1
|
Li Y, Du W, Yang R, Wei X, Li H, Zhang X. Copper Chaperone for Superoxide Dismutase Subtypes as a Prognostic Marker in Luminal B Breast Cancer. Clin Med Insights Oncol 2024; 18:11795549231219239. [PMID: 38187458 PMCID: PMC10771053 DOI: 10.1177/11795549231219239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 11/17/2023] [Indexed: 01/09/2024] Open
Abstract
Background Copper chaperone for superoxide dismutase (CCS) is an essential component of the oxidation-reduction system. In breast cancer cells, CCS expression is highly up-regulated, which contributes to cellular proliferation and migration. Breast cancer is a multifaceted disease with different tumor prognoses and responses to clinical treatments, which may be associated with multiple molecular subtypes of CCS. Methods The CCS expression patterns in breast cancer were investigated by TNMplot, cBioPortal, and HPA network database. The correlation of CCS expression with clinicopathological parameters was analyzed using the UALCAN database. The Cancer Genome Atlas (TCGA) data set was used to analyze the Clinical characteristics of CCS in luminal B patients. The bc-GenExMiner database was used to analyze the effects of BReast-CAncer susceptibility gene (BRCA)1/2, TP53 mutation status, estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor (HER) expression on CCS expression. The survival curves and prognostic value of CCS in luminal B breast cancer were performed through Kaplan-Meier curves, univariate and multivariate Cox regression using the PrognoScan, bc-GenExMiner, and Clinical bioinformatics analysis platform. Results We found that CCS expression was associated with patient age, race, ER, and PR status. We also discovered that BRCA1/2 mutations had an effect on CCS expression. The luminal B subtype had the highest CCS expression, which was linked to poor survival compared with other subtypes. In addition, Kaplan-Meier curve analysis showed that luminal B patients with high CCS mRNA expression showed a poor survival and the CCS gene is an independent predictor of outcome in patients with luminal B breast cancer by univariate and multivariate Cox regression. Conclusions Our findings emphasize the significant expression of CCS in luminal B breast cancer and its potential as an autonomous prognostic determinant for this specific molecular subtype. These findings suggest that CCS holds promise as a prospective marker for the treatment of luminal B breast cancer.
Collapse
Affiliation(s)
- Yanping Li
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Wenfei Du
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Rui Yang
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Xiaonan Wei
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Haibin Li
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Xiaoyuan Zhang
- Comprehensive Medical Training Center, Jining Medical University, Jining, China
| |
Collapse
|
2
|
Bhin J, Yemelyanenko J, Chao X, Klarenbeek S, Opdam M, Malka Y, Hoekman L, Kruger D, Bleijerveld O, Brambillasca CS, Sprengers J, Siteur B, Annunziato S, van Haren MJ, Martin NI, van de Ven M, Peters D, Agami R, Linn SC, Boven E, Altelaar M, Jonkers J, Zingg D, Wessels LF. MYC is a clinically significant driver of mTOR inhibitor resistance in breast cancer. J Exp Med 2023; 220:e20211743. [PMID: 37642941 PMCID: PMC10465700 DOI: 10.1084/jem.20211743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 05/18/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023] Open
Abstract
Targeting the PI3K-AKT-mTOR pathway is a promising therapeutic strategy for breast cancer treatment. However, low response rates and development of resistance to PI3K-AKT-mTOR inhibitors remain major clinical challenges. Here, we show that MYC activation drives resistance to mTOR inhibitors (mTORi) in breast cancer. Multiomic profiling of mouse invasive lobular carcinoma (ILC) tumors revealed recurrent Myc amplifications in tumors that acquired resistance to the mTORi AZD8055. MYC activation was associated with biological processes linked to mTORi response and counteracted mTORi-induced translation inhibition by promoting translation of ribosomal proteins. In vitro and in vivo induction of MYC conferred mTORi resistance in mouse and human breast cancer models. Conversely, AZD8055-resistant ILC cells depended on MYC, as demonstrated by the synergistic effects of mTORi and MYCi combination treatment. Notably, MYC status was significantly associated with poor response to everolimus therapy in metastatic breast cancer patients. Thus, MYC is a clinically relevant driver of mTORi resistance that may stratify breast cancer patients for mTOR-targeted therapies.
Collapse
Affiliation(s)
- Jinhyuk Bhin
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, Netherlands
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
- Department of Biomedical System Informatics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Julia Yemelyanenko
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Xue Chao
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Sjoerd Klarenbeek
- Experimental Animal Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Mark Opdam
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Yuval Malka
- Oncode Institute, Utrecht, Netherlands
- Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Liesbeth Hoekman
- Proteomics Facility, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Dinja Kruger
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Medical Oncology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam/Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Onno Bleijerveld
- Proteomics Facility, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Chiara S. Brambillasca
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Justin Sprengers
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Bjørn Siteur
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Stefano Annunziato
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Matthijs J. van Haren
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Nathaniel I. Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Dennis Peters
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Reuven Agami
- Oncode Institute, Utrecht, Netherlands
- Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Sabine C. Linn
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Epie Boven
- Department of Medical Oncology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam/Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Maarten Altelaar
- Proteomics Facility, Netherlands Cancer Institute, Amsterdam, Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Daniel Zingg
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Lodewyk F.A. Wessels
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
3
|
Yuan J, Yang L, Li Z, Zhang H, Wang Q, Huang J, Wang B, Mohan CD, Sethi G, Wang G. The role of the tumor microenvironment in endocrine therapy resistance in hormone receptor-positive breast cancer. Front Endocrinol (Lausanne) 2023; 14:1261283. [PMID: 37900137 PMCID: PMC10611521 DOI: 10.3389/fendo.2023.1261283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/29/2023] [Indexed: 10/31/2023] Open
Abstract
Endocrine therapy is the prominent strategy for the treatment of hormone-positive breast cancers. The emergence of resistance to endocrine therapy is a major health concern among hormone-positive breast cancer patients. Resistance to endocrine therapy demands the design of newer therapeutic strategies. The understanding of underlying molecular mechanisms of endocrine resistance, components of the tumor microenvironment (TME), and interaction of resistant breast cancer cells with the cellular/acellular components of the intratumoral environment are essential to formulate new therapeutic strategies for the treatment of endocrine therapy-resistant breast cancers. In the first half of the article, we have discussed the general mechanisms (including mutations in estrogen receptor gene, reregulated activation of signaling pathways, epigenetic changes, and cell cycle alteration) responsible for endocrine therapy resistance in hormone-positive breast cancers. In the latter half, we have emphasized the precise role of cellular (cancer-associated fibroblasts, immune cells, and cancer stem cells) and acellular components (collagen, fibronectin, and laminin) of TME in the development of endocrine resistance in hormone-positive breast cancers. In sum, the article provides an overview of the relationship between endocrine resistance and TME in hormone-positive breast cancers.
Collapse
Affiliation(s)
- Jie Yuan
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Li Yang
- Department of Clinical Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Zhi Li
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Hua Zhang
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Qun Wang
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Jun Huang
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Bei Wang
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Chakrabhavi Dhananjaya Mohan
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore Karnataka, India
- FEST Division, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Geng Wang
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
| |
Collapse
|
4
|
Fukuoka S, Koga Y, Yamauchi M, Koganemaru S, Yasunaga M, Shitara K, Doi T, Yoshino T, Kuronita T, Elenbaas B, Wahra P, Zhang H, Crowley L, Jenkins MH, Clark A, Kojima T. p70S6K/Akt dual inhibitor DIACC3010 is efficacious in preclinical models of gastric cancer alone and in combination with trastuzumab. Sci Rep 2023; 13:16017. [PMID: 37749105 PMCID: PMC10520030 DOI: 10.1038/s41598-023-40612-9] [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: 01/24/2023] [Accepted: 08/14/2023] [Indexed: 09/27/2023] Open
Abstract
The PI3K-Akt-mTOR (PAM) pathway is implicated in tumor progression in many tumor types, including metastatic gastric cancer (GC). The initial promise of PAM inhibitors has been unrealized in the clinic, presumably due, in part, to the up-regulation of Akt signaling that occurs when the pathway is inhibited. Here we present that DIACC3010 (formerly M2698), an inhibitor of two nodes in the PAM pathway, p70S6K and Akt 1/3, blocks the pathway in in vitro and in vivo preclinical models of GC while providing a mechanism that inhibits signaling from subsequent Akt up-regulation. Utilizing GC cell lines and xenograft models, we identified potential markers of DIACC3010-sensitivity in Her2-negative tumors, i.e., PIK3CA mutations, low basal pERK, and a group of differentially expressed genes (DEGs). The combination of DIACC3010 and trastuzumab was evaluated in Her2-positive cell lines and models. Potential biomarkers for the synergistic efficacy of the combination of DIACC3010 + trastuzumab also included DEGs as well as a lack of up-regulation of pERK. Of 27 GC patient-derived xenograft (PDX) models tested in BALB/c nu/nu mice, 59% were sensitive to DIACC3010 + trastuzumab. Of the 21 HER2-negative PDX models, DIACC3010 significantly inhibited the growth of 38%. Altogether, these results provide a path forward to validate the potential biomarkers of DIACC3010 sensitivity in GC and support clinical evaluation of DIACC3010 monotherapy and combination with trastuzumab in patients with HER2- negative and positive advanced GCs, respectively.
Collapse
Affiliation(s)
- Shota Fukuoka
- Division of Experimental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Yoshikatsu Koga
- Division of Developmental Therapeutics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center Hospital, Kashiwa, Japan
| | - Mayumi Yamauchi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Shigehiro Koganemaru
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masahiro Yasunaga
- Division of Developmental Therapeutics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center Hospital, Kashiwa, Japan
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Toshihiko Doi
- Division of Experimental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Toshio Kuronita
- Merck Biopharma Co., Ltd. (an affiliate of Merck KGaA), Tokyo, Japan
| | - Brian Elenbaas
- EMD Serono Research & Development Institute, Inc. (an affiliate of Merck KGaA), Billerica, MA, USA
| | - Pamela Wahra
- EMD Serono Research & Development Institute, Inc. (an affiliate of Merck KGaA), Billerica, MA, USA
| | - Hong Zhang
- EMD Serono Research & Development Institute, Inc. (an affiliate of Merck KGaA), Billerica, MA, USA
| | - Lindsey Crowley
- EMD Serono Research & Development Institute, Inc. (an affiliate of Merck KGaA), Billerica, MA, USA
| | - Molly H Jenkins
- EMD Serono Research & Development Institute, Inc. (an affiliate of Merck KGaA), Billerica, MA, USA
| | - Anderson Clark
- EMD Serono Research & Development Institute, Inc. (an affiliate of Merck KGaA), Billerica, MA, USA
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| |
Collapse
|
5
|
Aktas B, Fehm TN, Welslau M, Müller V, Lüftner D, Schütz F, Fasching PA, Janni W, Thomssen C, Witzel I, Belleville E, Untch M, Thill M, Tesch H, Ditsch N, Lux MP, Banys-Paluchowski M, Kolberg-Liedtke C, Hartkopf AD, Wöckel A, Kolberg HC, Stickeler E, Harbeck N, Schneeweiss A. Update Breast Cancer 2022 Part 4 - Advanced-Stage Breast Cancer. Geburtshilfe Frauenheilkd 2022; 82:922-931. [PMID: 36110891 PMCID: PMC9470282 DOI: 10.1055/a-1912-7362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 07/31/2022] [Indexed: 12/05/2022] Open
Abstract
For the treatment of patients with advanced HER2-negative hormone receptor-positive breast cancer, several substances have been introduced into practice in recent years. In addition, other drugs are under development. A number of studies have been published over the past year which have shown either an advantage for progression-free survival or for overall survival. This review summarizes the latest results, which have been published at current congresses or in specialist journals, and classifies them in the clinical treatment context. In particular, the importance of therapy with CDK4/6 inhibitors - trastuzumab deruxtecan, sacituzumab govitecan and capivasertib - is discussed. For trastuzumab deruxtecan, an overall survival benefit in HER2-negative breast cancer with low HER2 expression (HER2-low expression) was reported in the Destiny-Breast-04 study. Similarly, there was an overall survival benefit in the FAKTION study with capivasertib. The lack of overall survival benefit for palbociclib in the first line of therapy raises the question of clinical classification.
Collapse
Affiliation(s)
- Bahriye Aktas
- Department of Gynecology, University of Leipzig Medical Center, Leipzig, Germany
| | - Tanja N. Fehm
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Volkmar Müller
- Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
| | - Diana Lüftner
- Immanuel Hospital Märkische Schweiz & Medical University of Brandenburg Theodor-Fontane, Brandenburg, Buckow, Germany
| | - Florian Schütz
- Gynäkologie und Geburtshilfe, Diakonissen-Stiftungs-Krankenhaus Speyer, Speyer, Germany
| | - Peter A. Fasching
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen,
Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Christoph Thomssen
- Department of Gynaecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Isabell Witzel
- Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
| | | | - Michael Untch
- Clinic for Gynecology and Obstetrics, Breast Cancer Center, Gynecologic Oncology Center, Helios Klinikum Berlin Buch, Berlin, Germany
| | - Marc Thill
- Agaplesion Markus Krankenhaus, Department of Gynecology and Gynecological Oncology, Frankfurt am Main, Germany
| | - Hans Tesch
- Oncology Practice at Bethanien Hospital, Frankfurt am Main, Germany
| | - Nina Ditsch
- Department of Gynecology and Obstetrics, University Hospital Augsburg, Augsburg, Germany
| | - Michael P. Lux
- Klinik für Gynäkologie und Geburtshilfe, Frauenklinik St. Louise, Paderborn, St. Josefs-Krankenhaus, Salzkotten, St. Vincenz Krankenhaus GmbH, Germany
| | - Maggie Banys-Paluchowski
- Department of Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | | | - Andreas D. Hartkopf
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Achim Wöckel
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | | | - Elmar Stickeler
- Department of Gynecology and Obstetrics, RWTH University Hospital Aachen, Aachen, Germany
| | - Nadia Harbeck
- Breast Center, Department of Gynecology and Obstetrics and CCC Munich LMU, LMU University Hospital, Munich, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases (NCT), Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
6
|
Cuellar-Vite L, Weber-Bonk KL, Abdul-Karim FW, Booth CN, Keri RA. Focal Adhesion Kinase Provides a Collateral Vulnerability That Can Be Leveraged to Improve mTORC1 Inhibitor Efficacy. Cancers (Basel) 2022; 14:3374. [PMID: 35884439 PMCID: PMC9323520 DOI: 10.3390/cancers14143374] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 11/16/2022] Open
Abstract
The PI3K/AKT/mTORC1 pathway is a major therapeutic target for many cancers, particularly breast cancer. Everolimus is an mTORC1 inhibitor used in metastatic estrogen receptor-positive (ER+) and epidermal growth factor receptor 2-negative (HER2-) breast cancer. However, mTORC1 inhibitors have limited efficacy in other breast cancer subtypes. We sought to discover collateral sensitivities to mTORC1 inhibition that could be exploited to improve therapeutic response. Using a mouse model of breast cancer that is intrinsically resistant to mTORC1 inhibition, we found that rapamycin alters the expression of numerous extracellular matrix genes, suggesting a potential role for integrins/FAK in controlling mTORC1-inhibitor efficacy. FAK activation was also inversely correlated with rapamycin response in breast cancer cell lines. Supporting its potential utility in patients, FAK activation was observed in >50% of human breast cancers. While blocking FAK in mouse models of breast cancer that are highly responsive to rapamycin had no impact on tumor growth, FAK inhibition sensitized rapamycin-resistant tumors to mTORC1 inhibition. These data reveal an innate dependency on FAK when mTORC1 signaling is lost in tumors that are resistant to mTORC1 inhibitors. They also suggest a precision medicine approach to improving mTORC1 inhibitor efficacy in resistant cancers by suppressing FAK signaling.
Collapse
Affiliation(s)
- Leslie Cuellar-Vite
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA;
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Kristen L. Weber-Bonk
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Fadi W. Abdul-Karim
- Anatomic Pathology, Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (F.W.A.-K.); (C.N.B.)
| | - Christine N. Booth
- Anatomic Pathology, Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (F.W.A.-K.); (C.N.B.)
| | - Ruth A. Keri
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
- Department of General Medical Sciences-Oncology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
7
|
Voutsadakis IA. Biomarkers of everolimus efficacy in breast cancer therapy. J Oncol Pharm Pract 2022; 28:945-959. [PMID: 35018844 DOI: 10.1177/10781552211073673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Everolimus is an inhibitor of serine/ threonine kinase mTOR. The drug is approved for the treatment of metastatic ER positive, HER2 negative breast cancers and benefits a subset of patients with these breast cancers in combination with hormonal therapies. Despite extensive efforts, no additional predictive biomarkers to guide therapeutic decisions for everolimus have been introduced in clinical practice. DATA SOURCES This paper discusses predictive biomarkers for everolimus efficacy in breast cancer. A search of the medline and web of science databases was performed using the words "everolimus" and "biomarkers". References of retrieved articles were manually scanned for additional relevant articles. DATA SUMMARY Everolimus benefits a subset of patients with metastatic ER positive, HER2 negative breast cancers in combination with hormonal therapies. Despite extensive efforts no additional predictive biomarkers to guide therapeutic decisions for everolimus therapy have been confirmed for use in clinical practice. However, promising biomarker leads for everolimus efficacy in breast cancer have been suggested and include expression of proteins in the mTOR pathway in ER positive, HER2 negative breast cancers. In HER2 positive cancers PIK3CA mutations, and PTEN expression loss are prognostic. Other clinical predictive biomarkers with more limited data include characteristics derived from whole genome sequencing, subsets of circulating leukocytes and changes in Standardized Uptake Values (SUV) of Positron Emission Tomography (PET) scans. CONCLUSIONS Putative predictive biomarkers for everolimus efficacy in breast cancer patients, both genomic and clinical, deserve further study and could lead to a better selection of responsive patients.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Algoma District Cancer Program, 10066Sault Area Hospital, Sault Ste. Marie, Ontario, Canada, and Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| |
Collapse
|
8
|
Bilici A, Uysal M, Menekse S, Akin S, Yildiz F, Turan M, Sezgin Goksu S, Beypinar I, Sakalar T, Değirmenci M, Erdem D, Basaran G, Olmez OF, Avci N, Tural D, Sakin A, Turker S, Demir A, Temiz S, Kaplan MA, Dogan M, Tanriverdi O, Bilgetekin I, Cinkir HY, Acikgoz O, Paydas S, Uslu R, Turhal S. Real-Life Analysis of Efficacy and Safety of Everolimus Plus Exemestane in Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor-2-Negative Metastatic Breast Cancer Patients: A Turkish Oncology Group (TOG) Study. Cancer Invest 2021; 40:199-209. [PMID: 34894960 DOI: 10.1080/07357907.2021.2017952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE This study evaluated the efficacy and safety of everolimus (EVE) plus exemestane (EXE) in hormone-receptor positive (HR+), human epidermal growth factor receptor-2-negative (HER2-) metastatic breast cancer (MBC) patients in real-life settings. METHODS Overall, 204 HR+, HER2- MBC patients treated with EVE + EXE after progressing following prior endocrine treatment were included. Overall survival (OS) and progression-free survival (PFS) and safety data were analyzed. RESULTS The objective response rate, median PFS, and median OS were 33.4%, 8.9 months, and 23.4 months, respectively. Multivariate analysis revealed that negative progesterone receptor status was a significant determinant of poor treatment response (p = 0.035) and PFS (p = 0.024). The presence of bone-only metastasis was associated with better treatment response (p = 0.002), PFS (p < 0.001), and OS (p = 0.001). CONCLUSION We confirmed the favorable efficacy and safety profile of EVE + EXE for HR+, HER - MBC patients.
Collapse
Affiliation(s)
- Ahmet Bilici
- Department of Medical Oncology, Medical Faculty, Medipol University, Bagcilar, Turkey
| | - Mukremin Uysal
- Department of Medical Oncology, Medical Faculty, Afyon Kocatepe University, Afyon, Turkey
| | - Serkan Menekse
- Department of Medical Oncology, Manisa State Hospital, Manisa, Turkey
| | - Semih Akin
- Department of Medical Oncology, Medical Faculty, Ege University, Izmir, Turkey
| | - Fatih Yildiz
- Department of Medical Oncology, Dr Abdurrahman Yurtarslan Oncology Education and Research Hospital, Ankara, Turkey
| | - Merve Turan
- Department of Medical Oncology, Medical Faculty, Adnan Menderes University, Aydin, Turkey
| | - Sema Sezgin Goksu
- Department of Medical Oncology, Medical Faculty, Akdeniz University, Antalya, Turkey
| | - Ismail Beypinar
- Department of Medical Oncology, Medical Faculty, Afyon Kocatepe University, Afyon, Turkey
| | - Teoman Sakalar
- Department of Medical Oncology, Medical Faculty, Erciyes University, Kayseri, Turkey
| | - Mustafa Değirmenci
- Department of Medical Oncology, Izmir Tepecik Education and Research Hospital, Izmir, Turkey
| | - Dilek Erdem
- Department of Medical Oncology, Medical Faculty, Bahcesehir University, Samsun, Turkey
| | - Gul Basaran
- Department of Medical Oncology, Medical Faculty, Acibadem University, Istanbul, Turkey
| | - Omer Fatih Olmez
- Department of Medical Oncology, Medical Faculty, Medipol University, Bagcilar, Turkey
| | - Nilufer Avci
- Department of Medical Oncology, Medicana Bursa Hospital, Bursa, Turkey
| | - Deniz Tural
- Department of Medical Oncology, Bakirkoy Dr Sadi Konuk Education and Research Hospital, Istanbul, Turkey
| | - Abdullah Sakin
- Department of Medical Oncology, Medical Faculty, Yuzuncuyil University, Van, Turkey
| | - Sema Turker
- Department of Medical Oncology, Diskapi Yildirim Beyazit Education and Research Hospital, Ankara, Turkey
| | - Atakan Demir
- Department of Medical Oncology, Medical Faculty, Acibadem University, Istanbul, Turkey
| | - Suleyman Temiz
- Department of Medical Oncology, Kocaeli Acıbadem Hospital, Kocaeli, Turkey
| | - Muhammed Ali Kaplan
- Department of Medical Oncology, Medical Faculty, Dicle University, Diyarbakir, Turkey
| | - Mutlu Dogan
- Department of Medical Oncology, Ankara Numune Education and Research Hospital, Ankara, Turkey
| | - Ozgur Tanriverdi
- Department of Medical Oncology, Medical Faculty, Mugla Sitki Kocman University, Mugla, Turkey
| | - Irem Bilgetekin
- Department of Medical Oncology, Medical Faculty, Gazi University, Ankara, Turkey
| | - Havva Yesil Cinkir
- Department of Medical Oncology, Medical Faculty, Gaziantep University, Gaziantep, Turkey
| | - Ozgur Acikgoz
- Department of Medical Oncology, Medical Faculty, Medipol University, Bagcilar, Turkey
| | - Semra Paydas
- Department of Medical Oncology, Medical Faculty, Cukurova University, Adana, Turkey
| | - Ruchan Uslu
- Department of Medical Oncology, Medical Faculty, Ege University, Izmir, Turkey
| | - Serdar Turhal
- Anadolu Medical Center, Department of Medical Oncology, Istanbul, Turkey
| |
Collapse
|
9
|
Schettini F, Sobhani N, Ianza A, Triulzi T, Molteni A, Lazzari MC, Strina C, Milani M, Corona SP, Sirico M, Bernocchi O, Giudici F, Cappelletti MR, Ciruelos E, Jerusalem G, Loi S, Fox SB, Generali D. Immune system and angiogenesis-related potential surrogate biomarkers of response to everolimus-based treatment in hormone receptor-positive breast cancer: an exploratory study. Breast Cancer Res Treat 2020; 184:421-431. [PMID: 32770287 PMCID: PMC7599144 DOI: 10.1007/s10549-020-05856-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/31/2020] [Indexed: 01/08/2023]
Abstract
Purpose mTOR inhibitor everolimus is used for hormone receptor-positive (HR+)/HER2-negative metastatic breast cancer (mBC). No reliable predictive biomarker of response is available. Following evidences from other solid tumors, we aimed to assess the association between treatment-associated immune system features and everolimus activity. Methods We retrospectively explored a correlation with the therapeutic activity of everolimus and tumor-associated immune pathways with ingenuity pathway analysis (IPA), neutrophil-to-lymphocyte ratio (NLR), circulating lymphocytes, and endothelial cells (CECs) in 3 different HR+ mBC studies, including the BALLET phase IIIb study. Results The circulating levels of CD3+/CD8+, CD3+/CD4+, and overall T lymphocytes were higher in responders versus non-responders at baseline (p = 0.017, p < 0.001, p = 0.034) and after treatment (p = 0.01, p = 0.003, p = 0.023). Reduced CECs, a tumor neoangiogenesis marker, were observed in responders after treatment (p < 0.001). Patients with low NLR (≤ 4.4) showed a better progression-free survival compared to patients with high NLR (> 4.4) (p = 0.01). IPA showed that the majority of immunity-related genes were found upregulated in responders compared to non-responders before treatment, but not after. Conclusions Lymphocytes subpopulations, CECs and NLR could be interesting biomarkers predictive of response to everolimus-based regimens, potentially useful in daily clinical practice to select/monitor everolimus-based treatment in mBC. Further studies to confirm such hypotheses are warranted.
Collapse
Affiliation(s)
- Francesco Schettini
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy.,Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Navid Sobhani
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Anna Ianza
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alfredo Molteni
- UO Ematologia e CTMO, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | | | - Carla Strina
- UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Manuela Milani
- UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Silvia Paola Corona
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Marianna Sirico
- UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Ottavia Bernocchi
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy.,UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Fabiola Giudici
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Maria Rosaria Cappelletti
- UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Eva Ciruelos
- SOLTI Breast Cancer Research Group, Barcelona, Spain.,Department of Medical Oncology, Breast Cancer Unit, University Hospital, 12 de Octubre, Avda de Córdoba s/n, Madrid, Spain
| | - Guy Jerusalem
- Department of Medical Oncology, Centre Hospitalier Universitaire de Liège and Liège University, Avenue de L'Hòpital 1, 4000, Liège, Belgium
| | - Sherine Loi
- Peter MacCallum Cancer Center, 305 Grattan Street, Melbourne, VIC, Australia.,University of Melbourne, Melbourne, VIC, Australia
| | - Stephen B Fox
- Peter MacCallum Cancer Center, 305 Grattan Street, Melbourne, VIC, Australia.,University of Melbourne, Melbourne, VIC, Australia
| | - Daniele Generali
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy. .,UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy.
| |
Collapse
|
10
|
PI3K pathway protein analyses in metastatic breast cancer patients receiving standard everolimus and exemestane. J Cancer Res Clin Oncol 2020; 146:3013-3023. [PMID: 32566979 PMCID: PMC7519923 DOI: 10.1007/s00432-020-03291-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/12/2020] [Indexed: 11/26/2022]
Abstract
Purpose Everolimus plus exemestane (EVE/EXE) is a registered treatment option for ER-positive, HER2-negative (ER +/HER2-) metastatic breast cancer (MBC), but resistance mechanisms limit efficacy. We aimed to find markers that might help select patients with a higher chance on benefit from EVE/EXE. Methods Immunohistochemistry (IHC) of PTEN, p-AKT(Thr308), p-AKT(Ser473), p-4EBP1, p-p70S6K, p-S6RP(Ser240/244), p-ERK1/2 and p-S6RP (Ser235/236) was performed on primary tumour tissue and on biopsies immediately taken from ER +/HER2- MBC patients before the start of standard EVE/EXE (Eudract 2013-004120-11). Unsupervised hierarchical clustering was executed to create heatmaps to distinguish subgroups of preferentially activated and less-activated PI3K/MAPK proteins. Uni- and multivariate Cox models were used for associations with PFS. Results Primary tumour tissue from 145 patients was retrieved. Median PFS was 5.4 months. Patients without (neo)adjuvant therapy (p = 0.03) or bone only disease (p = 0.04) had longer PFS on EVE/EXE. In primary tumours, neither single proteins nor PI3K/MAPK-associated heatmap subgroups were significantly associated with PFS. In 21 patients a non-osseous biopsy obtained before dosing was useful for continuous scoring, which demonstrated upregulation of several proteins as compared to readings in corresponding primary tumour tissues. These comparisons revealed that increased expression of p-4EBP1 was significantly associated with worse PFS (multivariate HR 3.69, p = 0.05). Conclusions IHC of single proteins or heatmap subgroups of the differentially activated PI3K/MAPK pathways was not able to discriminate patients on EVE/EXE with poor or better PFS. Upregulation of p-4EBP1 in pre-treatment biopsies as compared to levels in primary tumours pointed towards shorter PFS. Electronic supplementary material The online version of this article (10.1007/s00432-020-03291-x) contains supplementary material, which is available to authorized users.
Collapse
|
11
|
Safety and efficacy of sirolimus combined with endocrine therapy in patients with advanced hormone receptor-positive breast cancer and the exploration of biomarkers. Breast 2020; 52:17-22. [PMID: 32335491 PMCID: PMC7375615 DOI: 10.1016/j.breast.2020.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/02/2020] [Accepted: 04/09/2020] [Indexed: 12/04/2022] Open
Abstract
Background We performed a retrospective study on the efficacy and safety of sirolimus (an mTOR inhibitor) in hormone receptor (HR)-positive advanced breast cancer and searched for biomarkers to predict its efficacy. Methods All patients with HR-positive metastatic breast cancer treated with sirolimus plus endocrine therapy between December 2017 and July 2018 at the Cancer Hospital, Chinese Academy of Medical Sciences were consecutively and retrospectively reviewed. Mutations in circulating tumour DNA (ctDNA) were assayed for 1021 tumour-related genes via gene panel target capture-based next-generation sequencing. Results Thirty-six patients with metastatic breast cancer treated with sirolimus plus endocrine therapy were included. The progression-free survival (PFS) rates between the sirolimus group and everolimus group were similar, and the median PFS was 4.9 months and 5.5 months, respectively (hazard ratio 1.56, 95% CI 0.86–2.81, P = 0.142). The objective response rate in the 36 patients was 19.4%, and the clinical benefit rate was 41.7%. Lipid metabolism disorder was the most common adverse event (69.4%), and 13.9% of patients had stomatitis. Most (94.4%) adverse events were grade 1–2. Twenty patients (55.6%) underwent ctDNA analysis before receiving sirolimus therapy. For patients who received less than 3 lines of chemotherapy, those with PI3K/Akt/mTOR pathway alterations had a better response to sirolimus than those without alterations, with a median PFS of 7.0 months vs 4.3 months (hazard ratio = 0.01, 95% CI 0.00–0.34, P = 0.010). Conclusions Sirolimus is a potentially effective treatment option for patients with HR-positive advanced breast cancer. Sirolimus is a potentially effective treatment option for patients with hormone receptor-positive advanced breast cancer. A previous chemotherapy line was a poor predictor of sirolimus efficacy. PI3K/Akt/mTOR pathway alterations may affect the efficacy of sirolimus.
Collapse
|
12
|
Challenges and future of precision medicine strategies for breast cancer based on a database on drug reactions. Biosci Rep 2019; 39:BSR20190230. [PMID: 31387972 PMCID: PMC6732363 DOI: 10.1042/bsr20190230] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 06/02/2019] [Accepted: 07/15/2019] [Indexed: 01/12/2023] Open
Abstract
Breast cancer (BC) is a malignancy with the highest incidence in women. Great progress has been made in research related to traditional precision medicine for BC. However, many reports have suggested that patients with BC have not benefited a lot from such progress. Thus, we analyze traditional precision medicine strategies for BC, sum up their limitations and challenges, and preliminarily propose future orientations of precision medicine strategies based on a database on drug reaction of patients with BC. According to related research, traditional precision medicine strategies for BC, which are based on molecular subtypes, perform pertinent treatments, new drug research and development according to molecular typing results. Nevertheless, these strategies still have some deficiencies. First, there are very few patients with each molecular subtype, the match ratio of drugs is low. Second, these strategies can not solve the problem of poor drug sensitivity resulting from heterogeneity. The main strategy we put forward in the present paper is based on patients’ varying drug reactions. Focusing on treating existing patients and maximizing the utilization of existing drugs, it is expected to not have deficiencies of traditional precision medicine for BC, including low match rate and poor therapeutic efficacy arising from tumor heterogeneity of BC.
Collapse
|
13
|
Chen Z, Zheng Y, Cao W, Zhang Y, Zhao Z, Wang G, Zhao J, Cai S, Shao X, Huang J, Ye W, Huang Y, Li W, Huang X, Wu H, Wang X, Yin Y. Everolimus-containing therapy vs conventional therapy in the treatment of refractory breast cancer patients with PI3K/AKT/mTOR mutations: A retrospective study. Cancer Med 2019; 8:5544-5553. [PMID: 31385461 PMCID: PMC6745827 DOI: 10.1002/cam4.2460] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/09/2019] [Accepted: 07/17/2019] [Indexed: 01/10/2023] Open
Abstract
Background Previous case reports have shown the promising antitumor activity of everolimus in solid tumors containing molecular aberrations in PI3K/ATK/mTOR pathway, however, whether it is effective in patients with breast cancer remains unknown. Therefore, we conducted this retrospective cohort study to compare the efficacy of molecularly matched targeted therapy with everolimus to conventional therapy in refractory breast cancer patients harboring PI3K/ATK/mTOR pathway activating mutations. Methods Refractory metastatic breast cancer patients who have received molecular screening using next‐generation sequencing (NGS) between September 8, 2015 and October 30, 2017 in two sites were screened for this study. The primary outcome was progression‐free survival (PFS). The secondary outcomes were overall response rate (ORR), disease control rate (DCR), and safety profile. Results A total of 78 patients were screened for analysis, amongst all, 52 (66.7%) had at least one gene mutation in PI3K/AKT/mTOR pathway. The most common mutation fell in PIK3CA (76.9%, 40/52) with a mutational prevalence of 51.3%. Of the 32 patients who were eligible for efficacy analysis, patients in the everolimus group (n = 19) exhibited shorter PFS than those in the conventional group (n = 13) (median, 1.9 vs 6.1 months; HR, 3.6; 95% CI, 1.48‐8.81; P = .0005). ORR was 15.4% (2/13) in the everolimus group and 23.1% (3/13) in the conventional group (P = 1.000), and DCR was 30.8% (4/13) and 100% (13/13) for each group, respectively (P = .000). The incidence of grade 3‐5 adverse events was relatively higher in the conventional group (38.5%, 5/13) than that in the everolimus group (26.3%, 5/19). Conclusions Our findings suggested that everolimus might not be effective for cancer patients harboring mutations in PI3K/ATK/mTOR pathway and physicians should be cautious about its off‐label use in clinical practice.
Collapse
Affiliation(s)
- Zhanhong Chen
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yabing Zheng
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Wenming Cao
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yuzi Zhang
- The Medical Department, 3D Medicines Inc, Shanghai, China
| | - Zhengyi Zhao
- The Medical Department, 3D Medicines Inc, Shanghai, China
| | - Guoqiang Wang
- The Medical Department, 3D Medicines Inc, Shanghai, China
| | - Jing Zhao
- The Medical Department, 3D Medicines Inc, Shanghai, China
| | - Shangli Cai
- The Medical Department, 3D Medicines Inc, Shanghai, China
| | - Xiying Shao
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jian Huang
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Weiwu Ye
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yuan Huang
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang Huang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Wu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaojia Wang
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Hesari A, Azizian M, Darabi H, Nesaei A, Hosseini SA, Salarinia R, Motaghi AA, Ghasemi F. Expression of circulating miR-17, miR-25, and miR-133 in breast cancer patients. J Cell Biochem 2019; 120:7109-7114. [PMID: 30485486 DOI: 10.1002/jcb.27984] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 10/08/2018] [Indexed: 01/24/2023]
Abstract
One of the most lethal cancers among women is breast cancer. MicroRNAs (miRNAs) can be of great importance in the early detection of breast cancer. This study aimed to investigate some miRNAs in the serum of patients with breast cancer compared with the control group. Total RNA was extracted from the serum of patients with breast cancer and healthy volunteers. The expression levels of miRNAs and the genes were assessed using real-time reverse transcriptase-polymerase chain reaction with specific primers. Our data showed that miR-25 and miR-133 were downregulated, and miR-17 was upregulated in patients with breast cancer. Upregulation of miR-17 is related to the poor survival time and increased cell proliferation. The reduced expression of miR-133 and miR-25 is significantly associated with clinical stage, metastasis, and survival time of patients with breast cancer. Expressions of miRNAs miR-17, miR-25, and miR-133 are altered in patients with clinical stage, metastasis, poor survival time.
Collapse
Affiliation(s)
- AmirReza Hesari
- Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Mitra Azizian
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Hassan Darabi
- Department of Medical Genetics, Medical Genetics Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abolfazl Nesaei
- Department of Basic Sciences, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Seyede Atefe Hosseini
- Department of Medical Biotechnology and Molecular Sciences, School of Medicine, North Khorasan University of Sciences, Bojnurd, Iran
| | - Reza Salarinia
- Department of Medical Biotechnology and Molecular Sciences, School of Medicine, North Khorasan University of Sciences, Bojnurd, Iran
| | - Amir Ali Motaghi
- Student Research Committee, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Faezeh Ghasemi
- Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran.,Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
15
|
Bazzichetto C, Conciatori F, Pallocca M, Falcone I, Fanciulli M, Cognetti F, Milella M, Ciuffreda L. PTEN as a Prognostic/Predictive Biomarker in Cancer: An Unfulfilled Promise? Cancers (Basel) 2019; 11:cancers11040435. [PMID: 30925702 PMCID: PMC6520939 DOI: 10.3390/cancers11040435] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 12/22/2022] Open
Abstract
Identifying putative biomarkers of clinical outcomes in cancer is crucial for successful enrichment, and for the selection of patients who are the most likely to benefit from a specific therapeutic approach. Indeed, current research in personalized cancer therapy focuses on the possibility of identifying biomarkers that predict prognosis, sensitivity or resistance to therapies. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a tumor suppressor gene that regulates several crucial cell functions such as proliferation, survival, genomic stability and cell motility through both enzymatic and non-enzymatic activities and phosphatidylinositol 3-kinase (PI3K)-dependent and -independent mechanisms. Despite its undisputed role as a tumor suppressor, assessment of PTEN status in sporadic human tumors has yet to provide clinically robust prognostic, predictive or therapeutic information. This is possibly due to the exceptionally complex regulation of PTEN function, which involves genetic, transcriptional, post-transcriptional and post-translational events. This review shows a brief summary of the regulation and function of PTEN and discusses its controversial aspects as a prognostic/predictive biomarker.
Collapse
Affiliation(s)
- Chiara Bazzichetto
- Medical Oncology 1, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
- Department of Molecular Medicine, University of Rome, La Sapienza, Rome 00185, Italy.
| | - Fabiana Conciatori
- Medical Oncology 1, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
| | - Matteo Pallocca
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
| | - Italia Falcone
- Medical Oncology 1, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
| | - Maurizio Fanciulli
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
| | - Francesco Cognetti
- Medical Oncology 1, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
| | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona 37126, Italy.
| | - Ludovica Ciuffreda
- Medical Oncology 1, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Rome 00144, Italy.
| |
Collapse
|
16
|
Li S, Wang X, Li Y, Lai H, Liu Y, Jin L. Non-invasive analysis of tumor mutation profiles and druggable mutations by sequencing of cell free DNA of Chinese metastatic breast cancer patients. Thorac Cancer 2019. [PMID: 30793491 DOI: 10.1111/1759‐7714.13002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Metastatic breast cancer (MBC) remains an incurable disease worldwide. Tumor gene mutations have evolved and led to drug resistance in the treatment course of MBC. However, data on the mutation profiles and druggable genomic alterations of MBC remain limited, particularly among Chinese patients. Our study aimed to depict the mutation profiles and identify druggable mutations in circulating tumor DNA (ctDNA) in Chinese MBC patients. METHODS Targeted deep sequencing of a 1021-gene panel was performed on 17 blood samples and 5 available tissue samples from 17 Chinese MBC patients. RESULTS We identified 60 somatic mutations in 17 blood samples (sensitivity 100%). Somatic mutations were identified in the blood samples of all patients, and 41.18% (7/17) of patients harbored at least one druggable mutation. A high ctDNA level in plasma is associated with shorter progression-free survival. CONCLUSION Targeted deep sequencing of cell free DNA is a highly sensitive, noninvasive method to depict tumor mutation profiles, identify druggable mutations in MBC, and predict patient outcome. Our study shed light on the utility of ctDNA as noninvasive "liquid biopsy" in the management of MBC.
Collapse
Affiliation(s)
- Shunying Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiaobao Wang
- Department of Otorhinolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yuquan Li
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Hongna Lai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yujie Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Liang Jin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| |
Collapse
|
17
|
Li S, Wang X, Li Y, Lai H, Liu Y, Jin L. Non-invasive analysis of tumor mutation profiles and druggable mutations by sequencing of cell free DNA of Chinese metastatic breast cancer patients. Thorac Cancer 2019; 10:807-814. [PMID: 30793491 PMCID: PMC6449225 DOI: 10.1111/1759-7714.13002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/11/2019] [Accepted: 01/13/2019] [Indexed: 11/27/2022] Open
Abstract
Background Metastatic breast cancer (MBC) remains an incurable disease worldwide. Tumor gene mutations have evolved and led to drug resistance in the treatment course of MBC. However, data on the mutation profiles and druggable genomic alterations of MBC remain limited, particularly among Chinese patients. Our study aimed to depict the mutation profiles and identify druggable mutations in circulating tumor DNA (ctDNA) in Chinese MBC patients. Methods Targeted deep sequencing of a 1021‐gene panel was performed on 17 blood samples and 5 available tissue samples from 17 Chinese MBC patients. Results We identified 60 somatic mutations in 17 blood samples (sensitivity 100%). Somatic mutations were identified in the blood samples of all patients, and 41.18% (7/17) of patients harbored at least one druggable mutation. A high ctDNA level in plasma is associated with shorter progression‐free survival. Conclusion Targeted deep sequencing of cell free DNA is a highly sensitive, noninvasive method to depict tumor mutation profiles, identify druggable mutations in MBC, and predict patient outcome. Our study shed light on the utility of ctDNA as noninvasive “liquid biopsy” in the management of MBC.
Collapse
Affiliation(s)
- Shunying Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiaobao Wang
- Department of Otorhinolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yuquan Li
- Division of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Hongna Lai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yujie Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Liang Jin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| |
Collapse
|
18
|
Therapeutic Targeting of mTOR in T-Cell Acute Lymphoblastic Leukemia: An Update. Int J Mol Sci 2018; 19:ijms19071878. [PMID: 29949919 PMCID: PMC6073309 DOI: 10.3390/ijms19071878] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 06/22/2018] [Accepted: 06/24/2018] [Indexed: 12/14/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive blood malignancy that arises from the clonal expansion of transformed T-cell precursors. Although T-ALL prognosis has significantly improved due to the development of intensive chemotherapeutic protocols, primary drug-resistant and relapsed patients still display a dismal outcome. In addition, lifelong irreversible late effects from conventional therapy are a growing problem for leukemia survivors. Therefore, novel targeted therapies are required to improve the prognosis of high-risk patients. The mechanistic target of rapamycin (mTOR) is the kinase subunit of two structurally and functionally distinct multiprotein complexes, which are referred to as mTOR complex 1 (mTORC1) and mTORC2. These two complexes regulate a variety of physiological cellular processes including protein, lipid, and nucleotide synthesis, as well as autophagy in response to external cues. However, mTOR activity is frequently deregulated in cancer, where it plays a key oncogenetic role driving tumor cell proliferation, survival, metabolic transformation, and metastatic potential. Promising preclinical studies using mTOR inhibitors have demonstrated efficacy in many human cancer types, including T-ALL. Here, we highlight our current knowledge of mTOR signaling and inhibitors in T-ALL, with an emphasis on emerging evidence of the superior efficacy of combinations consisting of mTOR inhibitors and either traditional or targeted therapeutics.
Collapse
|
19
|
Shea MP, O'Leary KA, Wegner KA, Vezina CM, Schuler LA. High collagen density augments mTOR-dependent cancer stem cells in ERα+ mammary carcinomas, and increases mTOR-independent lung metastases. Cancer Lett 2018; 433:1-9. [PMID: 29935374 DOI: 10.1016/j.canlet.2018.06.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/14/2018] [Accepted: 06/15/2018] [Indexed: 12/18/2022]
Abstract
Metastatic estrogen receptor alpha positive (ERα+) cancers account for most breast cancer mortality. Cancer stem cells (CSCs) and dense/stiff extracellular matrices are implicated in aggression and therapy resistance. We examined this interplay and response to mTOR inhibition using ERα+ adenocarcinomas from NRL-PRL females in combination with Col1a1tmJae/+ (mCol1a1) mice, which accumulate collagen-I around growing tumors. Orthotopic transplantation of tumor cells to mCol1a1 but not wildtype hosts resulted in striking desmoplasia. Mammary tumors in mCol1a1 recipients displayed higher CSC activity and enhanced AKT-mTOR and YAP activation, and these animals developed more and larger lung metastases. Treatment with the mTOR inhibitor, rapamycin, with or without the anti-estrogen, ICI182780, rapidly diminished mammary tumors, which rapidly reversed when treatment ceased. In contrast, lung metastases, which exhibited lower proliferation and pS6RP, indicating lower mTOR activity, were unresponsive, and mCol1a1 hosts continued to sustain greater metastatic burdens. These findings shed light on the influence of desmoplastic tumor microenvironments on the CSC niche and metastatic behavior in ERα+ breast cancer. The differential mTOR dependence of local mammary tumors and pulmonary lesions has implications for success of mTOR inhibitors in advanced ERα+ disease.
Collapse
Affiliation(s)
- Michael P Shea
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA; Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Kathleen A O'Leary
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Kyle A Wegner
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA; Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Chad M Vezina
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA; Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA; University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Linda A Schuler
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA; Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA; University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, WI, USA.
| |
Collapse
|
20
|
Petrossian K, Nguyen D, Lo C, Kanaya N, Somlo G, Cui YX, Huang CS, Chen S. Use of dual mTOR inhibitor MLN0128 against everolimus-resistant breast cancer. Breast Cancer Res Treat 2018; 170:499-506. [PMID: 29623577 DOI: 10.1007/s10549-018-4779-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 03/31/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE HR+/HER2- aromatase inhibitor-resistant metastatic breast cancer can be treated with everolimus and a second AI until the cancer recurs. Targeting these everolimus-resistant patients with the latest standard of care, CDK4/6 inhibitors, has not been clearly addressed. Understanding the signaling transduction pathways, which everolimus resistance activates, will elucidate the mechanisms and offer treatment strategies of everolimus resistance. METHODS To mimic the clinical setting, letrozole-resistant cells were used to generate an everolimus-resistant model (RAD-R). Reverse phase protein array (RPPA) was performed to reveal changes in the signaling transduction pathways, and expression levels of key proteins were analyzed. Inhibitors targeting the major signaling pathways, a CDK4/6 inhibitor palbociclib and a mTORC1/2 inhibitor (MLN0128), were evaluated to establish resistance mechanisms of RAD-R. RESULTS RPPA results from RAD-R indicated changes to significant regulatory pathways and upregulation of p-AKT expression level associating with everolimus resistance. MLN0128, that inhibits the AKT phosphorylation, effectively suppressed the proliferation of RAD-R cells while treatment with palbociclib had no effect. CONCLUSION Among the many signaling transduction pathways, which are altered post everolimus resistance, targeting dual mTORC1/2 is a possible option for patients who have recurrent disease from previous everolimus treatment.
Collapse
Affiliation(s)
- Karineh Petrossian
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 East Duarte Road, Kaplan CRB, Room 2002C, Duarte, CA, USA
| | - Duc Nguyen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 East Duarte Road, Kaplan CRB, Room 2002C, Duarte, CA, USA
| | - Chiao Lo
- Department of Breast Health, National Taiwan University Hospital, Taipei City, Taiwan
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 East Duarte Road, Kaplan CRB, Room 2002C, Duarte, CA, USA
| | - George Somlo
- Department of Medical Oncology and Therapeutics Research, City of Hope Medical Center, Duarte, CA, 91010, USA
| | - Yvonne Xiaoyong Cui
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 East Duarte Road, Kaplan CRB, Room 2002C, Duarte, CA, USA
| | - Chiun-Sheng Huang
- Department of Breast Health, National Taiwan University Hospital, Taipei City, Taiwan
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 East Duarte Road, Kaplan CRB, Room 2002C, Duarte, CA, USA.
| |
Collapse
|
21
|
Abstract
Mechanistic target of rapamycin (mTOR) is the kinase subunit of two structurally and functionally distinct large multiprotein complexes, referred to as mTOR complex 1 (mTORC1) and mTORC2. mTORC1 and mTORC2 play key physiological roles as they control anabolic and catabolic processes in response to external cues in a variety of tissues and organs. However, mTORC1 and mTORC2 activities are deregulated in widespread human diseases, including cancer. Cancer cells take advantage of mTOR oncogenic signaling to drive their proliferation, survival, metabolic transformation, and metastatic potential. Therefore, mTOR lends itself very well as a therapeutic target for innovative cancer treatment. mTOR was initially identified as the target of the antibiotic rapamycin that displayed remarkable antitumor activity in vitro Promising preclinical studies using rapamycin and its derivatives (rapalogs) demonstrated efficacy in many human cancer types, hence supporting the launch of numerous clinical trials aimed to evaluate the real effectiveness of mTOR-targeted therapies. However, rapamycin and rapalogs have shown very limited activity in most clinical contexts, also when combined with other drugs. Thus, novel classes of mTOR inhibitors with a stronger antineoplastic potency have been developed. Nevertheless, emerging clinical data suggest that also these novel mTOR-targeting drugs may have a weak antitumor activity. Here, we summarize the current status of available mTOR inhibitors and highlight the most relevant results from both preclinical and clinical studies that have provided valuable insights into both their efficacy and failure.
Collapse
|