Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Curr Pharm Teach Learn ; 16(7): 102106, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38744564

ABSTRACT

INTRODUCTION: Podcasts are a popular way to learn and engage at the convenience of the listener. Education is incorporating podcasts to supplement and reinforce students learning inside and outside the classroom. METHODS: The authors created a podcast covering the Top 200 commonly prescribed medications. This was to help students recall and reinforce medication knowledge they typically must learn on their own. RESULTS: Student performance on post-tests improved (p = 0.0011) compared to pre-tests with an effect size r of 0.39 (0.37, 0.32, and 0.42 for P1, P2 and P3 respectively). Students reported the content was easy to follow, and they enjoyed learning from other students. The total number of plays for the podcast as of 19 July 2023 were 882. Each episode had a range of one to 89 number of plays. DISCUSSION: The podcast was well received by students, and drug knowledge increased. While the podcasts were shorter in time, they still provided the foundational information for a first-year pharmacy student to know. Overall, podcasts provide another way to help students retain and reinforce material learned inside and outside the classroom.


Subject(s)
Education, Pharmacy , Educational Measurement , Students, Pharmacy , Webcasts as Topic , Humans , Webcasts as Topic/statistics & numerical data , Education, Pharmacy/methods , Education, Pharmacy/standards , Education, Pharmacy/statistics & numerical data , Students, Pharmacy/statistics & numerical data , Students, Pharmacy/psychology , Educational Measurement/methods , Educational Measurement/statistics & numerical data , Learning , Curriculum/trends , Curriculum/standards
2.
Curr Pharm Teach Learn ; 13(8): 935-944, 2021 08.
Article in English | MEDLINE | ID: mdl-34294257

ABSTRACT

INTRODUCTION: In fall 2017, West Coast University School of Pharmacy implemented ExamSoft for testing. Three courses in each didactic year employed ExamSoft. Prior to this, courses had Scantron-based exams. We surveyed the students to assess their perception of ExamSoft. We hypothesized that students' inherent bias towards technology affected their perception of ExamSoft. METHODS: To assess this hypothesis, we conducted a survey of all students. The survey contained questions about comfort with technology and nine questions on students' perceptions of ExamSoft and its usefulness. RESULTS: The survey responses were stratified according to the preference of respondents towards technology and its use in exams. Respondents were stratified into three groups: tech-embracers, tech-skeptics, and neutral. Our results showed that respondents classified as tech-skeptics tended to have a more negative view of ExamSoft and its perceived impact on their grades than students stratified as tech-embracers or neutral. CONCLUSIONS: Our study suggests that students' inherent bias towards technology plays an important role in their perception of computer-based testing. Assessing incoming students' comfort with technology and student orientation activities to help acquaint students with new technology could help improve their acceptance of educational technology used for testing.


Subject(s)
Computers , Students , Educational Technology , Humans , Perception , Surveys and Questionnaires
3.
J Steroid Biochem Mol Biol ; 202: 105697, 2020 09.
Article in English | MEDLINE | ID: mdl-32461092

ABSTRACT

Treatment of hormone sensitive breast cancer tumors with endocrine therapy such as antiestrogens or aromatase inhibitors has improved the outcome significantly. Studies including our own have shown that downregulation of ERα with pure antiestrogen fulvestrant in combination with aromatase inhibitors may prolong responsiveness of the tumors to endocrine therapy. Fulvestrant has been studied as second line or first line treatment for post-menopausal hormone receptor positive breast cancers as a single agent or in combination with AIs. Studies have also suggested that further escalation of dose may improve benefit. However, dose escalation of fulvestrant, which is administered via intramuscular injection, is difficult due to its poor solubility. To overcome this shortcoming of an injectable drug, a novel orally active antiestrogen, AZD9496 was developed. In addition to being orally active, AZD9496 is designed as a selective ERα downregulator (SERD). In the current study, we compared the effect of AZD9496 and fulvestrant on the growth of MCF-7Ca (human estrogen receptor positive MCF-7 cells stably transfected with human placental aromatase gene) xenografts grown in ovariectomized athymic nude mice. AZD9496 was also compared to fulvestrant in vitro as a single agent or in combination with anastrozole. Our current study shows that AZD9496 is equally effective as fulvestrant at controlling the growth of hormone sensitive human breast cancer tumors. Similar to fulvestrant, AZD9496 inhibits cellular aromatase activity through ERα mediated signaling. However, unlike fulvestrant, combination of AZD9496 with anastrozole did not produce increased tumor inhibition. Our results show that AZD9496 was significantly better at inhibiting cellular aromatase which contributed to its anticancer activity. Next, we measured the effect of AZD9496 on the mouse uterus. Uterine weight of mice treated with AZD9496 was significantly lower than that for mice treated with androstenedione. This reduction in uterine weight was due to AZD9496 mediated inhibition of aromatase activity and not a direct effect on uterine ERα expression. We also observed that anti-cancer efficacy of AZD9496 depended on its ability to inhibit cellular aromatase. These results suggest that AZD9496 may be a better alternative to fulvestrant due to its selectivity for mammary ER and ability to inhibit aromatase in addition of downregulating ERα that can be obtained upon oral administration. As such, AZD9496 may prove to be a better option than fulvestrant for the treatment of hormone sensitive human breast cancer.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Cinnamates/therapeutic use , Indoles/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Selective Estrogen Receptor Modulators/therapeutic use , Anastrozole/pharmacology , Anastrozole/therapeutic use , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Aromatase/metabolism , Aromatase Inhibitors/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cinnamates/pharmacology , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor Antagonists/therapeutic use , Estrogen Receptor alpha , Female , Fulvestrant/pharmacology , Fulvestrant/therapeutic use , Humans , Indoles/pharmacology , Mammary Neoplasms, Experimental/metabolism , Mice, Nude , Postmenopause , Selective Estrogen Receptor Modulators/pharmacology
4.
Adv Physiol Educ ; 41(3): 395-404, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28679578

ABSTRACT

This paper describes the development, implementation, and students' perceptions of a new trilayer approach of teaching (TLAT). The TLAT model involved blending lecture, in-class group activities, and out-of-class assignments on selected content areas and was implemented initially in a first-year integrated pharmacy course. Course contents were either delivered by traditional lectures or by the TLAT. A survey instrument was distributed by SurveyMonkey to determine students' perceptions of the TLAT model. Descriptive statistics were used for data analysis. Students' performance in a total of 225 examination and quiz questions was analyzed to evaluate whether the TLAT model improved students' learning. Students' (n = 98) performance scores for TLAT-based and lecture-based questions were 83.3 ± 10.2 and 79.5 ± 14.0, respectively (P < 0.05). Ninety-three percent of students believed that in-class group activities enhanced conceptual understanding of course materials, helped them take responsibility of their own learning, and enhanced their overall learning experiences. More than 80% of respondents felt that solving cases and developing concept maps helped them sharpen creative and critical thinking skills. In addition, 90% of the respondents indicated that the homework throughout the semester helped them stay up to date and focused with the progress of the course. The use of the TLAT model led to an improvement in student learning of complex concepts. Moreover, the results suggest that this model improves students' self-reliance and attitudes toward learning. Our findings should serve as an impetus for inclusion of diverse active learning strategies in pharmacy education.


Subject(s)
Education, Pharmacy/methods , Education, Pharmacy/standards , Pharmacology/education , Physiology/education , Teaching/standards , Curriculum/standards , Curriculum/trends , Educational Measurement , Humans , Learning , Teaching/trends
5.
Sci Rep ; 6: 30721, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27477453

ABSTRACT

Estrogen deprivation has a profound effect on the female brain. One of the most obvious examples of this condition is hot flushes. Although estrogens relieve these typical climacteric symptoms, many women do not want to take them owing to unwanted side-effects impacting, for example, the uterus, breast and blood. Therefore, there is a need for developing safer estrogen therapies. We show here that treatment with 10ß,17ß-dihydroxyestra-1,4-dien-3-one (DHED), a novel brain-targeting bioprecursor prodrug of the main human estrogen, 17ß-estradiol, alleviates hot flushes in rat models of thermoregulatory dysfunction of the brain. Oral administration of DHED elicits a significant reduction of tail skin temperature (TST) rise representing hot flushes in the morphine-dependent ovariectomized rat model and results in the restoration of estrogen deprivation-induced loss of diurnal rhythm in TST. These beneficial effects occur without detrimental peripheral hormonal exposure; thus, the treatment avoids potentially harmful stimulation of estrogen-sensitive peripheral organs, including the uterus and the anterior pituitary, or the proliferation of MCF-7a breast cancer cell xenografts. Our promising preclinical assessments warrant further considerations of DHED for the development of a brain-selective 17ß-estradiol therapy to relieve hot flushes without undesirable peripheral side-effects.


Subject(s)
Estradiol/administration & dosage , Estrogens/administration & dosage , Hot Flashes/drug therapy , Prodrugs/administration & dosage , Administration, Oral , Animals , Rats , Treatment Outcome
6.
Mol Cancer Ther ; 14(11): 2642-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26310543

ABSTRACT

Heregulin-driven ERBB3 signaling has been implicated as a mechanism of resistance to cytotoxic and antiendocrine therapies in preclinical breast cancer models. In this study, we evaluated the effects of seribantumab (MM-121), a heregulin-blocking anti-ERBB3 monoclonal antibody, alone and in combination with the aromatase inhibitor letrozole, on cell signaling and tumor growth in a preclinical model of postmenopausal estrogen receptor-positive (ER(+)) breast cancer. In vitro, heregulin treatment induced estrogen receptor phosphorylation in MCF-7Ca cells, and long-term letrozole-treated (LTLT-Ca) cells had increased expression and activation levels of EGFR, HER2, and ERBB3. Treatment with seribantumab, but not letrozole, inhibited basal and heregulin-mediated ERBB receptor phosphorylation and downstream effector activation in letrozole-sensitive (MCF-7Ca) and -refractory (LTLT-Ca) cells. Notably, in MCF-7Ca-derived xenograft tumors, cotreatment with seribantumab and letrozole had increased antitumor activity compared with letrozole alone, which was accompanied by downregulated PI3K/MTOR signaling both prior to and after the development of resistance to letrozole. Moreover, the addition of an MTOR inhibitor to this treatment regimen did not improve antitumor activity and was not well tolerated. Our results demonstrate that heregulin-driven ERBB3 signaling mediates resistance to letrozole in a preclinical model of ER(+) breast cancer, suggesting that heregulin-expressing ER(+) breast cancer patients may benefit from the addition of seribantumab to antiendocrine therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Nitriles/pharmacology , Receptor, ErbB-3/antagonists & inhibitors , Triazoles/pharmacology , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Immunoblotting , Letrozole , Mice, Inbred BALB C , Mice, Nude , Neuregulin-1/pharmacology , Ovariectomy , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Receptor, ErbB-3/immunology , Receptor, ErbB-3/metabolism , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
7.
Breast Cancer Res Treat ; 152(3): 499-508, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26133921

ABSTRACT

Resistance to aromatase inhibitors (AIs) involves increased HER2. One mechanism by which HER2 may mediate resistance is through expansion of the tumor initiating cell (TIC) population. This study investigates whether combining all-trans retinoic acid (ATRA) and histone deacetylase inhibitor entinostat (ENT) can inhibit TICs and HER2 in AI-resistant cells and tumors. Modulation of cell viability and HER2 expression were assessed in AI-resistant cells treated with ATRA + ENT. Letrozole-resistant LTLT-Ca cells treated with ATRA + ENT were assayed for changes in TIC characteristics, such as TIC markers (BCRP, ALDH, and BMI-1), side population (SP), and mammosphere formation. Xenograft tumors of MCF-7Ca cells made resistant to letrozole were treated with ATRA, ATRA + letrozole, ATRA + ENT, or ATRA + ENT + letrozole. Resulting tumors were assayed for changes in TIC characteristics. Patient samples taken pre- and post-AI treatment were analyzed for changes in ERα and HER2 protein expression. Treatment with ATRA + ENT reduced HER2 expression and viability (P < 0.001) in AI-resistant cells, as well as decreased SP (P < 0.0001), mammosphere formation (P < 0.01), and expression of TIC molecular markers (P < 0.01) in LTLT-Ca. A reduction in tumor growth rate was observed in mice treated with ENT + ATRA + letrozole when compared to mice treated with single agents (P < 0.0001) or ENT + ATRA (P = 0.02). Decreased TIC characteristics, including mammosphere formation (P < 0.05), were observed in tumors from the triple combination. An increase in HER2 and downregulation in ERα protein expression was observed in patients upon resistance to AI (P < 0.005). These studies indicate that the combination of ATRA and ENT inhibits the TIC population of AI-resistant cells and may be effective in reducing tumor recurrence.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Histone Deacetylase Inhibitors/pharmacology , Receptor, ErbB-2/metabolism , Animals , Aromatase Inhibitors/pharmacology , Benzamides/administration & dosage , Benzamides/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Histone Deacetylase Inhibitors/administration & dosage , Humans , Letrozole , Mice, Nude , Nitriles/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Tretinoin/administration & dosage , Triazoles/pharmacology , Xenograft Model Antitumor Assays
8.
Sci Transl Med ; 7(297): 297ra113, 2015 Jul 22.
Article in English | MEDLINE | ID: mdl-26203081

ABSTRACT

Many neurological and psychiatric maladies originate from the deprivation of the human brain from estrogens. However, current hormone therapies cannot be used safely to treat these conditions commonly associated with menopause because of detrimental side effects in the periphery. The latter also prevents the use of the hormone for neuroprotection. We show that a small-molecule bioprecursor prodrug, 10ß,17ß-dihydroxyestra-1,4-dien-3-one (DHED), converts to 17ß-estradiol in the brain after systemic administration but remains inert in the rest of the body. The localized and rapid formation of estrogen from the prodrug was revealed by a series of in vivo bioanalytical assays and through in vivo imaging in rodents. DHED treatment efficiently alleviated symptoms that originated from brain estrogen deficiency in animal models of surgical menopause and provided neuroprotection in a rat stroke model. Concomitantly, we determined that 17ß-estradiol formed in the brain from DHED elicited changes in gene expression and neuronal morphology identical to those obtained after direct 17ß-estradiol treatment. Together, complementary functional and mechanistic data show that our approach is highly relevant therapeutically, because administration of the prodrug selectively produces estrogen in the brain independently from the route of administration and treatment regimen. Therefore, peripheral responses associated with the use of systemic estrogens, such as stimulation of the uterus and estrogen-responsive tumor growth, were absent. Collectively, our brain-selective prodrug approach may safely provide estrogen neuroprotection and medicate neurological and psychiatric symptoms developing from estrogen deficiency, particularly those encountered after surgical menopause, without the adverse side effects of current hormone therapies.


Subject(s)
Androstenediols/pharmacology , Brain/metabolism , Estradiol/metabolism , Estrogens/metabolism , Prodrugs/pharmacology , Androstenediols/therapeutic use , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Biomarkers/metabolism , Brain/drug effects , Brain Ischemia/complications , Brain Ischemia/drug therapy , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Estradiol/chemistry , Estrogens/chemistry , Female , Humans , MCF-7 Cells , Neuroprotection/drug effects , Prodrugs/metabolism , Stroke/complications , Stroke/drug therapy , Uterus/drug effects
9.
Endocr Relat Cancer ; 22(4): 645-56, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26113604

ABSTRACT

Obesity is a risk factor for breast cancer progression. Breast cancer patients who are overweight or obese or have excess abdominal fat have an increased risk of local or distant recurrence and cancer-related death. Hormone depletion therapies can also cause weight gain, exacerbating the risk for these patients. To understand the effect of obesity on hormone-dependent human breast cancer tumors, we fed ovariectomized athymic nude mice a diet containing 45% kcal fat and 17% kcal sucrose (high fat sucrose diet (HFSD)), 10% kcal fat (low fat diet (LFD)), or a standard chow diet (chow). The mice fed the HFSD developed metabolic abnormalities consistent with the development of obesity such as weight gain, high fasting blood glucose, and impaired glucose tolerance. These mice also developed hyperinsulinemia and insulin resistance. The obese mice also had a higher tumor growth rate compared to the lean mice. Furthermore, the obese mice showed a significantly reduced responsiveness to letrozole. To understand the role of obesity in this reduced responsiveness, we examined the effect of insulin on the growth of MCF-7Ca cells in response to estrogen or letrozole. The presence of insulin rendered MCF-7Ca cells less responsive to estrogen and letrozole. Exogenous insulin treatment of MCF-7Ca cells also resulted in increased p-Akt as well as ligand-independent phosphorylation of ERα. These findings suggest that diet-induced obesity may result in reduced responsiveness of tumors to letrozole due to the development of hyperinsulinemia. We conclude that obesity influences the response and resistance of breast cancer tumors to aromatase inhibitor treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Aromatase Inhibitors/therapeutic use , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Nitriles/therapeutic use , Obesity/metabolism , Triazoles/therapeutic use , Adipose Tissue/metabolism , Animals , Aromatase/genetics , Aromatase/metabolism , Blood Glucose/analysis , Cell Line, Tumor , Cyclooxygenase 2/genetics , Diet , Disease Models, Animal , Estrogens/pharmacology , Humans , Insulin/blood , Letrozole , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Obesity/blood , Ovariectomy , Tumor Burden
10.
Mol Cancer Ther ; 14(8): 1848-57, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26037781

ABSTRACT

Mortality following breast cancer diagnosis is mainly due to the development of distant metastasis. To escape from the primary site, tumor cells undergo the epithelial-to-mesenchymal transition (EMT), which helps them acquire a more motile and invasive phenotype. In our previous study, we showed that class I selective HDAC inhibitor entinostat reverses the EMT phenotype through reversal of epigenetic repression of E-cadherin. Recent evidence suggests that a subset of cells within a breast tumor may drive the metastatic outgrowth following escape from the primary site. These cells, termed tumor-initiating cells (TIC), represent a great threat to overall prognosis. They are critical in terms of drug resistance and tumor initiation at metastatic sites. Acquisition of EMT traits has also been shown to impart TIC phenotype to the cells, making EMT a "dual-threat" for prognosis. In the current study, we show that entinostat treatment can reduce the percentage of TIC cells from triple-negative breast cancer (TNBC) cells. Entinostat treatment was able to reduce the CD44(high)/CD24(low) cell population, ALDH-1 activity, as well as protein and mRNA expression of known TIC markers such as Bmi-1, Nanog, and Oct-4. Next, we inoculated MDA-MB-231 cells transfected with firefly luciferase (231/Luc) in mammary fat pad of NSG mice. The mice were then treated with entinostat (2.5 mg/kg/d), and tumor development and formation of metastasis were assessed by bioluminescence imaging. Treatment with entinostat significantly reduced tumor formation at the primary site as well as lung metastasis. As such, entinostat may help prevent development of distant metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pyridines/pharmacology , Triple Negative Breast Neoplasms/metabolism , Animals , Biomarkers , CD24 Antigen/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronan Receptors/metabolism , Immunophenotyping , Mice , MicroRNAs/genetics , Neoplasm Metastasis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Curr Pharm Des ; 20(42): 6575-83, 2014.
Article in English | MEDLINE | ID: mdl-25341934

ABSTRACT

Aromatase inhibitors (AIs) have become one of the mainstays of treatment of postmenopausal women with hormone receptorpositive breast cancer. However, acquired resistance to treatment continues to be a significant clinical challenge. There is increasing evidence from preclinical studies that activation of growth factor signaling pathways, as well as cross-talk between these pathways and estrogen receptor-alpha signaling pathways are important mechanisms that contribute to AI resistance. These preclinical studies have been the foundation for multiple randomized clinical trials that have evaluated combination targeted therapy in patients with advanced breast cancer. While the clinical benefit observed in these trials has been variable, the preclinical studies were successful in predicting clinical outcomes. This review focuses on mechanisms of acquired AI resistance and describes preclinical studies that have evaluated combination targeted therapy to overcome AI resistance, as well as clinical trials that have translated this information to the clinical setting.


Subject(s)
Aromatase Inhibitors/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Aromatase Inhibitors/therapeutic use , Combined Modality Therapy , Humans
12.
Future Oncol ; 10(3): 443-56, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24559450

ABSTRACT

Most breast cancer (BC) patients have tumors that express hormone receptors (HRs). Although endocrine therapy, such as aromatase inhibitors, is very effective, most patients with metastatic HR-positive (HR(+)) BC become resistant to endocrine therapy at some point in their treatment and subsequently require chemotherapy. The PI3K/mTOR pathway is often upregulated in endocrine-resistant BC patients and, therefore, has been one of the targets for development of new agents. Recently, a Phase III trial (BOLERO-2) in aromatase inhibitor-resistant BC patients showed a significant improvement in time to progression with the combination of everolimus and exemestane compared with exemestane alone, confirming the importance of the PI3K/mTOR pathway in endocrine-resistant BC. Side effects from mTOR inhibitors are manageable, but early detection and proactive management are required to ensure patients' safety, compliance and continuity of treatment. Thus, mTOR inhibitors offer a new hope and promise for patients with HR(+) BC.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Antineoplastic Agents, Hormonal/pharmacology , Drug Synergism , Everolimus , Female , Humans , Indoles/pharmacology , Indoles/therapeutic use , Purines/pharmacology , Purines/therapeutic use , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors
13.
Expert Rev Anticancer Ther ; 14(4): 381-93, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24559291

ABSTRACT

Clinically, there are two distinct types of aromatase inhibitor (AI) resistance, namely acquired and innate resistance. Because the underlying mechanisms of these two types of resistance may not be mutually exclusive, strategies to tackle these resistances may not be effective when used interchangeably. Activation of growth factor receptor pathways is the hallmark of acquired AI resistance. These pathways can be targeted either at the cell surface receptor level or their downstream signaling cascades. Currently, everolimus in combination with exemestane represents a new standard of care for patients progressing on non-steroidal AIs. HDAC inhibitors have also shown promising results For innate resistance, the combination of fulvestrant and AI in the front line setting represents a new treatment option, particularly for patients who present with de novo metastatic disease. A Phase III trial is currently ongoing to evaluate the benefit of CDK 4/6 inhibitor, palbociclib, in the first line setting in combination with AI.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Animals , Aromatase Inhibitors/classification , Breast Neoplasms/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Histone Deacetylases/metabolism , Humans , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/metabolism , Receptors, Growth Factor/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , src-Family Kinases/metabolism
14.
Breast Cancer Res ; 16(1): R15, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-24472707

ABSTRACT

INTRODUCTION: Although aromatase inhibitors (AIs; for example, letrozole) are highly effective in treating estrogen receptor positive (ER+) breast cancer, a significant percentage of patients either do not respond to AIs or become resistant to them. Previous studies suggest that acquired resistance to AIs involves a switch from dependence on ER signaling to dependence on growth factor-mediated pathways, such as human epidermal growth factor receptor-2 (HER2). However, the role of HER2, and the identity of other relevant factors that may be used as biomarkers or therapeutic targets remain unknown. This study investigated the potential role of transcription factor hypoxia inducible factor 1 (HIF-1) in acquired AI resistance, and its regulation by HER2. METHODS: In vitro studies using AI (letrozole or exemestane)-resistant and AI-sensitive cells were conducted to investigate the regulation and role of HIF-1 in AI resistance. Western blot and RT-PCR analyses were conducted to compare protein and mRNA expression, respectively, of ERα, HER2, and HIF-1α (inducible HIF-1 subunit) in AI-resistant versus AI-sensitive cells. Similar expression analyses were also done, along with chromatin immunoprecipitation (ChIP), to identify previously known HIF-1 target genes, such as breast cancer resistance protein (BCRP), that may also play a role in AI resistance. Letrozole-resistant cells were treated with inhibitors to HER2, kinase pathways, and ERα to elucidate the regulation of HIF-1 and BCRP. Lastly, cells were treated with inhibitors or inducers of HIF-1α to determine its importance. RESULTS: Basal HIF-1α protein and BCRP mRNA and protein are higher in AI-resistant and HER2-transfected cells than in AI-sensitive, HER2- parental cells under nonhypoxic conditions. HIF-1α expression in AI-resistant cells is likely regulated by HER2 activated-phosphatidylinositide-3-kinase/Akt-protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway, as its expression was inhibited by HER2 inhibitors and kinase pathway inhibitors. Inhibition or upregulation of HIF-1α affects breast cancer cell expression of BCRP; AI responsiveness; and expression of cancer stem cell characteristics, partially through BCRP. CONCLUSIONS: One of the mechanisms of AI resistance may be through regulation of nonhypoxic HIF-1 target genes, such as BCRP, implicated in chemoresistance. Thus, HIF-1 should be explored further for its potential as a biomarker of and therapeutic target.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Aromatase Inhibitors/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Neoplasm Proteins/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Hypoxia , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Letrozole , MCF-7 Cells , Nitriles/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/biosynthesis , Receptor, ErbB-2/antagonists & inhibitors , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Spheroids, Cellular , TOR Serine-Threonine Kinases/metabolism , Triazoles/pharmacology , Tumor Cells, Cultured
15.
Breast Cancer Res Treat ; 143(1): 99-111, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24305977

ABSTRACT

Loss of ERα in breast cancer correlates with poor prognosis, increased recurrence rates, and higher incidence of metastasis. Epigenetic silencing of E-cadherin (loss of which is associated with more invasive phenotype) is observed in metastatic cell lines and invasive breast cancers. Here, we are showing that entinostat (ENT) can reverse epithelial to mesenchymal transition (EMT), which is considered to be a first step in the process of metastases formation. Triple-negative breast cancer cells such as MDA-MB-231 and Hs578T show a basal phenotype characterized by loss of E-cadherin expression and higher expression of mesenchymal markers such as N-cadherin and vimentin along with transcriptional repressors such as twist and snail. When MDA-MB-231 and Hs578T cells or tumors were treated with ENT, E-cadherin transcription was increased along with reduction in N-cadherin mRNA expression. Chromatin immunoprecipitation assay showed that treatment of MDA-MB-231 and Hs578T cells increased the activation of E-cadherin promoter by reducing the association of twist and snail with the E-cadherin (CDH1) promoter and downregulated both the snail and twist. ENT also inhibited cell migration in vitro. In addition, phosphorylation of vimentin was increased, as well as remodeling of vimentin filaments. ENT treatment also reduced formation of tubulin-based microtentacles, which help floating cells attach to other surfaces. These findings suggest that ENT can reverse EMT and may reduce the formation of metastasis.


Subject(s)
Benzamides/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cadherins/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Histone Deacetylase Inhibitors/pharmacology , Pyridines/pharmacology , Breast Neoplasms/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Promoter Regions, Genetic , Protein Binding , Snail Family Transcription Factors , Transcription Factors/metabolism , Twist-Related Protein 1/metabolism
16.
Mol Cancer Ther ; 12(12): 2804-16, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24092810

ABSTRACT

We previously showed that in innately resistant tumors, silencing of the estrogen receptor (ER) could be reversed by treatment with a histone deacetylase (HDAC) inhibitor, entinostat. Tumors were then responsive to aromatase inhibitor (AI) letrozole. Here, we investigated whether ER in the acquired letrozole-resistant tumors could be restored with entinostat. Ovariectomized athymic mice were inoculated with MCF-7Ca cells, supplemented with androstenedione (Δ(4)A), the aromatizable substrate. When the tumors reached about 300 mm(3), the mice were treated with letrozole. After initial response to letrozole, the tumors eventually became resistant (doubled their initial volume). The mice then were grouped to receive letrozole, exemestane (250 µg/d), entinostat (50 µg/d), or the combination of entinostat with letrozole or exemestane for 26 weeks. The growth rates of tumors of mice treated with the combination of entinostat with letrozole or exemestane were significantly slower than with the single agent (P < 0.05). Analysis of the letrozole-resistant tumors showed entinostat increased ERα expression and aromatase activity but downregulated Her-2, p-Her-2, p-MAPK, and p-Akt. However, the mechanism of action of entinostat in reversing acquired resistance did not involve epigenetic silencing but rather included posttranslational as well as transcriptional modulation of Her-2. Entinostat treatment reduced the association of the Her-2 protein with HSP-90, possibly by reducing the stability of Her-2 protein. In addition, entinostat also reduced Her-2 mRNA levels and its stability. Our results suggest that the HDAC inhibitor may reverse letrozole resistance in cells and tumors by modulating Her-2 expression and activity.


Subject(s)
Antineoplastic Agents/pharmacology , Aromatase Inhibitors/pharmacology , Benzamides/pharmacology , Drug Resistance, Neoplasm , Histone Deacetylase Inhibitors/pharmacology , Nitriles/pharmacology , Pyridines/pharmacology , Receptor, ErbB-2 , Triazoles/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Aromatase/genetics , Aromatase/metabolism , Aromatase Inhibitors/administration & dosage , Benzamides/administration & dosage , Disease Models, Animal , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase Inhibitors/administration & dosage , Humans , Letrozole , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Mice , Nitriles/administration & dosage , Pyridines/administration & dosage , RNA Stability/drug effects , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Triazoles/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
17.
Breast Cancer Res Treat ; 138(3): 699-708, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23508762

ABSTRACT

Despite significant improvement in the treatment outcome of hormone responsive postmenopausal breast cancer, some patients eventually acquire resistance to aromatase inhibitors (AIs). Using our MCF-7Ca xenograft model, we observed that although AIs such as anastrozole initially inhibit tumor growth effectively, tumors eventually began to grow. Our previous data show that anastrozole-resistant tumors upregulate growth factor receptor pathways as they adapt to grow in the low estrogen environment. Therefore, in the current study, we investigated the effect of inhibiting the growth factor receptor pathways with a MEK-1/2 inhibitor selumetinib (AZD6244, ARRY-142866). We treated the mice with anastrozole-resistant tumors with selumetinib alone or in combination with anastrozole. MCF-7Ca cells were inoculated sc into ovariectomized athymic nude mice supplemented throughout the experiment with androstenedione (100 µg/day), the substrate for aromatase conversion to estrogen. Once the tumors reached a measurable size (~300 mm(3)), the mice were treated with anastrozole (200 µg/day), supplemented with androstenedione (Δ(4)A). The tumors in the anastrozole group doubled in volume after 6 weeks, at which time the animals were regrouped to receive the following treatments: (i) anastrozole, (ii) anastrozole withdrawal (Δ(4)A alone), (iii) selumetinib (25 mg/kg/d, bid, po), and (iv) selumetinib + anastrozole, (n = 10 mice/group). The treatments were given for 6 weeks (till week 12) and then the mice were euthanized, the tumors were collected and analyzed. The tumors of mice treated with selumetinib + anastrozole had significantly lower growth rates than those treated with single agents (p = 0.008). Western blot analysis of the tumors showed that treatment with anastrozole resulted in upregulation of proteins in the growth factor receptor cascade such as p-mTOR, pAkt, pMEK, and pMAPK. This was accompanied by downregulation of ERα protein, consistent with previous findings. The treatment of mice with selumetinib resulted in downregulation of activated MAPK, along with p-mTOR, which likely resulted in upregulation of ERα. Our results suggest that inhibition of the growth factor receptor pathway with selumetinib can reverse anastrozole resistance.


Subject(s)
Benzimidazoles/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Nitriles/pharmacology , Triazoles/pharmacology , Anastrozole , Androstenedione/pharmacology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aromatase Inhibitors/pharmacology , Benzimidazoles/administration & dosage , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , Mice , Mice, Nude , Nitriles/administration & dosage , Ovariectomy , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Triazoles/administration & dosage , Xenograft Model Antitumor Assays
18.
Breast Cancer Res Treat ; 135(3): 681-92, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22878889

ABSTRACT

Aromatase inhibitors (AIs) are an effective therapy in treating estrogen receptor-positive breast cancer. Nonetheless, a significant percentage of patients either do not respond or become resistant to AIs. Decreased dependence on ER-signaling and increased dependence on growth factor receptor signaling pathways, particularly human epidermal growth factor receptor 2 (EGFR2/HER2), have been implicated in AI resistance. However, the role of growth factor signaling remains unclear. This current study investigates the possibility that signaling either through HER2 alone or through interplay between epidermal growth factor receptor 1 (EGFR/HER1) and HER2 mediates AI resistance by increasing the tumor initiating cell (TIC) subpopulation in AI-resistant cells via regulation of stem cell markers, such as breast cancer resistance protein (BCRP). TICs and BCRP are both known to be involved in drug resistance. Results from in vitro analyses of AI-resistant versus AI-sensitive cells and HER2-versus HER2+ cells, as well as from in vivo xenograft tumors, indicate that (1) AI-resistant cells overexpress both HER2 and BCRP and exhibit increased TIC characteristics compared to AI-sensitive cells; (2) inhibition of HER2 and/or BCRP decrease TIC characteristics in letrozole-resistant cells; and (3) HER2 and its dimerization partner EGFR/HER1 are involved in the regulation of BCRP. Overall, these results suggest that reducing or eliminating the TIC subpopulation with agents that target BCRP, HER2, EGFR/HER1, and/or their downstream kinase pathways could be effective in preventing and/or treating acquired AI resistance.


Subject(s)
Aromatase Inhibitors/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Nitriles/pharmacology , Receptor, ErbB-2/metabolism , Triazoles/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Female , Humans , Letrozole , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
19.
Macromol Biosci ; 12(1): 55-60, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22052593

ABSTRACT

Trastuzumab (TRZ) is a humanized monoclonal antibody that targets the extracellular domain of the human epidermal growth factor receptor type 2 (Her2). Semitelechelic (ST) poly[N-(2-hydroxypropyl)methacrylamide]-TRZ conjugates are successfully synthesized and evaluated as a potential drug delivery system that actively targets Her2-overexpressing cancer cells. The ST backbone shows favorable characteristics when conjugated to TRZ. The conjugate exhibits comparable and prolonged anticancer activity when compared to free TRZ in Her2 overexpressing breast cancer cell lines.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Breast Neoplasms/drug therapy , Drug Delivery Systems , Polymethacrylic Acids/administration & dosage , Polymethacrylic Acids/chemical synthesis , Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Agents/chemistry , Breast Neoplasms/pathology , Cell Survival , Drug Delivery Systems/methods , Female , Humans , Mitogen-Activated Protein Kinases/metabolism , Polymethacrylic Acids/chemistry , Receptor, ErbB-2/metabolism , Trastuzumab , Tumor Cells, Cultured
20.
BMC Cancer ; 11: 231, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21658239

ABSTRACT

BACKGROUND: Aromatase inhibitors (AI) that inhibit breast cancer cell growth by blocking estrogen synthesis have become the treatment of choice for post-menopausal women with estrogen receptor positive (ER+) breast cancer. However, some patients display de novo or acquired resistance to AI. Interactions between estrogen and growth factor signaling pathways have been identified in estrogen-responsive cells as one possible reason for acquisition of resistance. Our laboratory has characterized an autocrine growth factor overexpressed in invasive ductal carcinoma named PC-Cell Derived Growth Factor (GP88), also known as progranulin. In the present study, we investigated the role GP88 on the acquisition of resistance to letrozole in ER+ breast cancer cells METHODS: We used two aromatase overexpressing human breast cancer cell lines MCF-7-CA cells and AC1 cells and their letrozole resistant counterparts as study models. Effect of stimulating or inhibiting GP88 expression on proliferation, anchorage-independent growth, survival and letrozole responsiveness was examined. RESULTS: GP88 induced cell proliferation and conferred letrozole resistance in a time- and dose-dependent fashion. Conversely, naturally letrozole resistant breast cancer cells displayed a 10-fold increase in GP88 expression when compared to letrozole sensitive cells. GP88 overexpression, or exogenous addition blocked the inhibitory effect of letrozole on proliferation, and stimulated survival and soft agar colony formation. In letrozole resistant cells, silencing GP88 by siRNA inhibited cell proliferation and restored their sensitivity to letrozole. CONCLUSION: Our findings provide information on the role of an alternate growth and survival factor on the acquisition of aromatase inhibitor resistance in ER+ breast cancer.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/metabolism , Breast Neoplasms/enzymology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Intercellular Signaling Peptides and Proteins/metabolism , Nitriles/pharmacology , Triazoles/pharmacology , Aromatase/genetics , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Female , Gene Silencing , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Letrozole , Progranulins , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , RNA, Small Interfering/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...