13 results on '"Seth A. Wander"'
Search Results
2. Interplay between ESR1/PIK3CA codon variants, oncogenic pathway alterations and clinical phenotype in patients with metastatic breast cancer (MBC): comprehensive circulating tumor DNA (ctDNA) analysis
- Author
-
Lorenzo Gerratana, Andrew A. Davis, Marko Velimirovic, Katherine Clifton, Whitney L. Hensing, Ami N. Shah, Charles S. Dai, Carolina Reduzzi, Paolo D’Amico, Firas Wehbe, Arielle Medford, Seth A. Wander, William J. Gradishar, Amir Behdad, Fabio Puglisi, Cynthia X. Ma, Aditya Bardia, and Massimo Cristofanilli
- Subjects
Circulating biomarker ,Endocrine therapy ,Next generation sequencing ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background although being central for the biology and druggability of hormone-receptor positive, HER2 negative metastatic breast cancer (MBC), ESR1 and PIK3CA mutations are simplistically dichotomized as mutated or wild type in current clinical practice. Methods The study analyzed a multi-institutional cohort comprising 703 patients with luminal-like MBC characterized for circulating tumor DNA through next generation sequencing (NGS). Pathway classification was defined based on previous work (i.e., RTK, RAS, RAF, MEK, NRF2, ER, WNT, MYC, P53, cell cycle, notch, PI3K). Single nucleotide variations (SNVs) were annotated for their oncogenicity through OncoKB. Only pathogenic variants were included in the models. Associations among clinical characteristics, pathway classification, and ESR1/PIK3CA codon variants were explored. Results The results showed a differential pattern of associations for ESR1 and PIK3CA codon variants in terms of co-occurring pathway alterations patterns of metastatic dissemination, and prognosis. ESR1 537 was associated with SNVs in the ER and RAF pathways, CNVs in the MYC pathway and bone metastases, while ESR1 538 with SNVs in the cell cycle pathway and liver metastases. PIK3CA 1047 and 542 were associated with CNVs in the PI3K pathway and with bone metastases. Conclusions The study demonstrated how ESR1 and PIK3CA codon variants, together with alterations in specific oncogenic pathways, can differentially impact the biology and clinical phenotype of luminal-like MBC. As novel endocrine therapy agents such as selective estrogen receptor degraders (SERDS) and PI3K inhibitors are being developed, these results highlight the pivotal role of ctDNA NGS to describe tumor evolution and optimize clinical decision making.
- Published
- 2023
- Full Text
- View/download PDF
3. Adverse kidney outcomes of CDK 4/6 inhibitors for metastatic breast cancer
- Author
-
Paul E. Hanna, Ian A. Strohbehn, Daiana Moreno, Destiny Harden, Rituvanthikaa Seethapathy, Rani Sawtell, Qiyu Wang, Tianqi Ouyang, Nurit Katz-Agranov, James Dinulos, Seth A. Wander, Shruti Gupta, and Meghan E. Sise
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Cyclin-dependent kinase (CDK) 4/6 inhibitors have significantly improved overall and progression free survival of patients with metastatic breast cancer, but their effect on short and long-term kidney function is unknown. We found that early, mild estimated glomerular filtration rate (eGFR) decline was common in patients treated with CDK 4/6 inhibitors; however, severe kidney injury is rare and long-term eGFR decline is uncommon.
- Published
- 2023
- Full Text
- View/download PDF
4. ESR1 mutation as an emerging clinical biomarker in metastatic hormone receptor-positive breast cancer
- Author
-
Jamie O. Brett, Laura M. Spring, Aditya Bardia, and Seth A. Wander
- Subjects
Breast cancer ,Hormone receptor/estrogen receptor ,ESR1 mutation ,Resistance ,Combination ,SERD ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract In metastatic hormone receptor-positive breast cancer, ESR1 mutations are a common cause of acquired resistance to the backbone of therapy, estrogen deprivation by aromatase inhibition. How these mutations affect tumor sensitivity to established and novel therapies are active areas of research. These therapies include estrogen receptor-targeting agents, such as selective estrogen receptor modulators, covalent antagonists, and degraders (including tamoxifen, fulvestrant, and novel agents), and combination therapies, such as endocrine therapy plus CDK4/6, PI3K, or mTORC1 inhibition. In this review, we summarize existing knowledge surrounding the mechanisms of action of ESR1 mutations and roles in resistance to aromatase inhibition. We then analyze the recent literature on how ESR1 mutations affect outcomes in estrogen receptor-targeting and combination therapies. For estrogen receptor-targeting therapies such as tamoxifen and fulvestrant, ESR1 mutations cause relative resistance in vitro but do not clearly lead to resistance in patients, making novel agents in this category promising. Regarding combination therapies, ESR1 mutations nullify any aromatase inhibitor component of the combination. Thus, combinations using endocrine alternatives to aromatase inhibition, or combinations where the non-endocrine component is efficacious as monotherapy, are still effective against ESR1 mutations. These results emphasize the importance of investigating combinatorial resistance, challenging as these efforts are. We also discuss future directions and open questions, such as studying the differences among distinct ESR1 mutations, asking how to adjust clinical decisions based on molecular surveillance testing, and developing novel therapies that are effective against ESR1 mutations.
- Published
- 2021
- Full Text
- View/download PDF
5. Immunogenicity of SARS-CoV-2 vaccines in patients with breast cancer
- Author
-
Elyssa Denault, Erika Nakajima, Vivek Naranbhai, Jennifer A. Hutchinson, Lindsey Mortensen, Elizabeth Neihoff, Caroline Barabell, Amy Comander, Dejan Juric, Irene Kuter, Theresa Mulvey, Jeffrey Peppercorn, Aron S. Rosenstock, Jennifer Shin, Neelima Vidula, Seth A. Wander, Beverly Moy, Leif W. Ellisen, Steven J. Isakoff, A. John Iafrate, Justin F. Gainor, Aditya Bardia, and Laura M. Spring
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Purpose: To explore the immunogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines in patients with breast cancer based on type of anticancer treatment. Methods: Patients with breast cancer had anti-spike antibody concentrations measured ⩾14 days after receiving a full SARS-CoV-2 vaccination series. The primary endpoint was IgA/G/M anti-spike antibody concentration. Multiple regression analysis was used to analyze log 10 -transformed antibody titer concentrations. Results: Between 29 April and 20 July 2021, 233 patients with breast cancer were enrolled, of whom 212 were eligible for the current analysis. Patients who received mRNA-1273 (Moderna) had the highest antibody concentrations [geometric mean concentration (GMC) in log 10 : 3.0 U/mL], compared to patients who received BNT162b2 (Pfizer) (GMC: 2.6 U/mL) (multiple regression adjusted p = 0.013) and Ad26.COV2.S (Johnson & Johnson/Janssen) (GMC: 2.6 U/mL) ( p = 0.071). Patients receiving cytotoxic therapy had a significantly lower antibody titer GMC (2.5 U/mL) compared to patients on no therapy or endocrine therapy alone (3.0 U/mL) ( p = 0.005). Patients on targeted therapies (GMC: 2.7 U/mL) also had a numerically lower GMC compared to patients not receiving therapy/on endocrine therapy alone, although this result was not significant ( p = 0.364). Among patients who received an additional dose of vaccine ( n = 31), 28 demonstrated an increased antibody response that ranged from 0.2 to >4.4 U/ mL. Conclusion: Most patients with breast cancer generate detectable anti-spike antibodies following SARS-CoV-2 vaccination, though systemic treatments and vaccine type impact level of response. Further studies are needed to better understand the clinical implications of different antibody levels, the effectiveness of additional SARS-CoV-2 vaccine doses, and the risk of breakthrough infections among patients with breast cancer.
- Published
- 2022
- Full Text
- View/download PDF
6. Next-generation selective estrogen receptor degraders and other novel endocrine therapies for management of metastatic hormone receptor-positive breast cancer: current and emerging role
- Author
-
Maxwell R. Lloyd, Seth A. Wander, Erika Hamilton, Pedram Razavi, and Aditya Bardia
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Endocrine therapy (ET) is a pivotal strategy to manage early- and advanced-stage estrogen receptor-positive (ER+) breast cancer. In patients with metastatic breast cancer (MBC), progression of disease inevitably occurs due to the presence of acquired or intrinsic resistance mechanisms. ET resistance can be driven by ligand-independent, ER-mediated signaling that promotes tumor proliferation in the absence of hormone, or ER-independent oncogenic signaling that circumvents endocrine regulated transcription pathways. Estrogen receptor 1 ( ESR1 ) mutations induce constitutive ER activity and upregulate ER-dependent gene transcription, provoking resistance to estrogen deprivation and aromatase inhibitor therapy. The role ESR1 mutations play in regulating response to other therapies, such as the selective estrogen receptor degrader (SERD) fulvestrant and the available CDK4/6 inhibitors, is less clear. Novel oral SERDs and other next-generation ETs are in clinical development for ER+ breast cancer as single agents and in combination with established targeted therapies. Recent results from the phase III EMERALD trial demonstrated improved outcomes with the oral SERD elacestrant compared to standard anti-estrogen therapies in ER+ MBC after prior progression on ET, and other agents have shown promise in both the laboratory and early-phase clinical trials. In this review, we will discuss the emerging data related to oral SERDs and other novel ET in managing ER+ breast cancer. As clinical data continue to mature on these next-generation ETs, important questions will emerge related to the optimal sequence of therapeutic options and the genomic and molecular landscape of resistance to these agents.
- Published
- 2022
- Full Text
- View/download PDF
7. MicroRNA-Mediated Suppression of the TGF-β Pathway Confers Transmissible and Reversible CDK4/6 Inhibitor Resistance
- Author
-
Liam Cornell, Seth A. Wander, Tanvi Visal, Nikhil Wagle, and Geoffrey I. Shapiro
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: CDK4/6 inhibition is now part of the standard armamentarium for patients with estrogen receptor-positive (ER+) breast cancer, so that defining mechanisms of resistance is a pressing issue. Here, we identify increased CDK6 expression as a key determinant of acquired resistance after palbociclib treatment in ER+ breast cancer cells. CDK6 expression is critical for cellular survival during palbociclib exposure. The increased CDK6 expression observed in resistant cells is dependent on TGF-β pathway suppression via miR-432-5p expression. Exosomal miR-432-5p expression mediates the transfer of the resistance phenotype between neighboring cell populations. Levels of miR-432-5p are higher in primary breast cancers demonstrating CDK4/6 resistance compared to those that are sensitive. These data are further confirmed in pre-treatment and post-progression biopsies from a parotid cancer patient who had responded to ribociclib, demonstrating the clinical relevance of this mechanism. Finally, the CDK4/6 inhibitor resistance phenotype is reversible in vitro and in vivo by a prolonged drug holiday. : Cornell et al. demonstrate a mechanism of acquired CDK4/6 inhibitor resistance that is independent of inherent genetic mutations, is conferred through extracellular signaling, and is reversible in vitro and in vivo. Resistance was mediated by exosomal miRNA, causing increased expression of CDK6 to overcome G1 arrest and promote cell survival. Keywords: breast cancer, drug resistance, targeted therapy, exosomes, CDK6, microRNA, TGF-β, SMAD4, palbociclib, ribociclib
- Published
- 2019
- Full Text
- View/download PDF
8. Phase I study of the aurora A kinase inhibitor alisertib with induction chemotherapy in patients with acute myeloid leukemia
- Author
-
Amir T. Fathi, Seth A. Wander, Traci M. Blonquist, Andrew M. Brunner, Philip C. Amrein, Jeffrey Supko, Nicole M. Hermance, Amity L. Manning, Hossein Sadrzadeh, Karen K. Ballen, Eyal C. Attar, Timothy A. Graubert, Gabriela Hobbs, Christelle Joseph, Ashley M. Perry, Meghan Burke, Regina Silver, Julia Foster, Meghan Bergeron, Aura Y. Ramos, Tina T. Som, Kaitlyn M. Fishman, Kristin L. McGregor, Christine Connolly, Donna S. Neuberg, and Yi-Bin Chen
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Aberrant expression of aurora kinase A is implicated in the genesis of various neoplasms, including acute myeloid leukemia. Alisertib, an aurora A kinase inhibitor, has demonstrated efficacy as monotherapy in trials of myeloid malignancy, and this efficacy appears enhanced in combination with conventional chemotherapies. In this phase I, dose-escalation study, newly diagnosed patients received conventional induction with cytarabine and idarubicin, after which alisertib was administered for 7 days. Dose escalation occurred via cohorts. Patients could then receive up to four cycles of consolidation, incorporating alisertib, and thereafter alisertib maintenance for up to 12 months. Twenty-two patients were enrolled. One dose limiting toxicity occurred at dose level 2 (prolonged thrombocytopenia), and the recommended phase 2 dose was established at 30mg twice daily. Common therapy-related toxicities included cytopenias and mucositis. Only three (14%) patients had persistent disease at mid-cycle, requiring “5+2” reinduction. The composite remission rate (complete remission and complete remission with incomplete neutrophil recovery) was 86% (nineteen of twenty-two patients; 90% CI 68–96%). Among those over age 65 and those with high-risk disease (secondary acute leukemia or cytogenetically high-risk disease), the composite remission rate was 88% and 100%, respectively. The median follow up was 13.5 months. Of those treated at the recommended phase 2 dose, the 12-month overall survival and progression-free survival were 62% (90% CI 33–81%) and 42% (90% CI 17–65%), respectively. Alisertib is well tolerated when combined with induction chemotherapy in acute myeloid leukemia, with a promising suggestion of efficacy. (clinicaltrials.gov Identifier:01779843).
- Published
- 2017
- Full Text
- View/download PDF
9. Triple negative breast cancer initiating cell subsets differ in functional and molecular characteristics and in γ‐secretase inhibitor drug responses
- Author
-
Diana J. Azzam, Dekuang Zhao, Jun Sun, Andy J. Minn, Prathibha Ranganathan, Katherine Drews‐Elger, Xiaoqing Han, Manuel Picon‐Ruiz, Candace A. Gilbert, Seth A. Wander, Anthony J. Capobianco, Dorraya El‐Ashry, and Joyce M. Slingerland
- Subjects
breast cancer stem cells ,GSI ,metastasis ,Notch1 ,Sox2 ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Increasing evidence suggests that stem‐like cells mediate cancer therapy resistance and metastasis. Breast tumour‐initiating stem cells (T‐ISC) are known to be enriched in CD44+CD24neg/low cells. Here, we identify two T‐ISC subsets within this population in triple negative breast cancer (TNBC) lines and dissociated primary breast cancer cultures: CD44+CD24low+ subpopulation generates CD44+CD24neg progeny with reduced sphere formation and tumourigenicity. CD44+CD24low+ populations contain subsets of ALDH1+ and ESA+ cells, yield more frequent spheres and/or T‐ISC in limiting dilution assays, preferentially express metastatic gene signatures and show greater motility, invasion and, in the MDA‐MB‐231 model, metastatic potential. CD44+CD24low+ but not CD44+CD24neg express activated Notch1 intracellular domain (N1‐ICD) and Notch target genes. We show N1‐ICD transactivates SOX2 to increase sphere formation, ALDH1+ and CD44+CD24low+cells. Gamma secretase inhibitors (GSI) reduced sphere formation and xenograft growth from CD44+CD24low+ cells, but CD44+CD24neg were resistant. While GSI hold promise for targeting T‐ISC, stem cell heterogeneity as observed herein, could limit GSI efficacy. These data suggest a breast T‐ISC hierarchy in which distinct pathways drive developmentally related subpopulations with different anti‐cancer drug responsiveness.
- Published
- 2013
- Full Text
- View/download PDF
10. Targeting CDK4 and 6 in Cancer Therapy: Emerging Preclinical Insights Related to Abemaciclib
- Author
-
Seth A Wander, Neil O’Brien, Lacey M Litchfield, Declan O’Dea, Claudia Morato Guimaraes, Dennis J Slamon, and Shom Goel
- Subjects
Cancer Research ,CDK4 and 6 ,Tumor Suppressor Proteins ,Oncology and Carcinogenesis ,Aminopyridines ,Cyclin-Dependent Kinase 4 ,Breast Neoplasms ,Estrogens ,Cyclin-Dependent Kinase 6 ,abemaciclib ,Estrogen ,Oncology ,5.1 Pharmaceuticals ,Breast Cancer ,preclinical ,Humans ,Benzimidazoles ,Female ,Oncology & Carcinogenesis ,Mitogens ,Development of treatments and therapeutic interventions ,Protein Kinase Inhibitors ,antitumor ,Cancer - Abstract
Pharmacologic inhibitors of cyclin-dependent kinases 4 and 6 (CDK4 and 6) are approved for the treatment of subsets of patients with hormone receptor positive (HR+) breast cancer (BC). In metastatic disease, strategies involving endocrine therapy combined with CDK4 and 6 inhibitors (CDK4 and 6i) improve clinical outcomes in HR+ BCs. CDK4 and 6i prevent retinoblastoma tumor suppressor protein phosphorylation, thereby blocking the transcription of E2F target genes, which in turn inhibits both mitogen and estrogen-mediated cell proliferation. In this review, we summarize preclinical data pertaining to the use of CDK4 and 6i in BC, with a particular focus on several of the unique chemical, pharmacologic, and mechanistic properties of abemaciclib. As research efforts elucidate the novel mechanisms underlying abemaciclib activity, potential new applications are being identified. For example, preclinical studies have demonstrated abemaciclib can exert antitumor activity against multiple tumor types and can cross the blood-brain barrier. Abemaciclib has also demonstrated distinct activity as a monotherapeutic in the treatment of BC. Accordingly, we also discuss how a greater understanding of mechanisms related to CDK4 and 6 blockade highlight abemaciclib’s unique in-class properties, and could pave new avenues for enhancing its therapeutic efficacy.
- Published
- 2022
- Full Text
- View/download PDF
11. Cytoplasmic p27 promotes epithelial–mesenchymal transition and tumor metastasis via STAT3-mediated Twist1 upregulation
- Author
-
Joyce M. Slingerland, Bin Wang, Tan A. Ince, Dan Theodorescu, Wei Guo, Wen Zhou, Dekuang Zhao, Jun Sun, Seth A. Wander, Gordon B. Mills, Alexandra H. Besser, and Michael A. Durante
- Subjects
STAT3 Transcription Factor ,Cytoplasm ,Cancer Research ,Epithelial-Mesenchymal Transition ,animal structures ,Mice, Nude ,Breast Neoplasms ,Biology ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Growth factor receptor ,Cell Line, Tumor ,Proliferating Cell Nuclear Antigen ,Genetics ,Animals ,Humans ,Epithelial–mesenchymal transition ,Neoplasm Metastasis ,Promoter Regions, Genetic ,STAT3 ,Molecular Biology ,Protein kinase B ,Cell Proliferation ,030304 developmental biology ,Mice, Inbred BALB C ,0303 health sciences ,Janus kinase 2 ,Twist-Related Protein 1 ,Nuclear Proteins ,Epithelial Cells ,Janus Kinase 2 ,Cell cycle ,Up-Regulation ,HEK293 Cells ,Urinary Bladder Neoplasms ,030220 oncology & carcinogenesis ,Cancer cell ,MCF-7 Cells ,STAT protein ,biology.protein ,Cancer research ,Original Article ,Female ,Signal Transduction - Abstract
p27 restrains normal cell growth, but PI3K-dependent C-terminal phosphorylation of p27 at threonine 157 (T157) and T198 promotes cancer cell invasion. Here, we describe an oncogenic feedforward loop in which p27pT157pT198 binds Janus kinase 2 (JAK2) promoting STAT3 (signal transducer and activator of transcription 3) recruitment and activation. STAT3 induces TWIST1 to drive a p27-dependent epithelial-mesenchymal transition (EMT) and further activates AKT contributing to acquisition and maintenance of metastatic potential. p27 knockdown in highly metastatic PI3K-activated cells reduces STAT3 binding to the TWIST1 promoter, TWIST1 promoter activity and TWIST1 expression, reverts EMT and impairs metastasis, whereas activated STAT3 rescues p27 knockdown. Cell cycle-defective phosphomimetic p27T157DT198D (p27CK-DD) activates STAT3 to induce a TWIST1-dependent EMT in human mammary epithelial cells and increases breast and bladder cancer invasion and metastasis. Data support a mechanism in which PI3K-deregulated p27 binds JAK2, to drive STAT3 activation and EMT through STAT3-mediated TWIST1 induction. Furthermore, STAT3, once activated, feeds forward to further activate AKT.
- Published
- 2015
- Full Text
- View/download PDF
12. The evolving role of FLT3 inhibitors in acute myeloid leukemia: quizartinib and beyond
- Author
-
Amir T. Fathi, Seth A. Wander, and Mark J. Levis
- Subjects
biology ,business.industry ,Lestaurtinib ,Sunitinib ,medicine.medical_treatment ,Reviews ,Myeloid leukemia ,hemic and immune systems ,Hematology ,Hematopoietic stem cell transplantation ,Pharmacology ,Receptor tyrosine kinase ,Clinical trial ,chemistry.chemical_compound ,fluids and secretions ,chemistry ,hemic and lymphatic diseases ,embryonic structures ,Cancer research ,biology.protein ,Medicine ,Midostaurin ,business ,medicine.drug ,Quizartinib - Abstract
Acute myeloid leukemia remains associated with poor outcomes despite advances in our understanding of the complicated molecular events driving leukemogenesis and malignant progression. Those patients harboring mutations in the FLT3 receptor tyrosine kinase have a particularly poor prognosis; however, significant excitement has been generated by the emergence of a variety of targeted inhibitors capable of suppressing FLT3 signaling in vivo. Here we will review results from preclinical studies and early clinical trials evaluating both first- and second-generation FLT3 inhibitors. Early FLT3 inhibitors (including sunitinib, midostaurin, and lestaurtinib) demonstrated significant promise in preclinical models of FLT3 mutant AML. Unfortunately, many of these compounds failed to achieve robust and sustained FLT3 inhibition in early clinical trials, at best resulting in only transient decreases in peripheral blast counts. These results have prompted the development of second-generation FLT3 inhibitors, epitomized by the novel agent quizartinib. These second-generation inhibitors have demonstrated enhanced FLT3 specificity and have been generally well tolerated in early clinical trials. Several FLT3 inhibitors have reached phase III clinical trials, and a variety of phase I/II trials exploring a role for these novel compounds in conjunction with conventional chemotherapy or hematopoietic stem cell transplantation are ongoing. Finally, molecular insights provided by FLT3 inhibitors have shed light upon the variety of mechanisms underlying the acquisition of resistance and have provided a rationale supporting the use of combinatorial regimens with other emerging targeted therapies.
- Published
- 2014
- Full Text
- View/download PDF
13. PI3K/mTOR inhibition can impair tumor invasion and metastasis in vivo despite a lack of antiproliferative action in vitro: implications for targeted therapy
- Author
-
Clara Milikowski, Feng Hong, Jianqin Wei, Tan A. Ince, Dekuang Zhao, Andy J. Minn, Seth A. Wander, Joyce M. Slingerland, Nanette H. Bishopric, Alexandra H. Besser, and Chad J. Creighton
- Subjects
Cytoplasm ,Cancer Research ,medicine.medical_treatment ,Gene Expression ,Kaplan-Meier Estimate ,Metastasis ,Targeted therapy ,Mice ,Preclinical Study ,0302 clinical medicine ,Cell Movement ,Molecular Targeted Therapy ,RNA, Small Interfering ,Phosphoinositide-3 Kinase Inhibitors ,Mice, Inbred BALB C ,0303 health sciences ,TOR Serine-Threonine Kinases ,Micrometastasis ,p27 ,Motility ,Tumor Burden ,3. Good health ,Cell biology ,Oncology ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Female ,Erratum ,Signal transduction ,Cyclin-Dependent Kinase Inhibitor p27 ,Signal Transduction ,Pyridones ,Cancer invasion ,Mice, Nude ,Bone Neoplasms ,Breast Neoplasms ,Biology ,Disease-Free Survival ,03 medical and health sciences ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,030304 developmental biology ,Cell growth ,medicine.disease ,Xenograft Model Antitumor Assays ,PI3K/mTOR ,Pyrimidines ,Cancer research - Abstract
Oncogenic PI3K/mTOR activation is frequently observed in human cancers and activates cell motility via p27 phosphorylations at T157 and T198. Here we explored the potential for a novel PI3K/mTOR inhibitor to inhibit tumor invasion and metastasis. An MDA-MB-231 breast cancer line variant, MDA-MB-231-1833, with high metastatic bone tropism, was treated with a novel catalytic PI3K/mTOR inhibitor, PF-04691502, at nM doses that did not impair proliferation. Effects on tumor cell motility, invasion, p27 phosphorylation, localization, and bone metastatic outgrowth were assayed. MDA-MB-231-1833 showed increased PI3K/mTOR activation, high levels of cytoplasmic p27pT157pT198 and increased cell motility and invasion in vitro versus parental. PF-04691502 treatment, at a dose that did not affect proliferation, reduced total and cytoplasmic p27, decreased p27pT157pT198 and restored cell motility and invasion to levels seen in MDA-MB-231. p27 knockdown in MDA-MB-231-1833 phenocopied PI3K/mTOR inhibition, whilst overexpression of the phosphomimetic mutant p27T157DT198D caused resistance to the anti-invasive effects of PF-04691502. Pre-treatment of MDA-MB-231-1833 with PF-04691502 significantly impaired metastatic tumor formation in vivo, despite lack of antiproliferative effects in culture and little effect on primary orthotopic tumor growth. A further link between cytoplasmic p27 and metastasis was provided by a study of primary human breast cancers which showed cytoplasmic p27 is associated with increased lymph nodal metastasis and reduced survival. Novel PI3K/mTOR inhibitors may oppose tumor metastasis independent of their growth inhibitory effects, providing a rationale for clinical investigation of PI3K/mTOR inhibitors in settings to prevent micrometastasis. In primary human breast cancers, cytoplasmic p27 is associated with worse outcomes and increased nodal metastasis, and may prove useful as a marker of both PI3K/mTOR activation and PI3K/mTOR inhibitor efficacy. Electronic supplementary material The online version of this article (doi:10.1007/s10549-012-2389-6) contains supplementary material, which is available to authorized users.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.