1
|
Tanaka Y, Amano T, Nakamura A, Yoshino F, Takebayashi A, Takahashi A, Yamanaka H, Inatomi A, Hanada T, Yoneoka Y, Tsuji S, Murakami T. Rapamycin prevents cyclophosphamide-induced ovarian follicular loss and potentially inhibits tumour proliferation in a breast cancer xenograft mouse model. Hum Reprod 2024:deae085. [PMID: 38734930 DOI: 10.1093/humrep/deae085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 03/26/2024] [Indexed: 05/13/2024] Open
Abstract
STUDY QUESTION To what extent and via what mechanism does the concomitant administration of rapamycin (a follicle activation pathway inhibitor and antitumour agent) and cyclophosphamide (a highly toxic ovarian anticancer agent) prevent cyclophosphamide-induced ovarian reserve loss and inhibit tumour proliferation in a breast cancer xenograft mouse model? SUMMARY ANSWER Daily concomitant administration of rapamycin and a cyclic regimen of cyclophosphamide, which has sufficient antitumour effects as a single agent, suppressed cyclophosphamide-induced primordial follicle loss by inhibiting primordial follicle activation in a breast cancer xenograft mouse model, suggesting the potential of an additive inhibitory effect against tumour proliferation. WHAT IS KNOWN ALREADY Cyclophosphamide stimulates primordial follicles by activating the mammalian target of the rapamycin (mTOR) pathway, resulting in the accumulation of primary follicles, most of which undergo apoptosis. Rapamycin, an mTOR inhibitor, regulates primordial follicle activation and exhibits potential inhibitory effects against breast cancer cell proliferation. STUDY DESIGN, SIZE, DURATION To assess ovarian follicular apoptosis, 3 weeks after administering breast cancer cells, 8-week-old mice were randomized into three treatment groups: control, cyclophosphamide, and cyclophosphamide + rapamycin (Cy + Rap) (n = 5 or 6 mice/group). Mice were treated with rapamycin or vehicle control for 1 week, followed by a single dose of cyclophosphamide or vehicle control. Subsequently, the ovaries were resected 24 h after cyclophosphamide administration (short-term treatment groups). To evaluate follicle abundance and the mTOR pathway in ovaries, as well as the antitumour effects and impact on the mTOR pathway in tumours, 8-week-old xenograft breast cancer transplanted mice were randomized into three treatment groups: vehicle control, Cy, and Cy + Rap (n = 6 or 7 mice/group). Rapamycin (5 mg/kg) or the vehicle was administered daily for 29 days. Cyclophosphamide (120 mg/kg) or the vehicle was administered thrice weekly (long-term treatment groups). The tumour diameter was measured weekly. Seven days after the last cyclophosphamide treatment, the ovaries were harvested, fixed, and sectioned (for follicle counting) or frozen (for further analysis). Similarly, the tumours were resected and fixed or frozen. PARTICIPANTS/MATERIALS, SETTING, METHODS Terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) was performed to examine ovarian follicular apoptosis in the short-term treatment groups. All subsequent experiments were conducted in the long-term treatment groups. Tumour growth was evaluated using the tumour volume index. The tumour volume index indicates the relative volume, compared to the volume 3 weeks after tumour cell injection (at treatment initiation) set to 100%. Tumour cell proliferation was evaluated by Ki-67 immunostaining. Activation of the mTOR pathway in tumours was assessed using the protein extracts from tumours and analysed by western blotting. Haematoxylin and eosin staining of ovaries was used to perform differential follicle counts for primordial, primary, secondary, antral, and atretic follicles. Activation of the mTOR pathway in ovaries was assessed using protein extracts from whole ovaries and analysed by western blotting. Localization of mTOR pathway activation within ovaries was assessed by performing anti-phospho-S6 kinase (downstream of mTOR pathway) immunohistochemistry. MAIN RESULTS AND THE ROLE OF CHANCE Ovaries of the short-term treatment groups were resected 24 h after cyclophosphamide administration and subjected to TUNEL staining of apoptotic cells. No TUNEL-positive primordial follicles were detected in the control, Cy, and Cy + Rap groups. Conversely, many granulosa cells of growing follicles were TUNEL positive in the Cy group but negative in the control and Cy + Rap groups. All subsequent experimental results were obtained from the long-term treatment groups. The tumour volume index stabilized at a mean of 160-200% in the Cy group and 130% in the Cy + Rap group throughout the treatment period. In contrast, tumours in the vehicle control group grew continuously with a mean tumour volume index of 600%, significantly greater than that of the two treatment groups. Based on the western blot analysis of tumours, the mTOR pathway was activated in the vehicle control group and downregulated in the Cy + Rap group when compared with the control and Cy groups. Ki-67 immunostaining of tumours showed significant inhibition of cell proliferation in the Cy + Rap group when compared with that in the control and Cy groups. The ovarian follicle count revealed that the Cy group had significantly fewer primordial follicles (P < 0.001) than the control group, whereas the Cy + Rap group had significantly higher number of primordial follicles (P < 0.001, 2.5 times) than the Cy group. The ratio of primary to primordial follicles was twice as high in the Cy group than in the control group; however, no significant difference was observed between the control group and the Cy + Rap group. Western blot analysis of ovaries revealed that the mTOR pathway was activated by cyclophosphamide and inhibited by rapamycin. The phospho-S6 kinase (pS6K)-positive primordial follicle rate was 2.7 times higher in the Cy group than in the control group. However, this effect was suppressed to a level similar to the control group in the Cy + Rap group. LARGE SCALE DATA None. LIMITATIONS, REASONS FOR CAUTION The combinatorial treatment of breast cancer tumours with rapamycin and cyclophosphamide elicited inhibitory effects on cell proliferative potential compared to cyclophosphamide monotherapy. However, no statistically significant additive effect was observed on tumour volume. Thus, the beneficial antitumour effect afforded by rapamycin administration on breast cancer could not be definitively proven. Although rapamycin has ovarian-protective effects, it does not fully counteract the ovarian toxicity of cyclophosphamide. Nevertheless, rapamycin is advantageous as an ovarian protective agent as it can be used in combination with other ovarian protective agents, such as hormonal therapy. Hence, in combination with other agents, mTOR inhibitors may be sufficiently ovario-protective against high-dose and cyclic cyclophosphamide regimens. WIDER IMPLICATIONS OF THE FINDINGS Compared with a cyclic cyclophosphamide regimen that replicates human clinical practice under breast cancer-bearing conditions, the combination with rapamycin mitigates the ovarian follicle loss of cyclophosphamide without interfering with the anticipated antitumour effects. Hence, rapamycin may represent a new non-invasive treatment option for cyclophosphamide-induced ovarian dysfunction in breast cancer patients. STUDY FUNDING/COMPETING INTEREST(S) This work was not financially supported. The authors declare that they have no conflict of interest.
Collapse
Affiliation(s)
- Yuji Tanaka
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Tsukuru Amano
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Akiko Nakamura
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Fumi Yoshino
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Akie Takebayashi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Akimasa Takahashi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Hiroyuki Yamanaka
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Ayako Inatomi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Tetsuro Hanada
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Yutaka Yoneoka
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Shunichiro Tsuji
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Takashi Murakami
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
2
|
Parikh DA, Kody L, Brain S, Heditsian D, Lee V, Curtis C, Karin MR, Wapnir IL, Patel MI, Sledge GW, Caswell-Jin JL. Patient perspectives on window of opportunity clinical trials in early-stage breast cancer. Breast Cancer Res Treat 2022; 194:171-178. [PMID: 35538268 PMCID: PMC9090598 DOI: 10.1007/s10549-022-06611-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/13/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE Window of opportunity trials (WOT) are increasingly common in oncology research. In WOT participants receive a drug between diagnosis and anti-cancer treatment, usually for the purpose of investigating that drugs effect on cancer biology. This qualitative study aimed to understand patient perspectives on WOT. METHODS We recruited adults diagnosed with early-stage breast cancer awaiting definitive therapy at a single-academic medical center to participate in semi-structured interviews. Thematic and content analyses were performed to identify attitudes and factors that would influence decisions about WOT participation. RESULTS We interviewed 25 women diagnosed with early-stage breast cancer. The most common positive attitudes toward trial participation were a desire to contribute to research and a hope for personal benefit, while the most common concerns were the potential for side effects and how they might impact fitness for planned treatment. Participants indicated family would be an important normative factor in decision-making and, during the COVID-19 pandemic, deemed the absence of family members during clinic visits a barrier to enrollment. Factors that could hinder participation included delay in standard treatment and the requirement for additional visits or procedures. Ultimately, most interviewees stated they would participate in a WOT if offered (N = 17/25). CONCLUSION In this qualitative study, interviewees weighed altruism and hypothetical personal benefit against the possibility of side effect from a WOT. In-person family presence during trial discussion, challenging during COVID-19, was important for many. Our results may inform trial design and communication approaches in future window of opportunity efforts.
Collapse
Affiliation(s)
- Divya A Parikh
- Department of Medicine, Stanford University, 875 Blake Wilbur Drive, Stanford, CA, 94305, USA.
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Lisa Kody
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Susie Brain
- Department of Medicine, Stanford University, 875 Blake Wilbur Drive, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Diane Heditsian
- Department of Medicine, Stanford University, 875 Blake Wilbur Drive, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Vivian Lee
- Department of Medicine, Stanford University, 875 Blake Wilbur Drive, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Christina Curtis
- Department of Medicine, Stanford University, 875 Blake Wilbur Drive, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Mardi R Karin
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - Irene L Wapnir
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - Manali I Patel
- Department of Medicine, Stanford University, 875 Blake Wilbur Drive, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - George W Sledge
- Department of Medicine, Stanford University, 875 Blake Wilbur Drive, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Jennifer L Caswell-Jin
- Department of Medicine, Stanford University, 875 Blake Wilbur Drive, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
3
|
El Khoury A, Butchakdjian ZM, Lagha E, Semaan P, Soueidy M. Acute Multiple In-Stent Thromboses in a Patient With Clopidogrel-Fluconazole Interaction. Cureus 2022; 14:e23718. [PMID: 35510000 PMCID: PMC9060737 DOI: 10.7759/cureus.23718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2022] [Indexed: 11/12/2022] Open
Abstract
Clopidogrel is an anti-platelet that exerts its function by selectively inhibiting the binding of adenosine di-phosphate (ADP) to the P2Y12 receptor. Fluconazole is a fungistatic agent that alters fungal cell membranes. Both of these drugs act on the cytochrome P450 2C19. We report the case of an 83-year-old male that presented two days following coronary angioplasty with stent thrombosis, following the concomitant use of clopidogrel and fluconazole. We aim to study the interaction between clopidogrel and fluconazole. We hypothesize that fluconazole decreases the therapeutic level of clopidogrel, requiring an increase in dosage to achieve the same anti-thrombotic effect.
Collapse
|
4
|
Chang GR, Hou PH, Wang CM, Wu CF, Su HK, Liao HJ, Chen TP. Chronic everolimus treatment of high-fat diet mice leads to a reduction in obesity but impaired glucose tolerance. Pharmacol Res Perspect 2021; 9:e00732. [PMID: 33715287 PMCID: PMC7955951 DOI: 10.1002/prp2.732] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
Everolimus, which inhibits mTOR kinase activity and is clinically used in graft rejection treatment, may have a two‐sided influence on metabolic syndrome; its role in obesity and hyperglycemic in animals and humans, however, has been explored insufficiently. This study further determined how continual everolimus treatment affects glucose homeostasis and body weight control in C57BL6/J mice with obesity. An obesity mouse model was developed by administering a high‐fat diet (HFD) to C57BL6/J mice over 12 weeks. The experimental group, while continuing their HFD consumption, were administered everolimus daily for 8 weeks. Metabolic parameters, glucose tolerance, fatty liver score, endocrine profile, insulin sensitivity index (ISI), insulin resistance (IR) index, and Akt phosphorylation, GLUT4, TNF‐α, and IL‐1 levels were measured in vivo. Compared with the control group, the everolimus group gained less body weight and had smaller adipocytes and lower fat pad weight; triglyceride (serum and hepatic), patatin‐like phospholipase domain‐containing 3, and fatty acid synthase levels; fatty liver scores; and glucose tolerance test values—all despite consuming more food. However, the everolimus group exhibited decreased ISI and muscle Akt phosphorylation and GLUT4 expression as well as impaired glucose tolerance and serum TNF‐α and IL‐1β levels—even when insulin levels were high. In conclusion, continual everolimus treatment may lead to diabetes with glucose intolerance and IR.
Collapse
Affiliation(s)
- Geng-Ruei Chang
- Department of Veterinary Medicine, National Chiayi University, Chiayi, Taiwan
| | - Po-Hsun Hou
- Department of Psychiatry, Taichung Veterans General Hospital, Taichung, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chao-Min Wang
- Department of Veterinary Medicine, National Chiayi University, Chiayi, Taiwan
| | - Ching-Feng Wu
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung University, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Huang-Kai Su
- Department of Veterinary Medicine, National Chiayi University, Chiayi, Taiwan
| | - Huei-Jyuan Liao
- Department of Veterinary Medicine, National Chiayi University, Chiayi, Taiwan
| | - To-Pang Chen
- Division of Endocrinology and Metabolism, Show Chwan Memorial Hospital, Changhua, Taiwan
| |
Collapse
|
5
|
Iqbal MA, Siddiqui S, Ur Rehman A, Siddiqui FA, Singh P, Kumar B, Saluja D. Multiomics integrative analysis reveals antagonistic roles of CBX2 and CBX7 in metabolic reprogramming of breast cancer. Mol Oncol 2021; 15:1450-1465. [PMID: 33400401 PMCID: PMC8096797 DOI: 10.1002/1878-0261.12894] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/07/2020] [Accepted: 12/31/2020] [Indexed: 12/24/2022] Open
Abstract
Striking similarity exists between metabolic changes associated with embryogenesis and tumorigenesis. Chromobox proteins‐CBX2/4/6/7/8, core components of canonical polycomb repressor complex 1, play essential roles in embryonic development and aberrantly expressed in breast cancer. Understanding how altered CBX expression relates to metabolic reprogramming in breast cancer may reveal vulnerabilities of therapeutic pertinence. Using transcriptomic and metabolomic data from breast cancer patients (N > 3000 combined), we performed pathway‐based analysis and identified outstanding roles of CBX2 and CBX7 in positive and negative regulation of glucose metabolism, respectively. Genetic ablation experiments validated the contrasting roles of two isoforms in cancer metabolism and cell growth. Furthermore, we provide evidence for the role of mammalian target of rapamycin complex 1 signaling in mediating contrary effects of CBX2 and CBX7 on breast cancer metabolism. Underpinning the biological significance of metabolic roles, CBX2 and CBX7 were found to be the most up‐ and downregulated isoforms, respectively, in breast tumors compared with normal tissues. Moreover, CBX2 and CBX7 expression (not other isoforms) correlated strongly, but oppositely, with breast tumor subtype aggressiveness and the proliferation markers. Consistently, genomic data also showed higher amplification frequency of CBX2, not CBX7, in breast tumors. Highlighting the clinical significance of findings, disease‐specific survival and drug sensitivity analysis revealed that CBX2 and CBX7 predicted patient outcome and sensitivity to FDA‐approved/investigational drugs. In summary, this work identifies novel cross talk between CBX2/7 and breast tumor metabolism, and the results presented may have implications in strategies targeting breast cancer.
Collapse
Affiliation(s)
- Mohammad Askandar Iqbal
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi, India
| | - Shumaila Siddiqui
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi, India
| | - Asad Ur Rehman
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, India
| | - Farid Ahmad Siddiqui
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi, India.,Turku Centre for Biotechnology, BioCity, University of Turku and Abo Akademi, Finland
| | - Prithvi Singh
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi, India.,Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia (A Central University), New Delhi, India
| | - Bhupender Kumar
- Department of Biochemistry, Institute of Home Economics, University of Delhi, India
| | - Daman Saluja
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, India
| |
Collapse
|
6
|
Schettini F, Sobhani N, Ianza A, Triulzi T, Molteni A, Lazzari MC, Strina C, Milani M, Corona SP, Sirico M, Bernocchi O, Giudici F, Cappelletti MR, Ciruelos E, Jerusalem G, Loi S, Fox SB, Generali D. Immune system and angiogenesis-related potential surrogate biomarkers of response to everolimus-based treatment in hormone receptor-positive breast cancer: an exploratory study. Breast Cancer Res Treat 2020; 184:421-431. [PMID: 32770287 PMCID: PMC7599144 DOI: 10.1007/s10549-020-05856-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/31/2020] [Indexed: 01/08/2023]
Abstract
Purpose mTOR inhibitor everolimus is used for hormone receptor-positive (HR+)/HER2-negative metastatic breast cancer (mBC). No reliable predictive biomarker of response is available. Following evidences from other solid tumors, we aimed to assess the association between treatment-associated immune system features and everolimus activity. Methods We retrospectively explored a correlation with the therapeutic activity of everolimus and tumor-associated immune pathways with ingenuity pathway analysis (IPA), neutrophil-to-lymphocyte ratio (NLR), circulating lymphocytes, and endothelial cells (CECs) in 3 different HR+ mBC studies, including the BALLET phase IIIb study. Results The circulating levels of CD3+/CD8+, CD3+/CD4+, and overall T lymphocytes were higher in responders versus non-responders at baseline (p = 0.017, p < 0.001, p = 0.034) and after treatment (p = 0.01, p = 0.003, p = 0.023). Reduced CECs, a tumor neoangiogenesis marker, were observed in responders after treatment (p < 0.001). Patients with low NLR (≤ 4.4) showed a better progression-free survival compared to patients with high NLR (> 4.4) (p = 0.01). IPA showed that the majority of immunity-related genes were found upregulated in responders compared to non-responders before treatment, but not after. Conclusions Lymphocytes subpopulations, CECs and NLR could be interesting biomarkers predictive of response to everolimus-based regimens, potentially useful in daily clinical practice to select/monitor everolimus-based treatment in mBC. Further studies to confirm such hypotheses are warranted.
Collapse
Affiliation(s)
- Francesco Schettini
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy.,Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Navid Sobhani
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Anna Ianza
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alfredo Molteni
- UO Ematologia e CTMO, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | | | - Carla Strina
- UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Manuela Milani
- UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Silvia Paola Corona
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Marianna Sirico
- UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Ottavia Bernocchi
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy.,UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Fabiola Giudici
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Maria Rosaria Cappelletti
- UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Eva Ciruelos
- SOLTI Breast Cancer Research Group, Barcelona, Spain.,Department of Medical Oncology, Breast Cancer Unit, University Hospital, 12 de Octubre, Avda de Córdoba s/n, Madrid, Spain
| | - Guy Jerusalem
- Department of Medical Oncology, Centre Hospitalier Universitaire de Liège and Liège University, Avenue de L'Hòpital 1, 4000, Liège, Belgium
| | - Sherine Loi
- Peter MacCallum Cancer Center, 305 Grattan Street, Melbourne, VIC, Australia.,University of Melbourne, Melbourne, VIC, Australia
| | - Stephen B Fox
- Peter MacCallum Cancer Center, 305 Grattan Street, Melbourne, VIC, Australia.,University of Melbourne, Melbourne, VIC, Australia
| | - Daniele Generali
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy. .,UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy.
| |
Collapse
|
7
|
Influences of preoperative metformin on immunological factors in early breast cancer. Cancer Chemother Pharmacol 2020; 86:55-63. [PMID: 32533334 PMCID: PMC7338817 DOI: 10.1007/s00280-020-04092-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 06/03/2020] [Indexed: 10/27/2022]
Abstract
PURPOSE Metformin has been suggested to possibly reduce cancer risk. However, the mechanism underlying the positive effects of metformin on cancer treatment remains unclear. We conducted a prospective study to evaluate the effects of preoperative metformin in patients with early breast cancer. METHOD We evaluated the effects on immunological factors (TILs, CD4 + , CD8 + , PD-L1, IFNγ and IL-2) by comparing core needle biopsies (CNB) obtained before metformin treatment with surgical specimens. Seventeen patients were enrolled in this prospective study from January to December 2016. We also analyzed 59 patients undergoing surgery during the same period to reveal the correlation of immune factors between CNB and surgical specimen. RESULT There was a moderate correlation between CNB and surgical specimens on TILs and CD8 + lymphocyte. (TILs Rs = 0.63, CD4 + Rs = 0.224, CD8 + Rs = 0.42) In the metformin group, TILs increases were confirmed in five (29%) patients, while a decrease was confirmed in two (12%). The expressions of CD4 + and CD8 + by TILs were increased in 41% and 18% of surgical specimens, respectively. However, TILs number (p = 0.0554), CD4+ (p = 0.0613) and CD8 + (p = 0.0646) expressions did not significantly increased. Furthermore, IFNγ expression appeared to be increased in response to metformin (p = 0.08). CONCLUSION Preoperative metformin tends to increase TILs, as well as the numbers of CD4 and CD8 positive lymphocytes, and IFNγ levels. Metformin might improve immune function and have a possibility of chemo-sensitivity and thereby increase the effectiveness of immunotherapy, based on the results of this preliminary study.
Collapse
|
8
|
Ortega MA, Fraile-Martínez O, Asúnsolo Á, Buján J, García-Honduvilla N, Coca S. Signal Transduction Pathways in Breast Cancer: The Important Role of PI3K/Akt/mTOR. JOURNAL OF ONCOLOGY 2020; 2020:9258396. [PMID: 32211045 PMCID: PMC7085392 DOI: 10.1155/2020/9258396] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/25/2019] [Accepted: 01/11/2020] [Indexed: 12/15/2022]
Abstract
Breast cancer is the cancer with the highest prevalence in women and is the number-one cause of cancer mortality worldwide. Cell transduction is a fundamental process in the development and progression of cancer. Modifications in various cell signalling pathways promote tumour cell proliferation, progression, and survival. The PI3K/Akt/mTOR pathway is an example of that, and it is involved in growth, proliferation, survival, motility, metabolism, and immune response regulation. Activation of this pathway is one of the main causes of cancer cell resistance to antitumour therapies. This makes PI3K/Akt/mTOR signalling a crucial object of study for understanding the development and progression of this disease. Thus, this pathway may have a role as a potential therapeutic target, as well as prognostic and diagnostic value, in patients with breast cancer. Despite the existence of selective PI3K/Akt/mTOR pathway inhibitors and current clinical trials, the cellular mechanisms are not yet known. The present review aims to understand the current state of this important disease and the paths that must be forged.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences and Networking Biomedical Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Spain
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences and Networking Biomedical Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Ángel Asúnsolo
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Julia Buján
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences and Networking Biomedical Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences and Networking Biomedical Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Santiago Coca
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences and Networking Biomedical Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| |
Collapse
|
9
|
Aggelis V, Johnston SRD. Advances in Endocrine-Based Therapies for Estrogen Receptor-Positive Metastatic Breast Cancer. Drugs 2019; 79:1849-1866. [DOI: 10.1007/s40265-019-01208-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
10
|
Ishida N, Baba M, Hatanaka Y, Hagio K, Okada H, Hatanaka KC, Togashi K, Matsuno Y, Yamashita H. PIK3CA mutation, reduced AKT serine 473 phosphorylation, and increased ERα serine 167 phosphorylation are positive prognostic indicators in postmenopausal estrogen receptor-positive early breast cancer. Oncotarget 2018; 9:17711-17724. [PMID: 29707142 PMCID: PMC5915150 DOI: 10.18632/oncotarget.24845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 02/28/2018] [Indexed: 12/12/2022] Open
Abstract
Although endocrine therapy is the most important treatment option in estrogen receptor (ER)-positive breast cancer, new strategies, such as molecular targeted agents together with endocrine therapy are required to improve survival. PIK3CA is the most frequent mutated gene in ER-positive early breast cancers, and PIK3CA mutation status is reported to affect activation of AKT and ERα. Moreover, recent studies demonstrate that patients had a better prognosis when tumors expressed ER, androgen receptor (AR), and vitamin D receptor (VDR). In this study, we examined expression of AR and VDR, phosphorylation of AKT serine (Ser) 473 (AKT phospho-Ser473) and ERα Ser167 (ERα phospho-Ser167) by immunohistochemistry in ER-positive, HER2-negative early breast cancer. PIK3CA gene mutations were also detected in genomic DNA extracted from tumor blocks. Correlations between these biological markers, clinicopathological factors and prognosis were analyzed. Levels of AKT phospho-Ser473 were significantly higher in premenopausal women than in postmenopausal women. In contrast, AR expression was significantly higher in postmenopausal women than in premenopausal women. PIK3CA mutations were detected in 47% in premenopausal women and 47% in postmenopausal women. Postmenopausal women with PIK3CA wild-type tumors had significantly worse disease-free survival than patients with PIK3CA mutant tumors. Low levels of AKT phospho-Ser473 and high levels of ERα phospho-Ser167 were strongly associated with increased disease-free survival in postmenopausal women. Evaluation of ERα activation, in addition to PIK3CA mutation status, might be helpful in identifying patients who are likely to benefit from endocrine therapy alone versus those who are not in postmenopausal ER-positive early breast cancer.
Collapse
Affiliation(s)
- Naoko Ishida
- Department of Breast Surgery, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Motoi Baba
- Department of Breast Surgery, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Yutaka Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
- Research Division of Companion Diagnostics, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Kanako Hagio
- Department of Breast Surgery, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Hiromi Okada
- Department of Surgical Pathology, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Kanako C. Hatanaka
- Research Division of Companion Diagnostics, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Kenichi Togashi
- Roche Diagnostics K.K., Konan, Minato-ku, Tokyo 108-0075, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
- Research Division of Companion Diagnostics, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| | - Hiroko Yamashita
- Department of Breast Surgery, Hokkaido University Hospital, Kita-ku, Sapporo 060-8648, Japan
| |
Collapse
|
11
|
Du L, Li X, Zhen L, Chen W, Mu L, Zhang Y, Song A. Everolimus inhibits breast cancer cell growth through PI3K/AKT/mTOR signaling pathway. Mol Med Rep 2018; 17:7163-7169. [PMID: 29568883 PMCID: PMC5928673 DOI: 10.3892/mmr.2018.8769] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 10/30/2017] [Indexed: 01/14/2023] Open
Abstract
Breast cancer is one of the most prevalent malignancies and the leading cause of cancer‑associated mortality in women worldwide and in China. Everolimus (C53H83NO14) is an efficient anti-cancer drug for breast cancer which targets mammalian target of rapamycin (mTOR). The present study investigated the inhibitory effects of everolimus on breast cancer cells and an MCF‑7‑bearing mouse model. The potential mechanism of the everolimus‑mediated decrease in growth and aggressiveness of breast cancer cells was reported. Results demonstrated that everolimus significantly inhibited breast cancer cell growth, migration and invasion. It was demonstrated that everolimus induced apoptosis through decreasing B cell lymphoma (Bcl)‑2 and Bcl‑w and increasing caspase‑3 and caspase‑8 expression levels in breast cancer cells. It was observed that everolimus decreased phosphoinositide 3‑kinase (PI3K), protein kinase B (AKT) and mTOR expression levels in breast cancer cells. Results additionally demonstrated that PI3 K overexpression prevented that everolimus‑mediated inhibition of growth and aggressiveness in MCF‑7 cells. In vivo assays demonstrated that everolimus treatment markedly inhibited tumor growth in the MCF‑7 bearing mouse model. Overall, these data indicate that everolimus inhibits growth and aggressiveness of breast cancer cells through the PI3K/AKT/mTOR signaling pathways, suggesting the PI3K/AKT/mTOR signaling pathway may act as a therapeutic target for the treatment of human cancer.
Collapse
Affiliation(s)
- Liyan Du
- Department of Breast Surgery, Xingtai First Hospital, Xingtai, Hebei 054001, P.R. China
| | - Xiaomei Li
- Department of Breast Surgery, Xingtai First Hospital, Xingtai, Hebei 054001, P.R. China
| | - Linhong Zhen
- Department of Breast Surgery, Xingtai First Hospital, Xingtai, Hebei 054001, P.R. China
| | - Weiling Chen
- Department of Breast Surgery, Xingtai First Hospital, Xingtai, Hebei 054001, P.R. China
| | - Lingguang Mu
- Department of Breast Surgery, Xingtai First Hospital, Xingtai, Hebei 054001, P.R. China
| | - Yang Zhang
- Department of Breast Surgery, Xingtai First Hospital, Xingtai, Hebei 054001, P.R. China
| | - Ailin Song
- Department of Breast Surgery, Xingtai First Hospital, Xingtai, Hebei 054001, P.R. China
| |
Collapse
|
12
|
Schmid P, Pinder SE, Purushotham A, Thompson AM. Reply to R.F. Sweis et al. J Clin Oncol 2017; 35:261-262. [PMID: 28056198 DOI: 10.1200/jco.2016.69.8019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Peter Schmid
- Peter Schmid, Queen Mary University London, London, United Kingdom; Sarah E. Pinder and Arnie Purushotham, King's College London; Guys & St Thomas NHS Trust, London, United Kingdom; and Alastair M. Thompson, MD Anderson Cancer Center, Houston, TX
| | - Sarah E Pinder
- Peter Schmid, Queen Mary University London, London, United Kingdom; Sarah E. Pinder and Arnie Purushotham, King's College London; Guys & St Thomas NHS Trust, London, United Kingdom; and Alastair M. Thompson, MD Anderson Cancer Center, Houston, TX
| | - Arnie Purushotham
- Peter Schmid, Queen Mary University London, London, United Kingdom; Sarah E. Pinder and Arnie Purushotham, King's College London; Guys & St Thomas NHS Trust, London, United Kingdom; and Alastair M. Thompson, MD Anderson Cancer Center, Houston, TX
| | - Alastair M Thompson
- Peter Schmid, Queen Mary University London, London, United Kingdom; Sarah E. Pinder and Arnie Purushotham, King's College London; Guys & St Thomas NHS Trust, London, United Kingdom; and Alastair M. Thompson, MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
13
|
Neoadjuvant systemic therapy in breast cancer: Challenges and uncertainties. Eur J Obstet Gynecol Reprod Biol 2016; 210:144-156. [PMID: 28039758 DOI: 10.1016/j.ejogrb.2016.12.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 11/20/2016] [Accepted: 12/13/2016] [Indexed: 12/29/2022]
Abstract
The management of locally advanced breast cancer (LABC) remains a major clinical issue, despite progress achieved in diagnosis and therapy. Preoperative or neoadjuvant therapy has gained interest since breast cancer has been regarded as a systemic disease. Comparing adjuvant versus neoadjuvant treatment, the neoadjuvant approach offers the advantage of downstaging the disease and testing the efficacy of therapy administered to patients. A large number of clinical trials have attempted to define the optimal neoadjuvant treatment, but little attention has been paid to the sequence of chemotherapy. Moreover, the integration of antibodies against Human Epidermal Receptor-2 (HER-2) and other biological therapies that may improve the long-term control of breast cancer patients, have a special clinical interest. In this review, we will discuss these topics attempting to answer the questions why, when and which regimen to use for patients with LABC. Especially, the introduction of the platina derivatives in neoadjuvant trials with their exceptional high pathological complete response rates are challenging to rethink the optimal treatment options in early and locally advanced breast cancer.
Collapse
|
14
|
Hadad SM, Jordan LB, Roy PG, Purdie CA, Iwamoto T, Pusztai L, Moulder-Thompson SL, Thompson AM. A prospective comparison of ER, PR, Ki67 and gene expression in paired sequential core biopsies of primary, untreated breast cancer. BMC Cancer 2016; 16:745. [PMID: 27658825 PMCID: PMC5034430 DOI: 10.1186/s12885-016-2788-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/15/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Sequential biopsy of breast cancer is used to assess biomarker effects and drug efficacy. The preoperative "window of opportunity" setting is advantageous to test biomarker changes in response to therapeutic agents in previously untreated primary cancers. This study tested the consistency over time of paired, sequential biomarker measurements on primary, operable breast cancer in the absence of drug therapy. METHODS Immunohistochemistry was performed for ER, PR and Ki67 on paired preoperative/operative tumor samples taken from untreated patients within 2 weeks of each other. Microarray analysis on mRNA extracted from formalin fixed paraffin embedded cores was performed using Affymetrix based arrays on paired core biopsies analysed using Ingenuity Pathway Analysis (IPA) and Gene Set Analysis (GSA). RESULTS In 41 core/resection pairs, the recognised trend to lower ER, PR and Ki67 score on resected material was confirmed. Concordance for ER, PR and Ki67 without changing biomarker status (e.g. ER+ to ER-) was 90, 74 and 80 % respectively. However, in 23 paired core samples (diagnostic core v on table core), Ki67 using a cut off of 13.25 % was concordant in 22/23 (96 %) and differences in ER and PR immunohistochemistry by Allred or Quickscore between the pairs did not impact hormone receptor status. IPA and GSA demonstrated substantial gene expression changes between paired cores at the mRNA level, including reduced expression of ER pathway analysis on the second core, despite the absence of drug intervention. CONCLUSIONS Sequential core biopsies of primary breast cancer (but not core versus resection) was consistent and is appropriate to assess the effects of drug therapy in vivo on ER, PR and Ki67 using immunohistochemistry. Conversely, studies utilising mRNA expression may require non-treatment controls to distinguish therapeutic from biopsy differences.
Collapse
Affiliation(s)
| | - Lee B. Jordan
- Department of Pathology, Ninewells Hospital and Medical School, Dundee, DD1 9SY UK
| | | | - Colin A. Purdie
- Department of Pathology, Ninewells Hospital and Medical School, Dundee, DD1 9SY UK
| | - Takayuki Iwamoto
- Department of Breast and Endocrine Surgery, Okayama University, Okayama, Japan
| | - Lajos Pusztai
- Yale Medical Oncology, PO Box 208028, New Haven, 06520 CT USA
| | - Stacy L. Moulder-Thompson
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Houston, 77030 TX USA
| | - Alastair M. Thompson
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Houston, 77030 TX USA
| |
Collapse
|
15
|
Študentová H, Vitásková D, Melichar B. Safety of mTOR inhibitors in breast cancer. Expert Opin Drug Saf 2016; 15:1075-85. [DOI: 10.1080/14740338.2016.1192604] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Hana Študentová
- Department of Oncology, Palacký University Medical School & Teaching Hospital, Olomouc, Czech Republic
| | - Denisa Vitásková
- Department of Oncology, Palacký University Medical School & Teaching Hospital, Olomouc, Czech Republic
| | - Bohuslav Melichar
- Department of Oncology, Palacký University Medical School & Teaching Hospital, Olomouc, Czech Republic
- Institute of Molecular and Translational Medicine, Palacký University Medical School & Teaching Hospital, Olomouc, Czech Republic
| |
Collapse
|
16
|
Schmid P, Pinder SE, Wheatley D, Macaskill J, Zammit C, Hu J, Price R, Bundred N, Hadad S, Shia A, Sarker SJ, Lim L, Gazinska P, Woodman N, Korbie D, Trau M, Mainwaring P, Gendreau S, Lackner MR, Derynck M, Wilson TR, Butler H, Earl G, Parker P, Purushotham A, Thompson A. Phase II Randomized Preoperative Window-of-Opportunity Study of the PI3K Inhibitor Pictilisib Plus Anastrozole Compared With Anastrozole Alone in Patients With Estrogen Receptor-Positive Breast Cancer. J Clin Oncol 2016; 34:1987-94. [PMID: 26976426 DOI: 10.1200/jco.2015.63.9179] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Preclinical data support a key role for the PI3K pathway in estrogen receptor-positive breast cancer and suggest that combining PI3K inhibitors with endocrine therapy may overcome resistance. This preoperative window study assessed whether adding the PI3K inhibitor pictilisib (GDC-0941) can increase the antitumor effects of anastrozole in primary breast cancer and aimed to identify the most appropriate patient population for combination therapy. PATIENTS AND METHODS In this randomized, open-label phase II trial, postmenopausal women with newly diagnosed operable estrogen receptor-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancers were recruited. Participants were randomly allocated (2:1, favoring the combination) to 2 weeks of preoperative treatment with anastrozole 1 mg once per day (n = 26) or the combination of anastrozole 1 mg with pictilisib 260 mg once per day (n = 49). The primary end point was inhibition of tumor cell proliferation as measured by change in Ki-67 protein expression between tumor samples taken before and at the end of treatment. RESULTS There was significantly greater geometric mean Ki-67 suppression of 83.8% (one-sided 95% CI, ≥ 79.0%) for the combination and 66.0% (95% CI, ≤ 75.4%) for anastrozole (geometric mean ratio [combination:anastrozole], 0.48; 95% CI, ≤ 0.72; P = .004). PIK3CA mutations were not predictive of response to pictilisib, but there was significant interaction between response to treatment and molecular subtype (P = .03); for patients with luminal B tumors, the combination:anastrozole geometric mean ratio of Ki-67 suppression was 0.37 (95% CI, ≤ 0.67; P = .008), whereas no significant Ki-67 response was observed for pictilisib in luminal A tumors (1.01; P = .98). Multivariable analysis confirmed Ki-67 response to the combination treatment of patients with luminal B tumors irrespective of progesterone receptor status or baseline Ki-67 expression. CONCLUSION Adding pictilisib to anastrozole significantly increases suppression of tumor cell proliferation in luminal B primary breast cancer.
Collapse
Affiliation(s)
- Peter Schmid
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX.
| | - Sarah E Pinder
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Duncan Wheatley
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Jane Macaskill
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Charles Zammit
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Jennifer Hu
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Robert Price
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Nigel Bundred
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Sirwan Hadad
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Alice Shia
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Shah-Jalal Sarker
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Louise Lim
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Patrycja Gazinska
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Natalie Woodman
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Darren Korbie
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Matt Trau
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Paul Mainwaring
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Steven Gendreau
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Mark R Lackner
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Mika Derynck
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Timothy R Wilson
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Hannah Butler
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Gemma Earl
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Peter Parker
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Arnie Purushotham
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| | - Alastair Thompson
- Peter Schmid, Alice Shia, Shah-Jalal Sarker, and Louise Lim, Queen Mary University London; Sarah E. Pinder, Patrycja Gazinska, Natalie Woodman, Peter Parker, and Arnie Purushotham, Kings College London; Robert Price, Kings College Hospital; Jennifer Hu, Barts Health National Health Service (NHS) Trust, London; Duncan Wheatley, Royal Cornwall Hospital, Truro; Jane Macaskill, Ninewells Hospital Dundee, Dundee; Charles Zammit, Hannah Butler, and Gemma Earl, Brighton & Sussex University Hospitals NHS Trust, Brighton; Nigel Bundred, University Hospital of South Manchester, Manchester; Sirwan Hadad, Royal Hallamshire Sheffield, Sheffield; Darren Korbie and Matt Trau, Australian Institute for Bioengineering and Nanotechnology, and Matt Trau, University of Queensland; Paul Mainwaring, Mater Research Centre; Brisbane, Australia; Steven Gendreau, Mark R. Lackner, Mika Derynck, and Timothy R. Wilson, Genentech, South San Francisco, CA; and Alastair Thompson, MD Anderson Cancer Centre, Houston, TX
| |
Collapse
|
17
|
Izdebska M, Grzanka D, Gagat M, Hałas-Wiśniewska M, Grzanka A. Downregulation of importin-9 protects MCF-7 cells against apoptosis induced by the combination of garlic-derived alliin and paclitaxel. Oncol Rep 2016; 35:3084-93. [PMID: 26934847 DOI: 10.3892/or.2016.4628] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/11/2016] [Indexed: 11/06/2022] Open
Abstract
Numerous studies on the biological mechanism of breast cancer have identified a number of potential therapeutic molecular targets. In this context, one type of potential candidates appears to be agents that target the actin cytoskeleton of cancer cells or regulate actin cytoskeleton dynamics. The aim of the present study was to study the impact of altered actin transport between the cytoplasm and nucleus by the downregulation of importin-9 (IPO9) in breast adenocarcinoma MCF-7 cells exposed to an apoptosis-inducing combination of garlic-derived S-allyl-L-cysteine sulfoxide (alliin) and paclitaxel (PTX). The expression of IPO9 was downregulated by the transfection of non-aggressive breast cancer MCF-7 cells with siRNA against IPO9. The altered expression of IPO9 and cofilin-1 (CFL1) was examined using western blotting. Moreover, the effect of the downregulation of IPO9 on cell death induced by the combination of PTX and alliin was also investigated. The alterations of IPO9 and CFL1 levels were also related with F-actin organizational changes and F-actin fluorescence intensity in the nuclear/perinuclear area of the cells. The results presented here indicate that alliin and PTX act synergistically to promote and potentiate apoptosis in MCF-7 cells. Furthermore, using RNA interference technique, we showed that downregulation of IPO9 expression was correlated with a significant reduction in the apoptotic cell population as well as with a decrease in F-actin content in whole cells, and in the cortical and nuclear/perinuclear areas of the cells. Simultaneously, the downregulation of IPO9 was also accompanied by the increased post-translational expression of CFL1. Furthermore, the data obtained in the present study allow us to conclude that CFL1 itself does not translocate actin into the cell nucleus but this transport requires the functional expression of IPO9.
Collapse
Affiliation(s)
- Magdalena Izdebska
- Department of Histology and Embryology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Faculty of Medicine, 85-092 Bydgoszcz, Poland
| | - Dariusz Grzanka
- Department and Clinic of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Faculty of Medicine, 85-092 Bydgoszcz, Poland
| | - Maciej Gagat
- Department of Histology and Embryology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Faculty of Medicine, 85-092 Bydgoszcz, Poland
| | - Marta Hałas-Wiśniewska
- Department of Histology and Embryology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Faculty of Medicine, 85-092 Bydgoszcz, Poland
| | - Alina Grzanka
- Department of Histology and Embryology, Sexually Transmitted Diseases and Immunodermatology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Faculty of Medicine, 85-092 Bydgoszcz, Poland
| |
Collapse
|
18
|
Han HS, Magliocco AM. Molecular Testing and the Pathologist's Role in Clinical Trials of Breast Cancer. Clin Breast Cancer 2016; 16:166-79. [PMID: 27103546 DOI: 10.1016/j.clbc.2016.02.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 01/11/2016] [Accepted: 02/03/2016] [Indexed: 01/26/2023]
Abstract
Molecular characterization of breast cancer is pivotal for identifying new molecular targets and determining the appropriate treatment choices. Advances in molecular profiling technology have given greater insight into this heterogeneous disease, over and above hormone receptor and human epidermal growth factor receptor 2 status. Agents targeting recently characterized molecular biomarkers are under clinical development; the success of these targeted agents is likely to depend on identifying the patient population most likely to benefit. Therefore, clinical trials of breast cancer often require prescreening for, or stratification by, relevant molecular markers or exploratory analyses of biomarkers that can predict or monitor the response to treatment. Consequently, the role of the pathologist has become increasingly important. The key considerations for pathologists include tissue availability, ownership of archival tissue, type of diagnostic/biomarker test required, method of sample processing, concordance between different tests and testing centers, and tumor heterogeneity. In the present review, we explore how pathology is used in current clinical trials of breast cancer and describe the various technologies available for molecular testing. Furthermore, the factors required for the successful application of pathology in clinical trials of breast cancer and the issues that can arise and how these can be circumvented are discussed.
Collapse
Affiliation(s)
- Hyo Sook Han
- Department of Women's Oncology, Moffitt Cancer Center, Tampa, FL
| | | |
Collapse
|
19
|
Marous M, Bièche I, Paoletti X, Alt M, Razak A, Stathis A, Kamal M, Le Tourneau C. Designs of preoperative biomarkers trials in oncology: a systematic review of the literature. Ann Oncol 2015; 26:2419-28. [DOI: 10.1093/annonc/mdv378] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 08/19/2015] [Indexed: 01/06/2023] Open
|
20
|
Pouget M, Abrial C, Planchat E, Van Praagh I, Arbre M, Kwiatkowski F, Dubray-Longeras P, Devaud H, Dohou J, Herviou P, Mahammedi H, Durando X, Chollet P, Mouret-Reynier MA. Everolimus in Metastatic Breast Cancer: Clinical Experience as a Late Treatment Line. Oncology 2015; 89:319-31. [DOI: 10.1159/000437230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/18/2015] [Indexed: 11/19/2022]
|
21
|
González Espinoza IR, Villarreal Garza C, Juárez León OA, Adel Álvarez LA, Cruz López JC, Téllez Bernal E. Cáncer de mama con receptores hormonales positivos: tratamiento adyuvante, primera línea en cáncer metastásico y nuevas estrategias (inhibición de mTOR). GACETA MEXICANA DE ONCOLOGÍA 2015. [DOI: 10.1016/j.gamo.2015.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
22
|
Peng L, Zhou Y, Ye X, Zhao Q. Treatment-related fatigue with everolimus and temsirolimus in patients with cancer-a meta-analysis of clinical trials. Tumour Biol 2014; 36:643-54. [PMID: 25281033 DOI: 10.1007/s13277-014-2669-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 09/22/2014] [Indexed: 02/08/2023] Open
Abstract
Mammalian target of rapamycin (mTOR) inhibitors, everolimus and temsirolimus, are approved for the treatment of a variety of malignancies. Fatigue has been described with these agents as a common side effect, although the overall incidence and risk remain unclear. We performed a meta-analysis to calculate the overall incidence of fatigue in cancer patients treated with everolimus and temsirolimus and to compare the differences in incidence with placebo. The electronic databases PubMed, Embase, Web of Science, and Cochrane databases were searched for studies to include in the meta-analysis. Eligible studies were phase II and III prospective clinical trials of cancer patients treated with single drug everolimus or temsirolimus with toxicity data on fatigue. Overall incidence rates, relative risk (RR), and 95% confidence intervals (CI) were calculated employing fixed or random effects models depending on the heterogeneity of the included studies. A total of 9,760 patients with a variety of malignancies from 56 prospective clinical trials were included for the meta-analysis. The overall incidences of all-grade and high-grade fatigue in cancer patients treated with mTOR inhibitor (everolimus or temsirolimus) were 45.4% (95% CI 36.9-55.8%) and 8.7% (95% CI 7.2-10.4%), respectively. The relative risks of fatigue of mTOR inhibitor compared to placebo were increased for all-grade (RR = 1.22, 95% CI 1.08-1.38, P = 0.002) and high-grade (RR = 1.82, 95% CI 1.24-2.69, P = 0.002) fatigue. The incidence of all-grade fatigue of patients treated with everolimus was higher than those with temsirolimus (RR = 1.85, 95% CI 1.71-2.01, P < 0.001). No significant difference was detected with between everolimus and temsirolimus in terms of high-grade fatigue (RR = 1.15, 95% CI 0.94-1.41, P = 0.18). Treatment with mTOR inhibitor, everolimus and temsirolimus, is associated with an increased incidence of fatigue in patients with cancer. Early detection and management of fatigue is needed.
Collapse
Affiliation(s)
- Ling Peng
- Department of Thoracic Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | | | | | | |
Collapse
|
23
|
[Exemestane-everolimus in HER2-negative, hormonal receptor-positive, post-menopausal metastatic breast cancer with resistance to non-steroidal aromatase inhibitor: a new option]. Bull Cancer 2014; 101:325-33. [PMID: 24691195 DOI: 10.1684/bdc.2014.1910] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Phosphatidylinositol-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is commonly deregulated in breast cancer and has been involved in resistance to endocrine therapy. In the BOLERO-2 study, the addition of everolimus, a selective inhibitor of mTOR protein, to exemestane was associated with a significant improvement in progression-free survival, compared to exemestane plus placebo, in patients with hormone receptor-positive, HER2-negative metastatic breast cancer, and resistant to non-steroidal aromatase inhibitor therapy. However, adverse events and treatment stops were more often observed with the combination therapy, suggesting the need for a careful benefit/risk evaluation before initiating this new combination. This review aims at synthesizing the biological basis of the everolimus-exemestane association, presenting the main validated and ongoing therapeutic trials, interests and limits, as well as the multiple potential therapeutic perspectives.
Collapse
|
24
|
Gonzalez-Angulo AM, Akcakanat A, Liu S, Green MC, Murray JL, Chen H, Palla SL, Koenig KB, Brewster AM, Valero V, Ibrahim NK, Moulder-Thompson S, Litton JK, Tarco E, Moore J, Flores P, Crawford D, Dryden MJ, Symmans WF, Sahin A, Giordano SH, Pusztai L, Do KA, Mills GB, Hortobagyi GN, Meric-Bernstam F. Open-label randomized clinical trial of standard neoadjuvant chemotherapy with paclitaxel followed by FEC versus the combination of paclitaxel and everolimus followed by FEC in women with triple receptor-negative breast cancer†. Ann Oncol 2014; 25:1122-7. [PMID: 24669015 DOI: 10.1093/annonc/mdu124] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Everolimus synergistically enhances taxane-induced cytotoxicity in breast cancer cells in vitro and in vivo in addition to demonstrating a direct antiproliferative activity. We aim to determine pharmacodynamics changes and response of adding everolimus to standard neoadjuvant chemotherapy in triple-negative breast cancer (TNBC). PATIENTS AND METHODS Phase II study in patients with primary TNBC randomized to T-FEC (paclitaxel 80 mg/m(2) i.v. weekly for 12 weeks, followed by 5-fluorouracil 500 mg/m(2), epirubicin 100 mg/m(2), and cyclophosphamide 500 mg/m(2) every 3 weeks for four cycles) versus TR-FEC (paclitaxel 80 mg/m(2) i.v. and everolimus 30 mg PO weekly for 12 weeks, followed by FEC). Tumor samples were collected to assess molecular changes in the PI3K/AKT/mTOR pathway, at baseline, 48 h, 12 weeks, and at surgery by reverse phase protein arrays (RPPA). Clinical end points included 12-week clinical response rate (12-week RR), pathological complete response (pCR), and toxicity. RESULTS Sixty-two patients were registered, and 50 were randomized, 27 received T-FEC, and 23 received TR-FEC. Median age was 48 (range 31-75). There was downregulation of the mTOR pathway at 48 h in the TR-FEC arm. Twelve-week RR by ultrasound were 29.6% versus 47.8%, (P = 0.075), and pCR were 25.9% versus 30.4% (P = 0.76) for T-FEC and TR-FEC, respectively. mTOR downregulation at 48 h did not correlate with 12-week RR in the TR-FEC group (P = 0.58). Main NCI grade 3/4 toxicities included anemia, neutropenia, rash/desquamation, and vomiting in both arms. There was one case of grade 3 pneumonitis in the TR-FEC arm. No grade 3/4 stomatitis occurred. CONCLUSION The addition of everolimus to paclitaxel was well tolerated. Everolimus downregulated mTOR signaling but downregulation of mTOR at 48 h did not correlate with 12-week RR in the TR-FEC group. CLINICAL TRIAL NUMBER NCT00499603.
Collapse
Affiliation(s)
| | - A Akcakanat
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - S Liu
- Department of Breast Medical Oncology
| | | | | | - H Chen
- Department of Breast Medical Oncology
| | | | | | | | - V Valero
- Department of Breast Medical Oncology
| | | | | | | | - E Tarco
- Department of Breast Medical Oncology
| | - J Moore
- Department of Breast Medical Oncology
| | - P Flores
- Department of Breast Medical Oncology
| | | | | | - W F Symmans
- Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - A Sahin
- Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | | | - L Pusztai
- Division of Hematology-Oncology, Yale University, New Haven
| | - K-A Do
- Departments of Biostatistics
| | | | | | - F Meric-Bernstam
- Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
25
|
Triple negative breast carcinoma EGFR amplification is not associated with EGFR, Kras or ALK mutations. Br J Cancer 2014; 110:1045-52. [PMID: 24423920 PMCID: PMC3929875 DOI: 10.1038/bjc.2013.794] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 11/20/2013] [Accepted: 11/21/2013] [Indexed: 01/28/2023] Open
Abstract
Background: The amplification of epidermal growth factor receptor (EGFR) in triple negative breast carcinomas (TNBC) suggests its potential therapeutic application, as for HER-2, using standardised methods of measurement. In this regard, we aimed to compare several methods for evaluating EGFR amplification along with potential mutations for suitability in clinical practice. Methods: Tissue sections of 138 TNBCs were used (1) to compare EGFR amplification and expression by silver in situ hybridisation (SISH) to qPCR and immunohistochemistry (IHC) and (2) to search for EGFR mutations, along with Kras, PI3K, Braf and HER-2 mutations and echinoderm microtubule associated protein like 4-anaplastic lymphoma kinase (EML4-ALK) translocation. Results: (1) Amplification of EGFR was observed in well-characterised TNBCs (up to 92%); (2) qPCR correlated with SISH with 94% specificity and 75.6% sensitivity; (3) IHC correlated with SISH with 97% sensitivity and 78% specificity; (4) no EGFR, Kras mutations or EML4-ALK translocations were found, but PI3K and Braf mutations were observed in 26% of cases; and (5) small, acentric circular extrachromosomal DNA similar to ‘double minutes' in glioblastomas was observed in 18% of SISH sections. Conclusions: SISH and IHC are methods that are suitable in clinical practice to screen for EGFR amplification and overexpression, which are frequently observed in TNBC. Patients with TNBC are potential candidates for EGFR-targeted therapy combined with PI3K and Braf inhibitors.
Collapse
|
26
|
Lauring J, Park BH, Wolff AC. The phosphoinositide-3-kinase-Akt-mTOR pathway as a therapeutic target in breast cancer. J Natl Compr Canc Netw 2014; 11:670-8. [PMID: 23744866 DOI: 10.6004/jnccn.2013.0086] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The phosphoinositide-3-kinase (PI3-kinase)-Akt-mTOR pathway is a central signal transduction pathway that regulates many critical aspects of normal and cancer physiology, including cell proliferation, apoptosis, cell morphology and migration, protein synthesis, and integration of metabolism. In breast cancer, somatic mutations that activate the pathway occur in more than 50% of tumors, underscoring the potentially broad impact of targeting the pathway for therapy. A vast body of preclinical data demonstrates the efficacy of pathway inhibition on tumor growth, and evidence also shows that activation of the pathway occurs in models of acquired resistance to hormonal therapy. This preclinical work led to the investigation of allosteric mTOR inhibitors, everolimus and temsirolimus, in metastatic hormone receptor-positive breast cancer. The recent BOLERO-2 trial comparing everolimus plus exemestane versus placebo plus exemestane in women with resistance to nonsteroidal aromatase inhibitors demonstrated a 6-month improvement in progression-free survival and led to FDA approval of everolimus for this indication in the United States. This landmark trial is the first demonstration of significant clinical benefit using drugs targeting this pathway in breast cancer. Many questions remain about the role of everolimus and other pathway-targeting drugs in clinical development in breast cancer treatment. This article reviews the role of the PI3-kinase-Akt-mTOR pathway in breast cancer biology and the clinical trial evidence available to date.
Collapse
Affiliation(s)
- Josh Lauring
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21287, USA.
| | | | | |
Collapse
|
27
|
Eccles SA, Aboagye EO, Ali S, Anderson AS, Armes J, Berditchevski F, Blaydes JP, Brennan K, Brown NJ, Bryant HE, Bundred NJ, Burchell JM, Campbell AM, Carroll JS, Clarke RB, Coles CE, Cook GJR, Cox A, Curtin NJ, Dekker LV, dos Santos Silva I, Duffy SW, Easton DF, Eccles DM, Edwards DR, Edwards J, Evans DG, Fenlon DF, Flanagan JM, Foster C, Gallagher WM, Garcia-Closas M, Gee JMW, Gescher AJ, Goh V, Groves AM, Harvey AJ, Harvie M, Hennessy BT, Hiscox S, Holen I, Howell SJ, Howell A, Hubbard G, Hulbert-Williams N, Hunter MS, Jasani B, Jones LJ, Key TJ, Kirwan CC, Kong A, Kunkler IH, Langdon SP, Leach MO, Mann DJ, Marshall JF, Martin LA, Martin SG, Macdougall JE, Miles DW, Miller WR, Morris JR, Moss SM, Mullan P, Natrajan R, O’Connor JPB, O’Connor R, Palmieri C, Pharoah PDP, Rakha EA, Reed E, Robinson SP, Sahai E, Saxton JM, Schmid P, Smalley MJ, Speirs V, Stein R, Stingl J, Streuli CH, Tutt ANJ, Velikova G, Walker RA, Watson CJ, Williams KJ, Young LS, Thompson AM. Critical research gaps and translational priorities for the successful prevention and treatment of breast cancer. Breast Cancer Res 2013; 15:R92. [PMID: 24286369 PMCID: PMC3907091 DOI: 10.1186/bcr3493] [Citation(s) in RCA: 275] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 09/12/2013] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Breast cancer remains a significant scientific, clinical and societal challenge. This gap analysis has reviewed and critically assessed enduring issues and new challenges emerging from recent research, and proposes strategies for translating solutions into practice. METHODS More than 100 internationally recognised specialist breast cancer scientists, clinicians and healthcare professionals collaborated to address nine thematic areas: genetics, epigenetics and epidemiology; molecular pathology and cell biology; hormonal influences and endocrine therapy; imaging, detection and screening; current/novel therapies and biomarkers; drug resistance; metastasis, angiogenesis, circulating tumour cells, cancer 'stem' cells; risk and prevention; living with and managing breast cancer and its treatment. The groups developed summary papers through an iterative process which, following further appraisal from experts and patients, were melded into this summary account. RESULTS The 10 major gaps identified were: (1) understanding the functions and contextual interactions of genetic and epigenetic changes in normal breast development and during malignant transformation; (2) how to implement sustainable lifestyle changes (diet, exercise and weight) and chemopreventive strategies; (3) the need for tailored screening approaches including clinically actionable tests; (4) enhancing knowledge of molecular drivers behind breast cancer subtypes, progression and metastasis; (5) understanding the molecular mechanisms of tumour heterogeneity, dormancy, de novo or acquired resistance and how to target key nodes in these dynamic processes; (6) developing validated markers for chemosensitivity and radiosensitivity; (7) understanding the optimal duration, sequencing and rational combinations of treatment for improved personalised therapy; (8) validating multimodality imaging biomarkers for minimally invasive diagnosis and monitoring of responses in primary and metastatic disease; (9) developing interventions and support to improve the survivorship experience; (10) a continuing need for clinical material for translational research derived from normal breast, blood, primary, relapsed, metastatic and drug-resistant cancers with expert bioinformatics support to maximise its utility. The proposed infrastructural enablers include enhanced resources to support clinically relevant in vitro and in vivo tumour models; improved access to appropriate, fully annotated clinical samples; extended biomarker discovery, validation and standardisation; and facilitated cross-discipline working. CONCLUSIONS With resources to conduct further high-quality targeted research focusing on the gaps identified, increased knowledge translating into improved clinical care should be achievable within five years.
Collapse
Affiliation(s)
- Suzanne A Eccles
- The Institute of Cancer Research, 15 Cotswold Road, London SM2 5MG, UK
| | - Eric O Aboagye
- Imperial College London, Exhibition Rd, London SW7 2AZ, UK
| | - Simak Ali
- Imperial College London, Exhibition Rd, London SW7 2AZ, UK
| | | | - Jo Armes
- Kings College London, Strand, London WC2R 2LS, UK
| | | | - Jeremy P Blaydes
- University of Southampton, University Road, Southampton SO17 1BJ, UK
| | - Keith Brennan
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Nicola J Brown
- University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Helen E Bryant
- University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Nigel J Bundred
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | | | | | - Jason S Carroll
- Cancer Research UK, Cambridge Research Institute/University of Cambridge, Trinity Lane, Cambridge CB2 1TN, UK
| | - Robert B Clarke
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Charlotte E Coles
- Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, UK
| | - Gary JR Cook
- Kings College London, Strand, London WC2R 2LS, UK
| | - Angela Cox
- University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Nicola J Curtin
- Newcastle University, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | | | | | - Stephen W Duffy
- Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Douglas F Easton
- Cancer Research UK, Cambridge Research Institute/University of Cambridge, Trinity Lane, Cambridge CB2 1TN, UK
| | - Diana M Eccles
- University of Southampton, University Road, Southampton SO17 1BJ, UK
| | - Dylan R Edwards
- University of East Anglia, Earlham Road, Norwich NR4 7TJ, UK
| | - Joanne Edwards
- University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
| | - D Gareth Evans
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Deborah F Fenlon
- University of Southampton, University Road, Southampton SO17 1BJ, UK
| | | | - Claire Foster
- University of Southampton, University Road, Southampton SO17 1BJ, UK
| | | | | | - Julia M W Gee
- University of Cardiff, Park Place, Cardiff CF10 3AT, UK
| | - Andy J Gescher
- University of Leicester, University Road, Leicester LE1 4RH, UK
| | - Vicky Goh
- Kings College London, Strand, London WC2R 2LS, UK
| | - Ashley M Groves
- University College London, Gower Street, London WC1E 6BT, UK
| | | | - Michelle Harvie
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Bryan T Hennessy
- Royal College of Surgeons Ireland, 123, St Stephen’s Green, Dublin 2, Ireland
| | | | - Ingunn Holen
- University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Sacha J Howell
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Anthony Howell
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | | | | | | | - Bharat Jasani
- University of Cardiff, Park Place, Cardiff CF10 3AT, UK
| | - Louise J Jones
- Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Timothy J Key
- University of Oxford, Wellington Square, Oxford OX1 2JD, UK
| | - Cliona C Kirwan
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Anthony Kong
- University of Oxford, Wellington Square, Oxford OX1 2JD, UK
| | - Ian H Kunkler
- University of Edinburgh, South Bridge, Edinburgh EH8 9YL, UK
| | - Simon P Langdon
- University of Edinburgh, South Bridge, Edinburgh EH8 9YL, UK
| | - Martin O Leach
- The Institute of Cancer Research, 15 Cotswold Road, London SM2 5MG, UK
| | - David J Mann
- Imperial College London, Exhibition Rd, London SW7 2AZ, UK
| | - John F Marshall
- Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Lesley Ann Martin
- The Institute of Cancer Research, 15 Cotswold Road, London SM2 5MG, UK
| | - Stewart G Martin
- University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | | | | | | | | | - Sue M Moss
- Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Paul Mullan
- Queen’s University Belfast, University Road, Belfast BT7 1NN, UK
| | - Rachel Natrajan
- The Institute of Cancer Research, 15 Cotswold Road, London SM2 5MG, UK
| | | | | | - Carlo Palmieri
- The University of Liverpool, Brownlow Hill, Liverpool L69 7ZX, UK
| | - Paul D P Pharoah
- Cancer Research UK, Cambridge Research Institute/University of Cambridge, Trinity Lane, Cambridge CB2 1TN, UK
| | - Emad A Rakha
- University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Elizabeth Reed
- Princess Alice Hospice, West End Lane, Esher KT10 8NA, UK
| | - Simon P Robinson
- The Institute of Cancer Research, 15 Cotswold Road, London SM2 5MG, UK
| | - Erik Sahai
- London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - John M Saxton
- University of East Anglia, Earlham Road, Norwich NR4 7TJ, UK
| | - Peter Schmid
- Brighton and Sussex Medical School, University of Sussex, Brighton, East Sussex BN1 9PX, UK
| | | | | | - Robert Stein
- University College London, Gower Street, London WC1E 6BT, UK
| | - John Stingl
- Cancer Research UK, Cambridge Research Institute/University of Cambridge, Trinity Lane, Cambridge CB2 1TN, UK
| | | | | | | | | | - Christine J Watson
- Cancer Research UK, Cambridge Research Institute/University of Cambridge, Trinity Lane, Cambridge CB2 1TN, UK
| | - Kaye J Williams
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Leonie S Young
- Royal College of Surgeons Ireland, 123, St Stephen’s Green, Dublin 2, Ireland
| | | |
Collapse
|
28
|
Grosso SHG, Katayama MLH, Roela RA, Nonogaki S, Soares FA, Brentani H, Lima L, Folgueira MAAK, Waitzberg AFL, Pasini FS, Góes JCGS, Brentani MM. Breast cancer tissue slices as a model for evaluation of response to rapamycin. Cell Tissue Res 2013; 352:671-84. [DOI: 10.1007/s00441-013-1608-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 03/04/2013] [Indexed: 01/20/2023]
|
29
|
PIK3CA genotype and a PIK3CA mutation-related gene signature and response to everolimus and letrozole in estrogen receptor positive breast cancer. PLoS One 2013; 8:e53292. [PMID: 23301057 PMCID: PMC3534682 DOI: 10.1371/journal.pone.0053292] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 11/27/2012] [Indexed: 11/19/2022] Open
Abstract
The phosphatidylinositol 3′ kinase (PI3K) pathway is commonly activated in breast cancer and aberrations such as PI3K mutations are common. Recent exciting clinical trial results in advanced estrogen receptor-positive (ER) breast cancer support mTOR activation is a major means of estrogen-independent tumor growth. Hence the means to identify a responsive breast cancer population that would most benefit from these compounds in the adjuvant or earlier stage setting is of high interest. Here we study PIK3CA genotype as well as a previously reported PI3K/mTOR-pathway gene signature (PIK3CA-GS) and their ability to estimate the level of PI3K pathway activation in two clinical trials of newly diagnosed ER-positive breast cancer patients- a total of 81 patients- one of which was randomized between letrozole and placebo vs letrozole and everolimus. The main objectives were to correlate the baseline PIK3CA genotype and GS with the relative change from baseline to day 15 in Ki67 (which has been shown to be prognostic in breast cancer) and phosphorylated S6 (S240) immunohistochemistry (a substrate of mTOR). In the randomized dataset, the PIK3CA-GS could identify those patients with the largest relative decreases in Ki67 to letrozole/everolimus (R = −0.43, p = 0.008) compared with letrozole/placebo (R = 0.07, p = 0.58; interaction test p = 0.02). In a second dataset of pre-surgical everolimus alone, the PIK3CA-GS was not significantly correlated with relative change in Ki67 (R = −0.11, p = 0.37) but with relative change in phosphorlyated S6 (S240) (R = −0.46, p = 0.028). PIK3CA genotype was not significantly associated with any endpoint in either datasets. Our results suggest that the PIK3CA-GS has potential to identify those ER-positive BCs who may benefit from the addition of everolimus to letrozole. Further evaluation of the PIK3CA-GS as a predictive biomarker is warranted as it may facilitate better selection of responsive patient populations for mTOR inhibition in combination with letrozole.
Collapse
|
30
|
Zagouri F, Sergentanis TN, Chrysikos D, Filipits M, Bartsch R. mTOR inhibitors in breast cancer: a systematic review. Gynecol Oncol 2012; 127:662-72. [PMID: 22967800 DOI: 10.1016/j.ygyno.2012.08.040] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 08/20/2012] [Accepted: 08/26/2012] [Indexed: 10/27/2022]
Abstract
PI3K/AKT/mTOR pathway is a crucial mediator of tumor progression. As the PI3K/Akt pathway is heavily deregulated in breast cancer, the application of mTOR inhibitors in breast cancer patients seems warranted. This is the first systematic review according to PRISMA guidelines to synthesize all available data of mTOR inhibitors in all subcategories of breast cancer. The search strategy retrieved 16 studies evaluating everolimus (1492 patients), seven studies examining temsirolimus (1245 patients), one study evaluating sirolimus (400 patients) and two studies evaluating MKC-1 (60 patients). The Breast Cancer Trials of Oral Everolimus-2 (BOLERO-2) study has marked a turning point in the evaluation of everolimus in the treatment of estrogen receptor positive breast cancer. Given the positive results, everolimus has entered NCCN 2012 guidelines, and its approval of its combination with exemestane by FDA and EMA is imminent. In addition, the promising antitumor activity and long-term disease control further suggest that mTOR inhibition with everolimus may provide an avenue for achieving long-lasting benefit from trastuzumab-based therapy in HER2-positive patients. Regarding temsirolimus, it seems that the agent may play, in the future, a role in the treatment of metastatic breast cancer; importantly, however, there is an unmet need to find its optimal target subpopulation.
Collapse
Affiliation(s)
- Flora Zagouri
- Comprehensive Cancer Center Vienna, Department of Medicine I/Division of Oncology, Medical University of Vienna, Austria.
| | | | | | | | | |
Collapse
|
31
|
Sheri A, Dowsett M. Developments in Ki67 and other biomarkers for treatment decision making in breast cancer. Ann Oncol 2012; 23 Suppl 10:x219-27. [DOI: 10.1093/annonc/mds307] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
32
|
Abstract
Neoadjuvant treatment of breast cancer has become established as the safe and often effective therapeutic approach of choice for larger primary and for locally advanced breast cancer. The neoadjuvant approach offers the advantages of downstaging the disease, potentially reducing the extent of surgery and in an era of individualization of therapy, testing the efficacy of therapy administered to patients. The preoperative setting is also an effective way to study the activity of novel agents or therapeutic combinations in vivo against human breast cancer. For new therapies, preoperative trials avoid the issue of adaptive resistance and pretreatments that can be problematic in the advanced disease setting. For evidence of a drug targeting the cancer in vivo, comparisons of endocrine therapy, chemotherapy agents and/or targeted agents can provide data on activity and efficacy with a much shorter time frame and many fewer patients than for adjuvant trials; effects seen in neoadjuvant trials may even reflect what is found in the adjuvant setting. Patient benefits from the neoadjuvant approach may be greatest for those who experience complete pathologically documented response (and the consequent survival benefits) and women for whom breast conservation, rather than mastectomy, becomes possible.
Collapse
Affiliation(s)
- A M Thompson
- Ninewells Hospital and Medical School, Dundee Cancer Centre, Clinical Research Centre, University of Dundee, Dundee, UK.
| | | |
Collapse
|
33
|
Beaver JA, Park BH. The BOLERO-2 trial: the addition of everolimus to exemestane in the treatment of postmenopausal hormone receptor-positive advanced breast cancer. Future Oncol 2012; 8:651-7. [PMID: 22764762 PMCID: PMC3466807 DOI: 10.2217/fon.12.49] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The combination of the mTOR inhibitor everolimus with the aromatase inhibitor exemestane was evaluated in the randomized Phase III BOLERO-2 trial. Research has indicated that aberrant signaling through the mTOR pathway is associated with resistance to endocrine therapies. The BOLERO-2 trial examined the effects on progression-free survival of the addition of everolimus to exemestane in a patient population of postmenopausal, hormone receptor-positive, advanced breast cancer. At the interim analysis, the median progression-free survival assessed by local investigators was 6.9 months for everolimus plus exemestane versus 2.8 months for placebo plus exemestane (hazard ratio: 0.43; p < 0.001), and by central assessment was 10.6 versus 4.1 months, respectively (hazard ratio: 0.36; p < 0.001). The everolimus plus exemestane arm showed greater number of grade 3 and 4 adverse events. This study suggests that the addition of everolimus to exemestane is a potential viable treatment option for this patient population.
Collapse
Affiliation(s)
- Julia A Beaver
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, 1650 Orleans St, Room 151, Baltimore, MD 21287, USA
| | - Ben H Park
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, 1650 Orleans St, Room 151, Baltimore, MD 21287, USA
| |
Collapse
|
34
|
Villarreal-Garza C, Cortes J, Andre F, Verma S. mTOR inhibitors in the management of hormone receptor-positive breast cancer: the latest evidence and future directions. Ann Oncol 2012; 23:2526-2535. [PMID: 22553196 DOI: 10.1093/annonc/mds075] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND There is an unmet therapeutic need in endocrine-resistant, hormone receptor (HR)-positive, human epidermal growth factor receptor 2-negative advanced breast cancer (BC). Preclinical studies support the hypothesis that the mammalian target of rapamycin (mTOR) inhibition could potentially overcome resistance to endocrine therapy. MATERIALS AND METHODS A literature review regarding BC and mTOR inhibitors was undertaken. The reference lists from retrieved manuscripts were reviewed to identify further studies. RESULTS Phase II studies have reported that the combination of mTOR inhibitors with endocrine therapy shows efficacy in patients with advanced disease that progressed after treatment with aromatase inhibitors. The recent findings of the phase III BOLERO-2 confirmed that everolimus in combination with exemestane significantly improved progression-free survival and response rate, with a manageable safety profile. CONCLUSIONS The addition of everolimus to exemestane for women with HR-positive metastatic BC is now considered a new therapeutic strategy. However, a word of caution should be added regarding toxic effects, which might limit practical use and compliance. It is essential that clinicians are educated about key recommendations for toxicity management and specific guideline dose modifications. Additional research efforts with the addition of these compounds in the early-stage setting is greatly needed to improve the survival of patients with HR-positive BC.
Collapse
Affiliation(s)
- C Villarreal-Garza
- Department of Medical Oncology, Instituto Nacional de Cancerologia, Mexico DF, Mexico
| | - J Cortes
- Breast Cancer Unit, Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - F Andre
- Breast Cancer Unit, Department of Medical Oncology, University Paris XI and Institut Gustave Roussy, Villejuif, France
| | - S Verma
- Department of Medical Oncology, Sunnybrook Odette Cancer Centre, Toronto, Canada.
| |
Collapse
|
35
|
Newton M, Nagaiah G, Abraham J. mTOR as a target in breast cancer: the emerging role of everolimus. BREAST CANCER MANAGEMENT 2012. [DOI: 10.2217/bmt.12.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY The majority of patients with breast cancer are estrogen- and progesterone-receptor positive, and have benefited from the development of anti-estrogen therapies, such as tamoxifen and aromatase inhibitors. Unfortunately, metastatic patients will eventually develop resistance to these agents. Inhibitors of the mTOR, particularly everolimus, show promising activity in this group of patients. mTOR inhibition appears to reverse resistance to anti-estrogen therapy in the estrogen-receptor-/progesterone-receptor-positive subset. Additionally, they may have a similar effect by reversing anti-HER2 resistance in patients who overexpress HER2. This article reviews the mechanism of action of mTOR inhibitors and summarizes the available clinical data of their use in breast cancer.
Collapse
Affiliation(s)
- Michael Newton
- West Virginia University School of Pharmacy, Morgantown, WV, USA
- Mary Babb Randolph Cancer Center, Section of Hematology/Oncology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Govardhanan Nagaiah
- Mary Babb Randolph Cancer Center, Section of Hematology/Oncology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Jame Abraham
- Mary Babb Randolph Cancer Center, Section of Hematology/Oncology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
36
|
Lu X, Xiao L, Wang L, Ruden DM. Hsp90 inhibitors and drug resistance in cancer: the potential benefits of combination therapies of Hsp90 inhibitors and other anti-cancer drugs. Biochem Pharmacol 2012; 83:995-1004. [PMID: 22120678 PMCID: PMC3299878 DOI: 10.1016/j.bcp.2011.11.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 10/31/2011] [Accepted: 11/14/2011] [Indexed: 12/11/2022]
Abstract
Hsp90 is a chaperone protein that interacts with client proteins that are known to be in the cell cycle, signaling and chromatin-remodeling pathways. Hsp90 inhibitors act additively or synergistically with many other drugs in the treatment of both solid tumors and leukemias in murine tumor models and humans. Hsp90 inhibitors potentiate the actions of anti-cancer drugs that target Hsp90 client proteins, including trastuzumab (Herceptin™) which targets Her2/Erb2B, as Hsp90 inhibition elicits the drug effects in cancer cell lines that are otherwise resistant to the drug. A phase II study of the Hsp90 inhibitor 17-AAG and trastuzumab showed that this combination therapy has anticancer activity in patients with HER2-positive metastatic breast cancer progressing on trastuzumab. In this review, we discuss the results of Hsp90 inhibitors in combination with trastuzumab and other cancer drugs. We also discuss recent results from yeast focused on the genetics of drug resistance when Hsp90 is inhibited and the implications that this might have in understanding the effects of genetic variation in treating cancer in humans.
Collapse
Affiliation(s)
- Xiangyi Lu
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201
| | - Li Xiao
- University of Alabama at Birmingham, Department of Immunology and Rheumatology, Birmingham, AL 35294
| | - Luan Wang
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201
| | - Douglas M. Ruden
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201
| |
Collapse
|
37
|
Petrich AM, Leshchenko V, Kuo PY, Xia B, Thirukonda VK, Ulahannan N, Gordon S, Fazzari MJ, Ye BH, Sparano JA, Parekh S. Akt inhibitors MK-2206 and nelfinavir overcome mTOR inhibitor resistance in diffuse large B-cell lymphoma. Clin Cancer Res 2012; 18:2534-44. [PMID: 22338016 DOI: 10.1158/1078-0432.ccr-11-1407] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The mTOR pathway is constitutively activated in diffuse large B-cell lymphoma (DLBCL). mTOR inhibitors have activity in DLBCL, although response rates remain low. We evaluated DLBCL cell lines with differential resistance to the mTOR inhibitor rapamycin: (i) to identify gene expression profile(s) (GEP) associated with resistance to rapamycin, (ii) to understand mechanisms of rapamycin resistance, and (iii) to identify compounds likely to synergize with mTOR inhibitor. EXPERIMENTAL DESIGN We sought to identify a GEP of mTOR inhibitor resistance by stratification of eight DLBCL cell lines with respect to response to rapamycin. Then, using pathway analysis and connectivity mapping, we sought targets likely accounting for this resistance and compounds likely to overcome it. We then evaluated two compounds thus identified for their potential to synergize with rapamycin in DLBCL and confirmed mechanisms of activity with standard immunoassays. RESULTS We identified a GEP capable of reliably distinguishing rapamycin-resistant from rapamycin-sensitive DLBCL cell lines. Pathway analysis identified Akt as central to the differentially expressed gene network. Connectivity mapping identified compounds targeting Akt as having a high likelihood of reversing the GEP associated with mTOR inhibitor resistance. Nelfinavir and MK-2206, chosen for their Akt-inhibitory properties, yielded synergistic inhibition of cell viability in combination with rapamycin in DLBCL cell lines, and potently inhibited phosphorylation of Akt and downstream targets of activated mTOR. CONCLUSIONS GEP identifies DLBCL subsets resistant to mTOR inhibitor therapy. Combined targeting of mTOR and Akt suppresses activation of key components of the Akt/mTOR pathway and results in synergistic cytotoxicity. These findings are readily adaptable to clinical trials.
Collapse
MESH Headings
- Apoptosis/drug effects
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Drug Resistance, Neoplasm/drug effects
- Flow Cytometry
- Fluorescent Antibody Technique
- Gene Expression Profiling
- HIV Protease Inhibitors/pharmacology
- Heterocyclic Compounds, 3-Ring/pharmacology
- Humans
- Immunoenzyme Techniques
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Nelfinavir/pharmacology
- Oligonucleotide Array Sequence Analysis
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Phosphorylation/drug effects
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction/drug effects
- Sirolimus/pharmacology
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/metabolism
Collapse
|
38
|
Bago-Horvath Z, Sieghart W, Grusch M, Lackner A, Hayden H, Pirker C, Komina O, Węsierska-Gądek J, Haitel A, Filipits M, Berger W, Schmid K. Synergistic effects of erlotinib and everolimus on bronchial carcinoids and large-cell neuroendocrine carcinomas with activated EGFR/AKT/mTOR pathway. Neuroendocrinology 2012; 96:228-37. [PMID: 22378048 DOI: 10.1159/000337257] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 02/14/2012] [Indexed: 12/21/2022]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) and mammalian target of rapamycin (mTOR) are crucial targets in cancer therapy. Combined inhibition of both targets yielded synergistic effects in vitro and in vivo in several cancer entities. However, the impact of EGFR and mTOR expression and combined inhibition in neuroendocrine lung tumors other than small-cell lung cancer remains unclear. MATERIAL AND METHODS Expression and activation of EGFR/AKT/mTOR pathway constituents were investigated in typical and atypical bronchial carcinoid (AC) tumors and large-cell neuroendocrine lung carcinomas (LCNEC) by immunohistochemistry in 110 tumor samples, and correlated with clinicopathological parameters and patient survival. Cytotoxicity of mTOR inhibitor everolimus and EGFR inhibitor erlotinib alone and in combination was assessed using growth inhibition assay in NCI-H720 AC and SHP-77 LCNEC cells. Cell cycle phase distribution was determined by FACS. Apoptosis-associated activation of caspase-3/7 was measured by Caspase-Glo® assay. Activity status of EGFR and mTOR pathway components was analyzed by immunoblotting. RESULTS Activation of the EGFR/AKT/mTOR axis could be demonstrated in all entities and was significantly increased in higher grade tumors. Neoadjuvant chemotherapy correlated significantly with p-AKT expression and p-ERK loss. Erlotinib combined with everolimus exerted synergistic combination effects in AC and LCNEC cells by induction of apoptosis, while cell cycle phase distribution remained unaffected. These effects could be explained by synergistic downregulation of phospho-mTOR, phospho-p70S6 kinase and phospho-AKT expression by everolimus and erlotinib. CONCLUSIONS Our study indicates that EGFR and mTOR are clinically important targets in bronchial neuroendocrine tumors, and further in vivo and clinical exploration of combined inhibition is warranted.
Collapse
|
39
|
Abstract
In recent years the description of well-defined molecular subtypes of breast cancer, together with the identification of the driving genetic alterations and signaling pathways, has led to the clinical development of a number of successful molecular targeted agents. This is best exemplified in the subset of HER2-amplified breast cancers, in which an increasing number of active agents are changing the natural history of this aggressive disease. Other targets are under exploration, and the clinical development of these agents will require a change from the current large, randomized trials in unselected patient populations to smaller trials in groups with a molecularly defined tumor type. In addition, combinatorial approaches that act on the secondary mutations and/or compensatory pathways in resistant tumors may markedly improve on the effects of targeted agents used alone.
Collapse
Affiliation(s)
- Michaela J Higgins
- Division of Hematology and Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | |
Collapse
|
40
|
Goodson WH, Luciani MG, Sayeed SA, Jaffee IM, Moore DH, Dairkee SH. Activation of the mTOR pathway by low levels of xenoestrogens in breast epithelial cells from high-risk women. Carcinogenesis 2011; 32:1724-33. [PMID: 21890461 PMCID: PMC3204351 DOI: 10.1093/carcin/bgr196] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Breast cancer is an estrogen-driven disease. Consequently, hormone replacement therapy correlates with disease incidence. However, increasing male breast cancer rates over the past three decades implicate additional sources of estrogenic exposure including wide spread estrogen-mimicking chemicals or xenoestrogens (XEs), such as bisphenol-A (BPA). By exposing renewable, human, high-risk donor breast epithelial cells (HRBECs) to BPA at concentrations that are detectable in human blood, placenta and milk, we previously identified gene expression profile changes associated with activation of mammalian target of rapamycin (mTOR) pathway genesets likely to trigger prosurvival changes in human breast cells. We now provide functional validation of mTOR activation using pairwise comparisons of 16 independent HRBEC samples with and without BPA exposure. We demonstrate induction of key genes and proteins in the PI3K-mTOR pathway--AKT1, RPS6 and 4EBP1 and a concurrent reduction in the tumor suppressor, phosphatase and tensin homolog gene protein. Altered regulation of mTOR pathway proteins in BPA-treated HRBECs led to marked resistance to rapamycin, the defining mTOR inhibitor. Moreover, HRBECs pretreated with BPA, or the XE, methylparaben (MP), surmounted antiestrogenic effects of tamoxifen showing dose-dependent apoptosis evasion and induction of cell cycling. Overall, XEs, when tested in benign breast cells from multiple human subjects, consistently initiated specific functional changes of the kind that are attributed to malignant onset in breast tissue. Our observations demonstrate the feasibility of studying renewable human samples as surrogates and reinforce the concern that BPA and MP, at low concentrations detected in humans, can have adverse health consequences.
Collapse
Affiliation(s)
- William H Goodson
- California Pacific Medical Center, Research Institute, San Francisco, CA 94107, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Satheesha S, Cookson VJ, Coleman LJ, Ingram N, Madhok B, Hanby AM, Suleman CAB, Sabine VS, Macaskill EJ, Bartlett JMS, Dixon JM, McElwaine JN, Hughes TA. Response to mTOR inhibition: activity of eIF4E predicts sensitivity in cell lines and acquired changes in eIF4E regulation in breast cancer. Mol Cancer 2011; 10:19. [PMID: 21320304 PMCID: PMC3055230 DOI: 10.1186/1476-4598-10-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 02/14/2011] [Indexed: 11/10/2022] Open
Abstract
Background Inhibitors of the kinase mTOR, such as rapamycin and everolimus, have been used as cancer therapeutics with limited success since some tumours are resistant. Efforts to establish predictive markers to allow selection of patients with tumours likely to respond have centred on determining phosphorylation states of mTOR or its targets 4E-BP1 and S6K in cancer cells. In an alternative approach we estimated eIF4E activity, a key effector of mTOR function, and tested the hypothesis that eIF4E activity predicts sensitivity to mTOR inhibition in cell lines and in breast tumours. Results We found a greater than three fold difference in sensitivity of representative colon, lung and breast cell lines to rapamycin. Using an assay to quantify influences of eIF4E on the translational efficiency specified by structured 5'UTRs, we showed that this estimate of eIF4E activity was a significant predictor of rapamycin sensitivity, with higher eIF4E activities indicative of enhanced sensitivity. Surprisingly, non-transformed cell lines were not less sensitive to rapamycin and did not have lower eIF4E activities than cancer lines, suggesting the mTOR/4E-BP1/eIF4E axis is deregulated in these non-transformed cells. In the context of clinical breast cancers, we estimated eIF4E activity by analysing expression of eIF4E and its functional regulators within tumour cells and combining these scores to reflect inhibitory and activating influences on eIF4E. Estimates of eIF4E activity in cancer biopsies taken at diagnosis did not predict sensitivity to 11-14 days of pre-operative everolimus treatment, as assessed by change in tumour cell proliferation from diagnosis to surgical excision. However, higher pre-treatment eIF4E activity was significantly associated with dramatic post-treatment changes in expression of eIF4E and 4E-binding proteins, suggesting that eIF4E is further deregulated in these tumours in response to mTOR inhibition. Conclusions Estimates of eIF4E activity predict sensitivity to mTOR inhibition in cell lines but breast tumours with high estimated eIF4E activity gain changes in eIF4E regulation in order to enhance resistance.
Collapse
Affiliation(s)
- Sampoorna Satheesha
- Leeds Institute of Molecular Medicine, St, James's University Hospital, Leeds University, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Are we missing the mTOR target in breast cancer? Breast Cancer Res Treat 2010; 128:607-11. [PMID: 20953834 DOI: 10.1007/s10549-010-1207-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 09/28/2010] [Indexed: 01/14/2023]
|