1
|
Kazemi K, Jamshidi K, Naseri R, Shahriarirad R, Shamsaeefar A, Hosseinzadeh A. Comparison of the effect of Everolimus, Prednisolone, and a combination of both on experimentally induced peritoneal adhesions in rats. Sci Rep 2024; 14:11077. [PMID: 38745015 PMCID: PMC11093995 DOI: 10.1038/s41598-024-61620-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Postoperative intra-abdominal adhesions represent a significant post-surgical problem. Its complications can cause a considerable clinical and cost burden. Herein, our study aimed to investigate the effect of Everolimus on peritoneal adhesion formation after inducing adhesions in rats. In this experimental study, adhesion bands were induced by intraperitoneal injection of 3 ml of 10% sterile talc solution in 64 male albino rats. The first group served as the control group. The second one received oral Prednisolone (1 mg/kg/day), the third received Everolimus (0.1 mg/kg/day), and group four received both drugs with similar dosages for four consecutive weeks. The formation of adhesion bands was qualitatively graded according to the Nair classification. The rats in the control group had extensive adhesions between the abdominal wall and the organs. Regarding substantial adhesion formation, 50% (8/16) of animals in the control group had substantial adhesions, while this rate in the groups receiving Prednisolone, Everolimus, and combination treatment was 31%, 31%, and 31%, respectively. Also, 68.75% (5/11) of the Prednisolone recipients had insubstantial adhesions, the same as Everolimus recipients, while in the combination group, 66.66% (10/15) rats had insubstantial adhesions. Everolimus demonstrated satisfactory results in reducing the rates of induced peritoneal adhesion in an experimental model, similar to Prednisolone and superior to a combination regime.
Collapse
Affiliation(s)
- Kourosh Kazemi
- Shiraz Transplant Center, Abu Ali Sina Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kamran Jamshidi
- Department of Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reyhaneh Naseri
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Shahriarirad
- Thoracic and Vascular Surgery Research Center, Shiraz University of Medical Science, Shiraz, Iran
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Shamsaeefar
- Shiraz Transplant Center, Abu Ali Sina Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Hosseinzadeh
- Department of Surgery, Shiraz University of Medical Sciences, Shiraz, Iran.
- Thoracic and Vascular Surgery Research Center, Shiraz University of Medical Science, Shiraz, Iran.
| |
Collapse
|
2
|
Naseri B, Mardi A, Khosrojerdi A, Baghbani E, Aghebati-Maleki L, Hatami-Sadr A, Heris JA, Eskandarzadeh S, Kafshdouz M, Alizadeh N, Baradaran B. Everolimus treatment enhances inhibitory immune checkpoint molecules' expression in monocyte-derived dendritic cells. Hum Immunol 2024; 85:110798. [PMID: 38569354 DOI: 10.1016/j.humimm.2024.110798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Antigen-specific T-cell immunity is provided by dendritic cells (DCs), which are specialized antigen-presenting cells. Furthermore, they establish a link between innate and adaptive immune responses. Currently, DC modification is a new approach for the therapy of several disorders. During solid organ transplantation, Everolimus, which is a mammalian target of rapamycin (mTOR) inhibitor, was initially utilized to suppress the immune system's functionality. Due to the intervention of Everolimus in various signaling pathways in cells and its modulatory properties on the immune system, this study aims to investigate the effect of treatment with Everolimus on the maturation and expression of immune checkpoint genes in monocyte-derived DCs. METHODS To isolate monocytes from PBMCs, the CD14 marker was used via the MACS method. Monocytes were cultured and induced to differentiate into monocyte-derived DCs by utilizing GM-CSF and IL-4 cytokines. On the fifth day, immature DCs were treated with Everolimus and incubated for 24 h. On the sixth day, the flow cytometry technique was used to investigate the effect of Everolimus on the phenotypic characteristics of DCs. In the end, the expression of immune checkpoint genes in both the Everolimus-treated and untreated DCs groups was assessed using the real-time PCR method. RESULTS The findings of this research demonstrated that the administration of Everolimus to DCs led to a notable rise in human leukocyte antigen (HLA)-DR expression and a decrease in CD11c expression. Furthermore, there was a significant increase in the expression of immune checkpoint molecules, namely CTLA-4, VISTA, PD-L1, and BTLA, in DCs treated with Everolimus. CONCLUSION The findings of this study show that Everolimus can target DCs and affect their phenotype and function in order to shift them toward a partially tolerogenic state. However, additional research is required to gain a comprehensive understanding of the precise impact of Everolimus on the activation status of DCs.
Collapse
Affiliation(s)
- Bahar Naseri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezou Khosrojerdi
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | | - Mahshid Kafshdouz
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Li S, Chen JS, Li X, Bai X, Shi D. MNK, mTOR or eIF4E-selecting the best anti-tumor target for blocking translation initiation. Eur J Med Chem 2023; 260:115781. [PMID: 37669595 DOI: 10.1016/j.ejmech.2023.115781] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/29/2023] [Accepted: 08/29/2023] [Indexed: 09/07/2023]
Abstract
Overexpression of eIF4E is common in patients with various solid tumors and hematologic cancers. As a potential anti-cancer target, eIF4E has attracted extensive attention from researchers. At the same time, mTOR kinases inhibitors and MNK kinases inhibitors, which are directly related to regulation of eIF4E, have been rapidly developed. To explore the optimal anti-cancer targets among MNK, mTOR, and eIF4E, this review provides a detailed classification and description of the anti-cancer activities of promising compounds. In addition, the structures and activities of some dual-target inhibitors are briefly described. By analyzing the different characteristics of the inhibitors, it can be concluded that MNK1/2 and eIF4E/eIF4G interaction inhibitors are superior to mTOR inhibitors. Simultaneous inhibition of MNK and eIF4E/eIF4G interaction may be the most promising anti-cancer method for targeting translation initiation.
Collapse
Affiliation(s)
- Shuo Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, Shandong, PR China.
| | - Jia-Shu Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, Shandong, PR China.
| | - Xiangqian Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, Shandong, PR China.
| | - Xiaoyi Bai
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, Shandong, PR China.
| | - Dayong Shi
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, Shandong, PR China.
| |
Collapse
|
4
|
Puszkiel A, You B, Payen L, Lopez J, Guitton J, Rousset P, Fontaine J, Péron J, Maillet D, Tartas S, Bonnin N, Trillet-Lenoir V, Colomban O, Augu-Denechere D, Freyer G, Tod M. A PK-PD model linking biomarker dynamics to progression-free survival in patients treated with everolimus and sorafenib combination therapy, EVESOR phase I trial. Cancer Chemother Pharmacol 2023; 91:413-425. [PMID: 37010549 DOI: 10.1007/s00280-023-04520-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 03/08/2023] [Indexed: 04/04/2023]
Abstract
PURPOSE The objective was to develop a pharmacokinetic-pharmacodynamic (PK-PD) model linking everolimus and sorafenib exposure with biomarker dynamics and progression-free survival (PFS) based on data from EVESOR trial in patients with solid tumors treated with everolimus and sorafenib combination therapy and to simulate alternative dosing schedules for sorafenib. PATIENTS AND METHODS Everolimus (5-10 mg once daily, qd) and sorafenib (200-400 mg twice daily, bid) were administered according to four different dosing schedules in 43 solid tumor patients. Rich PK and PD sampling for serum angiogenesis biomarkers was performed. Baseline activation of RAS/RAF/ERK (MAPK) pathway was assessed by quantification of mRNA specific gene panel in tumor biopsies. The PK-PD modeling was performed using NONMEM® software. RESULTS An indirect response PK-PD model linking sorafenib plasma exposure with soluble vascular endothelial growth factor receptor 2 (sVEGFR2) dynamics was developed. Progression-free survival (PFS) was described by a parametric time-to-event model. Higher decreases in sVEGFR2 at day 21 and higher baseline activation of MAPK pathway were associated with longer PFS (p = 0.002 and p = 0.007, respectively). The simulated schedule sorafenib 200 mg bid 5 days-on/2 days-off + continuous everolimus 5 mg qd was associated with median PFS of 4.3 months (95% CI 1.6-14.4), whereas the median PFS in the EVESOR trial was 3.6 months (95% CI 2.7-4.2, n = 43). CONCLUSION Sorafenib 200 mg bid 5 days-on/2 days-off + everolimus 5 mg qd continuous was selected for an additional arm of EVESOR trial to evaluate whether this simulated schedule is associated with higher clinical benefit. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01932177.
Collapse
Affiliation(s)
- Alicja Puszkiel
- EA 3738 CICLY, Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon 1, Lyon, France
| | - Benoit You
- EA 3738 CICLY, Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon 1, Lyon, France.
- Medical Oncology, Institut de Cancérologie des Hospices Civils de Lyon (IC-HCL), CITOHL, Centre Hospitalier Lyon Sud, 165 Chemin du Grand Revoyet, Pierre-Benite, 69495, Lyon, France.
| | - Léa Payen
- Laboratoire d'Oncologie Moléculaire et Transfert, Centre de Biologie Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Jonathan Lopez
- Centre de Recherche en Cancérologie de Lyon - INSERM U1052 CNRS U5286, Service de Biochimie et Biologie moléculaire, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Guitton
- Laboratoire de Biochimie-Toxicologie, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Pascal Rousset
- Department of Radiology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Juliette Fontaine
- Department of Pathology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Julien Péron
- Medical Oncology, Institut de Cancérologie des Hospices Civils de Lyon (IC-HCL), CITOHL, Centre Hospitalier Lyon Sud, 165 Chemin du Grand Revoyet, Pierre-Benite, 69495, Lyon, France
| | - Denis Maillet
- Medical Oncology, Institut de Cancérologie des Hospices Civils de Lyon (IC-HCL), CITOHL, Centre Hospitalier Lyon Sud, 165 Chemin du Grand Revoyet, Pierre-Benite, 69495, Lyon, France
| | - Sophie Tartas
- Medical Oncology, Institut de Cancérologie des Hospices Civils de Lyon (IC-HCL), CITOHL, Centre Hospitalier Lyon Sud, 165 Chemin du Grand Revoyet, Pierre-Benite, 69495, Lyon, France
| | - Nathalie Bonnin
- Medical Oncology, Institut de Cancérologie des Hospices Civils de Lyon (IC-HCL), CITOHL, Centre Hospitalier Lyon Sud, 165 Chemin du Grand Revoyet, Pierre-Benite, 69495, Lyon, France
| | - Veronique Trillet-Lenoir
- Medical Oncology, Institut de Cancérologie des Hospices Civils de Lyon (IC-HCL), CITOHL, Centre Hospitalier Lyon Sud, 165 Chemin du Grand Revoyet, Pierre-Benite, 69495, Lyon, France
| | - Olivier Colomban
- EA 3738 CICLY, Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon 1, Lyon, France
| | - Diane Augu-Denechere
- Medical Oncology, Institut de Cancérologie des Hospices Civils de Lyon (IC-HCL), CITOHL, Centre Hospitalier Lyon Sud, 165 Chemin du Grand Revoyet, Pierre-Benite, 69495, Lyon, France
| | - Gilles Freyer
- EA 3738 CICLY, Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon 1, Lyon, France
- Medical Oncology, Institut de Cancérologie des Hospices Civils de Lyon (IC-HCL), CITOHL, Centre Hospitalier Lyon Sud, 165 Chemin du Grand Revoyet, Pierre-Benite, 69495, Lyon, France
| | - Michel Tod
- EA 3738 CICLY, Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon 1, Lyon, France
- Pharmacie de l'Hôpital de la Croix Rousse, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
5
|
Ali ES, Mitra K, Akter S, Ramproshad S, Mondal B, Khan IN, Islam MT, Sharifi-Rad J, Calina D, Cho WC. Recent advances and limitations of mTOR inhibitors in the treatment of cancer. Cancer Cell Int 2022; 22:284. [PMID: 36109789 PMCID: PMC9476305 DOI: 10.1186/s12935-022-02706-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
The PI3K-Akt-mechanistic (formerly mammalian) target of the rapamycin (mTOR) signaling pathway is important in a variety of biological activities, including cellular proliferation, survival, metabolism, autophagy, and immunity. Abnormal PI3K-Akt-mTOR signalling activation can promote transformation by creating a cellular environment conducive to it. Deregulation of such a system in terms of genetic mutations and amplification has been related to several human cancers. Consequently, mTOR has been recognized as a key target for the treatment of cancer, especially for treating cancers with elevated mTOR signaling due to genetic or metabolic disorders. In vitro and in vivo, rapamycin which is an immunosuppressant agent actively suppresses the activity of mTOR and reduces cancer cell growth. As a result, various sirolimus-derived compounds have now been established as therapies for cancer, and now these medications are being investigated in clinical studies. In this updated review, we discuss the usage of sirolimus-derived compounds and other drugs in several preclinical or clinical studies as well as explain some of the challenges involved in targeting mTOR for treating various human cancers.
Collapse
|
6
|
Melendez B, Shah S, Jiang Y, Dottino J, Watson E, Pearce H, Borthwick M, Schmandt RE, Zhang Q, Cumpian K, Celestino J, Fellman B, Yuan Y, Lu KH, Mikos AG, Yates MS. Novel polymer-based system for intrauterine delivery of everolimus for anti-cancer applications. J Control Release 2021; 339:521-530. [PMID: 34648891 DOI: 10.1016/j.jconrel.2021.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 09/22/2021] [Accepted: 10/07/2021] [Indexed: 11/20/2022]
Abstract
Non-surgical treatment options for low-grade endometrial cancer and precancerous lesions are a critical unmet need for women who wish to preserve fertility or are unable to undergo hysterectomy. The PI3K/AKT/mTOR pathway is frequently activated in endometrial cancers and has been associated with resistance to endocrine therapy, making it a compelling target for early stage disease. Oral everolimus, an inhibitor against mTORC1, has shown clinical benefit in advanced or recurrent disease but has severe adverse effects that may lead to treatment interruption or dose reduction. To overcome this, we developed a polymer-based intrauterine delivery system to achieve persistent, local delivery of everolimus without systemic exposure. In vivo studies, using a rat model, showed that a poly(propylene fumarate)-based rod loaded with everolimus achieved everolimus delivery to the endometrium with levels similar to oral administration, but with limited systemic exposure and up to 84 days of release. Biological activity of everolimus delivered with this system was confirmed, measured by reduced lumen epithelial cell height and PI3K pathway biomarkers. This study shows a promising new delivery approach for anti-cancer drugs for non-surgical treatment of low-grade endometrial cancer.
Collapse
Affiliation(s)
- Brenda Melendez
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Pressler St, Houston, TX 77030, United States of America
| | - Sarita Shah
- Department of Bioengineering, Rice University, 6500 Main St, Houston, TX 77030, United States of America
| | - Yunyun Jiang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Pressler St, Houston, TX 77030, United States of America
| | - Joseph Dottino
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Pressler St, Houston, TX 77030, United States of America
| | - Emma Watson
- Department of Bioengineering, Rice University, 6500 Main St, Houston, TX 77030, United States of America
| | - Hannah Pearce
- Department of Bioengineering, Rice University, 6500 Main St, Houston, TX 77030, United States of America
| | - Mikayla Borthwick
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Pressler St, Houston, TX 77030, United States of America
| | - Rosemarie E Schmandt
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Pressler St, Houston, TX 77030, United States of America
| | - Qian Zhang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Pressler St, Houston, TX 77030, United States of America
| | - Kayleah Cumpian
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Pressler St, Houston, TX 77030, United States of America
| | - Joseph Celestino
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Pressler St, Houston, TX 77030, United States of America
| | - Bryan Fellman
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Houston, TX 77030, United States of America
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Houston, TX 77030, United States of America
| | - Karen H Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Pressler St, Houston, TX 77030, United States of America
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, 6500 Main St, Houston, TX 77030, United States of America
| | - Melinda S Yates
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1155 Pressler St, Houston, TX 77030, United States of America.
| |
Collapse
|
7
|
Gao L, Zhang L, He F, Chen J, Zhao M, Li S, Wu H, Liu Y, Zhang Y, Ping Q, Hu L, Qiao H. Surfactant Assisted Rapid-Release Liposomal Strategies Enhance the Antitumor Efficiency of Bufalin Derivative and Reduce Cardiotoxicity. Int J Nanomedicine 2021; 16:3581-3598. [PMID: 34079251 PMCID: PMC8165102 DOI: 10.2147/ijn.s313153] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/06/2021] [Indexed: 12/18/2022] Open
Abstract
Background BF211, a derivative of bufalin (BF), shows significantly improved solubility and potent antitumor efficiency compared to BF. Unfortunately, the unwanted toxicity such as cardiotoxicity caused by unspecific distribution has hindered its clinical use. Methods PEGylated BF211 liposomes (BF211@Lipo) were designed and optimizely prepared based on the pre-prescription research. In vitro and in vivo cardiotoxicity was evaluated. In vivo pharmacokinetics and biodistribution of BF211@Lipo were investigated. In vivo antitumor activity and toxicity were evaluated in HepG2 cell xenograft models. The rapid-release triggered by Poloxamer 188 (P188) was assessed in vitro and in vivo. Results The optimized BF211@Lipo displayed a spherical morphology with a size of (164.6 ± 10.3) nm and a high encapsulation efficiency of (93.24 ± 2.15) %. The in vivo concentration–time curves of BF211 loaded in liposomes showed a prolonged half-life in plasma and increased tumor accumulation. No obvious abnormality in electrocardiograms was observed in guinea pigs even at 9 mg/kg. Moreover, to improve the efficient release of BF211@Lipo, a surfactant-assisted rapid-release strategy was developed, and the release-promoting mechanism was revealed by the fluorescence resonance energy transfer (FRET) and fluorescence nanoparticle tracking analysis (fl-NTA) technology. Sequential injection of BF211@Lipo and P188 could ignite the “cold” liposomes locally in tumor regions, facilitating the burst release of BF211 and enhancing the therapeutic index. Conclusion Our progressive efforts that begin with preparation technology and dosage regimen enable BF211 to like a drug, providing a promising nano platform to deliver the cardiac glycosides and alleviate the side effects by decreasing unspecific biodistribution.
Collapse
Affiliation(s)
- Lina Gao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Lei Zhang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Fengjun He
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Jing Chen
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Meng Zhao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Simin Li
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Hao Wu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yumeng Liu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yinan Zhang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Qineng Ping
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Hongzhi Qiao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.,Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| |
Collapse
|
8
|
Spiliotaki M, Kallergi G, Nikolaou C, Xenidis N, Politaki E, Apostolaki S, Georgoulia N, Koinis F, Tsoukalas N, Hatzidaki D, Kotsakis A, Georgoulias V. Dynamic changes of CTCs in patients with metastatic HR(+)/HER2(-) breast cancer receiving salvage treatment with everolimus/exemestane. Cancer Chemother Pharmacol 2021; 87:277-287. [PMID: 33515073 DOI: 10.1007/s00280-020-04227-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 12/29/2020] [Indexed: 01/23/2023]
Abstract
PURPOSE Detection of CTCs represents a poor prognostic factor in patients with early and metastatic breast cancer (mBC) and treatment with everolimus-exemestane (E/E) is an established effective treatment in hormone receptor-positive/HER2-negative mBC patients. The effect of E/E on CTCs in mBC patients was prospectively investigated. METHODS CTCs from 50 pre-treated patients with mBC receiving E/E were analyzed using the CellSearch (CS) platform and triple immunofluorescence (IF) staining for cytokeratin, M30 and Ki67 expression to assess their proliferative and apoptotic status. RESULTS CTCs (by CS) were detected in 64% of patients before treatment and E/E administration resulted in their decreased prevalence [(n = 18; 36%, p = 0.004) and (n = 7; 19.4%, p = 0.019) post-1st and post-3rd treatment cycle, respectively] whereas it was significantly increased at disease progression (PD: 61%) compared to post-1st and post-3rd cycle (p = 0.049 and p = 0.021, respectively). Ki67-positive CTCs were detected in 60%, 60%, 17% and 50% of patients before treatment, post-1st, post-3rd cycle and at PD, respectively, while the opposite was observed for M30-positive CTCs (0% at baseline, 10% after the 1st cycle, 50% after the 3rd cycle and 0% at PD). The detection of even ≥ 1 CTC/5 ml after one cycle was associated with decreased PFS (3.3 vs 9.0 months, p = 0.025) whereas the detection of even ≥ 2 CTCs at PD was associated with decreased OS (32.4 vs 19.5 months; p = 0.009). CONCLUSIONS The combination of E/E resulted in early elimination of proliferating CTCs in mBC patients and this effect was associated with a favorable clinical outcome.
Collapse
Affiliation(s)
- Maria Spiliotaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| | - Galatea Kallergi
- Department of Biochemistry, University of Crete Medical School, Heraklion, Crete, Greece
| | - Christos Nikolaou
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| | - Nikolaos Xenidis
- Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece.,Department of Medical Oncology, Medical School, Democritus University of Thrace, Xanthi, Greece
| | - Eleni Politaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| | - Stella Apostolaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| | - Nefeli Georgoulia
- Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece
| | - Filippos Koinis
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece.,Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece.,Department of Medical Oncology, Faculty of Medicine, School of Health Sciences, University Hospital of Larissa & Laboratory of Oncology, University of Thessaly Mezourlo, Larissa, Thessaly, Greece
| | - Nikolaos Tsoukalas
- Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece.,Medical Oncology Unit, NIMITS Hospital, Athens, Greece
| | - Dora Hatzidaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece.,Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece
| | - Athanasios Kotsakis
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece.,Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece.,Department of Medical Oncology, Faculty of Medicine, School of Health Sciences, University Hospital of Larissa & Laboratory of Oncology, University of Thessaly Mezourlo, Larissa, Thessaly, Greece
| | - Vassilis Georgoulias
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece. .,Hellenic Oncology Research Group (HORG), 55 Lombardou str, 11474, Athens, Greece.
| |
Collapse
|
9
|
Halamkova J, Kazda T, Adamkova-Krakorova D, Rybnickova S, Kiss I, Demlova R. Everolimus-related unilateral abdominal lymphedema in a renal cancer patient: A case report. Medicine (Baltimore) 2020; 99:e22634. [PMID: 33080700 PMCID: PMC7571978 DOI: 10.1097/md.0000000000022634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
RATIONALE Unilateral manifestation of lymphedema during everolimus therapy has been described only rarely, mostly in transplant recipients. PATIENT CONCERNS We report the first case of a patient who developed unilateral abdominal lymphedema, during a short period of everolimus treatment for renal cancer. DIAGNOSIS The abdominal asymmetry occurred only on the right side of the abdomen, neither ultrasound nor CT scan detected ascites but showed enlargement of the abdominal wall. The Naranjo Adverse Drug Reaction Probability scale was evaluated, in this case, a score of 6 indicated a probable adverse reaction to everolimus. INTERVENTIONS Discontinuation of everolimus therapy led to immediate alleviation and reduction of the lymphedema, with worsening once again after initiating retreatment with everolimus at a reduced dose. OUTCOMES The patient's lymphedema recovered after discontinuation of everolimus. LESSONS This rare case demonstrates the importance of the selection of mammalian target of rapamycin inhibitors using caution, especially for patients with a high risk of developing lymphedema.
Collapse
Affiliation(s)
- Jana Halamkova
- Department of Cancer Comprehensive Care, Masaryk Memorial Cancer Institute, Faculty of Medicine Masaryk University
- Department of Medical Ethics, Faculty of Medicine, Masaryk University
| | - Tomas Kazda
- Department of Radiation Oncology, Masaryk Memorial Cancer Institute, Faculty of Medicine Masaryk University
| | | | | | - Igor Kiss
- Department of Cancer Comprehensive Care, Masaryk Memorial Cancer Institute, Faculty of Medicine Masaryk University
| | - Regina Demlova
- Clinical Trial Unit, Masaryk Memorial Cancer Institute
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
10
|
Torniai M, Scortichini L, Tronconi F, Rubini C, Morgese F, Rinaldi S, Mazzanti P, Berardi R. Systemic treatment for lung carcinoids: from bench to bedside. Clin Transl Med 2019; 8:22. [PMID: 31273555 PMCID: PMC6609661 DOI: 10.1186/s40169-019-0238-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 06/21/2019] [Indexed: 12/13/2022] Open
Abstract
In the huge spectrum of lung neuroendocrine neoplasms, typical and atypical carcinoids should be considered as a separate biological entity from poorly differentiated forms, harboring peculiar molecular alterations. Despite their indolent behavior, lung carcinoids correlate with a worse survival. To date, only limited therapeutic options are available and novel drugs are strongly needed. In this work, we extensively reviewed scientific literature exploring available therapeutic options, new molecular targets and future perspectives in the management of well differentiated neoplasms of bronchopulmonary tree. Systemic therapy represents the main option in advanced and unresectable disease; accepted choices are somatostatin analogs, peptide receptor radionuclide therapy, everolimus and chemotherapy. To date, an univocal treatment strategy has not been identified yet, thus tailored therapeutic algorithms should consider treatment efficacy as well as safety profiles. Several molecular alterations found in carcinoid tumors might act as molecular targets leading to development of new therapeutic options. Further studies are necessary to identify new potential “druggable” molecular targets in the selected subset of low-grade lung carcinoids. Furthermore, evaluating the available therapies in more homogeneous population might improve their efficacy through a perfect tailoring of treatment options.
Collapse
Affiliation(s)
- Mariangela Torniai
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126, Ancona, Italy
| | - Laura Scortichini
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126, Ancona, Italy
| | - Francesca Tronconi
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126, Ancona, Italy
| | - Corrado Rubini
- Section of Pathological Anatomy and Histopathology, Department of Neuroscience, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Ancona, Italy
| | - Francesca Morgese
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126, Ancona, Italy
| | - Silvia Rinaldi
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126, Ancona, Italy
| | - Paola Mazzanti
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126, Ancona, Italy
| | - Rossana Berardi
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126, Ancona, Italy.
| |
Collapse
|
11
|
Abstract
Pharmacodynamic (PD) monitoring may complement routine pharmacokinetic monitoring of mTOR inhibitors (mTORis) in an attempt to better guide individualized sirolimus (SRL) or everolimus (EVR) treatment after organ transplantation. This review focuses on current knowledge about PD biomarkers for personalized mTORi therapies. Different strategies have already been used in the evaluation of the pharmacodynamics of SRL and EVR as a proxy for their effects on the immune response after transplantation. These include measuring p70S6K (70 kDa ribosomal protein S6 kinase) activity, p70S6K phosphorylation (P-p70S6K), or P-S6 protein expression. Compared with Western blot and ELISA, phosphoflow cytometry can detect phosphorylated proteins and differentiate activation-induced changes of signaling molecules inside the cell from unstimulated populations of identical cells in the same sample. Alternatively, in patients receiving a combined therapy, the other PD approach is to consider biomarkers such as NFAT residual expression for calcineurin inhibitors or to evaluate nonspecific effects of the drugs such as lymphocyte proliferation, interleukin synthesis, specific peripheral blood T regulatory subsets, or lymphocyte surface antigens, which have the advantage to reflect the overall immunosuppressive status achieved. Although limited, the available data on mTOR pathway biomarkers seem promising. Before clinical implementation, the analytical methodologies must be standardized and cross-validated, and the selected biomarkers will have to demonstrate their clinical utility for SRL or EVR dose individualization in multicenter clinical trials.
Collapse
|
12
|
Hurvitz S, Singh R, Adams B, Taguchi JA, Chan D, Dichmann RA, Castrellon A, Hu E, Berkowitz J, Mani A, DiCarlo B, Callahan R, Smalberg I, Wang X, Meglar I, Martinez D, Hobbs E, Slamon DJ. Phase Ib/II single-arm trial evaluating the combination of everolimus, lapatinib and capecitabine for the treatment of HER2-positive breast cancer with brain metastases (TRIO-US B-09). Ther Adv Med Oncol 2018; 10:1758835918807339. [PMID: 30542377 PMCID: PMC6236634 DOI: 10.1177/1758835918807339] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/18/2018] [Indexed: 01/01/2023] Open
Abstract
Background: Improving outcomes for patients with human epidermal growth factor 2-positive
(HER2+) central nervous system (CNS) metastases remains an unmet clinical
need. This trial evaluated a novel combination of everolimus, lapatinib and
capecitabine for this disease. Methods: Patients with trastuzumab-pretreated, HER2+ breast cancer brain metastasis
without prior therapy with a mammalian target of rapamycin (mTOR) inhibitor
were eligible. Patients received lapatinib and everolimus daily
(continuously) and capecitabine twice daily (d1–14) in 21-d cycles. The
primary endpoint was the 12-week CNS objective response rate (ORR).
Secondary endpoints included safety, progression-free survival (PFS),
overall survival (OS), best CNS ORR and extra-CNS ORR. Results: A total of 19 participants were enrolled and treated with ⩾1 dose of the
study drug. The median age was 58.5 years, the median number of therapies
for metastatic breast cancer was 2.5 (0–11). Pretrial, 74% of participants
had received prior lapatinib, capecitabine or both. A total of 63% had
received previous CNS radiation or surgical resection and CNS radiation. The
maximum tolerated doses were lapatinib at 1000 mg, everolimus at 10 mg, and
capecitabine at 1000 mg/m2. Phase II proceeded with capecitabine
at 750 mg/m2 due to better tolerability. The most common grade
3/4 adverse events were mucositis (16%), diarrhea, fatigue, and hypokalemia
(11% each). Of 11 participants evaluable for 12-week CNS ORR, 3 (27%) had
partial response and 7 (64%) had stable disease. The best CNS ORR in
eligible participants was 28% (5/18). The median PFS and OS were 6.2 and
24.2 months, respectively. Conclusions: This novel triplet combination of lapatinib, everolimus, and capecitabine is
well tolerated and yielded a 27% response rate in the CNS at 12 weeks in
heavily pretreated participants. Larger studies are warranted to further
evaluate this regimen. Trial registration: ClinicalTrials.gov: NCT01783756. Registered 05 February 2013, https://clinicaltrials.gov/ct2/show/NCT01783756
Collapse
Affiliation(s)
- Sara Hurvitz
- University of California, Los Angeles, 10945 Le Conte Avenue, PVUB Suite 3360, Los Angeles, CA 90095, USA
| | - Rashi Singh
- University of California, Irvine, Irvine, CA, USA
| | - Brad Adams
- University of California, Los Angeles, Los Angeles, CA, USA
| | | | - David Chan
- Torrance Health Association DBA, Torrance Memorial Physician Network, Redondo Beach, CA, USA
| | | | | | - Eddie Hu
- University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Aruna Mani
- Memorial Healthcare System, Hollywood, FL, USA
| | - Brian DiCarlo
- San Luis Obispo Oncology and Hematology Health Center, San Luis Obispo, CA, USA Coastal Integrative Cancer Care, San Luis Obispo, CA
| | - Rena Callahan
- University of California, Los Angeles, Los Angeles, CA, USA
| | - Ira Smalberg
- Tower Imaging Medical Group, Los Angeles, CA Tower Saint John's Imaging, Santa Monica, CA, USA
| | - Xiaoyan Wang
- University of California, Los Angeles, Los Angeles, CA, USA
| | - Ivana Meglar
- University of California, Los Angeles, Los Angeles, CA, USA
| | - Diego Martinez
- University of California, Los Angeles, Los Angeles, CA, USA
| | - Evthokia Hobbs
- University of California, Los Angeles, Los Angeles, CA, USA
| | | |
Collapse
|
13
|
RhoA G17V is sufficient to induce autoimmunity and promotes T-cell lymphomagenesis in mice. Blood 2018; 132:935-947. [DOI: 10.1182/blood-2017-11-818617] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 05/07/2018] [Indexed: 11/20/2022] Open
Abstract
Key Points
Expression of RhoA G17V in CD4+ cells results in cellular and humoral autoimmunity. RhoA G17V expression with Tet2 loss induces T-cell lymphomas with features of AITL.
Collapse
|
14
|
A pharmacodynamic study of sirolimus and metformin in patients with advanced solid tumors. Cancer Chemother Pharmacol 2018; 82:309-317. [PMID: 29948021 DOI: 10.1007/s00280-018-3619-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 06/04/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Sirolimus is a mammalian target of rapamycin (mTOR) inhibitor. Metformin may potentiate mTOR inhibition by sirolimus while mitigating its adverse effects. We conducted a pilot study to test this hypothesis. METHODS Patients with advanced solid tumor were treated with sirolimus for 7 days followed by randomization to either sirolimus with metformin (Arm A) or sirolimus (Arm B) until day 21. From day 22 onwards, all patients received sirolimus and metformin. The primary aim was to compare the change in phospho-p70S6K (pp70S6K) in peripheral blood mononuclear cells (PBMC) from day 8 to day 22 using a two-sample t test. Secondary aims were objective response rate, toxicity, and other serum pharmacodynamic biomarkers (e.g., fasting glucose, triglycerides, insulin, C-peptide, IGF-1, IGF-1R, IGF-BP, and leptin). RESULTS 24 patients were enrolled, with 18 evaluable for the primary endpoint. There was no significant difference in mean change in pp70S6K in arm A vs. arm B (- 0.12 vs. - 0.16; P = 0.64). Similarly, there were no significant differences in other serum pharmacodynamic biomarkers. There were no partial responses. There were no dose-limiting or unexpected toxicities. CONCLUSIONS Adding metformin to sirolimus, although well tolerated, was not associated with significant changes in pp70S6K in PBMC or other serum pharmacodynamic biomarkers. IMPACT Combining metformin with sirolimus did not improve mTOR inhibition.
Collapse
|
15
|
Lee L, Ito T, Jensen RT. Everolimus in the treatment of neuroendocrine tumors: efficacy, side-effects, resistance, and factors affecting its place in the treatment sequence. Expert Opin Pharmacother 2018; 19:909-928. [PMID: 29757017 PMCID: PMC6064188 DOI: 10.1080/14656566.2018.1476492] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Since the initial approval of everolimus in 2011, there have been a number of important changes in therapeutic/diagnostic modalities as well as classification/staging systems of neuroendocrine tumors (NETs), which can significantly impact the use of everolimus in patients with advanced NETs. Areas covered: The efficacy of everolimus monotherapy and combination therapy demonstrated in clinical studies involving patients with advanced NETs are reviewed. Several factors affecting everolimus use are described including: the development and routine use of NET classification/staging systems; widespread use of molecular imaging modalities; side effects; drug resistance; and the availability of other treatment options. Furthermore, the current position of everolimus in the treatment approach is discussed, taking into account the recommendations from the recent guidelines. Expert opinion: Although everolimus demonstrated its high efficacy and tolerability in the RADIANT trials and other clinical studies, there still remain a number of controversies related to everolimus treatment in the management of NETs. The synergistic anti-growth effect of other agents in combination with everolimus or its effect on overall survival have not been established. The appropriate order of the use of everolimus in the treatment of advanced NETs still remains unclear, which needs to be defined in further studies and will be addressed in the new guidelines.
Collapse
Affiliation(s)
- Lingaku Lee
- a Digestive Diseases Branch , National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda , MD , USA
| | - Tetsuhide Ito
- b Neuroendocrine Tumor Centre , Fukuoka Sanno Hospital, International University of Health and Welfare , Fukuoka , Japan
| | - Robert T Jensen
- a Digestive Diseases Branch , National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
16
|
Earwaker P, Anderson C, Willenbrock F, Harris AL, Protheroe AS, Macaulay VM. RAPTOR up-regulation contributes to resistance of renal cancer cells to PI3K-mTOR inhibition. PLoS One 2018; 13:e0191890. [PMID: 29389967 PMCID: PMC5794101 DOI: 10.1371/journal.pone.0191890] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 01/12/2018] [Indexed: 02/04/2023] Open
Abstract
The outlook for patients with advanced renal cell cancer (RCC) has been improved by targeted agents including inhibitors of the PI3 kinase (PI3K)-AKT-mTOR axis, although treatment resistance is a major problem. Here, we aimed to understand how RCC cells acquire resistance to PI3K-mTOR inhibition. We used the RCC4 cell line to generate a model of in vitro resistance by continuous culture in PI3K-mTOR kinase inhibitor NVP-BEZ235 (BEZ235, Dactolisib). Resistant cells were cross-resistant to mTOR inhibitor AZD2014. Sensitivity was regained after 4 months drug withdrawal, and resistance was partially suppressed by HDAC inhibition, supporting an epigenetic mechanism. BEZ235-resistant cells up-regulated and/or activated numerous proteins including MET, ABL, Notch, IGF-1R, INSR and MEK/ERK. However, resistance was not reversed by inhibiting or depleting these pathways, suggesting that many induced changes were passengers not drivers of resistance. BEZ235 blocked phosphorylation of mTOR targets S6 and 4E-BP1 in parental cells, but 4E-BP1 remained phosphorylated in resistant cells, suggesting BEZ235-refractory mTORC1 activity. Consistent with this, resistant cells over-expressed mTORC1 component RAPTOR at the mRNA and protein level. Furthermore, BEZ235 resistance was suppressed by RAPTOR depletion, or allosteric mTORC1 inhibitor rapamycin. These data reveal that RAPTOR up-regulation contributes to PI3K-mTOR inhibitor resistance, and suggest that RAPTOR expression should be included in the pharmacodynamic assessment of mTOR kinase inhibitor trials.
Collapse
Affiliation(s)
| | | | | | - Adrian L. Harris
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Trust, Churchill Hospital, Oxford, United Kingdom
| | - Andrew S. Protheroe
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Trust, Churchill Hospital, Oxford, United Kingdom
| | - Valentine M. Macaulay
- Department of Oncology, Oxford, United Kingdom
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Trust, Churchill Hospital, Oxford, United Kingdom
| |
Collapse
|
17
|
Molecular mechanism of inhibitory effects of bovine lactoferrin on the growth of oral squamous cell carcinoma. PLoS One 2018; 13:e0191683. [PMID: 29381751 PMCID: PMC5790278 DOI: 10.1371/journal.pone.0191683] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 01/09/2018] [Indexed: 12/18/2022] Open
Abstract
Background Lactoferrin (LF), a member of the transferrin family, recently has been demonstrated to have anticancer effects on various cancers including oral squamous cell carcinoma (OSCC). However, little is known about the underlying mechanisms of its effects on OSCC. Therefore, we aimed to investigate the mechanism of the suppressive effects of bovine LF (bLF) on the growth of OSCC cells. Methods In the current study, HSC2, HSC3, HSC4 and normal human oral keratinocytes (RT7) cell lines were tested with bLF 1, 10, and 100 μg/ml. The effects and detail mechanisms of bLF on proliferation and apoptosis of cells were investigated using flow cytometry and western blotting. Results We found that bLF (1, 10, and 100 μg/ml) induced activation of p53, a tumor suppressor gene, is associated with the induction of cell cycle arrest in G1/S phase and apoptosis in OSCC. Moreover, bLF downregulated the phosphorylation of Akt and activated suppressor of cytokine signaling 3 (SOCS3), thereby attenuating multiple signaling pathways including mTOR/S6K and JAK/STAT3. Interestingly, we revealed that bLF exerted its effect selectively against HSC3 but not on RT7 via different effects on the phosphorylation status of NF-κB and Akt. Conclusion This is the first report showing that bLF selectively suppresses proliferation through mTOR/S6K and JAK/STAT3 pathways and induction of apoptosis in OSCC. This study provides important new findings, which might be useful in the prevention and treatment of OSCC.
Collapse
|
18
|
Co-selected mutations in VCP: a novel mechanism of resistance to VCP inhibitors. Cell Death Dis 2018; 9:35. [PMID: 29348605 PMCID: PMC5833740 DOI: 10.1038/s41419-017-0049-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 09/22/2017] [Accepted: 09/25/2017] [Indexed: 11/08/2022]
|
19
|
Ghidini M, Petrelli F, Ghidini A, Tomasello G, Hahne JC, Passalacqua R, Barni S. Clinical development of mTor inhibitors for renal cancer. Expert Opin Investig Drugs 2017; 26:1229-1237. [DOI: 10.1080/13543784.2017.1384813] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Michele Ghidini
- Oncology Unit, Oncology Department, ASST Ospedale di Cremona, Cremona, Italy
| | - Fausto Petrelli
- Oncology Unit, Oncology Department, ASST Bergamo Ovest, Treviglio, Italy
| | | | - Gianluca Tomasello
- Oncology Unit, Oncology Department, ASST Ospedale di Cremona, Cremona, Italy
| | - Jens Claus Hahne
- Laboratory of Gastrointestinal Cancer Biology and Genomics, Division of Molecular Pathology, The Institute of Cancer Research, Sutton, UK
| | - Rodolfo Passalacqua
- Oncology Unit, Oncology Department, ASST Ospedale di Cremona, Cremona, Italy
| | - Sandro Barni
- Oncology Unit, Oncology Department, ASST Bergamo Ovest, Treviglio, Italy
| |
Collapse
|
20
|
Hatano T, Inaba H, Endo K, Egawa S. Intermittent everolimus administration for renal angiomyolipoma associated with tuberous sclerosis complex. Int J Urol 2017; 24:780-785. [PMID: 28905429 DOI: 10.1111/iju.13428] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/02/2017] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To evaluate the effects and utility of intermittent everolimus treatment for renal angiomyolipoma associated with tuberous sclerosis complex. METHODS We investigated a total of 26 patients with tuberous sclerosis complex who had angiomyolipoma ≥4 cm in diameter. For each patient, we analyzed the reduction in the size of the angiomyolipoma, the change in size after everolimus withdrawal, the size reduction rate on everolimus readministration and adverse events caused by everolimus. The volume of angiomyolipoma was measured using abdominal computed tomography or magnetic resonance imaging. Adverse events were evaluated according to CTCAE v4.0-JCOG. RESULTS The average size reduction rate of angiomyolipoma in the initial treatment with everolimus was 67%. Eight patients (31%) did not have enlarged angiomyolipoma after everolimus withdrawal. The other 18 patients (69%) restarted everolimus treatment because of enlargement of the angiomyolipoma. The average size reduction rate of angiomyolipoma in the everolimus retreatment group was 61%, which was equivalent to the rate for the initial treatment. There were fewer adverse events during everolimus retreatment than in the initial treatment. CONCLUSIONS This is the first report regarding intermittent everolimus treatment for renal angiomyolipoma associated with tuberous sclerosis complex. This treatment is effective for tumor control and adverse event management. This beneficial treatment option for patients can minimize the drug dosage and the occurrence of adverse events.
Collapse
Affiliation(s)
- Takashi Hatano
- Department of Urology, JR Tokyo General Hospital, Tokyo, Japan
| | - Hiroyuki Inaba
- Department of Urology, JR Tokyo General Hospital, Tokyo, Japan
| | - Katsuhisa Endo
- Department of Urology, JR Tokyo General Hospital, Tokyo, Japan
| | - Shin Egawa
- Department of Urology, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
21
|
Bajpai J, Ramaswamy A, Chandrasekharan A, Mishra S, Shet T, Gupta S, Badwe RA. Activation of phosphoinositide 3-kinase/Akt/mechanistic target of rapamycin pathway and response to everolimus in endocrine receptor-positive metastatic breast cancer - A retrospective pilot analysis and viewpoint. South Asian J Cancer 2017; 6:102-105. [PMID: 28975114 PMCID: PMC5615875 DOI: 10.4103/sajc.sajc_113_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Introduction: Biomarkers predictive of response to mechanistic target of rapamycin (mTOR) inhibitor, everolimus, in endocrine receptor (ER)-positive metastatic breast cancer (MBC) are a work in progress. We evaluated the feasibility of directly measuring mTOR activity and phosphatase and tensin homolog (PTEN) expression and correlating their expression with response and survival. Materials and Methods: MBC patients who received everolimus with endocrine therapy (ET) after progression on an aromatase inhibitor and had adequate tissue preservation for estimation of mTOR activity and PTEN expression were selected for analysis from a prospectively maintained database. Progression-free survival (PFS) and overall survival (OS) were estimated by Kaplan–Meier method, and correlation between mTOR activity and PTEN expression with survival was done by log-rank test. Results: Thirteen ER-positive MBC patients were available for analysis. PTEN expression was lost in 11/13 (84.6%) patients and retained in 2/13 patients (15.4%). mTOR activity was absent in four patients (30.7%), weak in six patients (46.1%), and moderate in 3 patients (23.2%). Median PFS for the entire population was 2.5 months while median OS was not reached. Patients with an absent mTOR activity showed a longer PFS (5 vs. 1.5 vs. 2 months) than those with weak and moderate activity, respectively (P = 0.043). There was no correlation between loss of PTEN expression and PFS. Conclusions: Measurement of direct mTOR activity in patients with MBC receiving everolimus/ET combination appears feasible. Absent mTOR activity may predict for longer PFS with everolimus-ET combination and requires further study.
Collapse
Affiliation(s)
- Jyoti Bajpai
- Deparment of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Anant Ramaswamy
- Deparment of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Arun Chandrasekharan
- Deparment of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Surya Mishra
- Deparment of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Tanuja Shet
- Deparment of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Sudeep Gupta
- Deparment of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - R A Badwe
- Deparment of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
22
|
Maiorana A, Dionisi-Vici C. Hyperinsulinemic hypoglycemia: clinical, molecular and therapeutical novelties. J Inherit Metab Dis 2017; 40:531-542. [PMID: 28656511 DOI: 10.1007/s10545-017-0059-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/25/2017] [Accepted: 05/29/2017] [Indexed: 01/01/2023]
Abstract
Hyperinsulinemic hypoglycemia (HI) is the most common cause of hypoglycemia in children. Impairment of cellular pathways involved in insulin secretion from pancreatic β-cells, broadly classified as channelopathies and metabolopathies, have been discovered in the past two decades. The increasing use of NGS target panels, combined with clinical, biochemical and imaging findings allows differentiating the diagnostic management of children with focal forms, surgically curable, from those with diffuse forms, more conservatively treated with pharmacological and nutritional interventions. Specific approaches according to the subtype of HI have been established and novel therapies are currently under investigation. Despite diagnostic and therapeutic advances, HI remains an important cause of morbidity in children, still accounting for 26-44% of permanent intellectual disabilities, especially in neonatal-onset patients. Initial insult from recurrent hypoglycemia in early life greatly contributes to the poor outcomes. Therefore, patients need to be rapidly identified and treated aggressively, and require at follow-up a complex and regular monitoring, managed by a multidisciplinary HI team. This review gives an overview on the more recent diagnostic and therapeutic tools, on the novel drug and nutritional therapies, and on the long-term neurological outcomes.
Collapse
Affiliation(s)
- Arianna Maiorana
- Division of Metabolic Diseases, Department of Pediatric Specialties, Bambino Gesù Children's Hospital, Piazza S. Onofrio 4, 00165, Rome, Italy.
| | - Carlo Dionisi-Vici
- Division of Metabolic Diseases, Department of Pediatric Specialties, Bambino Gesù Children's Hospital, Piazza S. Onofrio 4, 00165, Rome, Italy
| |
Collapse
|
23
|
Ji S, Lin W, Wang L, Ni Z, Jin F, Zha X, Fei G. Combined Targeting of mTOR and Akt Using Rapamycin and MK-2206 in The Treatment of Tuberous Sclerosis Complex. J Cancer 2017; 8:555-562. [PMID: 28367235 PMCID: PMC5370499 DOI: 10.7150/jca.17205] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/29/2016] [Indexed: 12/11/2022] Open
Abstract
Tuberous sclerosis complex (TSC), caused by loss-of-function mutations in the TSC1 or TSC2 genes, is an autosomal dominant disease characterized by benign tumor formation in multiple organs. Hyperactivation of mammalian target of rapamycin (mTOR) is the primary alteration underlying TSC tumor. Thus, rapamycin, as an mTOR specific inhibitor, has been assumed as a potential drug for the treatment of TSC. However, its application in TSC patients has been limited due to side effects. By analyzing Tsc1- or Tsc2-null mouse embryonic fibroblasts (MEFs), we found that loss of TSC1 or TSC2 led to a decreased sensitivity to MK-2206, a novel allosteric Akt inhibitor. Ectopic expression of a constitutively activated Akt (myristoylated Akt-1, myrAkt-1) sensitized Tsc2-null and Tsc1-null MEFs to MK-2206. Furthermore, MK-2206 increased the cytotoxicity of rapamycin in Tsc1-/- or Tsc2-/- MEFs. Moreover, the benefit of the combinatorial treatment was also demonstrated in a TSC xenograft mouse model. We conclude that the combination of rapamycin and MK-2206 may be utilized as a new therapeutic regimen for TSC.
Collapse
Affiliation(s)
- Shuang Ji
- Pulmonary Department, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China;; Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Wei Lin
- Department of Stomatology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China;; Department of Prosthodontics, Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Li Wang
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Zhaofei Ni
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Fuquan Jin
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Guanghe Fei
- Pulmonary Department, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
24
|
Abstract
In 2014, the Immunosuppressive Drugs Scientific Committee of the International Association of Therapeutic Drug Monitoring and Clinical Toxicology called a meeting of international experts to provide recommendations to guide therapeutic drug monitoring (TDM) of everolimus (EVR) and its optimal use in clinical practice. EVR is a potent inhibitor of the mammalian target of rapamycin, approved for the prevention of organ transplant rejection and for the treatment of various types of cancer and tuberous sclerosis complex. EVR fulfills the prerequisites for TDM, having a narrow therapeutic range, high interindividual pharmacokinetic variability, and established drug exposure-response relationships. EVR trough concentrations (C0) demonstrate a good relationship with overall exposure, providing a simple and reliable index for TDM. Whole-blood samples should be used for measurement of EVR C0, and sampling times should be standardized to occur within 1 hour before the next dose, which should be taken at the same time everyday and preferably without food. In transplantation settings, EVR should be generally targeted to a C0 of 3-8 ng/mL when used in combination with other immunosuppressive drugs (calcineurin inhibitors and glucocorticoids); in calcineurin inhibitor-free regimens, the EVR target C0 range should be 6-10 ng/mL. Further studies are required to determine the clinical utility of TDM in nontransplantation settings. The choice of analytical method and differences between methods should be carefully considered when determining EVR concentrations, and when comparing and interpreting clinical trial outcomes. At present, a fully validated liquid chromatography tandem mass spectrometry assay is the preferred method for determination of EVR C0, with a lower limit of quantification close to 1 ng/mL. Use of certified commercially available whole-blood calibrators to avoid calibration bias and participation in external proficiency-testing programs to allow continuous cross-validation and proof of analytical quality are highly recommended. Development of alternative assays to facilitate on-site measurement of EVR C0 is encouraged.
Collapse
|
25
|
Verlingue L, Dugourd A, Stoll G, Barillot E, Calzone L, Londoño‐Vallejo A. A comprehensive approach to the molecular determinants of lifespan using a Boolean model of geroconversion. Aging Cell 2016; 15:1018-1026. [PMID: 27613445 PMCID: PMC6398530 DOI: 10.1111/acel.12504] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2016] [Indexed: 12/11/2022] Open
Abstract
Altered molecular responses to insulin and growth factors (GF) are responsible for late‐life shortening diseases such as type‐2 diabetes mellitus (T2DM) and cancers. We have built a network of the signaling pathways that control S‐phase entry and a specific type of senescence called geroconversion. We have translated this network into a Boolean model to study possible cell phenotype outcomes under diverse molecular signaling conditions. In the context of insulin resistance, the model was able to reproduce the variations of the senescence level observed in tissues related to T2DM's main morbidity and mortality. Furthermore, by calibrating the pharmacodynamics of mTOR inhibitors, we have been able to reproduce the dose‐dependent effect of rapamycin on liver degeneration and lifespan expansion in wild‐type and HER2–neu mice. Using the model, we have finally performed an in silico prospective screen of the risk–benefit ratio of rapamycin dosage for healthy lifespan expansion strategies. We present here a comprehensive prognostic and predictive systems biology tool for human aging.
Collapse
Affiliation(s)
- Loic Verlingue
- Institut Curie CNRS, UMR3244 Telomere and Cancer Laboratory PSL Research University 75005 Paris France
- Department of Medical Oncology Institut Curie 75005 Paris France
| | - Aurélien Dugourd
- Institut Curie Mines Paris Tech, Inserm, U900 PSL Research University F‐75005 Paris France
| | - Gautier Stoll
- Sorbonne Paris Cité Université Paris Descartes 12 Rue de l'École de Médecine 75006 Paris France
- Equipe 11 labellisée Ligue contre le Cancer INSERM U 1138 Centre de Recherche des Cordeliers 15 rue de l'Ecole de Médecine 75006 Paris France
- Université Pierre et Marie Curie 4 Place Jussieu 75005 Paris France
| | - Emmanuel Barillot
- Institut Curie Mines Paris Tech, Inserm, U900 PSL Research University F‐75005 Paris France
| | - Laurence Calzone
- Institut Curie Mines Paris Tech, Inserm, U900 PSL Research University F‐75005 Paris France
| | - Arturo Londoño‐Vallejo
- Institut Curie CNRS, UMR3244 Telomere and Cancer Laboratory PSL Research University 75005 Paris France
- UPMC Univ Paris 06 CNRS, UMR3244 Sorbonne Universités 75005 Paris France
| |
Collapse
|
26
|
Rahman MA, Bishayee K, Sadra A, Huh SO. Oxyresveratrol activates parallel apoptotic and autophagic cell death pathways in neuroblastoma cells. Biochim Biophys Acta Gen Subj 2016; 1861:23-36. [PMID: 27815218 DOI: 10.1016/j.bbagen.2016.10.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 09/05/2016] [Accepted: 10/29/2016] [Indexed: 01/31/2023]
Abstract
BACKGROUND Drug resistance from apoptosis is a challenging issue with different cancer types, and there is an interest in identifying other means of inducing cytotoxicity. Here, treatment of neuroblastoma cells with oxyresveratrol (OXYRES), a natural antioxidant, led to dose-dependent cell death and increased autophagic flux along with activation of caspase-dependent apoptosis. METHODS For cell viability, we performed the CCK-8 assay. Protein expression changes were with Western blot and immunocytochemistry. Silencing of proteins was with siRNA. The readouts for cell cycle, mitochondria membrane potential, caspase-3, autophagy and apoptosis were performed with flow cytometry. RESULTS Phosphorylation of p38 MAPK increased with OXYRES treatment and inhibition of p38 reduced autophagy and cell death from OXYRES. In contrast, PI3K/AKT/mTOR signaling decreased in the target cells with OXYRES and inhibition of PI3K or mTOR enhanced OXYRES-mediated cytotoxicity with increased levels of autophagy. Modulation of either of the apoptosis and autophagy flux pathways affected the extent of cell death by OXYRES, but did not affect the indicators of these pathways with respect to each other. Both pathways were independent of ROS generation or p53 activation. CONCLUSION OXYRES led to cell death from autophagy, which was independent of apoptosis induction. The OXYRES effects were due to changes in the activity levels of p38 MAPK and PI3K/AKT/mTOR. GENERAL SIGNIFICANCE With two independent and parallel pathways for cytotoxicity induction in target cells, this study puts forward a potential utility for OXYRES or the pathways it represents as novel means of inducing cell death in neuroblastoma cells.
Collapse
Affiliation(s)
- Md Ataur Rahman
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, South Korea
| | - Kausik Bishayee
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, South Korea
| | - Ali Sadra
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, South Korea
| | - Sung-Oh Huh
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, South Korea.
| |
Collapse
|
27
|
Kurdi A, De Doncker M, Leloup A, Neels H, Timmermans JP, Lemmens K, Apers S, De Meyer GRY, Martinet W. Continuous administration of the mTORC1 inhibitor everolimus induces tolerance and decreases autophagy in mice. Br J Pharmacol 2016; 173:3359-3371. [PMID: 27638766 DOI: 10.1111/bph.13626] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/31/2016] [Accepted: 09/05/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Everolimus is an allosteric inhibitor of the mechanistic target of rapamycin complex 1 (mTORC1) widely known for its potent autophagy stimulating properties. Because everolimus shows poor solubility and stability in aqueous solutions, long-term in vivo administration in preclinical models is challenging. The aim of the present study was to evaluate the effects of short-term and long-term everolimus administration on mTORC1 inhibition and autophagy induction in mice. EXPERIMENTAL APPROACH We developed a vehicle in which everolimus was solubilized and stable at 37°C for at least 1 month. Using osmotic minipumps, GFP microtubule-associated protein light chain 3 transgenic mice were treated continuously either with vehicle or everolimus (1.5 mg·kg-1 per day) for 3 or 28 days. Alternatively, a regimen consisting of intermittent everolimus administration (every other day) for 56 days by oral gavage was used. Autophagy markers and mTORC1 activation status were investigated in the liver. KEY RESULTS As expected, everolimus inhibited mTORC1 and stimulated autophagy in the liver after 3 days of treatment. However, continuous administration for 28 days resulted in hyperactivation of the Akt1-mTORC1 pathway accompanied by a remarkable decrease in autophagy markers. Everolimus given intermittently for 56 days partially rescued mTORC1 sensitivity to the drug but without inducing autophagy. The failure to induce autophagy following long-term everolimus administration was due to uncoupling of the mTORC1 substrate unc-51 like autophagy activating kinase 1. CONCLUSIONS AND IMPLICATIONS Our data encourage the use of intermittent everolimus regimens to prevent tolerance and to extend its activity.
Collapse
Affiliation(s)
- Ammar Kurdi
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Arthur Leloup
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Hugo Neels
- Laboratory for TDM and Toxicology, ZNA Stuivenberg, Antwerp, Belgium.,Toxicological Centre, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Katrien Lemmens
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sandra Apers
- Natural Products and Food Research and Analysis (NatuRA), Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
28
|
Buti S, Leonetti A, Dallatomasina A, Bersanelli M. Everolimus in the management of metastatic renal cell carcinoma: an evidence-based review of its place in therapy. CORE EVIDENCE 2016; 11:23-36. [PMID: 27621699 PMCID: PMC5012611 DOI: 10.2147/ce.s98687] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Introduction Renal cell carcinoma (RCC) is the most common type of kidney cancer in adults, and its pathogenesis is strictly related to altered cellular response to hypoxia, in which mTOR signaling pathway is implicated. Everolimus, an mTOR serine/threonine kinase inhibitor, represents a therapeutic option for the treatment of advanced RCC. Aim The objective of this article is to review the evidence for the treatment of metastatic RCC with everolimus. Evidence review Everolimus was approved for second- and third-line therapy in patients with advanced RCC according to the results of a Phase III pivotal trial that demonstrated a benefit in median progression-free survival of ~2 months compared to placebo after failure of previous lines of therapy, of which at least one was an anti-VEGFR tyrosine kinase inhibitor (TKI). The role of this drug in first-line setting has been investigated in Phase II trials, with no significant clinical benefit, even in combination with bevacizumab. Everolimus activity in non-clear cell RCC is supported by two randomized Phase II trials that confirmed the benefit in second-line setting but not in first line. Recently, two randomized Phase III trials (METEOR and CheckMate 025) demonstrated the inferiority of everolimus in second-line setting compared to the TKI cabozantinib and to the immune checkpoint inhibitor nivolumab, respectively. Moreover, a recent Phase II study demonstrated a significant benefit for the second-line combination treatment with everolimus plus lenvatinib (a novel TKI) in terms of progression-free survival and overall survival compared to the single-agent everolimus. Basing on preclinical data, the main downstream effectors of mTOR cascade, S6RP and its phosphorylated form, could be good predictive biomarkers of response to everolimus. The safety profile of the drug is favorable, with a good cost-effectiveness compared to second-line sorafenib or axitinib, and no significant impact on the quality of life of treated patients has been found. Conclusion Everolimus still represents a current standard of treatment for RCC progressive to previous treatment lines with VEGFR-TKI. The evidence about two new molecules, cabozantinib and nivolumab, successfully tested head-to-head with everolimus in recently published Phase III trials, will determine the shift of everolimus to the third-line setting and subsequent lines of treatment.
Collapse
Affiliation(s)
| | | | - Alice Dallatomasina
- Division of Experimental Oncology, San Raffaele Scientific Institute, Milan, Italy
| | | |
Collapse
|
29
|
Safety analysis, association with response and previous treatments of everolimus and exemestane in 181 metastatic breast cancer patients: A multicenter Italian experience. Breast 2016; 29:96-101. [PMID: 27476084 DOI: 10.1016/j.breast.2016.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 07/04/2016] [Accepted: 07/08/2016] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The everolimus and exemestane combination represents a treatment option for the endocrine sensitive metastatic breast cancer (MBC) patients. The toxicity profile reported in the Bolero 2 trial showed the feasibility in the selected patients. Few data are available for the unselected population. METHODS In order to evaluate the safety in the unselected population of the clinical practice and to evaluate a possible association of toxicities with previous treatments, clinical data from 181 consecutive patients were retrospectively collected. RESULTS Due to toxic events, everolimus dosage was reduced to 5 mg in 27% of patients. No association was found in the analysis between toxicity and number of prior therapies, neither between toxicity and response. In the multivariate analysis the previous exposure to anthracyclines for advanced disease represents the only predictive factor of developing grade ≥2 toxicity (OR = 2.85 CI 95% 1.07-7.59, p = 0.036). CONCLUSIONS The association of everolimus and exemestane has confirmed to be a safe and effective treatment for endocrine sensitive MBC patients even in routine clinical practice. The rate of treatment discontinuation due to toxicity is low and none association between previous number of treatments and response or between toxicity and response was found.
Collapse
|
30
|
Detti B, Francolini G, Becherini C, Olmetto E, Giacomelli I, Scartoni D, Greto D, Baldazzi V, Simontacchi G, Meattini I, Livi L. Complete response in metastatic renal cell carcinoma after radiotherapy and everolimus: a clinical case and review of the literature. J Chemother 2016; 28:432-4. [PMID: 27376403 DOI: 10.1080/1120009x.2016.1173869] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We report the case of a man affected by renal cell carcinoma with vertebral metastases, who presented a radiological complete response after systemic treatment with everolimus.
Collapse
Affiliation(s)
- Beatrice Detti
- a Radioterapia , Azienda Ospedaliero-Universitaria Careggi , Florence , Italy
| | - Giulio Francolini
- a Radioterapia , Azienda Ospedaliero-Universitaria Careggi , Florence , Italy
| | - Carlotta Becherini
- a Radioterapia , Azienda Ospedaliero-Universitaria Careggi , Florence , Italy
| | - Emanuela Olmetto
- a Radioterapia , Azienda Ospedaliero-Universitaria Careggi , Florence , Italy
| | - Irene Giacomelli
- a Radioterapia , Azienda Ospedaliero-Universitaria Careggi , Florence , Italy
| | - Daniele Scartoni
- a Radioterapia , Azienda Ospedaliero-Universitaria Careggi , Florence , Italy
| | - Daniela Greto
- a Radioterapia , Azienda Ospedaliero-Universitaria Careggi , Florence , Italy
| | - Valentina Baldazzi
- a Radioterapia , Azienda Ospedaliero-Universitaria Careggi , Florence , Italy
| | | | - Icro Meattini
- a Radioterapia , Azienda Ospedaliero-Universitaria Careggi , Florence , Italy
| | - Lorenzo Livi
- a Radioterapia , Azienda Ospedaliero-Universitaria Careggi , Florence , Italy
| |
Collapse
|
31
|
Ortolani S, Ciccarese C, Cingarlini S, Tortora G, Massari F. Suppression of mTOR pathway in solid tumors: lessons learned from clinical experience in renal cell carcinoma and neuroendocrine tumors and new perspectives. Future Oncol 2016; 11:1809-28. [PMID: 26075448 DOI: 10.2217/fon.15.81] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The PI3K-AKT-mTOR pathway plays role in the regulation of many cellular processes. Hyperactivation of mTOR signaling has been implicated in human carcinogenesis, representing an attractive target for cancer therapy. Among other cancer subtypes, renal cell carcinoma (RCC) and neuroendocrine tumors are relevant settings in which the deregulation of mTOR pathway is of crucial importance. Different mTOR-inhibitory agents have been developed in recent years. Temsirolimus is approved for advanced RCC; everolimus is registered for the treatment of advanced RCC, pancreatic neuroendocrine tumors and postmenopausal, hormone receptor-positive/HER2-negative, advanced breast cancer. This review is focused on the description of the clinical experience with mTOR-inhibitor agents for the treatment of advanced RCC and neuroendocrine tumors, followed by an excursus on the landscape of the ongoing research in this field.
Collapse
Affiliation(s)
- Silvia Ortolani
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale LA Scuro 10, 37124 Verona, Italy
| | - Chiara Ciccarese
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale LA Scuro 10, 37124 Verona, Italy
| | - Sara Cingarlini
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale LA Scuro 10, 37124 Verona, Italy
| | - Giampaolo Tortora
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale LA Scuro 10, 37124 Verona, Italy
| | - Francesco Massari
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale LA Scuro 10, 37124 Verona, Italy
| |
Collapse
|
32
|
Kurdi A, De Meyer GRY, Martinet W. Potential therapeutic effects of mTOR inhibition in atherosclerosis. Br J Clin Pharmacol 2015; 82:1267-1279. [PMID: 26551391 DOI: 10.1111/bcp.12820] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 12/13/2022] Open
Abstract
Despite significant improvement in the management of atherosclerosis, this slowly progressing disease continues to affect countless patients around the world. Recently, the mechanistic target of rapamycin (mTOR) has been identified as a pre-eminent factor in the development of atherosclerosis. mTOR is a constitutively active kinase found in two different multiprotein complexes, mTORC1 and mTORC2. Pharmacological interventions with a class of macrolide immunosuppressive drugs, called rapalogs, have shown undeniable evidence of the value of mTORC1 inhibition to prevent the development of atherosclerotic plaques in several animal models. Rapalog-eluting stents have also shown extraordinary results in humans, even though the exact mechanism for this anti-atherosclerotic effect remains elusive. Unfortunately, rapalogs are known to trigger diverse undesirable effects owing to mTORC1 resistance or mTORC2 inhibition. These adverse effects include dyslipidaemia and insulin resistance, both known triggers of atherosclerosis. Several strategies, such as combination therapy with statins and metformin, have been suggested to oppose rapalog-mediated adverse effects. Statins and metformin are known to inhibit mTORC1 indirectly via 5' adenosine monophosphate-activated protein kinase (AMPK) activation and may hold the key to exploit the full potential of mTORC1 inhibition in the treatment of atherosclerosis. Intermittent regimens and dose reduction have also been proposed to improve rapalog's mTORC1 selectivity, thereby reducing mTORC2-related side effects.
Collapse
Affiliation(s)
- Ammar Kurdi
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
33
|
D'Arcangelo M, Margetts J, Greystoke A. The use of circulating biomarkers in early clinical trials in patients with cancer. Biomark Med 2015; 9:1011-23. [PMID: 26441037 DOI: 10.2217/bmm.15.51] [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: 01/27/2023] Open
Abstract
The development of targeted therapies has changed the approach to early oncological clinical trial design. Identification of patient populations most likely to derive benefit and the biologically effective dose are now as important as determination of the maximum tolerated dose. Completion of the 'pharmacological audit trail' highlights drugs most likely to progress through to license, so resources can be allocated appropriately. Key to the success of this changing model is the validation/qualification of circulating biomarkers. These might provide a readily accessible and dynamic picture of drug effect, tumor response and toxicity with minimum risk to patients. This review article examines circulating biomarkers currently used in early oncological clinical trials. It considers the evidence for their employment, limitations and challenges for future development.
Collapse
Affiliation(s)
- Manolo D'Arcangelo
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK.,Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, UK
| | - Jane Margetts
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, UK
| | - Alastair Greystoke
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK.,Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, UK
| |
Collapse
|
34
|
Abstract
mTOR (mechanistic target of rapamycin) functions as the central regulator for cell proliferation, growth and survival. Up-regulation of proteins regulating mTOR, as well as its downstream targets, has been reported in various cancers. This has promoted the development of anti-cancer therapies targeting mTOR, namely fungal macrolide rapamycin, a naturally occurring mTOR inhibitor, and its analogues (rapalogues). One such rapalogue, everolimus, has been approved in the clinical treatment of renal and breast cancers. Although results have demonstrated that these mTOR inhibitors are effective in attenuating cell growth of cancer cells under in vitro and in vivo conditions, subsequent sporadic response to rapalogues therapy in clinical trials has promoted researchers to look further into the complex understanding of the dynamics of mTOR regulation in the tumour environment. Limitations of these rapalogues include the sensitivity of tumour subsets to mTOR inhibition. Additionally, it is well known that rapamycin and its rapalogues mediate their effects by inhibiting mTORC (mTOR complex) 1, with limited or no effect on mTORC2 activity. The present review summarizes the pre-clinical, clinical and recent discoveries, with emphasis on the cellular and molecular effects of everolimus in cancer therapy.
Collapse
|
35
|
MacKeigan JP, Krueger DA. Differentiating the mTOR inhibitors everolimus and sirolimus in the treatment of tuberous sclerosis complex. Neuro Oncol 2015; 17:1550-9. [PMID: 26289591 DOI: 10.1093/neuonc/nov152] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 07/11/2015] [Indexed: 02/06/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a genetic autosomal dominant disorder characterized by benign tumor-like lesions, called hamartomas, in multiple organ systems, including the brain, skin, heart, kidneys, and lung. These hamartomas cause a diverse set of clinical problems based on their location and often result in epilepsy, learning difficulties, and behavioral problems. TSC is caused by mutations within the TSC1 or TSC2 genes that inactivate the genes' tumor-suppressive function and drive hamartomatous cell growth. In normal cells, TSC1 and TSC2 integrate growth signals and nutrient inputs to downregulate signaling to mammalian target of rapamycin (mTOR), an evolutionarily conserved serine-threonine kinase that controls cell growth and cell survival. The molecular connection between TSC and mTOR led to the clinical use of allosteric mTOR inhibitors (sirolimus and everolimus) for the treatment of TSC. Everolimus is approved for subependymal giant cell astrocytomas and renal angiomyolipomas in patients with TSC. Sirolimus, though not approved for TSC, has undergone considerable investigation to treat various aspects of the disease. Everolimus and sirolimus selectively inhibit mTOR signaling with similar molecular mechanisms, but with distinct clinical profiles. This review differentiates mTOR inhibitors in TSC while describing the molecular mechanisms, pathogenic mutations, and clinical trial outcomes for managing TSC.
Collapse
Affiliation(s)
- Jeffrey P MacKeigan
- Laboratory of Systems Biology, Van Andel Research Institute, Grand Rapids, Michigan (J.P.M.); Department of Pediatrics, Tuberous Sclerosis Clinic, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio (D.A.K.)
| | - Darcy A Krueger
- Laboratory of Systems Biology, Van Andel Research Institute, Grand Rapids, Michigan (J.P.M.); Department of Pediatrics, Tuberous Sclerosis Clinic, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio (D.A.K.)
| |
Collapse
|
36
|
Long-Term Everolimus Treatment in Individuals With Tuberous Sclerosis Complex: A Review of the Current Literature. Pediatr Neurol 2015; 53:23-30. [PMID: 26092412 DOI: 10.1016/j.pediatrneurol.2014.10.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/02/2014] [Accepted: 10/26/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Tuberous sclerosis complex is a genetic disease usually caused by mutations to either TSC1 or TSC2, where its gene products are involved in the inhibition of the mammalian target of rapamycin pathway. Under normal cellular conditions, mammalian target of rapamycin (mTOR) regulates cell growth and proliferation in response to signals from nutrients or growth factors, but loss of TSC1 or TSC2 leads to overactivation of mTOR and uncontrolled cellular proliferation. Everolimus is an mTOR inhibitor approved for use in a number of indications where mTOR overactivation is implicated, including tuberous sclerosis complex. METHODS AND PATIENTS We conducted a literature search of PubMed to identify published articles about the long-term efficacy and safety of everolimus in patients with tuberous sclerosis complex. RESULTS The short-term efficacy and safety of everolimus in patients with tuberous sclerosis complex has been demonstrated in placebo-controlled trials, and open-label extension studies are ongoing to monitor long-term effects, including safety. Examples of regrowth following discontinuation of mTOR inhibitors suggest that everolimus needs to be given indefinitely to maintain suppression of subependymal giant cell astrocytoma and other tuberous sclerosis complex-associated disease manifestations. No additional safety concerns have been reported to date with long-term administration of everolimus, but published long-term data (>1 year treatment) are currently limited to a small open-label trial and case reports for this relatively rare condition. CONCLUSIONS From the limited data available, long-term administration of everolimus appears feasible with few safety concerns beyond those associated with short-term use. Further investigation is needed to determine the long-term efficacy and safety of everolimus in patients with tuberous sclerosis complex.
Collapse
|
37
|
Rensing N, Han L, Wong M. Intermittent dosing of rapamycin maintains antiepileptogenic effects in a mouse model of tuberous sclerosis complex. Epilepsia 2015; 56:1088-97. [PMID: 26122303 DOI: 10.1111/epi.13031] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2015] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Inhibitors of the mechanistic target of rapamycin (mTOR) pathway have antiepileptogenic effects in preventing epilepsy and pathologic and molecular mechanisms of epileptogenesis in mouse models of tuberous sclerosis complex (TSC). However, long-term treatment with mTOR inhibitors may be required to maintain efficacy and potentially has chronic side effects, such as immunosuppression. Attempts to minimize drug exposure will facilitate translational efforts to develop mTOR inhibitors as antiepileptogenic agents for patients with TSC. In this study, we tested intermittent dosing paradigms of mTOR inhibitors for antiepileptogenic properties in a TSC mouse model. METHODS Western blot analysis of phosphorylation of S6 protein was used to assess the dose- and time-dependence of mTOR inhibition by rapamycin in control mice and conditional knockout mice with inactivation of the Tsc1 gene in glial fibrillary acidic protein (GFAP)-expressing cells (Tsc1(GFAP)CKO mice). Based on the Western blot studies, different dosing paradigms of rapamycin starting at postnatal day 21 were tested for their ability to prevent epilepsy or pathologic abnormalities in Tsc1(GFAP)CKO mice: 4 days of rapamycin only (4-∞), 4 days on-24 days off (4-24), and 4 days on-10 days off (4-10). RESULTS mTOR activity was inhibited by rapamycin in a dose-dependent fashion and recovered to baseline by about 10 days after the last rapamycin dose. The 4-10 and 4-24 dosing paradigms almost completely prevented epilepsy and the 4-10 paradigm inhibited glial proliferation and megalencephaly in Tsc1(GFAP)CKO mice. SIGNIFICANCE Intermittent dosing of rapamycin, with drug holidays of more than 3 weeks, maintains significant antiepileptogenic properties in mouse models of TSC. These findings have important translational applications in developing mTOR inhibitors as antiepileptogenic agents in TSC patients by minimizing drug exposure and potential side effects.
Collapse
Affiliation(s)
- Nicholas Rensing
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, U.S.A
| | - Lirong Han
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, U.S.A
| | - Michael Wong
- Department of Neurology and the Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, U.S.A
| |
Collapse
|
38
|
Lewis L, Taylor M, Suriya Ertugyrovna Y, Kuanysh Shadybayevich N, Kaldygul Kabakovna S, Ramil Zufarovich A. Budget impact analysis of everolimus for the treatment of hormone receptor positive, human epidermal growth factor receptor-2 negative (HER2-) advanced breast cancer in Kazakhstan. J Med Econ 2015; 18:189-99. [PMID: 25415641 DOI: 10.3111/13696998.2014.969432] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE The aim of this study was to determine the budget impact of everolimus (in combination with letrozole/anastrozole) as a second-line treatment for ER+ HER2- negative advanced and metastatic breast cancer in post-menopausal women. RESEARCH DESIGN AND METHODS A cumulative cohort model was developed to estimate the 5-year costs associated with introducing everolimus to the Kazakh healthcare system. Two alternative market share scenarios were compared: with everolimus and without everolimus. PFS and OS data were taken from the trial and extrapolated. The background costs of the pre-progressed and post-progressed health states, drug costs and costs associated with adverse events were included in the model. RESULTS The 5-year results from the budget impact analysis demonstrate that the introduction of everolimus leads to a 12% increase in drug costs, a 2% reduction in pre-progression health state costs, a 1% increase in post-progression health state costs, and a 2% reduction in adverse event costs. The net result is only a modest increase in total costs; a 2.69% increase of T201 million, from T7.5 billion to T7.7 billion over a period of 5 years. CONCLUSIONS The analysis estimated that, if everolimus were to be introduced to the Kazakh healthcare market for the treatment of ER+ HER2- advanced breast cancer, there would be minimal impact upon overall healthcare expenditure. An increase in drug acquisitions costs was almost exactly offset by a reduction in other healthcare costs, due to improved management of the disease.
Collapse
Affiliation(s)
- Lily Lewis
- York Health Economics Consortium Ltd, University of York , York , UK
| | | | | | | | | | | |
Collapse
|
39
|
Acevedo-Gadea C, Hatzis C, Chung G, Fishbach N, Lezon-Geyda K, Zelterman D, DiGiovanna MP, Harris L, Abu-Khalaf MM. Sirolimus and trastuzumab combination therapy for HER2-positive metastatic breast cancer after progression on prior trastuzumab therapy. Breast Cancer Res Treat 2015; 150:157-67. [DOI: 10.1007/s10549-015-3292-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/28/2015] [Indexed: 01/20/2023]
|
40
|
Owonikoko TK, Ramalingam SS, Miller DL, Force SD, Sica GL, Mendel J, Chen Z, Rogatko A, Tighiouart M, Harvey RD, Kim S, Saba NF, Pickens A, Behera M, Fu RW, Rossi MR, Auffermann WF, Torres WE, Bechara R, Deng X, Sun SY, Fu H, Gal AA, Khuri FR. A Translational, Pharmacodynamic, and Pharmacokinetic Phase IB Clinical Study of Everolimus in Resectable Non-Small Cell Lung Cancer. Clin Cancer Res 2015; 21:1859-68. [PMID: 25673697 DOI: 10.1158/1078-0432.ccr-14-1998] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/28/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE The altered PI3K/mTOR pathway is implicated in lung cancer, but mTOR inhibitors have failed to demonstrate efficacy in advanced lung cancer. We studied the pharmacodynamic effects of everolimus in resectable non-small cell lung cancer (NSCLC) to inform further development of these agents in lung cancer. EXPERIMENTAL DESIGN We enrolled 33 patients and obtained baseline tumor biopsy and 2[18F]fluoro-2-deoxy-D-glucose-positron emission tomography/computed tomography (FDG-PET/CT) imaging followed by everolimus treatment (5 or 10 mg daily, up to 28 days), or without intervening treatment for controls. Target modulation by everolimus was quantified in vivo and ex vivo by comparing metabolic activity on paired PET scans and expression of active phosphorylated forms of mTOR, Akt, S6, eIF4e, p70S6K, 4EBP1, and total Bim protein between pretreatment and posttreatment tissue samples. RESULTS There were 23 patients on the treatment arm and 10 controls; median age 64 years; 22 tumors (67%) were adenocarcinomas. There was a dose-dependent reduction in metabolic activity (SUVmax: 29.0%, -21%, -24%; P = 0.014), tumor size (10.1%, 5.8%, -11.6%; P = 0.047), and modulation of S6 (-36.1, -13.7, -77.0; P = 0.071) and pS6 (-41.25, -61.57, -47.21; P = 0.063) in patients treated in the control, 5-mg, and 10-mg cohorts, respectively. Targeted DNA sequencing in all patients along with exome and whole transcriptome RNA-seq in an index patient with hypersensitive tumor was employed to further elucidate the mechanism of everolimus activity. CONCLUSIONS This "window-of-opportunity" study demonstrated measurable, dose-dependent, biologic, metabolic, and antitumor activity of everolimus in early-stage NSCLC.
Collapse
Affiliation(s)
- Taofeek K Owonikoko
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia. Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia. Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Daniel L Miller
- Winship Cancer Institute of Emory University, Atlanta, Georgia. Department of Surgery, Emory University, Atlanta, Georgia
| | - Seth D Force
- Winship Cancer Institute of Emory University, Atlanta, Georgia. Department of Surgery, Emory University, Atlanta, Georgia
| | - Gabriel L Sica
- Winship Cancer Institute of Emory University, Atlanta, Georgia. Department of Pathology, Emory University, Atlanta, Georgia
| | - Jennifer Mendel
- Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Zhengjia Chen
- Winship Cancer Institute of Emory University, Atlanta, Georgia. Department of Statistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Andre Rogatko
- Cedars Sinai Medical Center, Los Angeles, California
| | | | - R Donald Harvey
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia. Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Sungjin Kim
- Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia. Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Allan Pickens
- Department of Surgery, Emory University, Atlanta, Georgia
| | - Madhusmita Behera
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia
| | - Robert W Fu
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia
| | - Michael R Rossi
- Department of Pathology, Emory University, Atlanta, Georgia. Department of Radiation Oncology, Emory University, Atlanta, Georgia
| | | | | | - Rabih Bechara
- Division of Interventional Pulmonology, Emory University, Atlanta, Georgia
| | - Xingming Deng
- Winship Cancer Institute of Emory University, Atlanta, Georgia. Department of Radiation Oncology, Emory University, Atlanta, Georgia
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia. Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Haian Fu
- Winship Cancer Institute of Emory University, Atlanta, Georgia. Department of Pharmacology, Emory University, Atlanta, Georgia
| | - Anthony A Gal
- Winship Cancer Institute of Emory University, Atlanta, Georgia. Department of Pathology, Emory University, Atlanta, Georgia
| | - Fadlo R Khuri
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia. Winship Cancer Institute of Emory University, Atlanta, Georgia.
| |
Collapse
|
41
|
In vitro activity of the mTOR inhibitor everolimus, in a large panel of breast cancer cell lines and analysis for predictors of response. Breast Cancer Res Treat 2015; 149:669-80. [DOI: 10.1007/s10549-015-3282-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/18/2015] [Indexed: 12/21/2022]
|
42
|
Gombos A, Barthélémy P, Awada A. Evaluating the pharmacokinetics and pharmacodynamics of everolimus for treating breast cancer. Expert Opin Drug Metab Toxicol 2015; 11:823-34. [PMID: 25659402 DOI: 10.1517/17425255.2015.1013464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The aberrant activation of the phosphoinositide 3-kinase-Akt-mTOR signaling pathway is a common mechanism of resistance to endocrine therapy and human epidermal growth factor receptor 2 (HER2)-targeted treatments in breast cancer. Data from large clinical trials have shown that the combination of everolimus , an orally bioavailable mTOR inhibitor with exemestane improves outcome of metastatic breast cancer resistant to non-steroidal aromatase inhibitors. On the other hand, the addition of everolimus to trastuzumab in order to overcome resistance did not show meaningful clinical benefit in recent reported Phase III clinical trials. Everolimus has a favorable pharmacokinetic (PK) profile in early breast cancer studies. The association of endocrine therapy and HER2-targeted agents did not influence the main PK parameters of the drugs. AREAS COVERED This review article focuses on the biological rationale of using everolimus in breast cancer and on latest advances in the field of everolimus-based combinations with an emphasis on the PK and pharmacodynamic parameters of the drug throughout different studies. EXPERT OPINION Better identification of patients who sustain benefit or who are resistant to everolimus-based combinations in the treatment of advanced breast cancer remains an unmet need. New combination strategies based on the understanding of resistance mechanisms and intracellular feedback loops should be studied further in the future.
Collapse
Affiliation(s)
- Andrea Gombos
- Université Libre de Bruxelles, Institut Jules Rue Héger-Bordet 1 , Medical Oncology Clinic , Rue Héger 1, Brussels 1000 , Belgium
| | | | | |
Collapse
|
43
|
Pea A, Hruban RH, Wood LD. Genetics of pancreatic neuroendocrine tumors: implications for the clinic. Expert Rev Gastroenterol Hepatol 2015; 9:1407-19. [PMID: 26413978 PMCID: PMC4890468 DOI: 10.1586/17474124.2015.1092383] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Pancreatic neuroendocrine tumors (PanNETs) are a common and deadly neoplasm of the pancreas. Although the importance of genetic alterations in PanNETs has been known for many years, recent comprehensive sequencing studies have greatly expanded our knowledge of neuroendocrine tumorigenesis in the pancreas. These studies have identified specific cellular processes that are altered in PanNETs, highlighted alterations with prognostic implications, and pointed to pathways for targeted therapies. In this review, we will discuss the genetic alterations that play a key role in PanNET tumorigenesis, with a specific focus on those alterations with the potential to change the way patients with these neoplasms are diagnosed and treated.
Collapse
Affiliation(s)
- Antonio Pea
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD USA, The Sol Goldman Pancreatic Cancer Research Center,Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD USA, The Sol Goldman Pancreatic Cancer Research Center,Unit of Surgery B, The Pancreas Institute, University of Verona Hospital Trust, Verona, Italy
| | - Ralph H. Hruban
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD USA, The Sol Goldman Pancreatic Cancer Research Center
| | - Laura D. Wood
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD USA, The Sol Goldman Pancreatic Cancer Research Center
| |
Collapse
|
44
|
Dorff T, Mack PC. The Role of mTOR Inhibitors and PI3K Pathway Blockade in Renal Cell Cancer. KIDNEY CANCER 2015. [DOI: 10.1007/978-3-319-17903-2_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
45
|
Fouladi M, Perentesis JP, Wagner LM, Vinks AA, Reid JM, Ahern C, Thomas G, Mercer CA, Krueger DA, Houghton PJ, Doyle LA, Chen H, Weigel B, Blaney SM. A Phase I Study of Cixutumumab (IMC-A12) in Combination with Temsirolimus (CCI-779) in Children with Recurrent Solid Tumors: A Children's Oncology Group Phase I Consortium Report. Clin Cancer Res 2014; 21:1558-65. [PMID: 25467181 DOI: 10.1158/1078-0432.ccr-14-0595] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 11/03/2014] [Indexed: 11/16/2022]
Abstract
PURPOSE To determine the MTD, dose-limiting toxicities (DLT), pharmacokinetics, and biologic effects of cixutumumab administered in combination with temsirolimus to children with refractory solid tumors. EXPERIMENTAL DESIGN Cixutumumab and temsirolimus were administered intravenously once every 7 days in 28-day cycles. Pharmacokinetic and biology studies, including assessment of mTOR downstream targets in peripheral blood mononuclear cells, were performed during the first cycle. RESULTS Thirty-nine patients, median age 11.8 years (range, 1-21.5), with recurrent solid or central nervous system tumors were enrolled, of whom 33 were fully assessable for toxicity. There were four dose levels, which included two dose reductions and a subsequent intermediated dose escalation: (i) IMC-A12 6 mg/kg, temsirolimus 15 mg/m(2); (ii) IMC-A12 6 mg/kg, temsirolimus 10 mg/m(2); (iii) IMC-A12 4 mg/kg, temsirolimus 8 mg/m(2); and (iv) IMC-A12 6 mg/kg, temsirolimus 8 mg/m(2). Mucositis was the predominant DLT. Other DLTs included hypercholesterolemia, fatigue, thrombocytopenia, and increased alanine aminotransferase. Target inhibition (decreased S6K1 and PAkt) in peripheral blood mononuclear cells was noted at all dose levels. Marked interpatient variability in temsirolimus pharmacokinetic parameters was noted. At 8 mg/m(2), the median temsirolimus AUC was 2,946 ng • h/mL (range, 937-5,536) with a median sirolimus AUC of 767 ng • h/mL (range, 245-3,675). CONCLUSIONS The recommended pediatric phase II doses for the combination of cixutumumab and temsirolimus are 6 mg/kg and 8 mg/m(2), respectively.
Collapse
Affiliation(s)
- Maryam Fouladi
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| | | | - Lars M Wagner
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | - Joel M Reid
- Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Charlotte Ahern
- Children's Oncology Group Operations Center, Arcadia, California
| | | | | | - Darcy A Krueger
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | - L Austin Doyle
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Helen Chen
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | | | - Susan M Blaney
- Texas Children's Cancer Center/Baylor College of Medicine, Houston, Texas
| |
Collapse
|
46
|
Evaluation of the impact of the cancer therapy everolimus on the central nervous system in mice. PLoS One 2014; 9:e113533. [PMID: 25436776 PMCID: PMC4250083 DOI: 10.1371/journal.pone.0113533] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 10/24/2014] [Indexed: 01/13/2023] Open
Abstract
Cancer and treatments may induce cognitive impairments in cancer patients, and the causal link between chemotherapy and cognitive dysfunctions was recently validated in animal models. New cancer targeted therapies have become widely used, and their impact on brain functions and quality of life needs to be explored. We evaluated the impact of everolimus, an anticancer agent targeting the mTOR pathway, on cognitive functions, cerebral metabolism, and hippocampal cell proliferation/vascular density in mice. Adult mice received everolimus daily for 2 weeks, and behavioral tests were performed from 1 week after the last treatment. Everolimus-treated mice displayed a marked reduction in weight gain from the last day of the treatment period. Ex vivo analysis showed altered cytochrome oxidase activity in selective cerebral regions involved in energy balance, food intake, reward, learning and memory modulation, sleep/wake cycle regulation, and arousal. Like chemotherapy, everolimus did not alter emotional reactivity, learning and memory performances, but in contrast to chemotherapy, did not affect behavioral flexibility or reactivity to novelty. In vivo hippocampal neural cell proliferation and vascular density were also unchanged after everolimus treatments. In conclusion, two weeks daily everolimus treatment at the clinical dose did not evoke alteration of cognitive performances evaluated in hippocampal- and prefrontal cortex-dependent tasks that would persist at one to four weeks after the end of the treatment completion. However, acute everolimus treatment caused selective CO modifications without altering the mTOR effector P70S6 kinase in cerebral regions involved in feeding behavior and/or the sleep/wake cycle, at least in part under control of the solitary nucleus and the parasubthalamic region of the hypothalamus. Thus, this area may represent a key target for everolimus-mediating peripheral modifications, which has been previously associated with symptoms such as weight loss and fatigue.
Collapse
|
47
|
Phase II study of temozolomide (TMZ) and everolimus (RAD001) therapy for metastatic melanoma: a North Central Cancer Treatment Group study, N0675. Am J Clin Oncol 2014; 37:369-76. [PMID: 23357973 DOI: 10.1097/coc.0b013e31827b45d4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Mammalian target of rapamycin (mTOR) pathway is activated in malignant melanoma and in situ lesions as opposed to benign nevi. Inhibition of PI3K-Akt-mTOR signaling is implicated in sensitization of melanoma cells to alkylating agents (temozolomide [TMZ]) and inhibition of tumor angiogenesis. METHODS We conducted a single-arm phase II multi-institution cooperative group study to assess the antitumor activity and safety profile of the combination of TMZ and the rapamycin derivative everolimus in patients with metastatic unresectable malignant melanoma. Patients received 10 mg/d of RAD001 for 5 of 7 days (ie, 50 mg/wk) and 200 mg/m/d of TMZ for 5 days each cycle. RESULTS Of the first 39 eligible patients, 17 were PFS-9 successes, for a predetermined threshold of 18/39 patients for a positive trial. Overall, 21 of 48 patients were progression free at 9 weeks, for an event-free survival rate of 44% (95% confidence interval, 29%-59%). The median progression-free survival was 2.4 months and the median overall survival was 8.6 months. Four patients achieved a partial response; the median duration of response was 15.1 months. No complete remissions were observed. Treatment was in general well tolerated with only 1 patient discontinuing therapy due to toxicity (hyperlipidemia). CONCLUSIONS The combination of TMZ and RAD001 was well tolerated but failed to meet/exceed our study threshold for promising clinical activity in patients with metastatic melanoma.
Collapse
|
48
|
Targeting the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway: an emerging treatment strategy for squamous cell lung carcinoma. Cancer Treat Rev 2014; 40:980-9. [PMID: 25037117 DOI: 10.1016/j.ctrv.2014.06.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 05/29/2014] [Accepted: 06/08/2014] [Indexed: 01/19/2023]
Abstract
Squamous cell lung carcinoma accounts for approximately 30% of all non-small cell lung cancers (NSCLCs). Despite progress in the understanding of the biology of cancer, cytotoxic chemotherapy remains the standard of care for patients with squamous cell lung carcinoma, but the prognosis is generally poor. The phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway is one of the most commonly activated signaling pathways in cancer, leading to cell proliferation, survival, and differentiation. It has therefore become a major focus of clinical research. Various alterations in the PI3K/AKT/mTOR pathway have been identified in squamous cell lung carcinoma and a number of agents targeting these alterations are in clinical development for use as single agents and in combination with other targeted and conventional treatments. These include pan-PI3K inhibitors, isoform-specific PI3K inhibitors, AKT inhibitors, mTOR inhibitors, and dual PI3K/mTOR inhibitors. These agents have demonstrated antitumor activity in preclinical models of NSCLC and preliminary clinical evidence is also available for some agents. This review will discuss the role of the PI3K/AKT/mTOR pathway in cancer and how the discovery of genetic alterations in this pathway in patients with squamous cell lung carcinoma can inform the development of targeted therapies for this disease. An overview of ongoing clinical trials investigating PI3K/AKT/mTOR pathway inhibitors in squamous cell lung carcinoma will also be included.
Collapse
|
49
|
Bison SM, Pool SE, Koelewijn SJ, van der Graaf LM, Groen HC, Melis M, de Jong M. Peptide receptor radionuclide therapy (PRRT) with [(177)Lu-DOTA(0),Tyr(3)]octreotate in combination with RAD001 treatment: further investigations on tumor metastasis and response in the rat pancreatic CA20948 tumor model. EJNMMI Res 2014; 4:21. [PMID: 24995150 PMCID: PMC4070081 DOI: 10.1186/s13550-014-0021-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 04/01/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Previously, we reported on the unexpected development of distant metastases in the subcutaneous rat pancreas CA20948 tumor model after 4.5 weeks of treatment with RAD001-only or in combination with [(177)Lu-DOTA(0),Tyr(3)]octreotate ((177)Lu-DOTATATE) (Cancer Res. 73:12-8, 2013). Moreover, the combination therapy was less effective compared to (177)Lu-DOTATATE-only. In the current study, we address the following questions: (1) Why was the combination therapy less effective? Is (177)Lu-DOTATATE tumor uptake affected by pretreatment with RAD001? (2) Could sudden cessation of RAD001 therapy cause the development of distant metastases? (3) Is (177)Lu-DOTATATE an effective treatment option for these metastases? METHODS Lewis rats (HanHsd or SsNHsd substrain with a slight difference in immune response) bearing subcutaneous CA20948 tumors were treated with either 125 or 275 MBq (177)Lu-DOTATATE, RAD001, or their combination. RAD001 was given twice a week for 4.5 or 12 weeks, whereas (177)Lu-DOTATATE was given as a single injection. When combined, RAD001 was started either 3 days prior to or 3 days post administration of (177)Lu-DOTATATE. SPECT/CT was performed to quantify (177)Lu-DOTATATE tumor uptake. Where indicated, primary tumors were surgically removed when tumor size is >6,000 mm(3) to enable monitoring for possible metastasis. If metastases were suspected, an (111)In-DTPA-octreotide SPECT/CT scan was performed. Seven rats with metastases were treated with 400 MBq (177)Lu-DOTATATE. RESULTS Lu-DOTATATE tumor uptake was not significantly affected by RAD001 pretreatment. The occurrence of metastases after RAD001 treatment was not dose dependent in the dose range tested, nor was it related to the duration of RAD001 treatment. In the experiment in which the LEW/SsNsd substrain was used, only 12.5% of RAD001-treated rats showed complete response (CR), compared to 50% tumor regression in the control group. Re-treatment with a high dose of (177)Lu-DOTATATE resulted in CR in only two out of seven animals. CONCLUSION Less effective anti-tumor effects after the combination of RAD001 + (177)Lu-DOTATATE could not be explained by reduced (177)Lu-DOTATATE tumor uptake after RAD001. Our current data support RAD001-induced immune suppression as the reason for this observation. No evidence was found that cessation of RAD001 treatment caused development of metastases. Metastases appeared to be less sensitive to (177)Lu-DOTATATE treatment than primary tumors.
Collapse
Affiliation(s)
- Sander M Bison
- Department of Nuclear Medicine, Erasmus MC, Postbus 2040, Rotterdam 3000, CA, the Netherlands ; Department of Radiology, Erasmus MC, Rotterdam 3000, CA, the Netherlands ; Department of Medical Informatics, Erasmus MC, Rotterdam 3000, CA, the Netherlands
| | - Stefan E Pool
- Department of Nuclear Medicine, Erasmus MC, Postbus 2040, Rotterdam 3000, CA, the Netherlands
| | - Stuart J Koelewijn
- Department of Nuclear Medicine, Erasmus MC, Postbus 2040, Rotterdam 3000, CA, the Netherlands
| | - Linda M van der Graaf
- Department of Nuclear Medicine, Erasmus MC, Postbus 2040, Rotterdam 3000, CA, the Netherlands
| | - Harald C Groen
- Department of Nuclear Medicine, Erasmus MC, Postbus 2040, Rotterdam 3000, CA, the Netherlands ; Department of Radiology, Erasmus MC, Rotterdam 3000, CA, the Netherlands
| | - Marleen Melis
- Department of Nuclear Medicine, Erasmus MC, Postbus 2040, Rotterdam 3000, CA, the Netherlands
| | - Marion de Jong
- Department of Nuclear Medicine, Erasmus MC, Postbus 2040, Rotterdam 3000, CA, the Netherlands ; Department of Radiology, Erasmus MC, Rotterdam 3000, CA, the Netherlands
| |
Collapse
|
50
|
Yee AJ, Hari P, Marcheselli R, Mahindra AK, Cirstea DD, Scullen TA, Burke JN, Rodig SJ, Hideshima T, Laubach JP, Ghobrial IM, Schlossman RL, Munshi NC, Anderson KC, Weller EA, Richardson PG, Raje NS. Outcomes in patients with relapsed or refractory multiple myeloma in a phase I study of everolimus in combination with lenalidomide. Br J Haematol 2014; 166:401-9. [PMID: 24761838 DOI: 10.1111/bjh.12909] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/17/2014] [Indexed: 11/30/2022]
Abstract
Everolimus, an oral mammalian target of rapamycin (mTOR) inhibitor, has been studied in multiple myeloma (MM) but lacks significant single agent activity. Based on preclinical studies showing synergistic activity of mTOR inhibitors with lenalidomide, we studied the combination of lenalidomide and everolimus in relapsed or refractory MM in a phase I clinical trial. We assessed patient samples using gene expression, Western blotting and immunohistochemistry to probe the mTOR pathway. Twenty-six patients were evaluable for toxicity. Dose-limiting toxicities included grade 4 neutropenia and thrombocytopenia. The maximum tolerated dose was lenalidomide 15 mg and everolimus 5 mg for 21 d with a 7 d rest period. Grade 3/4 adverse events included thrombocytopenia (35%) and neutropenia (42%). The overall response rate was 65% (1 complete response + 4 partial response + 10 minimal response). The median progression-free survival was 5·5 months and median overall survival was 29·5 months. Biomarker data demonstrated downregulation of phosphorylated p70S6K. Gene expression profiling suggested activation of mTOR in responders versus non-responders. The combination of lenalidomide and everolimus was well tolerated with predictable toxicities and showed responses in a heavily pretreated population. When confirmed with larger patient numbers, this analysis may guide patient selection for future clinical trials of mTOR inhibition in MM.
Collapse
Affiliation(s)
- Andrew J Yee
- Division of Hematology and Oncology, Massachusetts General Hospital Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|