Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Eur Acad Dermatol Venereol ; 37(2): 348-355, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36300769

ABSTRACT

INTRODUCTION/BACKGROUND: Manifestations of psoriasis in special areas are difficult to treat and are associated with a high disease burden and significant quality of life (QoL) impairment. Topical therapies may be inadequate for these patients, necessitating systemic treatment. OBJECTIVE: The objective of EMBRACE was to evaluate the impact on QoL, efficacy and safety of apremilast 30 mg BID in patients with limited skin involvement with plaque psoriasis manifestations in special areas and impaired QoL. METHODS: EMBRACE (NCT03774875) was a phase 4, randomized, placebo-controlled, multinational study. Patients had plaque psoriasis not controlled by topical therapy; lack of response, contraindication or intolerance to conventional first-line systemic therapy; psoriasis in ≥1 special area (including visible locations, scalp, nails, genital areas or palmoplantar areas); Psoriasis Area and Severity Index (PASI) ≥3 to ≤10; and Dermatology Life Quality Index (DLQI) >10. The primary endpoint was DLQI response (≥4-point reduction) at Week 16. RESULTS: Of 277 randomized patients (apremilast: n = 185; placebo: n = 92), 221 completed Week 16 (apremilast: n = 152; placebo: n = 69). The primary endpoint (≥4-point reduction in DLQI at Week 16) was met by significantly more patients receiving apremilast (73.3%) versus placebo (41.3%; p < 0.0001). Significantly greater improvement in affected body surface area (BSA) and PASI was observed with apremilast versus placebo at Week 16. There were also significantly greater improvements with apremilast versus placebo in itch numeric rating scale (-2.5 vs. -0.9, p < 0.0001) and skin discomfort/pain visual analog scale (-21.5 vs. -5.4, p = 0.0003) and greater achievement of Patient Benefit Index ≥1 (77% vs. 40%, p < 0.0001) at Week 16. No new safety signals were observed. CONCLUSIONS: Apremilast significantly improved skin-related QoL in patients with limited skin involvement with plaque psoriasis in special areas and highly impaired QoL. The safety profile was consistent with prior apremilast studies.


Subject(s)
Phosphodiesterase 4 Inhibitors , Psoriasis , Humans , Quality of Life , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Double-Blind Method , Phosphodiesterase 4 Inhibitors/therapeutic use , Severity of Illness Index , Psoriasis/complications , Psoriasis/drug therapy , Psoriasis/chemically induced , Treatment Outcome
2.
Pain ; 159(5): 987-994, 2018 May.
Article in English | MEDLINE | ID: mdl-29419653

ABSTRACT

The long-term safety of naldemedine, a peripherally acting µ-opioid receptor antagonist, was evaluated in patients with opioid-induced constipation and chronic noncancer pain in a 52-week, randomized, double-blind, phase 3 study. Eligible adults who could be on a routine laxative regimen were randomized 1:1 to receive once-daily oral naldemedine 0.2 mg (n = 623) or placebo (n = 623). The primary endpoint was summary measures of treatment-emergent adverse events (AEs). Additional endpoints included opioid withdrawal on the Clinical Opiate Withdrawal Scale and the Subjective Opiate Withdrawal Scale, pain intensity on Numeric Rating Scale, frequency of bowel movements, and constipation-related symptoms and quality of life on the Patient Assessment of Constipation Symptoms and Patient Assessment of Constipation Quality of Life scales, respectively. Treatment-emergent AEs (naldemedine, 68.4% vs placebo, 72.1%; difference: -3.6% [95% confidence interval: -8.7 to 1.5]) and treatment-emergent AEs leading to study discontinuation (6.3% vs 5.8%; difference: 0.5% [-2.2 to 3.1)] were reported for similar proportions of patients. Diarrhea was reported more frequently with naldemedine (11.0%) vs placebo (5.3%; difference: 5.6% [2.6-8.6]). There were no meaningful differences between groups in opioid withdrawal or pain intensity. Sustained significant improvements in bowel movement frequency and overall constipation-related symptoms and quality of life were observed with naldemedine (P ≤ 0.0001 vs placebo at all time points). Naldemedine was generally well tolerated for 52 weeks and did not interfere with opioid-mediated analgesia or precipitate opioid withdrawal. Naldemedine significantly increased bowel movement frequency, improved symptomatic burden of opioid-induced constipation, and increased patients' quality of life vs placebo.


Subject(s)
Analgesics, Opioid/adverse effects , Chronic Pain/drug therapy , Constipation/drug therapy , Naltrexone/analogs & derivatives , Narcotic Antagonists/therapeutic use , Adult , Aged , Analgesics, Opioid/therapeutic use , Constipation/chemically induced , Double-Blind Method , Female , Humans , Male , Middle Aged , Naltrexone/adverse effects , Naltrexone/therapeutic use , Narcotic Antagonists/adverse effects , Quality of Life , Treatment Outcome
3.
Lancet Gastroenterol Hepatol ; 2(8): 555-564, 2017 08.
Article in English | MEDLINE | ID: mdl-28576452

ABSTRACT

BACKGROUND: Opioid-induced constipation is a frequent side-effect of opioid treatment, and standard interventions have limited or inconsistent efficacy. This study assessed the efficacy and safety of naldemedine, a peripherally acting µ-opioid receptor antagonist, for the treatment of opioid-induced constipation in patients with chronic non-cancer pain. METHODS: We report two double-blind, randomised, placebo-controlled trials in adults with chronic non-cancer pain and opioid-induced constipation. The first (COMPOSE-1) was done in 68 outpatient sites in seven countries and the second (COMPOSE-2) at 69 outpatient sites in six countries; both studies were done in Europe and the USA. Eligible patients were aged 18-80 years, did not use laxatives, and had a stable opioid regimen for treatment of chronic non-cancer pain with a total daily dose averaging at least 30 mg (morphine equivalent) for at least 1 month before screening. Patients were randomly assigned (1:1) to receive either oral naldemedine 0·2 mg or matching placebo once a day for 12 weeks. Randomisation was stratified by average total daily opioid dose (30-100 mg and >100 mg equivalents of oral morphine sulphate). The primary endpoint was proportion of responders. A responder had at least three spontaneous bowel movements (SBMs) per week with an increase from baseline of at least one SBM per week for at least 9 weeks of the 12-week treatment period including at least three of the last 4 weeks. Efficacy endpoints were analysed by intention to treat and the safety population included all patients who received at least one dose of study drug. These trials have both been completed and are registered with ClinicalTrials.gov, numbers NCT01965158 and NCT01993940. FINDINGS: In COMPOSE-1, 547 patients were recruited between Aug 29, 2013, and Jan 22, 2015, and were randomly assigned to receive naldemedine (n=274) or placebo (n=273). Patients for COMPOSE-2 were recruited between Nov 4, 2013, and June 9, 2015; 553 patients were randomly assigned to receive naldemedine (n=277) or placebo (n=276). Five patients were enrolled at more than one site, so were excluded from the intention-to-treat population (COMPOSE-1: one per group; COMPOSE-2: one in the naldemedine group, two from the placebo group), with intention-to-treat group sizes of 273 in the naldemedine group and 272 in the placebo group in COMPOSE-1, and 276 in the naldemedine group and 274 in the placebo group in COMPOSE-2. The proportion of responders in both trials was significantly higher with naldemedine than with placebo in COMPOSE-1 (130 responders [47·6%] of 273 in the naldemedine group vs 94 responders [34·6%] of 272 in the placebo group, difference 13·0% [95% CI 4·8-21·3]; p=0·002) and in COMPOSE-2 (145 [52·5%] of 276 vs 92 [33·6%] of 274, difference 18·9% [10·8-27·0]; p<0·0001). Incidence of adverse events with naldemedine was similar to placebo (COMPOSE-1: 132 [49%] of 271 in the naldemedine group vs 123 [45%] of 272 in the placebo group; COMPOSE-2: 136 [50%] of 271 vs 132 [48%] of 274). Treatment-related adverse events were noted in 59 (22%) of 271 patients in the naldemedine group and 45 (17%) of 272 in the placebo group in COMOPOSE-1, and in 54 (20%) of 271 patients in the naldemedine group and 31 (11%) of 274 in the placebo group of COMPOSE-2; the between-group differences were largely due to gastrointestinal disorders, which were more common with naldemedine than placebo (COMPOSE-1: 40 [15%] patients in the naldemedine group vs 18 [7%] in the placebo group; COMPOSE-2: 42 [16%] vs 20 [7%]). INTERPRETATION: Naldemedine treatment led to a significantly higher responder rate than did placebo and was generally well tolerated. These results support that naldemedine could be a new option for the treatment of opioid-induced constipation in patients with chronic non-cancer pain. FUNDING: Shionogi & Co, Ltd.


Subject(s)
Analgesics, Opioid/adverse effects , Constipation/chemically induced , Constipation/drug therapy , Naltrexone/analogs & derivatives , Narcotic Antagonists/adverse effects , Narcotic Antagonists/therapeutic use , Receptors, Opioid, mu/antagonists & inhibitors , Administration, Oral , Adult , Aged , Aged, 80 and over , Chronic Pain/drug therapy , Chronic Pain/etiology , Defecation/drug effects , Double-Blind Method , Drug Administration Schedule , Female , Humans , Intention to Treat Analysis , Male , Middle Aged , Naltrexone/adverse effects , Naltrexone/therapeutic use , Young Adult
4.
Mo Med ; 113(1): 72-8, 2016.
Article in English | MEDLINE | ID: mdl-27039496

ABSTRACT

BACKGROUND: Prescribing opioids for chronic non-cancer pain (CNCP) is a challenge due to associated risks from abuse, addiction and adverse effects. We surveyed resident physicians on their knowledge, attitude and practices in opioid prescription practices in the ambulatory setting and conducted an educational module to address their knowledge gaps. METHODS: A phase 1 survey assessed knowledge, attitudes and practices of residents in the out-patient management of CNCP with opioids. Demographics, numbers of patients seen, those with concerns for risky behaviors, adverse effects and the reasons for concern were also recorded. In Phase 2, an educational module in the form of didactics and case based discussions addressed the perceived deficiencies noted from results of phase 1 survey. Pre and post module surveys assessed the effectiveness of the educational module. RESULTS: In the phase 1 study (45/49, 92% response rate, M:F = 30:15) 33.3% (15/45) were in Post-Graduate Year (PGY) 1, 35.6% (16/45) PGY2s and 31.1% (14/45) PGY3s; 80% (36/45) saw more than one patient with CNCP in the previous 3 months; 62.2% (28/45) had at least one patient with concerns for misuse and addiction; 77.8% (35/45) and 86.7% (39/45) reported a lack of training and consistent documentation respectively, and 82.2% (37/45) were uncomfortable to refill for other provider's patients. All (100%, 45/45) consulted the clinical pharmacist; 86.7% (39/45) believed that either focused education would be beneficial. In the phase 2 study (44/49, 89.7% response rate, M: F = 29: 15), the pre- and post-module responses showed that > 90% of the residents perceived improvement in knowledge and confidence in management of CNCP with opioids after the educational module. CONCLUSIONS: Internal medicine residents perceived deficits in their ability to manage CNCP. Following a focused educational training, residents' knowledge and confidence in prescription of opioids improved, demonstrating the need to include management of CNCP with opioids into their curriculum.


Subject(s)
Analgesics, Opioid/therapeutic use , Attitude of Health Personnel , Chronic Pain/drug therapy , Internship and Residency , Pain Management , Adult , Cross-Sectional Studies , Female , Humans , Male , Practice Patterns, Physicians'
5.
J Urol ; 190(3): 958-64, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23454404

ABSTRACT

PURPOSE: Previous studies suggest a lower dose of desmopressin orally disintegrating tablet may be effective in females compared to males with nocturia. We confirm the efficacy and safety of 25 µg desmopressin orally disintegrating tablet compared to placebo in female patients. MATERIALS AND METHODS: In this 3-month, randomized, double-blind, parallel group study 25 µg desmopressin once daily was compared to placebo in women with nocturia (2 or more nocturnal voids). The co-primary efficacy end points were change from baseline in mean number of nocturnal voids and proportion of patients achieving at least a 33% reduction from baseline in the mean number of nocturnal voids (33% responders). RESULTS: The full analysis set comprised 261 patients (age range 19 to 87 years). Desmopressin significantly reduced the mean number of nocturnal voids and increased the odds of a 33% or greater response compared to placebo during 3 months, assessed by longitudinal analysis (-0.22, p = 0.028 and OR 1.85, p = 0.006, respectively). Desmopressin increased the mean time to first nocturnal void by 49 minutes compared to placebo at 3 months (p = 0.003). The response to desmopressin was seen by week 1 of treatment and was sustained throughout the trial. Significant increases in health related quality of life and sleep quality were observed compared to placebo. Desmopressin was well tolerated. Serum sodium levels remained greater than 125 mmol/L throughout the trial and 3 transient decreases to less than 130 mmol/L were recorded. CONCLUSIONS: At a dose of 25 µg, desmopressin orally disintegrating tablet is an effective and well tolerated treatment for women with nocturia. Treatment provides rapid and sustained improvement in nocturia and quality of life.


Subject(s)
Antidiuretic Agents/therapeutic use , Deamino Arginine Vasopressin/therapeutic use , Nocturia/diagnosis , Nocturia/drug therapy , Administration, Oral , Adult , Aged , Aged, 80 and over , Antidiuretic Agents/adverse effects , Deamino Arginine Vasopressin/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Middle Aged , Multivariate Analysis , Patient Safety , Reference Values , Risk Assessment , Severity of Illness Index , Treatment Outcome , Young Adult
8.
Clin Cancer Res ; 17(4): 880-7, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21131556

ABSTRACT

PURPOSE: BMS-641988 is an androgen receptor antagonist with increased potency relative to bicalutamide in both in vitro and in vivo prostate cancer models. A first-in-man phase I study was conducted to define the safety and tolerability of oral BMS-641988 in patients with castration-resistant prostate cancer (CRPC). EXPERIMENTAL DESIGN: Doses were escalated from 5 to 150 mg based on discrete pharmacokinetic parameters in cohorts of three to six subjects. After establishing safety with 20 mg of BMS-641988 in the United States, a companion study was opened in Japan to assess differences in drug metabolism between populations. RESULTS: Sixty-one men with CRPC were treated with daily BMS-641988. The pharmacokinetics (PK) of BMS-641988 and its active metabolites were proportional to dose. One patient experienced an epileptic seizure at a dose of 60 mg administered twice. Despite achieving target drug exposures, antitumor activity was limited to one partial response. Seventeen of 23 evaluable patients (74%) exhibited stable disease on imaging (median 15 weeks; range 8-32), and 10 of 61 patients (16%) achieved a ≥ 30% decline in levels of prostate-specific antigen (PSA). Partial agonism was seen within the context of this study upon removal of the drug as evidenced by a decrease in PSA. CONCLUSIONS: Although the clinical outcomes of predominantly stable disease and partial agonism were similar to what was observed in the preclinical evaluation of the compound, the limited antitumor activity of BMS-641988 at therapeutic dose levels coupled with an episode of seizure activity led to study closure.


Subject(s)
Androgen Antagonists/administration & dosage , Bone Neoplasms/secondary , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Imides/administration & dosage , Prostatic Neoplasms/drug therapy , Aged , Aged, 80 and over , Androgen Antagonists/adverse effects , Androgen Antagonists/pharmacokinetics , Bone Neoplasms/pathology , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Humans , Imides/adverse effects , Imides/pharmacokinetics , Male , Middle Aged , Orchiectomy , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/pathology , Treatment Outcome
9.
Dig Dis Sci ; 55(5): 1221-9, 2010 May.
Article in English | MEDLINE | ID: mdl-19517233

ABSTRACT

BACKGROUND: Rectal cancers that are confined to the mucosa (T0) can be resected endoscopically. This can help the patient avoid transabdominal surgery. The published data on accuracy of endoscopic ultrasound (EUS) to predict T0 stage of rectal cancers has been varied. AIM: To evaluate the accuracy of EUS in T0 staging of rectal cancers. METHOD (STUDY SELECTION CRITERIA): Only EUS studies confirmed by surgery were selected. T0 was defined as tumor confined to the mucosa. DATA COLLECTION AND EXTRACTION: Articles were searched in Medline, PubMed, and CENTRAL. STATISTICAL METHOD: Pooling was conducted by both the fixed-effects model and random-effects model. RESULTS: An initial search identified 3,360 reference articles. Of these, 339 relevant articles were selected and reviewed. Eleven studies (N = 1,791) which met the inclusion criteria were included in this analysis. Pooled sensitivity of EUS in diagnosing T0 was 97.3% (95% CI: 93.7-99.1). EUS had a pooled specificity of 96.3% (95% CI: 95.3-97.2). The positive likelihood ratio of EUS was 21.9 (95% CI: 16.3-29.7) and negative likelihood ratio was 0.08 (95% CI: 0.04-0.15). All the pooled estimates, calculated by fixed and random effect models, were similar. The P-value for Chi-squared heterogeneity for all the pooled accuracy estimates was >0.10. CONCLUSIONS: EUS has excellent sensitivity and specificity, this helps accurately diagnose T0 stage of rectal cancers. Over the past two decades, the sensitivity and specificity of EUS to diagnose T0 stage of rectal cancers has remained high. This can help physicians offer endoscopic treatment to these patients, therefore EUS should be strongly considered for staging of early rectal cancers.


Subject(s)
Endosonography , Rectal Neoplasms/diagnostic imaging , Humans , Models, Statistical , Neoplasm Staging , Proctoscopy , Rectal Neoplasms/pathology , Rectal Neoplasms/surgery , Sensitivity and Specificity
10.
Ann Surg Oncol ; 16(5): 1255-65, 2009 May.
Article in English | MEDLINE | ID: mdl-19219506

ABSTRACT

BACKGROUND: Nodal staging in patients with rectal cancer predicts prognosis and directs therapy. Published data on the accuracy of endoscopic ultrasound (EUS) for diagnosing nodal invasion in patients with rectal cancer has been inconsistent. AIM: To evaluate the accuracy of EUS in diagnosing nodal metastasis of rectal cancers. METHOD: Study Selection Criteria: Only EUS studies confirmed by surgical histology were selected. Data Collection and Extraction: Articles were searched in Medline, Pubmed, and CENTRAL. STATISTICAL METHOD: Pooling was conducted by both fixed-effects model and random-effects model. RESULTS: The initial search identified 3610 reference articles in which 352 relevant articles were selected and reviewed. Data were extracted from 35 studies (N = 2732) that met the inclusion criteria. Pooled sensitivity of EUS in diagnosing nodal involvement by rectal cancers was 73.2% (95% confidence interval [95% CI] 70.6-75.6). EUS had a pooled specificity of 75.8% (95% CI 73.5-78.0). The positive likelihood ratio of EUS was 2.84 (95% CI 2.16-3.72), and negative likelihood ratio was 0.42 (95% CI 0.33-0.52). All the pooled estimates, calculated by fixed- and random-effect models, were similar. SROC curves showed an area under the curve of 0.79. The P for chi-squared heterogeneity for all the pooled accuracy estimates was >.10. CONCLUSIONS: EUS is an important and accurate diagnostic tool for evaluating nodal metastasis of rectal cancers. This meta-analysis shows that the sensitivity and specificity of EUS is moderate. Further refinement in EUS technologies and diagnostic criteria are needed to improve the diagnostic accuracy.


Subject(s)
Endosonography , Lymph Nodes/diagnostic imaging , Rectal Neoplasms/diagnostic imaging , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Neoplasm Staging , Rectal Neoplasms/pathology , Rectal Neoplasms/secondary , Sensitivity and Specificity
11.
Dig Dis Sci ; 54(11): 2330-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19137428

ABSTRACT

The objective of this study was to evaluate the efficacy of EUS-guided CPN for pain relief in patients with chronic pancreatitis and pancreatic cancer. An initial search identified 1,439 reference articles, of which 130 relevant articles were selected and reviewed. Data was extracted from 8 studies (N = 283) for EUS-guided CPN for pain due to pancreatic cancer and nine studies for chronic pancreatitis (N = 376) which met the inclusion criteria. With EUS-guided CPN, the pooled proportion of patients with pancreatic cancer that showed pain relief was 80.12% (95% CI = 74.47-85.22). In patients with pain due to chronic pancreatitis, EUS-guided CPN provided pain relief in 59.45% (95% CI = 54.51-64.30). In conclusion, EUS-guided CPN offers a safe alternative technique for pain relief in patients with chronic pancreatitis or pancreatic cancer. In patients with pain due to chronic pancreatitis, better techniques or injected materials are needed to improve the response.


Subject(s)
Abdominal Pain/surgery , Celiac Plexus/surgery , Endosonography , Nerve Block , Abdominal Pain/etiology , Celiac Plexus/diagnostic imaging , Humans , Pancreatic Neoplasms/complications , Pancreatitis, Chronic/complications
12.
Ann Surg Oncol ; 16(2): 254-65, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19018597

ABSTRACT

Published data on accuracy of endoscopic ultrasound (EUS) in differentiating T stages of rectal cancers is varied. Study selection criteria were to select only EUS studies confirmed with results of surgical pathology. Articles were searched in Medline and Pubmed. Pooling was conducted by both fixed and random effects models. Initial search identified 3,630 reference articles, of which 42 studies (N = 5,039) met the inclusion criteria and were included in this analysis. The pooled sensitivity and specificity of EUS to determine T1 stage was 87.8% [95% confidence interval (CI) 85.3-90.0%] and 98.3% (95% CI 97.8-98.7%), respectively. For T2 stage, EUS had a pooled sensitivity and specificity of 80.5% (95% CI 77.9-82.9%) and 95.6% (95% CI 94.9-96.3%), respectively. To stage T3 stage, EUS had a pooled sensitivity and specificity of 96.4% (95% CI 95.4-97.2%) and 90.6% (95% CI 89.5-91.7%), respectively. In determining the T4 stage, EUS had a pooled sensitivity of 95.4% (95% CI 92.4-97.5%) and specificity of 98.3% (95% CI 97.8-98.7%). The p value for chi-squared heterogeneity for all the pooled accuracy estimates was > 0.10. We conclude that, as a result of the demonstrated sensitivity and specificity, EUS should be the investigation of choice to T stage rectal cancers. The sensitivity of EUS is higher for advanced disease than for early disease. EUS should be strongly considered for T staging of rectal cancers.


Subject(s)
Endosonography , Rectal Neoplasms/diagnostic imaging , Gastroscopy , Humans , Lymphatic Metastasis , Neoplasm Staging , Prognosis , ROC Curve , Rectal Neoplasms/pathology , Rectal Neoplasms/surgery , Reproducibility of Results , Sensitivity and Specificity , Time Factors
13.
World J Gastroenterol ; 14(25): 4011-9, 2008 Jul 07.
Article in English | MEDLINE | ID: mdl-18609685

ABSTRACT

AIM: To evaluate the accuracy of endoscopic ultrasound (EUS) for staging of gastric cancers. METHODS: Only EUS studies confirmed by surgery were selected. Only studies from which a 2 x 2 table could be constructed for true positive, false negative, false positive and true negative values were included. Articles were searched in Medline, Pubmed, Ovid journals, Cumulative index for nursing and allied health literature, International pharmaceutical abstracts, old Medline, Medline nonindexed citations, and Cochrane control trial registry. Two reviewers independently searched and extracted data. The differences were resolved by mutual agreement. 2 x 2 tables were constructed with the data extracted from each study. Meta-analysis for the accuracy of EUS was analyzed by calculating pooled estimates of sensitivity, specificity, likelihood ratios, and diagnostic odds ratio. Pooling was conducted by both the Mantel-Haenszel method (fixed effects model) and DerSimonian Laird method (random effects model). The heterogeneity of studies was tested using Cochran's Q test based upon inverse variance weights. RESULTS: Initial search identified 1620 reference articles and of these, 376 relevant articles were selected and reviewed. Twenty-two studies (n=1896) which met the inclusion criteria were included in this analysis. Pooled sensitivity of T1 was 88.1% (95% CI: 84.5-91.1) and T2 was 82.3% (95% CI: 78.2-86.0). For T3, pooled sensitivity was 89.7% (95% CI: 87.1-92.0). T4 had a pooled sensitivity of 99.2% (95% CI: 97.1-99.9). For nodal staging, the pooled sensitivity for N1 was 58.2% (95% CI: 53.5-62.8) and N2 was 64.9% (95% CI: 60.8-68.8). Pooled sensitivity to diagnose distant metastasis was 73.2% (95% CI: 63.2-81.7). The P for chi-squared heterogeneity for all the pooled accuracy estimates was >0.10. CONCLUSION: EUS results are more accurate with advanced disease than early disease. If EUS diagnoses advanced disease, such as T4 disease, the patient is 500 times more likely to have true anatomic stage of T4 disease.


Subject(s)
Endosonography , Gastroscopy/methods , Stomach Neoplasms/diagnostic imaging , Humans , Neoplasm Metastasis , Neoplasm Staging , Predictive Value of Tests , Prognosis , Reproducibility of Results , Sensitivity and Specificity
14.
World J Gastroenterol ; 14(19): 3028-37, 2008 May 21.
Article in English | MEDLINE | ID: mdl-18494054

ABSTRACT

AIM: To evaluate the accuracy of endoscopic ultrasound (EUS), EUS-fine needle aspiration (FNA) in evaluating mediastinal lymphadenopathy. METHODS: Only EUS and EUS-FNA studies confirmed by surgery or with appropriate follow-up were selected. Articles were searched in Medline, Pubmed, and Cochrane control trial registry. Only studies from which a 2 multiply 2 table could be constructed for true positive, false negative, false positive and true negative values were included. Two reviewers independently searched and extracted data. The differences were resolved by mutual agreement. Meta-analysis for the accuracy of EUS was analyzed by calculating pooled estimates of sensitivity, specificity, likelihood ratios, and diagnostic odds ratios. Pooling was conducted by both Mantel-Haenszel method (fixed effects model) and DerSimonian Laird method (random effects model). The heterogeneity of studies was tested using Cochran's Q test based upon inverse variance weights. RESULTS: Data was extracted from 76 studies (n = 9310) which met the inclusion criteria. Of these, 44 studies used EUS alone and 32 studies used EUS-FNA. FNA improved the sensitivity of EUS from 84.7% (95% CI: 82.9-86.4) to 88.0% (95% CI: 85.8-90.0). With FNA, the specificity of EUS improved from 84.6% (95% CI: 83.2-85.9) to 96.4% (95% CI: 95.3-97.4). The P for chi-squared heterogeneity for all the pooled accuracy estimates was > 0.10. CONCLUSION: EUS is highly sensitive and specific for the evaluation of mediastinal lymphadenopathy and FNA substantially improves this. EUS with FNA should be the diagnostic test of choice for evaluating mediastinal lymphadenopathy.


Subject(s)
Endosonography , Lymphatic Diseases/diagnostic imaging , Mediastinal Diseases/diagnostic imaging , Bias , Biopsy, Fine-Needle , Humans , Lymphatic Diseases/pathology , Mediastinal Diseases/pathology , Predictive Value of Tests , Reproducibility of Results , Sensitivity and Specificity , Time Factors
15.
World J Gastroenterol ; 14(10): 1479-90, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18330935

ABSTRACT

AIM: To evaluate the accuracy of endoscopic ultrasound (EUS) in the staging of esophageal cancer. METHODS: Only EUS studies confirmed by surgery were selected. Articles were searched in Medline and Pubmed. Two reviewers independently searched and extracted data. Meta-analysis of the accuracy of EUS was analyzed by calculating pooled estimates of sensitivity, specificity, likelihood ratios, and diagnostic odds ratio. Pooling was conducted by both the Mantel-Haenszel method (fixed effects model) and DerSimonian Laird method (random effects model). The heterogeneity of studies was tested using Cochran's Q test based upon inverse variance weights. RESULTS: Forty-nine studies (n = 2558) which met the inclusion criteria were included in this analysis. Pooled sensitivity and specificity of EUS to diagnose T1 was 81.6% (95% CI: 77.8-84.9) and 99.4% (95% CI: 99.0-99.7), respectively. To diagnose T4, EUS had a pooled sensitivity of 92.4% (95% CI: 89.2-95.0) and specificity of 97.4% (95% CI: 96.6-98.0). With Fine Needle Aspiration (FNA), sensitivity of EUS to diagnose N stage improved from 84.7% (95% CI: 82.9-86.4) to 96.7% (95% CI: 92.4-98.9). The P value for the c2 test of heterogeneity for all pooled estimates was > 0.10. CONCLUSION: EUS has excellent sensitivity and specificity in accurately diagnosing the TN stage of esophageal cancer. EUS performs better with advanced (T4) than early (T1) disease. FNA substantially improves the sensitivity and specificity of EUS in evaluating N stage disease. EUS should be strongly considered for staging esophageal cancer.


Subject(s)
Endosonography , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/pathology , Neoplasm Staging/methods , Humans , Publication Bias , Sensitivity and Specificity
16.
Dig Dis Sci ; 53(9): 2405-14, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18097752

ABSTRACT

BACKGROUND: Published data on the accuracy of endoscopic ultrasound (EUS) for staging distant and celiac axis lymph node (CLN) metastasis in patients with esophageal cancer (ECA) has varied. AIM: To evaluate the accuracy of EUS in diagnosing distal and CLN metastasis in ECA patients. STUDY SELECTION: EUS studies confirmed by surgery were selected. STATISTICAL METHOD: Pooling was conducted by both fixed and random-effects models. RESULTS: Data were extracted from 25 studies (N = 2029) which met the inclusion criteria. In ECA patients, pooled sensitivity of EUS was 67.2% (95% CI: 62.6-71.6) in diagnosis of distal metastasis and 66.6% (95% CI: 61.9-71.1) in diagnosis of CLN metastasis. EUS had a pooled specificity of 97.9% (95% CI: 97.1-98.6) for distal metastasis and 98.1% (95% CI: 97.3-98.7) for CLN metastasis. CONCLUSIONS: Although EUS has excellent specificity in accurately diagnosing distal and CLN metastasis in patients with ECA, the sensitivity is low.


Subject(s)
Endosonography , Esophageal Neoplasms/pathology , Lymphatic Metastasis/diagnostic imaging , Abdomen , Humans , Peritoneum , Sensitivity and Specificity
17.
Gastrointest Endosc ; 65(6): 788-97, 2007 May.
Article in English | MEDLINE | ID: mdl-17350008

ABSTRACT

BACKGROUND: Vascular invasion (VI) in a patient with pancreatic or periampullary cancers precludes surgery and indicates a poor prognosis. Published data on the accuracy of EUS in diagnosing VI is varied. OBJECTIVE: The aim of this meta-analysis was to evaluate the accuracy of EUS in diagnosing VI in patients with pancreatic and periampullary cancers. DESIGN: Data from EUS studies were pooled according to the Mantel-Haenszel and DerSimonian Laird methods. PATIENTS: EUS studies in which VI was confirmed by surgery or angiography were selected. INTERVENTIONS: EUS. MAIN OUTCOME MEASURES: Pooled estimates of sensitivity, specificity, likelihood ratios, and diagnostic odds ratio of EUS. RESULTS: Data were extracted from 29 studies (N = 1308) that met the inclusion criteria. The pooled sensitivity of EUS in diagnosing VI was 73% (95% CI, 68.8-76.9) and the pooled specificity was 90.2% (95% CI, 87.9-92.2). The positive likelihood ratio for diagnosing VI by EUS was 9.1 (95% CI, 4.6-17.9) and the negative likelihood ratio was 0.3 (95% CI, 0.2-0.5). Diagnostic odds ratio, the odds of having VI in positive as compared with negative EUS studies, was 40.1 (95% CI, 16.1-99.9). The P value for chi(2) heterogeneity for all the pooled estimates was >.05. CONCLUSIONS: Although EUS is the best noninvasive test to diagnose VI in pancreatic and periampullary cancers, this meta-analysis showed that the specificity (90%) is high but the sensitivity (73%) is not as high as suggested. Further refinements in EUS technologies and interpretation may improve the sensitivity for detecting VI.


Subject(s)
Ampulla of Vater , Common Bile Duct Neoplasms/diagnostic imaging , Common Bile Duct Neoplasms/pathology , Endosonography , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/pathology , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/pathology , Biopsy, Fine-Needle , Endosonography/methods , Humans , Neoplasm Invasiveness , Prognosis , Reproducibility of Results , Sensitivity and Specificity , Vascular Neoplasms
18.
Cancer Lett ; 242(2): 222-30, 2006 Oct 28.
Article in English | MEDLINE | ID: mdl-16458425

ABSTRACT

We investigated aberrant methylation in 101 prostate cancers(PCa) and 32 histologically normal prostate tissues. We focused on genes largely in the apoptotic pathway. Methylation frequencies of the genes were Reprimo, 54%; TMS1, 47%; DcR1, 45%; RRAD, 37%; DcR2, 37%; CRBP1, 34%; HPP1, 32%; RIZ1, 31%; DRM/Gremlin, 21%; SOCS1, 20%; DR4, 5%; DR5, 1%. Methylation of Reprimo and TMS1 correlate with preoperative serum prostate-specific antigen. Methylation of TMS1, DcR1, DcR2, and CRBP1 correlate with Gleason score. Methylation of TMS1 and unmethylation of both DcR1 and DcR2 correlate with poorer disease free survival by univariate and multivariate analyses. Our data suggest that methylation of multiple genes may be involved in pathogenesis and correlate with prognosis of PCa.


Subject(s)
Apoptosis , DNA Methylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Base Sequence , DNA/chemistry , DNA/metabolism , DNA Primers/chemistry , Disease-Free Survival , Humans , Male , Middle Aged , Multivariate Analysis , Prognosis , Prostatic Neoplasms/diagnosis
19.
Int J Cancer ; 118(4): 924-31, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16108009

ABSTRACT

Aberrant methylation of 5'gene promoter regions associated with gene silencing is an epigenetic phenomenon responsible for silencing of tumor suppressor genes in many cancer types. The aims of our study were to study the role of methylation of a large panel of genes during multistage pathogenesis and to correlate our findings with patient age and other clinico-pathological features. We investigated the aberrant promoter methylation profile of 19 genes in 92 colorectal cancers (CRCs) and corresponding nonmalignant epithelia (NME) (n = 57), and selected 15 genes for studying 26 colorectal adenomas (CAs). On the Basis of our results, the genes could be divided into 3 groups. Group 1 consisted of 13 genes whose methylation was tumor-specific. For 8 of these genes, the methylation frequencies in CAs were similar to those of CRCs, but significantly different from the frequencies in NME. Group 2, consisting of 2 genes demonstrating little or no methylation, were present in any sample type. In Group 3, consisting of 4 genes, relatively frequent methylation was present in both CRCs and NME, and the differences between these specimen types were not significant. Methylation of Group 1 genes were tightly correlated with each other, and these genes demonstrated increased methylation frequencies in CRCs with increasing age. Methylation was not correlated with other clinico-pathological features. In general, methylation frequencies of CAs were intermediate between CRCs and NME. Our study constitutes the most comprehensive methylation profile of CRCs, demonstrates that methylation commences early during CRC pathogenesis and is an age-related phenomenon.


Subject(s)
Aging/physiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/physiopathology , DNA Methylation , Aged , Aging/genetics , Female , Humans , Male , Middle Aged , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...