501
|
|
502
|
Goetz MP, Toi M, Campone M, Sohn J, Paluch-Shimon S, Huober J, Park IH, Trédan O, Chen SC, Manso L, Freedman OC, Garnica Jaliffe G, Forrester T, Frenzel M, Barriga S, Smith IC, Bourayou N, Di Leo A. MONARCH 3: Abemaciclib As Initial Therapy for Advanced Breast Cancer. J Clin Oncol 2017; 35:3638-3646. [DOI: 10.1200/jco.2017.75.6155] [Citation(s) in RCA: 790] [Impact Index Per Article: 98.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Abemaciclib, a cyclin-dependent kinase 4 and 6 inhibitor, demonstrated efficacy as monotherapy and in combination with fulvestrant in women with hormone receptor (HR)–positive, human epidermal growth factor receptor 2 (HER2)–negative advanced breast cancer previously treated with endocrine therapy. Methods MONARCH 3 is a double-blind, randomized phase III study of abemaciclib or placebo plus a nonsteroidal aromatase inhibitor in 493 postmenopausal women with HR-positive, HER2-negative advanced breast cancer who had no prior systemic therapy in the advanced setting. Patients received abemaciclib or placebo (150 mg twice daily continuous schedule) plus either 1 mg anastrozole or 2.5 mg letrozole, daily. The primary objective was investigator-assessed progression-free survival. Secondary objectives included response evaluation and safety. A planned interim analysis occurred after 189 events. Results Median progression-free survival was significantly prolonged in the abemaciclib arm (hazard ratio, 0.54; 95% CI, 0.41 to 0.72; P = .000021; median: not reached in the abemaciclib arm, 14.7 months in the placebo arm). In patients with measurable disease, the objective response rate was 59% in the abemaciclib arm and 44% in the placebo arm ( P = .004). In the abemaciclib arm, diarrhea was the most frequent adverse effect (81.3%) but was mainly grade 1 (44.6%). Comparing abemaciclib and placebo, the most frequent grade 3 or 4 adverse events were neutropenia (21.1% v 1.2%), diarrhea (9.5% v 1.2%), and leukopenia (7.6% v 0.6%). Conclusion Abemaciclib plus a nonsteroidal aromatase inhibitor was effective as initial therapy, significantly improving progression-free survival and objective response rate and demonstrating a tolerable safety profile in women with HR-positive, HER2-negative advanced breast cancer.
Collapse
Affiliation(s)
- Matthew P. Goetz
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Masakazu Toi
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Mario Campone
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Joohyuk Sohn
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Shani Paluch-Shimon
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Jens Huober
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - In Hae Park
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Olivier Trédan
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Shin-Cheh Chen
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Luis Manso
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Orit C. Freedman
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Georgina Garnica Jaliffe
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Tammy Forrester
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Martin Frenzel
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Susana Barriga
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Ian C. Smith
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Nawel Bourayou
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| | - Angelo Di Leo
- Matthew P. Goetz, Mayo Clinic, Rochester, MN; Masakazu Toi, Kyoto University, Kyoto, Japan; Mario Campone, Institut de Cancerologie de l’Ouest, Angers Cedex; Olivier Trédan, Centre Léon Bérard, Lyon; Nawel Bourayou, Eli Lilly, Paris, France; Joohyuk Sohn, Yonsei Cancer Center, Seoul; In Hae Park, National Cancer Center, Goyangsi, South Korea; Shani Paluch-Shimon, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Jens Huober, University of Ulm, Ulm, Germany; Shin-Cheh Chen, Chang Gung University
| |
Collapse
|
503
|
Lim JSJ, Turner NC, Yap TA. CDK4/6 Inhibitors: Promising Opportunities beyond Breast Cancer. Cancer Discov 2017; 6:697-9. [PMID: 27371575 DOI: 10.1158/2159-8290.cd-16-0563] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Patnaik and colleagues report on the safety, pharmacokinetics, pharmacodynamics, and preliminary efficacy of abemaciclib for the treatment of advanced solid cancers, demonstrating antitumor activity in advanced breast cancers as well as glioblastoma, melanoma, non-small cell lung cancer, colorectal cancer, and ovarian cancer. The development of abemaciclib and other CDK4/6 inhibitors should now be fully optimized through the use of novel predictive biomarkers of response and rational combinations. Cancer Discov; 6(7); 697-9. ©2016 AACRSee related article by Patnaik et al., p. 740.
Collapse
Affiliation(s)
- Joline S J Lim
- The Institute of Cancer Research and Royal Marsden Hospital, London, United Kingdom
| | - Nicholas C Turner
- The Institute of Cancer Research and Royal Marsden Hospital, London, United Kingdom
| | - Timothy A Yap
- The Institute of Cancer Research and Royal Marsden Hospital, London, United Kingdom.
| |
Collapse
|
504
|
Mills CC, Kolb EA, Sampson VB. Recent Advances of Cell-Cycle Inhibitor Therapies for Pediatric Cancer. Cancer Res 2017; 77:6489-6498. [PMID: 29097609 DOI: 10.1158/0008-5472.can-17-2066] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/24/2017] [Accepted: 09/19/2017] [Indexed: 12/21/2022]
Abstract
This review describes the pivotal roles of cell-cycle and checkpoint regulators and discusses development of specific cell-cycle inhibitors for therapeutic use for pediatric cancer. The mechanism of action as well as the safety and tolerability of drugs in pediatric patients, including compounds that target CDK4/CDK6 (palbociclib, ribociclib, and abemaciclib), aurora kinases (AT9283 and MLN8237), Wee1 kinase (MK-1775), KSP (ispinesib), and tubulin (taxanes, vinca alkaloids), are presented. The design of mechanism-based combinations that exploit the cross-talk of signals activated by cell-cycle arrest, as well as pediatric-focused drug development, are critical for the advancement of drugs for rare childhood diseases. Cancer Res; 77(23); 6489-98. ©2017 AACR.
Collapse
Affiliation(s)
| | - E A Kolb
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Valerie B Sampson
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware.
| |
Collapse
|
505
|
Chen S, Guan X, Wang LL, Li B, Sang XB, Liu Y, Zhao Y. Fascaplysin inhibit ovarian cancer cell proliferation and metastasis through inhibiting CDK4. Gene 2017; 635:3-8. [DOI: 10.1016/j.gene.2017.09.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 08/30/2017] [Accepted: 09/05/2017] [Indexed: 12/29/2022]
|
506
|
de Groot A, Kuijpers C, Kroep J. CDK4/6 inhibition in early and metastatic breast cancer: A review. Cancer Treat Rev 2017; 60:130-138. [DOI: 10.1016/j.ctrv.2017.09.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/08/2017] [Accepted: 09/09/2017] [Indexed: 10/18/2022]
|
507
|
Guarducci C, Bonechi M, Boccalini G, Benelli M, Risi E, Di Leo A, Malorni L, Migliaccio I. Mechanisms of Resistance to CDK4/6 Inhibitors in Breast Cancer and Potential Biomarkers of Response. Breast Care (Basel) 2017; 12:304-308. [PMID: 29234249 DOI: 10.1159/000484167] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Randomized clinical trials demonstrated that CDK4/6 inhibitors are highly effective in patients with hormone receptor-positive (HR+), HER2-negative (HER2-) metastatic breast cancer in combination with endocrine therapy. The use of CDK4/6 inhibitors in clinics is becoming common for patients with HR+/HER2- metastatic breast cancer and will certainly increase in the near future. However, patients might show de novo or acquired resistance to these drugs. Molecular alterations have been suggested as determinants for de novo resistance to CDK4/6 inhibitors, but have never been validated in a clinical setting. In addition, molecular mechanisms of acquired resistance to palbociclib have been analyzed only in preclinical studies. Here we review the current knowledge on the available preclinical data about the mechanisms of de novo and acquired resistance to CDK4/6 inhibitors in breast cancer, and clinical data about potential biomarkers of response.
Collapse
Affiliation(s)
- Cristina Guarducci
- 'Sandro Pitigliani' Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Martina Bonechi
- 'Sandro Pitigliani' Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Giulia Boccalini
- 'Sandro Pitigliani' Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Matteo Benelli
- 'Sandro Pitigliani' Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Emanuela Risi
- 'Sandro Pitigliani' Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Angelo Di Leo
- 'Sandro Pitigliani' Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Luca Malorni
- 'Sandro Pitigliani' Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy.,'Sandro Pitigliani' Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Ilenia Migliaccio
- 'Sandro Pitigliani' Medical Oncology Department, Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| |
Collapse
|
508
|
Zardavas D, Pondé N, Tryfonidis K. CDK4/6 blockade in breast cancer: current experience and future perspectives. Expert Opin Investig Drugs 2017; 26:1357-1372. [PMID: 29027483 DOI: 10.1080/13543784.2017.1389896] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Dysregulated cellular proliferation, one of the hallmarks of cancer, is mediated by aberrant activation of the cell cycle machinery through the biological effects of cyclin-dependent kinases (CDKs). The clinical development of non-selective CDK inhibitors failed due to combined lack of efficacy and excessive toxicity reported by clinical trials across different cancer types. The clinical development of second generation, CDK4/6-selective inhibitors, namely palbociclib, abemaciclib and ribociclib, led to practice-changing results in the setting of breast cancer. Areas covered: This review illustrates how CDK4/6-selective inhibitors got approval for the treatment of patients with either newly diagnosed or pretreated advanced hormone receptor positive, HER2-negative breast cancer. Furthermore, data about potential predictive biomarkers, as well as preclinical and preliminary clinical evidence for potential antitumor activity of CDK4/6 inhibition in other breast cancer subtypes is provided. Expert opinion: Future clinical development of CDK4/6 inhibitors in breast cancer will focus on the following aspects: i) optimization of treatment sequencing for patients with advanced disease, ii) early-stage disease, iii) other subtypes of breast cancer in rationally chosen therapeutic combinations and iv) the identification of predictive biomarkers.
Collapse
Affiliation(s)
- Dimitrios Zardavas
- a Medical Department , Breast International Group (BIG) , Brussels , Belgium
| | - Noam Pondé
- b Institut Jules Bordet , Université Libre de Bruxelles (ULB) , Brussels , Belgium
| | - Konstantinos Tryfonidis
- c Medical Department , European Organization for Research and Treatment of Cancer (EORTC) , Brussels , Belgium
| |
Collapse
|
509
|
Chen SH, Gong X, Zhang Y, Van Horn RD, Yin T, Huber L, Burke TF, Manro J, Iversen PW, Wu W, Bhagwat SV, Beckmann RP, Tiu RV, Buchanan SG, Peng SB. RAF inhibitor LY3009120 sensitizes RAS or BRAF mutant cancer to CDK4/6 inhibition by abemaciclib via superior inhibition of phospho-RB and suppression of cyclin D1. Oncogene 2017; 37:821-832. [PMID: 29059158 DOI: 10.1038/onc.2017.384] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 08/21/2017] [Accepted: 09/07/2017] [Indexed: 12/15/2022]
Abstract
KRAS, NRAS and BRAF mutations are among the most important oncogenic drivers in many major cancer types, such as melanoma, lung, colorectal and pancreatic cancer. There is currently no effective therapy for the treatment of RAS mutant cancers. LY3009120, a pan-RAF and RAF dimer inhibitor advanced to clinical study has been shown to inhibit both RAS and BRAF mutant cell proliferation in vitro and xenograft tumor growth in vivo. Abemaciclib, a CDK4/6-selective inhibitor, is currently in phase III studies for ER-positive breast cancer and KRAS mutant lung cancer. In this study, we found that combinatory treatment with LY3009120 and abemaciclib synergistically inhibited proliferation of tumor cells in vitro and led to tumor growth regression in xenograft models with a KRAS, NRAS or BRAF mutation at the doses of two drugs that were well tolerated in combination. Further in vitro screen in 328 tumor cell lines revealed that tumor cells with KRAS, NRAS or BRAF mutation, or cyclin D activation are more sensitive, whereas tumor cells with PTEN, PIK3CA, PIK3R1 or retinoblastoma (Rb) mutation are more resistant to this combination treatment. Molecular analysis revealed that abemaciclib alone inhibited Rb phosphorylation partially and caused an increase of cyclin D1. The combinatory treatment cooperatively demonstrated more complete inhibition of Rb phosphorylation, and LY3009120 suppressed the cyclin D1 upregulation mediated by abemaciclib. These results were further verified by CDK4/6 siRNA knockdown. Importantly, the more complete phospho-Rb inhibition and cyclin D1 suppression by LY3009120 and abemaciclib combination led to more significant cell cycle G0/G1 arrest of tumor cells. These preclinical findings suggest that combined inhibition of RAF and d-cyclin-dependent kinases might provide an effective approach to treat patients with tumors harboring mutations in RAS or RAF genes.
Collapse
Affiliation(s)
- S-H Chen
- Oncology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | - X Gong
- Oncology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | - Y Zhang
- Oncology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | - R D Van Horn
- Oncology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | - T Yin
- Oncology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | - L Huber
- Oncology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | - T F Burke
- Oncology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | - J Manro
- Statistics, Eli Lilly and Company, Indianapolis, IN, USA
| | - P W Iversen
- Statistics, Eli Lilly and Company, Indianapolis, IN, USA
| | - W Wu
- Oncology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | - S V Bhagwat
- Oncology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | - R P Beckmann
- Oncology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | - R V Tiu
- Early Phase Oncology and Oncology Business Unit, Eli Lilly and Company, Indianapolis, IN, USA
| | - S G Buchanan
- Oncology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | - S-B Peng
- Oncology Research, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
510
|
Echavarria I, Jerez Y, Martin M, López-Tarruella S. Incorporating CDK4/6 Inhibitors in the Treatment of Advanced Luminal Breast Cancer. Breast Care (Basel) 2017; 12:296-302. [PMID: 29234248 PMCID: PMC5704690 DOI: 10.1159/000481656] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
After optimizing endocrine monotherapy modalities in the setting of advanced luminal breast cancer (BC), dual endocrine/targeted therapy combinations have been tested with positive results, and are transforming this BC subtype treatment landscape. Cell cycle deregulation is a hallmark of cancer that has become a key druggable target in hormone receptor (HR)-positive BC due to its role in endocrine resistance mechanisms. Cyclin dependent kinase (CDK)4/6 inhibitors have experienced a fast development in combination with endocrine therapy and have already been commercialized in some countries. In this review, we will summarize the development of these CDK4/6 inhibitors in luminal BC, from the preclinical data to the pivotal phase III trials that led to their approval, focusing on the efficacy and safety data for each of the treatment settings. Moreover, we will consider the challenges CDK4/6 inhibitors face in their positioning in the algorithm of treatment for advanced luminal BC and the considerations physicians should take into account when selecting these therapies for their patients. However, we are still in need of reliable predictive biomarkers in order to identify patients who will derive the greatest benefit from these drug combinations that are not exempt from toxicity.
Collapse
Affiliation(s)
- Isabel Echavarria
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), Universidad Complutense de Madrid, CiberOnc, Madrid, Spain
| | | | | | | |
Collapse
|
511
|
Daoud A, Chu QS. Targeting Novel but Less Common Driver Mutations and Chromosomal Translocations in Advanced Non-Small Cell Lung Cancer. Front Oncol 2017; 7:222. [PMID: 29034207 PMCID: PMC5626928 DOI: 10.3389/fonc.2017.00222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 09/01/2017] [Indexed: 01/25/2023] Open
Abstract
Discovery of the epidermal growth factor receptor gene mutation and the anaplastic lymphoma kinase chromosomal translocation in non-small cell lung cancer has prompted efforts around the world to identify many less common targetable oncogenic drivers. Such concerted efforts have been variably successful in both non-squamous and squamous cell carcinomas of the lung. Some of the targeted therapies for these oncogenic drivers have received regulatory approval for clinical use, while others have modest clinical benefit. In this mini-review, several of these targets will be reviewed.
Collapse
Affiliation(s)
- Alia Daoud
- Department of Medical Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Quincy S. Chu
- Department of Medical Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
512
|
An Y, Wang H, Jie J, Tang Y, Zhang W, Ji S, Guo X. Identification of distinct molecular subtypes of uterine carcinosarcoma. Oncotarget 2017; 8:15878-15886. [PMID: 28178664 PMCID: PMC5362530 DOI: 10.18632/oncotarget.15032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 01/06/2017] [Indexed: 12/14/2022] Open
Abstract
Uterine carcinosarcoma (UCS) is a rare but lethal neoplasm with high metastasis and recurrence rate, and to date, no molecular classification of UCS has been defined to achieve targeted therapies. In this study, we identified two distinct molecular subtypes of UCS with distinct gene expression patterns and clinicopathologic characteristics. Subtype I UCS recapitulates low-grade UCS, in contrast subtype II UCS represents high-grade UCS with higher tumor invasion rate and tumor weight. Interestingly, subtype I UCS is characterized by cell adhesion and apoptosis pathways, whereas genes over-expressed in subtype II UCS are more involved in myogenesis/muscle development. We also proposed certain potential subtype specific therapeutic targets, such as SYK (spleen tyrosine kinase) for subtype I and cell-cycle proteins for subtype II. Our findings provide a better recognition of UCS molecular subtypes and subtype specific oncogenesis mechanisms, and can help develop more specific targeted treatment options for these tumors.
Collapse
Affiliation(s)
- Yang An
- Department of Biochemistry and Molecular Biology, Medical School, Henan University, Kaifeng 475004, China.,Cell Signal Transduction Laboratory, Henan University, Kaifeng 475004, China
| | - Haojie Wang
- Department of Biochemistry and Molecular Biology, Medical School, Henan University, Kaifeng 475004, China.,Cell Signal Transduction Laboratory, Henan University, Kaifeng 475004, China
| | - Jingyao Jie
- Department of Biochemistry and Molecular Biology, Medical School, Henan University, Kaifeng 475004, China.,Cell Signal Transduction Laboratory, Henan University, Kaifeng 475004, China
| | - Yitai Tang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Weijuan Zhang
- Department of Biochemistry and Molecular Biology, Medical School, Henan University, Kaifeng 475004, China.,Cell Signal Transduction Laboratory, Henan University, Kaifeng 475004, China
| | - Shaoping Ji
- Department of Biochemistry and Molecular Biology, Medical School, Henan University, Kaifeng 475004, China.,Cell Signal Transduction Laboratory, Henan University, Kaifeng 475004, China.,Department of Oncology, The First Affiliated Hospital of Henan University, Kaifeng, 475001, China
| | - Xiangqian Guo
- Department of Biochemistry and Molecular Biology, Medical School, Henan University, Kaifeng 475004, China.,Cell Signal Transduction Laboratory, Henan University, Kaifeng 475004, China.,Department of Preventive Medicine, Medical School, Henan University, Kaifeng 475004, China.,Department of Burn and Plastic Surgery, The Affiliated Nanshi Hospital of Henan University, Nanyang, 473003, China
| |
Collapse
|
513
|
Sacco PC, Gridelli C. An update on the developing mitotic inhibitors for the treatment of non-small cell carcinoma. Expert Opin Emerg Drugs 2017; 22:213-222. [PMID: 28836854 DOI: 10.1080/14728214.2017.1369952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Mitosis is necessary to sustain life and is followed immediately by cell division into two daughter cells. Microtubules play a key role in the formation of the mitotic spindle apparatus and cytokinesis at the end of mitosis. Various anti-microtubule agents such as taxanes and vinca alkaloids are widely used in the treatment of advanced non-small cell lung cancer (NSCLC) but their use is associated with hematologic toxicity profile, acquired resistance and hypersensitivity reactions. Areas covered: The Nab-paclitaxels are the more recent antimitotic agents approved in NSCLC showing a better tolerability and activity when compared to previous ones. Despite this, the outcome of patients with advanced non-small cell lung cancer is poor. Due to the key role of mitosis, research is focused on the identification of new mitotic drug targets other than microtubule inhibitors, such as cell cycle targets, aurora kinases and Polo-like kinases. Expert opinion: Despite improvements in chemotherapeutic choices and supportive care, the majority of patients experience a deteriorating quality of life and significant toxicities associated to a poor outcome. Thus, the therapeutic management of patients with advanced NSCLC represents an ongoing challenge and novel agents targeting mitosis are under investigation.
Collapse
Affiliation(s)
| | - Cesare Gridelli
- a Division of Medical Oncology , 'S.G. Moscati' Hospital , Avellino , Italy
| |
Collapse
|
514
|
Zangardi ML, Spring LM, Blouin GC, Bardia A. Ribociclib for post-menopausal women with HR+/HER2- advanced or metastatic breast cancer. Expert Rev Clin Pharmacol 2017; 10:1169-1176. [DOI: 10.1080/17512433.2017.1376653] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Mark L. Zangardi
- Ambulatory Oncology, Department of Pharmacy, Massachusetts General Hospital, Boston, MA, USA
| | - Laura M. Spring
- Department of Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Gayle C. Blouin
- Ambulatory Oncology, Department of Pharmacy, Massachusetts General Hospital, Boston, MA, USA
| | - Aditya Bardia
- Department of Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
515
|
Iriyama N, Hino H, Moriya S, Hiramoto M, Hatta Y, Takei M, Miyazawa K. The cyclin-dependent kinase 4/6 inhibitor, abemaciclib, exerts dose-dependent cytostatic and cytocidal effects and induces autophagy in multiple myeloma cells. Leuk Lymphoma 2017; 59:1439-1450. [DOI: 10.1080/10428194.2017.1376741] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Noriyoshi Iriyama
- Department of Medicine, Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Hirotsugu Hino
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Shota Moriya
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Masaki Hiramoto
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Yoshihiro Hatta
- Department of Medicine, Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Masami Takei
- Department of Medicine, Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Keisuke Miyazawa
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
516
|
Griggs JJ, Wolff AC. Cyclin-Dependent Kinase 4/6 Inhibitors in the Treatment of Breast Cancer: More Breakthroughs and an Embarrassment of Riches. J Clin Oncol 2017; 35:2857-2859. [DOI: 10.1200/jco.2017.73.9375] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Jennifer J. Griggs
- Jennifer J. Griggs, University of Michigan Cancer Center, Ann Arbor, MI; and Antonio C. Wolff, The Johns Hopkins Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Antonio C. Wolff
- Jennifer J. Griggs, University of Michigan Cancer Center, Ann Arbor, MI; and Antonio C. Wolff, The Johns Hopkins Kimmel Comprehensive Cancer Center, Baltimore, MD
| |
Collapse
|
517
|
Abstract
The ability to sustain unscheduled proliferation is a hallmark of cancer. The normal process of cell division occurs via the cell cycle, a series of highly regulated steps that are orchestrated at the molecular level by specific cyclins that act in association with cyclin-dependent kinases (CDKs). Cyclin D and CDK4/6 play a key role in cell-cycle progression by phosphorylating and inactivating the retinoblastoma protein, a tumor suppressor that restrains G1- to S-phase progression. The first-generation CDK inhibitors demonstrated broad activity upon several CDKs, which likely explains their considerable toxicities and limited efficacy. Palbociclib, ribociclib, and abemaciclib represent a new class of highly specific ATP-competitive CDK4/6 inhibitors that induce reversible G1-phase cell-cycle arrest in retinoblastoma-positive tumor models. Both palbociclib and ribociclib have been approved in combination with hormone-based therapy for the treatment of naïve hormone receptor-positive advanced breast cancer on the basis of an improvement in progression-free survival. In general, CDK4/6 inhibitors are cytostatic as monotherapy but demonstrate favorable tolerability, which has prompted interest in combination approaches. Combinations with phosphatidylinositol 3-kinase and mammalian target of rapamycin inhibitors in breast cancer, and inhibitors of the RAS/RAF/mitogen-activated protein kinase pathway in RAS-mutant cancers are particularly promising approaches that are currently being evaluated. Although the subject of intense preclinical study, predictive biomarkers for response and resistance to these drugs remain largely undefined. CDK4/6 inhibitors have emerged as the most promising of the cell-cycle therapeutics and intense efforts are now underway to expand the reach of this paradigm.
Collapse
Affiliation(s)
- Matthew Ingham
- All authors: Columbia University School of Medicine, New York, NY
| | - Gary K. Schwartz
- All authors: Columbia University School of Medicine, New York, NY
| |
Collapse
|
518
|
Sabari JK, Paik PK. Relevance of genetic alterations in squamous and small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:373. [PMID: 29057233 PMCID: PMC5635252 DOI: 10.21037/atm.2017.06.72] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/22/2017] [Indexed: 01/10/2023]
Abstract
The precision medicine revolution has led to the development and US FDA approval of multiple targeted therapies in non-squamous non-small cell lung cancers, including tyrosine kinase inhibitors targeting EGFR, ALK, and ROS1. However, the development of targeted therapies for squamous cell lung cancers (SQCLCs) and small cell lung cancers (SCLCs) has lagged behind and the mainstay of systemic therapy for most patients with metastatic disease remains chemotherapy; which has seen little meaningful progress over the past three decades. The ideal of precision medicine in these diseases may appear elusive; however, recent comprehensive genomic analysis of SQCLC and SCLC has led to multiple breakthroughs in our understanding of the biology of these diseases and has led to new therapeutic approaches currently under active clinical investigation. This review will focus on the therapeutic relevance of these alterations in their respective diseases and new insights into promising therapeutics currently under investigation.
Collapse
Affiliation(s)
- Joshua K. Sabari
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul K. Paik
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
519
|
Bilgin B, Sendur MAN, Şener Dede D, Akıncı MB, Yalçın B. A current and comprehensive review of cyclin-dependent kinase inhibitors for the treatment of metastatic breast cancer. Curr Med Res Opin 2017; 33:1559-1569. [PMID: 28657360 DOI: 10.1080/03007995.2017.1348344] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Resistance to endocrine treatment generally occurs over time, especially in the metastatic stage. In this paper, we aimed to review the mechanisms of cyclin-dependent kinase (CDK) 4/6 inhibition and clinical usage of new agents in the light of recent literature updates. SCOPE A literature search was carried out using PubMed, Medline and ASCO and ESMO annual-meeting abstracts by using the following search keywords; "palbociclib", "abemaciclib", "ribociclib", "cyclin-dependent kinase inhibitors" and "CDK 4/6" in metastatic breast cancer (MBC). The last search was on 10 June 2017. FINDINGS CDKs and cyclins are two molecules that have a key role in cell cycle progression. Today, there are three highly selective CDK4/6 inhibitors in clinical development - palbociclib, ribociclib and abemaciclib. Palbociclib and ribociclib were recently approved by the US FDA in combination with letrozole for the treatment of MBC in a first-line setting, as well as palbociclib in combination with fulvestrant for hormone-receptor (HR)-positive MBC that had progressed while on previous endocrine therapy according to the PALOMA-1, MONALEESA-2 and PALOMA-3 trials, respectively. In the recently published randomized phase III MONARCH 2 trial, abemaciclib plus letrozole had longer progression-free survival and higher objective response rates with less serious adverse events in advanced HR-positive breast cancer previously treated with hormonal treatment. CONCLUSION CDK4/6 inhibition is a new and promising target for patients with hormone-receptor-positive MBC. Both palbociclib and ribociclib showed significant additive benefit for patients receiving first-line treatment for HR-positive, epidermal growth factor receptor-2-negative advanced breast cancer. Palbociclib and abemaciclib also had significant activity in combination with fulvestrant for patients with MBC that progressed on previous endocrine therapy.
Collapse
Affiliation(s)
- Burak Bilgin
- a Ankara Yıldırım Beyazıt University , Faculty of Medicine, Department of Medical Oncology , Ankara , Turkey
| | - Mehmet A N Sendur
- a Ankara Yıldırım Beyazıt University , Faculty of Medicine, Department of Medical Oncology , Ankara , Turkey
| | - Didem Şener Dede
- a Ankara Yıldırım Beyazıt University , Faculty of Medicine, Department of Medical Oncology , Ankara , Turkey
| | - Muhammed Bülent Akıncı
- a Ankara Yıldırım Beyazıt University , Faculty of Medicine, Department of Medical Oncology , Ankara , Turkey
| | - Bülent Yalçın
- a Ankara Yıldırım Beyazıt University , Faculty of Medicine, Department of Medical Oncology , Ankara , Turkey
| |
Collapse
|
520
|
Sledge GW, Toi M, Neven P, Sohn J, Inoue K, Pivot X, Burdaeva O, Okera M, Masuda N, Kaufman PA, Koh H, Grischke EM, Frenzel M, Lin Y, Barriga S, Smith IC, Bourayou N, Llombart-Cussac A. MONARCH 2: Abemaciclib in Combination With Fulvestrant in Women With HR+/HER2− Advanced Breast Cancer Who Had Progressed While Receiving Endocrine Therapy. J Clin Oncol 2017; 35:2875-2884. [DOI: 10.1200/jco.2017.73.7585] [Citation(s) in RCA: 824] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Purpose MONARCH 2 ( ClinicalTrials.gov identifier: NCT02107703) compared the efficacy and safety of abemaciclib, a selective cyclin-dependent kinase 4 and 6 inhibitor, plus fulvestrant with fulvestrant alone in patients with advanced breast cancer (ABC). Patients and Methods MONARCH 2 was a global, double-blind, phase III study of women with hormone receptor-positive and human epidermal growth factor receptor 2-negative ABC who had progressed while receiving neoadjuvant or adjuvant endocrine therapy (ET), ≤ 12 months from the end of adjuvant ET, or while receiving first-line ET for metastatic disease. Patients were randomly assigned 2:1 to receive abemaciclib or placebo (150 mg twice daily) on a continuous schedule and fulvestrant (500 mg, per label). The primary end point was investigator-assessed progression-free survival (PFS), and key secondary end points included overall survival, objective response rate (ORR), duration of response, clinical benefit rate, quality of life, and safety. Results Between August 2014 and December 2015, 669 patients were randomly assigned to receive abemaciclib plus fulvestrant (n = 446) or placebo plus fulvestrant (n = 223). Abemaciclib plus fulvestrant significantly extended PFS versus fulvestrant alone (median, 16.4 v 9.3 months; hazard ratio, 0.553; 95% CI, 0.449 to 0.681; P < .001). In patients with measurable disease, abemaciclib plus fulvestrant achieved an ORR of 48.1% (95% CI, 42.6% to 53.6%) compared with 21.3% (95% CI, 15.1% to 27.6%) in the control arm. The most common adverse events in the abemaciclib versus placebo arms were diarrhea (86.4% v 24.7%), neutropenia (46.0% v 4.0%), nausea (45.1% v 22.9%), and fatigue (39.9% v 26.9%). Conclusions Abemaciclib at 150 mg twice daily plus fulvestrant was effective, significantly improving PFS and ORR and demonstrating a tolerable safety profile in women with hormone receptor-positive and human epidermal growth factor receptor 2-negative ABC who progressed while receiving ET.
Collapse
Affiliation(s)
- George W. Sledge
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Masakazu Toi
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Patrick Neven
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Joohyuk Sohn
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Kenichi Inoue
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Xavier Pivot
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Olga Burdaeva
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Meena Okera
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Norikazu Masuda
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Peter A. Kaufman
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Han Koh
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Eva-Maria Grischke
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Martin Frenzel
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Yong Lin
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Susana Barriga
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Ian C. Smith
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Nawel Bourayou
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| | - Antonio Llombart-Cussac
- George W. Sledge Jr., Stanford University, Stanford; Han Koh, Kaiser Permanente Medical Group, Bellflower, CA; Masakazu Toi, Kyoto University; Kenichi Inoue, Saitama Cancer Center, Saitama; Norikazu Masuda, Osaka National Hospital, Osaka, Japan; Patrick Neven, Universitaire Ziekenhuizen Leuven – Campus Gasthuisberg, Leuven, Belgium; Joohyuk Sohn, Yonsei Cancer Center, Seoul, Korea; Xavier Pivot, CHU de Besancon Hopital Jean Minjoz, Besancon Cedex; Nawel Bourayou, Eli Lilly, Paris, France; Olga Burdaeva,
| |
Collapse
|
521
|
Kong Y, Sheng X, Wu X, Yan J, Ma M, Yu J, Si L, Chi Z, Cui C, Dai J, Li Y, Yu H, Xu T, Tang H, Tang B, Mao L, Lian B, Wang X, Yan X, Li S, Guo J. Frequent Genetic Aberrations in the CDK4 Pathway in Acral Melanoma Indicate the Potential for CDK4/6 Inhibitors in Targeted Therapy. Clin Cancer Res 2017; 23:6946-6957. [PMID: 28830923 DOI: 10.1158/1078-0432.ccr-17-0070] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 06/01/2017] [Accepted: 08/16/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Effective therapies for the majority of metastatic acral melanoma patients have not been established. Thus, we investigated genetic aberrations of CDK4 pathway in acral melanoma and evaluated the efficacy of CDK4/6 inhibitors in targeted therapy of acral melanoma.Experimental Design: A total of 514 primary acral melanoma samples were examined for the copy number variations (CNV) of CDK4 pathway-related genes, including Cdk4, Ccnd1, and P16INK4a , by QuantiGenePlex DNA Assay. The sensitivity of established acral melanoma cell lines and patient-derived xenograft (PDX) containing typical CDK4 aberrations to CDK4/6 inhibitors was evaluated.Results: Among the 514 samples, 203 cases, 137 cases, and 310 cases, respectively, showed Cdk4 gain (39.5%), Ccnd1 gain (26.7%), and P16INK4a loss (60.3%). The overall frequency of acral melanomas that contain at least one aberration in Cdk4, Ccnd1, and P16INK4a was 82.7%. The median overall survival time for acral melanoma patients with concurrent Cdk4 gain with P16INK4a loss was significantly shorter than that for patients without such aberrations (P = 0.005). The pan-CDK inhibitor AT7519 and selective CDK4/6 inhibitor PD0332991 could inhibit the cell viability of acral melanoma cells and the tumor growth of PDX with Cdk4 gain plus Ccnd1 gain, Cdk4 gain plus P16INK4a loss, and Ccnd1 gain plus P16INK4a loss.Conclusions: Genetic aberration of CDK4 pathway is a frequent event in acral melanoma. Acral melanoma cell lines and PDX containing CDK4 pathway aberrations are sensitive to CDK4/6 inhibitors. Our study provides evidence for the testing of CDK4/6 inhibitors in acral melanoma patients. Clin Cancer Res; 23(22); 6946-57. ©2017 AACR.
Collapse
Affiliation(s)
- Yan Kong
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xinan Sheng
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaowen Wu
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Junya Yan
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Meng Ma
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiayi Yu
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Lu Si
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhihong Chi
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuanliang Cui
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jie Dai
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yiqian Li
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Huan Yu
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Tianxiao Xu
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Huan Tang
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Bixia Tang
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Lili Mao
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Bin Lian
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xuan Wang
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xieqiao Yan
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Siming Li
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Guo
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
522
|
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4/6) are fundamental drivers of the cell cycle and are required for the initiation and progression of various malignancies1,2. Pharmacologic inhibitors of CDK4/6 have shown significant activity against several solid tumors3,4. Their primary mechanism of action is thought to be the inhibition of phosphorylation of the retinoblastoma (RB) tumor suppressor, inducing G1 cell cycle arrest in tumor cells5. Here, we use murine models of breast carcinoma and other solid tumors to show that selective CDK4/6 inhibitors not only induce tumor cell cycle arrest, but also promote anti-tumor immunity. We confirm this phenomenon through transcriptomic analysis of serial biopsies from a clinical trial of CDK4/6 inhibitor treatment for breast cancer. The enhanced anti-tumor immune response has two underpinnings. First, CDK4/6 inhibitors activate tumor cell expression of endogenous retroviral elements, thus increasing intracellular levels of double-stranded RNA. This in turn stimulates production of type III interferons and hence enhances tumor antigen presentation. Second, CDK4/6 inhibitors markedly suppress the proliferation of regulatory T cells (Tregs). Mechanistically, the effects of CDK4/6 inhibitors on both tumor cells and Tregs are associated with reduced activity of the E2F target, DNA methyltransferase 1. Ultimately, these events promote cytotoxic T cell-mediated clearance of tumor cells, which is further enhanced by the addition of immune checkpoint blockade. Our findings indicate that CDK4/6 inhibitors increase tumor immunogenicity and provide rationale for new combination regimens comprising CDK4/6 inhibitors and immunotherapies as anti-cancer treatment.
Collapse
|
523
|
Sacaan AI, Thibault S, Hong M, Kondegowda NG, Nichols T, Li R, Rosselot C, Evering W, Fenutria R, Vitsky A, Brown T, Finkelstein M, Garcia-Ocaña A, Khan N, Stewart AF, Vasavada RC. CDK4/6 Inhibition on Glucose and Pancreatic Beta Cell Homeostasis in Young and Aged Rats. Mol Cancer Res 2017; 15:1531-1541. [DOI: 10.1158/1541-7786.mcr-17-0172] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/27/2017] [Accepted: 07/25/2017] [Indexed: 11/16/2022]
|
524
|
López-Tarruella S, Jerez Y, Márquez-Rodas I, Echavarria I, Martin M. Ribociclib for the treatment of advanced hormone receptor-positive, HER2-negative breast cancer. Future Oncol 2017; 13:2137-2149. [PMID: 28758424 DOI: 10.2217/fon-2017-0183] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CDK4/6 inhibitors are a promising new class of drugs for hormone-receptor-positive breast cancer and have been shown to overcome and delay hormone resistance in advanced breast cancer. Ribociclib, a selective oral inhibitor of CDK4/6, was approved by the US FDA for first-line treatment of hormone-receptor-positive/HER2-negative metastatic breast cancer. This review summarizes the clinical evidence available for ribociclib, from preclinical data to the pivotal studies, with a special focus on toxicity and its management. In addition, this article reviews potential new combinations under study, as well as ongoing clinical trials both in the metastatic and early setting. Finally, this review compares ribociclib activity and toxicity with those of the drugs of the same class (palbociclib and abemaciclib).
Collapse
Affiliation(s)
- Sara López-Tarruella
- Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), CiberOnc, Madrid, Spain
| | - Yolanda Jerez
- Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), CiberOnc, Madrid, Spain
| | - Iván Márquez-Rodas
- Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), CiberOnc, Madrid, Spain
| | - Isabel Echavarria
- Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), CiberOnc, Madrid, Spain
| | - Miguel Martin
- Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), Universidad Complutense, CiberOnc, GEICAM, Madrid, Spain
| |
Collapse
|
525
|
The addition of abemaciclib to sunitinib induces regression of renal cell carcinoma xenograft tumors. Oncotarget 2017; 8:95116-95134. [PMID: 29221116 PMCID: PMC5707010 DOI: 10.18632/oncotarget.19618] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 06/29/2017] [Indexed: 01/05/2023] Open
Abstract
Multiple therapies currently exist for renal cell carcinoma, however, most do not result in cure and the development of acquired resistance is the rule rather than the exception. CDK4/6 and PIM1 kinases are potential new therapeutic targets in RCC. Abemaciclib is a potent CDK4/6 and PIM1 kinase inhibitor, thus we evaluated the effects of abemaciclib on renal cell carcinoma. In vitro, abemaciclib causes decreased cellular viability, increased apoptosis, and alterations in autophagy in renal cell carcinoma cell lines. A pre-clinical mouse model of RCC shows abemaciclib in combination with sunitinib to cause dramatic reduction in tumor sizes without overt toxicity. Thus abemaciclib is active in renal cell carcinoma and should be evaluated in a clinical trial in combination with sunitinib. Additionally, CDK4/6 and PIM1 kinase appear to be viable clinical targets in renal cell carcinoma.
Collapse
|
526
|
Garrido P, Olmedo ME, Gómez A, Paz Ares L, López-Ríos F, Rosa-Rosa JM, Palacios J. Treating KRAS-mutant NSCLC: latest evidence and clinical consequences. Ther Adv Med Oncol 2017; 9:589-597. [PMID: 29081842 PMCID: PMC5564881 DOI: 10.1177/1758834017719829] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 06/15/2017] [Indexed: 12/19/2022] Open
Abstract
KRAS mutations represent one of the most prevalent oncogenic driver mutations in non-small cell lung cancer (NSCLC). For many years we have unsuccessfully addressed KRAS mutation as a unique disease. The recent widespread use of comprehensive genomic profiling has identified different subgroups with prognostic implications. Moreover, recent data recognizing the distinct biology and therapeutic vulnerabilities of different KRAS subgroups have allowed us to explore different treatment approaches. Small molecules that selectively inhibit KRAS G12C or use of immune checkpoint inhibitors based on co-mutation status are some examples which anticipate that personalized treatment for this challenging disease is finally on the horizon.
Collapse
Affiliation(s)
- Pilar Garrido
- Head of Thoracic Tumor Unit, Medical Oncology Department, Hospital Universitario Ramón y Cajal, Facultad de Medicina. Universidad de Alcalá (IRYCIS) Carretera Colmenar Viejo KM 9100, 28034 Madrid, Spain
| | - María Eugenia Olmedo
- Medical Oncology Department, Hospital Universitario Ramón y Cajal. Facultad de Medicina. Universidad de Alcalá (IRYCIS), Madrid, Spain
| | - Ana Gómez
- Medical Oncology Department, Hospital Universitario Ramón y Cajal. Facultad de Medicina. Universidad de Alcalá (IRYCIS), Madrid, Spain
| | - Luis Paz Ares
- Centro de Investigaciones Biomédicas en Red en Cáncer (CIBER-ONC), Madrid, Spain; Medical Oncology Department, Hospital Universitario Doce de Octubre, Universidad Complutense and Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Fernando López-Ríos
- Centro de Investigaciones Biomédicas en Red en Cáncer (CIBER-ONC), Madrid, Spain Hospital Universitario HM Sanchinarro C/ Oña, 10. 28050 Madrid, España
| | | | - José Palacios
- Centro de Investigaciones Biomédicas en Red en Cáncer (CIBER-ONC), Madrid, Spain Servicio de Anatomía Patológica, Hospital Universitario Ramón y Cajal, Universidad de Alcalá (IRYCIS), Madrid, Spain
| |
Collapse
|
527
|
Kwapisz D. Cyclin-dependent kinase 4/6 inhibitors in breast cancer: palbociclib, ribociclib, and abemaciclib. Breast Cancer Res Treat 2017; 166:41-54. [PMID: 28741274 DOI: 10.1007/s10549-017-4385-3] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 07/07/2017] [Indexed: 11/27/2022]
Abstract
PURPOSE The cyclin D-cyclin dependent kinase (CDK) 4/6-inhibitor of CDK4 (INK4)-retinoblastoma (Rb) pathway plays a crucial role in cell cycle progression and its dysregulation is an important contributor to endocrine therapy resistance. CDK4/6 inhibitors trigger cell cycle arrest in Rb protein (pRb)-competent cells. Recent years have seen the development of selective CDK4/6 inhibitors, which have delivered promising results of efficacy and manageable safety profiles. The main objective of this review is to discuss preclinical and clinical data to date, and ongoing clinical trials with palbociclib, ribociclib, and abemaciclib in breast cancer. METHODS A literature search of above topics was carried out using PubMed and data reported at international oncology meetings and clinicaltrials.gov were included. RESULTS The highly selective oral CDK4/6 inhibitors have been tested in combination with endocrine therapy in Phase III studies in metastatic breast cancer. Results led to the US Food and Drug Administration approval of palbociclib (PD0332991) and ribociclib (LEE011), and abemaciclib (LY2835219) is in development. Studies of these agents, in combination with endocrine therapy, are also underway in ER-positive early breast cancer in the neoadjuvant and adjuvant settings. Moreover, they are also being investigated with other agents in the advanced setting and in triple negative breast cancer. CONCLUSIONS After having demonstrated impressive activity in ER-positive, HER2-negative metastatic breast cancer, currently CDK4/6 inhibitors are in further development. It is obvious that this class of agents with their efficacy, low and easily manageable toxicity, and oral dosage is a very important treatment option for breast cancer patients.
Collapse
|
528
|
Spring LM, Zangardi ML, Moy B, Bardia A. Clinical Management of Potential Toxicities and Drug Interactions Related to Cyclin-Dependent Kinase 4/6 Inhibitors in Breast Cancer: Practical Considerations and Recommendations. Oncologist 2017; 22:1039-1048. [PMID: 28706010 PMCID: PMC5599204 DOI: 10.1634/theoncologist.2017-0142] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 06/08/2017] [Indexed: 12/27/2022] Open
Abstract
Aberrations of the cell cycle are pervasive in cancer, and selective cell cycle inhibition of cancer cells is a target of choice for a number of novel cancer therapeutics. Cyclin-dependent kinases (CDKs) are key regulatory enzymes that control cell cycle transitions and the commitment to cell division. Palbociclib and ribociclib are both orally active, highly selective reversible inhibitors of CDK4 and CDK6 that are approved by the U.S. Food and Drug Administration (FDA) for hormone receptor-positive metastatic breast cancer in combination with specific endocrine therapies. A third oral CDK4/6 inhibitor, abemaciclib, received Breakthrough Therapy designation status from the FDA and is also being developed in breast cancer. The most common adverse events associated with palbociclib and ribociclib are hematologic, particularly neutropenia. However, the neutropenia associated with CDK4/6 inhibitors is distinct from chemotherapy-induced neutropenia in that it is rapidly reversible, reflecting a cytostatic effect on neutrophil precursors in the bone marrow. Most hematologic abnormalities seen with CDK4/6 inhibitors are not complicated and are adequately managed with standard supportive care and dose adjustments when indicated. Cytopenias are less prevalent with abemaciclib, although fatigue and gastrointestinal toxicity is more common with this agent. This review focuses on the clinical management of potential toxicities and drug interactions seen with the use of CDK4/6 inhibitors in breast cancer, with a focus on palbociclib and ribociclib, and summarizes practical management strategies for an oncologist. IMPLICATIONS FOR PRACTICE The emergence of modern cyclin-dependent kinase (CDK) inhibitors has changed the treatment paradigm for metastatic hormone receptor (HR)-positive breast cancer. Palbociclib, ribociclib, and abemaciclib are highly selective reversible inhibitors of CDK4 and CDK6. Palbociclib is U.S. Food and Drug Administration (FDA)-approved in the first- and second-line settings in combination with endocrine therapy for HR-positive metastatic breast cancer. Ribociclib is FDA-approved in the first-line setting. Abemaciclib has received FDA Breakthrough Therapy designation status. This review focuses on the clinical management of potential toxicities and drug interactions seen with the use of CDK4/6 inhibitors in breast cancer.
Collapse
Affiliation(s)
- Laura M Spring
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark L Zangardi
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Beverly Moy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
529
|
Tripathy D, Bardia A, Sellers WR. Ribociclib (LEE011): Mechanism of Action and Clinical Impact of This Selective Cyclin-Dependent Kinase 4/6 Inhibitor in Various Solid Tumors. Clin Cancer Res 2017; 23:3251-3262. [PMID: 28351928 PMCID: PMC5727901 DOI: 10.1158/1078-0432.ccr-16-3157] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 01/12/2017] [Accepted: 03/24/2017] [Indexed: 12/16/2022]
Abstract
The cyclin D-cyclin-dependent kinase (CDK) 4/6-p16-retinoblastoma (Rb) pathway is commonly disrupted in cancer, leading to abnormal cell proliferation. Therapeutics targeting this pathway have demonstrated antitumor effects in preclinical and clinical studies. Ribociclib is a selective, orally bioavailable inhibitor of CDK4 and CDK6, which received FDA approval in March 2017 and is set to enter the treatment landscape alongside other CDK4/6 inhibitors, including palbociclib and abemaciclib. Here, we describe the mechanism of action of ribociclib and review preclinical and clinical data from phase I, II, and III trials of ribociclib across different tumor types, within the context of other selective CDK4/6 inhibitors. The pharmacokinetics, pharmacodynamics, safety, tolerability, and clinical responses with ribociclib as a single agent or in combination with other therapies are discussed, and an overview of the broad portfolio of ongoing clinical trials with ribociclib across a wide range of indications is presented. On the basis of the available data, ribociclib has a manageable tolerability profile and therapeutic potential for a variety of cancer types. Its high selectivity makes it an important partner drug for other targeted therapies, and it has been shown to enhance the clinical activity of existing anticancer therapies and delay the development of treatment resistance, without markedly increasing toxicity. Ongoing trials of doublet and triplet targeted therapies containing ribociclib seek to identify optimal CDK4/6-based targeted combination regimens for various tumor types and advance the field of precision therapeutics in oncology. Clin Cancer Res; 23(13); 3251-62. ©2017 AACR.
Collapse
Affiliation(s)
- Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Aditya Bardia
- Department of Hematology/Oncology, Massachusetts General Hospital Cancer Center/Harvard Medical School, Boston, Massachusetts
| | - William R Sellers
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| |
Collapse
|
530
|
Turner NC, Neven P, Loibl S, Andre F. Advances in the treatment of advanced oestrogen-receptor-positive breast cancer. Lancet 2017; 389:2403-2414. [PMID: 27939057 DOI: 10.1016/s0140-6736(16)32419-9] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 07/28/2016] [Accepted: 08/24/2016] [Indexed: 12/12/2022]
Abstract
Oestrogen-receptor-positive breast cancer is the most common subtype of breast cancer. Endocrine therapies that target the dependence of this subtype on the oestrogen receptor have substantial activity, yet the development of resistance to therapy is inevitable in advanced cancer. Major progress has been made in identifying the drivers of oestrogen-receptor-positive breast cancer and the mechanisms of resistance to endocrine therapy. This progress has translated into major advances in the treatment of advanced breast cancer, with several targeted therapies that enhance the efficacy of endocrine therapy; inhibitors of mTOR and inhibitors of the cyclin-dependent kinases CDK4 and CDK6 substantially improve progression-free survival. A new wave of targeted therapies is being developed, including inhibitors of PI3K, AKT, and HER2, and a new generation of oestrogen-receptor degraders. Considerable challenges remain in patient selection, deciding on the most appropriate order in which to administer therapies, and establishing whether cross-resistance occurs between therapies.
Collapse
Affiliation(s)
| | - Patrick Neven
- Multidisciplinary Breast Centre and Department of Gynaecological Oncology, University Hospitals Leuven, Department of Oncology, Leuven, Belgium
| | - Sibylle Loibl
- German Breast Group (GBG), c/o GBG Forschungs GmbH, Neu-Isenburg, Germany; Centre for Haematology and Oncology, Bethanien, Frankfurt, Germany.
| | - Fabrice Andre
- INSERM U981, Gustave Roussy Cancer Center, Université Paris Sud, Villejuif, France
| |
Collapse
|
531
|
Costa R, Costa RB, Talamantes SM, Helenowski I, Peterson J, Kaplan J, Carneiro BA, Giles FJ, Gradishar WJ. Meta-analysis of selected toxicity endpoints of CDK4/6 inhibitors: Palbociclib and ribociclib. Breast 2017; 35:1-7. [PMID: 28618307 DOI: 10.1016/j.breast.2017.05.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 05/30/2017] [Accepted: 05/31/2017] [Indexed: 11/16/2022] Open
Abstract
PURPOSE Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors such as palbociclib and ribociclib are associated with distinct adverse effects (AEs) compared to other targeted therapies. This meta-analysis of clinical trials summarizes these agents' toxicity profile. METHODS A librarian-guided literature search was conducted in March of 2017. The trials needed to have at least one of the study arms consisting of palbociclib or ribociclib monotherapy at currently FDA approved dose regimens. Heterogeneity across studies was analyzed using I2 statistics. Data were analyzed using random effects meta-analysis for absolute risks. RESULTS Seven randomized trials and 1,332 patients were included in our meta-analysis. There was evidence of significant heterogeneity between studies for serious AEs but not for death. The pooled absolute risk (AR) for all-causality serious AEs and treatment-related death were 16% and 0%, respectively. Patients treated with CDK 4/6 inhibitors had an AR of grade 3/4 neutropenia of 61%; neutropenic fever and infections were rare (1% and 3%, respectively). Grade 3/4 nausea, vomiting, and rash were rare. There was no significant correlation between age of patients at study entry and the risk of grade 3/4 neutropenia. CONCLUSION Treatment with CDK 4/6 inhibitors is well tolerated and associated with a low risk of treatment-related deaths. There is an increased AR of grade 3/4 neutropenia but a low AR of associated infections.
Collapse
Affiliation(s)
- R Costa
- Division of Hematology Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - R B Costa
- Division of Hematology Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Irene Helenowski
- Northwestern University Department of Preventive Medicine, Chicago, IL, USA
| | - Jonna Peterson
- Galter Health Sciences Library, Northwestern University, Chicago, IL, USA
| | - Jason Kaplan
- Division of Hematology Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - B A Carneiro
- Division of Hematology Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Francis J Giles
- Division of Hematology Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - W J Gradishar
- Division of Hematology Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
532
|
Wander SA, Mayer EL, Burstein HJ. Blocking the Cycle: Cyclin-Dependent Kinase 4/6 Inhibitors in Metastatic, Hormone Receptor-Positive Breast Cancer. J Clin Oncol 2017; 35:2866-2870. [PMID: 28580881 DOI: 10.1200/jco.2017.73.9482] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The Oncology Grand Rounds series is designed to place original reports published in the Journal into clinical context. A case presentation is followed by a description of diagnostic and management challenges, a review of the relevant literature, and a summary of the authors' suggested management approaches. The goal of this series is to help readers better understand how to apply the results of key studies, including those published in Journal of Clinical Oncology, to patients seen in their own clinical practice. A 68-year-old postmenopausal woman was diagnosed with breast cancer 6 years ago when she presented with a stage II (T2N1), right-sided, invasive ductal carcinoma considered grade 2 of 3 on core biopsy, with a positive fine-needle aspiration of a palpable, ipsilateral axillary lymph node. Immunohistochemical analysis was positive for estrogen and progesterone receptor expression and negative for human epidermal growth factor receptor 2 (HER2) overexpression. She received neoadjuvant dose-dense doxorubicin, cyclophosphamide, and paclitaxel chemotherapy, followed by breast-conserving surgery and axillary lymph node dissection, which revealed residual disease in three of 11 nodes. She received adjuvant radiation therapy and initiated letrozole, with excellent compliance during the interval 6-year period. While receiving adjuvant letrozole therapy, she reported 3 months of worsening back pain. Skeletal scintigraphy and cross-sectional imaging confirmed widespread osseous metastatic disease and right supraclavicular lymph node enlargement ( Fig 1 ). Core biopsy of the involved lymph node confirmed estrogen receptor (ER)-positive (90%), progesterone receptor-negative, HER2-negative recurrent metastatic breast cancer. The patient reported mild pain that was adequately controlled with over-the-counter anti-inflammatory medications. She has remained active with an excellent performance status.
Collapse
Affiliation(s)
- Seth A Wander
- Seth A. Wander, Erica L. Mayer, and Harold J. Burstein, Dana-Farber Cancer Institute; Brigham & Women's Hospital; Harvard Medical School, Boston, MA
| | - Erica L Mayer
- Seth A. Wander, Erica L. Mayer, and Harold J. Burstein, Dana-Farber Cancer Institute; Brigham & Women's Hospital; Harvard Medical School, Boston, MA
| | - Harold J Burstein
- Seth A. Wander, Erica L. Mayer, and Harold J. Burstein, Dana-Farber Cancer Institute; Brigham & Women's Hospital; Harvard Medical School, Boston, MA
| |
Collapse
|
533
|
Matikas A, Foukakis T, Bergh J. Tackling endocrine resistance in ER-positive HER2-negative advanced breast cancer: A tale of imprecision medicine. Crit Rev Oncol Hematol 2017; 114:91-101. [DOI: 10.1016/j.critrevonc.2017.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/28/2017] [Accepted: 04/04/2017] [Indexed: 12/29/2022] Open
|
534
|
Dickler MN, Tolaney SM, Rugo HS, Cortés J, Diéras V, Patt D, Wildiers H, Hudis CA, O'Shaughnessy J, Zamora E, Yardley DA, Frenzel M, Koustenis A, Baselga J. MONARCH 1, A Phase II Study of Abemaciclib, a CDK4 and CDK6 Inhibitor, as a Single Agent, in Patients with Refractory HR +/HER2 - Metastatic Breast Cancer. Clin Cancer Res 2017; 23:5218-5224. [PMID: 28533223 DOI: 10.1158/1078-0432.ccr-17-0754] [Citation(s) in RCA: 493] [Impact Index Per Article: 61.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/04/2017] [Accepted: 05/17/2017] [Indexed: 01/19/2023]
Abstract
Purpose: The phase II MONARCH 1 study was designed to evaluate the single-agent activity and adverse event (AE) profile of abemaciclib, a selective inhibitor of CDK4 and CDK6, in women with refractory hormone receptor-positive (HR+), HER2- metastatic breast cancer (MBC).Experimental Design: MONARCH 1 was a phase II single-arm open-label study. Women with HR+/HER2- MBC who had progressed on or after prior endocrine therapy and had 1 or 2 chemotherapy regimens in the metastatic setting were eligible. Abemaciclib 200 mg was administered orally on a continuous schedule every 12 hours until disease progression or unacceptable toxicity. The primary objective of MONARCH 1 was investigator-assessed objective response rate (ORR). Other endpoints included clinical benefit rate, progression-free survival (PFS), and overall survival (OS).Results: Patients (n = 132) had a median of 3 (range, 1-8) lines of prior systemic therapy in the metastatic setting, 90.2% had visceral disease, and 50.8% had ≥3 metastatic sites. At the 12-month final analysis, the primary objective of confirmed objective response rate was 19.7% (95% CI, 13.3-27.5; 15% not excluded); clinical benefit rate (CR+PR+SD≥6 months) was 42.4%, median progression-free survival was 6.0 months, and median overall survival was 17.7 months. The most common treatment-emergent AEs of any grade were diarrhea, fatigue, and nausea; discontinuations due to AEs were infrequent (7.6%).Conclusions: In this poor-prognosis, heavily pretreated population with refractory HR+/HER2- metastatic breast cancer, continuous dosing of single-agent abemaciciclib was well tolerated and exhibited promising clinical activity. Clin Cancer Res; 23(17); 5218-24. ©2017 AACR.
Collapse
Affiliation(s)
| | | | - Hope S Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, California
| | - Javier Cortés
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Ramón y Cajal University Hospital, Madrid, Spain
| | | | - Debra Patt
- Texas Oncology, Austin, Texas
- Baylor University Medical Center, Texas Oncology, US Oncology, Dallas, Texas
| | - Hans Wildiers
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | | | - Joyce O'Shaughnessy
- Baylor University Medical Center, Texas Oncology, US Oncology, Dallas, Texas
| | - Esther Zamora
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Denise A Yardley
- Sarah Cannon Research Institute, Tennessee Oncology PLLC, Nashville, Tennessee
| | | | | | - José Baselga
- Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
535
|
Torres-Guzmán R, Calsina B, Hermoso A, Baquero C, Alvarez B, Amat J, McNulty AM, Gong X, Boehnke K, Du J, de Dios A, Beckmann RP, Buchanan S, Lallena MJ. Preclinical characterization of abemaciclib in hormone receptor positive breast cancer. Oncotarget 2017; 8:69493-69507. [PMID: 29050219 PMCID: PMC5642494 DOI: 10.18632/oncotarget.17778] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 04/24/2017] [Indexed: 11/28/2022] Open
Abstract
Abemaciclib is an ATP-competitive, reversible kinase inhibitor selective for CDK4 and CDK6 that has shown antitumor activity as a single agent in hormone receptor positive (HR+) metastatic breast cancer in clinical trials. Here, we examined the mechanistic effects of abemaciclib treatment using in vitro and in vivo breast cancer models. Treatment of estrogen receptor positive (ER+) breast cancer cells with abemaciclib alone led to a decrease in phosphorylation of Rb, arrest at G1, and a decrease in cell proliferation. Moreover, abemaciclib exposure led to durable inhibition of pRb, TopoIIα expression and DNA synthesis, which were maintained after drug removal. Treatment of ER+ breast cancer cells also led to a senescence response as indicated by accumulation of β-galactosidase, formation of senescence-associated heterochromatin foci, and a decrease in FOXM1 positive cells. Continuous exposure to abemaciclib altered breast cancer cell metabolism and induced apoptosis. In a xenograft model of ER+ breast cancer, abemaciclib monotherapy caused regression of tumor growth. Overall these data indicate that abemaciclib is a CDK4 and CDK6 inhibitor that, as a single agent, blocks breast cancer cell progression, and upon longer treatment can lead to sustained antitumor effects through the induction of senescence, apoptosis, and alteration of cellular metabolism.
Collapse
Affiliation(s)
| | - Bruna Calsina
- Quantitative Biology, Eli Lilly and Company, Madrid, Spain
| | - Ana Hermoso
- Quantitative Biology, Eli Lilly and Company, Madrid, Spain
| | - Carmen Baquero
- Quantitative Biology, Eli Lilly and Company, Madrid, Spain
| | | | - Joaquín Amat
- Quantitative Biology, Eli Lilly and Company, Madrid, Spain
| | - Ann M McNulty
- Oncology Research, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Xueqian Gong
- Oncology Research, Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Jian Du
- Oncology Research, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Alfonso de Dios
- Discovery Chemistry, Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Sean Buchanan
- Oncology Research, Eli Lilly and Company, Indianapolis, Indiana, USA
| | | |
Collapse
|
536
|
Abstract
Over the past two decades there has been a great deal of interest in the development of inhibitors of the cyclin-dependent kinases (CDKs). This attention initially stemmed from observations that different CDK isoforms have key roles in cancer cell proliferation through loss of regulation of the cell cycle, a hallmark feature of cancer. CDKs have now been shown to regulate other processes, particularly various aspects of transcription. The early non-selective CDK inhibitors exhibited considerable toxicity and proved to be insufficiently active in most cancers. The lack of patient selection biomarkers and an absence of understanding of the inhibitory profile required for efficacy hampered the development of these inhibitors. However, the advent of potent isoform-selective inhibitors with accompanying biomarkers has re-ignited interest. Palbociclib, a selective CDK4/6 inhibitor, is now approved for the treatment of ER+/HER2- advanced breast cancer. Current developments in the field include the identification of potent and selective inhibitors of the transcriptional CDKs; these include tool compounds that have allowed exploration of individual CDKs as cancer targets and the determination of their potential therapeutic windows. Biomarkers that allow the selection of patients likely to respond are now being discovered. Drug resistance has emerged as a major hurdle in the clinic for most protein kinase inhibitors and resistance mechanism are beginning to be identified for CDK inhibitors. This suggests that the selective inhibitors may be best used combined with standard of care or other molecularly targeted agents now in development rather than in isolation as monotherapies.
Collapse
Affiliation(s)
- Steven R Whittaker
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Aurélie Mallinger
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Paul Workman
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Paul A Clarke
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SW7 3RP, United Kingdom.
| |
Collapse
|
537
|
Kaklamani VG, Gradishar WJ. Endocrine Therapy in the Current Management of Postmenopausal Estrogen Receptor-Positive Metastatic Breast Cancer. Oncologist 2017; 22:507-517. [PMID: 28314835 PMCID: PMC5423509 DOI: 10.1634/theoncologist.2015-0464] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 12/18/2016] [Indexed: 12/16/2022] Open
Abstract
Metastatic breast cancer (MBC) results in substantial morbidity and mortality for women afflicted with this disease. A majority of MBCs are hormone-responsive and estrogen receptor-positive, making endocrine therapy (ET) an integral component of systemic therapy. With a primary goal of minimizing the effects of estrogen on hormone-responsive MBC, ETs are among the first targeted treatments that aim to inhibit the influence of estrogen receptor activation on tumor proliferation. Several biochemical mechanisms have been the focus of drug development for treatment, including selective estrogen-receptor modulation, aromatase inhibition, and selective estrogen-receptor degradation. Treatments that exploit these mechanisms have improved survival and quality of life for women with MBC. However, in many cases, resistance to ET limits their effectiveness. Elucidation of the complex cellular signal cascades involved in the development of acquired resistance to ET and the interrelationship of growth factor signaling and estrogen responsiveness have characterized components of these pathways as attractive targets for drug development. Based on these insights and with the aim of overcoming hormone resistance, targeted therapies are emerging as useful treatments for MBC. This article reviews current endocrine treatments of MBC as well as recent and ongoing study of combination treatments and targeted therapies that interfere with cellular proliferation pathways as means of overcoming resistance. The Oncologist 2017;22:507-517 IMPLICATIONS FOR PRACTICE: This review provides medical oncologists and other oncology health care providers with a current understanding of the rationale for endocrine therapy in estrogen receptor-positive metastatic breast cancer and the efficacy and safety profile of available treatment options. Additionally, current concepts regarding the development of treatment resistance and the treatment strategies for overcoming resistance are discussed. Enhancing the current information and the understanding of these topics will assist clinicians in evaluating optimal treatment options for their patients.
Collapse
Affiliation(s)
- Virginia G Kaklamani
- Cancer Treatment and Research Center at the University of Texas Health Science Center, San Antonio, Texas, USA
| | - William J Gradishar
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
538
|
Jansen VM, Bhola NE, Bauer JA, Formisano L, Lee KM, Hutchinson KE, Witkiewicz AK, Moore PD, Estrada MV, Sánchez V, Ericsson PG, Sanders ME, Pohlmann PR, Pishvaian MJ, Riddle DA, Dugger TC, Wei W, Knudsen ES, Arteaga CL. Kinome-Wide RNA Interference Screen Reveals a Role for PDK1 in Acquired Resistance to CDK4/6 Inhibition in ER-Positive Breast Cancer. Cancer Res 2017; 77:2488-2499. [PMID: 28249908 PMCID: PMC5421398 DOI: 10.1158/0008-5472.can-16-2653] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/23/2016] [Accepted: 02/22/2017] [Indexed: 12/13/2022]
Abstract
Acquired resistance to cyclin-dependent kinases 4 and 6 (CDK4/6) small-molecule inhibitors in breast cancer arises through mechanisms that are yet uncharacterized. In this study, we used a kinome-wide siRNA screen to identify kinases that, when downregulated, yield sensitivity to the CDK4/6 inhibitor ribociclib. In this manner, we identified 3-phosphoinositide-dependent protein kinase 1 (PDK1) as a key modifier of ribociclib sensitivity in estrogen receptor-positive MCF-7 breast cancer cells. Pharmacologic inhibition of PDK1 with GSK2334470 in combination with ribociclib or palbociclib, another CDK4/6 inhibitor, synergistically inhibited proliferation and increased apoptosis in a panel of ER-positive breast cancer cell lines. Ribociclib-resistant breast cancer cells selected by chronic drug exposure displayed a relative increase in the levels of PDK1 and activation of the AKT pathway. Analysis of these cells revealed that CDK4/6 inhibition failed to induce cell-cycle arrest or senescence. Mechanistic investigations showed that resistant cells coordinately upregulated expression of cyclins A, E, and D1, activated phospho-CDK2, and phospho-S477/T479 AKT. Treatment with GSK2334470 or the CDK2 inhibitor dinaciclib was sufficient to reverse these events and to restore the sensitivity of ribociclib-resistant cells to CDK4/6 inhibitors. Ribociclib, in combination with GSK2334470 or the PI3Kα inhibitor alpelisib, decreased xenograft tumor growth more potently than each drug alone. Taken together, our results highlight a role for the PI3K-PDK1 signaling pathway in mediating acquired resistance to CDK4/6 inhibitors. Cancer Res; 77(9); 2488-99. ©2017 AACR.
Collapse
Affiliation(s)
- Valerie M Jansen
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Neil E Bhola
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joshua A Bauer
- Department of Biochemistry, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Luigi Formisano
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kyung-Min Lee
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Katherine E Hutchinson
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Preston D Moore
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mónica Valéria Estrada
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Violeta Sánchez
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paula G Ericsson
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Melinda E Sanders
- Department of Pathology, Microbiology and Immunology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paula R Pohlmann
- Department of Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Michael J Pishvaian
- Department of Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - David A Riddle
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Teresa C Dugger
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Erik S Knudsen
- Department of Medicine, University of Arizona, Tucson, Arizona
| | - Carlos L Arteaga
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee.
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
539
|
Pan Q, Sathe A, Black PC, Goebell PJ, Kamat AM, Schmitz-Draeger B, Nawroth R. CDK4/6 Inhibitors in Cancer Therapy: A Novel Treatement Strategy for Bladder Cancer. Bladder Cancer 2017; 3:79-88. [PMID: 28516152 PMCID: PMC5409046 DOI: 10.3233/blc-170105] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Patients with metastatic bladder cancer (mBC) treated with cisplatin-based chemotherapy have a limited median survival of only around 14 months [1]. Despite over 30 years of basic and clinical research, until recently no therapeutic options beyond cisplatin-based therapy had entered clinical routine and, at least in the US, none of the tested agents had been approved for second-line treatment. This has changed with the advent of immune checkpoint blockade, including especially PD-1/PD-L1 inhibitors. The high response rates of 24% over a 14.4 month follow up led to the first US Food and Drug Administration (FDA) approval for a second line therapy for these patients, and it is likely that this marks the beginning of a new era in the systemic treatment of muscle-invasive bladder cancer [2–4]. The strong clinical need to improve the medical management of this disease for those patients, not responding to current therapy has led to an increased molecular understanding of bladder cancer and has forstered the development of many potential molecular manipulations and targeted strategies beyond the new immune-oncologic approaches. Among the molecular alterations indentified in bladder cancer, cell cycle deregulation appears to be a key driver of disease progression. Target-directed therapy against CDK4/6 is an emerging strategy to regain control of cell cycle deregulation. Here, we provide an overview of the current status of CDK4/6 inhibitors in cancer therapy, their potential use in mBC and the challenges for their clinical use.
Collapse
Affiliation(s)
- Qi Pan
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Anuja Sathe
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Peter C Black
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Peter J Goebell
- Department of Urology, Friedrich-Alexander University, Erlangen, Germany
| | - Ashish M Kamat
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bernd Schmitz-Draeger
- Department of Urology, Friedrich-Alexander University, Erlangen, Germany; Urologie(24)/Urology Schön Klinik Nürnberg Fürth, Fürth, Germany
| | - Roman Nawroth
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
540
|
Xu H, Yu S, Liu Q, Yuan X, Mani S, Pestell RG, Wu K. Recent advances of highly selective CDK4/6 inhibitors in breast cancer. J Hematol Oncol 2017; 10:97. [PMID: 28438180 PMCID: PMC5404666 DOI: 10.1186/s13045-017-0467-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 04/19/2017] [Indexed: 01/15/2023] Open
Abstract
Uncontrolled cell division is the hallmark of cancers. Full understanding of cell cycle regulation would contribute to promising cancer therapies. In particular, cyclin-dependent kinases 4/6 (CDK4/6), which are pivotal drivers of cell proliferation by combination with cyclin D, draw more and more attention. Subsequently, extensive studies were carried out to explore drugs inhibiting CDK4/6 and assess the efficacy and safety of these drugs in cancer, especially breast cancer. Due to the insuperable adverse events and the less activity observed in vivo, the drug development of the initial pan-CDK inhibitor flavopiridol was consequently discontinued, and then highly specific inhibitors were extensively researched and developed, including palbociclib (PD0332991), ribociclib (LEE011), and abemaciclib (LY2835219). Food and Drug Administration has approved palbociclib and ribociclib for the treatment of hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced or metastatic breast cancer, and recent clinical trial data suggest that palbociclib significantly improved clinical outcome when combined with letrozole or fulvestrant. Besides, the favorable effects of abemaciclib on prolonging survival of breast cancer patients have also been observed in clinical trials both for single-agent and combination strategy. In this review, we outline the preclinical and clinical advancement of these three orally bioavailable and highly selective CDK4/6 inhibitors in breast cancer.
Collapse
Affiliation(s)
- Hanxiao Xu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 People’s Republic of China
| | - Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 People’s Republic of China
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 People’s Republic of China
| | - Xun Yuan
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 People’s Republic of China
| | - Sridhar Mani
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, NY 10461 USA
| | - Richard G. Pestell
- Pennsylvania Center for Cancer and Regenerative Medicine, Wynnewood, PA 19096 USA
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 People’s Republic of China
| |
Collapse
|
541
|
Rondla R, Padma Rao LS, Ramatenki V, Vadija R, Mukkera T, Potlapally SR, Vuruputuri U. Azolium analogues as CDK4 inhibitors: Pharmacophore modeling, 3D QSAR study and new lead drug discovery. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2016.12.106] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
542
|
Soldera SV, Leighl NB. Update on the Treatment of Metastatic Squamous Non-Small Cell Lung Cancer in New Era of Personalized Medicine. Front Oncol 2017; 7:50. [PMID: 28396848 PMCID: PMC5366319 DOI: 10.3389/fonc.2017.00050] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/09/2017] [Indexed: 12/26/2022] Open
Abstract
Despite advances in molecular characterization and lung cancer treatment in recent years, treatment options for patients diagnosed with squamous cell carcinoma of the lung (SCC) remain limited as actionable mutations are rarely detected in this subtype. This article reviews potential molecular targets and associated novel agents for the treatment of advanced SCC in the era of personalized medicine. Elements of various pathways including epidermal growth factor receptor, PI3KCA, fibroblast growth factor receptor, retinoblastoma, cyclin-dependent kinases, discoidin domain receptor tyrosine kinase 2, and mesenchymal-to-epithelial transition may play pivotal roles in the development of SCC and are under investigation for drug development.
Collapse
Affiliation(s)
| | - Natasha B. Leighl
- Division of Medical Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
543
|
Zugazagoitia J, Molina-Pinelo S, Lopez-Rios F, Paz-Ares L. Biological therapies in nonsmall cell lung cancer. Eur Respir J 2017; 49:49/3/1601520. [PMID: 28254765 DOI: 10.1183/13993003.01520-2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/28/2016] [Indexed: 11/05/2022]
Abstract
Biological therapies have improved survival outcomes of advanced-stage nonsmall cell lung cancer (NSCLC). Genotype-directed therapies have changed treatment paradigms of patients with EGFR-mutant and ALK/ROS1-rearranged lung adenocarcinomas, and the list of druggable targets with demonstrated clinical actionability (BRAF, MET, RET, NTRK1 and HER2) continues to expand. Furthermore, we have incrementally understood the mechanisms of cancer immune evasion and foresee ways to effectively circumvent them, particularly at the immune checkpoint level. Drugs targeting the tumour immune-evasive PD-1 pathway have demonstrated remarkable treatment benefits in this disease, with a non-negligible fraction of patients potentially receiving long-term survival benefits. Herein, we briefly discuss the role of various medical disciplines in the management of advanced-stage NSCLC and review the most relevant biological therapies for this disease, with particular emphasis in genotype-directed therapies and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Jon Zugazagoitia
- Medical Oncology Dept, Hospital Universitario 12 de Octubre and Instituto de Investigación i+12, Madrid, Spain.,Lung Cancer Group, Clinical Research Program, CNIO (Centro Nacional de Investigaciones Oncológicas), Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain.,These authors contributed equally to this work
| | - Sonia Molina-Pinelo
- Medical Oncology Dept, Hospital Universitario 12 de Octubre and Instituto de Investigación i+12, Madrid, Spain.,Lung Cancer Group, Clinical Research Program, CNIO (Centro Nacional de Investigaciones Oncológicas), Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain.,These authors contributed equally to this work
| | - Fernando Lopez-Rios
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain.,Laboratorio de Dianas Terapéuticas, Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Luis Paz-Ares
- Medical Oncology Dept, Hospital Universitario 12 de Octubre and Instituto de Investigación i+12, Madrid, Spain .,Lung Cancer Group, Clinical Research Program, CNIO (Centro Nacional de Investigaciones Oncológicas), Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain.,Complutense University, Madrid, Spain
| |
Collapse
|
544
|
Garrido-Castro AC, Goel S. CDK4/6 Inhibition in Breast Cancer: Mechanisms of Response and Treatment Failure. CURRENT BREAST CANCER REPORTS 2017; 9:26-33. [PMID: 28479958 PMCID: PMC5414585 DOI: 10.1007/s12609-017-0232-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
PURPOSE OF REVIEW To describe the role of D-type cyclins and CDKs 4 and 6 in breast cancer, and to discuss potential biomarkers for sensitivity or resistance to CDK4/6 inhibitors. RECENT FINDINGS A small number of preclinical and clinical studies have explored potential mechanisms of CDK4/6 inhibitor response and resistance in breast cancer. Putative markers of response include ER-positivity, luminal patterns of gene expression, high cyclin D1 levels, and low p16 levels. Possible resistance mechanisms include loss of Rb function, overexpression/amplification of cyclin E, and CDK6 amplification. Most these remain speculative and have not been validated in clinical specimens. SUMMARY If early successes with CDK4/6 inhibitors are to be capitalized upon, it is critical that our understanding of CDK4/6 biology in breast cancer extends beyond its current rudimentary state. Only then we will be able to develop rational therapeutic combinations that further enhance the efficacy of these agents.
Collapse
Affiliation(s)
- Ana C Garrido-Castro
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Shom Goel
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
545
|
Corona SP, Ravelli A, Cretella D, Cappelletti MR, Zanotti L, Dester M, Gobbi A, Petronini PG, Generali D. CDK4/6 inhibitors in HER2-positive breast cancer. Crit Rev Oncol Hematol 2017; 112:208-214. [PMID: 28325261 DOI: 10.1016/j.critrevonc.2017.02.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 10/20/2022] Open
Abstract
Notwithstanding the continuous progress made in cancer treatment in the last 20 years, and the availability of new targeted therapies, metastatic Breast Cancer (BC) is still incurable. Targeting the cell cycle machinery has emerged as an attractive strategy to tackle cancer progression, showing very promising results in the preclinical and clinical settings. The first selective inhibitors of CDK4/6 received breakthrough status and FDA approval in combination with letrozole (February 2015) and fulvestrant (February 2016) as first-line therapy in ER-positive advanced and metastatic BC. Considering the success of this family of compounds in hormone-positive BC, new possible applications are being investigated in other molecular subtypes. This review summarizes the latest findings on the use of CDK4/6 inhibitors in HER2 positive BC.
Collapse
Affiliation(s)
- Silvia Paola Corona
- Peter MacCallum Cancer Centre, Radiation Oncology Department, Moorabbin Campus, East Bentleigh Victoria 3165, Australia.
| | - Andrea Ravelli
- Universita degli Studi di Parma, Department of Clinical and Experimental Medicine, Experimental Oncology Unit, Via Gramsci, 14, Parma, Italy
| | - Daniele Cretella
- Universita degli Studi di Parma, Department of Clinical and Experimental Medicine, Experimental Oncology Unit, Via Gramsci, 14, Parma, Italy
| | - Maria Rosa Cappelletti
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Laura Zanotti
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Martina Dester
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Angela Gobbi
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Pier Giorgio Petronini
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Daniele Generali
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy; Universita degli Studi di Trieste, Department of Medical, Surgery and Health Sciences, Trieste, Italy
| |
Collapse
|
546
|
Stice JP, Wardell SE, Norris JD, Yllanes AP, Alley HM, Haney VO, White HS, Safi R, Winter PS, Cocce KJ, Kishton RJ, Lawrence SA, Strum JC, McDonnell DP. CDK4/6 Therapeutic Intervention and Viable Alternative to Taxanes in CRPC. Mol Cancer Res 2017; 15:660-669. [PMID: 28209757 DOI: 10.1158/1541-7786.mcr-17-0028] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 01/26/2017] [Indexed: 12/11/2022]
Abstract
Resistance to second-generation androgen receptor (AR) antagonists and CYP17 inhibitors in patients with castration-resistant prostate cancer (CRPC) develops rapidly through reactivation of the androgen signaling axis and has been attributed to AR overexpression, production of constitutively active AR splice variants, or the selection for AR mutants with altered ligand-binding specificity. It has been established that androgens induce cell-cycle progression, in part, through upregulation of cyclin D1 (CCND1) expression and subsequent activation of cyclin-dependent kinases 4 and 6 (CDK4/6). Thus, the efficacy of the newly described CDK4/6 inhibitors (G1T28 and G1T38), docetaxel and enzalutamide, was evaluated as single agents in clinically relevant in vitro and in vivo models of hormone-sensitive and treatment-resistant prostate cancer. CDK4/6 inhibition (CDK4/6i) was as effective as docetaxel in animal models of treatment-resistant CRPC but exhibited significantly less toxicity. The in vivo effects were durable and importantly were observed in prostate cancer cells expressing wild-type AR, AR mutants, and those that have lost AR expression. CDK4/6i was also effective in prostate tumor models expressing the AR-V7 variant or the AR F876L mutation, both of which are associated with treatment resistance. Furthermore, CDK4/6i was effective in prostate cancer models where AR expression was lost. It is concluded that CDK4/6 inhibitors are a viable alternative to taxanes as therapeutic interventions in endocrine therapy-refractory CRPC.Implications: The preclinical efficacy of CDK4/6 monotherapy observed here suggests the need for near-term clinical studies of these agents in advanced prostate cancer. Mol Cancer Res; 15(6); 660-9. ©2017 AACR.
Collapse
Affiliation(s)
- James P Stice
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Suzanne E Wardell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - John D Norris
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Alexander P Yllanes
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Holly M Alley
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Victoria O Haney
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Hannah S White
- G1 Therapeutics, Inc., Research Triangle Park, North Carolina
| | - Rachid Safi
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Peter S Winter
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Kimberly J Cocce
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Rigel J Kishton
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Scott A Lawrence
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Jay C Strum
- G1 Therapeutics, Inc., Research Triangle Park, North Carolina
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina.
| |
Collapse
|
547
|
The Growing Role of CDK4/6 Inhibitors in Treating Hormone Receptor-Positive Advanced Breast Cancer. Curr Treat Options Oncol 2017; 18:6. [DOI: 10.1007/s11864-017-0443-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
548
|
Rocca A, Schirone A, Maltoni R, Bravaccini S, Cecconetto L, Farolfi A, Bronte G, Andreis D. Progress with palbociclib in breast cancer: latest evidence and clinical considerations. Ther Adv Med Oncol 2017; 9:83-105. [PMID: 28203301 PMCID: PMC5298405 DOI: 10.1177/1758834016677961] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Deregulation of the cell cycle is a hallmark of cancer, and research on cell cycle control has allowed identification of potential targets for anticancer treatment. Palbociclib is a selective inhibitor of the cyclin-dependent kinases 4 and 6 (CDK4/6), which are involved, with their coregulatory partners cyclin D, in the G1-S transition. Inhibition of this step halts cell cycle progression in cells in which the involved pathway, including the retinoblastoma protein (Rb) and the E2F family of transcription factors, is functioning, although having been deregulated. Among breast cancers, those with functioning cyclin D-CDK4/6-Rb-E2F are mainly hormone-receptor (HR) positive, with some HER2-positive and rare triple-negative cases. Deregulation results from genetic or otherwise occurring hyperactivation of molecules subtending cell cycle progression, or inactivation of cell cycle inhibitors. Based on results of randomized clinical trials, palbociclib was granted accelerated approval by the US Food and Drug Administration (FDA) for use in combination with letrozole as initial endocrine-based therapy for metastatic disease in postmenopausal women with HR-positive, HER2-negative breast cancer, and was approved for use in combination with fulvestrant in women with HR-positive, HER2-negative advanced breast cancer with disease progression following endocrine therapy. This review provides an update of the available knowledge on the cell cycle and its regulation, on the alterations in cyclin D-CDK4/6-Rb-E2F axis in breast cancer and their roles in endocrine resistance, on the preclinical activity of CDK4/6 inhibitors in breast cancer, both as monotherapy and as partners of combinatorial synergic treatments, and on the clinical development of palbociclib in breast cancer.
Collapse
Affiliation(s)
- Andrea Rocca
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Maroncelli 40, Meldola, FC 47014, Italy
| | - Alessio Schirone
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| | - Roberta Maltoni
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| | - Sara Bravaccini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| | - Lorenzo Cecconetto
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| | - Alberto Farolfi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| | - Giuseppe Bronte
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| | - Daniele Andreis
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| |
Collapse
|
549
|
[Cell cycle inhibitors in endocrine receptor positive breast cancer]. Bull Cancer 2017; 104:114-122. [PMID: 28126188 DOI: 10.1016/j.bulcan.2016.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/08/2016] [Accepted: 12/14/2016] [Indexed: 11/24/2022]
Abstract
Dysregulation of cellular cycle is a key component of carcinogenesis and its targeting represents an interesting approach. Recently, the development of selective inhibitors of the cycle targeting the cyclin-dependent kinases (CDK) 4 and 6 revived interest in this therapeutic class after the failure of pan-inhibitors. Palbociclib, ribociclib, and abemaciclib are the 3 drugs with the most advanced development. They demonstrated preclinical activity in luminal breast cancer models and are under clinical evaluation. The first available studies demonstrate the value of these compounds with an improved prognosis of metastatic patients in combination with endocrine therapy (palbociclib, ribociclib) or in monotherapy (abemaciclib). The results of ongoing studies will clarify the role of these agents in our new strategies and the individualisation of biomarkers will help to define patients who benefit most from this approach.
Collapse
|
550
|
Cai Z, Liu Q. Cell Cycle Regulation in Treatment of Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:251-270. [PMID: 29282688 DOI: 10.1007/978-981-10-6020-5_12] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell cycle progression and cell proliferation are under precise and orchestrated control in normal cells. However, uncontrolled cell proliferation caused by aberrant cell cycle progression is a crucial characteristic of cancer. Understanding cell cycle progression and its regulation sheds light on cancer treatment. Agents targeting cell cycle regulators (such as CDKs) have been considered as promising candidates in cancer treatment. Although the first-generation pan-CDK inhibitors failed in clinical trials because of their adverse events and low efficacy, new selective CDK 4/6 inhibitors showed potent efficacy with tolerable safety in preclinical and clinical studies. Here we will review the mechanisms of cell cycle regulation and targeting key cell cycle regulators (such as CDKs) in breast cancer treatment. Particularly, we will discuss the mechanism of CDK inhibitors in disrupting cell cycle progression, the use of selective CDK4/6 inhibitors in treatment of advanced, hormone receptor (HR)-positive postmenopausal breast cancer patients, and other clinical trials that aim to extend the utilization of these agents.
Collapse
Affiliation(s)
- Zijie Cai
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, 510120, Guangdong, China
| | - Qiang Liu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|