101
|
Konstantinopoulos PA, Ceccaldi R, Shapiro GI, D'Andrea AD. Homologous Recombination Deficiency: Exploiting the Fundamental Vulnerability of Ovarian Cancer. Cancer Discov 2015. [PMID: 26463832 DOI: 10.1158/2159-8290.cd-15-0714] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
UNLABELLED Approximately 50% of epithelial ovarian cancers (EOC) exhibit defective DNA repair via homologous recombination (HR) due to genetic and epigenetic alterations of HR pathway genes. Defective HR is an important therapeutic target in EOC as exemplified by the efficacy of platinum analogues in this disease, as well as the advent of PARP inhibitors, which exhibit synthetic lethality when applied to HR-deficient cells. Here, we describe the genotypic and phenotypic characteristics of HR-deficient EOCs, discuss current and emerging approaches for targeting these tumors, and present challenges associated with these approaches, focusing on development and overcoming resistance. SIGNIFICANCE Defective DNA repair via HR is a pivotal vulnerability of EOC, particularly of the high-grade serous histologic subtype. Targeting defective HR offers the unique opportunity of exploiting molecular differences between tumor and normal cells, thereby inducing cancer-specific synthetic lethality; the promise and challenges of these approaches in ovarian cancer are discussed in this review.
Collapse
Affiliation(s)
- Panagiotis A Konstantinopoulos
- Department of Medical Oncology, Medical Gynecologic Oncology Program, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Raphael Ceccaldi
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Geoffrey I Shapiro
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medical Oncology, Early Drug Development Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Alan D D'Andrea
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
102
|
Konstantinopoulos PA, Ceccaldi R, Shapiro GI, D'Andrea AD. Homologous Recombination Deficiency: Exploiting the Fundamental Vulnerability of Ovarian Cancer. Cancer Discov 2015; 5:1137-54. [PMID: 26463832 DOI: 10.1158/2159-8290.cd-15-0714] [Citation(s) in RCA: 669] [Impact Index Per Article: 66.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/11/2015] [Indexed: 12/14/2022]
Abstract
UNLABELLED Approximately 50% of epithelial ovarian cancers (EOC) exhibit defective DNA repair via homologous recombination (HR) due to genetic and epigenetic alterations of HR pathway genes. Defective HR is an important therapeutic target in EOC as exemplified by the efficacy of platinum analogues in this disease, as well as the advent of PARP inhibitors, which exhibit synthetic lethality when applied to HR-deficient cells. Here, we describe the genotypic and phenotypic characteristics of HR-deficient EOCs, discuss current and emerging approaches for targeting these tumors, and present challenges associated with these approaches, focusing on development and overcoming resistance. SIGNIFICANCE Defective DNA repair via HR is a pivotal vulnerability of EOC, particularly of the high-grade serous histologic subtype. Targeting defective HR offers the unique opportunity of exploiting molecular differences between tumor and normal cells, thereby inducing cancer-specific synthetic lethality; the promise and challenges of these approaches in ovarian cancer are discussed in this review.
Collapse
Affiliation(s)
- Panagiotis A Konstantinopoulos
- Department of Medical Oncology, Medical Gynecologic Oncology Program, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Raphael Ceccaldi
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Geoffrey I Shapiro
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Medical Oncology, Early Drug Development Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Alan D D'Andrea
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
103
|
Raja AM, Xu S, Zhuo S, Tai DCS, Sun W, So PTC, Welsch RE, Chen CS, Yu H. Differential remodeling of extracellular matrices by breast cancer initiating cells. JOURNAL OF BIOPHOTONICS 2015; 8:804-15. [PMID: 25597396 PMCID: PMC4761427 DOI: 10.1002/jbio.201400079] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 11/15/2014] [Accepted: 11/26/2014] [Indexed: 06/04/2023]
Abstract
Cancer initiating cells (CICs) have been the focus of recent anti-cancer therapies, exhibiting strong invasion capability via potentially enhanced ability to remodel extracellular matrices (ECM). We have identified CICs in a human breast cancer cell line, MX-1, and developed a xenograft model in SCID mice. We investigated the CICs' matrix-remodeling effects using Second Harmonic Generation (SHG) microscopy to identify potential phenotypic signatures of the CIC-rich tumors. The isolated CICs exhibit higher proliferation, drug efflux and drug resistant properties in vitro; were more tumorigenic than non-CICs, resulting in more and larger tumors in the xenograft model. The CIC-rich tumors have less collagen in the tumor interior than in the CIC-poor tumors supporting the idea that the CICs can remodel the collagen more effectively. The collagen fibers were preferentially aligned perpendicular to the CIC-rich tumor boundary while parallel to the CIC-poor tumor boundary suggesting more invasive behavior of the CIC-rich tumors. These findings would provide potential translational values in quantifying and monitoring CIC-rich tumors in future anti-cancer therapies. CIC-rich tumors remodel the collagen matrix more than CIC-poor tumors.
Collapse
Affiliation(s)
- Anju M Raja
- Biomedical Engineering Division, Department of Electronic and Computer Engineering, Ngee Ann Polytechnic, 535 Clementi Road, Singapore, 599489
- Institute of Bioengineering and Nanotechnology, A*STAR, Singapore, 138669
- NUS Graduate Programme in Bioengineering, NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, 117597
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shuoyu Xu
- Institute of Bioengineering and Nanotechnology, A*STAR, Singapore, 138669
- BioSystems and Micromechanics, Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, #04-13/14 Enterprise Wing, Singapore, 138602
| | - Shuangmu Zhuo
- BioSystems and Micromechanics, Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, #04-13/14 Enterprise Wing, Singapore, 138602
- Institute of Laser and Optoelectronics Technology, Fujian Normal University, Fuzhou, 350007, China
| | - Dean C S Tai
- Institute of Bioengineering and Nanotechnology, A*STAR, Singapore, 138669
| | - Wanxin Sun
- Institute of Bioengineering and Nanotechnology, A*STAR, Singapore, 138669
| | - Peter T C So
- BioSystems and Micromechanics, Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, #04-13/14 Enterprise Wing, Singapore, 138602
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Roy E Welsch
- Sloan School of Management, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Chien-Shing Chen
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, 28 Medical Drive, Singapore, 117456
- School of Medicine, Division of Hematology and Oncology, Loma Linda University, CA, 92354, USA
| | - Hanry Yu
- Institute of Bioengineering and Nanotechnology, A*STAR, Singapore, 138669.
- NUS Graduate Programme in Bioengineering, NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, 117597.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
- BioSystems and Micromechanics, Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, #04-13/14 Enterprise Wing, Singapore, 138602.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, Singapore, 117597.
- Mechanobiology Institute, National University of Singapore, T-Lab, #05-01, 5A Engineering Drive 1, Singapore, 117411.
| |
Collapse
|
104
|
Hendrikx JJMA, Lagas JS, Song JY, Rosing H, Schellens JHM, Beijnen JH, Rottenberg S, Schinkel AH. Ritonavir inhibits intratumoral docetaxel metabolism and enhances docetaxel antitumor activity in an immunocompetent mouse breast cancer model. Int J Cancer 2015; 138:758-69. [PMID: 26297509 DOI: 10.1002/ijc.29812] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 08/03/2015] [Indexed: 12/27/2022]
Abstract
Docetaxel (Taxotere(®)) is currently used intravenously as an anticancer agent and is primarily metabolized by Cytochrome P450 3A (CYP3A). The HIV protease inhibitor ritonavir, a strong CYP3A4 inhibitor, decreased first-pass metabolism of orally administered docetaxel. Anticancer effects of ritonavir itself have also been described. We here aimed to test whether ritonavir co-administration could decrease intratumoral metabolism of intravenously administered docetaxel and thus increase the antitumor activity of docetaxel in an orthotopic, immunocompetent mouse model for breast cancer. Spontaneously arising K14cre;Brca1(F/F) ;p53(F/F) mouse mammary tumors were orthotopically implanted in syngeneic mice lacking Cyp3a (Cyp3a(-/-)) to limit ritonavir effects on systemic docetaxel clearance. Over 3 weeks, docetaxel (20 mg/kg) was administered intravenously once weekly, with or without ritonavir (12.5 mg/kg) administered orally for 5 days per week. Untreated mice were used as control for tumor growth. Ritonavir treatment alone did not significantly affect the median time of survival (14 vs. 10 days). Median time of survival in docetaxel-treated mice was 54 days. Ritonavir co-treatment significantly increased this to 66 days, and substantially reduced relative average tumor size, without altering tumor histology. Concentrations of the major docetaxel metabolite M2 in tumor tissue were reduced by ritonavir co-administration, whereas tumor RNA expression of Cyp3a was unaltered. In this breast cancer model, we observed no direct antitumor effect of ritonavir alone, but we found enhanced efficacy of docetaxel treatment when combined with ritonavir. Our data, therefore, suggest that decreased docetaxel metabolism inside the tumor as a result of Cyp3a inhibition contributes to increased antitumor activity.
Collapse
Affiliation(s)
- Jeroen J M A Hendrikx
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jurjen S Lagas
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ji-Ying Song
- Department of Experimental Animal Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jan H M Schellens
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Sven Rottenberg
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alfred H Schinkel
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
105
|
Greenow KR, Smalley MJ. Overview of Genetically Engineered Mouse Models of Breast Cancer Used in Translational Biology and Drug Development. CURRENT PROTOCOLS IN PHARMACOLOGY 2015; 70:14.36.1-14.36.14. [PMID: 26331886 DOI: 10.1002/0471141755.ph1436s70] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Breast cancer is a heterogeneous condition with no single standard of treatment and no definitive method for determining whether a tumor will respond to therapy. The development of murine models that faithfully mimic specific human breast cancer subtypes is critical for the development of patient-specific treatments. While the artificial nature of traditional in vivo xenograft models used to characterize novel anticancer treatments has limited clinical predictive value, the development of genetically engineered mouse models (GEMMs) makes it possible to study the therapeutic responses in an intact microenvironment. GEMMs have proven to be an experimentally tractable platform for evaluating the efficacy of novel therapeutic combinations and for defining the mechanisms of acquired resistance. Described in this overview are several of the more popular breast cancer GEMMs, including details on their value in elucidating the molecular mechanisms of this disorder.
Collapse
Affiliation(s)
- Kirsty R Greenow
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, United Kingdom
- Current Address: Propath UK Ltd., Hereford, United Kingdom
| | - Matthew J Smalley
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, United Kingdom
- Corresponding Author:
| |
Collapse
|
106
|
Blatter S, Rottenberg S. Minimal residual disease in cancer therapy--Small things make all the difference. Drug Resist Updat 2015; 21-22:1-10. [PMID: 26307504 DOI: 10.1016/j.drup.2015.08.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 07/25/2015] [Accepted: 08/05/2015] [Indexed: 01/16/2023]
Abstract
Minimal residual disease (MRD) is a major hurdle in the eradication of malignant tumors. Despite the high sensitivity of various cancers to treatment, some residual cancer cells persist and lead to tumor recurrence and treatment failure. Obvious reasons for residual disease include mechanisms of secondary therapy resistance, such as the presence of mutant cells that are insensitive to the drugs, or the presence of cells that become drug resistant due to activation of survival pathways. In addition to such unambiguous resistance modalities, several patients with relapsing tumors do not show refractory disease and respond again when the initial therapy is repeated. These cases cannot be explained by the selection of mutant tumor cells, and the precise mechanisms underlying this clinical drug resistance are ill-defined. In the current review, we put special emphasis on cell-intrinsic and -extrinsic mechanisms that may explain mechanisms of MRD that are independent of secondary therapy resistance. In particular, we show that studying genetically engineered mouse models (GEMMs), which highly resemble the disease in humans, provides a complementary approach to understand MRD. In these animal models, specific mechanisms of secondary resistance can be excluded by targeted genetic modifications. This allows a clear distinction between the selection of cells with stable secondary resistance and mechanisms that result in the survival of residual cells but do not provoke secondary drug resistance. Mechanisms that may explain the latter feature include special biochemical defense properties of cancer stem cells, metabolic peculiarities such as the dependence on autophagy, drug-tolerant persisting cells, intratumoral heterogeneity, secreted factors from the microenvironment, tumor vascularization patterns and immunosurveillance-related factors. We propose in the current review that a common feature of these various mechanisms is cancer cell dormancy. Therefore, dormant cancer cells appear to be an important target in the attempt to eradicate residual cancer cells, and eventually cure patients who repeatedly respond to anticancer therapy but lack complete tumor eradication.
Collapse
Affiliation(s)
- Sohvi Blatter
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Laengassstrasse 122, 3012 Bern, Switzerland
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Laengassstrasse 122, 3012 Bern, Switzerland; Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands.
| |
Collapse
|
107
|
Wahba HA, El-Hadaad HA. Current approaches in treatment of triple-negative breast cancer. Cancer Biol Med 2015; 12:106-16. [PMID: 26175926 PMCID: PMC4493381 DOI: 10.7497/j.issn.2095-3941.2015.0030] [Citation(s) in RCA: 252] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 06/01/2015] [Indexed: 12/30/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is diagnosed more frequently in younger and premenopausal women and is highly prevalent in African American women. TNBC is a term derived from tumors that are characterized by the absence of ER, PgR, and HER2. So patients with TNBC do not benefit from hormonal or trastuzumab-based therapies. TNBCs are biologically aggressive, although some reports suggest that they respond to chemotherapy better than other types of breast cancer, prognosis remains poor. This is due to: shortened disease-free interval in the adjuvant and neoadjuvant setting and a more aggressive course in the metastatic setting.
Collapse
Affiliation(s)
- Hanan Ahmed Wahba
- Clinical Oncology and Nuclear Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Hend Ahmed El-Hadaad
- Clinical Oncology and Nuclear Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
108
|
Maia ARR, de Man J, Boon U, Janssen A, Song JY, Omerzu M, Sterrenburg JG, Prinsen MBW, Willemsen-Seegers N, de Roos JADM, van Doornmalen AM, Uitdehaag JCM, Kops GJPL, Jonkers J, Buijsman RC, Zaman GJR, Medema RH. Inhibition of the spindle assembly checkpoint kinase TTK enhances the efficacy of docetaxel in a triple-negative breast cancer model. Ann Oncol 2015; 26:2180-92. [PMID: 26153498 DOI: 10.1093/annonc/mdv293] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 06/29/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Triple-negative breast cancers (TNBC) are considered the most aggressive type of breast cancer, for which no targeted therapy exists at the moment. These tumors are characterized by having a high degree of chromosome instability and often overexpress the spindle assembly checkpoint kinase TTK. To explore the potential of TTK inhibition as a targeted therapy in TNBC, we developed a highly potent and selective small molecule inhibitor of TTK, NTRC 0066-0. RESULTS AND CONCLUSIONS The compound is characterized by long residence time on the target and inhibits the proliferation of a wide variety of human cancer cell lines with potency in the same range as marketed cytotoxic agents. In cell lines and in mice, NTRC 0066-0 inhibits the phosphorylation of a TTK substrate and induces chromosome missegregation. NTRC 0066-0 inhibits tumor growth in MDA-MB-231 xenografts as a single agent after oral application. To address the effect of the inhibitor in breast cancer, we used a well-defined mouse model that spontaneously develops breast tumors that share key morphologic and molecular features with human TNBC. Our studies show that combination of NTRC 0066-0 with a therapeutic dose of docetaxel resulted in doubling of mouse survival and extended tumor remission, without toxicity. Furthermore, we observed that treatment efficacy is only achieved upon co-administration of the two compounds, which suggests a synergistic in vivo effect. Therefore, we propose TTK inhibition as a novel therapeutic target for neoadjuvant therapy in TNBC.
Collapse
Affiliation(s)
- A R R Maia
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam
| | - J de Man
- Netherlands Translational Research Center B.V., Oss
| | - U Boon
- Division of Molecular Pathology and Cancer Genomics Centre
| | - A Janssen
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam
| | - J-Y Song
- Division of Experimental Animal Pathology, The Netherlands Cancer Institute, Amsterdam
| | | | | | | | | | | | | | | | - G J P L Kops
- Department of Medical Oncology Department of Cancer Genomics Netherlands, UMC Utrecht, Utrecht, The Netherlands
| | - J Jonkers
- Division of Molecular Pathology and Cancer Genomics Centre Department of Cancer Genomics Netherlands, UMC Utrecht, Utrecht, The Netherlands
| | - R C Buijsman
- Netherlands Translational Research Center B.V., Oss
| | - G J R Zaman
- Netherlands Translational Research Center B.V., Oss
| | - R H Medema
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam Department of Cancer Genomics Netherlands, UMC Utrecht, Utrecht, The Netherlands
| |
Collapse
|
109
|
Kang X, Chen K, Li Y, Li J, D'Amico TA, Chen X. Personalized targeted therapy for esophageal squamous cell carcinoma. World J Gastroenterol 2015; 21:7648-7658. [PMID: 26167067 PMCID: PMC4491954 DOI: 10.3748/wjg.v21.i25.7648] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/19/2015] [Accepted: 04/28/2015] [Indexed: 02/06/2023] Open
Abstract
Esophageal squamous cell carcinoma continues to heavily burden clinicians worldwide. Researchers have discovered the genomic landscape of esophageal squamous cell carcinoma, which holds promise for an era of personalized oncology care. One of the most pressing problems facing this issue is to improve the understanding of the newly available genomic data, and identify the driver-gene mutations, pathways, and networks. The emergence of a legion of novel targeted agents has generated much hope and hype regarding more potent treatment regimens, but the accuracy of drug selection is still arguable. Other problems, such as cancer heterogeneity, drug resistance, exceptional responders, and side effects, have to be surmounted. Evolving topics in personalized oncology, such as interpretation of genomics data, issues in targeted therapy, research approaches for targeted therapy, and future perspectives, will be discussed in this editorial.
Collapse
|
110
|
Selective resistance to the PARP inhibitor olaparib in a mouse model for BRCA1-deficient metaplastic breast cancer. Proc Natl Acad Sci U S A 2015; 112:8409-14. [PMID: 26100884 DOI: 10.1073/pnas.1500223112] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Metaplastic breast carcinoma (MBC) is a rare histological breast cancer subtype characterized by mesenchymal elements and poor clinical outcome. A large fraction of MBCs harbor defects in breast cancer 1 (BRCA1). As BRCA1 deficiency sensitizes tumors to DNA cross-linking agents and poly(ADP-ribose) polymerase (PARP) inhibitors, we sought to investigate the response of BRCA1-deficient MBCs to the PARP inhibitor olaparib. To this end, we established a genetically engineered mouse model (GEMM) for BRCA1-deficient MBC by introducing the MET proto-oncogene into a BRCA1-associated breast cancer model, using our novel female GEMM ES cell (ESC) pipeline. In contrast to carcinomas, BRCA1-deficient mouse carcinosarcomas resembling MBC show intrinsic resistance to olaparib caused by increased P-glycoprotein (Pgp) drug efflux transporter expression. Indeed, resistance could be circumvented by using another PARP inhibitor, AZD2461, which is a poor Pgp substrate. These preclinical findings suggest that patients with BRCA1-associated MBC may show poor response to olaparib and illustrate the value of GEMM-ESC models of human cancer for evaluation of novel therapeutics.
Collapse
|
111
|
van Asten JJA, Vettukattil R, Buckle T, Rottenberg S, van Leeuwen F, Bathen TF, Heerschap A. Increased levels of choline metabolites are an early marker of docetaxel treatment response in BRCA1-mutated mouse mammary tumors: an assessment by ex vivo proton magnetic resonance spectroscopy. J Transl Med 2015; 13:114. [PMID: 25890200 PMCID: PMC4404119 DOI: 10.1186/s12967-015-0458-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 03/10/2015] [Indexed: 01/05/2023] Open
Abstract
Background Docetaxel is one of the most frequently used drugs to treat breast cancer. However, resistance or incomplete response to docetaxel is a major challenge. The aim of this study was to utilize MR metabolomics to identify potential biomarkers of docetaxel resistance in a mouse model for BRCA1-mutated breast cancer. Methodology High resolution magic angle spinning (HRMAS) 1H MR spectroscopy was performed on tissue samples obtained from docetaxel-sensitive or -resistant BRCA1-mutated mammary tumors in mice. Measurements were performed on samples obtained before treatment and at 1-2, 3-5 and 6-7 days after a 25 mg/kg dose of docetaxel. The MR spectra were analyzed by multivariate analysis, followed by analysis of the signals of individual compounds by peak fitting and integration with normalization to the integral of the creatine signal and of all signals between 2.9 and 3.6 ppm. Results The HRMAS spectra revealed significant metabolic differences between sensitive and resistant tissue samples. In particular choline metabolites were higher in resistant tumors by more than 50% with respect to creatine and by more than 30% with respect to all signals between 2.9 and 3.6 ppm. Shortly after treatment (1-2 days) the normalized choline metabolite levels were significantly increased by more than 30% in the sensitive group coinciding with the time of highest apoptotic activity induced by docetaxel. Thereafter, choline metabolites in these tumors returned towards pre-treatment levels. No change in choline compounds was observed in the resistant tumors over the whole time of investigation. Conclusions Relative tissue concentrations of choline compounds are higher in docetaxel resistant than in sensitive BRCA1-mutated mouse mammary tumors, but in the first days after docetaxel treatment only in the sensitive tumors an increase of these compounds is observed. Thus both pre- and post-treatment tissue levels of choline compounds have potential to predict response to docetaxel treatment.
Collapse
Affiliation(s)
- Jack J A van Asten
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Riyas Vettukattil
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands. .,Department of Molecular Pathology, The Netherlands Cancer Institute- Antoni van Leeuwenhoek hospital (NKI-AvL), Amsterdam, The Netherlands.
| | - Sven Rottenberg
- Department of Molecular Pathology, The Netherlands Cancer Institute- Antoni van Leeuwenhoek hospital (NKI-AvL), Amsterdam, The Netherlands.
| | - Fijs van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Tone F Bathen
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
112
|
Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, Johns AL, Miller D, Nones K, Quek K, Quinn MCJ, Robertson AJ, Fadlullah MZH, Bruxner TJC, Christ AN, Harliwong I, Idrisoglu S, Manning S, Nourse C, Nourbakhsh E, Wani S, Wilson PJ, Markham E, Cloonan N, Anderson MJ, Fink JL, Holmes O, Kazakoff SH, Leonard C, Newell F, Poudel B, Song S, Taylor D, Waddell N, Wood S, Xu Q, Wu J, Pinese M, Cowley MJ, Lee HC, Jones MD, Nagrial AM, Humphris J, Chantrill LA, Chin V, Steinmann AM, Mawson A, Humphrey ES, Colvin EK, Chou A, Scarlett CJ, Pinho AV, Giry-Laterriere M, Rooman I, Samra JS, Kench JG, Pettitt JA, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N, Jamieson NB, Graham JS, Niclou SP, Bjerkvig R, Grützmann R, Aust D, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Wolfgang CL, Morgan RA, Lawlor RT, Corbo V, Bassi C, Falconi M, Zamboni G, Tortora G, Tempero MA, Gill AJ, Eshleman JR, Pilarsky C, Scarpa A, Musgrove EA, Pearson JV, Biankin AV, Grimmond SM. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015; 518:495-501. [PMID: 25719666 PMCID: PMC4523082 DOI: 10.1038/nature14169] [Citation(s) in RCA: 1963] [Impact Index Per Article: 196.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 12/18/2014] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer remains one of the most lethal of malignancies and a major health burden. We performed whole-genome sequencing and copy number variation (CNV) analysis of 100 pancreatic ductal adenocarcinomas (PDACs). Chromosomal rearrangements leading to gene disruption were prevalent, affecting genes known to be important in pancreatic cancer (TP53, SMAD4, CDKN2A, ARID1A and ROBO2) and new candidate drivers of pancreatic carcinogenesis (KDM6A and PREX2). Patterns of structural variation (variation in chromosomal structure) classified PDACs into 4 subtypes with potential clinical utility: the subtypes were termed stable, locally rearranged, scattered and unstable. A significant proportion harboured focal amplifications, many of which contained druggable oncogenes (ERBB2, MET, FGFR1, CDK6, PIK3R3 and PIK3CA), but at low individual patient prevalence. Genomic instability co-segregated with inactivation of DNA maintenance genes (BRCA1, BRCA2 or PALB2) and a mutational signature of DNA damage repair deficiency. Of 8 patients who received platinum therapy, 4 of 5 individuals with these measures of defective DNA maintenance responded.
Collapse
Affiliation(s)
- Nicola Waddell
- 1] Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia [2] QIMR Berghofer Medical Research Institute, Herston Road, Brisbane 4006, Australia
| | - Marina Pajic
- 1] The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia [2] St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, New South Wales 2010, Australia
| | - Ann-Marie Patch
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - David K Chang
- 1] The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia [2] Department of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, New South Wales 2200, Australia [3] South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Liverpool, New South Wales 2170, Australia [4] Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Karin S Kassahn
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Peter Bailey
- 1] Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia [2] Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Amber L Johns
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - David Miller
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Katia Nones
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Kelly Quek
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Michael C J Quinn
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Alan J Robertson
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Muhammad Z H Fadlullah
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Tim J C Bruxner
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Angelika N Christ
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Ivon Harliwong
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Senel Idrisoglu
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Suzanne Manning
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Craig Nourse
- 1] Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia [2] Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Ehsan Nourbakhsh
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Shivangi Wani
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Peter J Wilson
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Emma Markham
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Nicole Cloonan
- 1] Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia [2] QIMR Berghofer Medical Research Institute, Herston Road, Brisbane 4006, Australia
| | - Matthew J Anderson
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - J Lynn Fink
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Oliver Holmes
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Stephen H Kazakoff
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Conrad Leonard
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Felicity Newell
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Barsha Poudel
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Sarah Song
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Darrin Taylor
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Nick Waddell
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Scott Wood
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Qinying Xu
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Jianmin Wu
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Mark Pinese
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Mark J Cowley
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Hong C Lee
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Marc D Jones
- 1] The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia [2] Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Adnan M Nagrial
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Jeremy Humphris
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Lorraine A Chantrill
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Venessa Chin
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Angela M Steinmann
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Amanda Mawson
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Emily S Humphrey
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Emily K Colvin
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Angela Chou
- 1] The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia [2] Department of Anatomical Pathology, St Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | - Christopher J Scarlett
- 1] The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia [2] School of Environmental &Life Sciences, University of Newcastle, Ourimbah, New South Wales 2258, Australia
| | - Andreia V Pinho
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Marc Giry-Laterriere
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Ilse Rooman
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Jaswinder S Samra
- 1] Department of Surgery, Royal North Shore Hospital, St Leonards, Sydney, New South Wales 2065, Australia [2] University of Sydney, Sydney, New South Wales 2006, Australia
| | - James G Kench
- 1] The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia [2] University of Sydney, Sydney, New South Wales 2006, Australia [3] Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales 2050, Australia
| | - Jessica A Pettitt
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Neil D Merrett
- 1] Department of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, New South Wales 2200, Australia [2] School of Medicine, University of Western Sydney, Penrith, New South Wales 2175, Australia
| | - Christopher Toon
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Krishna Epari
- Department of Surgery, Fremantle Hospital, Alma Street, Fremantle, Western Australia 6160, Australia
| | - Nam Q Nguyen
- Department of Gastroenterology, Royal Adelaide Hospital, North Terrace, Adelaide, South Australia 5000, Australia
| | - Andrew Barbour
- Department of Surgery, Princess Alexandra Hospital, Ipswich Rd, Woollongabba, Queensland 4102, Australia
| | - Nikolajs Zeps
- 1] School of Surgery M507, University of Western Australia, 35 Stirling Highway, Nedlands 6009, Australia [2] St John of God Pathology, 12 Salvado Rd, Subiaco, Western Australia 6008, Australia [3] Bendat Family Comprehensive Cancer Centre, St John of God Subiaco Hospital, Subiaco, Western Australia 6008, Australia
| | - Nigel B Jamieson
- 1] Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK [2] Academic Unit of Surgery, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow G4 OSF, UK [3] West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow G31 2ER, UK
| | - Janet S Graham
- 1] Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK [2] Department of Medical Oncology, Beatson West of Scotland Cancer Centre, 1053 Great Western Road, Glasgow G12 0YN, UK
| | - Simone P Niclou
- Norlux Neuro-Oncology Laboratory, CRP-Santé Luxembourg, 84 Val Fleuri, L-1526, Luxembourg
| | - Rolf Bjerkvig
- Norlux Neuro-Oncology, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, N-5019 Bergen, Norway
| | - Robert Grützmann
- Departments of Surgery and Pathology, TU Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Daniela Aust
- Departments of Surgery and Pathology, TU Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Ralph H Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | - Anirban Maitra
- Departments of Pathology and Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston Texas 77030, USA
| | - Christine A Iacobuzio-Donahue
- The David M. Rubenstein Pancreatic Cancer Research Center and Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Christopher L Wolfgang
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | - Richard A Morgan
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | - Rita T Lawlor
- 1] ARC-NET Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona 37134, Italy [2] Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy
| | - Vincenzo Corbo
- ARC-NET Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona 37134, Italy
| | - Claudio Bassi
- Department of Surgery and Oncology, Pancreas Institute, University and Hospital Trust of Verona, Verona 37134, Italy
| | - Massimo Falconi
- 1] Department of Surgery and Oncology, Pancreas Institute, University and Hospital Trust of Verona, Verona 37134, Italy [2] Departments of Surgery and Pathology, Ospedale Sacro Cuore Don Calabria Negrar, Verona 37024, Italy
| | - Giuseppe Zamboni
- 1] Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy [2] Departments of Surgery and Pathology, Ospedale Sacro Cuore Don Calabria Negrar, Verona 37024, Italy
| | - Giampaolo Tortora
- Department of Oncology, University and Hospital Trust of Verona, Verona 37134, Italy
| | - Margaret A Tempero
- Division of Hematology and Oncology, University of California, San Francisco, California 94122, USA
| | - Anthony J Gill
- 1] The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia [2] University of Sydney, Sydney, New South Wales 2006, Australia
| | - James R Eshleman
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | - Christian Pilarsky
- Departments of Surgery and Pathology, TU Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Aldo Scarpa
- 1] ARC-NET Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona 37134, Italy [2] Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy
| | - Elizabeth A Musgrove
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - John V Pearson
- 1] Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia [2] QIMR Berghofer Medical Research Institute, Herston Road, Brisbane 4006, Australia
| | - Andrew V Biankin
- 1] The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, University of New South Wales, 384 Victoria St, Darlinghurst, Sydney, New South Wales 2010, Australia [2] Department of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, New South Wales 2200, Australia [3] South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Liverpool, New South Wales 2170, Australia [4] Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Sean M Grimmond
- 1] Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia [2] Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| |
Collapse
|
113
|
Jannasch K, Wegwitz F, Lenfert E, Maenz C, Deppert W, Alves F. Chemotherapy of WAP-T mouse mammary carcinomas aggravates tumor phenotype and enhances tumor cell dissemination. Int J Cancer 2014; 137:25-36. [PMID: 25449528 DOI: 10.1002/ijc.29369] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 11/13/2014] [Indexed: 12/27/2022]
Abstract
In this study, the effects of the standard chemotherapy, cyclophosphamide/adriamycin/5-fluorouracil (CAF) on tumor growth, dissemination and recurrence after orthotopic implantation of murine G-2 cells were analyzed in the syngeneic immunocompetent whey acidic protein-T mouse model (Wegwitz et al., PLoS One 2010; 5:e12103; Schulze-Garg et al., Oncogene 2000; 19:1028-37). Single-dose CAF treatment reduced tumor size significantly, but was not able to eradicate all tumor cells, as recurrent tumor growth was observed 4 weeks after CAF treatment. Nine days after CAF treatment, residual tumors showed features of regressive alterations and were composed of mesenchymal-like tumor cells, infiltrating immune cells and some tumor-associated fibroblasts with an intense deposition of collagen. Recurrent tumors were characterized by coagulative necrosis and less tumor cell differentiation compared with untreated tumors, suggesting a more aggressive tumor phenotype. In support, tumor cell dissemination was strongly enhanced in mice that had developed recurrent tumors in comparison with untreated controls, although only few disseminated tumor cells could be detected in various organs 9 days after CAF application. In vitro experiments revealed that CAF treatment of G-2 cells eliminates the vast majority of epithelial tumor cells, whereas tumor cells with a mesenchymal phenotype survive. These results together with the in vivo findings suggest that tumor cells that underwent epithelial-mesenchymal transition and/or exhibit stem-cell-like properties are difficult to eliminate using one round of CAF chemotherapy. The model system described here provides a valuable tool for the characterization of the effects of chemotherapeutic regimens on recurrent tumor growth and on tumor cell dissemination, thereby enabling the development and preclinical evaluation of novel therapeutic strategies to target mammary carcinomas.
Collapse
Affiliation(s)
- Katharina Jannasch
- Department of Hematology and Medical Oncology, University Medical Center, 37075, Goettingen, Germany
| | | | | | | | | | | |
Collapse
|
114
|
Jaspers JE, Sol W, Kersbergen A, Schlicker A, Guyader C, Xu G, Wessels L, Borst P, Jonkers J, Rottenberg S. BRCA2-deficient sarcomatoid mammary tumors exhibit multidrug resistance. Cancer Res 2014; 75:732-41. [PMID: 25511378 DOI: 10.1158/0008-5472.can-14-0839] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pan- or multidrug resistance is a central problem in clinical oncology. Here, we use a genetically engineered mouse model of BRCA2-associated hereditary breast cancer to study drug resistance to several types of chemotherapy and PARP inhibition. We found that multidrug resistance was strongly associated with an EMT-like sarcomatoid phenotype and high expression of the Abcb1b gene, which encodes the drug efflux transporter P-glycoprotein. Inhibition of P-glycoprotein could partly resensitize sarcomatoid tumors to the PARP inhibitor olaparib, docetaxel, and doxorubicin. We propose that multidrug resistance is a multifactorial process and that mouse models are useful to unravel this.
Collapse
Affiliation(s)
- Janneke E Jaspers
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands. Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Wendy Sol
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ariena Kersbergen
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Andreas Schlicker
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Charlotte Guyader
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Guotai Xu
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lodewyk Wessels
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Piet Borst
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sven Rottenberg
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands. Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| |
Collapse
|
115
|
Schouten PC, Marmé F, Aulmann S, Sinn HP, van Essen HF, Ylstra B, Hauptmann M, Schneeweiss A, Linn SC. Breast cancers with a BRCA1-like DNA copy number profile recur less often than expected after high-dose alkylating chemotherapy. Clin Cancer Res 2014; 21:763-70. [PMID: 25480832 DOI: 10.1158/1078-0432.ccr-14-1894] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Breast cancers in carriers of inactivating mutations of the BRCA1 gene carry a specific DNA copy-number signature ("BRCA1-like"). This signature is shared with cancers that inactivate BRCA1 through other mechanisms. Because BRCA1 is important in repair of DNA double-strand breaks through error-free homologous recombination, patients with a BRCA1-like tumor may benefit from high-dose alkylating (HD) chemotherapy, which induces DNA double-strand breaks. EXPERIMENTAL DESIGN We investigated a single institution cohort of high-risk patients that received tandem HD chemotherapy schedule comprising ifosfamide, epirubicin, and carboplatin or conventional chemotherapy. We classified copy-number profiles to be BRCA1-like or non-BRCA1-like and analyzed clinical associations and performed survival analysis with a treatment by biomarker interaction design. RESULTS BRCA1-like status associated with high-grade and triple-negative breast cancers. BRCA1-like cases benefitted from the HD compared with a conventional regimen on disease-free survival (DFS): [hazard ratio (HR), 0.05; 95% confidence interval (CI), 0.01-0.38; P = 0.003]; distant DFS (DDFS): (HR, 0.06; 95% CI, 0.01-0.43; P = 0.01); and overall survival (OS; HR, 0.15; 95% CI, 0.03-0.83; P = 0.03) after correction for prognostic factors. No such benefit was observed in the non-BRCA1-like cases on DFS (HR, 0.74; 95% CI, 0.38-1.46; P = 0.39), DDFS (HR, 0.79; 95% CI, 0.41-1.52; P = 0.47), and OS (HR, 0.93; 95% CI, 0.52-1.64; P = 0.79). The P values for interaction were 0.01 (DFS), 0.01 (DDFS), and 0.045 (OS). CONCLUSIONS BRCA1-like tumors recurred significantly less often after HD than conventional chemotherapy. BRCA1-like copy-number profile classification may be a predictive marker for HD alkylating chemotherapy.
Collapse
Affiliation(s)
- Philip C Schouten
- Department of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Frederik Marmé
- Universitäts-Frauenklinik, University of Heidelberg, Heidelberg, Germany. National Center for Tumour Diseases, Heidelberg, Germany
| | - Sebastian Aulmann
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Hans-Peter Sinn
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Hendrik F van Essen
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Michael Hauptmann
- Department of Epidemiology and Biostatistics, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Andreas Schneeweiss
- Universitäts-Frauenklinik, University of Heidelberg, Heidelberg, Germany. National Center for Tumour Diseases, Heidelberg, Germany
| | - Sabine C Linn
- Department of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands. Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands. Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
116
|
O'Shaughnessy J, Schwartzberg L, Danso MA, Miller KD, Rugo HS, Neubauer M, Robert N, Hellerstedt B, Saleh M, Richards P, Specht JM, Yardley DA, Carlson RW, Finn RS, Charpentier E, Garcia-Ribas I, Winer EP. Phase III study of iniparib plus gemcitabine and carboplatin versus gemcitabine and carboplatin in patients with metastatic triple-negative breast cancer. J Clin Oncol 2014; 32:3840-7. [PMID: 25349301 DOI: 10.1200/jco.2014.55.2984] [Citation(s) in RCA: 226] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
PURPOSE There is a lack of treatments providing survival benefit for patients with metastatic triple-negative breast cancer (mTNBC), with no standard of care. A randomized phase II trial showed significant benefit for gemcitabine, carboplatin, and iniparib (GCI) over gemcitabine and carboplatin (GC) in clinical benefit rate, response rate, progression-free survival (PFS), and overall survival (OS). Here, we formally compare the efficacy of these regimens in a phase III trial. PATIENTS AND METHODS Patients with stage IV/locally recurrent TNBC who had received no more than two previous chemotherapy regimens for mTNBC were randomly allocated to gemcitabine 1,000 mg/m(2) and carboplatin area under the curve 2 (days 1 and 8) alone or GC plus iniparib 5.6 mg/kg (days 1, 4, 8, and 11) every 3 weeks. Random assignment was stratified by the number of prior chemotherapies. The coprimary end points were OS and PFS. Patients receiving GC could cross over to iniparib on progression. RESULTS Five hundred nineteen patients were randomly assigned (261 GCI; 258 GC). In the primary analysis, no statistically significant difference was observed for OS (hazard ratio [HR] = 0.88; 95% CI, 0.69 to 1.12; P = .28) nor PFS (HR = 0.79; 95% CI, 0.65 to 0.98; P = .027). An exploratory analysis showed that patients in the second-/third-line had improved OS (HR = 0.65; 95% CI, 0.46 to 0.91) and PFS (HR = 0.68; 95% CI, 0.50 to 0.92) with GCI. The safety profile for GCI was similar to GC. CONCLUSION The trial did not meet the prespecified criteria for the coprimary end points of PFS and OS in the ITT population. The potential benefit with iniparib observed in second-/third-line subgroup warrants further evaluation.
Collapse
Affiliation(s)
- Joyce O'Shaughnessy
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA.
| | - Lee Schwartzberg
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Michael A Danso
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Kathy D Miller
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Hope S Rugo
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Marcus Neubauer
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Nicholas Robert
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Beth Hellerstedt
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Mansoor Saleh
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Paul Richards
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Jennifer M Specht
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Denise A Yardley
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Robert W Carlson
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Richard S Finn
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Eric Charpentier
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Ignacio Garcia-Ribas
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| | - Eric P Winer
- Joyce O'Shaughnessy, Baylor Charles A. Sammons Cancer Center, Texas Oncology; Joyce O'Shaughnessy, US Oncology, Dallas; Beth Hellerstedt Texas Oncology-Round Rock, Austin, TX; Lee Schwartzberg, Accelerated Community Oncology Research Network; Lee Schwartzberg, The West Clinic, Memphis; Denise A. Yardley, Sarah Cannon Research Institute; Denise A. Yardley, Tennessee Oncology, Nashville, TN; Michael A. Danso, US Oncology; Michael A. Danso, Virginia Oncology Associates, Norfolk; Nicholas Robert, Virginia Cancer Specialists, Fairfax; Paul Richards, Blue Ridge Cancer Care, Salem, VA; Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Hope S. Rugo, University of California, San Francisco Comprehensive Cancer Center, San Francisco; Robert W. Carlson, Stanford Comprehensive Cancer Center, Palo Alto, CA; Richard S. Finn, Geffen School of Medicine at University of California, Los Angeles; Richard S. Finn, Translational Research in Oncology, Los Angeles, CA; Marcus Neubauer, Kansas City Cancer City, Overland Park, KS; Mansoor Saleh, Georgia Cancer Specialists, Sandy Springs, GA; Jennifer M. Specht, University of Washington, Seattle Cancer Care Alliance, Seattle, WA; Eric Charpentier, Ignacio Garcia-Ribas, Sanofi, Cambridge; and Eric P. Winer, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
117
|
Maddalo D, Manchado E, Concepcion CP, Bonetti C, Vidigal JA, Han YC, Ogrodowski P, Crippa A, Rekhtman N, de Stanchina E, Lowe SW, Ventura A. In vivo engineering of oncogenic chromosomal rearrangements with the CRISPR/Cas9 system. Nature 2014; 516:423-7. [PMID: 25337876 PMCID: PMC4270925 DOI: 10.1038/nature13902] [Citation(s) in RCA: 477] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/30/2014] [Indexed: 12/24/2022]
Abstract
Chromosomal rearrangements play a central role in the pathogenesis of human cancers and often result in the expression of therapeutically actionable gene fusions1. A recently discovered example is a fusion between the Echinoderm Microtubule-associated Protein-like 4 (EML4) and the Anaplastic Lymphoma Kinase (ALK) genes, generated by an inversion on the short arm of chromosome 2: inv(2)(p21p23). The EML4-ALK oncogene is detected in a subset of human non-small cell lung cancers (NSCLC)2 and is clinically relevant because it confers sensitivity to ALK inhibitors3. Despite their importance, modeling such genetic events in mice has proven challenging and requires complex manipulation of the germline. Here we describe an efficient method to induce specific chromosomal rearrangements in vivo using viral-mediated delivery of the CRISPR/Cas9 system to somatic cells of adult animals. We apply it to generate a mouse model of Eml4-Alk-driven lung cancer. The resulting tumors invariably harbor the Eml4-Alkinversion, express the Eml4-Alk fusion gene, display histo-pathologic and molecular features typical of ALK+ human NSCLCs, and respond to treatment with ALK-inhibitors. The general strategy described here substantially expands our ability to model human cancers in mice and potentially in other organisms.
Collapse
Affiliation(s)
- Danilo Maddalo
- Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics Program, 1275 York Avenue, New York, New York 10065, USA
| | - Eusebio Manchado
- Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics Program, 1275 York Avenue, New York, New York 10065, USA
| | - Carla P Concepcion
- 1] Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics Program, 1275 York Avenue, New York, New York 10065, USA [2] Weill Cornell Graduate School of Medical Sciences of Cornell University, 1300 York Avenue, New York, New York 10065, USA
| | - Ciro Bonetti
- Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics Program, 1275 York Avenue, New York, New York 10065, USA
| | - Joana A Vidigal
- Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics Program, 1275 York Avenue, New York, New York 10065, USA
| | - Yoon-Chi Han
- Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics Program, 1275 York Avenue, New York, New York 10065, USA
| | - Paul Ogrodowski
- Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics Program, 1275 York Avenue, New York, New York 10065, USA
| | - Alessandra Crippa
- Milano-Bicocca University, Department of Medical Oncology, San Gerardo Hospital, 20052, Via G B Pergolesi 33, Monza, Italy
| | - Natasha Rekhtman
- Memorial Sloan Kettering Cancer Center, Thoracic Pathology and Cytopathology, 1275 York Avenue, New York, New York 10065, USA
| | - Elisa de Stanchina
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program, 1275 York Avenue, New York, New York 10065, USA
| | - Scott W Lowe
- 1] Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics Program, 1275 York Avenue, New York, New York 10065, USA [2] Howard Hughes Medical Institute, 1275 York Avenue, New York, New York 10065, USA
| | - Andrea Ventura
- Memorial Sloan Kettering Cancer Center, Cancer Biology and Genetics Program, 1275 York Avenue, New York, New York 10065, USA
| |
Collapse
|
118
|
BRCAness predicts resistance to taxane-containing regimens in triple negative breast cancer during neoadjuvant chemotherapy. Clin Breast Cancer 2014; 15:80-5. [PMID: 25445419 DOI: 10.1016/j.clbc.2014.08.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 08/12/2014] [Accepted: 08/25/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND To provide optimal treatment of heterogeneous triple negative breast cancer (TNBC), we need biomarkers that can predict the chemotherapy response. PATIENTS AND METHODS We retrospectively investigated BRCAness in 73 patients with breast cancer who had been treated with taxane- and/or anthracycline-based neoadjuvant chemotherapy (NAC). Using multiplex, ligation-dependent probe amplification on formalin-fixed core needle biopsy (CNB) specimens before NAC and surgical specimens after NAC. BRCAness status was assessed with the assessor unaware of the clinical information. RESULTS We obtained 45 CNB and 60 surgical specimens from the 73 patients. Of the 45 CNB specimens, 17 had BRCAness (38.6% of all subtypes). Of the 23 TNBC CNB specimens, 14 had BRCAness (61% of TNBC cases). The clinical response rates were significantly lower for BRCAness than for non-BRCAness tumors, both for all tumors (58.8% vs. 89.3%, P = .03) and for TNBC (50% vs. 100%, P = .02). All tumors that progressed with taxane therapy had BRCAness. Of the patients with TNBC, those with non-BRCAness cancer had pathologic complete responses significantly more often than did those with BRCAness tumors (77.8% vs. 14.3%, P = .007). After NAC, the clinical response rates were significant lower for BRCAness than for non-BRCAness tumors in all subtypes (P = .002) and in TNBC cases (P = .008). After a median follow-up of 26.4 months, 6 patients-all with BRCAness-had developed recurrence. Patients with BRCAness had shorter progression-free survival than did those with non- BRCAness (P = .049). CONCLUSION Identifying BRCAness can help predict the response to taxane, and changing regimens for BRCAness TNBC might improve patient survival. A larger prospective study is needed to further clarify this issue.
Collapse
|
119
|
Moran T, Wei J, Cobo M, Qian X, Domine M, Zou Z, Bover I, Wang L, Provencio M, Yu L, Chaib I, You C, Massuti B, Song Y, Vergnenegre A, Lu H, Lopez-Vivanco G, Hu W, Robinet G, Yan J, Insa A, Xu X, Majem M, Chen X, de Las Peñas R, Karachaliou N, Sala MA, Wu Q, Isla D, Zhou Y, Baize N, Zhang F, Garde J, Germonpre P, Rauh S, ALHusaini H, Sanchez-Ronco M, Drozdowskyj A, Sanchez JJ, Camps C, Liu B, Rosell R, Colinet B, De Grève J, Germonpré P, Chen H, Chen X, Du J, Gao Y, Hu J, Hu W, Kong W, Li L, Li R, Li X, Liu B, Liu J, Lu H, Qian X, Ren W, Song Y, Wang L, Wei J, Wen L, Wu Q, Xiao X, Xu X, Yan J, Yang J, Yang M, Yang Y, Yin J, You C, Yu L, Yue X, Zhang F, Zhang J, Zhou Y, Zhu L, Zou Z, Baize N, Bombaron P, Chouaid C, Dansin E, Fournel P, Fraboulet G, Gervais R, Hominal S, Kahlout S, Lecaer H, Lena H, LeTreut J, Locher C, Molinier O, Monnet I, Oliviero G, Robinet G, Schoot R, Thomas P, et alMoran T, Wei J, Cobo M, Qian X, Domine M, Zou Z, Bover I, Wang L, Provencio M, Yu L, Chaib I, You C, Massuti B, Song Y, Vergnenegre A, Lu H, Lopez-Vivanco G, Hu W, Robinet G, Yan J, Insa A, Xu X, Majem M, Chen X, de Las Peñas R, Karachaliou N, Sala MA, Wu Q, Isla D, Zhou Y, Baize N, Zhang F, Garde J, Germonpre P, Rauh S, ALHusaini H, Sanchez-Ronco M, Drozdowskyj A, Sanchez JJ, Camps C, Liu B, Rosell R, Colinet B, De Grève J, Germonpré P, Chen H, Chen X, Du J, Gao Y, Hu J, Hu W, Kong W, Li L, Li R, Li X, Liu B, Liu J, Lu H, Qian X, Ren W, Song Y, Wang L, Wei J, Wen L, Wu Q, Xiao X, Xu X, Yan J, Yang J, Yang M, Yang Y, Yin J, You C, Yu L, Yue X, Zhang F, Zhang J, Zhou Y, Zhu L, Zou Z, Baize N, Bombaron P, Chouaid C, Dansin E, Fournel P, Fraboulet G, Gervais R, Hominal S, Kahlout S, Lecaer H, Lena H, LeTreut J, Locher C, Molinier O, Monnet I, Oliviero G, Robinet G, Schoot R, Thomas P, Vergnènegre A, Berchem G, Rauh S, Al Husaini H, Aparisi F, Arriola E, Ballesteros I, Barneto I, Bernabé R, Blasco A, Bosch-Barrera J, Bover I, Calvo de Juan V, Camps C, Carcereny E, Catot S, Cobo M, De Las Peñas R, Dómine M, Felip E, García-Campelo MR, García-Girón C, García-Gómez R, Garcia-Sevila R, Garde J, Gasco A, Gil J, González-Larriba JL, Hernando-Polo S, Jantus E, Insa A, Isla D, Jiménez B, Lianes P, López-López R, López-Martín A, López-Vivanco G, Macias JA, Majem M, Marti-Ciriquian JL, Massuti B, Montoyo R, Morales-Espinosa D, Morán T, Moreno MA, Pallares C, Parera M, Pérez-Carrión R, Porta R, Provencio M, Reguart N, Rosell R, Rosillo F, Sala MA, Sanchez JM, Sullivan I, Terrasa J, Trigo JM, Valdivia J, Viñolas N, Viteri S, Botia-Castillo M, Mate JL, Perez-Cano M, Ramirez JL, Sanchez-Rodriguez B, Taron M, Tierno-Garcia M, Mijangos E, Ocaña J, Pereira E, Shao J, Sun X, O'Brate R. Two biomarker-directed randomized trials in European and Chinese patients with nonsmall-cell lung cancer: the BRCA1-RAP80 Expression Customization (BREC) studies. Ann Oncol 2014; 25:2147-2155. [PMID: 25164908 DOI: 10.1093/annonc/mdu389] [Show More Authors] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND In a Spanish Lung Cancer Group (SLCG) phase II trial, the combination of BRCA1 and receptor-associated protein 80 (RAP80) expression was significantly associated with outcome in Caucasian patients with nonsmall-cell lung cancer (NSCLC). The SLCG therefore undertook an industry-independent collaborative randomized phase III trial comparing nonselected cisplatin-based chemotherapy with therapy customized according to BRCA1/RAP80 expression. An analogous randomized phase II trial was carried out in China under the auspices of the SLCG to evaluate the effect of BRCA1/RAP80 expression in Asian patients. PATIENTS AND METHODS Eligibility criteria included stage IIIB-IV NSCLC and sufficient tumor specimen for molecular analysis. Randomization to the control or experimental arm was 1 : 1 in the SLCG trial and 1 : 3 in the Chinese trial. In both trials, patients in the control arm received docetaxel/cisplatin; in the experimental arm, patients with low RAP80 expression received gemcitabine/cisplatin, those with intermediate/high RAP80 expression and low/intermediate BRCA1 expression received docetaxel/cisplatin, and those with intermediate/high RAP80 expression and high BRCA1 expression received docetaxel alone. The primary end point was progression-free survival (PFS). RESULTS Two hundred and seventy-nine patients in the SLCG trial and 124 in the Chinese trial were assessable for PFS. PFS in the control and experimental arms in the SLCG trial was 5.49 and 4.38 months, respectively [log rank P = 0.07; hazard ratio (HR) 1.28; P = 0.03]. In the Chinese trial, PFS was 4.74 and 3.78 months, respectively (log rank P = 0.82; HR 0.95; P = 0.82). CONCLUSION Accrual was prematurely closed on the SLCG trial due to the absence of clinical benefit in the experimental over the control arm. However, the BREC studies provide proof of concept that an international, nonindustry, biomarker-directed trial is feasible. Thanks to the groundwork laid by these studies, we expect that ongoing further research on alternative biomarkers to elucidate DNA repair mechanisms will help define novel therapeutic approaches. TRIAL REGISTRATION NCT00617656/GECP-BREC and ChiCTR-TRC-12001860/BREC-CHINA.
Collapse
Affiliation(s)
- T Moran
- Catalan Institute of Oncology, Medical Oncology Service, Hospital Germans Trias i Pujol, Badalona, Spain
| | - J Wei
- The Comprehensive Cancer Centre, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - M Cobo
- Medical Oncology Service, Hospital Carlos Haya, Malaga
| | - X Qian
- The Comprehensive Cancer Centre, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - M Domine
- Medical Oncology Service, Fundacion Jimenez Diaz, Madrid
| | - Z Zou
- The Comprehensive Cancer Centre, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - I Bover
- Medical Oncology Service, Hospital Son Llatzer, Palma de Mallorca
| | - L Wang
- The Comprehensive Cancer Centre, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - M Provencio
- Medical Oncology Service, Hospital Puerta de Hierro, Madrid, Spain
| | - L Yu
- The Comprehensive Cancer Centre, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - I Chaib
- Catalan Institute of Oncology, Medical Oncology Service, Hospital Germans Trias i Pujol, Badalona, Spain
| | - C You
- Department of Oncology, Suqian General Hospital, Suqian, China
| | - B Massuti
- Medical Oncology Service, Hospital General de Alicante, Alicante, Spain
| | - Y Song
- Department of Pneumology, Jinling Hospital, Nanjing, China
| | - A Vergnenegre
- Service de Pathologie Respiratoire et d'Allergologie, CHU Limoges, Limoges, France
| | - H Lu
- Department of Pneumology, Taizhou General Hospital, Taizhou, China
| | | | - W Hu
- The Comprehensive Cancer Centre, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - G Robinet
- Service Pneumologie, CHU Brest, Brest, France
| | - J Yan
- The Comprehensive Cancer Centre, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - A Insa
- Medical Oncology Service, Hospital Clinico de Valencia, Valencia, Spain
| | - X Xu
- Department of Pneumology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - M Majem
- Medical Oncology Service, Hospital Sant Pau, Barcelona, Spain
| | - X Chen
- Department of Oncology, Huaian General Hospital, Huaian, China
| | - R de Las Peñas
- Medical Oncology Service, Hospital Provincial de Castellon, Castellon, Spain
| | - N Karachaliou
- Translational Research Unit, Dr Rosell Oncology Institute, Quiron-Dexeus University Hospital, Barcelona
| | - M A Sala
- Medical Oncology Service, Hospital de Basurto, Bilbao, Spain
| | - Q Wu
- Department of Oncology, Yixin General Hospital, Yixin, China
| | - D Isla
- Medical Oncology Service, Hospital Lozano Blesa, Zaragoza, Spain
| | - Y Zhou
- Department of Oncology, Yixin General Hospital, Yixin, China
| | - N Baize
- Department de Pneumologie, CHU Angers, Angers, France
| | - F Zhang
- Department of Oncology, Maanshan General Hospital, Maanshan, China
| | - J Garde
- Medical Oncology Service, Hospital Arnau de Vilanova, Valencia, Spain
| | - P Germonpre
- Department of Pulmonary Medicine, Antwerp University Hospital, Edegem, Belgium
| | - S Rauh
- Department of Internal Medicine and Oncology, Centre Hospitalier Emile Mayrisch, Luxembourg, Luxembourg
| | - H ALHusaini
- Oncology Center, King Faisal Cancer Center, Riyadh, Saudi Arabia
| | - M Sanchez-Ronco
- Department of Health and Medicosocial Sciences, University of Alcala, Madrid
| | | | - J J Sanchez
- Department of Preventive Medicine, Autonomous University of Madrid, Madrid
| | - C Camps
- Medical Oncology Service, Hospital General de Valencia, Valencia
| | - B Liu
- The Comprehensive Cancer Centre, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - R Rosell
- Catalan Institute of Oncology, Cancer Biology and Precision Medicine Program, Hospital Germans Trias i Pujol, Badalona; MORe Foundation, Barcelona, Spain; Cancer Therapeutic Innovation Group, New York,USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Vollebergh MA, Lips EH, Nederlof PM, Wessels LFA, Wesseling J, Vd Vijver MJ, de Vries EGE, van Tinteren H, Jonkers J, Hauptmann M, Rodenhuis S, Linn SC. Genomic patterns resembling BRCA1- and BRCA2-mutated breast cancers predict benefit of intensified carboplatin-based chemotherapy. Breast Cancer Res 2014; 16:R47. [PMID: 24887359 PMCID: PMC4076636 DOI: 10.1186/bcr3655] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 05/02/2014] [Indexed: 12/12/2022] Open
Abstract
Introduction BRCA-mutated breast cancer cells lack the DNA-repair mechanism homologous recombination that is required for error-free DNA double-strand break (DSB) repair. Homologous recombination deficiency (HRD) may cause hypersensitivity to DNA DSB-inducing agents, such as bifunctional alkylating agents and platinum salts. HRD can be caused by BRCA mutations, and by other mechanisms. To identify HRD, studies have focused on triple-negative (TN) breast cancers as these resemble BRCA1-mutated breast cancer closely and might also share this hypersensitivity. However, ways to identify HRD in non-BRCA-mutated, estrogen receptor (ER)-positive breast cancers have remained elusive. The current study provides evidence that genomic patterns resembling BRCA1- or BRCA2-mutated breast cancers can identify breast cancer patients with TN as well as ER-positive, HER2-negative tumors that are sensitive to intensified, DSB-inducing chemotherapy. Methods Array comparative genomic hybridization (aCGH) was used to classify breast cancers. Patients with tumors with similar aCGH patterns as BRCA1- and/or BRCA2-mutated breast cancers were defined as having a BRCA-likeCGH status, others as non-BCRA-likeCGH. Stage-III patients (n = 249) had participated in a randomized controlled trial of adjuvant high-dose (HD) cyclophosphamide-thiotepa-carboplatin (CTC) versus 5-fluorouracil-epirubicin-cyclophosphamide (FE90C) chemotherapy. Results Among patients with BRCA-likeCGH tumors (81/249, 32%), a significant benefit of HD-CTC compared to FE90C was observed regarding overall survival (adjusted hazard ratio 0.19, 95% CI: 0.08 to 0.48) that was not seen for patients with non-BRCA-likeCGH tumors (adjusted hazard ratio 0.90, 95% CI: 0.53 to 1.54) (P = 0.004). Half of all BRCA-likeCGH tumors were ER-positive. Conclusions Distinct aCGH patterns differentiated between HER2-negative patients with a markedly improved outcome after adjuvant treatment with an intensified DNA-DSB-inducing regimen (BRCA-likeCGH patients) and those without benefit (non-BRCA-likeCGH patients).
Collapse
|
121
|
Szabova L, Bupp S, Kamal M, Householder DB, Hernandez L, Schlomer JJ, Baran ML, Yi M, Stephens RM, Annunziata CM, Martin PL, Van Dyke TA, Ohler ZW, Difilippantonio S. Pathway-specific engineered mouse allograft models functionally recapitulate human serous epithelial ovarian cancer. PLoS One 2014; 9:e95649. [PMID: 24748377 PMCID: PMC3991711 DOI: 10.1371/journal.pone.0095649] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 03/28/2014] [Indexed: 12/22/2022] Open
Abstract
The high mortality rate from ovarian cancers can be attributed to late-stage diagnosis and lack of effective treatment. Despite enormous effort to develop better targeted therapies, platinum-based chemotherapy still remains the standard of care for ovarian cancer patients, and resistance occurs at a high rate. One of the rate limiting factors for translation of new drug discoveries into clinical treatments has been the lack of suitable preclinical cancer models with high predictive value. We previously generated genetically engineered mouse (GEM) models based on perturbation of Tp53 and Rb with or without Brca1 or Brca2 that develop serous epithelial ovarian cancer (SEOC) closely resembling the human disease on histologic and molecular levels. Here, we describe an adaptation of these GEM models to orthotopic allografts that uniformly develop tumors with short latency and are ideally suited for routine preclinical studies. Ovarian tumors deficient in Brca1 respond to treatment with cisplatin and olaparib, a PARP inhibitor, whereas Brca1-wild type tumors are non-responsive to treatment, recapitulating the relative sensitivities observed in patients. These mouse models provide the opportunity for evaluation of effective therapeutics, including prediction of differential responses in Brca1-wild type and Brca1-deficient tumors and development of relevant biomarkers.
Collapse
MESH Headings
- Allografts
- Animals
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/pharmacology
- BRCA1 Protein/genetics
- Carcinoma, Ovarian Epithelial
- Cell Line, Tumor
- Cluster Analysis
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/mortality
- Cystadenocarcinoma, Serous/pathology
- Disease Models, Animal
- Disease Progression
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression Profiling
- Humans
- Mice
- Mutation
- Neoplasms, Glandular and Epithelial/drug therapy
- Neoplasms, Glandular and Epithelial/genetics
- Neoplasms, Glandular and Epithelial/metabolism
- Neoplasms, Glandular and Epithelial/mortality
- Neoplasms, Glandular and Epithelial/pathology
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/mortality
- Ovarian Neoplasms/pathology
- Tumor Burden/drug effects
Collapse
Affiliation(s)
- Ludmila Szabova
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Sujata Bupp
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Muhaymin Kamal
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Deborah B. Householder
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Lidia Hernandez
- Medical Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jerome J. Schlomer
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Maureen L. Baran
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Ming Yi
- Advanced Biomedical Computing Center, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Robert M. Stephens
- Advanced Biomedical Computing Center, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Christina M. Annunziata
- Medical Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Philip L. Martin
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Terry A. Van Dyke
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- Mouse Cancer Genetics Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Zoe Weaver Ohler
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Simone Difilippantonio
- Center for Advanced Preclinical Research, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| |
Collapse
|
122
|
Spliethoff JW, Evers DJ, Jaspers JE, Hendriks BHW, Rottenberg S, Ruers TJM. Monitoring of tumor response to Cisplatin using optical spectroscopy. Transl Oncol 2014; 7:230-9. [PMID: 24726234 PMCID: PMC4101345 DOI: 10.1016/j.tranon.2014.02.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 12/07/2013] [Accepted: 01/02/2014] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Anatomic imaging alone is often inadequate for tuning systemic treatment for individual tumor response. Optically based techniques could potentially contribute to fast and objective response monitoring in personalized cancer therapy. In the present study, we evaluated the feasibility of dual-modality diffuse reflectance spectroscopy-autofluorescence spectroscopy (DRS-AFS) to monitor the effects of systemic treatment in a mouse model for hereditary breast cancer. METHODS Brca1(-/-); p53(-/-) mammary tumors were grown in 36 mice, half of which were treated with a single dose of cisplatin. Changes in the tumor physiology and morphology were measured for a period of 1 week using dual-modality DRS-AFS. Liver and muscle tissues were also measured to distinguish tumor-specific alterations from systemic changes. Model-based analyses were used to derive different optical parameters like the scattering and absorption coefficients, as well as sources of intrinsic fluorescence. Histopathologic analysis was performed for cross-validation with trends in optically based parameters. RESULTS Treated tumors showed a significant decrease in Mie-scattering slope and Mie-to-total scattering fraction and an increase in both fat volume fraction and tissue oxygenation after 2 days of follow-up. Additionally, significant tumor-specific changes in the fluorescence spectra were seen. These longitudinal trends were consistent with changes observed in the histopathologic analysis, such as vital tumor content and formation of fibrosis. CONCLUSIONS This study demonstrates that dual-modality DRS-AFS provides quantitative functional information that corresponds well with the degree of pathologic response. DRS-AFS, in conjunction with other imaging modalities, could be used to optimize systemic cancer treatment on the basis of early individual tumor response.
Collapse
Affiliation(s)
- Jarich W Spliethoff
- Department of Surgery, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Daniel J Evers
- Department of Surgery, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Janneke E Jaspers
- Division of Molecular Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Benno H W Hendriks
- Department of Minimally Invasive Healthcare, Philips Research, Eindhoven, The Netherlands
| | - Sven Rottenberg
- Division of Molecular Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Theo J M Ruers
- Department of Surgery, The Netherlands Cancer Institute, Amsterdam, The Netherlands; MIRA Institute, Technical University Twente, Enschede, The Netherlands
| |
Collapse
|
123
|
Olive KP, Politi K. Translational therapeutics in genetically engineered mouse models of cancer. Cold Spring Harb Protoc 2014; 2014:131-143. [PMID: 24492770 DOI: 10.1101/pdb.top069997] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Advances in knowledge of the molecular alterations of human cancers, refinements in technologies for the generation of genetically engineered mouse models (GEMMs), and the development of cancer therapies have accelerated in recent years. Progress in these fields provides the foundation for clinically relevant studies to be performed in GEMMs, through which it is possible to glean information on drug efficacy and to identify determinants of sensitivity and resistance to drugs and drug combinations. GEMMs used in pre-, co-, and postclinical studies must closely recapitulate the genetics, histopathology, and response to therapy of the human disease. Prevention and intervention trials can be designed in GEMMs to test the effects of drugs on tumor initiation, regression, and progression. Given their complexity, careful consideration of the infrastructure requirements and practical aspects of each individual experiment, including enrollment, tumor monitoring, and dose and schedule, must be considered in the design of therapeutic studies in GEMMs. Advantages of GEMMs include the ability to rapidly perform drug efficacy studies in a defined genetic background, the ease of pharmacodynamic and pharmacokinetic assessments, and the possibility of experimentally manipulating model systems to address questions that cannot be addressed in patients. In light of these features, GEMMs are useful tools for translational studies to inform clinical trials in cancer patients.
Collapse
Affiliation(s)
- Kenneth P Olive
- Departments of Medicine and Pathology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032
| | | |
Collapse
|
124
|
Hadjiandreou MM, Mitsis GD. Mathematical Modeling of Tumor Growth, Drug-Resistance, Toxicity, and Optimal Therapy Design. IEEE Trans Biomed Eng 2014; 61:415-25. [DOI: 10.1109/tbme.2013.2280189] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
125
|
Sharma P, Stecklein SR, Kimler BF, Sethi G, Petroff BK, Phillips TA, Tawfik OW, Godwin AK, Jensen RA. The prognostic value of BRCA1 promoter methylation in early stage triple negative breast cancer. ACTA ACUST UNITED AC 2014; 3:1-11. [PMID: 25177489 PMCID: PMC4147783 DOI: 10.7243/2049-7962-3-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction Methylation of the BRCA1 promoter is frequent in triple negative breast cancers (TNBC) and results in a tumor phenotype similar to BRCA1-mutated tumors. BRCA1 mutation-associated cancers are more sensitive to DNA damaging agents as compared to conventional chemotherapy agents. It is not known if there is an interaction between the presence of BRCA1 promoter methylation (PM) and response to chemotherapy agents in sporadic TNBC. We sought to investigate the prognostic significance of BRCA1 PM in TNBC patients receiving standard chemotherapy. Methods Subjects with stage I-III TNBC treated with chemotherapy were identified and their formalin-fixed paraffin-embedded (FFPE) tumor specimens retrieved. Genomic DNA was isolated and subjected to methylation-specific PCR (MSPCR). Results DNA was isolated from primary tumor of 39 subjects. BRCA1 PM was detected in 30% of patients. Presence of BRCA1 PM was associated with lower BRCA1 transcript levels, suggesting epigenetic BRCA1 silencing. All patients received chemotherapy (anthracycline:90%, taxane:69%). At a median follow-up of 64 months, 46% of patients have recurred and 36% have died. On univariate analysis, African-American race, node positivity, stage, and BRCA1 PM were associated with worse RFS and OS. Five year OS was 36% for patients with BRCA1 PM vs. 77% for patients without BRCA1 PM (p=0.004). On multivariable analysis, BRCA1 PM was associated with significantly worse RFS and OS. Conclusions We show that BRCA1 PM is common in TNBC and has the potential to identify a significant fraction of TNBC patients who have suboptimal outcomes with standard chemotherapy.
Collapse
Affiliation(s)
- Priyanka Sharma
- Division of Hematology/Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Shane R Stecklein
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Bruce F Kimler
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA.,Breast Cancer Prevention Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Geetika Sethi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Brian K Petroff
- Division of Hematology/Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Breast Cancer Prevention Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Teresa A Phillips
- Division of Hematology/Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Breast Cancer Prevention Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ossama W Tawfik
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Roy A Jensen
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
126
|
Stuurman FE, Nuijen B, Beijnen JH, Schellens JHM. Oral anticancer drugs: mechanisms of low bioavailability and strategies for improvement. Clin Pharmacokinet 2013; 52:399-414. [PMID: 23420518 DOI: 10.1007/s40262-013-0040-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The use of oral anticancer drugs has increased during the last decade, because of patient preference, lower costs, proven efficacy, lack of infusion-related inconveniences, and the opportunity to develop chronic treatment regimens. Oral administration of anticancer drugs is, however, often hampered by limited bioavailability of the drug, which is associated with a wide variability. Since most anticancer drugs have a narrow therapeutic window and are dosed at or close to the maximum tolerated dose, a wide variability in the bioavailability can have a negative impact on treatment outcome. This review discusses mechanisms of low bioavailability of oral anticancer drugs and strategies for improvement. The extent of oral bioavailability depends on many factors, including release of the drug from the pharmaceutical dosage form, a drug's stability in the gastrointestinal tract, factors affecting dissolution, the rate of passage through the gut wall, and the pre-systemic metabolism in the gut wall and liver. These factors are divided into pharmaceutical limitations, physiological endogenous limitations, and patient-specific limitations. There are several strategies to reduce or overcome these limitations. First, pharmaceutical adjustment of the formulation or the physicochemical characteristics of the drug can improve the dissolution rate and absorption. Second, pharmacological interventions by combining the drug with inhibitors of transporter proteins and/or pre-systemic metabolizing enzymes can overcome the physiological endogenous limitations. Third, chemical modification of a drug by synthesis of a derivative, salt form, or prodrug could enhance the bioavailability by improving the absorption and bypassing physiological endogenous limitations. Although the bioavailability can be enhanced by various strategies, the development of novel oral products with low solubility or cell membrane permeability remains cumbersome and is often unsuccessful. The main reasons are unacceptable variation in the bioavailability and high investment costs. Furthermore, novel oral anticancer drugs are frequently associated with toxic effects including unacceptable gastrointestinal adverse effects. Therefore, compliance is often suboptimal, which may negatively influence treatment outcome.
Collapse
Affiliation(s)
- Frederik E Stuurman
- Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
127
|
Abstract
Germline mutations of human breast cancer-associated gene 1 (BRCA1) predispose women to breast and ovarian cancers. In mice, over 20 distinct mutations, including null, hypomorphic, isoform, conditional, and point mutations, have been created to study functions of Brca1 in mammary development and tumorigenesis. Analyses using these mutant mice have yielded an enormous amount of information that greatly facilitates our understanding of the gender- and tissue-specific tumor suppressor functions of BRCA1, as well as enriches our insights into applying these preclinical models of disease to breast cancer research. Here, we review features of these mutant mice and their applications to cancer prevention and therapeutic treatment.
Collapse
|
128
|
Targeted therapies in breast cancer. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
129
|
Muggia F, Tommasi S, Lynch H, Paradiso A. Hereditary breast and ovarian cancer: lessening the burden. Ann Oncol 2013; 24 Suppl 8:viii5-viii6. [PMID: 24298633 DOI: 10.1093/annonc/mdt318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
130
|
Ai S, Jia T, Ai W, Duan J, Liu Y, Chen J, Liu X, Yang F, Tian Y, Huang Z. Targeted delivery of doxorubicin through conjugation with EGF receptor-binding peptide overcomes drug resistance in human colon cancer cells. Br J Pharmacol 2013; 168:1719-35. [PMID: 23146125 DOI: 10.1111/bph.12055] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 10/10/2012] [Accepted: 11/01/2012] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Induction of multidrug resistance by doxorubicin (DOX), together with non-specific toxicities, has restricted DOX-based chemotherapy. Recently, we demonstrated that DOX conjugated with an EGF receptor-binding peptide (DOX-EBP) had enhanced anticancer efficacy and reduced systemic toxicity when targeting EGF receptor-overexpressing tumours. Here we investigated whether DOX-EBP is able to overcome drug resistance and the underlying molecular mechanisms. EXPERIMENTAL APPROACH DOX-resistant SW480/DOX cells were derived from non-resistant SW480 cells by stepwise exposure to increasing concentrations of DOX, and P-glycoprotein overexpression induced by DOX was confirmed by Western blotting. Cytotoxicity and intracellular distribution of drugs were evaluated by MTT assay and fluorescence microscopy respectively. EGF receptor-mediated endocytosis was determined in EGF receptor and endocytosis inhibition assays. Drug accumulation in tumour cells and murine xenografts was determined by HPLC. KEY RESULTS The cytotoxicity and accumulation of DOX-EBP in SW480/DOX cells were almost the same as in SW480 cells, but those of free DOX were reduced. DOX-EBP accumulation was prevented by inhibitors of both EGF receptors and endocytosis, suggesting EGF receptors mediate endocytotic uptake. Tumour accumulation of DOX-EBP was significantly higher than free DOX in mice, and the levels of DOX-EBP were similar in DOX-resistant and non-resistant tumour tissues. Importantly, DOX-EBP, but not free DOX, was effective at inhibiting solid tumour growth and increased survival rate in both sensitive and resistant models. CONCLUSION AND IMPLICATIONS DOX-EBP can overcome DOX resistance of tumour cells and increase in vivo antitumour efficacy. Therefore, it has the potential to be a potent therapeutic agent for treating drug-resistant cancers.
Collapse
Affiliation(s)
- Shibin Ai
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Zhao Y, Alakhova DY, Kabanov AV. Can nanomedicines kill cancer stem cells? Adv Drug Deliv Rev 2013; 65:1763-83. [PMID: 24120657 DOI: 10.1016/j.addr.2013.09.016] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 09/30/2013] [Accepted: 09/30/2013] [Indexed: 12/12/2022]
Abstract
Most tumors are heterogeneous and many cancers contain small population of highly tumorigenic and intrinsically drug resistant cancer stem cells (CSCs). Like normal stem cell, CSCs have the ability to self-renew and differentiate to other tumor cell types. They are believed to be a source for drug resistance, tumor recurrence and metastasis. CSCs often overexpress drug efflux transporters, spend most of their time in non-dividing G0 cell cycle state, and therefore, can escape the conventional chemotherapies. Thus, targeting CSCs is essential for developing novel therapies to prevent cancer relapse and emerging of drug resistance. Nanocarrier-based therapeutic agents (nanomedicines) have been used to achieve longer circulation times, better stability and bioavailability over current therapeutics. Recently, some groups have successfully applied nanomedicines to target CSCs to eliminate the tumor and prevent its recurrence. These approaches include 1) delivery of therapeutic agents (small molecules, siRNA, antibodies) that affect embryonic signaling pathways implicated in self-renewal and differentiation in CSCs, 2) inhibiting drug efflux transporters in an attempt to sensitize CSCs to therapy, 3) targeting metabolism in CSCs through nanoformulated chemicals and field-responsive magnetic nanoparticles and carbon nanotubes, and 4) disruption of multiple pathways in drug resistant cells using combination of chemotherapeutic drugs with amphiphilic Pluronic block copolymers. Despite clear progress of these studies the challenges of targeting CSCs by nanomedicines still exist and leave plenty of room for improvement and development. This review summarizes biological processes that are related to CSCs, overviews the current state of anti-CSCs therapies, and discusses state-of-the-art nanomedicine approaches developed to kill CSCs.
Collapse
|
132
|
Hadjiandreou MM, Mitsis GD. Taking a break from chemotherapy to fight drug-resistance: The cases of cancer and HIV/AIDS. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2013; 2013:197-200. [PMID: 24109658 DOI: 10.1109/embc.2013.6609471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In this work, we present how optimized treatment interruptions during chemotherapy may be used to control drug-resistance, a major challenge for clinicians worldwide. Specifically, we examine resistance in cancer and HIV/AIDS. For each disease, we use mathematical models alongside real data to represent the respective complex biological phenomena and optimal control algorithms to design optimized treatment schedules aiming at controlling disease progression and patient death. In both diseases, it is shown that the key to controlling resistance is the optimal management of the frequency and magnitude of treatment interruptions as a way to facilitate the interplay between the competitive resistant/sensitive strains.
Collapse
|
133
|
Pereira L, Igea A, Canovas B, Dolado I, Nebreda AR. Inhibition of p38 MAPK sensitizes tumour cells to cisplatin-induced apoptosis mediated by reactive oxygen species and JNK. EMBO Mol Med 2013; 5:1759-74. [PMID: 24115572 PMCID: PMC3840490 DOI: 10.1002/emmm.201302732] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 08/26/2013] [Accepted: 08/27/2013] [Indexed: 12/11/2022] Open
Abstract
The p38 MAPK pathway is an important regulator of many cellular responses. It is well established that p38 MAPK signalling negatively regulates epithelial cell transformation, but enhanced p38 MAPK activity has been also correlated with bad clinical prognosis in some tumour types. Here, we provide genetic and pharmacological evidence showing that p38 MAPK inhibition cooperates with the chemotherapeutic agent cisplatin to kill tumour cells. We show that p38 MAPK inhibition results in ROS upregulation, which in turn activates the JNK pathway via inactivation of phosphatases, sensitizing human tumour cells to cisplatin-induced apoptosis. Using a mouse model for breast cancer, we confirm that inhibition of p38 MAPK cooperates with cisplatin treatment to reduce tumour size and malignancy in vivo. Taken together, our results illustrate a new function of p38 MAPK that helps tumour cells to survive chemotherapeutic drug treatments, and reveal that the combination of p38 MAPK inhibitors with cisplatin can be potentially exploited for cancer therapy.
Collapse
Affiliation(s)
- Lorena Pereira
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
| | | | | | | | | |
Collapse
|
134
|
Opportunities and hurdles in the treatment of BRCA1-related breast cancer. Oncogene 2013; 33:3753-63. [PMID: 23955079 DOI: 10.1038/onc.2013.329] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/13/2013] [Accepted: 06/21/2013] [Indexed: 12/11/2022]
Abstract
BRCA1 functions as a classical tumor suppressor in breast and ovarian cancer. While the role of BRCA1 in homology-directed repair of DNA double-strand breaks contributes to its tumor suppressive activity, it also renders BRCA1-deficient cells highly sensitive to DNA-damaging agents. Although BRCA1 deficiency is therefore considered to be an attractive therapeutic target, re-activation of BRCA1 by secondary mutations has been shown to cause therapy resistance. In this review, we will assess the role of BRCA1 in both hereditary and sporadic breast cancer and discuss how different functionalities of the BRCA1 protein can contribute to its tumor suppressor function. In addition, we will discuss how this knowledge on BRCA1 function can help to overcome the hurdles encountered in the clinic and improve current treatment strategies for patients with BRCA1-related breast cancer.
Collapse
|
135
|
Chalasani P, Livingston R. Differential chemotherapeutic sensitivity for breast tumors with "BRCAness": a review. Oncologist 2013; 18:909-16. [PMID: 23881989 DOI: 10.1634/theoncologist.2013-0039] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BRCA1 or BRCA2 mutations predispose to cancer development, primarily through their loss of role in the repair of DNA double-strand breaks. They play a key role in homologous recombination repair, which is a conservative, error-free DNA repair mechanism. When mutated, other alternative, error-prone mechanisms for DNA repair take over, leading to genomic instability. Somatic mutations are rare in sporadic breast tumors, but expression of BRCA1 and BRCA2 genes can be downregulated in other mechanistic ways. These tumors have similar features in terms of their phenotypic and genotypic profiles, which are normally regulated by these genes, and mutations lead to defective DNA repair capacity, called "BRCAness." Attempts have been made to exploit this differentially expressed feature between tumors and normal tissues by treatment with DNA-damaging chemotherapy agents. Cells with this functional BRCA deficiency should be selectively susceptible to DNA-damaging drugs. Preclinical and early clinical (primarily retrospective) evidence supports this approach. In contrast, there is emerging evidence of relative resistance of tumors containing BRCA1 or BRCA2 mutations (or BRCAness) to taxanes. In this review, we summarize the data supporting differential chemotherapeutic sensitivity on the basis of defective DNA repair. If confirmed with available, clinically applicable techniques, this differential chemosensitivity could lead to treatment choices in breast cancer that have a more individualized biologic basis.
Collapse
Affiliation(s)
- Pavani Chalasani
- University of Arizona Cancer Center, Tucson, Arizona 85724, USA.
| | | |
Collapse
|
136
|
Abstract
Given the malleable nature of cancer cells, we should expect, and do see, the development of resistance to any new chemotherapeutic agent. Models that help us understand how this happens are a first step to better and more effective chemotherapeutics.
Collapse
Affiliation(s)
- Tito Fojo
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | | |
Collapse
|
137
|
von Minckwitz G, Martin M. Neoadjuvant treatments for triple-negative breast cancer (TNBC). Ann Oncol 2013; 23 Suppl 6:vi35-9. [PMID: 23012300 DOI: 10.1093/annonc/mds193] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Neoadjuvant chemotherapy provides a means both of improving subsequent surgical intervention and of testing novel therapies or combinations. Historically, triple-negative breast cancer (TNBC) has responded well in the neoadjuvant setting, with rates of pathological complete response (pCR) commonly higher than for other breast tumour types. However, more than half of TNBC patients do not achieve a pCR and have a very poor prognosis. The lack of drug-targetable receptors on TNBC tumours has made improving the available interventions in TNBC an area of important medical need. The routine use of neoadjuvant anthracycline/taxane combinations in TNBC is currently being supplemented by ongoing investigations of their use with other types of agent. In particular, the substantial proportion of TNBC tumours associated with BRCA1 mutations is driving clinical research into the use of DNA-damaging agents such as platinums, as well as of potentiators of DNA damage such as the investigational agent iniparib and inhibitors of poly-ADP ribose polymerase such as olaparib. Tyrosine kinase receptor inhibitors and microtubule-targeting inhibitors of cell cycling are also under active investigation. The use of neoadjuvant treatment with pCR as a surrogate of overall survival will allow the rapid evaluation and comparison of these and other much-needed new treatments for TNBC.
Collapse
Affiliation(s)
- G von Minckwitz
- German Breast Group, GBG Forschungs GmbH, Neu-Isenburg, Germany.
| | | |
Collapse
|
138
|
Warmoes M, Jaspers JE, Xu G, Sampadi BK, Pham TV, Knol JC, Piersma SR, Boven E, Jonkers J, Rottenberg S, Jimenez CR. Proteomics of genetically engineered mouse mammary tumors identifies fatty acid metabolism members as potential predictive markers for cisplatin resistance. Mol Cell Proteomics 2013; 12:1319-34. [PMID: 23397111 DOI: 10.1074/mcp.m112.024182] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In contrast to various signatures that predict the prognosis of breast cancer patients, markers that predict chemotherapy response are still elusive. To detect such predictive biomarkers, we investigated early changes in protein expression using two mouse models for distinct breast cancer subtypes who have a differential knock-out status for the breast cancer 1, early onset (Brca1) gene. The proteome of cisplatin-sensitive BRCA1-deficient mammary tumors was compared with that of cisplatin-resistant mammary tumors resembling pleomorphic invasive lobular carcinoma. The analyses were performed 24 h after administration of the maximum tolerable dose of cisplatin. At this time point, drug-sensitive BRCA1-deficient tumors showed DNA damage, but cells were largely viable. By applying paired statistics and quantitative filtering, we identified highly discriminatory markers for the sensitive and resistant model. Proteins up-regulated in the sensitive model are involved in centrosome organization, chromosome condensation, homology-directed DNA repair, and nucleotide metabolism. Major discriminatory markers that were up-regulated in the resistant model were predominantly involved in fatty acid metabolism, such as fatty-acid synthase. Specific inhibition of fatty-acid synthase sensitized resistant cells to cisplatin. Our data suggest that exploring the functional link between the DNA damage response and cancer metabolism shortly after the initial treatment may be a useful strategy to predict the efficacy of cisplatin.
Collapse
Affiliation(s)
- Marc Warmoes
- OncoProteomics Laboratory, Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Jackson JG, Lozano G. The mutant p53 mouse as a pre-clinical model. Oncogene 2013; 32:4325-30. [PMID: 23318424 DOI: 10.1038/onc.2012.610] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/24/2012] [Accepted: 11/07/2012] [Indexed: 11/09/2022]
Abstract
The p53 tumor-suppressor pathway is dismantled in the development of most cancers. Mice with various p53 mutant alleles either singly or in combination with other genetic alterations are predisposed to tumor development. Here, we review studies utilizing p53 mutant mice that have recapitulated and informed clinical observations. These studies have demonstrated the p53 contribution, sometimes beneficial and sometimes detrimental, to treatment response in lymphomas, and lung and breast cancers. Further, we examine how p53 mutant mouse models have been used to test the efficacy of p53 reactivation as a therapeutic strategy.
Collapse
Affiliation(s)
- J G Jackson
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
140
|
Jaspers JE, Kersbergen A, Boon U, Sol W, van Deemter L, Zander SA, Drost R, Wientjens E, Ji J, Aly A, Doroshow JH, Cranston A, Martin NM, Lau A, O’Connor MJ, Ganesan S, Borst P, Jonkers J, Rottenberg S. Loss of 53BP1 causes PARP inhibitor resistance in Brca1-mutated mouse mammary tumors. Cancer Discov 2013; 3:68-81. [PMID: 23103855 PMCID: PMC7518105 DOI: 10.1158/2159-8290.cd-12-0049] [Citation(s) in RCA: 421] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
UNLABELLED Inhibition of PARP is a promising therapeutic strategy for homologous recombination-deficient tumors, such as BRCA1-associated cancers. We previously reported that BRCA1-deficient mouse mammary tumors may acquire resistance to the clinical PARP inhibitor (PARPi) olaparib through activation of the P-glycoprotein drug efflux transporter. Here, we show that tumor-specific genetic inactivation of P-glycoprotein increases the long-term response of BRCA1-deficient mouse mammary tumors to olaparib, but these tumors eventually developed PARPi resistance. In a fraction of cases, this resistance is caused by partial restoration of homologous recombination due to somatic loss of 53BP1. Importantly, PARPi resistance was minimized by long-term treatment with the novel PARP inhibitor AZD2461, which is a poor P-glycoprotein substrate. Together, our data suggest that restoration of homologous recombination is an important mechanism for PARPi resistance in BRCA1-deficient mammary tumors and that the risk of relapse of BRCA1-deficient tumors can be effectively minimized by using optimized PARP inhibitors. SIGNIFICANCE In this study, we show that loss of 53BP1 causes resistance to PARP inhibition in mouse mammary tumors that are deficient in BRCA1. We hypothesize that low expression or absence of 53BP1 also reduces the response of patients with BRCA1-deficient tumors to PARP inhibitors.
Collapse
Affiliation(s)
- Janneke E. Jaspers
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ariena Kersbergen
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ute Boon
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Wendy Sol
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Liesbeth van Deemter
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Serge A. Zander
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rinske Drost
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ellen Wientjens
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jiuping Ji
- National Clinical Target Validation Laboratory, National Cancer Institute, Frederick
| | - Amal Aly
- Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - James H. Doroshow
- Division of Cancer Treatment and Diagnosis and Laboratory of Molecular Pharmacology, National Cancer Institute, Bethesda, Maryland
| | | | | | - Alan Lau
- AstraZeneca, Macclesfield, United Kingdom
| | | | | | - Piet Borst
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sven Rottenberg
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
141
|
Doornebal CW, Klarenbeek S, Braumuller TM, Klijn CN, Ciampricotti M, Hau CS, Hollmann MW, Jonkers J, de Visser KE. A preclinical mouse model of invasive lobular breast cancer metastasis. Cancer Res 2013; 73:353-63. [PMID: 23151903 DOI: 10.1158/0008-5472.can-11-4208] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Metastatic disease accounts for more than 90% of cancer-related deaths, but the development of effective antimetastatic agents has been hampered by the paucity of clinically relevant preclinical models of human metastatic disease. Here, we report the development of a mouse model of spontaneous breast cancer metastasis, which recapitulates key events in its formation and clinical course. Specifically, using the conditional K14cre;Cdh1(F/F);Trp53(F/F) model of de novo mammary tumor formation, we orthotopically transplanted invasive lobular carcinoma (mILC) fragments into mammary glands of wild-type syngeneic hosts. Once primary tumors were established in recipient mice, we mimicked the clinical course of treatment by conducting a mastectomy. After surgery, recipient mice succumbed to widespread overt metastatic disease in lymph nodes, lungs, and gastrointestinal tract. Genomic profiling of paired mammary tumors and distant metastases showed that our model provides a unique tool to further explore the biology of metastatic disease. Neoadjuvant and adjuvant intervention studies using standard-of-care chemotherapeutics showed the value of this model in determining therapeutic agents that can target early- and late-stage metastatic disease. In obtaining a more accurate preclinical model of metastatic lobular breast cancer, our work offers advances supporting the development of more effective treatment strategies for metastatic disease.
Collapse
Affiliation(s)
- Chris W Doornebal
- Division of Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Sotiropoulou PA, Karambelas AE, Debaugnies M, Candi A, Bouwman P, Moers V, Revenco T, Rocha AS, Sekiguchi K, Jonkers J, Blanpain C. BRCA1 deficiency in skin epidermis leads to selective loss of hair follicle stem cells and their progeny. Genes Dev 2012; 27:39-51. [PMID: 23271346 DOI: 10.1101/gad.206573.112] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The accurate maintenance of genomic integrity is essential for tissue homeostasis. Deregulation of this process leads to cancer and aging. BRCA1 is a critical mediator of this process. Here, we performed conditional deletion of Brca1 during epidermal development and found that BRCA1 is specifically required for hair follicle (HF) formation and for development of adult HF stem cells (SCs). Mice deficient for Brca1 in the epidermis are hairless and display a reduced number of HFs that degenerate progressively. Surprisingly, the interfollicular epidermis and the sebaceous glands remain unaffected by Brca1 deletion. Interestingly, HF matrix transient amplifying progenitors present increased DNA damage, p53 stabilization, and caspase-dependent apoptosis compared with the interfollicular and sebaceous progenitors, leading to hyperproliferation, apoptosis, and subsequent depletion of the prospective adult HF SCs. Concomitant deletion of p53 and Brca1 rescues the defect of HF morphogenesis and loss of HF SCs. During adult homeostasis, BRCA1 is dispensable for quiescent bulge SCs, but upon their activation during HF regeneration, Brca1 deletion causes apoptosis and depletion of Brca1-deficient bulge SCs. Our data reveal a major difference in the requirement of BRCA1 between different types of epidermal SCs and progenitors and during the different activation stages of adult HF SCs.
Collapse
Affiliation(s)
- Panagiota A Sotiropoulou
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moleculaire, Université Libre de Bruxelles, Brussels, Belgium.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
EZN-2208 (PEG-SN38) overcomes ABCG2-mediated topotecan resistance in BRCA1-deficient mouse mammary tumors. PLoS One 2012; 7:e45248. [PMID: 23028879 PMCID: PMC3444454 DOI: 10.1371/journal.pone.0045248] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 08/16/2012] [Indexed: 12/12/2022] Open
Abstract
BRCA1 dysfunction in hereditary breast cancer causes defective homology-directed DNA repair and sensitivity towards DNA damaging agents like the clinically used topoisomerase I inhibitors topotecan and irinotecan. Using our conditional K14cre;Brca1F/F;p53F/F mouse model, we showed previously that BRCA1;p53-deficient mammary tumors initially respond to topotecan, but frequently acquire resistance by overexpression of the efflux transporter ABCG2. Here, we tested the pegylated SN38 compound EZN-2208 as a novel approach to treat BRCA1-mutated tumors that express ABCG2. We found that EZN-2208 therapy resulted in more pronounced and durable responses of ABCG2-positive tumors than topotecan or irinotecan therapy. We also evaluated tumor-specific ABCG2 inhibition by Ko143 in Abcg2−/− host animals that carried tumors with topotecan-induced ABCG2 expression. Addition of Ko143 moderately increased overall survival of these animals, but did not yield tumor responses like those seen after EZN-2208 therapy. Our results suggest that pegylation of Top1 inhibitors may be a useful strategy to circumvent efflux transporter-mediated resistance and to improve their efficacy in the clinic.
Collapse
|
144
|
Oonk AMM, van Rijn C, Smits MM, Mulder L, Laddach N, Savola SP, Wesseling J, Rodenhuis S, Imholz ALT, Lips EH. Clinical correlates of 'BRCAness' in triple-negative breast cancer of patients receiving adjuvant chemotherapy. Ann Oncol 2012; 23:2301-2305. [PMID: 22357256 DOI: 10.1093/annonc/mdr621] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We have previously reported an array comparative genomic hybridization profile that identifies triple-negative breast cancers (TNBC), with BRCA1 dysfunction and a high sensitivity to intensified dose bifunctional alkylating agents. To determine the effect of conventional-dose chemotherapy in patients with this so-called BRCA1-like profile, clinical characteristics and survival were studied in a large group of TNBC patients. PATIENTS AND METHODS DNA was isolated and BRCA1-like status was assessed in 101 patients with early-stage TNBC receiving adjuvant cyclophosphamide-based chemotherapy. Clinical characteristics and survival were compared between BRCA1-like and non-BRCA1-like groups. Results Sixty-six tumors (65%) had a BRCA1-like profile. Patients with BRCA1-like tumors tended to be younger and had more often node-negative disease (P = 0.06 and P = 0.03, respectively). Five-year recurrence-free survival was 80% for the BRCA1-like group and 75% for the non-BRCA1-like group (P = 0.35). T stage was the only variable significantly associated with survival. CONCLUSIONS BRCA1-like tumors share clinical features, like young age at diagnosis and similar nodal status, with breast cancers in BRCA1 mutation carriers. Their prognosis is similar to that of non-BRCA1-like tumors when conventional-dose chemotherapy is administered. TNBCs that are classified as BRCA1-like may contain a defect in homologous recombination and could, in theory, benefit from the addition of poly ADP ribose polymerase inhibitors.
Collapse
Affiliation(s)
| | | | - M M Smits
- Pathology, Deventer Hospital, Deventer
| | - L Mulder
- Departments of Experimental Therapy; Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam
| | | | | | - J Wesseling
- Departments of Experimental Therapy; Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam
| | - S Rodenhuis
- Department of Medical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
| | | | - E H Lips
- Departments of Experimental Therapy; Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam
| |
Collapse
|
145
|
Bouwman P, Jonkers J. The effects of deregulated DNA damage signalling on cancer chemotherapy response and resistance. Nat Rev Cancer 2012; 12:587-98. [PMID: 22918414 DOI: 10.1038/nrc3342] [Citation(s) in RCA: 485] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tumours with specific DNA repair defects can be completely dependent on back-up DNA repair pathways for their survival. This dependence can be exploited therapeutically to induce synthetic lethality in tumour cells. For instance, homologous recombination (HR)-deficient tumours can be effectively targeted by DNA double-strand break-inducing agents. However, not all HR-defective tumours respond equally well to this type of therapy. Tumour cells may acquire resistance by invoking biochemical mechanisms that reduce drug action or by acquiring additional alterations in DNA damage response pathways. A thorough understanding of these processes is important for predicting treatment response and for the development of novel treatment strategies that prevent the emergence of therapy-resistant tumours.
Collapse
Affiliation(s)
- Peter Bouwman
- Division of Molecular Pathology and Cancer Systems Biology Center, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | |
Collapse
|
146
|
Zander SAL, Kersbergen A, Sol W, Gonggrijp M, van de Wetering K, Jonkers J, Borst P, Rottenberg S. Lack of ABCG2 shortens latency of BRCA1-deficient mammary tumors and this is not affected by genistein or resveratrol. Cancer Prev Res (Phila) 2012; 5:1053-60. [PMID: 22767648 DOI: 10.1158/1940-6207.capr-12-0050] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In addition to their role in drug resistance, the ATP-binding cassette (ABC) transporters ABCG2 and ABCB1 have been suggested to protect cells from a broad range of substances that may foster tumorigenesis. Phytoestrogens or their metabolites are substrates of these transporters and the influence of these compounds on breast cancer development is controversial. Estrogen-like properties might accelerate tumorigenesis on the one hand, whereas their proposed health-protective properties might antagonize tumorigenesis on the other. To address this issue, we used a newer generation mouse model of BRCA1-mutated breast cancer and examined tumor latency in K14cre;Brca1(F/F); p53(F/F), Abcb1a/b(-/-);K14cre;Brca1(F/F); p53(F/F), or Abcg2(-/-);K14cre;Brca1(F/F); p53(F/F) animals, fed with genistein- or resveratrol-supplemented diets. Ovariectomized K14cre;Brca1(F/F); p53(F/F) animals were included to evaluate whether any estrogen-mimicking effects can restore mammary tumor development in the absence of endogenous estrogens. Compared with the ABC transporter proficient model, ABCG2-deficient animals showed a reduced median tumor latency of 17.5 days (P < 0.001), whereas no significant difference was observed for ABCB1-deficient animals. Neither genistein nor resveratrol altered this latency reduction in Abcg2(-/-);K14cre;Brca1(F/F); p53(F/F) animals. Ovariectomy resulted in nearly complete loss of mammary tumor development, which was not restored by genistein or resveratrol. Our results show that ABCG2 contributes to the protection of genetically instable epithelial cells against carcinogenesis. Diets containing high levels of genistein or resveratrol had no effect on mammary tumorigenesis, whether mice were lacking ABCG2 or not. Because genistein and resveratrol only delayed skin tumor development of ovariectomized animals, we conclude that these phytoestrogens are no effective modulators of mammary tumor development in our mouse model.
Collapse
Affiliation(s)
- Serge A L Zander
- Division of Molecular Oncology, The Netherlands Cancer Institute (Antoni van Leeuwenhoek Hospital), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
147
|
Meiser B, Gleeson M, Watts K, Peate M, Zilliacus E, Barlow-Stewart K, Saunders C, Mitchell G, Kirk J. Getting to the point: what women newly diagnosed with breast cancer want to know about treatment-focused genetic testing. Oncol Nurs Forum 2012; 39:E101-11. [PMID: 22374498 DOI: 10.1188/12.onf.e101-e111] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE/OBJECTIVES To identify young women's information preferences regarding treatment-focused genetic testing (TFGT) and to develop and evaluate a novel educational resource. RESEARCH APPROACH Qualitative interview study and pilot testing of a novel resource. SETTING Two familial cancer services and one outpatient oncology clinic in Sydney and Melbourne, Australia. PARTICIPANTS 26 women with breast cancer aged 50 years and younger who either previously had TFGT (n = 14) or had a diagnosis of breast cancer within the previous 6-12 months. METHODOLOGIC APPROACH Participants were asked about their views of TFGT in semistructured interviews. A brief pamphlet on TFGT then was developed and pilot tested with 17 of the 26 women. MAIN RESEARCH VARIABLES Women's attitudes and preferences with regard to timing, mode of delivery, and amount and format of information regarding TFGT were explored. FINDINGS Most women wanted to be informed about TFGT at or around the time of their cancer diagnosis via a face-to-face consultation. No clear preference existed for which type of healthcare professional should provide information on TFGT. Brief written information about TFGT was viewed as important supporting material. The educational resource developed was well received. CONCLUSIONS The potential for more widespread TFGT in the future indicates a need for patient educational materials that enable women to make informed choices about TFGT. This pilot study has provided timely initial evidence on the efficacy of a brief written resource in preparing women for decision making about TFGT. INTERPRETATION The resource developed in this study will assist oncology nurses to make important genetic risk information available to women newly diagnosed with breast cancer at a stressful time.
Collapse
Affiliation(s)
- Bettina Meiser
- Department of Medical Oncology at Prince of Wales Hospital, Prince of Wales Clinical School at University of New South Wales, Sydney, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Wang S, Li W, Xue Z, Lu Y, Narsinh K, Fan W, Li X, Bu Q, Wang F, Liang J, Wu K, Cao F. Molecular imaging of p53 signal pathway in lung cancer cell cycle arrest induced by cisplatin. Mol Carcinog 2012; 52:900-7. [PMID: 22674879 DOI: 10.1002/mc.21930] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 05/03/2012] [Accepted: 05/08/2012] [Indexed: 01/03/2023]
Abstract
Cisplatin is a commonly employed chemotherapy drug for lung malignancy. However its efficacy is limited by acquired drug resistance and lacking of an in vivo real-time monitoring approach. The aim of this study is to investigate the effect of cisplatin on lung adenocarcinoma cell line p53-RE-Fluc/A549 in vivo via non-invasive reporter gene by molecular imaging. For this study, we employed p53-RE-Fluc/A549 cells that overexpressed a vector with three tandem repeats of p53 response element followed by the luciferase reporter gene. P53 activity was evaluated by optical imaging and verified by Western blot after cells were exposed to 10 µM cisplatin for 72 h. The cell cycle was mainly blocked at the S- and G2/M-phases after cisplatin treatment, whereas no significant change was observed in cell apoptotic index. Increased expression of p21 and Bcl-2 as well as decreased expression of Bax were observed after cisplatin treatment by Western blotting. Longitudinal in vivo bioluminescent imaging (BLI) revealed that the p53 activity was increased from 24 to 48 h after transient cisplatin treatment in p53-RE-Fluc/A549-bearing nude mice. RNA sequencing further revealed that cell cycle and p53 signaling pathway genes, such as E2F1, CCNA2, CDK1, and CCNE2 were significantly downregulated after long-term cisplatin treatment. Thus, our study showed that cisplatin exerts its cytotoxic effect through blockage of the cell cycle and may be partly regulated by the p53 signaling pathway. Furthermore, molecular imaging is a useful tool to investigate the mechanism and evaluate the effect of chemotherapy drugs both in vivo and in vitro.
Collapse
Affiliation(s)
- Shenxu Wang
- Cardiology and Molecular Imaging Department, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Singh M, Murriel CL, Johnson L. Genetically engineered mouse models: closing the gap between preclinical data and trial outcomes. Cancer Res 2012; 72:2695-700. [PMID: 22593194 DOI: 10.1158/0008-5472.can-11-2786] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The high failure rate of late-stage human clinical trials, particularly in oncology, predicates the need for improved translation of preclinical data from mouse tumor models into clinical predictions. Genetically engineered mouse models (GEMM) may fulfill this need, because they mimic spontaneous and autochthonous disease progression. Using oncogenic Kras-driven GEMMs of lung and pancreatic adenocarcinoma, we recently showed that these models can closely phenocopy human therapeutic responses to standard-of-care treatment regimens. Here we review the successful preclinical application of such GEMMs, as well as the potential for discovering predictive biomarkers and gaining mechanistic insights into clinical outcomes and drug resistance in human cancers.
Collapse
Affiliation(s)
- Mallika Singh
- Genentech, Inc., South San Francisco, California 94080, USA.
| | | | | |
Collapse
|
150
|
Borst P. Cancer drug pan-resistance: pumps, cancer stem cells, quiescence, epithelial to mesenchymal transition, blocked cell death pathways, persisters or what? Open Biol 2012; 2:120066. [PMID: 22724067 PMCID: PMC3376736 DOI: 10.1098/rsob.120066] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 04/27/2012] [Indexed: 12/11/2022] Open
Abstract
Although chemotherapy of tumours has scored successes, drug resistance remains the major cause of death of cancer patients. Initial treatment often leaves residual disease, from which the tumour regrows. Eventually, most tumours become resistant to all available chemotherapy. I call this pan-resistance to distinguish it from multi-drug resistance, usually describing resistance caused by upregulation of drug transporters, such as P-glycoprotein. In this review, I discuss mechanisms proposed to explain both residual disease and pan-resistance. Although plausible explanations are at hand for residual disease, pan-resistance is still a mystery. My conclusion is that it is time for a major effort to solve this mystery using the new genetically modified mouse tumour models that produce real tumours resembling cancer in human patients.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology
- ATP-Binding Cassette Transporters/physiology
- Animals
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Biological Availability
- Blood-Brain Barrier
- Cell Cycle
- Cell Death/drug effects
- Chromatin Assembly and Disassembly
- Clonal Evolution
- DNA Repair
- DNA, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/physiology
- Epigenesis, Genetic
- Epithelial-Mesenchymal Transition
- Humans
- Mice
- Models, Biological
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/physiology
- Neoplasm, Residual
- Neoplasms, Experimental/drug therapy
- Neoplastic Stem Cells/cytology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
Collapse
Affiliation(s)
- Piet Borst
- Molecular Oncology , NKI-AVL , Plesmanlaan 121, Amsterdam, The Netherlands.
| |
Collapse
|