Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
3.
Lung Cancer ; 173: 116-123, 2022 11.
Article in English | MEDLINE | ID: mdl-36198244

ABSTRACT

CONTEXTE: The Epidermal Growth Factor Receptor (EGFR) is mutated in 10-15% of patients with lung adenocarcinoma. At metastatic stage EGFR tyrosine kinase inhibitors (TKIs) are used front line for patients harboring targetable mutations. Novel anti-EGFR therapies are being developed. Amivantamab is a bispecific anti-EGFR and anti-MET antibody with expected skin toxicities. OBJECTIVE: We developed here guidelines for prevention and treatment of cutaneous toxicities under amivantamab according to our experience at Institut Curie. MATERIEL & METHOD: The first patients with metastatic lung cancer harboring EGFR Exon20ins mutation, included in the phase 1 CHRYSALIS trial and cured at Institute Curie from November 1st 2019 until December 31st 2021 were selected for this work. Retrospectively, all cutaneous adverse events were registered and classified according to the CTCAE 6.0 classification, and actions we implemented to minimize and treat these adverse events were collected. We then developed guidelines based on these datas. RESULTS: A total of seven patients started amivantamab as monotherapy. The two most frequent dermatological adverse events were: acneiform rash and paronychia (100 % of patients). Other adverse events presented by the patients were reported: modification of hair growth with hypertrichosis in 50 % of men (n = 1/2) and hirsutism in 80 % of women (n = 4/5); skin abrasion of the scalp in 71 % (n = 5/7); and skin fissure in 57 % (n = 4/7). We recommend first a rigorous inspection of the skin and teguments to determine the risk rate to have dryer skin under treatment; second a prevention of paronychia/acneiform rash/and skin fissures with prophylactic tetracycline, skin moisturizing, and hygienic measures starting at least 14 days before treatment initiation; third a particular attention to the psychological impact of skin toxicities with access to psychological support. CONCLUSION: We propose here guidelines for the management of dermatological toxicities under amivantamab with a multidisciplinary approach for the proactive management of cutaneous toxicities with a focus on preventive actions.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Protein Kinase Inhibitors , Skin Diseases , Female , Humans , Male , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/genetics , Exanthema/chemically induced , Exanthema/prevention & control , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Paronychia/chemically induced , Paronychia/prevention & control , Protein Kinase Inhibitors/toxicity , Skin Diseases/chemically induced , Skin Diseases/prevention & control
4.
Pediatr Pulmonol ; 57(7): 1779-1781, 2022 07.
Article in English | MEDLINE | ID: mdl-35451238

ABSTRACT

As elexacaftor/tezacaftor/ivacaftor has proven to have robust clinical efficacy for eligible persons with cystic fibrosis, desensitization should be offered to those with maculopapular eruption hypersensitivity reactions to achieve tolerance. As presented in this case, if provided with tools for crushing and mixing the medication, a successful escalation protocol can be completed at home without coordinating the help of a compound pharmacy.


Subject(s)
Chloride Channel Agonists , Cystic Fibrosis , Exanthema , Hypersensitivity, Delayed , Aminophenols/adverse effects , Aminophenols/therapeutic use , Benzodioxoles/adverse effects , Benzodioxoles/therapeutic use , Chloride Channel Agonists/adverse effects , Chloride Channel Agonists/therapeutic use , Cystic Fibrosis/drug therapy , Drug Combinations , Exanthema/chemically induced , Exanthema/prevention & control , Humans , Hypersensitivity, Delayed/chemically induced , Hypersensitivity, Delayed/prevention & control , Mutation , Pyrazoles/adverse effects , Pyrazoles/therapeutic use , Pyridines/adverse effects , Pyridines/therapeutic use , Pyrrolidines/adverse effects , Pyrrolidines/therapeutic use , Quinolones/adverse effects , Quinolones/therapeutic use
5.
Oncologist ; 27(7): e554-e560, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35325241

ABSTRACT

BACKGROUND: Rash eruptions are a common side-effect of pemetrexed, for which the administration of 8 mg/day of dexamethasone for 3 days from the day preceding pemetrexed administration is recommended. This study aimed to prospectively assess the effectiveness of prophylactic administration of low-dose dexamethasone for pemetrexed-induced rashes. METHODS: This single-arm, phase II study recruited patients with non-squamous non-small cell lung cancer and malignant pleural mesothelioma scheduled to receive chemotherapy including pemetrexed. Patients received 2 mg of dexamethasone daily from days 2 to 6 after chemotherapy with pemetrexed. The primary endpoint was the 3-week incidence of rash eruptions. RESULTS: Twenty-five patients were enrolled between September 2017 and May 2019. The incidence of rash after 3 weeks was 16.7%. Rashes erupted mainly on the upper half of the body, such as the chest and neck, and were of grades 1 and 2 in 2 patients each. No rashes of grade 3 or higher were observed, and there were no adverse events associated with additional corticosteroids. CONCLUSION: Prophylactic administration of low-dose dexamethasone for 5 days from the day after pemetrexed administration resulted in a milder incidence and severity of rash. These findings may provide a standard preventative strategy for pemetrexed-induced rashes. (Trial identifier: UMIN000025666).


Subject(s)
Carcinoma, Non-Small-Cell Lung , Dexamethasone , Exanthema , Lung Neoplasms , Mesothelioma, Malignant , Pemetrexed , Adrenal Cortex Hormones/therapeutic use , Carcinoma, Non-Small-Cell Lung/complications , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin , Dexamethasone/therapeutic use , Exanthema/chemically induced , Exanthema/drug therapy , Exanthema/prevention & control , Humans , Lung Neoplasms/complications , Lung Neoplasms/drug therapy , Mesothelioma, Malignant/complications , Mesothelioma, Malignant/drug therapy , Pemetrexed/adverse effects
6.
J Clin Pharm Ther ; 47(4): 477-482, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34778985

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: It was previously reported that the incidence of lenalidomide (LEN)-induced skin rash is reduced by administration of bortezomib (BOR) prior to LEN administration in patients with multiple myeloma (MM). Therefore, we investigated whether LEN-induced skin rash is affected by the duration of BOR administration and the dosing interval between BOR and LEN administration. METHOD: A retrospective investigation was conducted among MM patients who received BOR treatment prior to LEN treatment in Eiju General Hospital from May 2010 to December 2020. We investigated whether the BOR administration duration and interval duration from the completion of BOR administration to the initial LEN administration affect the development of LEN-induced skin rash. RESULT AND DISCUSSION: Twenty-eight of the 81 patients exhibited LEN-induced skin rash (34.6%). The administered duration, but not the interval, was significantly longer in the group without skin rash. Cut-off values were set for the duration of administration and interval, which were 35 days and 30 days, respectively. Multivariate analysis was performed on patients which are administered duration of more than 35 days and intervals of less than 30 days, and those who are not applicable. A significant difference was observed in the incidence of skin rash for each factor. WHAT IS NEW AND CONCLUSION: The risk of reduced LEN-induced skin rash is affected not only by the presence of prior BOR administration, but also by the duration of BOR and the interval from the completion of BOR to the initial LEN administration.


Subject(s)
Exanthema , Multiple Myeloma , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bortezomib/adverse effects , Dexamethasone/therapeutic use , Exanthema/chemically induced , Exanthema/epidemiology , Exanthema/prevention & control , Humans , Lenalidomide/adverse effects , Multiple Myeloma/drug therapy , Retrospective Studies
7.
J Med Chem ; 64(16): 12163-12180, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34375113

ABSTRACT

Rash is one of the primary dose-limiting toxicities of Akt (protein kinase B) inhibitors in clinical trials. Here, we demonstrate the inhibition of Akt2 isozyme may be a driver for keratinocyte apoptosis, which promotes us to search for new selective Akt inhibitors with an improved cutaneous safety property. According to our previous research, compound 2 is selected for further optimization for overcoming the disadvantages of compound 1, including high Akt2 inhibition and high toxicity against HaCaT keratinocytes. The dihedral angle-based design and molecular dynamics simulation lead to the identification of Hu7691 (B5) that achieves a 24-fold selectivity between Akt1 and Akt2. Hu7691 exhibits low activity in inducing HaCaT apoptosis, promising kinase selectivity, and excellent anticancer cell proliferation potencies. Based on the superior results of safety property, pharmacokinetic profile, and in vivo efficacy, the National Medical Products Administration (NMPA) approved the investigational new drug (IND) application of Hu7691.


Subject(s)
Benzamides/therapeutic use , Exanthema/prevention & control , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazoles/therapeutic use , Animals , Benzamides/chemistry , Benzamides/metabolism , Benzamides/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Dogs , Female , HEK293 Cells , Humans , Keratinocytes/drug effects , Male , Mice, Inbred BALB C , Mice, Nude , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Neoplasms/complications , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/chemistry , Pyrazoles/metabolism , Pyrazoles/toxicity , Rats, Sprague-Dawley , Structure-Activity Relationship
8.
Dermatology ; 237(6): 988-994, 2021.
Article in English | MEDLINE | ID: mdl-33378750

ABSTRACT

BACKGROUND: The incidence of epidermal growth factor receptor inhibitor (EGFRI)-induced papulopustular rash is 60-85%. OBJECTIVE: To investigate prophylactic topical treatment for EGFRI-induced rash. METHODS: A single-center, randomized, double-blind, placebo-controlled trial. Adult cancer patients initiating treatment with EGFRIs were randomized to receive facial topical treatment with chloramphenicol 3% + prednisolone 0.5% (CHL-PRED) ointment, chloramphenicol 3% (CHL) ointment, or aqua cream (AQUA). The primary end points were the incidence of ≥grade 3 rash using the Common Terminology Criteria for Adverse Events (CTCAE), on days 14 and 30. A subanalysis was conducted for incidence of a protocol-specified significant rash, defined as ≥10 facial papulopustular lesions. RESULTS: The per-protocol analysis on day 14 included 69 patients, who received CHL-PRED (21), CHL (23), or AQUA (25). The incidence of CTCAE ≥grade 3 rash was not statistically significant between arms; however, the incidence of the protocol-specified significant rash was: CHL-PRED 14%, CHL 39%, and AQUA 48% (p = 0.03, CHL-PRED vs. AQUA). At 30 days, the CTCAE ≥grade 3 incidence was similar, but the incidences of protocol-specified significant rash were 6%, 16%, and 43% (p = 0.03, CHL-PRED vs. AQUA). No significant differences were found between CHL and CHL-PRED and between CHL and AQUA. CONCLUSIONS: Prophylactic topical CHL-PRED was efficacious when compared to AQUA, in the treatment of EGFRI-induced facial papulopustular rash.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Chloramphenicol/therapeutic use , ErbB Receptors/adverse effects , ErbB Receptors/antagonists & inhibitors , Exanthema/prevention & control , Protein Kinase Inhibitors/adverse effects , Administration, Topical , Adult , Aged , Aged, 80 and over , Anti-Inflammatory Agents/therapeutic use , Double-Blind Method , Exanthema/chemically induced , Female , Humans , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/pathology , Prednisolone/therapeutic use
11.
J Eur Acad Dermatol Venereol ; 34(2): 319-324, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31419350

ABSTRACT

BACKGROUND: Gut microbiome influences cutaneous diseases including atopic dermatitis. Possible impact of intrauterine exposure to meconium on the occurrence of dermatitis and skin rash was proposed. OBJECTIVE: We investigated the possible influence of intrauterine exposure to meconium-stained amniotic fluid (MSAF) on the occurrence of dermatitis and skin rash-related hospitalizations throughout childhood. METHODS: Singleton deliveries occurring between 1991 and 2014 at a single medical centre were divided into two study groups based on presence or lack of MSAF during delivery. Population-based cohort analysis, Kaplan-Meier survival analysis and Cox proportional hazards model were used to study the association between MSAF and cutaneous morbidity-related hospitalizations. RESULTS: A lower rate of the total dermatitis or skin eruption-related hospitalization was documented in the MSAF-exposed group; 0.78 per 1000-person years (0.9%, n = 312), as compared to 0.98 per 1000-person years in the unexposed group (1.0%, n = 1992) with a hazard ratio of 0.86 (95% CI 0.76-0.96, P = 0.011). The survival curve showed lower cumulative hospitalization rate in the MSAF-exposed group as compared to the unexposed group (log rank P = 0.01). The Cox analysis, controlled for confounders, demonstrated MSAF exposure to be an independent protective factor for dermatitis and skin rash-related hospitalizations during childhood (adjusted HR 0.878 (95% CI 0.779-0.990, P = 0.034). CONCLUSION: Fetal exposure to MSAF appears to be an independent protective factor for dermatitis and skin rash-related hospitalizations in the offspring throughout childhood and adolescence.


Subject(s)
Amniotic Fluid , Dermatitis/prevention & control , Exanthema/prevention & control , Hospitalization , Meconium , Protective Factors , Adult , Cohort Studies , Female , Humans , Infant, Newborn , Pregnancy , Retrospective Studies
12.
Allergy Asthma Proc ; 40(5): 321-328, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31345280

ABSTRACT

Background: Although clinical trials documented omalizumab's efficacy in U.S. patients with chronic idiopathic urticaria (CIU), the real-world evidence on its long-term effectiveness is lacking. Objective: To assess omalizumab use and the long-term response in a large sample of U.S. real-world patients. Methods: Patients with CIU and ≥ 12 years old who were initiated on omalizumab (index date) and with ≥ 6 months of postindex data were identified in an electronic medical record system (2007-2018). Omalizumab use was described. Provider assessments of disease control and course, and patient-reported symptoms were compared at 6-month intervals postindex versus baseline in the patients with values available at both time points. Results: A total of 1096 patients (mean age, 44.1 years; 74.7% women) were followed up for a mean of 19 months postindex. Patients, predominantly initiated on a 300-mg dose, received a mean of 15 omalizumab administrations and were treated continuously for a mean of 14.2 months. At 6 months postindex versus baseline, the patients (n = 708) were more likely to be well controlled (odds ratio [OR] 31.68 [95% confidence interval {CI}, 17.20-58.36]) with an improved disease course (OR 15.73 [95% CI, 11.33-21.85]). Moreover, the patients (n = 373) were less likely to report itching (OR 0.39 [95% CI, 0.21-0.76]), rash (OR 0.59 [95% CI, 0.45-0.78]), and swelling (OR 0.46 [95% CI, 0.36-0.59]). Benefits associated with omalizumab treatment were sustained through month 24 and beyond. Conclusion: This real-world study showed that the patients who received a mean of 15 omalizumab administrations over a mean of 14.2 months experienced, starting at 6 and through 24 months after omalizumab initiation and beyond, improved CIU control, course, and symptoms.


Subject(s)
Chronic Urticaria/therapy , Omalizumab/therapeutic use , Adolescent , Adult , Child , Chronic Urticaria/etiology , Edema/prevention & control , Electronic Health Records , Exanthema/prevention & control , Female , Follow-Up Studies , Humans , Male , Middle Aged , Odds Ratio , Omalizumab/administration & dosage , Pruritus/prevention & control , Time Factors , Treatment Outcome , United States
13.
Cochrane Database Syst Rev ; 7: CD010891, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31314143

ABSTRACT

BACKGROUND: Drug-induced skin reactions present with a range of clinical symptoms, from mild maculopapular skin rashes to potentially fatal blistering skin rashes - such as Stevens-Johnson syndrome (SJS) or toxic epidermal necrolysis (TEN) - which may result in death. Milder reactions may be troublesome and lead to low drug compliance. The pathogenesis of these drug reactions is not yet fully understood; however, there is evidence that pretreatment genetic testing may help to predict and prevent these reactions in some cases. OBJECTIVES: To assess the effects of prospective pharmacogenetic screening to reduce drug-associated skin reactions in a patient population. SEARCH METHODS: We searched the following databases up to July 2018: the Cochrane Skin Specialised Register, CENTRAL, MEDLINE, Embase and LILACS. We also searched five trials registers, and checked the reference lists of included studies and relevant reviews for further references to relevant randomised controlled trials (RCTs). SELECTION CRITERIA: We included RCTs of participants who had prospective pharmacogenetic screening to determine genetic variants associated with hypersensitivity reactions, compared with those who did not have prospective pharmacogenetic screening. We included participants in any setting, who were of any age, gender, and ethnicity, who had been prescribed drugs known to cause delayed type hypersensitivity reactions. DATA COLLECTION AND ANALYSIS: We used standard methodological procedures expected by Cochrane. To assess studies for inclusion, two review authors independently screened all of the titles and abstracts of publications identified by the searches. Because there was only one included study, many of the planned data analyses were not applicable to the review. We used GRADE to assess the quality of the included study.The review's primary outcomes were the incidence of severe skin rashes with systemic symptoms (such as fever and multiple organ involvement), and long-term effects (such as scarring of eyelids or lung tissue). Secondary outcomes were hospitalisation for drug-induced skin reactions, blistering skin reactions (such as SJS, hypersensitivity (HSS) syndrome), and death. MAIN RESULTS: One study, which was a randomised, double-blind, controlled, multicentre trial, fulfilled our inclusion criteria. The trial included 1956 adult participants (74% men, with a mean age of 42 years) across 265 centres (medical centres, hospitals, outpatient clinics) in 19 countries around the world who were infected with HIV-type 1 and who had not received abacavir previously. The participants, who had a clinical need for treatment with an antiretroviral-drug regimen containing abacavir, were randomly assigned to undergo prospective human leukocyte antigen (HLA) Class I, locus B, allele 57:01 (HLA-B*57:01) screening (prospective-screening group) before this treatment, or to undergo a standard-care approach of abacavir use without prospective HLA-B*57:01 screening (control group). Participants who tested positive for HLA-B*57:01 were not given abacavir; instead, they received antiretroviral therapy that did not include abacavir. The control group did have retrospective HLA-B*57:01 pharmacogenetic testing. The trial duration was six months. Each participant was observed for six weeks. Assessments were performed at the time of study entry, at baseline (day one of abacavir treatment), and at weeks one, two and six. This study was funded by the manufacturer of abacavir, GlaxoSmithKline.The study did not assess any of our primary outcomes, and it measured none of our secondary outcomes in isolation. However, it did assess an outcome of (characteristically severe) hypersensitivity reaction which included (but was not limited to) our secondary outcomes of HSS and SJS/TEN.The study demonstrated that prospective HLA-B*57:01 screening probably reduces the incidence of hypersensitivity reaction to abacavir. The incidence of clinically diagnosed HSS reaction to abacavir was lower in the screening arm (risk ratio (RR) 0.43, 95% confidence interval (CI) 0.28 to 0.67; 1650 participants; moderate-quality evidence), as was immunologically confirmed HSS reaction (RR 0.02, 95% 0.00 to 0.37; 1644 participants; moderate-quality evidence). A positive result from an epicutaneous patch test performed six to ten weeks after clinical diagnosis provided immunological confirmation.Overall, the study demonstrates a low risk of bias across five out of seven domains. There was a high risk of detection bias because hypersensitivity reactions were diagnosed by the principal investigator at the recruitment site without the use of predefined clinical criteria. Although there was also high risk of attrition bias due to excluding participants with incomplete follow-up from analyses, the authors did undertake a series of sensitivity analyses based on the intention-to-treat population, which demonstrated consistent results with the primary analysis. We rated the study quality as moderate-quality using GRADE criteria. AUTHORS' CONCLUSIONS: Prospective screening for HLA-B*57:01 probably reduces severe hypersensitivity skin reactions to abacavir in patients positive for HIV-type 1. However, these results are only based on one study, which was at high risk of attrition and detection bias.Our primary outcomes (incidence of severe skin rashes with systemic symptoms, and long-term effects) were not assessed by the trial, and only one of the review's secondary outcomes was measured (hypersensitivity reaction); thus, we found no evidence relating to hospitalisation, death, or long-term conditions resulting from drug injury.We found no eligible evidence on genetic testing for severe drug-induced skin rash in relation to different drugs and classes of drugs. Further clinical trials based on other drugs, and in different patient populations, would be useful for advising policy changes for improving the prevention of adverse skin reactions to drug treatments.


Subject(s)
Exanthema/genetics , Exanthema/prevention & control , Genetic Testing , Humans , Randomized Controlled Trials as Topic , Stevens-Johnson Syndrome/genetics , Stevens-Johnson Syndrome/prevention & control
15.
Exp Cell Res ; 376(2): 210-220, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30690028

ABSTRACT

Cetuximab, an inhibitor of the epidermal growth factor receptor that is used widely to treat human cancers including oral squamous cell carcinoma (OSCC), has characteristic side effects of skin rash and hypomagnesemia. However, the mechanisms of and therapeutic agents for skin rashes and hypomagnesemia are still poorly understood. Our gene expression profiling analyses showed that cetuximab activates the p38 MAPK pathways in human skin cells (human keratinocyte cell line [HaCaT]) and inhibits c-Fos-related signals in human embryonic kidney cells (HEK293). We found that while the p38 inhibitor SB203580 inhibited the expression of p38 MAPK targets in HaCaT cells, flavagline reactivated c-Fos-related factors in HEK293 cells. It is noteworthy that, in addition to not interfering with the effect of cetuximab by both compounds, flavagline has additive effect for OSCC growth inhibition in vivo. Collectively, our results indicate that combination of cetuximab and these potential therapeutic agents for cetuximab-related toxicities could be a promising therapeutic strategy for patients with OSCC.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Carcinoma, Squamous Cell/drug therapy , Cetuximab/adverse effects , Growth Inhibitors/therapeutic use , Imidazoles/therapeutic use , Mouth Neoplasms/drug therapy , Pyridines/therapeutic use , Animals , Carcinoma, Squamous Cell/complications , Cell Line, Tumor , Drug Therapy, Combination , ErbB Receptors/antagonists & inhibitors , Exanthema/chemically induced , Exanthema/genetics , Exanthema/prevention & control , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Growth Inhibitors/adverse effects , Growth Inhibitors/antagonists & inhibitors , HEK293 Cells , Humans , Hypercalciuria/chemically induced , Hypercalciuria/genetics , Hypercalciuria/prevention & control , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/complications , Mouth Neoplasms/genetics , Nephrocalcinosis/chemically induced , Nephrocalcinosis/genetics , Nephrocalcinosis/prevention & control , Renal Tubular Transport, Inborn Errors/chemically induced , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/prevention & control , Transcriptome , Xenograft Model Antitumor Assays
16.
Int Immunopharmacol ; 69: 79-87, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30682720

ABSTRACT

Cutaneous toxicities are the commonest side effects in patients with cancer treated using epidermal growth factor receptor inhibitors such as erlotinib. For patients with such toxicities, there is a lack of safe, effective pharmacological agents. Here we established a skin toxicity model and investigated the preventive and therapeutic effect of Diallyl Trisulfide (DATS) in vivo. The mouse skin toxicities model was established through continuous administration of erlotinib for 49 days. Meanwhile, the mice in the experimental group underwent DATS treatment for 49 days. Hematoxylin and eosin (HE) staining and oil red O staining of back and limb skin was performed to determine whether DATS aqueous extract can reverse the skin toxicities caused by erlotinib. Compared with the erlotinib group, the incidence of rash in the DATS group was lower. In addition, in the DATS group, the degree of skin redness and herpes was mild, the body weight was stable, and the activity was favorable. By comparing the HE and oil red O staining results for the mouse skin, the degree of keratin hyperplasia was determined to be lower in the experimental group than in the erlotinib group, and the number of purulent neutrophils decreased. The number of follicles was relatively less. The release of TNF-α, IL-6 and other inflammatory factors was reduced by DATS. Erlotinib hydrochloride can cause severe skin toxicities, and DATS prevents skin toxicities, its mechanism may be related to DATS reduced erlotinib-induced inflammatory injury.


Subject(s)
Allyl Compounds/therapeutic use , Antineoplastic Agents/adverse effects , Drug-Related Side Effects and Adverse Reactions/prevention & control , Erlotinib Hydrochloride/adverse effects , Exanthema/prevention & control , Neoplasms/drug therapy , Protein Kinase Inhibitors/adverse effects , Skin/drug effects , Sulfides/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Disease Models, Animal , ErbB Receptors , Erlotinib Hydrochloride/therapeutic use , Exanthema/etiology , Female , Humans , Mice , Mice, Inbred BALB C , Protein Kinase Inhibitors/therapeutic use , Skin/pathology
17.
Mult Scler Relat Disord ; 29: 7-14, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30654246

ABSTRACT

BACKGROUND: Alemtuzumab is a humanized IgG monoclonal antibody approved in more than 60 countries for patients with relapsing remitting multiple sclerosis (RRMS). In phase 2 and 3 clinical trials (CAMMS223 (NCT00050778), CARE-MS I (NCT00530348), and CARE-MS II (NCT00548405)), patients receiving alemtuzumab demonstrated significantly greater improvements on clinical and MRI outcomes versus SC IFNß-1a; mild to moderate infusion-associated reactions (IARs) were the most frequently reported adverse events (AEs) associated with alemtuzumab. EMERALD (NCT02205489) was a phase 4, multicenter, multinational, single-arm study designed to assess an algorithm for the prevention and management of IARs in RRMS patients treated with alemtuzumab. METHODS: Patients were treated with a study regimen of enhanced IAR prophylaxis relative to phase 2 and 3 studies. H1 and/or H2 antagonists or equivalent gastroprotection (proton pump inhibitors) were given 1 day before alemtuzumab infusion, 1 h prior to the infusion, and post-infusion. Methylprednisolone was given orally 1 day before infusion, 1 h prior to the infusion, and as needed post-infusion. Antipyretics were given 1 h before infusion and as needed post-infusion. Anti-emetics and normal saline were given as needed during and post-infusion. RESULTS: Of the 61 patients screened, 58 (95.1%) were enrolled into the study. Of the 58 patients who received the first infusion of Period 1, 57 (98.3%) completed the 5 days of Course 1. A total of 54 patients received the first infusion of Period 2 and 53 completed the 3-day course. All patients (n = 58) completed the Month 6 visit and 54 the Month 12 visit. 93.1% of patients had at least one IAR (91.4% in Period 1 and 81.5% in Period 2), the majority of which were grade 1 (69.1%) or grade 2 (28.0%). The three most common IARs of headache, pyrexia, and rash occurred in 48.8%, 40.7%, and 24.1% of patients during the first course and 14.8%, 17.2%, and 5.6% of patients during the second course, respectively. The majority of IARs occurred within 6 h after the start of alemtuzumab infusion, with a peak during the first 2 h. The types and overall incidence of IARs were consistent with phase 2 and 3 trials. Frequency and distribution of rash were reduced in the EMERALD study compared with previous clinical trials. Serious IARs occurred in 15.5%, a higher rate than reported in clinical trials of alemtuzumab. CONCLUSION: Although most alemtuzumab-treated patients experienced IARs as in previous controlled clinical studies, there was an improvement in the frequency and distribution of alemtuzumab-associated rash, which may have been associated with this study's prophylaxis regimen.


Subject(s)
Alemtuzumab/adverse effects , Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal, Humanized/adverse effects , Drug-Related Side Effects and Adverse Reactions , Exanthema , Immunologic Factors/adverse effects , Infusions, Intravenous/adverse effects , Methylprednisolone/pharmacology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Outcome Assessment, Health Care , Adult , Alemtuzumab/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antipyretics/pharmacology , Drug Therapy, Combination , Drug-Related Side Effects and Adverse Reactions/drug therapy , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/etiology , Drug-Related Side Effects and Adverse Reactions/prevention & control , Exanthema/chemically induced , Exanthema/drug therapy , Exanthema/epidemiology , Exanthema/prevention & control , Female , Histamine Antagonists/pharmacology , Humans , Immunologic Factors/administration & dosage , Incidence , Male , Methylprednisolone/administration & dosage , Middle Aged , Proton Pump Inhibitors/pharmacology
18.
Dermatol Ther ; 32(4): e12665, 2019 07.
Article in English | MEDLINE | ID: mdl-30216601

ABSTRACT

Tropical regions receive a significant part of the traveling population. It is very important that health professionals are familiar with the main tropical skin diseases and able to advice patients appropriately. This article reviews the main tropical diseases of travelers, with an emphasis on diagnosis, management, and prevention. Among others, cutaneous larva migrans, myiasis, tungiasis, Chagas disease, Dengue fever, African trypanosomiasis, filariasis, and leishmaniasis are discussed. Increasing awareness among travelers and health care professionals can help reduce morbidity and mortality. Continued research on new drugs and vaccines is needed to reduce the risks of tropical diseases.


Subject(s)
Skin Diseases/therapy , Travel , Chagas Disease/diagnosis , Chagas Disease/prevention & control , Chagas Disease/therapy , Exanthema/diagnosis , Exanthema/prevention & control , Exanthema/therapy , Humans , Larva Migrans/diagnosis , Larva Migrans/prevention & control , Larva Migrans/therapy , Leishmaniasis/diagnosis , Leishmaniasis/prevention & control , Leishmaniasis/therapy , Myiasis/diagnosis , Myiasis/prevention & control , Myiasis/therapy , Scabies/diagnosis , Scabies/prevention & control , Scabies/therapy , Skin Diseases/diagnosis , Skin Diseases/prevention & control , Trypanosomiasis, African/diagnosis , Trypanosomiasis, African/prevention & control , Trypanosomiasis, African/therapy , Tungiasis/diagnosis , Tungiasis/prevention & control , Tungiasis/therapy , Yellow Fever/diagnosis , Yellow Fever/prevention & control , Yellow Fever/therapy
19.
Oncology (Williston Park) ; 32(7): 339-43, 2018 07 15.
Article in English | MEDLINE | ID: mdl-30080919

ABSTRACT

Poly(ADP-ribose) polymerase (PARP) proteins are used by cells in several DNA repair processes. PARP inhibition can result in preferential death of cancer cells when another mechanism for repairing DNA is defective. Two PARP inhibitors, olaparib and rucaparib, have been approved by the US Food and Drug Administration (FDA) for the treatment of recurrent, BRCA-associated ovarian cancer. More recently, these two and a third PARP inhibitor, niraparib, were approved by the FDA as maintenance therapy following platinum-based chemotherapy for recurrent ovarian cancer. This has caused a paradigm shift in disease management and a challenge for clinicians, who must decide how best to use these agents in individualized treatment. The oral formulation is attractive to patients, but adverse effects such as nausea and fatigue can impact quality of life. As clinicians become comfortable selecting PARP inhibitors and managing associated toxicities, future steps will be to investigate how to safely administer them in combination with other therapies.


Subject(s)
Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Anemia/chemically induced , Anemia/prevention & control , Creatinine/blood , Drug Administration Schedule , Drug Interactions , Exanthema/chemically induced , Exanthema/prevention & control , Fatigue/chemically induced , Fatigue/prevention & control , Female , Genes, BRCA1 , Genes, BRCA2 , Heart Rate/drug effects , Humans , Hypertension/chemically induced , Hypertension/prevention & control , Indazoles/therapeutic use , Leukopenia/chemically induced , Leukopenia/prevention & control , Mutation , Myelodysplastic Syndromes/chemically induced , Nasopharyngitis/chemically induced , Nasopharyngitis/prevention & control , Nausea/chemically induced , Nausea/prevention & control , Ovarian Neoplasms/genetics , Piperidines/therapeutic use , Pneumonia/chemically induced , Poly(ADP-ribose) Polymerase Inhibitors/economics , Thrombocytopenia/chemically induced , Thrombocytopenia/prevention & control , Transaminases/blood , Vomiting/chemically induced , Vomiting/prevention & control
20.
Ann Ig ; 30(4 Supple 1): 23-27, 2018.
Article in English | MEDLINE | ID: mdl-30062376

ABSTRACT

The Herpes Zoster vaccine is strongly recommended by the World Health Organization to promote healthy aging by preventing the corresponding age- related disease, also named shingles. The disease is due to the endogenous reactivation of Varicella Zoster Virus, the causal agent of chickenpox, that becomes latent at the peripheral nervous system level. Here, owing to the host's cell-mediated immunity, it may be confined for several decades. However, the immune senescence allows the possibility of virus reactivation, causing the onset of neuropathic pain and skin rash that characterize the acute disease. Sometimes, the neuralgia becomes chronic causing postherpetic neuralgia that has a significant impact on the quality of patient life, analogously to the ophthalmic HZ, a particularly feared form of disease. Due to the causal relationship between decreasing immune defenses and virus reactivation with disease onset, the incidence of Herpes Zoster, in Italy now equal to 6.42 (95%CI: 5.93-6.95) cases per 1,000 people per year will increase steadily in the future due to the longevity rise of the population. Considering epidemiological impact, complications and sequelae in the short- and long-term, costs of clinical-therapeutical management of patients, and, above all, the poor effectiveness of available therapy the only effective intervention is vaccination of the elderly. Currently in the European Union, there is only one vaccine for Herpes Zoster prevention, formed by live attenuated OKA-Merck virus strain that is also used for paediatric vaccine. According to the Health Technology Assessment surveys, the intervention cost (based on "Quality Adjusted Life Years") is clearly below the discriminating threshold value to judge the feasibility and, as predicted by the Italian National Plan of Vaccinal Prevention 2017-2019, the vaccine is offered free to all subjects >65 years.


Subject(s)
Herpes Zoster Vaccine/administration & dosage , Herpes Zoster/prevention & control , Neuralgia, Postherpetic/prevention & control , Secondary Prevention , Skin Diseases, Viral/prevention & control , Age Factors , Aged , Aging/immunology , Exanthema/prevention & control , Exanthema/virology , Herpes Zoster/epidemiology , Herpes Zoster/virology , Humans , Immunocompromised Host , Italy/epidemiology , Quality-Adjusted Life Years , Skin Diseases, Viral/epidemiology , Skin Diseases, Viral/virology , Virus Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...