26 results on '"Shee, K"'
Search Results
2. Abstract P5-04-08: Timing provides context for the paradoxical effects of AMPK activation in ER+ breast cancer: Suppressing growing tumors, but promoting dormant tumor cell survival and recurrence
- Author
-
Miller, TW, primary, Hampsch, RA, additional, McCleery, CF, additional, Wells, JD, additional, Fields, JL, additional, Dillon, LM, additional, and Shee, K, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Abstract PD4-08: A microenvironment secretome screen reveals FGF2 as a mediator of resistance to anti-estrogens and PI3K/mTOR pathway inhibitors in ER+ breast cancer
- Author
-
Shee, K, primary, Hinds, JW, additional, Yang, W, additional, Hampsch, RA, additional, Patel, K, additional, Varn, FS, additional, Cheng, C, additional, Jenkins, NP, additional, Kettenbach, AN, additional, Demidenko, E, additional, Owens, P, additional, Lanari, C, additional, Faber, AC, additional, Golub, TR, additional, Straussman, R, additional, and Miller, TW, additional
- Published
- 2018
- Full Text
- View/download PDF
4. Abstract P6-07-03: Broken promise of liquid biopsy: Plasma DNA does not accurately reflect tumor DNA in metastatic breast cancer
- Author
-
Shee, K, primary, Chamberlin, MD, additional, Varn, FS, additional, Bean, JR, additional, Marotti, JD, additional, Wells, WA, additional, Trask, HW, additional, Hamilton, JS, additional, West, RJ, additional, Kaufman, PA, additional, Schwartz, GN, additional, Gemery, JM, additional, McNulty, NJ, additional, Tsapakos, MJ, additional, Barth, RJ, additional, Arrick, BA, additional, Gui, J, additional, Cheng, C, additional, and Miller, TW, additional
- Published
- 2017
- Full Text
- View/download PDF
5. Abstract P1-07-04: Unique overlapping subtypes of triple-negative breast and ovarian cancers and sensitivity of “mesenchymal-like” cancers to HSP90 inhibition is revealed by integrated gene expression and drug sensitivity profiling
- Author
-
Shee, K, primary, Ung, MH, additional, Cheng, C, additional, and Miller, TW, additional
- Published
- 2017
- Full Text
- View/download PDF
6. Abstract P3-03-01: A novel high-throughput secreted factor screen and bioinformatics pipeline identifies microenvironment-derived FGF2 as a mechanism of resistance to anti-estrogens, PI3K, and mTOR inhibitors in ER+ breast cancer
- Author
-
Shee, K, primary, Hinds, JW, additional, Hampsch, RA, additional, Golub, TR, additional, Straussman, R, additional, and Miller, TW, additional
- Published
- 2017
- Full Text
- View/download PDF
7. Abstract P5-03-01: Therapeutic targeting of Rac GTPases in ER+ and HER2+ breast cancer
- Author
-
Hampsch, RA, primary, Shee, K, additional, and Miller, TW, additional
- Published
- 2016
- Full Text
- View/download PDF
8. P-REX1 creates a positive feedback loop to activate growth factor receptor, PI3K/AKT and MEK/ERK signaling in breast cancer
- Author
-
Dillon, L M, primary, Bean, J R, additional, Yang, W, additional, Shee, K, additional, Symonds, L K, additional, Balko, J M, additional, McDonald, W H, additional, Liu, S, additional, Gonzalez-Angulo, A M, additional, Mills, G B, additional, Arteaga, C L, additional, and Miller, T W, additional
- Published
- 2014
- Full Text
- View/download PDF
9. Preventive Pharmacologic Therapy for Kidney Stone Disease.
- Author
-
Sui W, Shee K, and Stoller M
- Subjects
- Humans, Kidney Calculi prevention & control, Kidney Calculi drug therapy
- Published
- 2024
- Full Text
- View/download PDF
10. Voided volume may not impact stone outcomes: Review of a large institutional nephrolithiasis cohort.
- Author
-
Shee K, Chan C, Yang H, Sui W, Bowman M, Hamouche F, Charondo LB, Ho S, Chi T, and Stoller ML
- Abstract
Background: Urologic guidelines universally recommend increasing fluid intake for kidney stone prevention. Increased voided volume is thought to help reduce stone recurrence and severity, but supporting evidence is limited., Patients and Methods: Nephrolithiasis outcomes and 24-h urine data for patients from the Registry for Stones of the Kidney and Ureter (ReSKU), a registry of nephrolithiasis patients collected between 2015 and 2020, were retrospectively analysed. Outcome was stone events, either an office visit where a patient reports symptomatic passage of stones or surgery for stone removal., Results: We identified 450 stone patients with 24-h urine and kidney stone outcome data. There was no significant difference in 24-h voided volume between patients with one stone event and patients with two or more stone events. On multivariable logistic regression, after controlling for age, gender, BMI, and 24-h sodium and creatinine per kilogram, no significant associations were found between voided volume and stone events. There was a statistically significant negative correlation noted between voided volume and stone events in calcium oxalate dihydrate stone formers (Spearman R = -0.42, p = 0.04), but not others., Conclusions: Twenty-four-hour voided volume was not associated with stone events in a large institutional cohort, and subset analysis reveals that some stone formers may benefit more from increased voided volume than others; identifying such patients represents a novel precision medicine opportunity., Competing Interests: All authors declare that they have no conflict of interest regarding the material presented in this manuscript. All authors declare that they approve submission of the final article., (© 2023 The Authors. BJUI Compass published by John Wiley & Sons Ltd on behalf of BJU International Company.)
- Published
- 2023
- Full Text
- View/download PDF
11. Addition of Enzalutamide to Leuprolide and Definitive Radiation Therapy Is Tolerable and Effective in High-Risk Localized or Regional Nonmetastatic Prostate Cancer: Results From a Phase 2 Trial.
- Author
-
Shee K, de la Calle CM, Chang AJ, Wong AC, Feng FY, Gottschalk AR, Carroll PR, and Nguyen HG
- Abstract
Background: Enzalutamide is an antiandrogen used to treat both metastatic and nonmetastatic prostate cancer. Here we present results from a phase 2 trial designed to determine the safety, tolerability, and efficacy of adding enzalutamide to standard androgen deprivation therapy with radiation therapy in high-risk localized or regional, nonmetastatic patients with prostate cancer., Methods and Materials: Enrollment criteria included at least 2 of the following: stage cT3a/b, prostate specific antigen (PSA) ≥20 ng/mL, Gleason grade 8 to 10, ≥33% core involvement on biopsy, or pelvic lymph node involvement on computed tomography or magnetic resonance imaging. Patients with metastatic disease were excluded. All patients received 24 months of leuprolide and enzalutamide, and 5 weeks of intensity modulated radiation therapy followed by a brachytherapy boost. Adverse events (AE), PSA, testosterone, and basic laboratory tests were then followed for up to 36 months. Primary outcomes were safety and tolerability and PSA complete response rate (PSA-CR, defined as PSA ≤0.3). Secondary outcomes included time to biochemical recurrence (BCR; nadir + 2 ng/mL)., Results: Sixteen patients were enrolled; 2 were ineligible and 3 withdrew before starting treatment. Median age at enrollment was 69.0 years (interquartile range [IQR] 11.5). Median treatment duration was 24.0 months (IQR 11.9). Median follow-up time was 35.5 months (IQR 11.2), and 9 of 11 (81.8%) patients completed the 36 months of follow-up. One of 11 (9%) patients had grade 4 AE (seizure), and no grade 5 AE were reported. Four of 11 (36.4%) patients had grade 3 AE, such as erectile dysfunction and hot flashes. All patients achieved PSA-CR, and median time to PSA-CR was 4.2 months (IQR 1.4). At 24 months follow-up, 0 of 11 (0%) patients had a biochemical recurrence. At 36 months, 1 of 9 (11.1%) patient had a biochemical recurrence. Of note, this patient did not complete the full 24 months of enzalutamide and leuprolide due to AEs., Conclusions: Enzalutamide in combination with standard androgen deprivation therapy and radiation therapy was well-tolerated and effective warranting further study in a randomized controlled trial., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
12. Phase Ib Study of the Oral Proteasome Inhibitor Ixazomib (MLN9708) and Fulvestrant in Advanced ER+ Breast Cancer Progressing on Fulvestrant.
- Author
-
Schwartz G, Shee K, Romo B, Marotti J, Kisselev A, Lewis L, and Miller T
- Subjects
- Antineoplastic Agents, Hormonal pharmacology, Antineoplastic Agents, Hormonal therapeutic use, Boron Compounds, Estradiol pharmacology, Estradiol therapeutic use, Female, Fulvestrant pharmacology, Fulvestrant therapeutic use, Glycine analogs & derivatives, Humans, Receptors, Estrogen, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Proteasome Inhibitors pharmacology, Proteasome Inhibitors therapeutic use
- Abstract
Lessons Learned: Fulvestrant is a selective estrogen receptor (ER)-downregulating antiestrogen that blocks ER transcriptional activity and is approved for ER-positive breast cancer. Fulvestrant also induces accumulation of insoluble ER and activates an unfolded protein response; proteasome inhibitors have been shown to enhance these effects in preclinical models., Background: Fulvestrant is a selective estrogen receptor (ER)-downregulating antiestrogen that blocks ER transcriptional activity and is approved for ER-positive (+) breast cancer. Fulvestrant also induces accumulation of insoluble ER and activates an unfolded protein response; proteasome inhibitors have been shown to enhance these effects in preclinical models., Methods: This is a single-center phase Ib study with a 3+3 design of fulvestrant and the proteasome inhibitor ixazomib (MLN9708) in patients with advanced ER+ breast cancer that was progressing on fulvestrant. A dose-escalation design allowed establishment of the ixazomib maximum tolerated dose (MTD). Secondary objectives included progression-free survival, pharmacokinetics, and tumor molecular analyses., Results: Among nine evaluable subjects, treatment was well-tolerated without dose-limiting toxicities The MTD of ixazomib was 4 mg in combination with fulvestrant. Plasma concentrations of the active form of ixazomib (MLN2238) in the 4-mg dose cohort had a median (range) maximal concentration (C
max ) of 155 (122-171) ng/mL, time of maximal concentration (Tmax ) of 1 (1-1.5) hour, terminal elimination half-life of 66.6 (57.3-102.6) hour after initial dose, and area under the curve (AUC) of 5,025 (4,160-5,345) ng*h/mL. One partial response was observed, and median progression-free survival was 51 days (range, 47-137)., Conclusion: This drug combination has a favorable safety profile and antitumor activity in patients with fulvestrant-resistant advanced ER+ breast cancer that justifies future testing., (© AlphaMed Press; the data published online to support this summary are the property of the authors.)- Published
- 2021
- Full Text
- View/download PDF
13. Changes in Peripheral and Local Tumor Immunity after Neoadjuvant Chemotherapy Reshape Clinical Outcomes in Patients with Breast Cancer.
- Author
-
Axelrod ML, Nixon MJ, Gonzalez-Ericsson PI, Bergman RE, Pilkinton MA, McDonnell WJ, Sanchez V, Opalenik SR, Loi S, Zhou J, Mackay S, Rexer BN, Abramson VG, Jansen VM, Mallal S, Donaldson J, Tolaney SM, Krop IE, Garrido-Castro AC, Marotti JD, Shee K, Miller TW, Sanders ME, Mayer IA, Salgado R, and Balko JM
- Subjects
- Adult, Aged, Albumins adverse effects, Antineoplastic Combined Chemotherapy Protocols, B7-H1 Antigen antagonists & inhibitors, CD8-Positive T-Lymphocytes drug effects, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Lymphocytes, Tumor-Infiltrating drug effects, Lymphocytes, Tumor-Infiltrating immunology, Middle Aged, Neoadjuvant Therapy adverse effects, Neoplasm Proteins genetics, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local immunology, Neoplasm Recurrence, Local pathology, Paclitaxel adverse effects, Prognosis, Programmed Cell Death 1 Receptor antagonists & inhibitors, Progression-Free Survival, Treatment Outcome, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms immunology, Triple Negative Breast Neoplasms pathology, Tumor Microenvironment drug effects, Albumins administration & dosage, B7-H1 Antigen genetics, Paclitaxel administration & dosage, Programmed Cell Death 1 Receptor genetics, Triple Negative Breast Neoplasms drug therapy
- Abstract
Purpose: The recent approval of anti-programmed death-ligand 1 immunotherapy in combination with nab-paclitaxel for metastatic triple-negative breast cancer (TNBC) highlights the need to understand the role of chemotherapy in modulating the tumor immune microenvironment (TIME)., Experimental Design: We examined immune-related gene expression patterns before and after neoadjuvant chemotherapy (NAC) in a series of 83 breast tumors, including 44 TNBCs, from patients with residual disease (RD). Changes in gene expression patterns in the TIME were tested for association with recurrence-free (RFS) and overall survival (OS). In addition, we sought to characterize the systemic effects of NAC through single-cell analysis (RNAseq and cytokine secretion) of programmed death-1-high (PD-1
HI ) CD8+ peripheral T cells and examination of a cytolytic gene signature in whole blood., Results: In non-TNBC, no change in expression of any single gene was associated with RFS or OS, while in TNBC upregulation of multiple immune-related genes and gene sets were associated with improved long-term outcome. High cytotoxic T-cell signatures present in the peripheral blood of patients with breast cancer at surgery were associated with persistent disease and recurrence, suggesting active antitumor immunity that may indicate ongoing disease burden., Conclusions: We have characterized the effects of NAC on the TIME, finding that TNBC is uniquely sensitive to the immunologic effects of NAC, and local increases in immune genes/sets are associated with improved outcomes. However, expression of cytotoxic genes in the peripheral blood, as opposed to the TIME, may be a minimally invasive biomarker of persistent micrometastatic disease ultimately leading to recurrence., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
14. AMPK Activation by Metformin Promotes Survival of Dormant ER + Breast Cancer Cells.
- Author
-
Hampsch RA, Wells JD, Traphagen NA, McCleery CF, Fields JL, Shee K, Dillon LM, Pooler DB, Lewis LD, Demidenko E, Huang YH, Marotti JD, Goen AE, Kinlaw WB, and Miller TW
- Subjects
- Animals, Aromatase Inhibitors therapeutic use, Breast Neoplasms pathology, Cell Line, Tumor, Cell Proliferation drug effects, Chemotherapy, Adjuvant methods, Estrogens biosynthesis, Female, Humans, Metformin therapeutic use, Mice, Neoadjuvant Therapy methods, Receptors, Estrogen metabolism, Signal Transduction drug effects, Xenograft Model Antitumor Assays, AMP-Activated Protein Kinases metabolism, Aromatase Inhibitors pharmacology, Breast Neoplasms therapy, Cell Survival drug effects, Metformin pharmacology
- Abstract
Purpose: Despite adjuvant endocrine therapy for patients with estrogen receptor alpha (ER)-positive breast cancer, dormant residual disease can persist for years and eventually cause tumor recurrence. We sought to deduce mechanisms underlying the persistence of dormant cancer cells to identify therapeutic strategies., Experimental Design: Mimicking the aromatase inhibitor-induced depletion of estrogen levels used to treat patients, we developed preclinical models of dormancy in ER
+ breast cancer induced by estrogen withdrawal in mice. We analyzed tumor xenografts and cultured cancer cells for molecular and cellular responses to estrogen withdrawal and drug treatments. Publicly available clinical breast tumor gene expression datasets were analyzed for responses to neoadjuvant endocrine therapy., Results: Dormant breast cancer cells exhibited upregulated 5' adenosine monophosphate-activated protein kinase (AMPK) levels and activity, and upregulated fatty acid oxidation. While the antidiabetes AMPK-activating drug metformin slowed the estrogen-driven growth of cells and tumors, metformin promoted the persistence of estrogen-deprived cells and tumors through increased mitochondrial respiration driven by fatty acid oxidation. Pharmacologic or genetic inhibition of AMPK or fatty acid oxidation promoted clearance of dormant residual disease, while dietary fat increased tumor cell survival., Conclusions: AMPK has context-dependent effects in cancer, cautioning against the widespread use of an AMPK activator across disease settings. The development of therapeutics targeting fat metabolism is warranted in ER+ breast cancer., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
15. A Transcriptionally Definable Subgroup of Triple-Negative Breast and Ovarian Cancer Samples Shows Sensitivity to HSP90 Inhibition.
- Author
-
Shee K, Wells JD, Ung M, Hampsch RA, Traphagen NA, Yang W, Liu SC, Zeldenrust MA, Wang L, Kalari KR, Yu J, Boughey JC, Demidenko E, Kettenbach AN, Cheng C, Goetz MP, and Miller TW
- Subjects
- Animals, Apoptosis, Breast Neoplasms classification, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Female, Humans, Mice, Mice, Inbred NOD, Triple Negative Breast Neoplasms classification, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Biomarkers, Tumor genetics, Breast Neoplasms drug therapy, Gene Expression Profiling methods, Gene Expression Regulation, Neoplastic, HSP90 Heat-Shock Proteins antagonists & inhibitors, Triple Negative Breast Neoplasms drug therapy
- Abstract
Purpose: We hypothesized that integrated analysis of cancer types from different lineages would reveal novel molecularly defined subgroups with unique therapeutic vulnerabilities. On the basis of the molecular similarities between subgroups of breast and ovarian cancers, we analyzed these cancers as a single cohort to test our hypothesis., Experimental Design: Identification of transcriptional subgroups of cancers and drug sensitivity analyses were performed using mined data. Cell line sensitivity to Hsp90 inhibitors (Hsp90i) was tested in vitro . The ability of a transcriptional signature to predict Hsp90i sensitivity was validated using cell lines, and cell line- and patient-derived xenograft (PDX) models. Mechanisms of Hsp90i sensitivity were uncovered using immunoblot and RNAi., Results: Transcriptomic analyses of breast and ovarian cancer cell lines uncovered two mixed subgroups comprised primarily of triple-negative breast and multiple ovarian cancer subtypes. Drug sensitivity analyses revealed that cells of one mixed subgroup are significantly more sensitive to Hsp90i compared with cells from all other cancer lineages evaluated. A gene expression classifier was generated that predicted Hsp90i sensitivity in vitro , and in cell line- and PDXs. Cells from the Hsp90i-sensitive subgroup underwent apoptosis mediated by Hsp90i-induced upregulation of the proapoptotic proteins Bim and PUMA., Conclusions: Our findings identify Hsp90i as a potential therapeutic strategy for a transcriptionally defined subgroup of ovarian and breast cancers. This study demonstrates that gene expression profiles may be useful to identify therapeutic vulnerabilities in tumor types with limited targetable genetic alterations, and to identify molecularly definable cancer subgroups that transcend lineage., (©2019 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
16. Integrated pan-cancer gene expression and drug sensitivity analysis reveals SLFN11 mRNA as a solid tumor biomarker predictive of sensitivity to DNA-damaging chemotherapy.
- Author
-
Shee K, Wells JD, Jiang A, and Miller TW
- Subjects
- Antineoplastic Agents therapeutic use, Biomarkers, Tumor metabolism, Cell Line, Tumor, DNA Damage drug effects, Datasets as Topic, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Neoplasms genetics, Neoplasms mortality, Precision Medicine methods, Predictive Value of Tests, Prognosis, Progression-Free Survival, RNA, Messenger metabolism, Survival Analysis, Time Factors, Antineoplastic Agents pharmacology, Biomarkers, Tumor genetics, Drug Resistance, Neoplasm genetics, Neoplasms drug therapy, Nuclear Proteins genetics
- Abstract
Background: Precision oncology seeks to integrate multiple layers of data from a patient's cancer to effectively tailor therapy. Conventional chemotherapies are sometimes effective but accompanied by adverse events, warranting the identification of a biomarker of chemosensitivity., Objective: Identify an mRNA biomarker that predicts chemosensitivity across solid tumor subtypes., Methods: We performed a pan-solid tumor analysis integrating gene expression and drug sensitivity profiles from 3 cancer cell line datasets to identify transcripts correlated with sensitivity to a panel of chemotherapeutics. We then tested the ability of an mRNA biomarker to predictive clinical outcomes in cohorts of patients with breast, lung, or ovarian cancer., Results: Expression levels of several mRNA transcripts were significantly correlated with sensitivity or resistance chemotherapeutics in cancer cell line datasets. The only mRNA transcript significantly correlated with sensitization to multiple classes of DNA-damaging chemotherapeutics in all 3 cell line datasets was encoded by Schlafen Family Member 11 (SLFN11). Analyses of multiple breast, lung, and ovarian cancer patient cohorts treated with chemotherapy confirmed SLFN11 mRNA expression as a predictive biomarker of longer overall survival and improved tumor response., Conclusions: Tumor SLFN11 mRNA expression is a biomarker of sensitivity to an array of DNA-damaging chemotherapeutics across solid tumor subtypes., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
17. Estrogen therapy induces an unfolded protein response to drive cell death in ER+ breast cancer.
- Author
-
Hosford SR, Shee K, Wells JD, Traphagen NA, Fields JL, Hampsch RA, Kettenbach AN, Demidenko E, and Miller TW
- Subjects
- Animals, Breast Neoplasms enzymology, Breast Neoplasms genetics, Cell Death drug effects, Cell Nucleus drug effects, Cell Nucleus metabolism, Drug Resistance, Neoplasm drug effects, Estrogen Receptor Modulators pharmacology, Estrogen Receptor Modulators therapeutic use, Estrogens pharmacology, Female, Fulvestrant pharmacology, Fulvestrant therapeutic use, Humans, JNK Mitogen-Activated Protein Kinases metabolism, MCF-7 Cells, Mice, Signal Transduction drug effects, Time Factors, Transcriptional Activation drug effects, Transcriptional Activation genetics, Transcriptome genetics, Tumor Suppressor Protein p53 metabolism, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Estrogens therapeutic use, Receptors, Estrogen metabolism, Unfolded Protein Response drug effects
- Abstract
Estrogens have been shown to elicit anticancer effects against estrogen receptor α (ER)-positive breast cancer. We sought to determine the mechanism underlying the therapeutic response. Response to 17β-estradiol was assessed in ER+ breast cancer models with resistance to estrogen deprivation: WHIM16 patient-derived xenografts, C7-2-HI and C4-HI murine mammary adenocarcinomas, and long-term estrogen-deprived MCF-7 cells. As another means to reactivate ER, the anti-estrogen fulvestrant was withdrawn from fulvestrant-resistant MCF-7 cells. Transcriptional, growth, apoptosis, and molecular alterations in response to ER reactivation were measured. 17β-estradiol treatment and fulvestrant withdrawal induced transcriptional activation of ER, and cells adapted to estrogen deprivation or fulvestrant were hypersensitive to 17β-estradiol. ER transcriptional response was followed by an unfolded protein response and apoptosis. Such apoptosis was dependent upon the unfolded protein response, p53, and JNK signaling. Anticancer effects were most pronounced in models exhibiting genomic amplification of the gene encoding ER (ESR1), suggesting that engagement of ER at high levels is cytotoxic. These data indicate that long-term adaptation to estrogen deprivation or ER inhibition alters sensitivity to ER reactivation. In such adapted cells, 17β-estradiol treatment and anti-estrogen withdrawal hyperactivate ER, which drives an unfolded protein response and subsequent growth inhibition and apoptosis. 17β-estradiol treatment should be considered as a therapeutic option for anti-estrogen-resistant disease, particularly in patients with tumors harboring ESR1 amplification or ER overexpression. Furthermore, therapeutic strategies that enhance an unfolded protein response may increase the therapeutic effects of ER reactivation., (© 2019 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
18. Ductal Carcinoma in Situ Biomarkers in a Precision Medicine Era: Current and Future Molecular-Based Testing.
- Author
-
Shee K, Muller KE, Marotti J, Miller TW, Wells WA, and Tsongalis GJ
- Subjects
- Breast Neoplasms genetics, Carcinoma, Intraductal, Noninfiltrating genetics, Disease Progression, Female, Humans, Neoplasm Invasiveness, Prognosis, Biomarkers, Tumor genetics, Breast Neoplasms diagnosis, Carcinoma, Intraductal, Noninfiltrating diagnosis, Precision Medicine
- Abstract
Historically, ductal carcinoma in situ (DCIS) of the breast has been managed aggressively with surgery and radiotherapy because of a risk of progression to invasive ductal carcinoma. However, this treatment paradigm has been challenged by overtreatment concerns and evidence that suggests that DCIS can be stratified according to risk of recurrence or risk of progression to invasive disease. Traditional methods of risk stratification include histologic grade and hormone receptor status. Recent technological advancements have enabled an era of precision medicine, where DCIS can be molecularly analyzed by tools, such as next-generation DNA and RNA sequencing, to identify molecular biomarkers for risk stratification. These findings have led to the development of tools such as the Oncotype DX Breast DCIS Score, a gene expression-based assay with the potential to prevent overtreatment in low-risk disease., (Copyright © 2019 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
19. The curcumin analog HO-3867 selectively kills cancer cells by converting mutant p53 protein to transcriptionally active wildtype p53.
- Author
-
Madan E, Parker TM, Bauer MR, Dhiman A, Pelham CJ, Nagane M, Kuppusamy ML, Holmes M, Holmes TR, Shaik K, Shee K, Kiparoidze S, Smith SD, Park YA, Gomm JJ, Jones LJ, Tomás AR, Cunha AC, Selvendiran K, Hansen LA, Fersht AR, Hideg K, Gogna R, and Kuppusamy P
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Proliferation drug effects, Curcumin pharmacology, Female, Humans, Mice, Mice, Nude, Mutant Proteins metabolism, Neoplasms drug therapy, Neoplasms genetics, Tumor Cells, Cultured, Tumor Suppressor Protein p53 metabolism, Xenograft Model Antitumor Assays, Apoptosis drug effects, Curcumin analogs & derivatives, Mutant Proteins genetics, Mutation, Neoplasms pathology, Piperidones pharmacology, Tumor Suppressor Protein p53 genetics
- Abstract
p53 is an important tumor-suppressor protein that is mutated in more than 50% of cancers. Strategies for restoring normal p53 function are complicated by the oncogenic properties of mutant p53 and have not met with clinical success. To counteract mutant p53 activity, a variety of drugs with the potential to reconvert mutant p53 to an active wildtype form have been developed. However, these drugs are associated with various negative effects such as cellular toxicity, nonspecific binding to other proteins, and inability to induce a wildtype p53 response in cancer tissue. Here, we report on the effects of a curcumin analog, HO-3867, on p53 activity in cancer cells from different origins. We found that HO-3867 covalently binds to mutant p53, initiates a wildtype p53-like anticancer genetic response, is exclusively cytotoxic toward cancer cells, and exhibits high anticancer efficacy in tumor models. In conclusion, HO-3867 is a p53 mutant-reactivating drug with high clinical anticancer potential., (© 2018 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
20. Therapeutically targeting tumor microenvironment-mediated drug resistance in estrogen receptor-positive breast cancer.
- Author
-
Shee K, Yang W, Hinds JW, Hampsch RA, Varn FS, Traphagen NA, Patel K, Cheng C, Jenkins NP, Kettenbach AN, Demidenko E, Owens P, Faber AC, Golub TR, Straussman R, and Miller TW
- Subjects
- Animals, Apoptosis drug effects, Bcl-2-Like Protein 11 metabolism, Breast Neoplasms metabolism, Cell Cycle Checkpoints drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cyclin D1 metabolism, Cytokines metabolism, Down-Regulation drug effects, Female, Fibroblast Growth Factor 2 pharmacology, Humans, Ligands, Mechanistic Target of Rapamycin Complex 1 antagonists & inhibitors, Mechanistic Target of Rapamycin Complex 1 metabolism, Mice, Models, Biological, Neoplasm Recurrence, Local pathology, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Receptors, Fibroblast Growth Factor metabolism, Signal Transduction, Transcriptome genetics, Treatment Outcome, Up-Regulation drug effects, Breast Neoplasms pathology, Drug Resistance, Neoplasm drug effects, Molecular Targeted Therapy, Receptors, Estrogen metabolism, Tumor Microenvironment drug effects
- Abstract
Drug resistance to approved systemic therapies in estrogen receptor-positive (ER+) breast cancer remains common. We hypothesized that factors present in the human tumor microenvironment (TME) drive drug resistance. Screening of a library of recombinant secreted microenvironmental proteins revealed fibroblast growth factor 2 (FGF2) as a potent mediator of resistance to anti-estrogens, mTORC1 inhibition, and phosphatidylinositol 3-kinase inhibition in ER+ breast cancer. Phosphoproteomic analyses identified ERK1/2 as a major output of FGF2 signaling via FGF receptors (FGFRs), with consequent up-regulation of Cyclin D1 and down-regulation of Bim as mediators of drug resistance. FGF2-driven drug resistance in anti-estrogen-sensitive and -resistant models, including patient-derived xenografts, was reverted by neutralizing FGF2 or FGFRs. A transcriptomic signature of FGF2 signaling in primary tumors predicted shorter recurrence-free survival independently of age, grade, stage, and FGFR amplification status. These findings delineate FGF2 signaling as a ligand-based drug resistance mechanism and highlights an underdeveloped aspect of precision oncology: characterizing and treating patients according to their TME constitution., (© 2018 Shee et al.)
- Published
- 2018
- Full Text
- View/download PDF
21. Trailblazing Precision Oncology for Rare Tumor Subtypes.
- Author
-
Shee K and Miller TW
- Subjects
- Ambulatory Care Facilities, Humans, Medical Oncology, Molecular Targeted Therapy, Precision Medicine, Neoplasms
- Abstract
Competing Interests: Disclosures of potential conflicts of interest may be found at the end of this article.
- Published
- 2018
- Full Text
- View/download PDF
22. Therapeutic sensitivity to Rac GTPase inhibition requires consequential suppression of mTORC1, AKT, and MEK signaling in breast cancer.
- Author
-
Hampsch RA, Shee K, Bates D, Lewis LD, Désiré L, Leblond B, Demidenko E, Stefan K, Huang YH, and Miller TW
- Subjects
- Animals, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm physiology, Female, Humans, MAP Kinase Kinase Kinases metabolism, Mechanistic Target of Rapamycin Complex 1, Mice, Mice, Inbred NOD, Mice, SCID, Multiprotein Complexes metabolism, Proto-Oncogene Proteins c-akt metabolism, TOR Serine-Threonine Kinases metabolism, Xenograft Model Antitumor Assays, Breast Neoplasms metabolism, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Signal Transduction drug effects
- Abstract
Rac GTPases have oncogenic roles in cell growth, survival, and migration. We tested response to the Rac inhibitor EHT1864 in a panel of breast cancer cell lines. EHT1864-induced growth inhibition was associated with dual inhibition of the PI3K/AKT/mTORC1 and MEK/ERK pathways. Breast cancer cells harboring PIK3CA mutations or HER2 overexpression were most sensitive to Rac inhibition, suggesting that such oncogenic alterations link Rac activation with PI3K/AKT/mTORC1 and MEK/ERK signaling. Interestingly, EHT1864 decreased activation of the mTORC1 substrate p70S6K earlier than AKT inhibition, suggesting that Rac may activate mTORC1/p70S6K independently of AKT. Comparison of the growth-inhibitory profile of EHT1864 to 137 other anti-cancer drugs across 656 cancer cell lines revealed significant correlation with the p70S6K inhibitor PF-4708671. We confirmed that Rac complexes contain MEK1/2 and ERK1/2, but also contain p70S6K; these interactions were disrupted by EHT1864. Pharmacokinetic profiles revealed that EHT1864 was present in mouse plasma at concentrations effective in vitro for approximately 1 h after intraperitoneal administration. EHT1864 suppressed growth of HER2+ tumors, and enhanced response to anti-estrogen treatment in ER+ tumors. Further therapeutic development of Rac inhibitors for HER2+ and PIK3CA-mutant cancers is warranted.
- Published
- 2017
- Full Text
- View/download PDF
23. Maximizing the Benefit-Cost Ratio of Anthracyclines in Metastatic Breast Cancer: Case Report of a Patient with a Complete Response to High-Dose Doxorubicin.
- Author
-
Shee K, Kono AT, D'Anna SP, Seltzer MA, Lu X, Miller TW, and Chamberlin MD
- Abstract
Despite the clinical efficacy of anthracycline agents such as doxorubicin, dose-limiting cardiac toxicities significantly limit their long-term use. Here, we present the case of a 33-year-old female patient with extensive metastatic ER+/PR+/HER2- mucinous adenocarcinoma of the breast, who was started on doxorubicin/cyclophosphamide therapy after progressing on paclitaxel and ovarian suppressor goserelin with aromatase inhibitor exemestane. The patient was comanaged by cardiology, who carefully monitored measures of cardiac function, including EKGs, serial echocardiograms, and profiling of lipids, troponin, and pro-BNP every 2 months. The patient was treated with the cardioprotective agent dexrazoxane, and changes in cardiac markers [e.g. decreases in ejection fraction (EF)] were immediately addressed by therapeutic intervention with the ACE inhibitor lisinopril and beta-blocker metoprolol. The patient had a complete response to doxorubicin therapy, with a cumulative dose of 1,350 mg/m
2 , which is significantly above the recommended limits, and to our knowledge, the highest dose reported in literature. Two and a half years after the last doxorubicin cycle, the patient is asymptomatic with no cardiotoxicity and an excellent quality of life. This case highlights the importance of careful monitoring and management of doxorubicin-mediated cardiotoxicity, and that higher cumulative doses of anthracyclines can be considered in patients with ongoing clinical benefit., Competing Interests: The authors have no conflicts of interest to disclose.- Published
- 2016
- Full Text
- View/download PDF
24. Strategically Timing Inhibition of Phosphatidylinositol 3-Kinase to Maximize Therapeutic Index in Estrogen Receptor Alpha-Positive, PIK3CA-Mutant Breast Cancer.
- Author
-
Yang W, Hosford SR, Dillon LM, Shee K, Liu SC, Bean JR, Salphati L, Pang J, Zhang X, Nannini MA, Demidenko E, Bates D, Lewis LD, Marotti JD, Eastman AR, and Miller TW
- Subjects
- Animals, Apoptosis drug effects, Breast Neoplasms metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Drug Resistance, Neoplasm drug effects, Estradiol analogs & derivatives, Estradiol pharmacology, Estrogen Receptor Modulators pharmacology, Estrogen Receptor alpha metabolism, Female, Fulvestrant, Humans, Indazoles pharmacology, MCF-7 Cells, Mice, Mice, Inbred NOD, Mutation drug effects, Signal Transduction drug effects, Sulfonamides pharmacology, Therapeutic Index, Breast Neoplasms drug therapy, Class I Phosphatidylinositol 3-Kinases antagonists & inhibitors, Class I Phosphatidylinositol 3-Kinases genetics, Estrogen Receptor alpha genetics, Mutation genetics, Phosphoinositide-3 Kinase Inhibitors, Protein Kinase Inhibitors pharmacology
- Abstract
Purpose: Phosphatidylinositol 3-kinase (PI3K) inhibitors are being developed for the treatment of estrogen receptor α (ER)-positive breast cancer in combination with antiestrogens. Understanding the temporal response and pharmacodynamic effects of PI3K inhibition in ER(+) breast cancer will provide a rationale for treatment scheduling to maximize therapeutic index., Experimental Design: Antiestrogen-sensitive and antiestrogen-resistant ER(+) human breast cancer cell lines and mice bearing PIK3CA-mutant xenografts were treated with the antiestrogen fulvestrant, the PI3K inhibitor GDC-0941 (pictilisib; varied doses/schedules that provided similar amounts of drug each week), or combinations. Cell viability, signaling pathway inhibition, proliferation, apoptosis, tumor volume, and GDC-0941 concentrations in plasma and tumors were temporally measured., Results: Treatment with the combination of fulvestrant and GDC-0941, regardless of dose/schedule, was significantly more effective than that with single-agent treatments in fulvestrant-resistant tumors. Short-term, complete PI3K inhibition blocked cell growth in vitro more effectively than chronic, incomplete inhibition. Longer-term PI3K inhibition hypersensitized cells to growth factor signaling upon drug withdrawal. Different schedules of GDC-0941 elicited similar tumor responses. While weekly high-dose GDC-0941 with fulvestrant continuously suppressed PI3K signaling for 72 hours, inducing a bolus of apoptosis and inhibiting proliferation, PI3K reactivation upon GDC-0941 washout induced a proliferative burst. Fulvestrant with daily low-dose GDC-0941 metronomically suppressed PI3K for 6 to 9 hours/day, repeatedly inducing small amounts of apoptosis and temporarily inhibiting proliferation, followed by proliferative rebound compared with fulvestrant alone., Conclusions: Continuous and metronomic PI3K inhibition elicits robust anticancer effects in ER(+), PIK3CA-mutant breast cancer. Clinical exploration of alternate treatment schedules of PI3K inhibitors with antiestrogens is warranted. Clin Cancer Res; 22(9); 2250-60. ©2016 AACRSee related commentary by Toska and Baselga, p. 2099., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
25. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion.
- Author
-
Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR, Du J, Davis A, Mongare MM, Gould J, Frederick DT, Cooper ZA, Chapman PB, Solit DB, Ribas A, Lo RS, Flaherty KT, Ogino S, Wargo JA, and Golub TR
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols, Cell Line, Tumor, Coculture Techniques, Humans, Indoles pharmacology, Indoles therapeutic use, Melanoma drug therapy, Melanoma genetics, Melanoma pathology, Molecular Targeted Therapy, Mutation, Phosphatidylinositol 3-Kinases metabolism, Prognosis, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Proteomics, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins c-met antagonists & inhibitors, Proto-Oncogene Proteins c-met metabolism, Signal Transduction drug effects, Stromal Cells cytology, Stromal Cells drug effects, Stromal Cells metabolism, Sulfonamides pharmacology, Sulfonamides therapeutic use, Vemurafenib, Drug Resistance, Neoplasm drug effects, Hepatocyte Growth Factor metabolism, Melanoma metabolism, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Tumor Microenvironment physiology
- Abstract
Drug resistance presents a challenge to the treatment of cancer patients. Many studies have focused on cell-autonomous mechanisms of drug resistance. By contrast, we proposed that the tumour micro-environment confers innate resistance to therapy. Here we developed a co-culture system to systematically assay the ability of 23 stromal cell types to influence the innate resistance of 45 cancer cell lines to 35 anticancer drugs. We found that stroma-mediated resistance is common, particularly to targeted agents. We characterized further the stroma-mediated resistance of BRAF-mutant melanoma to RAF inhibitors because most patients with this type of cancer show some degree of innate resistance. Proteomic analysis showed that stromal cell secretion of hepatocyte growth factor (HGF) resulted in activation of the HGF receptor MET, reactivation of the mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-OH kinase (PI(3)K)-AKT signalling pathways, and immediate resistance to RAF inhibition. Immunohistochemistry experiments confirmed stromal cell expression of HGF in patients with BRAF-mutant melanoma and showed a significant correlation between HGF expression by stromal cells and innate resistance to RAF inhibitor treatment. Dual inhibition of RAF and either HGF or MET resulted in reversal of drug resistance, suggesting RAF plus HGF or MET inhibitory combination therapy as a potential therapeutic strategy for BRAF-mutant melanoma. A similar resistance mechanism was uncovered in a subset of BRAF-mutant colorectal and glioblastoma cell lines. More generally, this study indicates that the systematic dissection of interactions between tumours and their micro-environment can uncover important mechanisms underlying drug resistance.
- Published
- 2012
- Full Text
- View/download PDF
26. Research, collaboration, and open science using web 2.0.
- Author
-
Shee K, Strong M, Guido NJ, Lue RA, Church GM, and Viel A
- Abstract
There is little doubt that the Internet has transformed the world in which we live. Information that was once archived in bricks and mortar libraries is now only a click away, and people across the globe have become connected in a manner inconceivable only 20 years ago. Although many scientists and educators have embraced the Internet as an invaluable tool for research, education and data sharing, some have been somewhat slower to take full advantage of emerging Web 2.0 technologies. Here we discuss the benefits and challenges of integrating Web 2.0 applications into undergraduate research and education programs, based on our experience utilizing these technologies in a summer undergraduate research program in synthetic biology at Harvard University. We discuss the use of applications including wiki-based documentation, digital brainstorming, and open data sharing via the Web, to facilitate the educational aspects and collaborative progress of undergraduate research projects. We hope to inspire others to integrate these technologies into their own coursework or research projects.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.