Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.058
Filter
1.
Nature ; 633(8031): 743, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39304757
2.
Front Immunol ; 15: 1408892, 2024.
Article in English | MEDLINE | ID: mdl-39234256

ABSTRACT

Introduction: Ciltacabtagene autoleucel (cilta-cel) is a chimeric antigen receptor T-cell therapy approved for patients with relapsed/refractory multiple myeloma (RRMM). In the phase 3 trial, CARTITUDE-4 (NCT04181827), cilta-cel demonstrated improved efficacy vs. standard of care (SOC; daratumumab plus pomalidomide and dexamethasone [DPd] or pomalidomide plus bortezomib and dexamethasone [PVd]) with a ≥ complete response (≥CR) rate of 73.1% vs. 21.8%. Methods: A cost-per-responder model was developed to assess the value of cilta-cel and SOC (87% DPd and 13% PVd) based on the CARTITUDE-4 trial data from a US mixed payer perspective (76.7% commercial, 23.3% Medicare). The model was developed using progression-free survival (PFS), overall survival (OS), and ≥CR endpoints from CARTITUDE-4 over a period of 25.4 months. Inpatient stays, outpatient visits, drug acquisition, administration, and monitoring costs were included. The base-case model assumed an inpatient setting for each cilta-cel infusion; another scenario included 30% outpatient and 70% inpatient infusions. Costs of managing grade 3-4 adverse events (AEs) and grade 1-4 cytokine release syndrome and neurotoxicity were included. Subsequent therapy costs were incurred after disease progression; terminal care costs were considered upon death events. Outcomes included total cost per treated patient, total cost per complete responder, and cost per month in PFS between cilta-cel and SOC. Costs were adjusted to 2024 US dollars. Results: Total cost per treated patient, total cost per complete responder, and total cost per month in PFS were estimated at $704,641, $963,941, and $30,978 for cilta-cel, respectively, and $840,730, $3,856,559, and $42,520 for SOC over the 25.4-month period. Cost drivers included treatment acquisition costs before progression and subsequent treatment costs ($451,318 and $111,637 for cilta-cel; $529,795 and $265,167 for SOC). A scenario analysis in which 30% of patients received an outpatient infusion (assuming the same payer mix) showed a lower cost per complete responder for cilta-cel ($956,523) than those with an infusion in the inpatient setting exclusively. Discussion: This analysis estimated that cost per treated patient, cost per complete responder, and cost per month in PFS for cilta-cel were remarkably lower than for DPd or PVd, highlighting the substantial clinical and economic benefit of cilta-cel for patients with RRMM.


Subject(s)
Cost-Benefit Analysis , Immunotherapy, Adoptive , Lenalidomide , Multiple Myeloma , Thalidomide , Humans , Multiple Myeloma/therapy , Multiple Myeloma/mortality , Multiple Myeloma/drug therapy , Lenalidomide/therapeutic use , Lenalidomide/administration & dosage , Immunotherapy, Adoptive/economics , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Thalidomide/economics , Thalidomide/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/economics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dexamethasone/therapeutic use , Dexamethasone/administration & dosage , Receptors, Chimeric Antigen/therapeutic use , Receptors, Chimeric Antigen/immunology , Male , Female , Bortezomib/therapeutic use , Bortezomib/administration & dosage , Middle Aged , Progression-Free Survival , Treatment Outcome , Aged , Drug Resistance, Neoplasm , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/economics
4.
N Engl J Med ; 391(11): 1015-1027, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39292928

ABSTRACT

BACKGROUND: Hereditary hemorrhagic telangiectasia (HHT) is characterized by extensive telangiectasias and arteriovenous malformations. The primary clinical manifestation is epistaxis that results in iron-deficiency anemia and reduced health-related quality of life. METHODS: We conducted a randomized, placebo-controlled trial to evaluate the safety and efficacy of pomalidomide for the treatment of HHT. We randomly assigned patients, in a 2:1 ratio, to receive pomalidomide at a dose of 4 mg daily or matching placebo for 24 weeks. The primary outcome was the change from baseline through week 24 in the Epistaxis Severity Score (a validated bleeding score in HHT; range, 0 to 10, with higher scores indicating worse bleeding). A reduction of 0.71 points or more is considered clinically significant. A key secondary outcome was the HHT-specific quality-of-life score (range, 0 to 16, with higher scores indicating more limitations). RESULTS: The trial was closed to enrollment in June 2023 after a planned interim analysis met a prespecified threshold for efficacy. A total of 144 patients underwent randomization; 95 patients were assigned to receive pomalidomide and 49 to receive placebo. The baseline mean (±SD) Epistaxis Severity Score was 5.0±1.5, a finding consistent with moderate-to-severe epistaxis. At 24 weeks, the mean difference between the pomalidomide group and the placebo group in the change from baseline in the Epistaxis Severity Score was -0.94 points (95% confidence interval [CI], -1.57 to -0.31; P = 0.004). The mean difference in the changes in the HHT-specific quality-of-life score between the groups was -1.4 points (95% CI, -2.6 to -0.3). Adverse events that were more common in the pomalidomide group than in the placebo group included neutropenia, constipation, and rash. CONCLUSIONS: Among patients with HHT, pomalidomide treatment resulted in a significant, clinically relevant reduction in epistaxis severity. No unexpected safety signals were identified. (Funded by the National Heart, Lung, and Blood Institute; PATH-HHT Clinicaltrials.gov number, NCT03910244).


Subject(s)
Epistaxis , Telangiectasia, Hereditary Hemorrhagic , Thalidomide , Aged , Female , Humans , Male , Middle Aged , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Double-Blind Method , Epistaxis/diagnosis , Epistaxis/drug therapy , Epistaxis/etiology , Epistaxis/psychology , Quality of Life , Severity of Illness Index , Telangiectasia, Hereditary Hemorrhagic/complications , Telangiectasia, Hereditary Hemorrhagic/drug therapy , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/analogs & derivatives , Treatment Outcome , Neutropenia/chemically induced , Neutropenia/epidemiology , Constipation/chemically induced , Constipation/epidemiology , Drug Eruptions/epidemiology , Drug Eruptions/etiology
5.
BMJ Case Rep ; 17(9)2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266024

ABSTRACT

Two men in their 60s and 40s were diagnosed with erythema nodosum leprosum based on the development of recurrent painful ulcers and nodules, respectively, for the previous 6 months. Thalidomide 100 mg four times a day, along with MB-MDT, was started in both patients. Both patients experienced severe dizziness on rising from a seated posture soon after initiation of thalidomide and a decrease in blood pressure and heart rate. Cardiovascular/neurology examination and routine blood investigations were normal. An autonomic nervous system assessment indicated bradycardia, postural hypotension and decreased cardiac autonomic function. The dosage of thalidomide was then gradually reduced over 4-5 days to 100 mg/day following a suspicion that thalidomide was the cause of postural hypotension. The dizziness subsided, and blood pressure and heart rate returned to normal.We concluded that thalidomide was the culprit behind bradycardia and dose- dependent postural hypotension.


Subject(s)
Bradycardia , Erythema Nodosum , Hypotension, Orthostatic , Thalidomide , Humans , Thalidomide/adverse effects , Thalidomide/therapeutic use , Thalidomide/administration & dosage , Bradycardia/chemically induced , Bradycardia/drug therapy , Male , Erythema Nodosum/drug therapy , Hypotension, Orthostatic/drug therapy , Hypotension, Orthostatic/chemically induced , Adult , Middle Aged , Leprosy, Lepromatous/drug therapy , Leprosy, Lepromatous/complications , Leprostatic Agents/adverse effects , Leprostatic Agents/therapeutic use , Leprostatic Agents/administration & dosage
6.
Blood Cancer J ; 14(1): 155, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261451

ABSTRACT

Given the early use of triplet and quadruplet regimens, most patients with multiple myeloma (MM) will be exposed and/or refractory to PIs, IMiDs, and anti-CD38 mAbs after first- or second-line treatment. Effective treatment for this group of triple class exposed/refractory (TCE/R) patients is crucial. Here we present a post-hoc subgroup analysis of TCE/R patients treated on the ALGONQUIN study of belantamab mafodotin plus pomalidomide-dexamethasone (belamaf-Pd) for relapsed MM. Of the 99 patients treated on the ALGONQUIN study, 69 were TCE and 56 were TCR and were included in this analysis. Patients had a median of three prior lines of therapy. The ORR was 86.4% in TCE patients and 84.9% in TCR patients, with ≥ very good partial response rates of 64% and 68% respectively. The median progression free survival was 18.3 months in TCE patients and 19.6 months in TCR patients, with overall survival not yet reached and 34.4 months, respectively for TCE and TCR patients. No new safety signals were identified. The most common Grade ≥ 3 AEs were keratopathy (48%), decreased visual acuity (42%), neutropenia (36%), thrombocytopenia (27%), and infection (25%). In this subgroup analysis of the ALGONQUIN study, patients with TCE/TCR disease treated with belamaf-Pd achieved high clinical response rates with durable remissions, comparable to other novel therapeutics in this space.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Dexamethasone , Multiple Myeloma , Thalidomide , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Dexamethasone/therapeutic use , Dexamethasone/administration & dosage , Male , Female , Middle Aged , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Thalidomide/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Adult , Boron Compounds/therapeutic use , Boron Compounds/administration & dosage , Aged, 80 and over , Neoplasm Recurrence, Local/drug therapy , Drug Resistance, Neoplasm
7.
Front Immunol ; 15: 1459185, 2024.
Article in English | MEDLINE | ID: mdl-39170613

ABSTRACT

Psoriatic disease, encompassing both psoriasis (Pso) and psoriatic arthritis (PsA), is closely intertwined with a significantly elevated risk of developing cardiovascular diseases. This connection is further compounded by a higher prevalence of cardiometabolic comorbidities, including type 2 diabetes, obesity, insulin resistance, arterial hypertension, and dysregulated lipid profiles. These comorbidities exceed the rates seen in the general population and compound the potential for increased mortality among those living with this condition. Recognizing the heightened cardiometabolic risk inherent in psoriatic disease necessitates a fundamental shift in the treatment paradigm. It is no longer sufficient to focus solely on mitigating inflammation. Instead, there is an urgent need to address and effectively manage the metabolic parameters that have a substantial impact on cardiovascular health. Within this context, apremilast emerges as a pivotal treatment option for psoriatic disease. What sets apremilast apart is its dual-action potential, addressing not only inflammation but also the critical metabolic parameters. This comprehensive treatment approach opens up new opportunities to improve the well-being of people living with psoriatic disease. This review delves into the multifaceted aspects involved in the development of cardiovascular disease and its intricate association with psoriatic disease. We then provide an in-depth exploration of the pleiotropic effects of apremilast, highlighting its potential to simultaneously mitigate metabolic complications and inflammation in individuals affected by these conditions.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Cardiovascular Diseases , Psoriasis , Thalidomide , Humans , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/prevention & control , Psoriasis/drug therapy , Psoriasis/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthritis, Psoriatic/drug therapy , Animals
8.
J Comp Eff Res ; 13(9): e240080, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39162049

ABSTRACT

Aim: The phase III randomized controlled trial (RCT) CARTITUDE-4 (NCT04181827) demonstrated superiority of CARVYKTI (ciltacabtagene autoleucel; cilta-cel) over daratumumab, pomalidomide and dexamethasone (DPd) and pomalidomide, bortezomib and dexamethasone (PVd) for relapsed/refractory multiple myeloma (RRMM) patients who have received one to three prior line(s) of therapy (LOT[s]) including an immunomodulatory agent and a proteasome inhibitor, and are refractory to lenalidomide. These analyses estimate the relative efficacy between cilta-cel and other common treatment regimens, for which no direct comparative evidence is available. Materials & methods: Patient data were available from the CARTITUDE-4, CASTOR, CANDOR and APOLLO RCTs. Imbalances between cohorts on key patient characteristics were adjusted for using inverse probability of treatment weighting (IPTW). Relative efficacies were estimated with response rate ratios (RRs) and 95% confidence intervals (CIs) for overall response rate (ORR), very good partial response or better rate (≥VGPR) and complete response or better rate (≥CR), and with hazard ratios (HRs) and 95% CIs for progression-free survival (PFS). Sensitivity analyses using different analytical methods and additional covariates were explored. Results: Key characteristics were well balanced across cohorts after IPTW. Cilta-cel showed statistically significant benefit in PFS (HRs: 0.11-0.51), ≥VGPR (RRs: 1.51-5.13) and ≥CR (RRs: 2.90-35.24) versus all comparators, and statistically significant improvements in ORR over most comparator regimens (RRs: 1.22-1.90). Results were consistent across sensitivity analyses. Conclusion: Cilta-cel demonstrated benefit over other common treatment regimens, highlighting its potential to become a new standard of care option for lenalidomide-refractory RRMM patients with one to three prior LOT(s). These comparisons help to demonstrate the improved efficacy of cilta-cel in countries where the standard of care may differ from DPd/PVd.


Subject(s)
Dexamethasone , Lenalidomide , Multiple Myeloma , Thalidomide , Humans , Multiple Myeloma/drug therapy , Lenalidomide/therapeutic use , Male , Female , Middle Aged , Aged , Dexamethasone/therapeutic use , Thalidomide/therapeutic use , Thalidomide/analogs & derivatives , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Immunotherapy, Adoptive/methods , Bortezomib/therapeutic use , Treatment Outcome , Antibodies, Monoclonal
9.
Medicine (Baltimore) ; 103(33): e39174, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39151531

ABSTRACT

RATIONALE: Light chain proximal tubulopathy (LCPT) is a rare form of renal impairment associated with multiple myeloma (MM). LCPT is caused by inclusions formed of free light chains that are typically crystalline, but can also be noncrystalline structures. PATIENT CONCERNS: A 62-year-old man was hospitalized for the investigation of abnormal urine test results lasting for 1 year and kidney-function abnormalities persisting for more than 1 month. DIAGNOSES: Noncrystalline LCPT and MM. INTERVENTIONS: The patient was treated with the lenalidomide, bortezomib, and dexamethasone and pomalidomide, bortezomib, and dexamethasone chemotherapy regimens. OUTCOMES: Complete remission of MM was achieved, and the patient's renal function returned to normal. LESSONS: This case report highlights the importance of renal pathology in the diagnosis of patients with unexplained chronic kidney disease and proteinuria.


Subject(s)
Multiple Myeloma , Humans , Male , Middle Aged , Multiple Myeloma/complications , Multiple Myeloma/drug therapy , Multiple Myeloma/diagnosis , Multiple Myeloma/pathology , Immunoglobulin Light Chains/urine , Kidney Tubules, Proximal/pathology , Dexamethasone/therapeutic use , Inclusion Bodies/pathology , Thalidomide/therapeutic use , Thalidomide/analogs & derivatives , Lenalidomide/therapeutic use , Lenalidomide/administration & dosage , Bortezomib/therapeutic use
10.
Nat Commun ; 15(1): 7388, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39191755

ABSTRACT

Functional blockade of the transforming growth factor-beta (TGFß) signalling pathway improves the efficacy of cytotoxic and immunotherapies. Here, we conducted a phase 1b study (ClinicalTrials.gov., NCT03143985) to determine the primary endpoints of safety, tolerability, and maximal tolerated dose (200 mg twice daily) for the orally-available TGFß type I receptor kinase inhibitor vactosertib in combination with pomalidomide in relapsed and/or refractory multiple myeloma (RRMM) patients who had received ≥2 lines of chemoimmunotherapy. Secondary endpoints demonstrated sustained clinical responses, favorable pharmacokinetic parameters and a 6-month progression-free survival of 82%. Vactosertib combined with pomalidomide was well-tolerated at all dose levels and displayed a manageable adverse event profile. Exploratory analysis indicated that vactosertib co-treatment with pomalidomide also reduced TGFß levels in patient bone marrow as well as the level of CD8+ T-cells that expressed the immunoinhibitory marker PD-1. In vitro experiments indicated that vactosertib+pomalidomide co-treatment decreased the viability of MM cell lines and patient tumor cells, and increased CD8+ T-cell cytotoxic activity. Vactosertib is a safe therapeutic that demonstrates tumor-intrinsic activity and can overcome immunosuppressive challenges within the tumor microenvironment to reinvigorate T-cell fitness. Vactosertib offers promise to improve immunotherapeutic responses in heavily-pretreated MM patients refractory to conventional agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Multiple Myeloma , Receptor, Transforming Growth Factor-beta Type I , Thalidomide , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Thalidomide/analogs & derivatives , Thalidomide/administration & dosage , Thalidomide/therapeutic use , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/metabolism , Middle Aged , Female , Male , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Transforming Growth Factor beta/metabolism , Neoplasm Recurrence, Local/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Adult , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Aniline Compounds , Triazoles
11.
Lupus ; 33(10): 1109-1115, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39118350

ABSTRACT

OBJECTIVE: Thalidomide is an effective medication for refractory mucocutaneous lesions of systemic lupus erythematosus (SLE) and can treat arthritis in some autoimmune diseases, but it has some adverse reactions. Recently, the effectiveness of tofacitinib in treating mucocutaneous lesions of SLE has been reported. We aimed to compare the efficacy and safety of tofacitinib with thalidomide in treating mucocutaneous and musculoskeletal lesions in patients with SLE. METHODS: This study was a real-world cohort study based on the Chinese SLE Treatment and Research group (CSTAR) registry. SLE patients who manifested mucocutaneous and/or musculoskeletal symptoms and were prescribed tofacitinib or thalidomide were included. We retrospectively conducted comparisons between the tofacitinib and thalidomide groups regarding clinical improvements, SLE disease activity, serological indicators, glucocorticoid doses, and adverse events at the 1, 3, and 6-months time points. RESULTS: At 3 and 6 months, the tofacitinib group exhibited a higher proportion of patients with improvement in mucocutaneous and musculoskeletal issues. Additionally, a greater percentage of patients in the tofacitinib group achieved remission or a low disease activity state (LLDAS) at these time points. No significant serological improvements were observed in either the tofacitinib or thalidomide groups. Fewer adverse events were observed in the tofacitinib group than in the thalidomide group. CONCLUSIONS: Tofacitinib might be superior to thalidomide in the improvement of mucocutaneous and musculoskeletal lesions in SLE, and had a good safety profile.


Subject(s)
Lupus Erythematosus, Systemic , Piperidines , Pyrimidines , Thalidomide , Humans , Lupus Erythematosus, Systemic/drug therapy , Piperidines/therapeutic use , Piperidines/adverse effects , Thalidomide/therapeutic use , Thalidomide/adverse effects , Pyrimidines/therapeutic use , Pyrimidines/adverse effects , Female , Retrospective Studies , Male , Adult , Treatment Outcome , Middle Aged , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/adverse effects , Registries , China , Cohort Studies
12.
J Agric Food Chem ; 72(34): 18809-18815, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39145990

ABSTRACT

Novel approaches for pest control are essential to ensure a sufficient food supply for the growing global population. The development of new insecticides must meet rigorous regulatory requirements for safety and address the resistance issues of existing insecticides. Proteolysis-targeting chimeras (PROTACs), originally developed for human diseases, show promise in agriculture. They offer innovative insecticides tailored to overcome resistance, opening avenues for agricultural applications. In this study, we developed small-molecule degraders by incorporating pomalidomide as an E3 ligand. These degraders were linked to a ligand (spirotetratmat enol) targeting the ACC protein through a flexible chain, aiming to achieve the efficient control of insects. Compounds 9a-9d were designed, synthesized, and evaluated for biological activities and mechanisms. Among them, 9b exhibited superior potency against Aphis craccivora (LC50 = 107.8 µg mL-1) compared to others and effectively degraded ACC proteins through the ubiquitin-proteasome system. These findings highlight the potential of utilizing PROTAC-based approaches in the development of insecticides for efficient pest control.


Subject(s)
Acetyl-CoA Carboxylase , Insecticides , Proteolysis , Insecticides/chemistry , Insecticides/pharmacology , Animals , Acetyl-CoA Carboxylase/metabolism , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/antagonists & inhibitors , Acetyl-CoA Carboxylase/chemistry , Insect Proteins/metabolism , Insect Proteins/genetics , Insect Proteins/chemistry , Drug Design , Thalidomide/chemistry , Thalidomide/analogs & derivatives , Thalidomide/pharmacology
13.
Acta Oncol ; 63: 689-700, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143719

ABSTRACT

BACKGROUND AND PURPOSE: The poor delivery of drugs to infiltrating tumor cells contributes to therapeutic failure in glioblastoma. During the early phase of an anti-angiogenic treatment, a remodeling of the tumor vasculature could occur, leading to a more functional vessel network that could enhance drug delivery. However, the restructuration of blood vessels could increase the proportion of normal endothelial cells that could be a barrier for the free diffusion of drugs. The net balance, in favor or not, of a better delivery of compounds during the course of an antiangiogenic treatment remains to be established. This study explored whether cediranib and thalidomide could modulate perfusion and vessel permeability in the brain U87 tumor mouse model. METHODS: The dynamic evolution of the diffusion of agents outside the tumor core using the fluorescent dye Evans Blue in histology and Gd-DOTA using dynamic contrast-enhanced (DCE)-MRI. CD31 labelling of endothelial cells was used to measure the vascular density. RESULTS AND INTERPRETATION: Cediranib and thalidomide effectively reduced tumor size over time. The accessibility of Evans Blue outside the tumor core continuously decreased over time. The vascular density was significantly decreased after treatment while the proportion of normal vessels remained unchanged over time. In contrast to histological studies, DCE-MRI did not tackle any significant change in hemodynamic parameters, in the core or margins of the tumor, whatever the parameter used or the pharmacokinetic model used. While cediranib and thalidomide were effective in decreasing the tumor size, they were ineffective in transiently increasing the delivery of agents in the core and the margins of the tumor.


Subject(s)
Angiogenesis Inhibitors , Brain Neoplasms , Glioblastoma , Quinazolines , Thalidomide , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/blood supply , Thalidomide/pharmacology , Thalidomide/therapeutic use , Animals , Angiogenesis Inhibitors/pharmacology , Mice , Quinazolines/pharmacology , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use , Humans , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Magnetic Resonance Imaging , Xenograft Model Antitumor Assays , Capillary Permeability/drug effects , Mice, Nude , Cell Line, Tumor , Indoles
14.
J Med Chem ; 67(17): 15168-15198, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39189384

ABSTRACT

Yes-associated protein (YAP) is a key oncogene in the Hippo tumor suppression pathway, historically challenging to target due to its intrinsically disordered nature. Leveraging recent advances in high-throughput screening that identified several YAP binders, we employed proteolysis-targeting chimera technology to develop a series of YAP degraders. Utilizing NSC682769, a known YAP binder, linked with VHL ligand 2 or pomalidomide via diverse linkers, we synthesized degraders including YZ-6. This degrader not only recruits the E3 ligase VHL for the rapid and sustained degradation of YAP but also effectively inhibits its nuclear localization, curtailing YAP/TEAD-mediated transcription in cancer cell lines such as NCI-H226 and Huh7. This dual action significantly diminishes YAP's oncogenic activity, contributing to the potent antiproliferative effects observed both in vitro and in a Huh7 xenograft mouse model. These results underscore the potential of PROTAC-mediated YAP degradation as a strategy for treating YAP-driven cancers.


Subject(s)
Adaptor Proteins, Signal Transducing , Proteolysis , Transcription Factors , YAP-Signaling Proteins , Humans , Proteolysis/drug effects , Animals , Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , Cell Line, Tumor , Mice , Adaptor Proteins, Signal Transducing/metabolism , YAP-Signaling Proteins/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/chemistry , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Thalidomide/chemical synthesis , Thalidomide/chemistry , Cell Proliferation/drug effects , Mice, Nude , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Xenograft Model Antitumor Assays , Proteolysis Targeting Chimera
15.
Front Immunol ; 15: 1408116, 2024.
Article in English | MEDLINE | ID: mdl-39139566

ABSTRACT

Pemphigus foliaceus (PF) is a superficial form of pemphigus. Treatment options for PF resemble pemphigus vulgaris, including glucocorticosteroids, immunosuppressive agents and rituximab et al. These treatment approaches can effectively improve the condition but may also be accompanied by high risks of side effects. Therefore, it is crucial to find a safe and effective treatment options for patients with PF. It will not only benefit/be necessary for patients who refuse glucocorticosteroids or immunosuppressive agents treatments, but also for patients who cannot be treated with glucocorticosteroids or immunosuppressive agents. Herein, we reported a case of PF that was treated with apremilast without systemic glucocorticosteroids or immunosuppressive agents. A 54-year-old woman presented with itchy erythema and erosions on the trunk for more than 1 month. The patient applied mometasonefuroate cream without improvement for a duration of two weeks. The past history of diabetes mellitus and atrophic gastritis was reported. Physical examination revealed scattered erythematous macules and erosions on the trunk. No mucosal involvement was observed. The condition was assessed by the pemphigus disease area index and numerical rating scale, with baseline scores of 7 and 8, respectively. Histopathological examination showed acantholysis and intraepithelial blister. Direct immunofluorescence revealed the presence of IgG and Complement 3 deposition between the acanthocytes with the reticular distribution. Based on enzyme-linked immunosorbent assay results, the levels of Dsg1 and Dsg3 antibodies were 28.18 and 0.26 kU/L respectively. The diagnosis of PF was made. This patient was successfully treated with apremilast without systemic glucocorticosteroids or immunosuppressive agents. The patient has continued with apremilast 30mg once daily for maintenance and no adverse events related to apremilast such as gastrointestinal side effects were observed during the 9-month follow-up period. In conclusion, apremilast therapy without systemic glucocorticosteroids nor immunosuppressive agents might provide an effective alternative to management of mild PF without obvious side effect.


Subject(s)
Pemphigus , Thalidomide , Humans , Pemphigus/drug therapy , Female , Middle Aged , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Immunosuppressive Agents/therapeutic use , Treatment Outcome , Glucocorticoids/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
16.
Rinsho Ketsueki ; 65(7): 628-632, 2024.
Article in Japanese | MEDLINE | ID: mdl-39098012

ABSTRACT

A 62-year-old woman with adult T-cell leukemia/lymphoma (ATL) received umbilical cord blood transplantation (CBT) in first complete remission. However, relapse of ATL was detected on day 74 post-transplantation, as evidenced by the rapid growth of lymphoma cells in peripheral blood and an increase in soluble interleukin-2 receptor (sIL2R) levels. Discontinuation of immunosuppressant therapy alone did not improve ATL findings, but treatment with lenalidomide caused lymphoma cells to disappear from the peripheral blood and sIL2R levels to return to normal. Pancytopenia was observed as a lenalidomide-associated adverse effect, but lymphocyte counts were not reduced. The patient was judged to be in complete remission based on results of Southern blot analysis and human T-cell leukemia virus 1 (HTLV-1)-infected cell analysis using flow cytometry (HAS-Flow). Flow cytometric analysis of peripheral blood and FISH analysis of X and Y chromosomes revealed that the therapeutic effect of lenalidomide was associated with an increase in the number of donor-derived peripheral natural killer cells. ATL relapse was not observed at 13 months into lenalidomide treatment. Our results suggest that lenalidomide is an effective option for the treatment of post-transplant relapsed ATL.


Subject(s)
Cord Blood Stem Cell Transplantation , Lenalidomide , Leukemia-Lymphoma, Adult T-Cell , Recurrence , Remission Induction , Humans , Lenalidomide/administration & dosage , Lenalidomide/therapeutic use , Leukemia-Lymphoma, Adult T-Cell/therapy , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Female , Middle Aged , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use
18.
Skinmed ; 22(3): 218-219, 2024.
Article in English | MEDLINE | ID: mdl-39090018

ABSTRACT

OtezlaTM was first approved on March 21, 2014 for the treatment of psoriatic arthritis, on September 23, 2014 for moderate to severe plaque psoriasis and on July 19, 2019 for the treatment of oral ulcers associated with Behcet's disease (BD). Apremilast is an inhibitor of phosphodi-esterase-4, an enzyme involved in the pathogenesis of several dermatologic conditions. This review explores the potential utility of apremilast in the treatment of other unapproved dermatologic indications.


Subject(s)
Thalidomide , Humans , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Behcet Syndrome/drug therapy , Psoriasis/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Tablets , Arthritis, Psoriatic/drug therapy
19.
Blood Adv ; 8(19): 5039-5050, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39058954

ABSTRACT

ABSTRACT: Optimal therapy for the growing number of patients with lenalidomide (LEN)-refractory multiple myeloma in their first relapse remains poorly defined. We therefore undertook a randomized phase 2 study to evaluate the efficacy and safety of combining the oral proteasome inhibitor ixazomib (IXA) with pomalidomide (POM) and dexamethasone (DEX) in this patient population. The overall response rate (ORR) for POM-DEX was 43.6%, and for IXA-POM-DEX, it was 63.2%. The depth of response, measured by the attainment of at least a very good partial response, favored triplet therapy over doublet therapy (28.9% vs 5.1%; P = .0063). A preplanned interim analysis after 75% of the progression events had occurred demonstrated an improvement in progression-free survival (PFS) that favored IXA-POM-DEX and that crossed the predefined boundary of superiority, leading to release of the study results. With additional follow-up, the median PFS for POM-DEX was 7.5 months (95% confidence interval [CI], 4.8-13.6 months) vs 20.3 months for IXA-POM-DEX (95% CI, 7.7-26.0 months; hazard ratio, 0.437; upper 90% bound = 0.657). The ORR and median PFS for 26 of 30 eligible patients who crossed over from the doublet to the triplet therapy at disease progression was 23.1% and 5.6 months, respectively. Overall survival was similar between the 2 groups. More hematologic toxicities were seen with the triplet therapy, but nonhematologic adverse events were similar between the 2 arms. Our data support further testing of this all-oral triplet therapy in comparison with current standard triplet therapy in the context of phase 3 studies for patients with LEN-refractory disease at first relapse. This trial was registered at www.clinicaltrials.gov as #NCT02004275.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Boron Compounds , Dexamethasone , Glycine , Lenalidomide , Multiple Myeloma , Thalidomide , Humans , Boron Compounds/therapeutic use , Boron Compounds/administration & dosage , Boron Compounds/adverse effects , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Thalidomide/administration & dosage , Dexamethasone/therapeutic use , Dexamethasone/administration & dosage , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Glycine/analogs & derivatives , Glycine/therapeutic use , Glycine/administration & dosage , Glycine/adverse effects , Lenalidomide/therapeutic use , Lenalidomide/administration & dosage , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Middle Aged , Male , Female , Drug Resistance, Neoplasm , Aged, 80 and over , Recurrence , Treatment Outcome , Adult
20.
Int J Hematol ; 120(3): 278-289, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38995485

ABSTRACT

Cytopenia is a well-documented complication in the treatment of hematological malignancies with lenalidomide and pomalidomide. Although prior studies have highlighted direct effects on hematopoietic cells to explain this adverse effect, the involvement of hematopoietic-supportive stroma remains less understood. This study examined the effects of lenalidomide/pomalidomide on the expansion and differentiation of human CD34+ hematopoietic stem/progenitor cells (HSPCs) in vitro, in co-culture with human bone-marrow mesenchymal stromal/stem cells (MSCs). Our findings indicate that lenalidomide/pomalidomide increases the population of immature CD34+CD38- cells while decreasing the number of mature CD34+CD38+ cells, suggesting a mechanism that inhibits early HSPC maturation. This effect persisted across myeloid, megakaryocytic, and erythroid lineages, with MSCs playing a key role in preserving immature progenitors and inhibiting their differentiation. Furthermore, in myeloid differentiation assays augmented by granulocyte-colony stimulating factor, lenalidomide/pomalidomide not only enhanced the presence of CD34+ cells with mature myeloid markers such as CD11b but also reduced the populations lacking CD34 yet positive for these markers, irrespective of MSC presence. Thus, while MSCs support the presence of these immature cell populations, they simultaneously inhibit their maturation. This finding provides novel mechanistic insights into lenalidomide- and pomalidomide-induced cytopenia, and could guide therapeutic strategies for its mitigation.


Subject(s)
Antigens, CD34 , Cell Differentiation , Coculture Techniques , Hematopoietic Stem Cells , Lenalidomide , Mesenchymal Stem Cells , Thalidomide , Lenalidomide/pharmacology , Humans , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Mesenchymal Stem Cells/drug effects , Cell Differentiation/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/cytology , Antigens, CD34/metabolism , Cell Proliferation/drug effects , Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL