Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43.535
Filter
Add more filters

Uruguay Oncology Collection
Publication year range
1.
Cell ; 185(2): 283-298.e17, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35021065

ABSTRACT

Gasdermins are a family of structurally related proteins originally described for their role in pyroptosis. Gasdermin B (GSDMB) is currently the least studied, and while its association with genetic susceptibility to chronic mucosal inflammatory disorders is well established, little is known about its functional relevance during active disease states. Herein, we report increased GSDMB in inflammatory bowel disease, with single-cell analysis identifying epithelial specificity to inflamed colonocytes/crypt top colonocytes. Surprisingly, mechanistic experiments and transcriptome profiling reveal lack of inherent GSDMB-dependent pyroptosis in activated epithelial cells and organoids but instead point to increased proliferation and migration during inĀ vitro wound closure, which arrests in GSDMB-deficient cells that display hyper-adhesiveness and enhanced formation of vinculin-based focal adhesions dependent on PDGF-A-mediated FAK phosphorylation. Importantly, carriage of disease-associated GSDMB SNPs confers functional defects, disrupting epithelial restitution/repair, which, altogether, establishes GSDMB as a critical factor for restoration of epithelial barrier function and the resolution of inflammation.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis , Base Sequence , Case-Control Studies , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Epithelial Cells/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , HEK293 Cells , HT29 Cells , Humans , Inflammatory Bowel Diseases/genetics , Methotrexate/pharmacology , Mutation/genetics , Phosphorylation/drug effects , Polymorphism, Single Nucleotide/genetics , Pyroptosis/drug effects , Pyroptosis/genetics , Reproducibility of Results , Transcriptome/drug effects , Transcriptome/genetics , Up-Regulation/drug effects , Wound Healing/drug effects , Wound Healing/genetics
2.
Cell ; 176(1-2): 2-4, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30633904

ABSTRACT

With the advent of more successful chemotherapies, the number of cancer survivors continues toĀ increase. Unfortunately, many of these patients will exhibit long-term sequelae from their treatments, including serious cognitive deficits that impair daily function. In this issue of Cell, Gibson etĀ al. (2019) demonstrate that chemotherapy-related cognitive impairment is orchestrated by microglia.


Subject(s)
Cognition Disorders , Cognitive Dysfunction , Humans , Methotrexate
3.
Cell ; 176(1-2): 43-55.e13, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30528430

ABSTRACT

Chemotherapy results in a frequent yet poorly understood syndrome of long-term neurological deficits. Neural precursor cell dysfunction and white matterĀ dysfunction are thought to contribute to thisĀ debilitating syndrome. Here, we demonstrate persistent depletion of oligodendrocyte lineage cells in humans who received chemotherapy. Developing aĀ mouse model of methotrexate chemotherapy-induced neurological dysfunction, we find a similar depletion of white matter OPCs, increased but incomplete OPC differentiation, and a persistent deficit in myelination. OPCs from chemotherapy-naive mice similarly exhibit increased differentiation when transplanted into the microenvironment of previously methotrexate-exposed brains, indicating an underlying microenvironmental perturbation. Methotrexate results in persistent activation of microglia and subsequent astrocyte activation that isĀ dependent on inflammatory microglia. MicroglialĀ depletion normalizes oligodendroglial lineage dynamics, myelin microstructure, and cognitive behavior after methotrexate chemotherapy. These findings indicate that methotrexate chemotherapy exposure is associated with persistent tri-glial dysregulation and identify inflammatory microglia as aĀ therapeutic target to abrogate chemotherapy-related cognitive impairment. VIDEO ABSTRACT.


Subject(s)
Cognitive Dysfunction/chemically induced , Methotrexate/adverse effects , Oligodendroglia/drug effects , Animals , Brain/metabolism , Cell Differentiation , Cell Lineage , Cognitive Dysfunction/metabolism , Disease Models, Animal , Drug Therapy , Drug-Related Side Effects and Adverse Reactions , Humans , Methotrexate/pharmacology , Mice , Microglia/metabolism , Myelin Sheath/metabolism , Nerve Fibers, Myelinated , Neurogenesis/physiology , Neuroglia/metabolism , Neurons/drug effects , Oligodendroglia/metabolism , White Matter/metabolism
4.
Cell ; 175(6): 1546-1560.e17, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30500537

ABSTRACT

Mammalian folate metabolism is comprised of cytosolic and mitochondrial pathways with nearly identical core reactions, yet the functional advantages of such an organization are not well understood. Using genome-editing and biochemical approaches, we find that ablating folate metabolism in the mitochondria of mammalian cell lines results in folate degradation in the cytosol. Mechanistically, we show that QDPR, an enzyme in tetrahydrobiopterin metabolism, moonlights to repair oxidative damage to tetrahydrofolate (THF). This repair capacity is overwhelmed when cytosolic THF hyperaccumulates in the absence of mitochondrially produced formate, leading to THF degradation. Unexpectedly, we also find that the classic antifolate methotrexate, by inhibiting its well-known target DHFR, causes even more extensive folate degradation in nearly all tested cancer cell lines. These findings shed light on designĀ features of folate metabolism, provide a biochemical basis for clinically observed folate deficiency in QDPR-deficient patients, and reveal a hitherto unknown and unexplored cellular effect of methotrexate.


Subject(s)
Carbon/metabolism , Cytosol/metabolism , Formates/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Tetrahydrofolates/metabolism , Cytosol/pathology , HCT116 Cells , HeLa Cells , Humans , MCF-7 Cells , Methotrexate/pharmacokinetics , Methotrexate/pharmacology , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Tetrahydrofolate Dehydrogenase/metabolism
5.
Immunity ; 56(5): 998-1012.e8, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37116499

ABSTRACT

Cytosolic innate immune sensing is critical for protecting barrier tissues. NOD1 and NOD2 are cytosolic sensors of small peptidoglycan fragments (muropeptides) derived from the bacterial cell wall. These muropeptides enter cells, especially epithelial cells, through unclear mechanisms. We previously implicated SLC46 transporters in muropeptide transport in Drosophila immunity. Here, we focused on Slc46a2, which was highly expressed in mammalian epidermal keratinocytes, and showed that it was critical for the delivery of diaminopimelic acid (DAP)-muropeptides and activation of NOD1 in keratinocytes, whereas the related transporter Slc46a3 was critical for delivering the NOD2 ligand MDP to keratinocytes. In a mouse model, Slc46a2 and Nod1 deficiency strongly suppressed psoriatic inflammation, whereas methotrexate, a commonly used psoriasis therapeutic, inhibited Slc46a2-dependent transport of DAP-muropeptides. Collectively, these studies define SLC46A2 as a transporter of NOD1-activating muropeptides, with critical roles in the skin barrier, and identify this transporter as an important target for anti-inflammatory intervention.


Subject(s)
Dermatitis , Methotrexate , Mice , Animals , Methotrexate/pharmacology , Inflammation , Peptidoglycan/metabolism , Epithelial Cells/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Immunity, Innate , Mammals
6.
Nature ; 609(7929): 1056-1062, 2022 09.
Article in English | MEDLINE | ID: mdl-36071163

ABSTRACT

Folates are essential nutrients with important roles as cofactors in one-carbon transfer reactions, being heavily utilized in the synthesis of nucleic acids and the metabolism of amino acids during cell division1,2. Mammals lack de novo folate synthesis pathways and thus rely on folate uptake from the extracellular milieu3. The human reduced folate carrier (hRFC, also known as SLC19A1) is the major importer of folates into the cell1,3, as well as chemotherapeutic agents such as methotrexate4-6. As an anion exchanger, RFC couples the import of folates and antifolates to anion export across the cell membrane and it is a major determinant in methotrexate (antifolate) sensitivity, as genetic variants and its depletion result in drug resistance4-8. Despite its importance, the molecular basis of substrate specificity by hRFC remains unclear. Here we present cryo-electron microscopy structures of hRFC in the apo state and captured in complex with methotrexate. Combined with molecular dynamics simulations and functional experiments, our study uncovers key determinants of hRFC transport selectivity among folates and antifolate drugs while shedding light on important features of anion recognition by hRFC.


Subject(s)
Cryoelectron Microscopy , Folic Acid Antagonists , Methotrexate , Reduced Folate Carrier Protein , Anions/metabolism , Apoproteins/genetics , Apoproteins/metabolism , Biological Transport , Carbon/metabolism , Folic Acid/metabolism , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/metabolism , Humans , Methotrexate/chemistry , Methotrexate/metabolism , Molecular Dynamics Simulation , Reduced Folate Carrier Protein/genetics , Reduced Folate Carrier Protein/metabolism , Reduced Folate Carrier Protein/ultrastructure , Substrate Specificity
7.
PLoS Genet ; 20(4): e1011252, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38683847

ABSTRACT

Pneumocystis jirovecii is a fungal pathogen that causes pneumocystis pneumonia, a disease that mainly affects immunocompromised individuals. This fungus has historically been hard to study because of our inability to grow it in vitro. One of the main drug targets in P. jirovecii is its dihydrofolate reductase (PjDHFR). Here, by using functional complementation of the baker's yeast ortholog, we show that PjDHFR can be inhibited by the antifolate methotrexate in a dose-dependent manner. Using deep mutational scanning of PjDHFR, we identify mutations conferring resistance to methotrexate. Thirty-one sites spanning the protein have at least one mutation that leads to resistance, for a total of 355 high-confidence resistance mutations. Most resistance-inducing mutations are found inside the active site, and many are structurally equivalent to mutations known to lead to resistance to different antifolates in other organisms. Some sites show specific resistance mutations, where only a single substitution confers resistance, whereas others are more permissive, as several substitutions at these sites confer resistance. Surprisingly, one of the permissive sites (F199) is without direct contact to either ligand or cofactor, suggesting that it acts through an allosteric mechanism. Modeling changes in binding energy between F199 mutants and drug shows that most mutations destabilize interactions between the protein and the drug. This evidence points towards a more important role of this position in resistance than previously estimated and highlights potential unknown allosteric mechanisms of resistance to antifolate in DHFRs. Our results offer unprecedented resources for the interpretation of mutation effects in the main drug target of an uncultivable fungal pathogen.


Subject(s)
Drug Resistance, Fungal , Folic Acid Antagonists , Methotrexate , Mutation , Pneumocystis carinii , Tetrahydrofolate Dehydrogenase , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Tetrahydrofolate Dehydrogenase/chemistry , Pneumocystis carinii/genetics , Pneumocystis carinii/enzymology , Pneumocystis carinii/drug effects , Folic Acid Antagonists/pharmacology , Drug Resistance, Fungal/genetics , Methotrexate/pharmacology , Allosteric Regulation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/drug effects , Humans , Fungal Proteins/genetics , Fungal Proteins/metabolism , Fungal Proteins/chemistry , Catalytic Domain/genetics
8.
N Engl J Med ; 388(25): 2338-2348, 2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37342922

ABSTRACT

BACKGROUND: In patients undergoing allogeneic hematopoietic stem-cell transplantation (HSCT), a calcineurin inhibitor plus methotrexate has been a standard prophylaxis against graft-versus-host disease (GVHD). A phase 2 study indicated the potential superiority of a post-transplantation regimen of cyclophosphamide, tacrolimus, and mycophenolate mofetil. METHODS: In a phase 3 trial, we randomly assigned adults with hematologic cancers in a 1:1 ratio to receive cyclophosphamide-tacrolimus-mycophenolate mofetil (experimental prophylaxis) or tacrolimus-methotrexate (standard prophylaxis). The patients underwent HSCT from an HLA-matched related donor or a matched or 7/8 mismatched (i.e., mismatched at only one of the HLA-A, HLA-B, HLA-C, and HLA-DRB1 loci) unrelated donor, after reduced-intensity conditioning. The primary end point was GVHD-free, relapse-free survival at 1 year, assessed in a time-to-event analysis, with events defined as grade III or IV acute GVHD, chronic GVHD warranting systemic immunosuppression, disease relapse or progression, and death from any cause. RESULTS: In a multivariate Cox regression analysis, GVHD-free, relapse-free survival was significantly more common among the 214 patients in the experimental-prophylaxis group than among the 217 patients in the standard-prophylaxis group (hazard ratio for grade III or IV acute GVHD, chronic GVHD, disease relapse or progression, or death, 0.64; 95% confidence interval [CI], 0.49 to 0.83; P = 0.001). At 1 year, the adjusted GVHD-free, relapse-free survival was 52.7% (95% CI, 45.8 to 59.2) with experimental prophylaxis and 34.9% (95% CI, 28.6 to 41.3) with standard prophylaxis. Patients in the experimental-prophylaxis group appeared to have less severe acute or chronic GVHD and a higher incidence of immunosuppression-free survival at 1 year. Overall and disease-free survival, relapse, transplantation-related death, and engraftment did not differ substantially between the groups. CONCLUSIONS: Among patients undergoing allogeneic HLA-matched HSCT with reduced-intensity conditioning, GVHD-free, relapse-free survival at 1 year was significantly more common among those who received cyclophosphamide-tacrolimus-mycophenolate mofetil than among those who received tacrolimus-methotrexate. (Funded by the National Heart, Lung, and Blood Institute and others; BMT CTN 1703 ClinicalTrials.gov number, NCT03959241.).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Bronchiolitis Obliterans Syndrome , Cyclophosphamide , Graft vs Host Disease , Hematologic Neoplasms , Hematopoietic Stem Cell Transplantation , Adult , Humans , Bronchiolitis Obliterans Syndrome/etiology , Bronchiolitis Obliterans Syndrome/prevention & control , Cyclophosphamide/administration & dosage , Graft vs Host Disease/prevention & control , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Methotrexate/administration & dosage , Mycophenolic Acid/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Tacrolimus/administration & dosage , Unrelated Donors , Hematologic Neoplasms/surgery , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
9.
Blood ; 143(1): 21-31, 2024 01 04.
Article in English | MEDLINE | ID: mdl-37647633

ABSTRACT

ABSTRACT: Patients who undergo human leukocyte antigen-matched unrelated donor (MUD) allogeneic hematopoietic stem cell transplantation (HSCT) with myeloablative conditioning for hematologic malignancies often develop acute graft-versus-host disease (GVHD) despite standard calcineurin inhibitor-based prophylaxis in combination with methotrexate. This trial evaluated a novel human CD24 fusion protein (CD24Fc/MK-7110) that selectively targets and mitigates inflammation due to damage-associated molecular patterns underlying acute GVHD while preserving protective immunity after myeloablative conditioning. This phase 2a, multicenter study evaluated the pharmacokinetics, safety, and efficacy of CD24Fc in combination with tacrolimus and methotrexate in preventing acute GVHD in adults undergoing MUD HSCT for hematologic malignancies. A double-blind, placebo-controlled, dose-escalation phase to identify a recommended dose was followed by an open-label expansion phase with matched controls to further evaluate the efficacy and safety of CD24Fc in preventing acute GVHD. A multidose regimen of CD24Fc produced sustained drug exposure with similar safety outcomes when compared with single-dose regimens. Grade 3 to 4 acute GVHD-free survival at day 180 was 96.2% (95% confidence interval [CI], 75.7-99.4) in the CD24Fc expansion cohort (CD24Fc multidose), compared with 73.6% (95% CI, 63.2-81.4) in matched controls (hazard ratio, 0.1 [95% CI, 0.0-0.6]; log-rank test, PĀ = .03). No participants in the CD24Fc escalation or expansion phases experienced dose-limiting toxicities (DLTs). The multidose regimen of CD24Fc was well tolerated with no DLTs and was associated with high rates of severe acute GVHD-free survival after myeloablative MUD HSCT. This trial was registered at ClinicalTrials.gov as #NCT02663622.


Subject(s)
Graft vs Host Disease , Hematologic Neoplasms , Hematopoietic Stem Cell Transplantation , Adult , Humans , Methotrexate/therapeutic use , Hematopoietic Stem Cell Transplantation/adverse effects , Transplantation, Homologous , Neoplasm Recurrence, Local/drug therapy , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Transplantation Conditioning/adverse effects
10.
Blood ; 144(17): 1834-1845, 2024 Oct 24.
Article in English | MEDLINE | ID: mdl-39028876

ABSTRACT

ABSTRACT: Abatacept plus calcineurin inhibitors/methotrexate (CNI/MTX) is the first US Food and Drug Administration (FDA)-approved regimen for acute graft-versus-host disease (aGVHD) prophylaxis during unrelated-donor hematopoietic cell transplantation (URD-HCT). Using Center for International Blood and Marrow Transplant Research data, we investigated its impact in patients receiving 7/8 HLA-mismatched unrelated donor (MMUD) or 8/8 HLA-matched unrelated donor (MUD) URD-HCT between 2011 and 2018. Primary outcomes included day-180, 1-year, and 2-year overall survival (OS) and relapse-free survival (RFS) for abataceptĀ + CNI/MTX vsĀ CNI/MTX, CNI/MTXĀ + antithymocyte globulin (ATG), and posttransplant cyclophosphamide-based prophylaxis (PT-Cy). For 7/8 MMUDs, day-180 OS (primary end point supporting FDA approval) was significantly higher for abataceptĀ + CNI/MTX vs CNI/MTX (98% vs 75%; PĀ =Ā .0028). Two-year RFS was significantly higher for abataceptĀ + CNI/MTX vs CNI/MTX (74% vs 49%; PĀ =Ā .0098) and CNI/MTXĀ + ATG (77% vs 35%; PĀ =Ā .0002), and similar vs PT-Cy (72% vs 56%; PĀ =Ā .1058). For 8/8 MUDs, 2-year RFS for abataceptĀ + CNI/MTX was numerically higher vs CNI/MTX (63% vs 52%; PĀ =Ā .1497), with an improved hazard ratio (HR) of 0.46 (0.25-0.86), and vs CNI/MTXĀ + ATG (66% vs 55%; PĀ =Ā .1193; HR, 0.39 [0.21-0.73]), and was similar vs PT-Cy (68% vs 57%; PĀ =Ā .2356; HR, 0.54 [0.26-1.11]). For 7/8 MMUD and 8/8 MUD recipients, abataceptĀ + CNI/MTX prophylaxis improved survival outcomes vs CNI/MTX and CNI/MTXĀ + ATG; outcomes were similar to PT-Cy-based regimens. AbataceptĀ + CNI/MTX may facilitate unrelated donor pool expansion for HCT.


Subject(s)
Abatacept , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Unrelated Donors , Humans , Graft vs Host Disease/prevention & control , Graft vs Host Disease/etiology , Graft vs Host Disease/mortality , Abatacept/therapeutic use , Abatacept/administration & dosage , Hematopoietic Stem Cell Transplantation/adverse effects , Female , Middle Aged , Male , Adult , Aged , Methotrexate/therapeutic use , Methotrexate/administration & dosage , Immunosuppressive Agents/therapeutic use , Young Adult , Acute Disease , Antilymphocyte Serum/therapeutic use , Antilymphocyte Serum/administration & dosage
11.
N Engl J Med ; 387(8): 715-726, 2022 08 25.
Article in English | MEDLINE | ID: mdl-36001712

ABSTRACT

BACKGROUND: The cytokine interleukin-6 is involved in the pathogenesis of rheumatoid arthritis. Olokizumab, a humanized monoclonal antibody targeting the interleukin-6 cytokine directly, is being tested for the treatment of rheumatoid arthritis. METHODS: In a 24-week, phase 3, multicenter, placebo- and active-controlled trial, we randomly assigned (in a 2:2:2:1 ratio) patients with rheumatoid arthritis and an inadequate response to methotrexate to receive subcutaneous olokizumab at a dose of 64 mg every 2 or 4 weeks, adalimumab (40 mg every 2 weeks), or placebo; all patients continued methotrexate therapy. The primary end point was an American College of Rheumatology 20 (ACR20) response (≥20% fewer tender and swollen joints and ≥20% improvement in three of five other domains) at week 12, with each olokizumab dose tested for superiority to placebo. We also tested the noninferiority of each olokizumab dose to adalimumab with respect to the percentage of patients with an ACR20 response (noninferiority margin, -12 percentage points in the lower boundary of the 97.5% confidence interval for the difference between groups). RESULTS: A total of 464 patients were assigned to receive olokizumab every 2 weeks, 479 to receive olokizumab every 4 weeks, 462 to receive adalimumab, and 243 to receive placebo. An ACR20 response at week 12 occurred in 44.4% of the patients receiving placebo, in 70.3% receiving olokizumab every 2 weeks (difference vs. placebo, 25.9 percentage points; 97.5% confidence interval [CI], 17.1 to 34.1), in 71.4% receiving olokizumab every 4 weeks (difference vs. placebo, 27.0 percentage points; 97.5% CI, 18.3 to 35.2), and in 66.9% receiving adalimumab (difference vs. placebo, 22.5 percentage points; 95% CI, 14.8 to 29.8) (P<0.001 for the superiority of each olokizumab dose to placebo). Both olokizumab doses were noninferior to adalimumab with respect to the percentage of patients with an ACR20 response at week 12 (difference, 3.4 percentage points [97.5% CI, -3.5 to 10.2] with olokizumab every 2 weeks and 4.5 percentage points [97.5% CI, -2.2 to 11.2] with olokizumab every 4 weeks). Adverse events, most commonly infections, occurred in approximately 70% of the patients who received olokizumab. Antibodies against olokizumab were detected in 3.8% of the patients receiving the drug every 2 weeks and in 5.1% of those receiving it every 4 weeks. CONCLUSIONS: In patients with rheumatoid arthritis who were receiving maintenance methotrexate, olokizumab was superior to placebo and noninferior to adalimumab in producing an ACR20 response at 12 weeks. Larger and longer trials are required to determine the efficacy and safety of olokizumab in patients with rheumatoid arthritis. (Supported by R-Pharm; CREDO2 ClinicalTrials.gov number, NCT02760407.).


Subject(s)
Adalimumab , Antibodies, Monoclonal, Humanized , Antirheumatic Agents , Arthritis, Rheumatoid , Methotrexate , Adalimumab/administration & dosage , Adalimumab/adverse effects , Adalimumab/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/adverse effects , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Double-Blind Method , Drug Therapy, Combination , Humans , Interleukin-6/antagonists & inhibitors , Methotrexate/administration & dosage , Methotrexate/adverse effects , Methotrexate/therapeutic use , Treatment Outcome , Tumor Necrosis Factor-alpha
12.
Blood ; 141(15): 1802-1811, 2023 04 13.
Article in English | MEDLINE | ID: mdl-36603187

ABSTRACT

To determineĀ the prognostic significance of central nervous system (CNS) leukemic involvement in newly diagnosed T-cell acute lymphoblastic leukemia (T-ALL), outcomes on consecutive, phase 3 Children's Oncology Group clinical trials were examined. AALL0434 and AALL1231 tested efficacy of novel agents within augmented-Berlin-Frankfurt-MĆ¼nster (aBFM) therapy. In addition to testing study-specific chemotherapy through randomization, the AALL0434 regimen delivered cranial radiation therapy (CRT) to most participants (90.8%), whereas AALL1231 intensified chemotherapy to eliminate CRT in 88.2% of participants. In an analysis of 2164 patients with T-ALL (AALL0434, 1550; AALL1231, 614), 1564 had CNS-1 (72.3%), 441 CNS-2 (20.4%), and 159 CNS-3 (7.3%). The 4-year event-free-survival (EFS) was similar for CNS-1 (85.1%Ā Ā±Ā 1.0%) and CNS-2 (83.2%Ā Ā±Ā 2.0%), but lower for CNS-3 (71.8%Ā Ā±Ā 4.0%;Ā PĀ = .0004). Patients with CNS-1 and CNS-2 had similar 4-year overall survival (OS) (90.1%Ā Ā±Ā 0.8% and 90.5%Ā Ā±Ā 1.5%, respectively), with OS for CNS-3 being 82.7%Ā Ā±Ā 3.4% (PĀ = .005). Despite therapeutic differences, outcomes for CNS-1 and CNS-2 were similar regardless of CRT, intensified corticosteroids, or novel agents. Except for significantly superior outcomes with nelarabine on AALL0434 (4-year disease-free survival, 93.1%Ā Ā±Ā 5.2%), EFS/OS was inferior with CNS-3 status, all of whom received CRT. Combined analyses of >2000 patients with T-ALL identified that CNS-1 and CNS-2 status at diagnosis had similar outcomes. Unlike B-ALL, CNS-2 status in T-ALL does not impact outcome with aBFM therapy, without additional intrathecal therapy, with or without CRT. Although nelarabine improved outcomes for those with CNS-3 status, novel approaches are needed. These trials were registered at www.clinicaltrials.gov as #NCT00408005 (AALL0434) and #NCT02112916 (AALL1231).


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Infant , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Central Nervous System , Disease-Free Survival , Methotrexate , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Prognosis , T-Lymphocytes , Treatment Outcome
13.
Ann Intern Med ; 177(9): 1145-1156, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39074374

ABSTRACT

BACKGROUND: Treatments for osteoarthritis (OA) are limited. Previous small studies suggest that the antirheumatic drug methotrexate may be a potential treatment for OA pain. OBJECTIVE: To assess symptomatic benefits of methotrexate in knee OA (KOA). DESIGN: A multicenter, randomized, double-blind, placebo-controlled trial done between 13 June 2014 and 13 October 2017. (ISRCTN77854383; EudraCT: 2013-001689-41). SETTING: 15 secondary care musculoskeletal clinics in the United Kingdom. PARTICIPANTS: A total of 207 participants with symptomatic, radiographic KOA and knee pain (severity ≥4 out of 10) on most days in the past 3 months with inadequate response to current medication were approached for inclusion. INTERVENTION: Participants were randomly assigned 1:1 to oral methotrexate once weekly (6-week escalation 10 to 25 mg) or matched placebo over 12 months and continued usual analgesia. MEASUREMENTS: The primary end point was average knee pain (numerical rating scale [NRS] 0 to 10) at 6 months, with 12-month follow-up to assess longer-term response. Secondary end points included knee stiffness and function outcomes and adverse events (AEs). RESULTS: A total of 155 participants (64% women; mean age, 60.9 years; 50% Kellgren-Lawrence grade 3 to 4) were randomly assigned to methotrexate (n = 77) or placebo (n = 78). Follow-up was 86% (n = 134; methotrexate: 66, placebo: 68) at 6 months. Mean knee pain decreased from 6.4 (SD, 1.80) at baseline to 5.1 (SD, 2.32) at 6 months in the methotrexate group and from 6.8 (SD, 1.62) to 6.2 (SD, 2.30) in the placebo group. The primary intention-to-treat analysis showed a statistically significant pain reduction of 0.79 NRS points in favor of methotrexate (95% CI, 0.08 to 1.51; P = 0.030). There were also statistically significant treatment group differences in favor of methotrexate at 6 months for Western Ontario and McMaster Universities Osteoarthritis Index stiffness (0.60 points [CI, 0.01 to 1.18]; P = 0.045) and function (5.01 points [CI, 1.29 to 8.74]; P = 0.008). Treatment adherence analysis supported a dose-response effect. Four unrelated serious AEs were reported (methotrexate: 2, placebo: 2). LIMITATION: Not permitting oral methotrexate to be changed to subcutaneous delivery for intolerance. CONCLUSION: Oral methotrexate added to usual medications demonstrated statistically significant reduction in KOA pain, stiffness, and function at 6 months. PRIMARY FUNDING SOURCE: Versus Arthritis.


Subject(s)
Antirheumatic Agents , Methotrexate , Osteoarthritis, Knee , Pain Measurement , Humans , Methotrexate/administration & dosage , Methotrexate/adverse effects , Methotrexate/therapeutic use , Osteoarthritis, Knee/drug therapy , Osteoarthritis, Knee/complications , Double-Blind Method , Female , Male , Middle Aged , Administration, Oral , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/adverse effects , Antirheumatic Agents/therapeutic use , Aged , Treatment Outcome , Arthralgia/drug therapy
14.
Ann Intern Med ; 177(5_Supplement): S82-S90, 2024 May.
Article in English | MEDLINE | ID: mdl-38621248

ABSTRACT

Many patients with rheumatologic conditions receive care from physicians other than rheumatologists. Here we note key findings from 6 studies in rheumatology published in 2023 that offer valuable insights for internal medicine specialists and subspecialists outside of rheumatology. The first study investigated the effect of low-dose glucocorticoids on patients with rheumatoid arthritis (RA) over 2 years and challenged existing perceptions about the risks of glucocorticoids in this setting. The second study focused on the updated guideline for preventing and treating glucocorticoid-induced osteoporosis. With the chronic and widespread use of glucocorticoids, the American College of Rheumatology emphasized the importance of assessing fracture risk and initiating pharmacologic therapy when appropriate. The third study explored the potential use of methotrexate in treating inflammatory hand osteoarthritis, suggesting a novel approach to managing this challenging and common condition. The results of the fourth article we highlight suggest that sarilumab has promise as an adjunct treatment of polymyalgia rheumatica relapse during glucocorticoid dosage tapering. The fifth study evaluated sublingual cyclobenzaprine for fibromyalgia treatment, noting both potential benefits and risks. Finally, the sixth article is a systematic review and meta-analysis that assessed the therapeutic equivalence of biosimilars and reference biologics in the treatment of patients with RA. Knowledge of this recent literature will be useful to clinicians regardless of specialty who care for patients with these commonly encountered conditions.


Subject(s)
Glucocorticoids , Humans , Glucocorticoids/therapeutic use , Glucocorticoids/adverse effects , Glucocorticoids/administration & dosage , Osteoporosis/drug therapy , Arthritis, Rheumatoid/drug therapy , Antirheumatic Agents/therapeutic use , Antirheumatic Agents/adverse effects , Methotrexate/therapeutic use , Methotrexate/adverse effects , Rheumatology/standards , Rheumatic Diseases/drug therapy , Rheumatic Diseases/complications , Biosimilar Pharmaceuticals/therapeutic use , Biosimilar Pharmaceuticals/adverse effects , Polymyalgia Rheumatica/drug therapy , Fibromyalgia/drug therapy
15.
Lancet Oncol ; 25(2): 255-264, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38142702

ABSTRACT

BACKGROUND: The optimal perioperative chemotherapy for patients with muscle-invasive bladder cancer is not defined. The VESPER (French Genito-Urinary Tumor Group and French Association of Urology V05) trial reported improved 3-year progression-free survival with dose-dense methotrexate, vinblastine, doxorubicin and cisplatin (dd-MVAC) versus gemcitabine and cisplatin (GC) in patients who received neoadjuvant therapy, but not in the overall perioperative setting. In this Article, we report on the secondary endpoints of overall survival and time to death due to bladder cancer at 5-year follow-up. METHODS: VESPER was an open-label, randomised, phase 3 trial done at 28 university hospitals or comprehensive cancer centres in France, in which adults (age ≤18 years and ≤80 years) with primary bladder cancer and histologically confirmed muscle-invasive urothelial carcinoma were randomly allocated (1:1; block size four) to treatment with dd-MVAC (every 2 weeks for a total of six cycles) or GC (every 3 weeks for a total of four cycles). Overall survival and time to death due to bladder cancer (presented as 5-year cumulative incidence of death due to bladder cancer) was analysed by intention to treat (ITT) in all randomly assigned patients. Overall survival was assessed by the Kaplan-Meier method with the treatment groups compared with log-rank test stratified for mode of administration of chemotherapy (neoadjuvant or adjuvant) and lymph node involvement. Time to death due to bladder cancer was analysed with an Aalen model for competing risks and a Fine and Gray regression model stratified for the same two covariates. Results were presented for the total perioperative population and for the neoadjuvant and adjuvant subgroups. The trial is registered with ClinicalTrials.gov, NCT01812369, and is complete. FINDINGS: From Feb 25, 2013, to March 1, 2018, 500 patients were randomly assigned, of whom 493 were included in the final ITT population (245 [50%] in the GC group and 248 [50%] in the dd-MVAC group; 408 [83%] male and 85 [17%] female). 437 (89%) patients received neoadjuvant chemotherapy. Median follow-up was 5Ā·3 years (IQR 5Ā·1-5Ā·4); 190 deaths at the 5-year cutoff were reported. In the perioperative setting (total ITT population), we found no evidence of association of overall survival at 5 years with dd-MVAC treatment versus GC treatment (64% [95% CI 58-70] vs 56% [50-63], stratified hazard ratio [HRstrat] 0Ā·79 [95% CI 0Ā·59-1Ā·05]). Time to death due to bladder cancer was increased in the dd-MVAC group compared with in the GC group (5-year cumulative incidence of death: 27% [95% CI 21-32] vs 40% [34-46], HRstrat 0Ā·61 [95% CI 0Ā·45-0Ā·84]). In the neoadjuvant subgroup, overall survival at 5 years was improved in the dd-MVAC group versus the GC group (66% [95% CI 60-73] vs 57% [50-64], HR 0Ā·71 [95% CI 0Ā·52-0Ā·97]), as was time to death due to bladder cancer (5-year cumulative incidence: 24% [18-30] vs 38% [32-45], HR 0Ā·55 [0Ā·39-0Ā·78]). In the adjuvant subgroup, the results were not conclusive due to the small sample size. Bladder cancer progression was the cause of death for 157 (83%) of the 190 deaths; other causes of death included cardiovascular events (eight [4%] deaths), deaths related to chemotherapy toxicity (four [2%]), and secondary cancers (four [2%]). INTERPRETATION: Our results on overall survival at 5 years were in accordance with the primary endpoint analysis (3-year progression-free survival). We found no evidence of improved overall survival with dd-MVAC over GC in the perioperative setting, but the data support the use of six cycles of dd-MVAC over four cycles of GC in the neoadjuvant setting. These results should impact practice and future trials of immunotherapy in bladder cancer. FUNDING: French National Cancer Institute.


Subject(s)
Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , Adult , Humans , Male , Female , Adolescent , Urinary Bladder Neoplasms/pathology , Cisplatin , Vinblastine/adverse effects , Methotrexate/adverse effects , Carcinoma, Transitional Cell/drug therapy , Gemcitabine , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Doxorubicin , Deoxycytidine , Neoadjuvant Therapy/adverse effects , Muscles/pathology
16.
Lancet Oncol ; 25(9): 1213-1221, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39127062

ABSTRACT

BACKGROUND: Breast-conserving surgery, adjuvant systemic therapy, and radiotherapy are the standard of care for most women with early breast cancer. There are few reports of clinical outcomes beyond the first decade of follow-up of randomised trials comparing breast-conserving surgery with or without radiotherapy. We present a 30-year update of the Scottish Breast Conservation Trial. METHODS: In this randomised, controlled, phase 3 trial across 14 hospitals in Scotland, women aged younger than 70 years with early breast cancer (tumours ≤4 cm [T1 or T2 and N0 or N1]) were included. They underwent breast-conserving surgery (1 cm margin) with axillary node sampling or clearance. Oestrogen receptor (ER)-rich patients (≥20 fmol/mg protein) received 20 mg oral tamoxifen daily for 5 years. ER-poor patients (<20 fmol/mg protein) received chemotherapy (cyclophosphamide 600 mg/m2, methotrexate 50 mg/m2, and fluorouracil 600 mg/m2 every 21 days intravenously in eight courses). Stratification was by menstrual status (within or more than 12 months from last menstrual period) and ER status (oestrogen concentration ≥20 fmol/mg protein, <20 fmol/mg protein, or unknown) and patients were randomly assigned (1:1) to high-dose (50 Gy in 20-25 fractions) local or locoregional radiotherapy versus no radiotherapy. No blinding was possible due to the nature of the treatment. We report the primary endpoint of the original trial, ipsilateral breast tumour recurrence, and the co-primary endpoint, overall survival. Clinical outcomes were compared by the log-rank test. Hazard ratios (HRs) are reported, with no radiotherapy as the reference group. Failures of the proportional hazards assumption are reported if significant. All analyses are by intention to treat. FINDINGS: Between April 1, 1985, and Oct 2, 1991, 589 patients were enrolled and randomly assigned to the two treatment groups (293 to radiotherapy and 296 to no radiotherapy). After exclusion of four ineligible patients (two in each group), there were 291 patients in the radiotherapy group and 294 patients in the no radiotherapy group. Median follow-up was 17Ā·5 years (IQR 8Ā·4-27Ā·9). Ipsilateral breast tumour recurrence was significantly lower in the radiotherapy group than in the no radiotherapy group (46 [16%] of 291 vs 107 [36%] of 294; HR 0Ā·39 [95% CI 0Ā·28-0Ā·55], p<0Ā·0001). Although there were differences in the hazard rate for ipsilateral breast tumour recurrence in the first decade after treatment (HR 0Ā·24 [95% CI 0Ā·15-0Ā·38], p<0Ā·0001), subsequent risks of ipsilateral breast tumour recurrence were similar in both groups (0Ā·98 [0Ā·54-1Ā·79], p=0Ā·95). There was no difference in overall survival between the two groups (median 18Ā·7 years [95% CI 16Ā·5-21Ā·5] in the no radiotherapy group vs 19Ā·2 years [16Ā·9-21Ā·3] in the radiotherapy group; HR 1Ā·08 [95% CI 0Ā·89-1 Ā·30], log-rank p=0Ā·43). INTERPRETATION: Our findings suggest that patients whose biology predicts a late relapse a decade or more after breast-conserving surgery for early breast cancer might gain little from adjuvant radiotherapy. FUNDING: Breast Cancer Institute (part of Edinburgh and Lothian Health Foundation) and PFS Genomics (now part of Exact Sciences).


Subject(s)
Breast Neoplasms , Mastectomy, Segmental , Humans , Female , Breast Neoplasms/pathology , Breast Neoplasms/mortality , Breast Neoplasms/radiotherapy , Breast Neoplasms/therapy , Breast Neoplasms/surgery , Middle Aged , Adult , Radiotherapy, Adjuvant , Aged , Neoplasm Recurrence, Local/pathology , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Tamoxifen/therapeutic use , Scotland , Disease-Free Survival , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemotherapy, Adjuvant , Treatment Outcome , Receptors, Estrogen/metabolism , Neoplasm Staging , Methotrexate/administration & dosage , Methotrexate/therapeutic use , Cyclophosphamide/administration & dosage , Cyclophosphamide/therapeutic use
17.
J Cell Physiol ; 239(5): e31256, 2024 May.
Article in English | MEDLINE | ID: mdl-38591855

ABSTRACT

Osteosarcoma (OS) cancer treatments include systemic chemotherapy and surgical resection. In the last years, novel treatment approaches have been proposed, which employ a drug-delivery system to prevent offside effects and improves treatment efficacy. Locally delivering anticancer compounds improves on high local concentrations with more efficient tumour-killing effect, reduced drugs resistance and confined systemic effects. Here, the synthesis of injectable strontium-doped calcium phosphate (SrCPC) scaffold was proposed as drug delivery system to combine bone tissue regeneration and anticancer treatment by controlled release of methotrexate (MTX) and doxorubicin (DOX), coded as SrCPC-MTX and SrCPC-DOX, respectively. The drug-loaded cements were tested in an in vitro model of human OS cell line SAOS-2, engineered OS cell line (SAOS-2-eGFP) and U2-OS. The ability of doped scaffolds to induce OS cell death and apoptosis was assessed analysing cell proliferation and Caspase-3/7 activities, respectively. To determine if OS cells grown on doped-scaffolds change their migratory ability and invasiveness, a wound-healing assay was performed. In addition, the osteogenic potential of SrCPC material was evaluated using human adipose derived-mesenchymal stem cells. Osteogenic markers such as (i) the mineral matrix deposition was analysed by alizarin red staining; (ii) the osteocalcin (OCN) protein expression was investigated by enzyme-linked immunosorbent assay test, and (iii) the osteogenic process was studied by real-time polymerase chain reaction array. The delivery system induced cell-killing cytotoxic effects and apoptosis in OS cell lines up to Day 7. SrCPC demonstrates a good cytocompatibility and it induced upregulation of osteogenic genes involved in the skeletal development pathway, together with OCN protein expression and mineral matrix deposition. The proposed approach, based on the local, sustained release of anticancer drugs from nanostructured biomimetic drug-loaded cements is promising for future therapies aiming to combine bone regeneration and anticancer local therapy.


Subject(s)
Antineoplastic Agents , Apoptosis , Bone Neoplasms , Calcium Phosphates , Doxorubicin , Methotrexate , Osteogenesis , Osteosarcoma , Tissue Scaffolds , Humans , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Calcium Phosphates/administration & dosage , Calcium Phosphates/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/metabolism , Strontium/pharmacology , Strontium/chemistry , Tissue Scaffolds/chemistry , Drug Delivery Systems , Methotrexate/administration & dosage , Methotrexate/pharmacology
18.
J Am Chem Soc ; 146(42): 28685-28695, 2024 Oct 23.
Article in English | MEDLINE | ID: mdl-39394997

ABSTRACT

The sensitivity to protein inhibitors is altered by modifications or protein mutations, as represented by drug resistance. The mode of stable drug binding to the protein pocket has been experimentally clarified. However, the nature of the binding of inhibitors with reduced sensitivity remains unclear at the atomic level. In this study, we analyzed the thermodynamics and kinetics of inhibitor binding to the surface of wild-type and mutant dihydrofolate reductase (DHFR) using molecular dynamics simulations combined with Markov state modeling. A strong inhibitor of methotrexate (MTX) showed a preference for the active site of wild-type DHFR with minimal binding to unrelated (secondary) sites. Deletion of a side-chain fragment in MTX largely destabilized the active site-bound state, with clear evidence of binding to secondary sites. Similarly, the F31V mutation in DHFR diminished the specificity of MTX binding to the active site. These results reveal the presence of multiple-bound states whose stabilities are comparable to or higher than those of the unbound state, suggesting that a reduction in the binding affinity for the active site significantly elevates the fractions of these states. This study presents a theoretical model that more accurately interprets the altered drug sensitivity than the traditional two-state model.


Subject(s)
Folic Acid Antagonists , Methotrexate , Molecular Dynamics Simulation , Tetrahydrofolate Dehydrogenase , Tetrahydrofolate Dehydrogenase/metabolism , Tetrahydrofolate Dehydrogenase/chemistry , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Folic Acid Antagonists/metabolism , Methotrexate/chemistry , Methotrexate/pharmacology , Methotrexate/metabolism , Binding Sites , Thermodynamics , Protein Binding , Kinetics , Mutation , Catalytic Domain
19.
Br J Cancer ; 130(11): 1819-1827, 2024 May.
Article in English | MEDLINE | ID: mdl-38594370

ABSTRACT

BACKGROUND: Although DHFR gene amplification has long been known as a major mechanism for methotrexate (MTX) resistance in cancer, the early changes and detailed development of the resistance are not yet fully understood. METHODS: We performed genomic, transcriptional and proteomic analyses of human colon cancer cells with sequentially increasing levels of MTX-resistance. RESULTS: The genomic amplification evolved in three phases (pre-amplification, homogenously staining region (HSR) and extrachromosomal DNA (ecDNA)). We confirm that genomic amplification and increased expression of DHFR, with formation of HSRs and especially ecDNAs, is the major driver of resistance. However, DHFR did not play a detectable role in the early phase. In the late phase (ecDNA), increase in FAM151B protein level may also have an important role by decreasing sensitivity to MTX. In addition, although MSH3 and ZFYVE16 may be subject to different posttranscriptional regulations and therefore protein expressions are decreased in ecDNA stages compared to HSR stages, they still play important roles in MTX resistance. CONCLUSION: The study provides a detailed evolutionary trajectory of MTX-resistance and identifies new targets, especially ecDNAs, which could help to prevent drug resistance. It also presents a proof-of-principal approach which could be applied to other cancer drug resistance studies.


Subject(s)
Drug Resistance, Neoplasm , Gene Amplification , Methotrexate , Tetrahydrofolate Dehydrogenase , Humans , Methotrexate/pharmacology , Drug Resistance, Neoplasm/genetics , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Antimetabolites, Antineoplastic/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genomics/methods
20.
BMC Immunol ; 25(1): 40, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965465

ABSTRACT

BACKGROUND: Psoriasis has a global prevalence of 1-3%, with variations observed across different ethnic groups and geographical areas. Disease susceptibility and response to anti-tumor necrosis factor-α (TNFα) drugs suggest different genetic regulatory mechanisms which may include NLR family pyrin domain containing 3 (NLRP3) polymorphism. Evaluation of the NLRP3 gene polymorphism, the serum level of CRP and TNFα in psoriasis patients and assessment of the NLRP3 (rs10754558) gene polymorphism, CRP and TNFα with disease severity and their role as biomarkers for response to Methotrexate and Adalimumab in psoriasis. The study had a total of 75 patients diagnosed with psoriasis vulgaris, who were compared to a control group of 75 healthy individuals. RESULTS: There was a highly significant difference in NLRP3 genotypes and alleles distribution between psoriasis patients and controls (P = 0.002,0.004). The heterozygote genotype GC (OR = 3.67,95%CI:1.75-7.68, P = 0.0006), was linked with increased risk of psoriasis. Additionally, The GC genotype was significantly associated with nonresponse to psoriasis therapy (OR = 11.7,95%CI:3.24-42.28, P = 0.0002). Regarding serum CRP and TNFα levels, there was a highly statistically significant difference between psoriasis patients and controls (P < 0.0001), and there was also a highly statistically significant difference between responders and non-responders in psoriasis patients regarding PASI 50 (P < 0.0001). CONCLUSIONS: The NLRP3 (rs10754558) genotypes GC was associated with the severe form of psoriasis and with nonresponse to psoriasis medication. Therefore, NLRP3 (rs10754558) gene polymorphism is an important prognostic biomarker in psoriasis patients. The serum TNFα can be used as a predictor for response to therapy in psoriasis patients. More research for evaluation of role of the NLRP3 gene polymorphism in the genetic risks and treatment outcomes associated with psoriasis is still required.


Subject(s)
Adalimumab , Methotrexate , NLR Family, Pyrin Domain-Containing 3 Protein , Polymorphism, Single Nucleotide , Psoriasis , Tumor Necrosis Factor-alpha , Humans , Psoriasis/genetics , Psoriasis/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Adalimumab/therapeutic use , Methotrexate/therapeutic use , Female , Tumor Necrosis Factor-alpha/genetics , Male , Adult , Middle Aged , Genotype , Genetic Predisposition to Disease , Treatment Outcome , C-Reactive Protein/metabolism , Biomarkers/blood , Alleles , Severity of Illness Index , Gene Frequency
SELECTION OF CITATIONS
SEARCH DETAIL