1
|
Athwal H, Kochiyanil A, Bhat V, Allan AL, Parsyan A. Centrosomes and associated proteins in pathogenesis and treatment of breast cancer. Front Oncol 2024; 14:1370565. [PMID: 38606093 PMCID: PMC11007099 DOI: 10.3389/fonc.2024.1370565] [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: 01/14/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
Breast cancer is the most prevalent malignancy among women worldwide. Despite significant advances in treatment, it remains one of the leading causes of female mortality. The inability to effectively treat advanced and/or treatment-resistant breast cancer demonstrates the need to develop novel treatment strategies and targeted therapies. Centrosomes and their associated proteins have been shown to play key roles in the pathogenesis of breast cancer and thus represent promising targets for drug and biomarker development. Centrosomes are fundamental cellular structures in the mammalian cell that are responsible for error-free execution of cell division. Centrosome amplification and aberrant expression of its associated proteins such as Polo-like kinases (PLKs), Aurora kinases (AURKs) and Cyclin-dependent kinases (CDKs) have been observed in various cancers, including breast cancer. These aberrations in breast cancer are thought to cause improper chromosomal segregation during mitosis, leading to chromosomal instability and uncontrolled cell division, allowing cancer cells to acquire new genetic changes that result in evasion of cell death and the promotion of tumor formation. Various chemical compounds developed against PLKs and AURKs have shown meaningful antitumorigenic effects in breast cancer cells in vitro and in vivo. The mechanism of action of these inhibitors is likely related to exacerbation of numerical genomic instability, such as aneuploidy or polyploidy. Furthermore, growing evidence demonstrates enhanced antitumorigenic effects when inhibitors specific to centrosome-associated proteins are used in combination with either radiation or chemotherapy drugs in breast cancer. This review focuses on the current knowledge regarding the roles of centrosome and centrosome-associated proteins in breast cancer pathogenesis and their utility as novel targets for breast cancer treatment.
Collapse
Affiliation(s)
- Harjot Athwal
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Arpitha Kochiyanil
- Faculty of Science, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Vasudeva Bhat
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
| | - Alison L. Allan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Armen Parsyan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Division of General Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Surgery, St. Joseph’s Health Care London and London Health Sciences Centre, London, ON, Canada
| |
Collapse
|
2
|
Masci D, Naro C, Puxeddu M, Urbani A, Sette C, La Regina G, Silvestri R. Recent Advances in Drug Discovery for Triple-Negative Breast Cancer Treatment. Molecules 2023; 28:7513. [PMID: 38005235 PMCID: PMC10672974 DOI: 10.3390/molecules28227513] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most heterogeneous and aggressive breast cancer subtypes with a high risk of death on recurrence. To date, TNBC is very difficult to treat due to the lack of an effective targeted therapy. However, recent advances in the molecular characterization of TNBC are encouraging the development of novel drugs and therapeutic combinations for its therapeutic management. In the present review, we will provide an overview of the currently available standard therapies and new emerging therapeutic strategies against TNBC, highlighting the promises that newly developed small molecules, repositioned drugs, and combination therapies have of improving treatment efficacy against these tumors.
Collapse
Affiliation(s)
- Domiziana Masci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (D.M.); (A.U.)
| | - Chiara Naro
- Department of Neurosciences, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (C.N.); (C.S.)
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Michela Puxeddu
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (G.L.R.)
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (D.M.); (A.U.)
| | - Claudio Sette
- Department of Neurosciences, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (C.N.); (C.S.)
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giuseppe La Regina
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (G.L.R.)
| | - Romano Silvestri
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (G.L.R.)
| |
Collapse
|
3
|
Haddad TC, Suman VJ, D'Assoro AB, Carter JM, Giridhar KV, McMenomy BP, Santo K, Mayer EL, Karuturi MS, Morikawa A, Marcom PK, Isaacs CJ, Oh SY, Clark AS, Mayer IA, Keyomarsi K, Hobday TJ, Peethambaram PP, O'Sullivan CC, Leon-Ferre RA, Liu MC, Ingle JN, Goetz MP. Evaluation of Alisertib Alone or Combined With Fulvestrant in Patients With Endocrine-Resistant Advanced Breast Cancer: The Phase 2 TBCRC041 Randomized Clinical Trial. JAMA Oncol 2023; 9:815-824. [PMID: 36892847 PMCID: PMC9999287 DOI: 10.1001/jamaoncol.2022.7949] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/23/2022] [Indexed: 03/10/2023]
Abstract
Importance Aurora A kinase (AURKA) activation, related in part to AURKA amplification and variants, is associated with downregulation of estrogen receptor (ER) α expression, endocrine resistance, and implicated in cyclin-dependent kinase 4/6 inhibitor (CDK 4/6i) resistance. Alisertib, a selective AURKA inhibitor, upregulates ERα and restores endocrine sensitivity in preclinical metastatic breast cancer (MBC) models. The safety and preliminary efficacy of alisertib was demonstrated in early-phase trials; however, its activity in CDK 4/6i-resistant MBC is unknown. Objective To assess the effect of adding fulvestrant to alisertib on objective tumor response rates (ORRs) in endocrine-resistant MBC. Design, Setting, and Participants This phase 2 randomized clinical trial was conducted through the Translational Breast Cancer Research Consortium, which enrolled participants from July 2017 to November 2019. Postmenopausal women with endocrine-resistant, ERBB2 (formerly HER2)-negative MBC who were previously treated with fulvestrant were eligible. Stratification factors included prior treatment with CDK 4/6i, baseline metastatic tumor ERα level measurement (<10%, ≥10%), and primary or secondary endocrine resistance. Among 114 preregistered patients, 96 (84.2%) registered and 91 (79.8%) were evaluable for the primary end point. Data analysis began after January 10, 2022. Interventions Alisertib, 50 mg, oral, daily on days 1 to 3, 8 to 10, and 15 to 17 of a 28-day cycle (arm 1) or alisertib same dose/schedule with standard-dose fulvestrant (arm 2). Main Outcomes and Measures Improvement in ORR in arm 2 of at least 20% greater than arm 1 when the expected ORR for arm 1 was 20%. Results All 91 evaluable patients (mean [SD] age, 58.5 [11.3] years; 1 American Indian/Alaskan Native [1.1%], 2 Asian [2.2%], 6 Black/African American [6.6%], 5 Hispanic [5.5%], and 79 [86.8%] White individuals; arm 1, 46 [50.5%]; arm 2, 45 [49.5%]) had received prior treatment with CDK 4/6i. The ORR was 19.6%; (90% CI, 10.6%-31.7%) for arm 1 and 20.0% (90% CI, 10.9%-32.3%) for arm 2. In arm 1, the 24-week clinical benefit rate and median progression-free survival time were 41.3% (90% CI, 29.0%-54.5%) and 5.6 months (95% CI, 3.9-10.0), respectively, and in arm 2 they were 28.9% (90% CI, 18.0%-42.0%) and 5.4 months (95% CI, 3.9-7.8), respectively. The most common grade 3 or higher adverse events attributed to alisertib were neutropenia (41.8%) and anemia (13.2%). Reasons for discontinuing treatment were disease progression (arm 1, 38 [82.6%]; arm 2, 31 [68.9%]) and toxic effects or refusal (arm 1, 5 [10.9%]; arm 2, 12 [26.7%]). Conclusions and Relevance This randomized clinical trial found that adding fulvestrant to treatment with alisertib did not increase ORR or PFS; however, promising clinical activity was observed with alisertib monotherapy among patients with endocrine-resistant and CDK 4/6i-resistant MBC. The overall safety profile was tolerable. Trial Registration ClinicalTrials.gov Identifier: NCT02860000.
Collapse
Affiliation(s)
- Tufia C Haddad
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Vera J Suman
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | | | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Katelyn Santo
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Erica L Mayer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Meghan S Karuturi
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Aki Morikawa
- Department of Medicine, University of Michigan, Ann Arbor
| | - P Kelly Marcom
- Department of Medicine, Duke University Cancer Institute, Durham, North Carolina
| | | | - Sun Young Oh
- Department of Medical Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, New York
| | - Amy S Clark
- Department of Medicine, University of Pennsylvania, Philadelphia
| | - Ingrid A Mayer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Khandan Keyomarsi
- Department of Experimental Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | - Minetta C Liu
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
4
|
Investigational Drug Treatments for Triple-Negative Breast Cancer. J Pers Med 2021; 11:jpm11070652. [PMID: 34357119 PMCID: PMC8303312 DOI: 10.3390/jpm11070652] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/25/2021] [Accepted: 07/08/2021] [Indexed: 02/05/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer (BC) and accounts for 10–20% of cases. Due to the lack of expression of several receptors, hormone therapy is largely ineffective for treatment purposes. Nevertheless, TNBC often responds very well to chemotherapy, which constitutes the most often recommended treatment. New beneficial targeted therapies are important to be investigated in order to achieve enhanced outcomes in patients with TNBC. This review will focus on recent therapeutic innovations for TNBC, focusing on various inhibitors such as phosphoinositide 3-kinase (PI3K) pathway inhibitors, poly-ADP-ribosyl polymerase (PARP) inhibitors, aurora kinase inhibitors, histone deacetylase inhibitors (HDACIs), and immune checkpoint inhibitors.
Collapse
|
5
|
Serrano-Del Valle A, Reina-Ortiz C, Benedi A, Anel A, Naval J, Marzo I. Future prospects for mitosis-targeted antitumor therapies. Biochem Pharmacol 2021; 190:114655. [PMID: 34129859 DOI: 10.1016/j.bcp.2021.114655] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 11/17/2022]
Abstract
Dysregulation of cell cycle progression is a hallmark of cancer cells. In recent years, efforts have been devoted to the development of new therapies that target proteins involved in cell cycle regulation and mitosis. Novel targeted antimitotic drugs include inhibitors of aurora kinase family, polo-like kinase 1, Mps1, Eg5, CENP-5 and the APC/cyclosome complex. While certain new inhibitors reached the clinical trial stage, most were discontinued due to negative results. However, these therapies should not be readily dismissed. Based on recent advances concerning their mechanisms of action, new strategies could be devised to increase their efficacy and promote further clinical trials. Here we discuss three main lines of action to empower these therapeutic approaches: increasing cell death signals during mitotic arrest, targeting senescent cells and facilitating antitumor immune response through immunogenic cell death (ICD).
Collapse
Affiliation(s)
| | - Chantal Reina-Ortiz
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Andrea Benedi
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Alberto Anel
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Javier Naval
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Isabel Marzo
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain.
| |
Collapse
|
6
|
Byun WS, Bae ES, Cui J, Park HJ, Oh DC, Lee SK. Antitumor Activity of Pulvomycin via Targeting Activated-STAT3 Signaling in Docetaxel-Resistant Triple-Negative Breast Cancer Cells. Biomedicines 2021; 9:436. [PMID: 33920736 PMCID: PMC8074004 DOI: 10.3390/biomedicines9040436] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 01/14/2023] Open
Abstract
Although docetaxel-based regimens are common and effective for early-stage triple-negative breast cancer (TNBC) treatment, acquired drug resistance frequently occurs. Therefore, a novel therapeutic strategy for docetaxel-resistant TNBC is urgently required. Signal transducer and activator of transcription 3 (STAT3) plays a pivotal role in the tumorigenesis and metastasis of numerous cancers, and STAT3 signaling is aberrantly activated in TNBC cells. In this study, a docetaxel-resistant TNBC cell line (MDA-MB-231-DTR) was established, and mechanisms for the antitumor activity of pulvomycin, a novel STAT3 inhibitor isolated from marine-derived actinomycete, were investigated. Levels of activated STAT3 (p-STAT3 (Y705)) increased in docetaxel-resistant cells, and knockdown of STAT3 recovered the sensitivity to docetaxel in MDA-MB-231-DTR cells. Pulvomycin effectively inhibited the proliferation of both cell lines. In addition, pulvomycin suppressed the activation of STAT3 and subsequently induced G0/G1 cell cycle arrest and apoptosis. Pulvomycin also significantly inhibited the invasion and migration of MDA-MB-231-DTR cells through the modulation of epithelial-mesenchymal transition markers. In an MDA-MB-231-DTR-bearing xenograft mouse model, the combination of pulvomycin and docetaxel effectively inhibited tumor growth through STAT3 regulation. Thus, our findings demonstrate that the combination of docetaxel and STAT3 inhibitors is an effective strategy for overcoming docetaxel resistance in TNBC.
Collapse
Affiliation(s)
| | | | | | | | | | - Sang Kook Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Korea; (W.S.B.); (E.S.B.); (J.C.); (H.J.P.); (D.-C.O.)
| |
Collapse
|
7
|
O’Shaughnessy J, McIntyre K, Wilks S, Ma L, Block M, Andorsky D, Danso M, Locke T, Scales A, Wang Y. Efficacy and Safety of Weekly Paclitaxel With or Without Oral Alisertib in Patients With Metastatic Breast Cancer: A Randomized Clinical Trial. JAMA Netw Open 2021; 4:e214103. [PMID: 33877311 PMCID: PMC8058641 DOI: 10.1001/jamanetworkopen.2021.4103] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
IMPORTANCE Elevated expression of AURKA adversely affects prognosis in estrogen receptor (ER)-positive and ERBB2 (formerly HER2)-negative and triple-negative breast cancer and is associated with resistance to taxanes. OBJECTIVE To compare paclitaxel alone vs paclitaxel plus alisertib in patients with ER-positive and ERBB2-negative or triple-negative metastatic breast cancer (MBC). DESIGN, SETTING, AND PARTICIPANTS In this randomized clinical trial conducted with the US Oncology Network, participants were randomized to intravenous (IV) paclitaxel 90 mg/m2 on days 1, 8, and 15 on a 28-day cycle or IV paclitaxel 60 mg/m2 on days 1, 8, and 15 plus oral alisertib 40 mg twice daily on days 1 to 3, 8 to 10, and 15 to 17 on a 28-day cycle. Stratification was by prior neo or adjuvant taxane and by line of metastatic therapy. Eligible patients were those who had undergone endocrine therapy, 0 or 1 prior chemotherapy regimens for MBC, more than 12 months treatment-free interval from neo or adjuvant taxane therapy, and with measurable or evaluable lytic bone-disease. Data were analyzed from March 2019 through May 2019. MAIN OUTCOMES AND MEASURES The main outcome was progression-free survival (PFS) with secondary end points of overall survival (OS), overall response rate, clinical benefit rate, safety, and analysis of archival breast cancer tissues for molecular markers associated with benefit from alisertib. RESULTS A total of 174 patients were randomized, including with 86 randomized to paclitaxel and 88 patients randomized to paclitaxel plus alisertib, and 169 patients received study treatment. The final cohort included 139 patients with a median (interquartile range [IQR]) age of 62 (27-84) years with ER-positive and ERBB2-negative MBC, with 70 randomized to paclitaxel and 69 randomized to paclitaxel plus alisertib. The TNBC cohort closed with only 35 patients enrolled due to slow accrual and were not included in efficacy analyses. The median (IQR) follow-up was 22 (10.6-25.1) months, and median (IQR) PFS was 10.2 (3.8-15.7) months with paclitaxel plus alisertib vs 7.1 (3.8-10.6) months with paclitaxel alone (HR, 0.56; 95% CI, 0.37-0.84; P = .005). Median (IQR) OS was 26.3 (12.4-37.2) months for patients who received paclitaxel plus alisertib vs 25.1 (11.0-31.4) months for paclitaxel alone (HR, 0.89; 95% CI, 0.58-1.38; P = .61). Grade 3 or 4 adverse events occurred in 56 patients (84.8%) receiving paclitaxel plus alisertib vs 34 patients (48.6%) receiving paclitaxel alone. The main grade 3 or 4 adverse events with paclitaxel plus alisertib vs paclitaxel alone were neutropenia (50 patients [59.5%] vs 14 patients [16.4%]), anemia (8 patients [9.5%] vs 1 patient [1.2%]), diarrhea (9 patients [10.7%] vs 0 patients), and stomatitis or oral mucositis (13 patients [15.5%] vs 0 patients). One patient receiving paclitaxel plus alisertib died of sepsis. CONCLUSIONS AND RELEVANCE This randomized clinical trial found that the addition of oral alisertib to a reduced dose of weekly paclitaxel significantly improved PFS compared with paclitaxel alone, and toxic effects with paclitaxel plus alisertib were manageable with alisertib dose reduction. These data support further evaluation of alisertib in patients with ER-positive, ERBB2-negative MBC. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02187991.
Collapse
Affiliation(s)
- Joyce O’Shaughnessy
- Baylor University Medical Center, Dallas, Texas
- Texas Oncology, Dallas
- US Oncology, Houston, Texas
| | | | - Sharon Wilks
- Texas Oncology, Dallas
- US Oncology, Houston, Texas
| | - Ling Ma
- US Oncology, Houston, Texas
- Rocky Mountain Cancer Centers, Lakewood, Colorado
| | - Margaret Block
- US Oncology, Houston, Texas
- Nebraska Cancer Specialists, Omaha
| | - David Andorsky
- US Oncology, Houston, Texas
- Rocky Mountain Cancer Centers, Boulder, Colorado
| | - Michael Danso
- US Oncology, Houston, Texas
- Virginia Oncology Associates, Norfolk
| | | | | | | |
Collapse
|
8
|
Kalev P, Hyer ML, Gross S, Konteatis Z, Chen CC, Fletcher M, Lein M, Aguado-Fraile E, Frank V, Barnett A, Mandley E, Goldford J, Chen Y, Sellers K, Hayes S, Lizotte K, Quang P, Tuncay Y, Clasquin M, Peters R, Weier J, Simone E, Murtie J, Liu W, Nagaraja R, Dang L, Sui Z, Biller SA, Travins J, Marks KM, Marjon K. MAT2A Inhibition Blocks the Growth of MTAP-Deleted Cancer Cells by Reducing PRMT5-Dependent mRNA Splicing and Inducing DNA Damage. Cancer Cell 2021; 39:209-224.e11. [PMID: 33450196 DOI: 10.1016/j.ccell.2020.12.010] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 10/30/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
The methylthioadenosine phosphorylase (MTAP) gene is located adjacent to the cyclin-dependent kinase inhibitor 2A (CDKN2A) tumor-suppressor gene and is co-deleted with CDKN2A in approximately 15% of all cancers. This co-deletion leads to aggressive tumors with poor prognosis that lack effective, molecularly targeted therapies. The metabolic enzyme methionine adenosyltransferase 2α (MAT2A) was identified as a synthetic lethal target in MTAP-deleted cancers. We report the characterization of potent MAT2A inhibitors that substantially reduce levels of S-adenosylmethionine (SAM) and demonstrate antiproliferative activity in MTAP-deleted cancer cells and tumors. Using RNA sequencing and proteomics, we demonstrate that MAT2A inhibition is mechanistically linked to reduced protein arginine methyltransferase 5 (PRMT5) activity and splicing perturbations. We further show that DNA damage and mitotic defects ensue upon MAT2A inhibition in HCT116 MTAP-/- cells, providing a rationale for combining the MAT2A clinical candidate AG-270 with antimitotic taxanes.
Collapse
Affiliation(s)
- Peter Kalev
- Biology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Marc L Hyer
- Pharmacology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Stefan Gross
- Biochemistry and Biophysics, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Zenon Konteatis
- Chemistry, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Chi-Chao Chen
- Bioinformatics, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Mark Fletcher
- Bioinformatics, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Max Lein
- Drug Metabolism and Pharmacokinetics, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Elia Aguado-Fraile
- Clinical Biomarkers, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Victoria Frank
- Biology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Amelia Barnett
- Biology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Everton Mandley
- Pharmacology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Joshua Goldford
- Cell Metabolism, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Yue Chen
- Drug Metabolism and Pharmacokinetics, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Katie Sellers
- Cell Metabolism, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Sebastian Hayes
- Cell Metabolism, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Kate Lizotte
- Cell Metabolism, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Phong Quang
- Biology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Yesim Tuncay
- Biology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Michelle Clasquin
- Cell Metabolism, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Rachel Peters
- Toxicology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Jaclyn Weier
- Biology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Eric Simone
- Chemistry, Manufacturing and Control, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Joshua Murtie
- Biology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA; Pharmacology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Wei Liu
- Bioinformatics, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Raj Nagaraja
- Drug Metabolism and Pharmacokinetics, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Lenny Dang
- Biochemistry and Biophysics, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Zhihua Sui
- Chemistry, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Scott A Biller
- Chemistry, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Jeremy Travins
- Chemistry, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Kevin M Marks
- Biology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA
| | - Katya Marjon
- Biology, Agios Pharmaceuticals, Inc., Cambridge, MA 02139, USA.
| |
Collapse
|
9
|
Ganapathi RN, Norris EJ, Sutker AP, Klotz KE, Ganapathi MK. Targeting Aurora A Kinase (AAK) in Platinum-Resistant High Grade Serous Ovarian Cancer. Front Oncol 2020; 10:1354. [PMID: 32974133 PMCID: PMC7466726 DOI: 10.3389/fonc.2020.01354] [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: 03/26/2020] [Accepted: 06/29/2020] [Indexed: 11/13/2022] Open
Abstract
Aurora A kinase (AAK) involved in G2-M transition is functionally involved in centrosome maturation and maintaining an active spindle assembly checkpoint. We tested the hypothesis that in platinum-taxane resistant high grade serous ovarian cancer (HGSOC) inhibition of AAK involved in G2-M transition would enhance the anti-tumor activity of cisplatin (CP) or paclitaxel (PT). Using HGSOC cell lines from platinum-taxane refractory patients that do not harbor BRCA1/2 mutations, we tested the anti-tumor activity of CP, or PT alone or in combination with the AAK inhibitor alisertib (AL). Treatment with CP for 3 h or PT for 6 h followed sequentially by AL for 48 h led to a significant decrease in cell survival (p < 0.001) compared to treatment with either drug alone in HGSOC cells but not in immortalized normal human ovarian surface epithelium or normal human fallopian tube secretory epithelium cells. The treatment with CP or PT followed by AL also led to a significant increase in reactive oxygen species (p < 0.05), apoptosis (p < 0.001) and accumulation of cells in G2/M that was accompanied by a modest increase in expression of AAK. Downregulation of AAK, but not aurora B kinase, with targeted siRNAs also significantly enhanced apoptosis by CP or PT, suggesting that AL specifically targeted AAK. In summary, in HGSOC without BRCA1/2 mutations, CP, or PT resistance can potentially be circumvented by sequential treatment with AL that inhibits AAK involved in G2-M transition.
Collapse
Affiliation(s)
- Ram N Ganapathi
- Carolinas Medical Center, Levine Cancer Institute, Charlotte, NC, United States
| | - Eric J Norris
- Carolinas Medical Center, Levine Cancer Institute, Charlotte, NC, United States
| | - Ashley P Sutker
- Carolinas Medical Center, Levine Cancer Institute, Charlotte, NC, United States
| | - Kaitlin E Klotz
- Carolinas Medical Center, Levine Cancer Institute, Charlotte, NC, United States
| | - Mahrukh K Ganapathi
- Carolinas Medical Center, Levine Cancer Institute, Charlotte, NC, United States
| |
Collapse
|
10
|
Sootome H, Miura A, Masuko N, Suzuki T, Uto Y, Hirai H. Aurora A Inhibitor TAS-119 Enhances Antitumor Efficacy of Taxanes In Vitro and In Vivo: Preclinical Studies as Guidance for Clinical Development and Trial Design. Mol Cancer Ther 2020; 19:1981-1991. [PMID: 32788206 DOI: 10.1158/1535-7163.mct-20-0036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 06/04/2020] [Accepted: 07/30/2020] [Indexed: 11/16/2022]
Abstract
TAS-119 is a novel orally active, selective inhibitor of Aurora kinase A identified as a clinical candidate for efficacy testing in combination with taxanes. In vitro, TAS-119 enhanced cell growth inhibition of paclitaxel in multiple human cancer cell lines derived from various tissues, including paclitaxel-resistant cell lines. Interestingly, TAS-119 did not enhance paclitaxel antitumor activity in normal lung diploid fibroblast cell lines WI-38 and MRC5. In vivo, TAS-119 enhanced the antitumor efficacy of paclitaxel and docetaxel in multiple models at doses inhibitory to Aurora A in tumors. Moreover, the drug combination was well tolerated, and TAS-119 did not exaggerate clinically documented side effects of taxanes, neutropenia and neurotoxicity, in rats. The same TAS-119 concentration enhanced the cell growth inhibitory activity of three clinically approved taxanes, paclitaxel, docetaxel, and cabazitaxel. The degree of enhancement calculated as fold of change of the IC50 value for each taxane was almost the same among the three taxanes. We conducted in vitro and in vivo experiments to develop an optimized combination therapy regimen for TAS-119 with paclitaxel/docetaxel. Using in vitro and in vivo models, we tested the drug administration order for TAS-119 combined with paclitaxel and the TAS-119 treatment duration. The best regimen in preclinical models was combining paclitaxel or docetaxel treatment with 4 days of TAS-119 dosing, which was initiated on the same day as the paclitaxel or docetaxel administration or one day later. This information provided guidance for the design of a clinical trial of TAS-119 and paclitaxel or docetaxel combination.
Collapse
Affiliation(s)
- Hiroshi Sootome
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, Tsukuba, Japan
| | - Akihiro Miura
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, Tsukuba, Japan.,Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | - Norio Masuko
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, Tsukuba, Japan
| | - Takamasa Suzuki
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, Tsukuba, Japan
| | - Yoshihiro Uto
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | - Hiroshi Hirai
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd, Tsukuba, Japan.
| |
Collapse
|
11
|
Bernatz S, Ilina EI, Devraj K, Harter PN, Mueller K, Kleber S, Braun Y, Penski C, Renner C, Halder R, Jennewein L, Solbach C, Thorsen F, Pestalozzi BC, Mischo A, Mittelbronn M. Impact of Docetaxel on blood-brain barrier function and formation of breast cancer brain metastases. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:434. [PMID: 31665089 PMCID: PMC6819416 DOI: 10.1186/s13046-019-1427-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 09/23/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND Breast cancer (BC) is the most frequent malignant tumor in females and the 2nd most common cause of brain metastasis (BM), that are associated with a fatal prognosis. The increasing incidence from 10% up to 40% is due to more effective treatments of extracerebral sites with improved prognosis and increasing use of MRI in diagnostics. A frequently administered, potent chemotherapeutic group of drugs for BC treatment are taxanes usually used in the adjuvant and metastatic setting, which, however, have been suspected to be associated with a higher incidence of BM. The aim of our study was to experimentally analyze the impact of the taxane docetaxel (DTX) on brain metastasis formation, and to elucidate the underlying molecular mechanism. METHODS A monocentric patient cohort was analyzed to determine the association of taxane treatment and BM formation. To identify the specific impact of DTX, a murine brain metastatic model upon intracardial injection of breast cancer cells was conducted. To approach the functional mechanism, dynamic contrast-enhanced MRI and electron microscopy of mice as well as in-vitro transendothelial electrical resistance (TEER) and tracer permeability assays using brain endothelial cells (EC) were carried out. PCR-based, immunohistochemical and immunoblotting analyses with additional RNA sequencing of murine and human ECs were performed to explore the molecular mechanisms by DTX treatment. RESULTS Taxane treatment was associated with an increased rate of BM formation in the patient cohort and the murine metastatic model. Functional studies did not show unequivocal alterations of blood-brain barrier properties upon DTX treatment in-vivo, but in-vitro assays revealed a temporary DTX-related barrier disruption. We found disturbance of tubulin structure and upregulation of tight junction marker claudin-5 in ECs. Furthermore, upregulation of several members of the tubulin family and downregulation of tetraspanin-2 in both, murine and human ECs, was induced. CONCLUSION In summary, a higher incidence of BM was associated with prior taxane treatment in both a patient cohort and a murine mouse model. We could identify tubulin family members and tetraspanin-2 as potential contributors for the destabilization of the blood-brain barrier. Further analyses are needed to decipher the exact role of those alterations on tumor metastatic processes in the brain.
Collapse
Affiliation(s)
- Simon Bernatz
- Edinger Institute, Institute of Neurology, University of Frankfurt am Main, Frankfurt, Germany
| | - Elena I Ilina
- Edinger Institute, Institute of Neurology, University of Frankfurt am Main, Frankfurt, Germany.,Luxembourg Center of Neuropathology (LCNP), Luxembourg, Luxembourg.,Department of Oncology, Luxembourg Institute of Health (LIH), NORLUX Neuro-Oncology Laboratory, Luxembourg, Luxembourg
| | - Kavi Devraj
- Edinger Institute, Institute of Neurology, University of Frankfurt am Main, Frankfurt, Germany.,Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
| | - Patrick N Harter
- Edinger Institute, Institute of Neurology, University of Frankfurt am Main, Frankfurt, Germany.,Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Klaus Mueller
- Edinger Institute, Institute of Neurology, University of Frankfurt am Main, Frankfurt, Germany
| | - Sascha Kleber
- Oncology Centre Hirslanden and Zurich, Zurich, Switzerland
| | - Yannick Braun
- Edinger Institute, Institute of Neurology, University of Frankfurt am Main, Frankfurt, Germany
| | - Cornelia Penski
- Edinger Institute, Institute of Neurology, University of Frankfurt am Main, Frankfurt, Germany
| | | | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Lukas Jennewein
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, D-60590, Frankfurt, Germany
| | - Christine Solbach
- Department of Gynecology and Obstetrics, School of Medicine, J. W. Goethe-University, Theodor-Stern-Kai 7, D-60590, Frankfurt, Germany
| | - Frits Thorsen
- KG Jebsen Brain Tumor Research Centre, University of Bergen, Bergen, Norway.,Molecular Imaging Center, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Bernhard C Pestalozzi
- Department of Medical Oncology and Hematology, University Hospital Zurich (USZ), Rämistrasse 100, CH-8891, Zurich, Switzerland
| | - Axel Mischo
- Department of Medical Oncology and Hematology, University Hospital Zurich (USZ), Rämistrasse 100, CH-8891, Zurich, Switzerland.
| | - Michel Mittelbronn
- Edinger Institute, Institute of Neurology, University of Frankfurt am Main, Frankfurt, Germany. .,Luxembourg Center of Neuropathology (LCNP), Luxembourg, Luxembourg. .,Department of Oncology, Luxembourg Institute of Health (LIH), NORLUX Neuro-Oncology Laboratory, Luxembourg, Luxembourg. .,Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg. .,National Center of Pathology (NCP), Luxembourg Center of Neuropathology (LCNP), Laboratoire national de santé (LNS), 1, Rue Louis Rech, L-3555, Dudelange, Luxembourg.
| |
Collapse
|
12
|
Liu N, Wang YA, Sun Y, Ecsedy J, Sun J, Li X, Wang P. Inhibition of Aurora A enhances radiosensitivity in selected lung cancer cell lines. Respir Res 2019; 20:230. [PMID: 31647033 PMCID: PMC6813099 DOI: 10.1186/s12931-019-1194-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 09/22/2019] [Indexed: 12/15/2022] Open
Abstract
Background In mammalian cells, Aurora serine/threonine kinases (Aurora A, B, and C) are expressed in a cell cycle-dependent fashion as key mitotic regulators required for the maintenance of chromosomal stability. Aurora-A (AURKA) has been proven to be an oncogene in a variety of cancers; however, whether its expression relates to patient survival and the association with radiotherapy remains unclear in non-small cell lung cancer (NSCLC). Methods Here, we first analyzed AURKA expression in 63 NSCLC tumor samples by immunohistochemistry (IHC) and used an MTS assay to compare cell survival by targeting AURKA with MLN8237 (Alisertib) in H460 and HCC2429 (P53-competent), and H1299 (P53-deficient) cell lines. The radiosensitivity of MLN8237 was further evaluated by clonogenic assay. Finally, we examined the effect of combining radiation and AURKA inhibition in vivo with a xenograft model and explored the potential mechanism. Results We found that increased AURKA expression correlated with decreased time to progression and overall survival (p = 0.0447 and 0.0096, respectively). AURKA inhibition using 100 nM MLN8237 for 48 h decreases cell growth in a partially P53-dependent manner, and the survival rates of H460, HCC2429, and H1299 cells were 56, 50, and 77%, respectively. In addition, the survival of H1299 cells decreased 27% after ectopic restoration of P53 expression, and the radiotherapy enhancement was also influenced by P53 expression (DER H460 = 1.33; HCC2429 = 1.35; H1299 = 1.02). Furthermore, tumor growth of H460 was delayed significantly in a subcutaneous mouse model exposed to both MLN8237 and radiation. Conclusions Taken together, our results confirmed that the expression of AURKA correlated with decreased NSCLC patient survival, and it might be a promising inhibition target when combined with radiotherapy, especially for P53-competent lung cancer cells. Modulation of P53 function could provide a new option for reversing cell resistance to the AURKA inhibitor MLN8237, which deserves further investigation.
Collapse
Affiliation(s)
- Ningbo Liu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin, 300060, China.
| | - Yong Antican Wang
- Biomed Innovation Center of Yehoo Group Co. Ltd., Shenzhen, 518000, China.,Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Yunguang Sun
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jeffrey Ecsedy
- Takeda Pharmaceuticals International Co, Cambridge, MA, UK
| | - Jifeng Sun
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Xue Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin, 300060, China.
| |
Collapse
|
13
|
Marvin CM, Ding S, White RE, Orlova N, Wang Q, Zywot EM, Vickerman BM, Harr L, Tarrant TK, Dayton PA, Lawrence DS. On Command Drug Delivery via Cell-Conveyed Phototherapeutics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1901442. [PMID: 31353802 PMCID: PMC6739139 DOI: 10.1002/smll.201901442] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/04/2019] [Indexed: 05/08/2023]
Abstract
Herein, the use of red blood cells (RBCs) as carriers of cytoplasmically interned phototherapeutic agents is described. Photolysis promotes drug release from the RBC carrier thereby providing the means to target specific diseased sites. This strategy is realized with a vitamin B12-taxane conjugate (B12-TAX), in which the drug is linked to the vitamin via a photolabile CoC bond. The conjugate is introduced into mouse RBCs (mRBCs) via a pore-forming/pore-resealing procedure and is cytoplasmically retained due to the membrane impermeability of B12. Photolysis separates the taxane from the B12 cytoplasmic anchor, enabling the drug to exit the RBC carrier. A covalently appended Cy5 antenna sensitizes the conjugate (Cy5-B12-TAX) to far red light, thereby circumventing the intense light absorbing properties of hemoglobin (350-600 nm). Microscopy and imaging flow cytometry reveal that Cy5-B12-TAX-loaded mRBCs act as drug carriers. Furthermore, intravital imaging of mice furnish a real time assessment of circulating phototherapeutic-loaded mRBCs as well as evidence of the targeted photorelease of the taxane upon photolysis. Histopathology confirms that drug release occurs in a well resolved spatiotemporal fashion. Finally, acoustic angiography is employed to assess the consequences of taxane release at the tumor site in Nu/Nu-tumor-bearing mice.
Collapse
Affiliation(s)
- Christina M Marvin
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Song Ding
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Rachel E White
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, 27599, USA
| | - Natalia Orlova
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Qunzhao Wang
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Emilia M Zywot
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Brianna M Vickerman
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Lauren Harr
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Teresa K Tarrant
- Department of Medicine, Division of Rheumatology and Immunology, Duke University, Durham, NC, 27710, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, 27599, USA
| | - David S Lawrence
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
14
|
Yang N, Wang C, Wang J, Wang Z, Huang D, Yan M, Kamran M, Liu Q, Xu B. Aurora kinase A stabilizes FOXM1 to enhance paclitaxel resistance in triple-negative breast cancer. J Cell Mol Med 2019; 23:6442-6453. [PMID: 31359594 PMCID: PMC6714217 DOI: 10.1111/jcmm.14538] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/21/2019] [Accepted: 06/03/2019] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) has a relatively poor outcome. Acquired chemoresistance is a major clinical challenge for TNBC patients. Previously, we reported that kinase-dead Aurora kinase A (Aurora-A) could effectively transactivate the FOXM1 promoter. Here, we demonstrate an additional pathway through which Aurora-A stabilizes FOXM1 by attenuating its ubiquitin in TNBC. Specifically, Aurora-A stabilizes FOXM1 in late M phase and early G1 phase of the cell cycle, which promotes proliferation of TNBC cells. Knock-down of Aurora-A significantly suppresses cell proliferation in TNBC cell lines and can be rescued by FOXM1 overexpression. We observe that paclitaxel-resistant TNBC cells exhibit high expression of Aurora-A and FOXM1. Overexpression of Aurora-A offers TNBC cells an additional growth advantage and protection against paclitaxel. Moreover, Aurora-A and FOXM1 could be simultaneously targeted by thiostrepton. Combination of thiostrepton and paclitaxel treatment reverses paclitaxel resistance and significantly inhibits cell proliferation. In conclusion, our study reveals additional mechanism through which Aurora-A regulates FOXM1 and provides a new therapeutic strategy to treat paclitaxel-resistant triple-negative breast cancer.
Collapse
Affiliation(s)
- Na Yang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chang Wang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jian Wang
- Department of Pathology, GanZhou Municipal People's Hospital, NanChang University, GanZhou, China
| | - Zifeng Wang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Di Huang
- Department of Breast Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Min Yan
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Muhammad Kamran
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Quentin Liu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.,State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - BangLao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
15
|
van Gijn SE, Wierenga E, van den Tempel N, Kok YP, Heijink AM, Spierings DCJ, Foijer F, van Vugt MATM, Fehrmann RSN. TPX2/Aurora kinase A signaling as a potential therapeutic target in genomically unstable cancer cells. Oncogene 2019; 38:852-867. [PMID: 30177840 PMCID: PMC6367211 DOI: 10.1038/s41388-018-0470-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/30/2018] [Accepted: 07/30/2018] [Indexed: 12/17/2022]
Abstract
Genomic instability is a hallmark feature of cancer cells, and can be caused by defective DNA repair, for instance due to inactivation of BRCA2. Paradoxically, loss of Brca2 in mice results in embryonic lethality, whereas cancer cells can tolerate BRCA2 loss. This holds true for multiple DNA repair genes, and suggests that cancer cells are molecularly "rewired" to cope with defective DNA repair and the resulting high levels of genomic instability. In this study, we aim to identify genes that genomically unstable cancer cells rely on for their survival. Using functional genomic mRNA (FGmRNA) profiling, 16,172 cancer samples were previously ranked based on their degree of genomic instability. We analyzed the top 250 genes that showed a positive correlation between FGmRNA levels and the degree of genomic instability, in a co-functionality network. Within this co-functionality network, a strong cluster of 11 cell cycle-related genes was identified, including TPX2. We then assessed the dependency on these 11 genes in the context of survival of genomically unstable cancer cells, induced by BRCA2 inactivation. Depletion of TPX2 or its associated kinase Aurora-A preferentially reduced cell viability in a panel of BRCA2-deficient cancer cells. In line with these findings, BRCA2-depleted and BRCA2-mutant human cell lines, or tumor cell lines derived from Brca2-/-;p53-/- mice showed increased sensitivity to the Aurora-A kinase inhibitor alisertib, with delayed mitotic progression and frequent mitotic failure. Our findings reveal that BRCA2-deficient cancer cells show enhanced sensitivity to inactivation of TPX2 or its partner Aurora-A, which points at an actionable dependency of genomically unstable cancers.
Collapse
Affiliation(s)
- Stephanie E van Gijn
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Elles Wierenga
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nathalie van den Tempel
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Yannick P Kok
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anne Margriet Heijink
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Diana C J Spierings
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marcel A T M van Vugt
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Rudolf S N Fehrmann
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
16
|
Falchook G, Coleman RL, Roszak A, Behbakht K, Matulonis U, Ray-Coquard I, Sawrycki P, Duska LR, Tew W, Ghamande S, Lesoin A, Schwartz PE, Buscema J, Fabbro M, Lortholary A, Goff B, Kurzrock R, Martin LP, Gray HJ, Fu S, Sheldon-Waniga E, Lin HM, Venkatakrishnan K, Zhou X, Leonard EJ, Schilder RJ. Alisertib in Combination With Weekly Paclitaxel in Patients With Advanced Breast Cancer or Recurrent Ovarian Cancer: A Randomized Clinical Trial. JAMA Oncol 2019; 5:e183773. [PMID: 30347019 DOI: 10.1001/jamaoncol.2018.3773] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Importance There is an unmet medical need for the treatment of recurrent ovarian cancer, and new approaches are needed to improve progression-free survival (PFS) and overall survival. Objective This phase 1/2 study evaluated the activity of alisertib in combination with weekly paclitaxel in patients with breast (phase 1) and ovarian cancer (phase 1 and phase 2). Design, Setting, and Participants An open-label phase 1 and randomized phase 2 clinical trial conducted from April 16, 2010, for phase 1 and March 28, 2012, to August 12, 2013, for phase 2 was conducted at 33 sites (United States, France, and Poland). Data are reported from a cutoff date of August 12, 2014, with a median duration of follow-up of 7.2 months in the alisertib plus paclitaxel arm and 4.6 months in the paclitaxel arm. A total of 191 women with advanced breast (phase 1 only) or recurrent ovarian cancer were enrolled, including 142 patients randomized to alisertib plus paclitaxel (n = 73) or paclitaxel alone (n = 69) in the phase 2 study. Interventions Patients were randomized 1:1 stratified by platinum-free interval (refractory, 0-6 months, 6-12 months) and prior weekly taxane treatment (yes, no) to receive alisertib 40 mg twice per day orally and 3 days on and 4 days off for 3 weeks, plus paclitaxel (60 mg/m2 intravenously, days 1, 8, and 15), or weekly paclitaxel 80 mg/m2 intravenously in 28-day cycles. Main Outcomes and Measures Primary endpoint was PFS; primary efficacy analysis and safety analysis used modified intention to treat (mITT) population (all randomized patients who received ≥1 dose of study drug). Results The median age for the 191 patients enrolled in phase 1 was 59 (range, 29-75) years. The median age for the 142 patients enrolled in phase 2 was 63 (range, 30-81) years for patients receiving alisertib plus paclitaxel and 61 (range, 41-81) years for patients receiving paclitaxel. At data cutoff, 107 (75%) patients had a documented PFS event; 52 (71%) in the alisertib plus paclitaxel arm, and 55 (80%) in the paclitaxel arm. Median PFS was 6.7 months with alisertib plus paclitaxel vs 4.7 months with paclitaxel (HR, 0.75; 80% CI, 0.58-0.96; P = .14; 2-sided P value cutoff = .20 to be considered worthy of further investigation). Drug-related grade 3 or higher adverse events were reported in 63 (86%) vs 14 (20%) patients in the alisertib plus paclitaxel and paclitaxel arms, including 56 (77%) vs 7 (10%) neutropenia, 18 (25%) vs 0 stomatitis, and 10 (14%) vs 2 (3%) anemia; 54 (74%) vs 17 (25%) had adverse events leading to dose reductions. Two patients died during the study (1 in each arm); neither death was considered related to study drug. Conclusions and Relevance The primary endpoint, PFS, significantly favored alisertib plus paclitaxel over paclitaxel alone. Further investigation is warranted. Trial Registration ClinicalTrials.gov identifier: NCT01091428.
Collapse
Affiliation(s)
- Gerald Falchook
- Sarah Cannon Research Institute at HealthONE, Denver, Colorado
| | | | - Andrzej Roszak
- Greater Poland Cancer Centre/University of Medical Sciences, Poznan, Poland
| | - Kian Behbakht
- Department of Gynecologic Oncology, University of Colorado School of Medicine, Aurora
| | - Ursula Matulonis
- Gynecologic Oncology Program, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Piotr Sawrycki
- Department of Oncology and Chemotherapy, L. Rydygiera District Hospital, Torun, Poland
| | - Linda R Duska
- University of Virginia Health System, Charlottesville
| | - William Tew
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sharad Ghamande
- Georgia Cancer Center at Augusta University, Augusta, Georgia
| | | | | | | | | | | | | | | | | | | | - Siqing Fu
- University of Texas, MD Anderson Cancer Center, Houston
| | - Emily Sheldon-Waniga
- Millennium Pharmaceuticals Inc (a wholly owned subsidiary of Takeda Pharmaceutical Company Limited), Cambridge, Massachusetts
| | - Huamao Mark Lin
- Millennium Pharmaceuticals Inc (a wholly owned subsidiary of Takeda Pharmaceutical Company Limited), Cambridge, Massachusetts
| | - Karthik Venkatakrishnan
- Millennium Pharmaceuticals Inc (a wholly owned subsidiary of Takeda Pharmaceutical Company Limited), Cambridge, Massachusetts
| | - Xiaofei Zhou
- Millennium Pharmaceuticals Inc (a wholly owned subsidiary of Takeda Pharmaceutical Company Limited), Cambridge, Massachusetts
| | - E Jane Leonard
- Millennium Pharmaceuticals Inc (a wholly owned subsidiary of Takeda Pharmaceutical Company Limited), Cambridge, Massachusetts
| | - Russell J Schilder
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Quantitative translational modeling to facilitate preclinical to clinical efficacy & toxicity translation in oncology. Future Sci OA 2018; 4:FSO306. [PMID: 29796306 PMCID: PMC5961452 DOI: 10.4155/fsoa-2017-0152] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/12/2018] [Indexed: 12/12/2022] Open
Abstract
Significant scientific advances in biomedical research have expanded our knowledge of the molecular basis of carcinogenesis, mechanisms of cancer growth, and the importance of the cancer immunity cycle. However, despite scientific advances in the understanding of cancer biology, the success rate of oncology drug development remains the lowest among all therapeutic areas. In this review, some of the key translational drug development objectives in oncology will be outlined. The literature evidence of how mathematical modeling could be used to build a unifying framework to answer these questions will be summarized with recommendations on the strategies for building such a mathematical framework to facilitate the prediction of clinical efficacy and toxicity of investigational antineoplastic agents. Together, the literature evidence suggests that a rigorous and unifying preclinical to clinical translational framework based on mathematical models is extremely valuable for making go/no-go decisions in preclinical development, and for planning early clinical studies.
Collapse
|
18
|
The Aurora kinase A inhibitor TC-A2317 disrupts mitotic progression and inhibits cancer cell proliferation. Oncotarget 2018; 7:84718-84735. [PMID: 27713168 PMCID: PMC5356694 DOI: 10.18632/oncotarget.12448] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/16/2016] [Indexed: 01/22/2023] Open
Abstract
Mitotic progression is crucial for the maintenance of chromosomal stability. A proper progression is ensured by the activities of multiple kinases. One of these enzymes, the serine/threonine kinase Aurora A, is required for proper mitosis through the regulation of centrosome and spindle assembly. In this study, we functionally characterized a newly developed Aurora kinase A inhibitor, TC-A2317. In human lung cancer cells, TC-A2317 slowed proliferation by causing aberrant formation of centrosome and microtubule spindles and prolonging the duration of mitosis. Abnormal mitotic progression led to accumulation of cells containing micronuclei or multinuclei. Furthermore, TC-A2317–treated cells underwent apoptosis, autophagy or senescence depending on cell type. In addition, TC-A2317 inactivated the spindle assembly checkpoint triggered by paclitaxel, thereby exacerbating mitotic catastrophe. Consistent with this, the expression level of Aurora A in tumors was inversely correlated with survival in lung cancer patients. Collectively, these data suggest that inhibition of Aurora kinase A using TC-A2317 is a promising target for anti-cancer therapeutics.
Collapse
|
19
|
Phase I trial to evaluate the addition of alisertib to fulvestrant in women with endocrine-resistant, ER+ metastatic breast cancer. Breast Cancer Res Treat 2018; 168:639-647. [PMID: 29289986 PMCID: PMC5842248 DOI: 10.1007/s10549-017-4616-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/07/2017] [Indexed: 02/01/2023]
Abstract
PURPOSE In estrogen receptor-positive (ER+) breast cancer models, activation of Aurora A kinase (AURKA) is associated with downregulation of ERα expression and resistance to endocrine therapy. Alisertib is an oral selective inhibitor of AURKA. The primary objectives of this phase I trial were to determine the recommended phase II dose (RP2D) and evaluate the toxicities and clinical activity of alisertib combined with fulvestrant in patients with ER+ metastatic breast cancer (MBC). METHODS In this standard 3 + 3 dose-escalation phase I study, postmenopausal patients with endocrine-resistant, ER+ MBC previously treated with endocrine therapy were assigned to one of two dose levels of alisertib (40 or 50 mg) in combination with fixed-dose fulvestrant. RESULTS Ten patients enrolled, of which nine were evaluable for the primary endpoint. The median patient age was 59. All patients had secondary (acquired) endocrine resistance, and all had received prior aromatase inhibitor. Six had experienced disease progression on fulvestrant. There were no severe (grade 3+) toxicities reported during cycle 1 at either dose level. The median progression-free survival time was 12.4 months (95% CI 5.3-not met), and the 6-month clinical benefit rate was 77.8% (95% CI 40.0-87.2%). CONCLUSIONS In patients with endocrine-resistant, ER+ MBC, alisertib in combination with fulvestrant was well tolerated. A favorable safety profile was observed. The RP2D is 50 mg twice daily on days 1-3, 8-10, and 15-17 of a 28-day cycle with standard dose fulvestrant. Promising antitumor activity was observed, including activity among patients with prior progression on fulvestrant.
Collapse
|
20
|
Chen W, Chen R, Li J, Fu Y, Yang L, Su H, Yao Y, Li L, Zhou T, Lu W. Pharmacokinetic/Pharmacodynamic Modeling of Schedule-Dependent Interaction between Docetaxel and Cabozantinib in Human Prostate Cancer Xenograft Models. J Pharmacol Exp Ther 2017; 364:13-25. [PMID: 29084815 DOI: 10.1124/jpet.117.243931] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 10/26/2017] [Indexed: 01/03/2023] Open
Abstract
In this work, a semimechanistic pharmacokinetic/pharmacodynamic (PK/PD) model to quantitatively describe the antitumor activity of docetaxel (Doc) and cabozantinib (Cab) under monotherapy, concurrent therapy, interval therapy, and different sequential therapy in mouse xenograft models of castration-resistant prostate cancer was developed and evaluated. The pharmacokinetics (PK) of Doc and Cab when administered separately and simultaneously were investigated in nude mice, and PD study was conducted in tumor-bearing mice treated with different dosing schedules. The PK interaction between Doc and Cab was expressed by adding the effect of Cab on the clearance of Doc in the PK model. And the PD interaction between the two drugs was demonstrated by the developed PK/PD model through the combination index "φ" Our results showed that the concurrent therapy and Doc followed by Cab (Doc ∼ Cab) sequential therapy exhibited better tumor inhibitory efficacy than monotherapy. The Cab followed by Doc (Cab ∼ Doc) sequential schedule was less effective than monotherapy, and the interval therapy did not enhance the antitumor efficacy compared with the concurrent therapy. The parameter φ estimated from the PK/PD model quantitatively characterized the action between Doc and Cab. There was no significant PD interaction between Doc and Cab in both the concurrent schedule and the interval schedule, whereas the effect of the two drugs in the "Doc ∼ Cab" and "Cab ∼ Doc" sequential schedule was synergistic and antagonistic, respectively. The proposed model properly described the antitumor effects of Doc and Cab under different treatment schedules and could be used for dose optimization through model-based simulation.
Collapse
Affiliation(s)
- Wenjun Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Rong Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Jian Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Yu Fu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Liang Yang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Hong Su
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Ye Yao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Liang Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Tianyan Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Wei Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| |
Collapse
|
21
|
Paolini M, Poul L, Darmon A, Germain M, Pottier A, Levy L, Vibert E. A new opportunity for nanomedicines: Micellar cytochrome P450 inhibitors to improve drug efficacy in a cancer therapy model. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1715-1723. [DOI: 10.1016/j.nano.2017.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 01/17/2017] [Accepted: 03/17/2017] [Indexed: 12/25/2022]
|
22
|
Fathi AT, Wander SA, Blonquist TM, Brunner AM, Amrein PC, Supko J, Hermance NM, Manning AL, Sadrzadeh H, Ballen KK, Attar EC, Graubert TA, Hobbs G, Joseph C, Perry AM, Burke M, Silver R, Foster J, Bergeron M, Ramos AY, Som TT, Fishman KM, McGregor KL, Connolly C, Neuberg DS, Chen YB. Phase I study of the aurora A kinase inhibitor alisertib with induction chemotherapy in patients with acute myeloid leukemia. Haematologica 2016; 102:719-727. [PMID: 28034990 PMCID: PMC5395112 DOI: 10.3324/haematol.2016.158394] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 12/26/2016] [Indexed: 12/17/2022] Open
Abstract
Aberrant expression of aurora kinase A is implicated in the genesis of various
neoplasms, including acute myeloid leukemia. Alisertib, an aurora A kinase
inhibitor, has demonstrated efficacy as monotherapy in trials of myeloid
malignancy, and this efficacy appears enhanced in combination with conventional
chemotherapies. In this phase I, dose-escalation study, newly diagnosed patients
received conventional induction with cytarabine and idarubicin, after which
alisertib was administered for 7 days. Dose escalation occurred
via cohorts. Patients could then receive up to four cycles
of consolidation, incorporating alisertib, and thereafter alisertib maintenance
for up to 12 months. Twenty-two patients were enrolled. One dose limiting
toxicity occurred at dose level 2 (prolonged thrombocytopenia), and the
recommended phase 2 dose was established at 30mg twice daily. Common
therapy-related toxicities included cytopenias and mucositis. Only three
(14%) patients had persistent disease at mid-cycle, requiring
“5+2” reinduction. The composite remission rate (complete
remission and complete remission with incomplete neutrophil recovery) was
86% (nineteen of twenty-two patients; 90% CI
68–96%). Among those over age 65 and those with high-risk
disease (secondary acute leukemia or cytogenetically high-risk disease), the
composite remission rate was 88% and 100%, respectively. The
median follow up was 13.5 months. Of those treated at the recommended phase 2
dose, the 12-month overall survival and progression-free survival were
62% (90% CI 33–81%) and 42% (90%
CI 17–65%), respectively. Alisertib is well tolerated when
combined with induction chemotherapy in acute myeloid leukemia, with a promising
suggestion of efficacy. (clinicaltrials.gov Identifier:01779843).
Collapse
Affiliation(s)
- Amir T Fathi
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Seth A Wander
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | | | - Andrew M Brunner
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Philip C Amrein
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Jeffrey Supko
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Nicole M Hermance
- Worcester Polytechnic Institute, Department of Biology, Worcester, MA, USA
| | - Amity L Manning
- Worcester Polytechnic Institute, Department of Biology, Worcester, MA, USA
| | - Hossein Sadrzadeh
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Karen K Ballen
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Eyal C Attar
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Timothy A Graubert
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Gabriela Hobbs
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Christelle Joseph
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Ashley M Perry
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Meghan Burke
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Regina Silver
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Julia Foster
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Meghan Bergeron
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Aura Y Ramos
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Tina T Som
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Kaitlyn M Fishman
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Kristin L McGregor
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Christine Connolly
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| | - Donna S Neuberg
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Yi-Bin Chen
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA
| |
Collapse
|
23
|
Venkatakrishnan K, Ecsedy JA. Enhancing value of clinical pharmacodynamics in oncology drug development: An alliance between quantitative pharmacology and translational science. Clin Pharmacol Ther 2016; 101:99-113. [PMID: 27804123 DOI: 10.1002/cpt.544] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/23/2016] [Accepted: 10/23/2016] [Indexed: 01/08/2023]
Abstract
Clinical pharmacodynamic evaluation is a key component of the "pharmacologic audit trail" in oncology drug development. We posit that its value can and should be greatly enhanced via application of a robust quantitative pharmacology framework informed by biologically mechanistic considerations. Herein, we illustrate examples of intersectional blindspots across the disciplines of quantitative pharmacology and translational science and offer a roadmap aimed at enhancing the caliber of clinical pharmacodynamic research in the development of oncology therapeutics.
Collapse
Affiliation(s)
- K Venkatakrishnan
- Quantitative Clinical Pharmacology, Takeda Pharmaceuticals International Co, Cambridge, Massachusetts, USA
| | - J A Ecsedy
- Translational and Biomarker Research, Takeda Pharmaceuticals International Co, Cambridge, Massachusetts, USA
| |
Collapse
|
24
|
Nakayama Y, Inoue T. Antiproliferative Fate of the Tetraploid Formed after Mitotic Slippage and Its Promotion; A Novel Target for Cancer Therapy Based on Microtubule Poisons. Molecules 2016; 21:molecules21050663. [PMID: 27213315 PMCID: PMC6274067 DOI: 10.3390/molecules21050663] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/10/2016] [Accepted: 05/13/2016] [Indexed: 12/20/2022] Open
Abstract
Microtubule poisons inhibit spindle function, leading to activation of spindle assembly checkpoint (SAC) and mitotic arrest. Cell death occurring in prolonged mitosis is the first target of microtubule poisons in cancer therapies. However, even in the presence of microtubule poisons, SAC and mitotic arrest are not permanent, and the surviving cells exit the mitosis without cytokinesis (mitotic slippage), becoming tetraploid. Another target of microtubule poisons-based cancer therapy is antiproliferative fate after mitotic slippage. The ultimate goal of both the microtubule poisons-based cancer therapies involves the induction of a mechanism defined as mitotic catastrophe, which is a bona fide intrinsic oncosuppressive mechanism that senses mitotic failure and responds by driving a cell to an irreversible antiproliferative fate of death or senescence. This mechanism of antiproliferative fate after mitotic slippage is not as well understood. We provide an overview of mitotic catastrophe, and explain new insights underscoring a causal association between basal autophagy levels and antiproliferative fate after mitotic slippage, and propose possible improved strategies. Additionally, we discuss nuclear alterations characterizing the mitotic catastrophe (micronuclei, multinuclei) after mitotic slippage, and a possible new type of nuclear alteration (clustered micronuclei).
Collapse
Affiliation(s)
- Yuji Nakayama
- Division of Functional Genomics, Research Center for Bioscience and Technology, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
| | - Toshiaki Inoue
- Division of Human Genome Science, Department of Molecular and Cellular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
| |
Collapse
|
25
|
Graff JN, Higano CS, Hahn NM, Taylor MH, Zhang B, Zhou X, Venkatakrishnan K, Leonard EJ, Sarantopoulos J. Open-label, multicenter, phase 1 study of alisertib (MLN8237), an aurora A kinase inhibitor, with docetaxel in patients with solid tumors. Cancer 2016; 122:2524-33. [PMID: 27192055 DOI: 10.1002/cncr.30073] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 03/03/2016] [Accepted: 03/07/2016] [Indexed: 11/08/2022]
Abstract
BACKGROUND This study was designed to determine the safety, tolerability, and pharmacokinetics (PK) of alisertib (MLN8237) in combination with docetaxel and to identify a recommended dose for the combination. METHODS Adults with metastatic cancer were treated on 21-day cycles with alisertib (10, 20, 30, or 40 mg) twice daily on days 1 to 7 or days 1 to 5 and with docetaxel (75 or 60 mg/m(2) ) on day 1. The primary objectives were to assess the safety and tolerability of the combination and to determine the recommended phase 2 dose (RP2D) for future studies. Secondary objectives included an efficacy assessment and PK analyses of docetaxel and alisertib. RESULTS Forty-one patients participated. Eight dose levels were explored with various doses of alisertib and docetaxel. The dose-limiting toxicities were neutropenic fever, neutropenia without fever, stomatitis, and urinary tract infection. The RP2D of this combination was 20 mg of alisertib twice daily on days 1 to 7 and intravenous docetaxel at 75 mg/m(2) on day 1 in 21-day cycles. Eight of the 28 patients (29%) who were efficacy-evaluable had objective responses. These included 1 complete response in a patient with bladder cancer, 6 partial responses in patients with castration-resistant prostate cancer, and 1 partial response in a patient with angiosarcoma. Concomitant administration of alisertib did not produce any clinically meaningful change in docetaxel PK. CONCLUSIONS Alisertib at 20 mg twice daily on days 1 to 7 with intravenous docetaxel at 75 mg/m(2) on day 1 in a 21-day cycle was well tolerated, and the combination demonstrated antitumor activity. Cancer 2016;122:2524-33. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Julie N Graff
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | | | - Noah M Hahn
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Matthew H Taylor
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Bin Zhang
- Millennium Pharmaceuticals, Inc, Takeda Pharmaceutical Company, Limited, Cambridge, Massachusetts
| | - Xiaofei Zhou
- Millennium Pharmaceuticals, Inc, Takeda Pharmaceutical Company, Limited, Cambridge, Massachusetts
| | - Karthik Venkatakrishnan
- Millennium Pharmaceuticals, Inc, Takeda Pharmaceutical Company, Limited, Cambridge, Massachusetts
| | - E Jane Leonard
- Millennium Pharmaceuticals, Inc, Takeda Pharmaceutical Company, Limited, Cambridge, Massachusetts
| | - John Sarantopoulos
- Institute for Drug Development, Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, Texas
| |
Collapse
|
26
|
Durlacher CT, Li ZL, Chen XW, He ZX, Zhou SF. An update on the pharmacokinetics and pharmacodynamics of alisertib, a selective Aurora kinase A inhibitor. Clin Exp Pharmacol Physiol 2016; 43:585-601. [DOI: 10.1111/1440-1681.12571] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 03/12/2016] [Accepted: 03/15/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Cameron T Durlacher
- Department of Pharmaceutical Sciences; College of Pharmacy; University of South Florida; Tampa FL USA
| | - Zhi-Ling Li
- Department of Pharmacy; Shanghai Children's Hospital; Shanghai Jiao Tong University; Shanghai China
| | - Xiao-Wu Chen
- Department of General Surgery; The First People's Hospital of Shunde Affiliated to Southern Medical University; Shunde Foshan Guangdong
| | - Zhi-Xu He
- Guizhou Provincial Key Laboratory for Regenerative Medicine; Stem Cell and Tissue Engineering Research Centre & Sino-US Joint Laboratory for Medical Sciences; Guizhou Medical University; Guiyang Guizhou China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences; College of Pharmacy; University of South Florida; Tampa FL USA
| |
Collapse
|
27
|
D'Assoro AB, Haddad T, Galanis E. Aurora-A Kinase as a Promising Therapeutic Target in Cancer. Front Oncol 2016; 5:295. [PMID: 26779440 PMCID: PMC4701905 DOI: 10.3389/fonc.2015.00295] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/11/2015] [Indexed: 12/14/2022] Open
Abstract
Mammalian Aurora family of serine/threonine kinases are master regulators of mitotic progression and are frequently overexpressed in human cancers. Among the three members of the Aurora kinase family (Aurora-A, -B, and -C), Aurora-A and Aurora-B are expressed at detectable levels in somatic cells undergoing mitotic cell division. Aberrant Aurora-A kinase activity has been implicated in oncogenic transformation through the development of chromosomal instability and tumor cell heterogeneity. Recent studies also reveal a novel non-mitotic role of Aurora-A activity in promoting tumor progression through activation of epithelial-mesenchymal transition reprograming resulting in the genesis of tumor-initiating cells. Therefore, Aurora-A kinase represents an attractive target for cancer therapeutics, and the development of small molecule inhibitors of Aurora-A oncogenic activity may improve the clinical outcomes of cancer patients. In the present review, we will discuss mitotic and non-mitotic functions of Aurora-A activity in oncogenic transformation and tumor progression. We will also review the current clinical studies, evaluating small molecule inhibitors of Aurora-A activity and their efficacy in the management of cancer patients.
Collapse
Affiliation(s)
- Antonino B D'Assoro
- Department of Medical Oncology, Mayo Clinic College of Medicine, Rochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Tufia Haddad
- Department of Medical Oncology, Mayo Clinic College of Medicine , Rochester, MN , USA
| | - Evanthia Galanis
- Department of Medical Oncology, Mayo Clinic College of Medicine, Rochester, MN, USA; Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
28
|
Niu H, Manfredi M, Ecsedy JA. Scientific Rationale Supporting the Clinical Development Strategy for the Investigational Aurora A Kinase Inhibitor Alisertib in Cancer. Front Oncol 2015; 5:189. [PMID: 26380220 PMCID: PMC4547019 DOI: 10.3389/fonc.2015.00189] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/07/2015] [Indexed: 01/08/2023] Open
Abstract
Alisertib (MLN8237) is a selective small molecule inhibitor of Aurora A kinase that is being developed in multiple cancer indications as a single agent and in combination with other therapies. A significant amount of research has elucidated a role for Aurora A in orchestrating numerous activities of cells transiting through mitosis and has begun to shed light on potential non-mitotic roles for Aurora A as well. These biological insights laid the foundation for multiple clinical trials evaluating the antitumor activity of alisertib in both solid cancers and heme-lymphatic malignancies. Several key facets of Aurora A biology as well as empirical data collected in experimental systems and early clinical trials have directed the development of alisertib toward certain cancer types, including neuroblastoma, small cell lung cancer, neuroendocrine prostate cancer, atypical teratoid/rhabdoid tumors, and breast cancer among others. In addition, these scientific insights provided the rationale for combining alisertib with other therapies, including microtubule perturbing agents, such as taxanes, EGFR inhibitors, hormonal therapies, platinums, and HDAC inhibitors among others. Here, we link the key aspects of the current clinical development of alisertib to the originating scientific rationale and provide an overview of the alisertib clinical experience to date.
Collapse
Affiliation(s)
- Huifeng Niu
- Department of Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA, USA
| | - Mark Manfredi
- Department of Oncology Biology, Takeda Pharmaceuticals International Co, Cambridge, MA, USA
| | - Jeffrey A. Ecsedy
- Department of Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA, USA
| |
Collapse
|
29
|
Li JP, Yang YX, Liu QL, Pan ST, He ZX, Zhang X, Yang T, Chen XW, Wang D, Qiu JX, Zhou SF. The investigational Aurora kinase A inhibitor alisertib (MLN8237) induces cell cycle G2/M arrest, apoptosis, and autophagy via p38 MAPK and Akt/mTOR signaling pathways in human breast cancer cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:1627-52. [PMID: 25834401 PMCID: PMC4365748 DOI: 10.2147/dddt.s75378] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alisertib (ALS) is an investigational potent Aurora A kinase inhibitor currently undergoing clinical trials for the treatment of hematological and non-hematological malignancies. However, its antitumor activity has not been tested in human breast cancer. This study aimed to investigate the effect of ALS on the growth, apoptosis, and autophagy, and the underlying mechanisms in human breast cancer MCF7 and MDA-MB-231 cells. In the current study, we identified that ALS had potent growth-inhibitory, pro-apoptotic, and pro-autophagic effects in MCF7 and MDA-MB-231 cells. ALS arrested the cells in G2/M phase in MCF7 and MDA-MB-231 cells which was accompanied by the downregulation of cyclin-dependent kinase (CDK)1/cell division cycle (CDC) 2, CDK2, and cyclin B1 and upregulation of p21 Waf1/Cip1, p27 Kip1, and p53, suggesting that ALS induces G2/M arrest through modulation of p53/p21/CDC2/cyclin B1 pathways. ALS induced mitochondria-mediated apoptosis in MCF7 and MDA-MB-231 cells; ALS significantly decreased the expression of B-cell lymphoma 2 (Bcl-2), but increased the expression of B-cell lymphoma 2-associated X protein (Bax) and p53-upregulated modulator of apoptosis (PUMA), and increased the expression of cleaved caspases 3 and 9. ALS significantly increased the expression level of membrane-bound microtubule-associated protein 1 light chain 3 (LC3)-II and beclin 1 and induced inhibition of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and p38 mitogen-activated protein kinase (MAPK) pathways in MCF7 and MDA-MB-231 cells as indicated by their altered phosphorylation, contributing to the pro-autophagic activities of ALS. Furthermore, treatment with wortmannin markedly downregulated ALS-induced p38 MAPK activation and LC3 conversion. In addition, knockdown of the p38 MAPK gene by ribonucleic acid interference upregulated Akt activation and resulted in LC3-II accumulation. These findings indicate that ALS promotes cellular apoptosis and autophagy in breast cancer cells via modulation of p38 MAPK/Akt/mTOR pathways. Further studies are warranted to further explore the molecular targets of ALS in the treatment of breast cancer.
Collapse
Affiliation(s)
- Jin-Ping Li
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China ; Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Yin-Xue Yang
- Department of Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Qi-Lun Liu
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Shu-Ting Pan
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China ; Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Zhi-Xu He
- Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Xueji Zhang
- Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, People's Republic of China
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Salt Lake Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Xiao-Wu Chen
- Department of General Surgery, The First People's Hospital of Shunde, Southern Medical University, Shunde, Foshan, Guangdong, People's Republic of China
| | - Dong Wang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People's Republic of China
| | - Jia-Xuan Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA ; Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| |
Collapse
|
30
|
Venkatakrishnan K, Friberg LE, Ouellet D, Mettetal JT, Stein A, Trocóniz IF, Bruno R, Mehrotra N, Gobburu J, Mould DR. Optimizing oncology therapeutics through quantitative translational and clinical pharmacology: challenges and opportunities. Clin Pharmacol Ther 2014; 97:37-54. [PMID: 25670382 DOI: 10.1002/cpt.7] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 10/15/2014] [Indexed: 01/01/2023]
Abstract
Despite advances in biomedical research that have deepened our understanding of cancer hallmarks, resulting in the discovery and development of targeted therapies, the success rates of oncology drug development remain low. Opportunities remain for objective dose selection informed by exposure-response understanding to optimize the benefit-risk balance of novel therapies for cancer patients. This review article discusses the principles and applications of modeling and simulation approaches across the lifecycle of development of oncology therapeutics. Illustrative examples are used to convey the value gained from integration of quantitative clinical pharmacology strategies from the preclinical-translational phase through confirmatory clinical evaluation of efficacy and safety.
Collapse
Affiliation(s)
- K Venkatakrishnan
- Clinical Pharmacology, Takeda Pharmaceuticals International Co., Cambridge, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|