Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Clin Transl Sci ; 16(6): 1039-1048, 2023 06.
Article in English | MEDLINE | ID: mdl-37085998

ABSTRACT

Letermovir is approved for use in cytomegalovirus-seropositive hematopoietic stem cell transplant recipients and is investigated in other transplant settings. Nonlinear pharmacokinetics (PKs) were observed in clinical studies after intravenous and oral dosing across a wide dose range, including the efficacious doses of 240 and 480 mg. A physiologically-based PK (PBPK) model for letermovir was built to develop a plausible explanation for the nonlinear PKs observed in clinical studies. In vitro studies suggested that letermovir elimination and distribution are mediated by saturable uridine glucuronosyltransferases (UGT)-metabolism and by saturable hepatic uptake via organic anion-transporting polypeptides (OATP) 1B. A sensitivity analysis of parameters describing the metabolism and distribution mechanisms indicated that the greater than dose-proportional increase in letermovir exposure is best described by a saturable OATP1B-mediated transport. This PBPK model was further used to evaluate the drug interaction potential between letermovir and everolimus, an immunosuppressant that may be co-administered with letermovir depending on regions. Because letermovir inhibits cytochrome P450 (CYP) 3A and everolimus is a known CYP3A substrate, an interaction when concomitantly administered is anticipated. The drug-drug interaction simulation confirmed that letermovir will likely increase everolimus are under the curve by 2.5-fold, consistent with the moderate increase in exposure observed with midazolam in the clinic. The output highlights the importance of drug monitoring, which is common clinical practice for everolimus to maintain safe and efficacious drug concentrations in the targeted patient population when concomitantly administered with letermovir.


Subject(s)
Everolimus , Immunosuppressive Agents , Humans , Everolimus/adverse effects , Drug Interactions , Immunosuppressive Agents/pharmacokinetics , Acetates , Cytochrome P-450 CYP3A/metabolism , Models, Biological
3.
J Pharmacokinet Pharmacodyn ; 49(1): 5-18, 2022 02.
Article in English | MEDLINE | ID: mdl-35103884

ABSTRACT

Quantitative systems pharmacology (QSP) modeling is applied to address essential questions in drug development, such as the mechanism of action of a therapeutic agent and the progression of disease. Meanwhile, machine learning (ML) approaches also contribute to answering these questions via the analysis of multi-layer 'omics' data such as gene expression, proteomics, metabolomics, and high-throughput imaging. Furthermore, ML approaches can also be applied to aspects of QSP modeling. Both approaches are powerful tools and there is considerable interest in integrating QSP modeling and ML. So far, a few successful implementations have been carried out from which we have learned about how each approach can overcome unique limitations of the other. The QSP + ML working group of the International Society of Pharmacometrics QSP Special Interest Group was convened in September, 2019 to identify and begin realizing new opportunities in QSP and ML integration. The working group, which comprises 21 members representing 18 academic and industry organizations, has identified four categories of current research activity which will be described herein together with case studies of applications to drug development decision making. The working group also concluded that the integration of QSP and ML is still in its early stages of moving from evaluating available technical tools to building case studies. This paper reports on this fast-moving field and serves as a foundation for future codification of best practices.


Subject(s)
Drug Development , Network Pharmacology , Drug Development/methods , Machine Learning
4.
Clin Pharmacol Drug Dev ; 11(4): 420-428, 2022 04.
Article in English | MEDLINE | ID: mdl-35157785

ABSTRACT

Letermovir (MK-8228/AIC246) is a cytomegalovirus (CMV) DNA terminase complex inhibitor for CMV prophylaxis in adult patients undergoing hematopoietic stem cell transplant. It is cytochrome P450 (CYP) 3A inhibitor and inhibits organic anion transporting polypeptide 1B1/3 and breast cancer resistance protein transporters. Atorvastatin (ATV), a commonly used treatment for hypercholesterolemia, is a substrate of organic anion transporting polypeptide 1B1, potentially breast cancer resistance protein, and CYP3A. As letermovir may be coadministered with ATV, the effect of multiple-dose letermovir 480 mg once daily on the pharmacokinetics of single-dose ATV 20 mg and its metabolites (ortho-hydroxyatorvastatin [o-OH-ATV] and para-hydroxyatorvastatin [p-OH-ATV]) was evaluated in an open-label trial in healthy female adults (N = 14). ATV area under the plasma concentration-time curve from time 0 to infinity and maximum plasma concentration (Cmax ) increased ≈3-fold with letermovir coadministration. The time to ATV Cmax also increased, while apparent clearance decreased. The exposures of o-OH-ATV and p-OH-ATV were comparable in the presence versus absence of letermovir; however, o-OH-ATV Cmax decreased by 60% with coadministration, while p-OH-ATV Cmax was similar. Due to the increase in ATV exposure with letermovir coadministration, statin-associated adverse events such as myopathy should be closely monitored following coadministration. The dose of ATV should not exceed 20 mg daily when coadministered with letermovir.


Subject(s)
Neoplasm Proteins , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Acetates , Adult , Atorvastatin , Drug Interactions , Female , Healthy Volunteers , Humans , Quinazolines
5.
Clin Pharmacol Ther ; 111(2): 485-495, 2022 02.
Article in English | MEDLINE | ID: mdl-34674258

ABSTRACT

The cytomegalovirus (CMV) viral terminase inhibitor letermovir is approved for prophylaxis of CMV infection and disease in adult CMV-seropositive allogeneic hematopoietic stem cell transplantation recipients. In a phase III trial (NCT02137772), letermovir significantly reduced clinically significant CMV infection (CS-CMVi) rate vs. placebo through Week 24 (primary end point) and Week 14 (secondary end point) post transplantation. Here, exposure-response relationships were investigated using efficacy and selected safety end points from the phase III trial to inform the proposed clinical dose. Post hoc exposure estimates were derived from a population pharmacokinetic model. No significant exposure dependencies were found for CS-CMVi through Week 24 or Week 14 among letermovir-treated participants. Evaluated covariates had no impact on exposure-efficacy relationships and letermovir plasma exposure did not affect time of CS-CMVi onset. There was no dependence between adverse event incidence and letermovir exposure. These results support current dosing recommendations in several countries and regions, including the United States and European Union.


Subject(s)
Acetates/administration & dosage , Antiviral Agents/administration & dosage , Cytomegalovirus Infections/prevention & control , Hematopoietic Stem Cell Transplantation , Quinazolines/administration & dosage , Acetates/pharmacokinetics , Administration, Intravenous , Administration, Oral , Adolescent , Adult , Aged , Antiviral Agents/pharmacokinetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Dose-Response Relationship, Drug , Double-Blind Method , Drug Dosage Calculations , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Male , Middle Aged , Models, Biological , Quinazolines/pharmacokinetics , Transplantation, Homologous , Treatment Outcome , Young Adult
6.
J Pharmacokinet Pharmacodyn ; 49(1): 81-99, 2022 02.
Article in English | MEDLINE | ID: mdl-34791577

ABSTRACT

Network inference is a valuable approach for gaining mechanistic insight from high-dimensional biological data. Existing methods for network inference focus on ranking all possible relations (edges) among all measured quantities such as genes, proteins, metabolites (features) observed, which yields a dense network that is challenging to interpret. Identifying a sparse, interpretable network using these methods thus requires an error-prone thresholding step which compromises their performance. In this article we propose a new method, DEKER-NET, that addresses this limitation by directly identifying a sparse, interpretable network without thresholding, improving real-world performance. DEKER-NET uses a novel machine learning method for feature selection in an iterative framework for network inference. DEKER-NET is extremely flexible, handling linear and nonlinear relations while making no assumptions about the underlying distribution of data, and is suitable for categorical or continuous variables. We test our method on the Dialogue for Reverse Engineering Assessments and Methods (DREAM) challenge data, demonstrating that it can directly identify sparse, interpretable networks without thresholding while maintaining performance comparable to the hypothetical best-case thresholded network of other methods.


Subject(s)
Algorithms , Gene Regulatory Networks , Machine Learning , Proteins
7.
Clin Pharmacol Ther ; 112(2): 210-223, 2022 08.
Article in English | MEDLINE | ID: mdl-34656074

ABSTRACT

Changes that accompany older age can alter the pharmacokinetics (PK), pharmacodynamics (PD), and likelihood of adverse effects (AEs) of a drug. However, older adults, especially the oldest or those with multiple chronic health conditions, polypharmacy, or frailty, are often under-represented in clinical trials of new drugs. Deficits in the current conduct of clinical evaluation of drugs for older adults and potential steps to fill those knowledge gaps are presented in this communication. The most important step is to increase clinical trial enrollment of older adults who are representative of the target treatment population. Unnecessary eligibility criteria should be eliminated. Physical and financial barriers to participation should be removed. Incentives could be created for inclusion of older adults. Enrollment goals should be established based on intended treatment indications, prevalence of the condition, and feasibility. Relevant clinical pharmacology data need to be obtained early enough to guide dosing and reduce risk for participation of older adults. Relevant PK and PD data as well as patient-centered outcomes should be measured during trials. Trial data should be analyzed for differences in PK, PD, effectiveness, and safety arising from differences in age or from the presence of conditions common in older adults. Postmarket evaluations with real-world evidence and drug labeling updates throughout the product lifecycle reflecting new knowledge are also needed. A comprehensive plan is needed to ensure adequate evaluation of the safety and effectiveness of drugs in older adults.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Polypharmacy , Aged , Drug Evaluation , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/prevention & control , Humans , Prevalence
8.
CPT Pharmacometrics Syst Pharmacol ; 10(3): 255-267, 2021 03.
Article in English | MEDLINE | ID: mdl-33440077

ABSTRACT

Letermovir is indicated for prophylaxis of cytomegalovirus infection and disease in allogeneic hematopoietic stem cell transplant (HSCT) recipients. Two-stage population pharmacokinetic (PK) modeling of letermovir was conducted to support dose rationale and evaluate the impact of intrinsic/extrinsic factors. Data from healthy phase I study participants over a wide dose range were modeled to evaluate the effects of selected intrinsic factors, including pharmacogenomics; next, phase III HSCT-recipient data at steady-state following clinical doses were modeled. The model in HSCT recipients adequately described letermovir PK following both oral or i.v. administration, and was consistent with the healthy participant model at steady-state clinical doses. Intrinsic factor effects were not clinically meaningful. These staged analyses indicate that letermovir PK in HSCT recipients and healthy participants differ only with respect to bioavailability and absorption rate. The HSCT recipient model was suitable for predicting exposure for exposure-response analysis supporting final dose selection.


Subject(s)
Acetates/pharmacokinetics , Antiviral Agents/pharmacokinetics , Cytomegalovirus Infections/prevention & control , Hematopoietic Stem Cell Transplantation/methods , Quinazolines/pharmacokinetics , Acetates/administration & dosage , Acetates/therapeutic use , Administration, Intravenous , Administration, Oral , Adult , Aged , Antiviral Agents/administration & dosage , Antiviral Agents/therapeutic use , Biological Availability , Case-Control Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers/statistics & numerical data , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Male , Middle Aged , Pharmacogenetics , Placebos/administration & dosage , Quinazolines/administration & dosage , Quinazolines/therapeutic use , Transplant Recipients/statistics & numerical data
9.
Clin Pharmacol Ther ; 107(6): 1296-1311, 2020 06.
Article in English | MEDLINE | ID: mdl-31889297

ABSTRACT

A model-informed drug discovery and development strategy played a key role in the novel glucose-responsive insulin MK-2640's early clinical development strategy and supported a novel clinical trial paradigm to assess glucose responsiveness. The development and application of in silico modeling approaches by leveraging substantial published clinical insulin pharmacokinetic-pharmacodynamic (PKPD) data and emerging preclinical and clinical data enabled rapid quantitative decision making. Learnings can be applied to define PKPD properties of novel insulins that could become therapeutically meaningful for diabetic patients.


Subject(s)
Blood Glucose/drug effects , Hypoglycemic Agents/administration & dosage , Insulin/analogs & derivatives , Models, Biological , Animals , Computer Simulation , Decision Making , Diabetes Mellitus/drug therapy , Drug Development , Drug Discovery/methods , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Insulin/administration & dosage , Insulin/pharmacokinetics , Insulin/pharmacology
10.
Int J Clin Pharmacol Ther ; 57(9): 450-457, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31232280

ABSTRACT

OBJECTIVE: Letermovir is an inhibitor of the terminase complex of cytomegalovirus (CMV) used as prophylactic therapy in CMV-seropositive allogeneic hematopoietic stem cell transplant recipients. As the combination oral contraceptive (COC) levonorgestrel/ethinyl estradiol (LNG/EE) may be coadministered in this target transplant population, the effects of letermovir on the pharmacokinetics (PK) of LNG and EE were investigated. MATERIALS AND METHODS: This was a phase I, open-label, fixed-sequence, two-period study conducted in healthy women (18 - 65 years old) of non-childbearing potential (protocol number: MK-8228 035). On day 1 of period 1, participants received a single dose of COC (LNG 0.15 mg/EE 0.03 mg). Following a 7-day washout, oral letermovir 480 mg was administered once-daily on days 1 - 12 of period 2, with a single dose of COC coadministered on day 8. Blood samples were collected to determine LNG and EE PK, and safety was assessed. RESULTS: The AUC0-∞ geometric mean ratios (90% confidence intervals) for COC + letermovir/COC alone were 1.36 (1.30, 1.43) for LNG and 1.42 (1.32, 1.52) for EE, indicating that letermovir coadministration increased COC exposure. Coadministration had no clinically-meaningful effect on Cmax, tmax, or apparent terminal T1/2 for either LNG or EE. All treatments were generally well tolerated. CONCLUSION: Letermovir coadministration with COC resulted in an increase in LNG and EE exposure in healthy adult women; however, levels were within the established safety margins. There was no decrease in LNG or EE exposure with no apparent risk of contraceptive failure on coadministration of letermovir and COC.
.


Subject(s)
Acetates/pharmacology , Contraceptives, Oral, Combined/pharmacokinetics , Ethinyl Estradiol/pharmacokinetics , Levonorgestrel/pharmacokinetics , Quinazolines/pharmacology , Adolescent , Adult , Aged , Drug Interactions , Female , Humans , Middle Aged , Young Adult
11.
J Clin Pharmacol ; 59(9): 1236-1243, 2019 09.
Article in English | MEDLINE | ID: mdl-31022310

ABSTRACT

The cytomegalovirus (CMV) viral terminase inhibitor letermovir is indicated for prevention of CMV infection in CMV-seropositive allogeneic hematopoietic stem cell transplant recipients. In this analysis, functional variants in solute carrier organic anion transporter family member 1B1 (SLCO1B1), uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1), and breast cancer resistance protein (BCRP) were assessed for effects on letermovir pharmacokinetics (PK) using pooled genetic information from 296 participants in 12 phase 1 studies. Letermovir area under the plasma concentration-time curve (AUC) was increased in carriers of the SLCO1B1 variant rs4149056 C allele relative to noncarriers with a geometric mean ratio (GMR) of 1.18 (95% confidence interval [CI], 1.06-1.30) for carriers of 1 copy and 1.42 (1.10-1.84) for carriers of 2 copies of the risk allele C compared with noncarriers. The SLCO1B1 variant rs4149032 T allele was associated with a decrease in letermovir AUC with GMR (95%CI) of 0.93 (0.85-1.02) and 0.82 (0.73-0.92) for carriers of 1 and 2 copies of the risk allele T, respectively, compared with noncarriers. The UGT1A1*6 variant rs4148323 A allele was present predominantly in Asian participants and was associated with an increase in letermovir AUC compared with noncarriers (GMR, 1.36; 95%CI, 1. 1.07-1.74). SLCO1B1 variant rs2306283, UGT1A1*28 TA promoter repeat, and BCRP variant rs2231142 had no effect on letermovir PK. Together, these data suggest that variants of enzymes and transporters that are involved in the disposition of letermovir in vitro may account for some variability in letermovir PK, but do not affect exposure to a clinically relevant extent.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Acetates/pharmacokinetics , Genetic Variation/genetics , Glucuronosyltransferase/genetics , Liver-Specific Organic Anion Transporter 1/genetics , Neoplasm Proteins/genetics , Quinazolines/pharmacokinetics , Adolescent , Adult , Aged , Alleles , Area Under Curve , Female , Humans , Male , Middle Aged , Pharmacogenomic Testing/methods , Promoter Regions, Genetic/genetics , Young Adult
12.
J Clin Pharmacol ; 59(10): 1331-1339, 2019 10.
Article in English | MEDLINE | ID: mdl-30990905

ABSTRACT

Letermovir (AIC246, MK-8228) is a human cytomegalovirus terminase inhibitor indicated for the prophylaxis of cytomegalovirus infection and disease in allogeneic hematopoietic stem cell transplant recipients that is also being investigated for use in other transplant settings. Many transplant patients receive immunosuppressant drugs, of which several have narrow therapeutic ranges. There is a potential for the coadministration of letermovir with these agents, and any potential effect on their pharmacokinetics (PK) must be understood. Five phase 1 trials were conducted in 73 healthy female participants to evaluate the effect of letermovir on the PK of cyclosporine, tacrolimus, sirolimus, and mycophenolic acid (active metabolite of mycophenolate mofetil [MMF]), as well as the effect of cyclosporine and MMF on letermovir PK. Safety and tolerability were also assessed. Coadministration of letermovir with cyclosporine, tacrolimus, and sirolimus resulted in 1.7-, 2.4-, and 3.4-fold increases in area under the plasma concentration-time curve and 1.1-, 1.6-, and 2.8-fold increases in maximum plasma concentration, respectively, of the immunosuppressants. Coadministration of letermovir and MMF had no meaningful effect on the PK of mycophenolic acid. Coadministration with cyclosporine increased letermovir area under the plasma concentration-time curve by 2.1-fold and maximum plasma concentration by 1.5-fold, while coadministration with MMF did not meaningfully affect the PK of letermovir. Given the increased exposures of cyclosporine, tacrolimus, and sirolimus, frequent monitoring of concentrations should be performed during administration and at discontinuation of letermovir, with dose adjustment as needed. These data support the reduction in clinical dosage of letermovir (to 240 mg) upon coadministration with cyclosporine.


Subject(s)
Acetates/pharmacokinetics , Cyclosporine/pharmacokinetics , Drug Interactions/physiology , Immunosuppressive Agents/pharmacokinetics , Mycophenolic Acid/pharmacokinetics , Quinazolines/pharmacokinetics , Sirolimus/pharmacokinetics , Tacrolimus/pharmacokinetics , Adolescent , Adult , Aged , Antiviral Agents/pharmacokinetics , Area Under Curve , Double-Blind Method , Female , Humans , Kidney Transplantation/methods , Middle Aged , Young Adult
13.
Clin Pharmacol Ther ; 105(2): 515-523, 2019 02.
Article in English | MEDLINE | ID: mdl-29901213

ABSTRACT

Letermovir is a human cytomegalovirus (CMV) terminase inhibitor for the prophylaxis of CMV infection in allogeneic hematopoietic stem-cell transplant (HSCT) recipients. In vitro, letermovir is a time-dependent inhibitor and an inducer of cytochrome P450 (CYP)3A, and an inhibitor of CYP2C8 and organic anion transporting polypeptide (OATP)1B. A stepwise approach was taken to qualify the interaction model of an existing letermovir physiologically based pharmacokinetic model to predict letermovir interactions with CYP3A and OATP1B. The model was then used to prospectively predict the interaction between letermovir and CYP2C8 substrates such as repaglinide, a substrate of CYP2C8, CYP3A, and OATP1B. The results showed that letermovir modestly increased the exposure of CYP2C8 substrates. These results were used to inform the US prescribing information in the absence of clinical drug-drug interaction studies. In addition, midazolam interactions with letermovir at therapeutic doses were also simulated to confirm that letermovir is a moderate CYP3A inhibitor.


Subject(s)
Acetates/pharmacokinetics , Antiviral Agents/pharmacokinetics , Drug Labeling , Quinazolines/pharmacokinetics , Adult , Cytochrome P-450 CYP2C8/metabolism , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytomegalovirus Infections/prevention & control , Drug Interactions , Female , Hematopoietic Stem Cell Transplantation/methods , Humans , Hypnotics and Sedatives/adverse effects , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Male , Midazolam/adverse effects , Models, Biological , Young Adult
14.
J Clin Pharmacol ; 58(7): 897-904, 2018 07.
Article in English | MEDLINE | ID: mdl-29578577

ABSTRACT

Letermovir is a human cytomegalovirus terminase inhibitor for cytomegalovirus infection prophylaxis in hematopoietic stem cell transplant recipients. Posaconazole (POS), a substrate of glucuronosyltransferase and P-glycoprotein, and voriconazole (VRC), a substrate of CYP2C9/19, are commonly administered to transplant recipients. Because coadministration of these azoles with letermovir is expected, the effect of letermovir on exposure to these antifungals was investigated. Two trials were conducted in healthy female subjects 18 to 55 years of age. In trial 1, single-dose POS 300 mg was administered alone, followed by a 7-day washout; then letermovir 480 mg once daily was given for 14 days with POS 300 mg coadministered on day 14. In trial 2, on day 1 VRC 400 mg was given every 12 hours; on days 2 and 3, VRC 200 mg was given every 12 hours, and on day 4 VRC 200 mg. On days 5 to 8, letermovir 480 mg was given once daily. Days 9 to 12 repeated days 1 to 4 coadministered with letermovir 480 mg once daily. In both trials, blood samples were collected for the assessment of the pharmacokinetic profiles of the antifungals, and safety was assessed. The geometric mean ratios (90%CIs) for POS+letermovir/POS area under the curve and peak concentration were 0.98 (0.83, 1.17) and 1.11 (0.95, 1.29), respectively. Voriconazole+letermovir/VRC area under the curve and peak concentration geometric mean ratios were 0.56 (0.51, 0.62) and 0.61 (0.53, 0.71), respectively. All treatments were generally well tolerated. Letermovir did not affect POS pharmacokinetics to a clinically meaningful extent but decreased VRC exposure. These results suggest that letermovir may be a perpetrator of CYP2C9/19-mediated drug-drug interactions.


Subject(s)
Acetates/pharmacokinetics , Antifungal Agents/pharmacokinetics , Antiviral Agents/pharmacokinetics , Quinazolines/pharmacokinetics , Triazoles/pharmacokinetics , Voriconazole/pharmacokinetics , Acetates/administration & dosage , Acetates/blood , Administration, Oral , Adult , Antifungal Agents/administration & dosage , Antiviral Agents/administration & dosage , Area Under Curve , Drug Combinations , Drug Interactions , Female , Healthy Volunteers , Humans , Middle Aged , Quinazolines/administration & dosage , Quinazolines/blood , Triazoles/administration & dosage , Triazoles/blood , Voriconazole/administration & dosage , Voriconazole/blood
15.
Breast ; 22(5): 839-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23523179

ABSTRACT

AIM: There is debate as to what constitutes an adequate excision margin to reduce the risk of locoregional recurrence (LRR) after breast cancer surgery. We have investigated the relationship between surgical margin distance and LRR in women with invasive breast cancer (IBC). METHODS: Tumour free margin distances were extracted from histopathology reports for women with IBC, treated by either breast conserving surgery or mastectomy, enrolled in the Breast Cancer Treatment Group Quality Assurance Project from July 1997 to June 2007. Cox proportional hazards regression analyses were conducted to compare the risk of LRR for involved margins compared with negative margins, measured in increments rounded to the nearest mm. RESULTS: 88 of 2300 patients (3.8%) experienced an LRR after a mean follow-up of 7.9 years. An involved margin, or a margin of 1 mm was associated with an increased risk of LRR (HR 2.72, 95% CI 1.30-5.69), whilst margin distances of 2 mm or greater were not. Risk of LRR with margin distances <2 mm was particularly high amongst those not receiving radiotherapy (RT). CONCLUSION: Based on our findings, we recommend that a tumour free margin distance of 2 mm be adopted as an adequate margin of excision for IBC, in the setting of patients receiving standard adjuvant RT and adjuvant drug therapies as dictated by the current clinical treatment paradigms.


Subject(s)
Breast Neoplasms/surgery , Carcinoma, Ductal, Breast/surgery , Carcinoma, Intraductal, Noninfiltrating/surgery , Mastectomy, Segmental/standards , Neoplasm Recurrence, Local/pathology , Quality Assurance, Health Care , Aged , Australia , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/radiotherapy , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Intraductal, Noninfiltrating/radiotherapy , Female , Humans , Middle Aged , Neoplasm, Residual , Radiotherapy, Adjuvant , Retrospective Studies
17.
Can J Gastroenterol ; 22(7): 637-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18629395

ABSTRACT

Malignant colonic polyps can be removed endoscopically but surgical resection is sometimes required. However, the polypectomy site can be difficult to locate. Current methods use various tattooing agents, with varying degrees of success. A new technique using preoperative injection of technetium-99m-labelled antimony colloid, with intraoperative localization using a handheld gamma probe, is described. Although unsuccessful in terms of localizing a previously partially resected polyp, the technique itself proved safe and simple, and has some advantages over other endoscopic approaches.


Subject(s)
Adenocarcinoma/diagnostic imaging , Antimony , Colonic Neoplasms/diagnostic imaging , Colonic Polyps/diagnostic imaging , Colonoscopy , Technetium Compounds , Adenocarcinoma/surgery , Aged , Antimony/administration & dosage , Colonic Neoplasms/surgery , Colonic Polyps/surgery , Gastric Mucosa , Humans , Injections , Male , Radionuclide Imaging , Technetium Compounds/administration & dosage
18.
PLoS Med ; 4(1): e23, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17378698

ABSTRACT

BACKGROUND: Graft-versus-host disease (GVHD) results from recognition of host antigens by donor T cells following allogeneic hematopoietic cell transplantation (AHCT). Notably, histoincompatibility between donor and recipient is necessary but not sufficient to elicit GVHD. Therefore, we tested the hypothesis that some donors may be "stronger alloresponders" than others, and consequently more likely to elicit GVHD. METHODS AND FINDINGS: To this end, we measured the gene-expression profiles of CD4(+) and CD8(+) T cells from 50 AHCT donors with microarrays. We report that pre-AHCT gene-expression profiling segregates donors whose recipient suffered from GVHD or not. Using quantitative PCR, established statistical tests, and analysis of multiple independent training-test datasets, we found that for chronic GVHD the "dangerous donor" trait (occurrence of GVHD in the recipient) is under polygenic control and is shaped by the activity of genes that regulate transforming growth factor-beta signaling and cell proliferation. CONCLUSIONS: These findings strongly suggest that the donor gene-expression profile has a dominant influence on the occurrence of GVHD in the recipient. The ability to discriminate strong and weak alloresponders using gene-expression profiling could pave the way to personalized transplantation medicine.


Subject(s)
Gene Expression Profiling , Graft vs Host Disease/diagnosis , Tissue Donors , Adult , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Discriminant Analysis , Female , Gene Expression Regulation , Hematopoietic Stem Cell Transplantation , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Perforin , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Time Factors , Transplantation, Homologous
19.
Curr Opin Chem Biol ; 10(4): 294-302, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16822703

ABSTRACT

Recent advances in the 'omics' technologies, scientific computing and mathematical modeling of biological processes have started to fundamentally impact the way we approach drug discovery. Recent years have witnessed the development of genome-scale functional screens, large collections of reagents, protein microarrays, databases and algorithms for data and text mining. Taken together, they enable the unprecedented descriptions of complex biological systems, which are testable by mathematical modeling and simulation. While the methods and tools are advancing, it is their iterative and combinatorial application that defines the systems biology approach.


Subject(s)
Drug Evaluation, Preclinical/methods , Genomics , Proteomics , Systems Biology/methods , Animals , Databases, Genetic , Models, Biological
20.
Bone ; 37(2): 159-69, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15921971

ABSTRACT

In this paper, we propose a mathematical model for parathyroid hormone receptor (PTH1R) kinetics, focusing on the receptor's response to PTH dosing to discern bone formation responses from bone resorption. The PTH1R is a major target for new osteoporosis treatments, as pulsatile PTH dosing has been shown to induce net bone formation in both animals and humans, and PTH(1-34) was recently FDA approved for the treatment of post-menopausal osteoporosis. PTH has also been shown to cause net bone loss when given continuously, so that the net action of PTH on bone is dependent on the dosing pattern. We have developed a simplified two-state receptor kinetics model for the PTH1R, based on the concepts of Segel et al., to distinguish the activity of active and inactive receptor and receptor-ligand complexes. The goal is to develop a plausible model of the minimal essential biological relationships necessary for understanding the responses to PTH dosing. A two-state model is able to effectively discriminate between continuous and pulsatile PTH dosing using the active species as surrogates for the downstream anabolic response. For continuous PTH dosing, the model predicts a desensitized system dominated by the inactive receptor and complex, consistent with downstream net bone loss that has been demonstrated experimentally. Using pulsatile PTH dosing, the model system predicts a highly sensitized state dominated by the active receptor and complex, corresponding to net bone formation. These results are consistent with the hypothesis that the kinetics of the receptor plays a critical role in the downstream effects of PTH dosing. Moreover, these results indicate that within a range of biologically relevant PTH doses, the two-state model is able to capture the differential behavior of the system for both continuous and pulsatile PTH dosing. The development of such a model provides a rational basis for developing more biologically extensive models that may support the design of optimal dosing strategies for PTH-based anti-osteoporosis treatments. Moreover, this model provides a unique starting point from which to design experiments investigating PTH receptor biology.


Subject(s)
Bone Resorption/therapy , Models, Biological , Parathyroid Hormone/administration & dosage , Receptor, Parathyroid Hormone, Type 1/physiology , Animals , Humans , Kinetics , Parathyroid Hormone/therapeutic use , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...