Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Clin Breast Cancer ; 22(2): 103-114, 2022 02.
Article in English | MEDLINE | ID: mdl-34903452

ABSTRACT

PRECLINICAL STUDIES: have demonstrated a complex cross-talk between Notch and estrogen signaling in ERα-positive breast cancer. Gamma-secretase inhibitors (GSIs) are investigational agents that block the cleavage and activation of Notch receptors. In animal models of endocrine-resistant breast cancer, combinations of tamoxifen and GSIs produce additive or synergistic efficacy, while decreasing the intestinal toxicity of GSIs. However, results of a clinical trial of a GSI-endocrine therapy combination in the metastatic setting have not been published to date, nor had the safety of such combinations been investigated with longer term treatment. We conducted a phase 1b dose escalation trial (NCT01149356) of GSI RO4929097 with exemestane in patients with ERα+, metastatic breast cancer (MBC) STUDY OBJECTIVES: To determine the safety, tolerability and maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D) of RO4929097 when administered in combination with exemestane in patients with estrogen receptor positive metastatic breast cancer RESULTS: We enrolled 15 patients with MBC. Of 14 evaluable patients, one had a partial response, 6 had stable disease and 7 progressive disease. Twenty % of patients had stable disease for ≥ 6 months. Common toxicities included nausea (73.3%), anorexia (60%), hyperglycemia (53.3%), hypophosphatemia (46.7%), fatigue (66.7%) and cough (33.0%). Grade 3 toxicities were uncommon, and included hypophosphatemia (13%) and rash (6.3%). Rash was the only DLT observed at 140 mg/d. Results suggest a possible recommended phase 2 dose of 90 mg/d. Ten patients with evaluable archival tissue showed expression of PKCα, which correlated with expression of Notch4. Mammospheres from a PKCα-expressing, endocrine-resistant T47D cell line were inhibited by a GSI-fulvestrant combination CONCLUSIONS: Our data indicate that combinations including endocrine therapy and Notch inhibitors deserve further investigation in endocrine-resistant ERα-positive breast cancer.


Subject(s)
Androstadienes/therapeutic use , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzazepines/therapeutic use , Breast Neoplasms/drug therapy , Fluorocarbons/therapeutic use , Aged , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis , Receptor, Notch3 , Receptors, Notch/therapeutic use
2.
Cancer Chemother Pharmacol ; 85(4): 673-683, 2020 04.
Article in English | MEDLINE | ID: mdl-32062691

ABSTRACT

PURPOSE: This study aimed to determine the safety, tolerability, and recommended phase II doses of trametinib plus uprosertib (GSK2141795) in patients with solid tumors likely to be sensitive to MEK and/or AKT inhibition. METHODS: This was a phase I, open-label, dose-escalation, and dose-expansion study in patients with triple-negative breast cancer or BRAF-wild type advanced melanoma. The primary outcome of the expansion study was investigator-assessed response. Among 126 enrolled patients, 63 received continuous oral daily dosing of trametinib and uprosertib, 29 received various alternative dosing schedules, and 34 were enrolled into expansion cohorts. Doses tested in the expansion cohort were trametinib 1.5 mg once daily (QD) + uprosertib 50 mg QD. RESULTS: Adverse events (AEs) were consistent with those reported in monotherapy studies but occurred at lower doses and with greater severity. Diarrhea was the most common dose-limiting toxicity; diarrhea and rash were particularly difficult to tolerate. Overall, 59% and 6% of patients reported AEs with a maximum severity of grade 3 and 4, respectively. Poor tolerability prevented adequate delivery of uprosertib with trametinib at a concentration predicted to have clinical activity. The study was terminated early based on futility in the continuous-dosing expansion cohorts and a lack of pharmacological or therapeutic advantage with intermittent dosing. The objective response rate was < 5% (1 complete response, 5 partial responses). CONCLUSIONS: Continuous and intermittent dosing of trametinib in combination with uprosertib was not tolerated, and minimal clinical activity was observed in all schedules tested.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Melanoma/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Cohort Studies , Diamines/administration & dosage , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Male , Maximum Tolerated Dose , Melanoma/pathology , Middle Aged , Prognosis , Pyrazoles/administration & dosage , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Tissue Distribution , Triple Negative Breast Neoplasms/pathology , Young Adult
3.
Invest Radiol ; 50(4): 195-204, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25360603

ABSTRACT

OBJECTIVES: The purpose of this study was to determine whether multiparametric magnetic resonance imaging (MRI) using dynamic contrast-enhanced MRI (DCE-MRI) and diffusion-weighted MRI (DWI), obtained before and after the first cycle of neoadjuvant chemotherapy (NAC), is superior to single-parameter measurements for predicting pathologic complete response (pCR) in patients with breast cancer. MATERIALS AND METHODS: Patients with stage II/III breast cancer were enrolled in an institutional review board-approved study in which 3-T DCE-MRI and DWI data were acquired before (n = 42) and after 1 cycle (n = 36) of NAC. Estimates of the volume transfer rate (K), extravascular extracellular volume fraction (ve), blood plasma volume fraction (vp), and the efflux rate constant (kep = K/ve) were generated from the DCE-MRI data using the Extended Tofts-Kety model. The apparent diffusion coefficient (ADC) was estimated from the DWI data. The derived parameter kep/ADC was compared with single-parameter measurements for its ability to predict pCR after the first cycle of NAC. RESULTS: The kep/ADC after the first cycle of NAC discriminated patients who went on to achieve a pCR (P < 0.001) and achieved a sensitivity, specificity, positive predictive value, and area under the receiver operator curve (AUC) of 0.92, 0.78, 0.69, and 0.88, respectively. These values were superior to the single parameters kep (AUC, 0.76) and ADC (AUC, 0.82). The AUCs between kep/ADC and kep were significantly different on the basis of the bootstrapped 95% confidence intervals (0.018-0.23), whereas the AUCs between kep/ADC and ADC trended toward significance (-0.11 to 0.24). CONCLUSIONS: The multiparametric analysis of DCE-MRI and DWI was superior to the single-parameter measurements for predicting pCR after the first cycle of NAC.


Subject(s)
Breast Neoplasms/drug therapy , Magnetic Resonance Imaging , Neoadjuvant Therapy/methods , Adult , Aged , Area Under Curve , Breast/pathology , Contrast Media , Diffusion Magnetic Resonance Imaging , Female , Humans , Image Enhancement , Middle Aged , Reproducibility of Results , Sensitivity and Specificity , Treatment Outcome
4.
Invest New Drugs ; 33(1): 159-68, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25294187

ABSTRACT

PURPOSE: This phase I study evaluated the safety, tolerability, maximum tolerated dose (MTD), and recommended phase II dose (RP2D) of tivantinib combined with sorafenib in patients with advanced solid tumors. MATERIALS AND METHODS: A standard 3 + 3 dose escalation design was used. At the RP2D, expansion cohorts in 5 tumor types could be enrolled. Pharmacogenetic and pharmacodynamic analysis were performed. RESULTS: Eighty-seven patients received the study treatment. The combination had no unexpected toxicities. The most common treatment-related adverse events (AE) were rash (40 %), diarrhea (38 %), and anorexia (33 %). The RP2D was tivantinib 360 mg BID and sorafenib 400 mg BID for all cancer histologies, except in hepatocellular carcinoma (HCC) patients tivantinib was 240 mg BID plus sorafenib 400 mg BID. The overall response rate was 12 % in all patients, 26 % in melanoma, 15 % in renal cell carcinoma (RCC), 10 % in HCC, and 0 % in other patients. Disease control rate (CR, PR and SD ≥8 weeks) was 58 % in all patients, 90 % in RCC, 65 % in HCC, 63 % in melanoma, 40 % in breast cancer, and 8 % in NSCLC patients. CONCLUSIONS: The combination treatment could be administered at full standard single-agent doses in all patients except those with HCC, where tivantinib was lowered to 240 mg BID. Preliminary evidence of anticancer activity was observed in patients with RCC, HCC, and melanoma, including patients refractory to sorafenib and/or other anti-VEGF pathway therapies. The combination treatment has therapeutic potential in treating a variety of solid tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cytochrome P-450 CYP2C19/genetics , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/genetics , Neoplasms/metabolism , Niacinamide/administration & dosage , Niacinamide/adverse effects , Niacinamide/analogs & derivatives , Niacinamide/pharmacokinetics , Niacinamide/pharmacology , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/adverse effects , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/pharmacology , Polymorphism, Genetic , Proto-Oncogene Proteins c-met/metabolism , Pyrrolidinones/administration & dosage , Pyrrolidinones/adverse effects , Pyrrolidinones/pharmacokinetics , Pyrrolidinones/pharmacology , Quinolines/administration & dosage , Quinolines/adverse effects , Quinolines/pharmacokinetics , Quinolines/pharmacology , Sorafenib , Treatment Outcome , Young Adult
5.
Ecancermedicalscience ; 8: 446, 2014.
Article in English | MEDLINE | ID: mdl-25114718

ABSTRACT

BACKGROUND: HER2 antagonists (anti-HER2; e.g., trastuzumab and lapatinib) are effective in treating an aggressive form of breast cancer (BC), but can cause cardiotoxicity due to the disruption in neuregulin (NRG)/HER2+ ligand receptor signalling. The recent data show that NRG-HER2 receptors located in the medulla oblongata are important regulators of vasomotor tone. Disrupting the NRG-HER2 signalling in mouse medulla results in increased sympathetic nerve output and blood pressure. We hypothesized that anti-HER2 agents would cause increased sympathetic tone with changes in plasma catecholamines and NRG. METHODS: In 15 newly diagnosed HER2+ BC patients receiving anti-HER2 agents, vital signs were measured along with supine plasma epinephrine (EPI), norepinephrine (NE), and NRG at baseline and three months. Serial echocardiography was performed. RESULTS: With three months of anti-HER2 treatment, NE increased (2.334 ± 1.294 nmol/L vs. 3.262 ± 2.103 nmol/L; p = 0.004) and NRG decreased (12.7±15.7 ng/ml vs. 10.9 ± 13.3 ng/ml; p = 0.036) with a corresponding increase in systolic blood pressure (110 ± 10 mmHg vs. 120 ± 16 mmHg, p = 0.049) and diastolic blood pressure (67 ± 14 vs. 77 ± 10, p = 0.009). There was no change, however, in EPI (0.183 ± 0.151 nmol/L vs. 0.159 ± 0.174 nmol/L; p = 0.519) or heart rate (73 ± 12 bpm vs. 77 ± 10 bpm, p = 0.146). Left ventricular ejection function declined over the follow-up period (baseline 63 ± 6% vs. follow-up 56 ± 5%). CONCLUSIONS: Anti-HER2 treatment results in increased NE, blood pressure, and decreased NRG; this suggests that the inhibition of NRGHER2 signalling leads to increased sympathoneural tone. Larger studies are needed to determine if these observations have prognostic value and may be offset with medical interventions, such as beta-blockers. CLINICAL TRIAL REGISTRATION: The study was registered with www.clinicaltrials.gov (NCT00875238).

6.
Oncologist ; 19(6): 616-22, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24797823

ABSTRACT

BACKGROUND: Oncogenic genetic alterations "drive" neoplastic cell proliferation. Small molecule inhibitors and antibodies are being developed that target an increasing number of these altered gene products. Next-generation sequencing (NGS) is a powerful tool to identify tumor-specific genetic changes. To determine the clinical impact of extensive genetic analysis, we reviewed our experience using a targeted NGS platform (FoundationOne) in advanced cancer patients. PATIENTS AND METHODS: We retrospectively assessed demographics, NGS results, and therapies received for patients undergoing targeted NGS (exonic sequencing of 236 genes and selective intronic sequencing from 19 genes) between April 2012 and August 2013. Coprimary endpoints were the percentage of patients with targeted therapy options uncovered by mutational profiling and the percentage who received genotype-directed therapy. RESULTS: Samples from 103 patients were tested, most frequently breast carcinoma (26%), head and neck cancers (23%), and melanoma (10%). Most patients (83%) were found to harbor potentially actionable genetic alterations, involving cell-cycle regulation (44%), phosphatidylinositol 3-kinase-AKT (31%), and mitogen-activated protein kinase (19%) pathways. With median follow-up of 4.1 months, 21% received genotype-directed treatments, most in clinical trials (61%), leading to significant benefit in several cases. The most common reasons for not receiving genotype-directed therapy were selection of standard therapy (35%) and clinical deterioration (13%). CONCLUSION: Mutational profiling using a targeted NGS panel identified potentially actionable alterations in a majority of advanced cancer patients. The assay identified additional therapeutic options and facilitated clinical trial enrollment. As time progresses, NGS results will be used to guide therapy in an increasing proportion of patients.


Subject(s)
Breast Neoplasms/genetics , Head and Neck Neoplasms/genetics , Melanoma/genetics , Neoplasm Proteins/genetics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Female , Head and Neck Neoplasms/pathology , High-Throughput Nucleotide Sequencing , Humans , Male , Melanoma/pathology , Middle Aged , Neoplasm Staging
7.
Magn Reson Med ; 71(4): 1592-602, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23661583

ABSTRACT

PURPOSE: The purpose of this pilot study is to determine (1) if early changes in both semiquantitative and quantitative DCE-MRI parameters, observed after the first cycle of neoadjuvant chemotherapy in breast cancer patients, show significant difference between responders and nonresponders and (2) if these parameters can be used as a prognostic indicator of the eventual response. METHODS: Twenty-eight patients were examined using DCE-MRI pre-, post-one cycle, and just prior to surgery. The semiquantitative parameters included longest dimension, tumor volume, initial area under the curve, and signal enhancement ratio related parameters, while quantitative parameters included K(trans), v(e), k(ep), v(p), and τ(i) estimated using the standard Tofts-Kety, extended Tofts-Kety, and fast exchange regime models. RESULTS: Our preliminary results indicated that the signal enhancement ratio washout volume and k(ep) were significantly different between pathologic complete responders from nonresponders (P < 0.05) after a single cycle of chemotherapy. Receiver operator characteristic analysis showed that the AUC of the signal enhancement ratio washout volume was 0.75, and the AUCs of k(ep) estimated by three models were 0.78, 0.76, and 0.73, respectively. CONCLUSION: In summary, the signal enhancement ratio washout volume and k(ep) appear to predict breast cancer response after one cycle of neoadjuvant chemotherapy. This observation should be confirmed with additional prospective studies.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Adult , Chemotherapy, Adjuvant/methods , Female , Humans , Image Enhancement/methods , Middle Aged , Neoadjuvant Therapy/methods , Pilot Projects , Prognosis , Reproducibility of Results , Sensitivity and Specificity , Treatment Outcome
8.
J Card Fail ; 19(1): 10-5, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23273589

ABSTRACT

BACKGROUND: Breast cancer (BC) treatments can cause heart failure (HF) in a subset of patients. ACC/AHA guidelines classify patients receiving cardiotoxic medications as stage A, a high-risk population for the development of HF. Circulating neuregulin (NRG) correlates with outcomes in stage C and D HF. We examined the levels of NRG in a BC cohort receiving cardiotoxic chemotherapy and its relationship with adverse cardiac effects during the transition from stage A to stage B or C HF. METHODS AND RESULTS: In an ongoing prospective study, a planned interim analysis of 78 BC women receiving either anthracycline (AC) or trastuzumab (Tsz) was performed. Biometric data, cardiac risk factors, and NRG levels, were collected before chemotherapy and after completion of AC therapy and/or 3 months into Tsz therapy. Cardiac function was measured by left ventricular ejection fraction (LVEF) by echocardiography at the above time points and longitudinally as standard of care. The interim cohort was predominately white with stage II BC and a median age of 50 years. A reduction of >10 absolute percentage points in LVEF was observed in 21.4% of the cohort, representing a transition from stage A to stage B or C HF. A statistically significant drop in plasma NRG was observed in women treated with AC and/or Tsz (P < .001). Additionally, baseline NRG correlated with the maximal change in LVEF. CONCLUSIONS: More than 20% of women experienced cardiac dysfunction, detected by decline in LVEF, and were reclassified as stage B or C HF. Plasma NRG levels were reduced after exposure to cardiotoxic chemotherapy, suggesting a loss in a cardioprotective growth factor. Higher baseline NRG levels were observed in those with the greatest decline in LVEF, supporting the continued investigation of NRG as a potential prognostic marker in early-stage HF.


Subject(s)
Antineoplastic Agents/adverse effects , Breast Neoplasms/drug therapy , Heart Failure/chemically induced , Heart Failure/pathology , Neuregulins/blood , Adult , Aged , Anthracyclines/adverse effects , Anthracyclines/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/administration & dosage , Biomarkers/blood , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cohort Studies , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Heart Failure/blood , Heart Failure/mortality , Humans , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Staging , Neuregulins/metabolism , Predictive Value of Tests , Prognosis , Prospective Studies , Risk Assessment , Severity of Illness Index , Statistics, Nonparametric , Stroke Volume/drug effects , Survival Rate , Trastuzumab
9.
Magn Reson Med ; 68(1): 261-71, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22127821

ABSTRACT

By fitting dynamic contrast-enhanced MRI data to an appropriate pharmacokinetic model, quantitative physiological parameters can be estimated. In this study, we compare four different models by applying four statistical measures to assess their ability to describe dynamic contrast-enhanced MRI data obtained in 28 human breast cancer patient sets: the chi-square test (χ(2)), Durbin-Watson statistic, Akaike information criterion, and Bayesian information criterion. The pharmacokinetic models include the fast exchange limit model with (FXL_v(p)) and without (FXL) a plasma component, and the fast and slow exchange regime models (FXR and SXR, respectively). The results show that the FXL_v(p) and FXR models yielded the smallest χ(2) in 45.64 and 47.53% of the voxels, respectively; they also had the smallest number of voxels showing serial correlation with 0.71 and 2.33%, respectively. The Akaike information criterion indicated that the FXL_v(p) and FXR models were preferred in 42.84 and 46.59% of the voxels, respectively. The Bayesian information criterion also indicated the FXL_v(p) and FXR models were preferred in 39.39 and 45.25% of the voxels, respectively. Thus, these four metrics indicate that the FXL_v(p) and the FXR models provide the most complete statistical description of dynamic contrast-enhanced MRI time courses for the patients selected in this study.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Gadolinium DTPA/pharmacokinetics , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Models, Biological , Computer Simulation , Contrast Media/pharmacokinetics , Data Interpretation, Statistical , Female , Humans , Image Enhancement/methods , Models, Statistical , Reproducibility of Results , Sensitivity and Specificity
10.
Phys Med Biol ; 56(17): 5753-69, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21841212

ABSTRACT

Quantitative analysis of dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) data requires the accurate determination of the arterial input function (AIF). A novel method for obtaining the AIF is presented here and pharmacokinetic parameters derived from individual and population-based AIFs are then compared. A Philips 3.0 T Achieva MR scanner was used to obtain 20 DCE-MRI data sets from ten breast cancer patients prior to and after one cycle of chemotherapy. Using a semi-automated method to estimate the AIF from the axillary artery, we obtain the AIF for each patient, AIF(ind), and compute a population-averaged AIF, AIF(pop). The extended standard model is used to estimate the physiological parameters using the two types of AIFs. The mean concordance correlation coefficient (CCC) for the AIFs segmented manually and by the proposed AIF tracking approach is 0.96, indicating accurate and automatic tracking of an AIF in DCE-MRI data of the breast is possible. Regarding the kinetic parameters, the CCC values for K(trans), v(p) and v(e) as estimated by AIF(ind) and AIF(pop) are 0.65, 0.74 and 0.31, respectively, based on the region of interest analysis. The average CCC values for the voxel-by-voxel analysis are 0.76, 0.84 and 0.68 for K(trans), v(p) and v(e), respectively. This work indicates that K(trans) and v(p) show good agreement between AIF(pop) and AIF(ind) while there is a weak agreement on v(e).


Subject(s)
Axillary Artery/physiopathology , Breast Neoplasms/blood supply , Contrast Media/pharmacokinetics , Gadolinium DTPA/pharmacokinetics , Magnetic Resonance Imaging/methods , Models, Biological , Perfusion Imaging/methods , Algorithms , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Computer Simulation , Female , Humans , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Reproducibility of Results , Sensitivity and Specificity
11.
J Magn Reson Imaging ; 33(5): 1063-70, 2011 May.
Article in English | MEDLINE | ID: mdl-21509862

ABSTRACT

PURPOSE: To provide a quantitative assessment of motion and distortion correction of diffusion-weighted images (DWIs) of the breast and to evaluate the effects of registration on the mean apparent diffusion coefficient (mADC). MATERIALS AND METHODS: Eight datasets from four patients with breast cancer and eight datasets from six healthy controls were acquired on a 3T scanner. A 3D affine registration was used to align each set of images and principal component analysis was used to assess the results. Variance in tumor ADC measurements, tumor mADC values, and voxel-wise tumor mADC values were compared before and after registration for each patient. RESULTS: Image registration significantly (P = 0.008) improved image alignment for both groups and significantly (P < 0.001) reduced the variance across individual tumor ADC measurements. While misalignment led to potential under- and overestimation of mADC values for individual voxels, average tumor mADC values did not significantly change (P > 0.09) after registration. CONCLUSION: 3D affine registration improved the alignment of DWIs of the breast and reduced the variance between ADC measurements. Although the reduced variance did not significantly change tumor region-of-interest measures of mADC, it may have a significant impact on voxel-based analyses.


Subject(s)
Breast Neoplasms/diagnosis , Breast/pathology , Diffusion Magnetic Resonance Imaging/methods , Adult , Algorithms , Artifacts , Breast Neoplasms/pathology , Female , Humans , Image Processing, Computer-Assisted/methods , Middle Aged , Models, Statistical , Motion , Principal Component Analysis
12.
Med Phys ; 37(6): 2541-52, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20632566

ABSTRACT

PURPOSE: The authors present a method to validate coregistration of breast magnetic resonance images obtained at multiple time points during the course of treatment. In performing sequential registration of breast images, the effects of patient repositioning, as well as possible changes in tumor shape and volume, must be considered. The authors accomplish this by extending the adaptive bases algorithm (ABA) to include a tumor-volume preserving constraint in the cost function. In this study, the authors evaluate this approach using a novel validation method that simulates not only the bulk deformation associated with breast MR images obtained at different time points, but also the reduction in tumor volume typically observed as a response to neoadjuvant chemotherapy. METHODS: For each of the six patients, high-resolution 3D contrast enhanced T1-weighted images were obtained before treatment, after one cycle of chemotherapy and at the conclusion of chemotherapy. To evaluate the effects of decreasing tumor size during the course of therapy, simulations were run in which the tumor in the original images was contracted by 25%, 50%, 75%, and 95%, respectively. The contracted area was then filled using texture from local healthy appearing tissue. Next, to simulate the post-treatment data, the simulated (i.e., contracted tumor) images were coregistered to the experimentally measured post-treatment images using a surface registration. By comparing the deformations generated by the constrained and unconstrained version of ABA, the authors assessed the accuracy of the registration algorithms. The authors also applied the two algorithms on experimental data to study the tumor volume changes, the value of the constraint, and the smoothness of transformations. RESULTS: For the six patient data sets, the average voxel shift error (mean +/- standard deviation) for the ABA with constraint was 0.45 +/- 0.37, 0.97 +/- 0.83, 1.43 +/- 0.96, and 1.80 +/- 1.17 mm for the 25%, 50%, 75%, and 95% contraction simulations, respectively. In comparison, the average voxel shift error for the unconstrained ABA was 0.46 +/- 0.29, 1.13 +/- 1.17, 2.40 +/- 2.04, and 3.53 +/- 2.89 mm, respectively. These voxel shift errors translate into compression of the tumor volume: The ABA with constraint returned volumetric errors of 2.70 +/- 4.08%, 7.31 +/- 4.52%, 9.28 +/- 5.55%, and 13.19 +/- 6.73% for the 25%, 50%, 75%, and 95% contraction simulations, respectively, whereas the unconstrained ABA returned volumetric errors of 4.00 +/- 4.46%, 9.93 +/- 4.83%, 19.78 +/- 5.657%, and 29.75 +/- 15.18%. The ABA with constraint yields a smaller mean shift error, as well as a smaller volume error (p = 0.031 25 for the 75% and 95% contractions), than the unconstrained ABA for the simulated sets. Visual and quantitative assessments on experimental data also indicate a good performance of the proposed algorithm. CONCLUSIONS: The ABA with constraint can successfully register breast MR images acquired at different time points with reasonable error. To the best of the authors' knowledge, this is the first report of an attempt to quantitatively assess in both phantoms and a set of patients the accuracy of a registration algorithm for this purpose.


Subject(s)
Algorithms , Breast Neoplasms/pathology , Breast/pathology , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/instrumentation , Phantoms, Imaging , Subtraction Technique , Equipment Design , Equipment Failure Analysis , Female , Humans , Image Enhancement/methods , Magnetic Resonance Imaging/methods , Reproducibility of Results , Sensitivity and Specificity
13.
Clin Cancer Res ; 16(2): 681-90, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20068102

ABSTRACT

PURPOSE: To identify molecular markers of pathologic response to neoadjuvant paclitaxel/radiation treatment, protein and gene expression profiling were done on pretreatment biopsies. EXPERIMENTAL DESIGN: Patients with high-risk, operable breast cancer were treated with three cycles of paclitaxel followed by concurrent paclitaxel/radiation. Tumor tissue from pretreatment biopsies was obtained from 19 of the 38 patients enrolled in the study. Protein and gene expression profiling were done on serial sections of the biopsies from patients that achieved a pathologic complete response (pCR) and compared to those with residual disease, non-pCR (NR). RESULTS: Proteomic and validation immunohistochemical analyses revealed that alpha-defensins (DEFA) were overexpressed in tumors from patients with a pCR. Gene expression analysis revealed that MAP2, a microtubule-associated protein, had significantly higher levels of expression in patients achieving a pCR. Elevation of MAP2 in breast cancer cell lines led to increased paclitaxel sensitivity. Furthermore, expression of genes that are associated with the basal-like, triple-negative phenotype were enriched in tumors from patients with a pCR. Analysis of a larger panel of tumors from patients receiving presurgical taxane-based treatment showed that DEFA and MAP2 expression as well as histologic features of inflammation were all statistically associated with response to therapy at the time of surgery. CONCLUSION: We show the utility of molecular profiling of pretreatment biopsies to discover markers of response. Our results suggest the potential use of immune signaling molecules such as DEFA as well as MAP2, a microtubule-associated protein, as tumor markers that associate with response to neoadjuvant taxane-based therapy.


Subject(s)
Biomarkers, Pharmacological/analysis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Paclitaxel/therapeutic use , Taxoids/therapeutic use , Biomarkers, Pharmacological/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Carcinoma/drug therapy , Carcinoma/radiotherapy , Combined Modality Therapy , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Genomics , Humans , Metabolome/drug effects , Neoadjuvant Therapy , Oligonucleotide Array Sequence Analysis , Proteomics , Taxoids/pharmacology , Tumor Cells, Cultured
14.
J Magn Reson Imaging ; 30(1): 121-34, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19557727

ABSTRACT

PURPOSE: To assess the temporal sampling requirements needed for quantitative analysis of dynamic contrast-enhanced MRI (DCE-MRI) data with a reference region (RR) model in human breast cancer. MATERIALS AND METHODS: Simulations were used to study errors in pharmacokinetic parameters (K(trans) and v(e)) estimated by the RR model using six DCE-MRI acquisitions over a range of pharmacokinetic parameter values, arterial input functions, and temporal samplings. DCE-MRI data were acquired on 12 breast cancer patients and parameters were estimated using the native resolution data (16.4 seconds) and compared to downsampled 32.8-second and 65.6-second data. RESULTS: Simulations show that, in the majority of parameter combinations, the RR model results in an error less than 20% in the extracted parameters with temporal sampling as poor as 35.6 seconds. The experimental results show a high correlation between K(trans) and v(e) estimates from data acquired at 16.4-second temporal resolution compared to the downsampled 32.8-second data: the slope of the regression line was 1.025 (95% confidence interval [CI]: 1.021, 1.029), Pearson's correlation r = 0.943 (95% CI: 0.940, 0.945) for K(trans), and 1.023 (95% CI: 1.021. 1.025), r = 0.979 (95% CI: 0.978, 0.980) for v(e). For the 64-second temporal resolution data the results were: 0.890 (95% CI: 0.894, 0.905), r = 0.8645, (95% CI: 0.858, 0.871) for K(trans), and 1.041 (95% CI: 1.039, 1.043), r = 0.970 (95% CI: 0.968, 0.971) for v(e). CONCLUSION: RR analysis allows for a significant reduction in temporal sampling requirements and this lends itself to analyze DCE-MRI data acquired in practical situations.


Subject(s)
Breast Neoplasms/pathology , Computer Simulation , Contrast Media , Gadolinium DTPA , Image Enhancement/methods , Magnetic Resonance Imaging/methods , Models, Biological , Breast/pathology , Computer Simulation/statistics & numerical data , Contrast Media/pharmacokinetics , Female , Gadolinium DTPA/pharmacokinetics , Humans , Image Processing, Computer-Assisted/methods , Reproducibility of Results , Sensitivity and Specificity , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...