Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jennifer M. Specht is active.

Publication


Featured researches published by Jennifer M. Specht.


Journal of Clinical Oncology | 2014

Phase III Study of Iniparib Plus Gemcitabine and Carboplatin Versus Gemcitabine and Carboplatin in Patients With Metastatic Triple-Negative Breast Cancer

Joyce O'Shaughnessy; Lee S. Schwartzberg; Michael A. Danso; Kathy D. Miller; Hope S. Rugo; Marcus A. Neubauer; Nicholas J. Robert; Beth A. Hellerstedt; Mansoor N. Saleh; Paul Richards; Jennifer M. Specht; Denise A. Yardley; Robert W. Carlson; Richard S. Finn; Eric Charpentier; Ignacio Garcia-Ribas

PURPOSE There is a lack of treatments providing survival benefit for patients with metastatic triple-negative breast cancer (mTNBC), with no standard of care. A randomized phase II trial showed significant benefit for gemcitabine, carboplatin, and iniparib (GCI) over gemcitabine and carboplatin (GC) in clinical benefit rate, response rate, progression-free survival (PFS), and overall survival (OS). Here, we formally compare the efficacy of these regimens in a phase III trial. PATIENTS AND METHODS Patients with stage IV/locally recurrent TNBC who had received no more than two previous chemotherapy regimens for mTNBC were randomly allocated to gemcitabine 1,000 mg/m(2) and carboplatin area under the curve 2 (days 1 and 8) alone or GC plus iniparib 5.6 mg/kg (days 1, 4, 8, and 11) every 3 weeks. Random assignment was stratified by the number of prior chemotherapies. The coprimary end points were OS and PFS. Patients receiving GC could cross over to iniparib on progression. RESULTS Five hundred nineteen patients were randomly assigned (261 GCI; 258 GC). In the primary analysis, no statistically significant difference was observed for OS (hazard ratio [HR] = 0.88; 95% CI, 0.69 to 1.12; P = .28) nor PFS (HR = 0.79; 95% CI, 0.65 to 0.98; P = .027). An exploratory analysis showed that patients in the second-/third-line had improved OS (HR = 0.65; 95% CI, 0.46 to 0.91) and PFS (HR = 0.68; 95% CI, 0.50 to 0.92) with GCI. The safety profile for GCI was similar to GC. CONCLUSION The trial did not meet the prespecified criteria for the coprimary end points of PFS and OS in the ITT population. The potential benefit with iniparib observed in second-/third-line subgroup warrants further evaluation.


Journal of Clinical Oncology | 2008

Tumor Metabolism and Blood Flow Changes by Positron Emission Tomography: Relation to Survival in Patients Treated With Neoadjuvant Chemotherapy for Locally Advanced Breast Cancer

Lisa K. Dunnwald; Julie R. Gralow; Georgiana K. Ellis; Robert B. Livingston; Hannah M. Linden; Jennifer M. Specht; Robert K. Doot; Thomas J. Lawton; William E. Barlow; Brenda F. Kurland; Erin K. Schubert; David A. Mankoff

PURPOSE Patients with locally advanced breast carcinoma (LABC) receive preoperative chemotherapy to provide early systemic treatment and assess in vivo tumor response. Serial positron emission tomography (PET) has been shown to predict pathologic response in this setting. We evaluated serial quantitative PET tumor blood flow (BF) and metabolism as in vivo measurements to predict patient outcome. PATIENTS AND METHODS Fifty-three women with primary LABC underwent dynamic [(18)F]fluorodeoxyglucose (FDG) and [(15)O]water PET scans before and at midpoint of neoadjuvant chemotherapy. The FDG metabolic rate (MRFDG) and transport (FDG K(1)) parameters were calculated; BF was estimated from the [(15)O]water study. Associations between BF, MRFDG, FDG K(1), and standardized uptake value and disease-free survival (DFS) and overall survival (OS) were evaluated using the Cox proportional hazards model. RESULTS Patients with persistent or elevated BF and FDG K(1) from baseline to midtherapy had higher recurrence and mortality risks than patients with reductions. In multivariable analyses, BF and FDG K(1) changes remained independent prognosticators of DFS and OS. For example, in the association between BF and mortality, a patient with a 5% increase in tumor BF had a 67% higher mortality risk compared with a patient with a 5% decrease in tumor BF (hazard ratio = 1.67; 95% CI, 1.24 to 2.24; P < .001). CONCLUSION LABC patients with limited or no decline in BF and FDG K(1) experienced higher recurrence and mortality risks that were greater than the effects of clinical tumor characteristics. Tumor perfusion changes over the course of neoadjuvant chemotherapy measured directly by [(15)O]water or indirectly by dynamic FDG predict DFS and OS.


Clinical Cancer Research | 2011

Fluoroestradiol Positron Emission Tomography Reveals Differences in Pharmacodynamics of Aromatase Inhibitors, Tamoxifen, and Fulvestrant in Patients with Metastatic Breast Cancer

Hannah M. Linden; Brenda F. Kurland; Lanell M. Peterson; Erin K. Schubert; Julie R. Gralow; Jennifer M. Specht; Georgiana K. Ellis; Thomas J. Lawton; Robert B. Livingston; Philip H. Petra; Jeanne M. Link; Kenneth A. Krohn; David A. Mankoff

Purpose: To determine, by molecular imaging, how in vivo pharmacodynamics of estrogen-estrogen receptor (ER) binding differ between types of standard endocrine therapy. Experimental Design: The ER has been a highly successful target for breast cancer treatment. ER-directed treatments include lowering ligand concentration by using aromatase inhibitors (AI) and blocking the receptor with agents like tamoxifen (TAM) or fulvestrant (FUL). We measured regional estrogen-ER binding by using positron emission tomography with 18F-fluoroestradiol (FES PET) prior to and during treatment with AI, TAM, or FUL in a series of 30 metastatic breast cancer patients. FES PET measured in vivo estrogen binding at all tumor sites in heavily pretreated women with metastatic bone soft tissue–dominant breast cancer. In patients with uterus (n = 16) changes in uterine FES uptake were also measured. Results: As expected, tumor FES uptake declined more markedly on ER blockers (TAM and FUL, average 54% decline) compared with a less than 15% average decline on estrogen-depleting AIs (P < 0.001). The rate of complete tumor blockade [FES standardized uptake value (SUV) ≤1.5] following TAM (5/5 patients) was greater than the blockade rate following FUL (4/11; 2-sided mid P = 0.019). Percent FES SUV change in the uterus showed a strong association with tumoral change (ρ = 0.63, P = 0.01). Conclusions: FES PET can assess the in vivo pharmacodynamics of ER-targeted agents and may give insight into the activity of established therapeutic agents. Imaging revealed significant differences between agents, including differences in the efficacy of blockade by different ER antagonists in current clinical use. Clin Cancer Res; 17(14); 4799–805. ©2011 AACR.


The Journal of Nuclear Medicine | 2010

Kinetic analysis of 18F-fluoride PET images of breast cancer bone metastases

Robert K. Doot; Mark Muzi; Lanell M. Peterson; Erin K. Schubert; Julie R. Gralow; Jennifer M. Specht; David A. Mankoff

The most common site of metastasis for breast cancer is bone. Quantitative 18F-fluoride PET can estimate the kinetics of fluoride incorporation into bone as a measure of fluoride transport, bone formation, and turnover. The purpose of this analysis was to evaluate the accuracy and precision of 18F-fluoride model parameter estimates for characterizing regional kinetics in metastases and normal bone in breast cancer patients. Methods: Twenty metastatic breast cancer patients underwent dynamic 18F-fluoride PET. Mean activity concentrations were measured from serial blood samples and regions of interest placed over bone metastases, normal vertebrae, and cardiac blood pools. This study examined parameter identifiability, model sensitivity, error, and accuracy using parametric values from the patient cohort. Results: Representative time–activity curves and model parameter ranges were obtained from the patient cohort. Model behavior analyses of these data indicated 18F-fluoride transport and flux (K1 and Ki, respectively) into metastatic and normal osseous tissue could be independently estimated with a reasonable bias of 9% or less and reasonable precision (coefficients of variation ≤ 16%). Average 18F-fluoride transport and flux into metastases from 20 patients (K1 = 0.17 ± 0.08 mL·cm−3·min−1 and Ki = 0.10 ± 0.05 mL·cm−3·min−1) were both significantly higher than for normal bone (K1 = 0.09 ± 0.03 mL·cm−3·min−1 and Ki = 0.05 ± 0.02 mL·cm−3·min−1, P < 0.001). Conclusion: Fluoride transport and flux can be accurately and independently estimated for bone metastases and normal vertebrae. Reasonable bias and precision for estimates of K1 and Ki from simulations and significant differences in values from patient modeling results in metastases and normal bone suggest that 18F-fluoride PET images may be useful for assessing changes in bone turnover in response to therapy. Future studies will examine the correlation of parameters to biologic features of bone metastases and to response to therapy.


Clinical Cancer Research | 2011

PET Tumor Metabolism in Locally Advanced Breast Cancer Patients Undergoing Neoadjuvant Chemotherapy: Value of Static versus Kinetic Measures of Fluorodeoxyglucose Uptake

Lisa K. Dunnwald; Robert K. Doot; Jennifer M. Specht; Julie R. Gralow; Georgiana K. Ellis; Robert B. Livingston; Hannah M. Linden; Vijayakrishna K. Gadi; Brenda F. Kurland; Erin K. Schubert; Mark Muzi; David A. Mankoff

Purpose: Changes in tumor metabolism from positron emission tomography (PET) in locally advanced breast cancer (LABC) patients treated with neoadjuvant chemotherapy (NC) are predictive of pathologic response. Serial dynamic [18F]-FDG (fluorodeoxyglucose) PET scans were used to compare kinetic parameters with the standardized uptake value (SUV) as predictors of pathologic response, disease-free survival (DFS), and overall survival (OS). Experimental Design: Seventy-five LABC patients underwent FDG PET prior to and at midpoint of NC. FDG delivery (K1), FDG flux (Ki), and SUV measures were calculated and compared by clinical and pathologic tumor characteristics using regression methods and area under the receiver operating characteristic curve (AUC). Associations between K1, Ki, and SUV and DFS and OS were evaluated using the Cox proportional hazards model. Results: Tumors that were hormone receptor negative, high grade, highly proliferative, or of ductal histology had higher FDG Ki and SUV values; on an average, FDG K1 did not differ systematically by tumor features. Predicting pathologic response in conjunction with estrogen receptor (ER) and axillary lymph node positivity, kinetic measures (AUC = 0.97) were more robust predictors than SUV (AUC = 0.84, P = 0.005). Changes in K1 and Ki predicted both DFS and OS, whereas changes in SUV predicted OS only. In multivariate modeling, only changes in K1 remained an independent prognosticator of DFS and OS. Conclusion: Kinetic measures of FDG PET for LABC patients treated with NC accurately measured treatment response and predicted outcome compared with static SUV measures, suggesting that kinetic analysis may hold advantage of static uptake measures for response assessment. Clin Cancer Res; 17(8); 2400–9. ©2011 AACR.


Clinical Cancer Research | 2010

Tumor Metabolism and Blood Flow as Assessed by Positron Emission Tomography Varies by Tumor Subtype in Locally Advanced Breast Cancer

Jennifer M. Specht; Brenda F. Kurland; Susan Montgomery; Lisa K. Dunnwald; Robert K. Doot; Julie R. Gralow; Georgina K. Ellis; Hannah M. Linden; Robert B. Livingston; Kimberly H. Allison; Erin K. Schubert; David A. Mankoff

Purpose: Dynamic positron emission tomography (PET) imaging can identify patterns of breast cancer metabolism and perfusion in patients receiving neoadjuvant chemotherapy (NC) that are predictive of response. This analysis examines tumor metabolism and perfusion by tumor subtype. Experimental Design: Tumor subtype was defined by immunohistochemistry in 71 patients with locally advanced breast cancer undergoing NC. Subtype was defined as luminal [estrogen receptor (ER)/progesterone receptor (PR) positive], triple negative [TN; ER/PR negative, human epidermal growth factor receptor 2 (HER2) negative], and HER2 (ER/PR negative, HER2 overexpressing). Metabolic rate (MRFDG) and blood flow (BF) were calculated from PET imaging before NC. Pathologic complete response (pCR) to NC was classified as pCR versus other. Results: Twenty-five (35%) of 71 patients had TN tumors; 6 (8%) were HER2 and 40 (56%) were luminal. MRFDG for TN tumors was on average 67% greater than for luminal tumors (95% confidence interval, 9-156%) and average MRFDG/BF ratio was 53% greater in TN compared with luminal tumors (95% confidence interval, 9-114%; P < 0.05 for both). Average BF levels did not differ by subtype (P = 0.73). Most luminal tumors showed relatively low MRFDG and BF (and did not achieve pCR); high MRFDG was generally matched with high BF in luminal tumors and predicted pCR. This was not true in TN tumors. Conclusion: The relationship between breast tumor metabolism and perfusion differed by subtype. The high MRFDG/BF ratio that predicts poor response to NC was more common in TN tumors. Metabolism and perfusion measures may identify subsets of tumors susceptible and resistant to NC and may help direct targeted therapy. Clin Cancer Res; 16(10); 2803–10. ©2010 AACR.


Seminars in Radiation Oncology | 2009

Neoadjuvant chemotherapy for locally advanced breast cancer.

Jennifer M. Specht; Julie R. Gralow

Preoperative systemic therapy is the standard of care in locally advanced breast cancer. In this setting, the intent of preoperative systemic therapy is to expand surgical options and to improve survival. Locally advanced and inflammatory breast cancer have different biological features, but they share the use of preoperative (primary, neoadjuvant) systemic therapy as the initial treatment of choice. The management of these patients necessitates involvement of a multidisciplinary team from the onset and during therapy. The eradication of invasive cancer from the breast and axillary lymph nodes, pathologic complete response, is a predictor of outcome associated with improved disease-free and overall survival. However, conventional chemotherapy regimens result in pathologic complete response in only a minority of patients. The management of patients with residual invasive disease after preoperative therapy is a common clinical problem for which additional research is necessary. The differential expression of genes and pathways in locally advanced and inflammatory breast cancer allows for the exploitation of targeted therapy, and early trials have shown exciting target and tumor effects. Much work remains, and future trials combining targeted and conventional therapies based on molecular subtypes and/or specific targets are needed if we hope to improve survival for patients with locally advanced breast cancer.


Journal of Magnetic Resonance Imaging | 2010

Association between serial dynamic contrast‐enhanced MRI and dynamic 18F‐FDG PET measures in patients undergoing neoadjuvant chemotherapy for locally advanced breast cancer

Savannah C. Partridge; Risa K. Vanantwerp; Robert K. Doot; Xiaoyu Chai; Brenda F. Kurland; Peter R. Eby; Jennifer M. Specht; Lisa K. Dunnwald; Erin K. Schubert; Constance D. Lehman; David A. Mankoff

To investigate the relationship between changes in vascularity and metabolic activity measured by dynamic contrast‐enhanced magnetic resonance imaging (DCE‐MRI) and dynamic 18F‐FDG‐positron emission tomography (PET) in breast tumors undergoing neoadjuvant chemotherapy.


Clinical Cancer Research | 2009

Blood Flow-Metabolism Mismatch: Good for the Tumor, Bad for the Patient

David A. Mankoff; Lisa K. Dunnwald; Savannah C. Partridge; Jennifer M. Specht

Although tightly coupled in most normal tissues, blood flow and metabolism are often not well matched in tumors. A flow-metabolism mismatch, specifically, high metabolism relative to blood flow, can be recognized in tumors by functional and molecular imaging and is associated with poor response to treatment and early relapse or disease progression. (Clin Cancer Res 2009;15(17):5294–6)


Clinical Cancer Research | 2016

Phase I Study of Veliparib (ABT-888) Combined with Cisplatin and Vinorelbine in Advanced Triple-Negative Breast Cancer and/or BRCA Mutation–Associated Breast Cancer

Eve Rodler; Brenda F. Kurland; Melissa Griffin; Julie R. Gralow; Peggy L. Porter; Rosa Yeh; Vijayakrishna K. Gadi; Jamie Guenthoer; Jan H. Beumer; Larissa A. Korde; Sandra Strychor; Brian F. Kiesel; Hannah M. Linden; John A. Thompson; Elizabeth M. Swisher; Xiaoyu Chai; Stacie Peacock Shepherd; Vincent L. Giranda; Jennifer M. Specht

Purpose: Cisplatin is synergistic with vinorelbine and the PARP inhibitor veliparib, and has antineoplastic activity in triple-negative breast cancer (TNBC) and BRCA mutation–associated breast cancer. This phase I study assessed veliparib with cisplatin and vinorelbine. Experimental Design: A 3+3 dose-escalation design evaluated veliparib administered twice daily for 14 days with cisplatin (75 mg/m2 day 1) and vinorelbine (25 mg/m2 days 1, 8) every 21 days, for 6 to 10 cycles, followed by veliparib monotherapy. Pharmacokinetics, measurement of poly(ADP-ribose) in peripheral blood mononuclear cells, and preliminary efficacy were assessed. IHC and gene-expression profiling were evaluated as potential predictors of response. Results: Forty-five patients enrolled in nine dose cohorts plus five in an expansion cohort at the highest dose level and recommended phase II dose, 300 mg twice daily. The MTD of veliparib was not reached. Neutropenia (36%), anemia (30%), and thrombocytopenia (12%) were the most common grade 3/4 adverse events. Best overall response for 48 patients was radiologic response with 9-week confirmation for 17 (35%; 2 complete, 15 partial), and stable disease for 21 (44%). Germline BRCA mutation presence versus absence was associated with 6-month progression-free survival [PFS; 10 of 14 (71%) vs. 8 of 27 (30%), mid-P = 0.01]. Median PFS for all 50 patients was 5.5 months (95% confidence interval, 4.1–6.7). Conclusions: Veliparib at 300 mg twice daily combined with cisplatin and vinorelbine is well tolerated with encouraging response rates. A phase II randomized trial is planned to assess veliparibs contribution to cisplatin chemotherapy in metastatic TNBC and BRCA mutation–associated breast cancer. Clin Cancer Res; 22(12); 2855–64. ©2016 AACR.

Collaboration


Dive into the Jennifer M. Specht's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vijayakrishna K. Gadi

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert K. Doot

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge