Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
J Surg Res ; 299: 195-204, 2024 May 17.
Article En | MEDLINE | ID: mdl-38761678

INTRODUCTION: Identifying contributors to lung transplant survival is vital in mitigating mortality. To enhance individualized mortality estimation and determine variable interaction, we employed a survival tree algorithm utilizing recipient and donor data. METHODS: United Network Organ Sharing data (2000-2021) were queried for single and double lung transplants in adult patients. Graft survival time <7 d was excluded. Sixty preoperative and immediate postoperative factors were evaluated with stepwise logistic regression on mortality; final model variables were included in survival tree modeling. Data were split into training and testing sets and additionally validated with 10-fold cross validation. Survival tree pruning and model selection was based on Akaike information criteria and log-likelihood values. Estimated survival probabilities and log-rank pairwise comparisons between subgroups were calculated. RESULTS: A total of 27,296 lung transplant patients (8175 single; 19,121 double lung) were included. Stepwise logistic regression yielded 47 significant variables associated with mortality. Survival tree modeling returned six significant factors: recipient age, length of stay from transplant to discharge, recipient ventilator duration post-transplant, double lung transplant, recipient reintubation post-transplant, and donor cytomegalovirus status. Eight subgroups consisting of combinations of these factors were identified with distinct Kaplan-Meier survival curves. CONCLUSIONS: Survival trees provide the ability to understand the effects and interactions of covariates on survival after lung transplantation. Individualized survival probability with this technique found that preoperative and postoperative factors influence survival after lung transplantation. Thus, preoperative patient counseling should acknowledge a degree of uncertainty given the influence of postoperative factors.

2.
Clin Cancer Res ; 29(18): 3659-3667, 2023 09 15.
Article En | MEDLINE | ID: mdl-37363962

PURPOSE: To evaluate DS-6157a, an antibody-drug conjugate targeting G protein-coupled receptor 20 (GPR20), in gastrointestinal stromal tumors (GIST). PATIENTS AND METHODS: In this phase I multicenter, open-label, multiple-dose study, patients with previously treated advanced GIST received intravenous DS-6157a on Day 1 of 21-day cycles, with a starting dose of 1.6 mg/kg. The primary objective evaluated the safety and tolerability of DS-6157a, while determining dose-limiting toxicity (DLT) and the MTD. Secondary objectives included plasma pharmacokinetics parameters, plasma antidrug antibodies (ADA), and efficacy. RESULTS: A total of 34 patients enrolled. DS-6157a was well tolerated, with DLTs in 4 patients (11.8%) at doses of 6.4 mg/kg, 9.6 mg/kg, and 12.8 mg/kg; the MTD was determined to be 6.4 mg/kg. Treatment-emergent adverse events (TEAE) grade ≥3 occurred in 17 patients (50.0%), including decreased platelet count (23.5%), anemia (20.6%), decreased neutrophil count (14.7%), and decreased white blood cell count (11.8%). Four patients (11.8%) experienced serious adverse events related to DS-6157a. Six patients died with 5 due to disease progression and 1 due to DS-6157a-related TEAE. Tumor shrinkage was observed in 7 patients (20.6%), and 1 patient (2.9%) achieved a partial response. Plasma concentrations and exposure of intact DS-6157a, DXd, and total anti-GPR20 antibody all demonstrated a dose-dependent profile. No treatment-emergent ADAs were observed. CONCLUSIONS: Targeting GPR20 with DS-6157a was tolerated in patients with advanced GIST with tumor shrinkage demonstrated in KIT/PDGFRA wild-type GIST. However, the study did not proceed further due to lower efficacy outcomes than anticipated.


Antineoplastic Agents , Gastrointestinal Stromal Tumors , Immunoconjugates , Neoplasms , Humans , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/pathology , Antineoplastic Agents/adverse effects , Neoplasms/drug therapy , Immunoconjugates/therapeutic use , Antibodies/therapeutic use , Maximum Tolerated Dose
3.
J Am Coll Surg ; 236(4): 563-572, 2023 04 01.
Article En | MEDLINE | ID: mdl-36728472

BACKGROUND: Elucidating contributors affecting liver transplant survival is paramount. Current methods offer crude global group outcomes. To refine patient-specific mortality probability estimation and to determine covariate interaction using recipient and donor data, we generated a survival tree algorithm, Recipient Survival After Orthotopic Liver Transplantation (ReSOLT), using United Network Organ Sharing (UNOS) transplant data. STUDY DESIGN: The UNOS database was queried for liver transplants in patients ≥18 years old between 2000 and 2021. Preoperative factors were evaluated with stepwise logistic regression; 43 significant factors were used in survival tree modeling. Graft survival of <7 days was excluded. The data were split into training and testing sets and further validated with 10-fold cross-validation. Survival tree pruning and model selection was achieved based on Akaike information criterion and log-likelihood values. Log-rank pairwise comparisons between subgroups and estimated survival probabilities were calculated. RESULTS: A total of 122,134 liver transplant patients were included for modeling. Multivariable logistic regression (area under the curve = 0.742, F1 = 0.822) and survival tree modeling returned 8 significant recipient survival factors: recipient age, donor age, recipient primary payment, recipient hepatitis C status, recipient diabetes, recipient functional status at registration and at transplantation, and deceased donor pulmonary infection. Twenty subgroups consisting of combinations of these factors were identified with distinct Kaplan-Meier survival curves (p < 0.001 among all by log rank test) with 5- and 10-year survival probabilities. CONCLUSIONS: Survival trees are a flexible and effective approach to understand the effects and interactions of covariates on survival. Individualized survival probability following liver transplant is possible with ReSOLT, allowing for more coherent patient and family counseling and prediction of patient outcome using both recipient and donor factors.


Liver Transplantation , Humans , Adolescent , Retrospective Studies , Tissue Donors , Liver , Risk Factors , Graft Survival , Treatment Outcome
4.
Neuro Oncol ; 25(2): 339-350, 2023 02 14.
Article En | MEDLINE | ID: mdl-35849035

BACKGROUND: Approximately 50% of newly diagnosed glioblastomas (GBMs) harbor epidermal growth factor receptor gene amplification (EGFR-amp). Preclinical and early-phase clinical data suggested efficacy of depatuxizumab mafodotin (depatux-m), an antibody-drug conjugate comprised of a monoclonal antibody that binds activated EGFR (overexpressed wild-type and EGFRvIII-mutant) linked to a microtubule-inhibitor toxin in EGFR-amp GBMs. METHODS: In this phase III trial, adults with centrally confirmed, EGFR-amp newly diagnosed GBM were randomized 1:1 to radiotherapy, temozolomide, and depatux-m/placebo. Corneal epitheliopathy was treated with a combination of protocol-specified prophylactic and supportive measures. There was 85% power to detect a hazard ratio (HR) ≤0.75 for overall survival (OS) at a 2.5% 1-sided significance level (ie traditional two-sided p ≤ 0.05) by log-rank testing. RESULTS: There were 639 randomized patients (median age 60, range 22-84; 62% men). Prespecified interim analysis found no improvement in OS for depatux-m over placebo (median 18.9 vs. 18.7 months, HR 1.02, 95% CI 0.82-1.26, 1-sided p = 0.63). Progression-free survival was longer for depatux-m than placebo (median 8.0 vs. 6.3 months; HR 0.84, 95% confidence interval [CI] 0.70-1.01, p = 0.029), particularly among those with EGFRvIII-mutant (median 8.3 vs. 5.9 months, HR 0.72, 95% CI 0.56-0.93, 1-sided p = 0.002) or MGMT unmethylated (HR 0.77, 95% CI 0.61-0.97; 1-sided p = 0.012) tumors but without an OS improvement. Corneal epitheliopathy occurred in 94% of depatux-m-treated patients (61% grade 3-4), causing 12% to discontinue. CONCLUSIONS: Interim analysis demonstrated no OS benefit for depatux-m in treating EGFR-amp newly diagnosed GBM. No new important safety risks were identified.


Brain Neoplasms , Glioblastoma , Adult , Male , Humans , Middle Aged , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Antibodies, Monoclonal, Humanized , Temozolomide/therapeutic use , ErbB Receptors , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology
5.
Clin Lung Cancer ; 23(3): 214-225, 2022 05.
Article En | MEDLINE | ID: mdl-35331641

BACKGROUND: This open-label Phase III trial (NCT02264990) evaluated the PARP inhibitor, veliparib, combined with carboplatin/paclitaxel versus chemotherapy alone for first-line treatment of patients with advanced non-squamous non-small cell lung cancers (NSCLC). A 52-gene expression classifier (LP52) previously shown to identify patients more likely to respond to veliparib was evaluated as a planned correlative analysis. MATERIALS AND METHODS: Adult current or former smokers with advanced non-squamous NSCLC were randomized 1:1 to veliparib (120 mg daily for 7 days/cycle) with carboplatin and paclitaxel or to investigators' choice of platinum doublet chemotherapy (up to 6, 21-day cycles), with optional pemetrexed maintenance. Prospective analysis of the LP52 signature was conducted using a clinical Qiagen/HTG assay. The primary endpoint was overall survival (OS) in LP52+ patients. RESULTS: Overall, 595 patients received veliparib + carboplatin/paclitaxel (n = 298) or chemotherapy alone (n = 297); 13% (n = 40) in each arm were LP52+. The primary endpoint was not met; median OS was 11.2 months with veliparib + carboplatin/paclitaxel versus 9.2 months with chemotherapy alone in the LP52+ subgroup (hazard ratio [HR] 0.644, 95% confidence interval [CI]: 0.396-1.048; P = .113). In the overall population, median OS was 12.1 months in both arms (HR 0.986, 95% CI: 0.827-1.176; P = .846). No new safety signals were observed. CONCLUSION: In patients with non-squamous NSCLC, there was no significant improvement in OS with veliparib + carboplatin/paclitaxel versus chemotherapy alone, although a trend toward improved OS in the LP52+ population suggests this subgroup may benefit from veliparib. Statistical power was limited due to the small sample size.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles , Carboplatin , Humans , Paclitaxel
6.
Eur J Cancer ; 154: 35-45, 2021 09.
Article En | MEDLINE | ID: mdl-34243076

BACKGROUND: Addition of veliparib to carboplatin-paclitaxel, with continuation of veliparib monotherapy if carboplatin-paclitaxel was discontinued, improved progression-free survival (PFS) in patients with germline BRCA-associated locally advanced/metastatic HER2- breast cancer and ≤2 lines of previous cytotoxic therapy for metastatic disease in BROCADE3. A pre-planned subgroup analysis evaluated efficacy and safety in patients without previous cytotoxic therapy for metastatic disease. METHODS: Patients were randomised 2:1 to receive veliparib (120 mg orally BID) or placebo on days -2 to 5. Carboplatin (AUC 6) was administered on day 1, and paclitaxel (80 mg/m2) on days 1, 8 and 15 (21-day cycles). Patients discontinuing carboplatin-paclitaxel for reasons besides progression could continue veliparib/placebo monotherapy (300 mg BID, increasing to 400 mg BID if tolerated) until progression. The primary end-point was PFS assessed by investigator. RESULTS: Of 509 patients in the intention-to-treat population (98.6% female; mean age 47, standard deviation 11), 413 (81%) had no previous cytotoxic therapy for metastatic disease (274, veliparib; 139, placebo). In the first-line subgroup, median PFS was 16.6 months (95% confidence interval [CI] 13.4-18.7) versus 13.1 months (95% CI 11.4-14.5) for the veliparib versus control groups (hazard ratio 0.70, 95% CI 0.54-0.89, P = .004). More patients were alive and progression-free at 2 years (36% versus 23.2%) and 3 years (27.9% versus 13.3%) in the veliparib versus control group. Adverse events unrelated to progression leading to study drug discontinuation occurred in 25 (9.1%) and 8 (5.8%) patients. CONCLUSIONS: Veliparib with carboplatin-paclitaxel led to durable disease control among first-line patients, suggesting a benefit of this treatment approach in early lines. CLINICAL TRIAL REGISTRATION: NCT02163694.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Genes, BRCA1 , Genes, BRCA2 , Germ-Line Mutation , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Carboplatin/administration & dosage , Carboplatin/adverse effects , Female , Humans , Male , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Receptor, ErbB-2/analysis
7.
Clin Cancer Res ; 27(18): 4983-4993, 2021 09 15.
Article En | MEDLINE | ID: mdl-34131001

PURPOSE: Safety, efficacy, and exploratory biomarker analyses were evaluated in patients with advanced HER2-negative germline breast cancer susceptibility gene (gBRCA)-associated breast cancer enrolled in the BROCADE3 trial who received crossover veliparib monotherapy after disease progression on placebo plus carboplatin/paclitaxel. PATIENTS AND METHODS: Eligible patients (N = 513) were randomized 2:1 to veliparib plus carboplatin/paclitaxel or placebo plus carboplatin/paclitaxel; patients had variable platinum-free intervals (PFI) at progression. In the placebo arm, patients were eligible to receive crossover veliparib monotherapy (300-400 mg twice daily continuous). Antitumor activity and adverse events were assessed during crossover veliparib treatment. BRCA reversion mutations at crossover were analyzed retrospectively using next-generation sequencing on plasma circulating tumor DNA (ctDNA). RESULTS: Seventy-five patients in the placebo plus carboplatin/paclitaxel arm received ≥1 dose of crossover veliparib postprogression (mean treatment duration: 154 days). Eight of 50 (16%) patients with measurable disease had a RECIST v1.1 response. Activity was greater in patients with PFI ≥180 days compared with <180 days [responses in 23.1% (3/13) vs. 13.5% (5/37) of patients]. BRCA reversion mutations that restored protein function were detected in ctDNA from 4 of 28 patients tested, and the mean duration of crossover veliparib monotherapy was <1 month in these 4 patients versus 7.49 months in patients lacking reversion mutations. The most frequent adverse events were nausea (61%), vomiting (29%), and fatigue (24%). CONCLUSIONS: Crossover veliparib monotherapy demonstrated limited antitumor activity in patients who experienced disease progression on placebo plus carboplatin/paclitaxel. PFI appeared to affect veliparib activity. BRCA reversion mutations may promote cross-resistance and limit veliparib activity following progression on platinum.


Breast Neoplasms , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzimidazoles , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carboplatin/adverse effects , Female , Humans , Mutation , Paclitaxel/adverse effects , Platinum/therapeutic use , Retrospective Studies
8.
Stat Med ; 40(10): 2321-2338, 2021 05 10.
Article En | MEDLINE | ID: mdl-33624861

When comparing survival times of treatment and control groups under a more realistic nonproportional hazards scenario, the standard log-rank (SLR) test may be replaced by a more efficient weighted log-rank (WLR) test, such as the Fleming-Harrington (FH) test. Designing a group-sequential clinical trial with one or more interim looks during which a FH test will be performed, necessitates correctly quantifying the information fraction (IF). For SLR test, IF is defined simply as the ratio of interim to final numbers of events; but for FH test, it can deviate substantially from this ratio. In this article, we separate the effect of weight function (of FH test) alone on IF from the effect of censoring. We have shown that, without considering the effect of censoring, IF can be derived analytically for FH test using information available at the design stage and the additional effect due to censoring is relatively smaller. This article intends to serve two major purposes: first, to emphasize and rationalize the deviation of IF in weighted log-rank test from that of SLR test which is often overlooked (Jiménez, Stalbovskaya, and Jones); second, although it is impossible to predict IF for a weighted log-rank test at the design stage, our decomposition of effects on IF provides a reasonable and practically feasible range of IF to work with. We illustrate our approach with an example and provide simulation results to evaluate operating characteristics.


Clinical Trials as Topic , Computer Simulation , Survival Analysis
9.
Eur J Cancer ; 147: 1-12, 2021 04.
Article En | MEDLINE | ID: mdl-33601293

BACKGROUND: In the EORTC 1410/INTELLANCE 2 randomised, phase II study (NCT02343406), with the antibody-drug conjugate depatuxizumab mafodotin (Depatux-M, ABT-414) in patients with recurrent EGFR-amplified glioblastoma, the primary end-point (overall survival) was not met, and the drug had ocular dose-limiting toxicity. This study reports results from the prespecified health-related quality of life (HRQoL) and neurological deterioration-free survival (NDFS) exploratory analysis. PATIENTS AND METHODS: Patients (n = 260) were randomised 1:1:1 to receive either Depatux-M 1.25 mg/kg or 1.0 mg/kg intravenously every 2 weeks with oral temozolomide (TMZ) 150 mg/m2, Depatux-M alone, or TMZ or oral lomustine (CCNU) 110 mg/m2 (TMZ/CCNU). HRQoL outcomes were recorded using the EORTC core Quality of Life QLQ-C30, and brain cancer-specific QLQ-BN20 questionnaires. Questionnaires were completed at baseline, weeks 8 and 16, and month 6, and changes from baseline to each time point were calculated. NDFS was defined as time to first deterioration in World Health Organisation performance status. RESULTS: Compliance with HRQoL was 88.1% at baseline and decreased to 37.9% at month 6. Differences from baseline between Depatux-M arms and TMZ/CCNU in global health/QoL status throughout treatment did not reach clinical relevance (≥10 points). Self-reported visual disorders deteriorated to a clinically relevant extent with Depatux-M arms versus TMZ/CCNU at all timepoints (mean differences range: 24.6-35.1 points). Changes from baseline for other HRQoL scales and NDFS were generally similar between treatment arms. CONCLUSIONS: Depatux-M had no impact on HRQoL and NDFS in patients with EGFR-amplified recurrent glioblastoma, except for more visual disorders, an expected side-effect of the study drug. CLINICAL TRIAL REGISTRATION: NCT02343406.


Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Gene Amplification , Glioblastoma/drug therapy , Neoplasm Recurrence, Local , Quality of Life , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/mortality , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Europe , Female , Functional Status , Glioblastoma/diagnosis , Glioblastoma/genetics , Glioblastoma/mortality , Humans , Male , Middle Aged , Neurologic Examination , Progression-Free Survival , Surveys and Questionnaires , Time Factors , Vision Disorders/chemically induced , Vision Disorders/physiopathology , Vision, Ocular/drug effects
10.
Lancet Oncol ; 21(10): 1269-1282, 2020 10.
Article En | MEDLINE | ID: mdl-32861273

BACKGROUND: BRCA1 or BRCA2-mutated breast cancers are sensitive to poly(ADP-ribose) polymerase (PARP) inhibitors and platinum agents owing to deficiency in homologous recombination repair of DNA damage. In this trial, we compared veliparib versus placebo in combination with carboplatin and paclitaxel, and continued as monotherapy if carboplatin and paclitaxel were discontinued before progression, in patients with HER2-negative advanced breast cancer and a germline BRCA1 or BRCA2 mutation. METHODS: BROCADE3 was a randomised, double-blind, placebo-controlled, phase 3 trial done at 147 hospitals in 36 countries. Eligible patients (aged ≥18 years) had deleterious germline BRCA1 or BRCA2 mutation-associated, histologically or cytologically confirmed advanced HER2-negative breast cancer, an Eastern Cooperative Oncology Group performance status of 0-2, and had received up to two previous lines of chemotherapy for metastatic disease. Patients were randomly assigned (2:1) by interactive response technology by means of permuted blocks within strata (block size of 3 or 6) to carboplatin (area under the concentration curve 6 mg/mL per min intravenously) on day 1 and paclitaxel (80 mg/m2 intravenously) on days 1, 8, and 15 of 21-day cycles combined with either veliparib (120 mg orally twice daily, on days -2 to 5) or matching placebo. If patients discontinued carboplatin and paclitaxel before progression, they could continue veliparib or placebo at an intensified dose (300 mg twice daily continuously, escalating to 400 mg twice daily if tolerated) until disease progression. Patients in the control group could receive open-label veliparib monotherapy after disease progression. Randomisation was stratified by previous platinum use, history of CNS metastases, and oestrogen and progesterone receptor status. The primary endpoint was investigator-assessed progression-free survival per Response Evaluation Criteria in Solid Tumors version 1.1. Efficacy analyses were done by intention to treat, which included all randomly assigned patients with a centrally confirmed BRCA mutation, and safety analyses included all patients who received at least one dose of velilparib or placebo. This study is ongoing and is registered with ClinicalTrials.gov, NCT02163694. FINDINGS: Between July 30, 2014, and Jan 17, 2018, 2202 patients were screened, of whom 513 eligible patients were enrolled and randomly assigned. In the intention-to-treat population (n=509), 337 patients were assigned to receive veliparib plus carboplatin-paclitaxel (veliparib group) and 172 were assigned to receive placebo plus carboplatin-paclitaxel (control group). Median follow-up at data cutoff (April 5, 2019) was 35·7 months (IQR 24·9-43·6) in the veliparib group and 35·5 months (23·1-45·9) in the control group. Median progression-free survival was 14·5 months (95% CI 12·5-17·7) in the veliparib group versus 12·6 months (10·6-14·4) in the control group (hazard ratio 0·71 [95% CI 0·57-0·88], p=0·0016). The most common grade 3 or worse adverse events were neutropenia (272 [81%] of 336 patients in the veliparib group vs 143 [84%] of 171 patients in the control group), anaemia (142 [42%] vs 68 [40%]), and thrombocytopenia (134 [40%] vs 48 [28%]). Serious adverse events occurred in 115 (34%) patients in the veliparib group versus 49 (29%) patients in the control group. There were no study drug-related deaths. INTERPRETATION: The addition of veliparib to a highly active platinum doublet, with continuation as monotherapy if the doublet were discontinued, resulted in significant and durable improvement in progression-free survival in patients with germline BRCA mutation-associated advanced breast cancer. These data indicate the utility of combining platinum and PARP inhibitors in this patient population. FUNDING: AbbVie.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles/therapeutic use , Breast Neoplasms/drug therapy , Carboplatin/therapeutic use , Paclitaxel/therapeutic use , Adult , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Double-Blind Method , Drug Administration Schedule , Drug-Related Side Effects and Adverse Reactions , Female , Genes, BRCA1 , Genes, BRCA2 , Germ-Line Mutation , Humans , Male , Middle Aged , Organoplatinum Compounds/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Progression-Free Survival , Treatment Outcome
13.
Onco Targets Ther ; 10: 3177-3186, 2017.
Article En | MEDLINE | ID: mdl-28721067

This phase I study aimed at determining the maximum tolerated dose (MTD) and characterizing the safety, tolerability, pharmacokinetics (PKs), and efficacy of pasireotide in patients with advanced neuroendocrine tumors (NETs). Patients were enrolled in two phases: dose-escalation phase (to determine the MTD) at a starting dose of 80 mg pasireotide long-acting release (LAR) i.m. followed by a dose-expansion phase (to evaluate safety and prelimi-nary efficacy). Associations between PK/pharmacodynamic parameters and clinical outcomes were evaluated using linear regression analysis. A total of 29 patients were treated with 80 mg (n=13) and 120 mg (n=16) doses. Most common primary tumor sites included small intestine (44.8%), pancreas (24.1%), and lung (17.2%). No protocol-defined dose-limiting toxicities were observed in the study; however, in post hoc analysis, a higher incidence of bradycardia (heart rate [HR] <40 beats per minute [bpm]) was observed with 120 mg (31.3%) vs 80 mg (0%). Two partial responses (PRs) were observed, both in the 120 mg dose cohort. Pasireotide concentrations correlated with tumor shrinkage, although the association was not statistically significant (P=0.08). Among the biomarkers analyzed, insulin-like growth factor 1 (IGF-1) showed a decreasing trend with increasing pasireotide concentration, while chromogranin A (CgA) and neuron-specific enolase (NSE) levels did not show any dose-response relationship. The most common adverse events in any dose group were hyperglycemia, fatigue, and nausea. MTD was defined at 120 mg for pasireotide LAR in patients with advanced NETs. Although objective radiographic responses were rarely observed with somatostatin analogs, two PRs were observed among 16 patients in the 120 mg cohort. Bradycardia (HR <40 bpm) appears to be a dose-limiting effect; however, the mechanism and clinical significance are uncertain. This study was registered with clinicaltrials.gov (NCT01364415).

14.
J Chemom ; 31(4)2017 Apr.
Article En | MEDLINE | ID: mdl-30369716

Tikhonov regularization was proposed for multivariate calibration by Andries and Kalivas [1]. We use this framework for modeling the statistical association between spectroscopy data and a scalar outcome. In both the calibration and regression settings this regularization process has advantages over methods of spectral pre-processing and dimension-reduction approaches such as feature extraction or principal component regression. We propose an extension of this penalized regression framework by adaptively refining the penalty term to optimally focus the regularization process. We illustrate the approach using simulated spectra and compare it with other penalized regression models and with a two-step method that first pre-processes the spectra then fits a dimension-reduced model using the processed data. The methods are also applied to magnetic resonance spectroscopy data to identify brain metabolites that are associated with cognitive function.

15.
Contemp Clin Trials ; 53: 20-28, 2017 02.
Article En | MEDLINE | ID: mdl-27940185

PURPOSE: PFS is often used as a surrogate endpoint for OS in metastatic breast cancer studies. We have evaluated the association of treatment effect on PFS with significant HROS (and how this association is affected by other factors) in published prospective metastatic breast cancer studies. METHODS: A systematic literature search in PubMed identified prospective metastatic breast cancer studies. Treatment effects on PFS were determined using hazard ratio (HRPFS), increase in median PFS (ΔMEDPFS) and % increase in median PFS (%ΔMEDPFS). Diagnostic accuracy of PFS measures (HRPFS, ΔMEDPFS and %ΔMEDPFS) in predicting significant HROS was assessed using receiver operating characteristic (ROC) curves and classification tree approach (CART). RESULTS: Seventy-four cases (i.e., treatment to control comparisons) from 65 individual publications were identified for the analyses. Of these, 16 cases reported significant treatment effect on HROS at 5% level of significance. Median number of deaths reported in these cases were 153. Area under the ROC curve (AUC) for diagnostic measures as HRPFS, ΔMEDPFS and %ΔMEDPFS were 0.69, 0.70 and 0.75, respectively. Classification tree results identified %ΔMEDPFS and number of deaths as diagnostic measure for significant HROS. Only 7.9% (3/39) cases with ΔMEDPFS shorter than 48.27% reported significant HROS. There were 7 cases with ΔMEDPFS of 48.27% or more and number of deaths reported as 227 or more - of these 5 cases reported significant HROS. CONCLUSION: %ΔMEDPFS was found to be a better diagnostic measure for predicting significant HROS. Our analysis results also suggest that consideration of total number of deaths may further improve its diagnostic performance. Based on our study results, the studies with 50% improvement in median PFS are more likely to produce significant HROS if the total number of OS events at the time of analysis is 227 or more.


Breast Neoplasms/mortality , Disease-Free Survival , Survival Rate , Area Under Curve , Breast Neoplasms/pathology , Female , Humans , Neoplasm Metastasis , Proportional Hazards Models , ROC Curve
16.
Stat Modelling ; 16(2): 114-139, 2016 Apr.
Article En | MEDLINE | ID: mdl-28316508

This article addresses estimation in regression models for longitudinally-collected functional covariates (time-varying predictor curves) with a longitudinal scaler outcome. The framework consists of estimating a time-varying coefficient function that is modeled as a linear combination of time-invariant functions with time-varying coefficients. The model uses extrinsic information to inform the structure of the penalty, while the estimation procedure exploits the equivalence between penalized least squares estimation and a linear mixed model representation. The process is empirically evaluated with several simulations and it is applied to analyze the neurocognitive impairment of HIV patients and its association with longitudinally-collected magnetic resonance spectroscopy (MRS) curves.

17.
Childs Nerv Syst ; 31(4): 563-8, 2015 Apr.
Article En | MEDLINE | ID: mdl-25712743

OBJECT: The distal catheter of a ventriculoatrial (VA) cerebrospinal fluid shunt is potentially exposed to bacterial seeding from a subclavian central line. The risk of blood stream infections (BSIs) from central lines increases with administration of total parenteral nutrition (TPN). The potential risks of shunt malfunction or infection in patients with a VA shunt and a concurrent subclavian central line and/or TPN administration have not been studied. METHODS: A retrospective review of 49 pediatric patients with placement of a VA shunt was performed. Three outcome measures were studied: shunt malfunction, shunt infection, and bacteremia/fungemia requiring shunt removal. All outcomes were measured by 1 year after shunt insertion. We analyzed the following potential risk factors: age at shunt insertion, prior ventriculoperitoneal (VP) shunt, prior shunt infection, abdominal infection/necrotizing enterocolitis (NEC), concurrent subclavian central line, and administration of TPN. The association between each risk factor and outcome was evaluated using Fisher's exact test to generate the relative risk. Additionally, a logistic regression analysis was performed to evaluate the odds ratio of the outcomes to risk factors considering age as a covariate. RESULTS: The average age at shunt insertion was 6.3 ± 7.6 years. The most common diagnosis was posthemorrhagic hydrocephalus of prematurity (53.1 %). Fifteen patients (30.1 %) had a shunt malfunction within 1 year, 6 (12.2 %) had a shunt infection, and 3 (6.1 %) required removal of the shunt due to bacteremia/fungemia. The age at shunt insertion was not a statistically significant independent risk factor for any of the three outcomes. Prior shunt infection predicted an increased risk for both future shunt malfunction and infection in both the associative relative risk analysis and the age-dependent logistic regression analysis, although the correlation did not reach statistical significance. The presence of a subclavian central line or TPN administration did not statistically increase the risk over baseline for any of the outcomes in either analysis. CONCLUSIONS: The relatively small number of patients limits the power of the study. Considering this limitation, the data suggests that the presence of a concurrent subclavian central line or administration of TPN does not increase the risk of shunt malfunction or infection over the baseline of this high-risk cohort.


Bacteremia/etiology , Hydrocephalus/etiology , Parenteral Nutrition, Total , Treatment Failure , Ventriculoperitoneal Shunt/adverse effects , Adolescent , Age Factors , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Infant, Premature, Diseases , Logistic Models , Male , Outcome Assessment, Health Care , Retrospective Studies , Risk Assessment
18.
Infect Control Hosp Epidemiol ; 36(1): 34-9, 2015 Jan.
Article En | MEDLINE | ID: mdl-25627759

OBJECTIVE We sought to determine whether the bacterial burden in the nares, as determined by the cycle threshold (CT) value from real-time MRSA PCR, is predictive of environmental contamination with MRSA. METHODS Patients identified as MRSA nasal carriers per hospital protocol were enrolled within 72 hours of room admission. Patients were excluded if (1) nasal mupirocin or chlorhexidine body wash was used within the past month or (2) an active MRSA infection was suspected. Four environmental sites, 6 body sites and a wound, if present, were cultured with premoistened swabs. All nasal swabs were submitted for both a quantitative culture and real-time PCR (Roche Lightcycler, Indianapolis, IN). RESULTS At study enrollment, 82 patients had a positive MRSA-PCR. A negative correlation of moderate strength was observed between the CT value and the number of MRSA colonies in the nares (r=-0.61; P<0.01). Current antibiotic use was associated with lower levels of MRSA nasal colonization (CT value, 30.2 vs 27.7; P<0.01). Patients with concomitant environmental contamination had a higher median log MRSA nares count (3.9 vs 2.5, P=0.01) and lower CT values (28.0 vs 30.2; P<0.01). However, a ROC curve was unable to identify a threshold MRSA nares count that reliably excluded environmental contamination. CONCLUSIONS Patients with a higher burden of MRSA in their nares, based on the CT value, were more likely to contaminate their environment with MRSA. However, contamination of the environment cannot be predicted solely by the degree of MRSA nasal colonization.


Carrier State/diagnosis , Fomites/microbiology , Methicillin-Resistant Staphylococcus aureus , Nose/microbiology , Real-Time Polymerase Chain Reaction , Skin/microbiology , Abdominal Wall/microbiology , Aged , Axilla/microbiology , Carrier State/microbiology , Colony Count, Microbial , Female , Forearm/microbiology , Groin/microbiology , Humans , Male , Methicillin-Resistant Staphylococcus aureus/genetics , Middle Aged , Patients' Rooms , Predictive Value of Tests , ROC Curve , Thoracic Wall/microbiology
...