these drugs on the immune system were not fully elucidated. Here, we report a patient with large-vessel vasculitis diagnosed after initiation of treatment with dabrafenib and trametinib for BRAF V600E-positive metastatic lung adenocarcinoma. She was a never-smoker woman in her early 70s who presented with a chronic cough and was diagnosed with BRAF V600E-positive metastatic lung adenocarcinoma by transbronchial lung biopsy. She was successfully treated with prednisolone and methotrexate while BRAF and MEK inhibitors were continued. We should be careful about autoimmune diseases using BRAF and MEK inhibitors.


Subject(s)
Adenocarcinoma of Lung , Imidazoles , Lung Neoplasms , Oximes , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Pyridones , Pyrimidinones , Vasculitis , Humans , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Female , Pyridones/adverse effects , Pyridones/therapeutic use , Pyrimidinones/therapeutic use , Pyrimidinones/adverse effects , Lung Neoplasms/drug therapy , Aged , Adenocarcinoma of Lung/drug therapy , Imidazoles/adverse effects , Imidazoles/therapeutic use , Oximes/adverse effects , Oximes/therapeutic use , Vasculitis/chemically induced , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Prednisolone/therapeutic use , Methotrexate/therapeutic use , Methotrexate/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
12.
Nat Commun ; 15(1): 3882, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719809

ABSTRACT

In this randomized phase II clinical trial, we evaluated the effectiveness of adding the TLR agonists, poly-ICLC or resiquimod, to autologous tumor lysate-pulsed dendritic cell (ATL-DC) vaccination in patients with newly-diagnosed or recurrent WHO Grade III-IV malignant gliomas. The primary endpoints were to assess the most effective combination of vaccine and adjuvant in order to enhance the immune potency, along with safety. The combination of ATL-DC vaccination and TLR agonist was safe and found to enhance systemic immune responses, as indicated by increased interferon gene expression and changes in immune cell activation. Specifically, PD-1 expression increases on CD4+ T-cells, while CD38 and CD39 expression are reduced on CD8+ T cells, alongside an increase in monocytes. Poly-ICLC treatment amplifies the induction of interferon-induced genes in monocytes and T lymphocytes. Patients that exhibit higher interferon response gene expression demonstrate prolonged survival and delayed disease progression. These findings suggest that combining ATL-DC with poly-ICLC can induce a polarized interferon response in circulating monocytes and CD8+ T cells, which may represent an important blood biomarker for immunotherapy in this patient population.Trial Registration: ClinicalTrials.gov Identifier: NCT01204684.


Subject(s)
CD8-Positive T-Lymphocytes , Cancer Vaccines , Carboxymethylcellulose Sodium/analogs & derivatives , Dendritic Cells , Glioma , Interferons , Poly I-C , Polylysine/analogs & derivatives , Humans , Dendritic Cells/immunology , Dendritic Cells/drug effects , Glioma/immunology , Glioma/therapy , Female , Male , Middle Aged , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Poly I-C/administration & dosage , Poly I-C/pharmacology , Adult , Toll-Like Receptors/agonists , Imidazoles/pharmacology , Imidazoles/therapeutic use , Aged , Vaccination , Monocytes/immunology , Monocytes/drug effects , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/drug therapy , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , Immunotherapy/methods , Toll-Like Receptor Agonists
13.
Am J Hematol ; 99(8): 1576-1585, 2024 08.
Article in English | MEDLINE | ID: mdl-38727135

ABSTRACT

Ponatinib is a third-generation BCR::ABL1 tyrosine kinase inhibitor (TKI) with high potency against Philadelphia chromosome (Ph)-positive leukemias, including T315I-mutated disease, which is resistant to first- and second-generation TKIs. Ponatinib was approved for T315I-mutated chronic myeloid leukemia (CML), CML resistant/intolerant to ≥2 prior TKIs, advanced phase CML and Ph-positive acute lymphoblastic leukemia (ALL) where no other TKIs are indicated, and T315I-mutated CML and Ph-positive ALL. The response-based dosing of ponatinib in chronic phase CML (CP-CML) improved treatment tolerance and reduced the risk of toxicities, including cardiovascular risks. Ponatinib-based therapy also resulted in significantly better outcomes in frontline Ph-positive ALL compared with prior TKIs and is becoming a new standard of care in this setting. As the clinical development of third-generation TKIs and their rational combinations progresses, we envision further transformative changes in the treatment of CML and Ph-positive ALL.


Subject(s)
Fusion Proteins, bcr-abl , Imidazoles , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Protein Kinase Inhibitors , Pyridazines , Humans , Pyridazines/therapeutic use , Pyridazines/adverse effects , Imidazoles/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/therapeutic use , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm
14.
J Gynecol Oncol ; 35(4): e104, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38768941

ABSTRACT

Mutations in genes encoding for proteins along the RAS-RAF-MEK-ERK pathway have been detected in a variety of tumor entities including ovarian carcinomas. In the recent years, several inhibitors of this pathway have been developed, whose antitumor potential is currently being assessed in different clinical trials. Low grade serous ovarian carcinoma, is a rare gynecological tumor which shows favorable overall survival, compared to the general ovarian cancer population, but worrying resistance to conventional chemotherapies. The clinical behavior of low grade serous ovarian carcinoma reflects the different gene profile compared to high-grade serous carcinoma: KRAS/BRAF mutations. BRAF inhibitors as single agents were approved for the treatment of BRAF mutated tumors. Nevertheless, many patients face progressive disease. The understanding of the mechanisms of resistance to BRAF inhibitors therapy and preclinical studies showing that BRAF and mitogen-activated protein kinase kinase (MEK) inhibitors combined therapy delays the onset of resistance compared to BRAF inhibitor single agent, led to the clinical investigation of combined therapy. The aim of this paper is to review the efficacy and safety of the combination of BRAF plus MEK inhibitors on ovarian carcinomas, in particularly focusing on low grade serous ovarian carcinoma.


Subject(s)
Cystadenocarcinoma, Serous , Ovarian Neoplasms , Proto-Oncogene Proteins B-raf , Humans , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Molecular Targeted Therapy , Neoplasm Grading , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , MAP Kinase Signaling System/drug effects , Sulfonamides/therapeutic use , Imidazoles/therapeutic use , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Oximes
15.
J Assoc Physicians India ; 72(3): 75-78, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38736121

ABSTRACT

For >3 decades now, angiotensin receptor blockers (ARB) have been used in the management of hypertension (HTN) and HTN-related cardiovascular (CV) diseases. Olmesartan medoxomil (OLM) is an angiotensin II type 1 (AT1) receptor antagonist (or blocker) that binds tightly to the AT1 receptor with long-lasting efficacy over the 24-hour period and safety demonstrated in several trials. It is well tolerated and effective in reducing blood pressure (BP) in mono and combination therapy with thiazide diuretics or calcium channel blockers across a wide range of patient subgroups. The effectiveness and safety of OLM-based combination therapies have good and tolerable profiles with high adherence in the fixed single-pill formulation. Consistent antihypertensive efficacy and good tolerability when used as monotherapy or as a combined therapy make OLM a valuable treatment option for adults with HTN. In this review, we discuss the important clinical implications of OLM as an optimal choice as monotherapy and combination therapy in managing patients with HTN.


Subject(s)
Angiotensin II Type 1 Receptor Blockers , Antihypertensive Agents , Blood Pressure , Drug Therapy, Combination , Hypertension , Imidazoles , Humans , Hypertension/drug therapy , Antihypertensive Agents/therapeutic use , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Blood Pressure/drug effects , Imidazoles/therapeutic use , Tetrazoles/therapeutic use , Olmesartan Medoxomil/therapeutic use
16.
Thorac Cancer ; 15(18): 1454-1456, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38766698

ABSTRACT

Although dabrafenib plus trametinib has been approved for BRAF V600E mutation positive advanced non-small cell lung cancer (NSCLC), data on its efficacy against uncommon BRAF mutations are still limited due to their rare frequency. We report a case of 70-year-old woman with BRAF V600_W604 deletion-insertion R-positive stage IVA lung adenocarcinoma, who was successfully treated with dabrafenib plus trametinib. Herein, we discuss the oncogenic role of uncommon BRAF mutations and highlight the importance of performing comprehensive genomic profiling on patients without any targetable gene alterations in companion diagnostics.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Non-Small-Cell Lung , Imidazoles , Lung Neoplasms , Oximes , Proto-Oncogene Proteins B-raf , Pyridones , Pyrimidinones , Aged , Female , Humans , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Imidazoles/therapeutic use , Imidazoles/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Oximes/therapeutic use , Oximes/administration & dosage , Proto-Oncogene Proteins B-raf/genetics , Pyridones/therapeutic use , Pyridones/administration & dosage , Pyrimidinones/therapeutic use , Pyrimidinones/administration & dosage
17.
J Am Chem Soc ; 146(27): 18241-18252, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38815248

ABSTRACT

Aberrant DNA repair is a hallmark of cancer, and many tumors display reduced DNA repair capacities that sensitize them to genotoxins. Here, we demonstrate that the differential DNA repair capacities of healthy and transformed tissue may be exploited to obtain highly selective chemotherapies. We show that the novel N3-(2-fluoroethyl)imidazotetrazine "KL-50" is a selective toxin toward tumors that lack the DNA repair protein O6-methylguanine-DNA-methyltransferase (MGMT), which reverses the formation of O6-alkylguanine lesions. We establish that KL-50 generates DNA interstrand cross-links (ICLs) by a multistep process comprising DNA alkylation to generate an O6-(2-fluoroethyl)guanine (O6FEtG) lesion, slow unimolecular displacement of fluoride to form an N1,O6-ethanoguanine (N1,O6EtG) intermediate, and ring-opening by the adjacent cytidine. The slow rate of N1,O6EtG formation allows healthy cells expressing MGMT to reverse the initial O6FEtG lesion before it evolves to N1,O6EtG, thereby suppressing the formation of toxic DNA-MGMT cross-links and reducing the amount of DNA ICLs generated in healthy cells. In contrast, O6-(2-chloroethyl)guanine lesions produced by agents such as lomustine and the N3-(2-chloroethyl)imidazotetrazine mitozolomide rapidly evolve to N1,O6EtG, resulting in the formation of DNA-MGMT cross-links and DNA ICLs in healthy tissue. These studies suggest that careful consideration of the rates of chemical DNA modification and biochemical DNA repair may lead to the identification of other tumor-specific genotoxic agents.


Subject(s)
Brain Neoplasms , Drug Resistance, Neoplasm , Humans , Drug Resistance, Neoplasm/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , DNA Repair/drug effects , O(6)-Methylguanine-DNA Methyltransferase/metabolism , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Imidazoles/chemistry , Imidazoles/pharmacology , Imidazoles/therapeutic use
18.
JAMA ; 331(21): 1814-1823, 2024 06 04.
Article in English | MEDLINE | ID: mdl-38722621

ABSTRACT

Importance: In newly diagnosed Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL), disease progression due to acquired resistance to first- or second-generation BCR::ABL1 tyrosine kinase inhibitors is common. Ponatinib inhibits BCR::ABL1 and all single-mutation variants, including T315I. Objective: To compare frontline ponatinib vs imatinib in adults with newly diagnosed Ph+ ALL. Design, Setting, and Participants: Global registrational, phase 3, open-label trial in adults aged 18 years or older with newly diagnosed Ph+ ALL. From January 2019 to May 2022, eligible patients at 77 sites were randomized 2:1 to ponatinib (30 mg/d) or imatinib (600 mg/d) with reduced-intensity chemotherapy, followed by single-agent ponatinib or imatinib after the cycle 20 phase of the trial. The last date of follow-up for this analysis was August 12, 2022. Intervention: Patients received ponatinib, 30 mg/d, or imatinib, 600 mg/d, with reduced-intensity chemotherapy, followed by single-agent ponatinib or imatinib after cycle 20. The ponatinib dose was reduced to 15 mg on achievement of minimal residual disease-(MRD) negative complete remission. Main Outcomes and Measures: The primary end point of this interim analysis was MRD-negative complete remission (≤0.01% BCR::ABL1 [MR4] centrally assessed by reverse transcriptase-quantitative polymerase chain reaction), with complete remission maintained for at least 4 weeks at the end of cycle 3. The key secondary end point was event-free survival. Results: Of 245 patients randomized (median age, 54 years; 133 [54.3%] female), 232 (ponatinib, n = 154; imatinib, n = 78) who had p190 or p210 dominant isoforms verified by the central laboratory were analyzed for the primary end point. The MRD-negative complete remission rate (primary end point) was significantly higher with ponatinib (34.4% [53/154]) vs imatinib (16.7% [13/78]) (risk difference, 0.18 [95% CI, 0.06-0.29]; P = .002). At the data cutoff, event-free survival had not met the prespecified number of events. Median event-free survival was not reached in the ponatinib group and was 29 months in the imatinib group. The most common adverse events were similar between treatment groups. Arterial occlusive events were infrequent and comparable between groups (ponatinib, 2.5%; imatinib, 1.2%). Conclusions and Relevance: Ponatinib demonstrated a superior rate of MRD-negative complete remission at the end of induction vs imatinib when combined with reduced-intensity chemotherapy in adults with newly diagnosed Ph+ ALL. The safety profile of ponatinib was comparable with imatinib. Trial Registration: ClinicalTrials.gov Identifier: NCT03589326.


Subject(s)
Antineoplastic Agents , Imatinib Mesylate , Imidazoles , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Pyridazines , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Fusion Proteins, bcr-abl/genetics , Imatinib Mesylate/therapeutic use , Imatinib Mesylate/adverse effects , Imidazoles/therapeutic use , Imidazoles/adverse effects , Imidazoles/administration & dosage , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Progression-Free Survival , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/adverse effects , Pyridazines/therapeutic use , Pyridazines/adverse effects , Remission Induction , Adolescent
19.
J Med Virol ; 96(5): e29678, 2024 May.
Article in English | MEDLINE | ID: mdl-38751128

ABSTRACT

Death due to severe influenza is usually a fatal complication of a dysregulated immune response more than the acute virulence of an infectious agent. Although spleen tyrosine kinase (SYK) as a critical immune signaling molecule and therapeutic target plays roles in airway inflammation and acute lung injury, the role of SYK in influenza virus infection is not clear. Here, we investigated the antiviral and anti-inflammatory effects of SYK inhibitor R406 on influenza infection through a coculture model of human alveolar epithelial (A549) and macrophage (THP-1) cell lines and mouse model. The results showed that R406 treatment increased the viability of A549 and decreased the pathogenicity and mortality of lethal influenza virus in mice with influenza A infection, decreased levels of intracellular signaling molecules under the condition of inflammation during influenza virus infection. Combination therapy with oseltamivir further ameliorated histopathological damage in the lungs of mice and further delayed the initial time to death compared with R406 treatment alone. This study demonstrated that phosphorylation of SYK is involved in the pathogenesis of influenza, and R406 has antiviral and anti-inflammatory effects on the treatment of the disease, which may be realized through multiple pathways, including the already reported SYK/STAT/IFNs-mediated antiviral pathway, as well as TNF-α/SYK- and SYK/Akt-based immunomodulation pathway.


Subject(s)
Anti-Inflammatory Agents , Antiviral Agents , Disease Models, Animal , Orthomyxoviridae Infections , Oxazines , Syk Kinase , Animals , Humans , Syk Kinase/antagonists & inhibitors , Mice , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Oxazines/pharmacology , Oxazines/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Imidazoles/pharmacology , Imidazoles/therapeutic use , Lung/pathology , Lung/virology , Lung/drug effects , Lung/immunology , A549 Cells , Influenza A virus/drug effects , Mice, Inbred BALB C , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Influenza, Human/drug therapy , Influenza, Human/immunology , THP-1 Cells , Female , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL