101
|
McAndrew NP, Finn RS. Management of ER positive metastatic breast cancer. Semin Oncol 2020; 47:270-277. [PMID: 32958261 DOI: 10.1053/j.seminoncol.2020.07.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/31/2020] [Accepted: 07/31/2020] [Indexed: 12/29/2022]
Abstract
There are over 2 million cases a year of breast cancer, leading to over 600,000 deaths globally [1]. Despite these large numbers, increasingly more women are being cured with early stage disease and women with advanced disease are living longer [2]. The appreciation for molecular subtypes of the disease has led to significant therapeutic advances and estrogen receptor positive (ER+) breast cancer represents the largest of these subgroups. An appreciation for the importance of estrogen signaling in ER+ dates back to 1896 when Dr. George Thomas Beatson observed impressive disease responses after performing bilateral oophorectomy in 3 women at Glasgow Cancer Hospital [3]. The evolution of treatment for advanced disease from progestins, to the selective estrogen receptor modulator tamoxifen, and subsequently the aromatase inhibitors and the selective estrogen receptor degrader fulvestrant, has been accompanied by improved efficacy and decreased side effects. While the use of these drugs has changed the natural history of both early and advanced disease, it has been long recognized that many patients will develop resistance to this approach. After many years of trying to improve on single-agent endocrine treatment, since 2012 there has been an explosion of new drugs that have shown improved efficacy in combination with endocrine approaches. The first of these to receive FDA approval was the mTOR inhibitor everolimus (2012) [4], followed by the approval of 3 cyclin-dependent kinase 4 and 6 (CDK 4/6) inhibitors [palbociclib (2015) [5], ribociclib (2018) [6], and abemaciclib (2018) [7]], and more recently the PI3-kinase inhibitor alpelisib (2019) [8]. In addition, chemotherapy is still used frequently when endocrine manipulations have been exhausted. Like other incurable malignancies, the goal in advanced ER+ breast cancer is to prolong survival and maintain quality of life. Currently, we have more tools available to achieve this than ever before and we will review the efficacy and side effect data with these agents that are driving physician choices for individual patients.
Collapse
Affiliation(s)
- Nicholas P McAndrew
- Division of Hematology Oncology, Department of Medicine, Geffen School of Medicine at UCLA, Santa Monica, CA 90404, United States
| | - Richard S Finn
- Division of Hematology Oncology, Department of Medicine, Geffen School of Medicine at UCLA, Santa Monica, CA 90404, United States.
| |
Collapse
|
102
|
Walsh EM, Smith KL, Stearns V. Management of hormone receptor-positive, HER2-negative early breast cancer. Semin Oncol 2020; 47:187-200. [PMID: 32546323 PMCID: PMC7374796 DOI: 10.1053/j.seminoncol.2020.05.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 12/24/2022]
Abstract
The majority of breast cancers are diagnosed at an early stage and are hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-negative. Significant advances have been made in the management of early stage HR-positive, HER2-negative breast cancer, resulting in improved survival outcomes. In this review, we discuss important factors to consider in the management of this disease. In particular, we discuss the role of adjuvant endocrine therapy, specific endocrine therapy agents, the duration of adjuvant endocrine therapy, treatment-related side effects, and the role of genomic assays and other biomarkers when considering treatment recommendations for individuals with HR-positive, HER2-negative early breast cancer. Finally, we address emerging data to individualize therapeutic decision-making and provide future considerations.
Collapse
Affiliation(s)
- Elaine M Walsh
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Karen L Smith
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Vered Stearns
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD.
| |
Collapse
|
103
|
Wander SA, Cohen O, Gong X, Johnson GN, Buendia-Buendia JE, Lloyd MR, Kim D, Luo F, Mao P, Helvie K, Kowalski KJ, Nayar U, Waks AG, Parsons SH, Martinez R, Litchfield LM, Ye XS, Yu C, Jansen VM, Stille JR, Smith PS, Oakley GJ, Chu QS, Batist G, Hughes ME, Kremer JD, Garraway LA, Winer EP, Tolaney SM, Lin NU, Buchanan SG, Wagle N. The Genomic Landscape of Intrinsic and Acquired Resistance to Cyclin-Dependent Kinase 4/6 Inhibitors in Patients with Hormone Receptor-Positive Metastatic Breast Cancer. Cancer Discov 2020; 10:1174-1193. [PMID: 32404308 PMCID: PMC8815415 DOI: 10.1158/2159-8290.cd-19-1390] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/29/2020] [Accepted: 05/08/2020] [Indexed: 11/16/2022]
Abstract
Mechanisms driving resistance to cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) in hormone receptor-positive (HR+) breast cancer have not been clearly defined. Whole-exome sequencing of 59 tumors with CDK4/6i exposure revealed multiple candidate resistance mechanisms including RB1 loss, activating alterations in AKT1, RAS, AURKA, CCNE2, ERBB2, and FGFR2, and loss of estrogen receptor expression. In vitro experiments confirmed that these alterations conferred CDK4/6i resistance. Cancer cells cultured to resistance with CDK4/6i also acquired RB1, KRAS, AURKA, or CCNE2 alterations, which conferred sensitivity to AURKA, ERK, or CHEK1 inhibition. Three of these activating alterations-in AKT1, RAS, and AURKA-have not, to our knowledge, been previously demonstrated as mechanisms of resistance to CDK4/6i in breast cancer preclinically or in patient samples. Together, these eight mechanisms were present in 66% of resistant tumors profiled and may define therapeutic opportunities in patients. SIGNIFICANCE: We identified eight distinct mechanisms of resistance to CDK4/6i present in 66% of resistant tumors profiled. Most of these have a therapeutic strategy to overcome or prevent resistance in these tumors. Taken together, these findings have critical implications related to the potential utility of precision-based approaches to overcome resistance in many patients with HR+ metastatic breast cancer.This article is highlighted in the In This Issue feature, p. 1079.
Collapse
Affiliation(s)
- Seth A. Wander
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Harvard Medical School, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Ofir Cohen
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA
| | | | - Gabriela N. Johnson
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Jorge E. Buendia-Buendia
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Maxwell R. Lloyd
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Dewey Kim
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Flora Luo
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Harvard Medical School, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Pingping Mao
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Harvard Medical School, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Karla Helvie
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Kailey J. Kowalski
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Utthara Nayar
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Harvard Medical School, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA
| | - Adrienne G. Waks
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Harvard Medical School, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA
| | | | | | | | | | | | | | | | | | | | | | - Gerald Batist
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Canada
| | - Melissa E. Hughes
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Levi A. Garraway
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Harvard Medical School, Boston, MA,Broad Institute of MIT and Harvard, Cambridge, MA,Eli Lilly and Co., Indianapolis, IN
| | - Eric P. Winer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Harvard Medical School, Boston, MA
| | - Sara M. Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Harvard Medical School, Boston, MA
| | - Nancy U. Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA,Harvard Medical School, Boston, MA
| | | | - Nikhil Wagle
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|
104
|
Bafna D, Ban F, Rennie PS, Singh K, Cherkasov A. Computer-Aided Ligand Discovery for Estrogen Receptor Alpha. Int J Mol Sci 2020; 21:E4193. [PMID: 32545494 PMCID: PMC7352601 DOI: 10.3390/ijms21124193] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/30/2020] [Accepted: 06/09/2020] [Indexed: 02/08/2023] Open
Abstract
Breast cancer (BCa) is one of the most predominantly diagnosed cancers in women. Notably, 70% of BCa diagnoses are Estrogen Receptor α positive (ERα+) making it a critical therapeutic target. With that, the two subtypes of ER, ERα and ERβ, have contrasting effects on BCa cells. While ERα promotes cancerous activities, ERβ isoform exhibits inhibitory effects on the same. ER-directed small molecule drug discovery for BCa has provided the FDA approved drugs tamoxifen, toremifene, raloxifene and fulvestrant that all bind to the estrogen binding site of the receptor. These ER-directed inhibitors are non-selective in nature and may eventually induce resistance in BCa cells as well as increase the risk of endometrial cancer development. Thus, there is an urgent need to develop novel drugs with alternative ERα targeting mechanisms that can overcome the limitations of conventional anti-ERα therapies. Several functional sites on ERα, such as Activation Function-2 (AF2), DNA binding domain (DBD), and F-domain, have been recently considered as potential targets in the context of drug research and discovery. In this review, we summarize methods of computer-aided drug design (CADD) that have been employed to analyze and explore potential targetable sites on ERα, discuss recent advancement of ERα inhibitor development, and highlight the potential opportunities and challenges of future ERα-directed drug discovery.
Collapse
Affiliation(s)
| | | | | | | | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; (D.B.); (F.B.); (P.S.R.); (K.S.)
| |
Collapse
|
105
|
Lipton NJ, Jesin J, Warner E, Cao X, Kiss A, Desautels D, Jerzak KJ. Willingness of women with early estrogen receptor-positive breast cancer to take adjuvant CDK4/6 inhibitors. Curr Oncol 2020; 27:127-134. [PMID: 32669921 PMCID: PMC7339836 DOI: 10.3747/co.27.6131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background The steady decline in breast cancer (bca) mortality has come at the cost of increasingly toxic and expensive adjuvant therapies. Trials evaluating the addition of 2 or 3 years of cyclin-dependent kinase 4/6 (cdk4/6) inhibitors to adjuvant endocrine therapy (et) are ongoing, but the willingness of patients to take such additional therapy is unknown. Methods We surveyed 100 consecutive postmenopausal women with nonmetastatic estrogen receptor-positive bca who had initiated adjuvant et within the preceding 2 years. Participants were asked about perceived recurrence risk, bca worry, and overall health. They were then asked about their willingness to accept 2 years of treatment with an additional oral drug that would reduce recurrence by 40% for a range of baseline recurrence risks in 2 hypothetical scenarios. Results Mean age of the 99 evaluable participants was 61.7 years. In the scenario with no drug toxicity, 85% of respondents were likely to accept the new drug for a reduction in recurrence to 30% from 50%, but only 49% would take the drug if risk was reduced to 3% from 5%. In a scenario with drug-induced fatigue, the corresponding drug acceptance rates were 55% and 39% respectively. For the second scenario, bca worry was correlated with increased willingness to take the drug, even for only a 2% absolute reduction in recurrence risk. Conclusions The willingness of patients with estrogen receptor-positive bca to take an adjuvant cdk4/6 inhibitor will greatly depend on the expected benefit and toxicities described to them as well as on worry about bca recurrence.
Collapse
Affiliation(s)
- N J Lipton
- Division of Medical Oncology, Sunnybrook Odette Cancer Centre, Toronto, ON
| | - J Jesin
- Division of Medical Oncology, Sunnybrook Odette Cancer Centre, Toronto, ON
| | - E Warner
- Division of Medical Oncology, Sunnybrook Odette Cancer Centre, Toronto, ON
- Sunnybrook Research Institute, Toronto, ON
| | - X Cao
- Sunnybrook Research Institute, Toronto, ON
| | - A Kiss
- Sunnybrook Research Institute, Toronto, ON
| | - D Desautels
- CancerCare Manitoba, University of Manitoba, Winnipeg, MB
- Department of Medicine, University of Manitoba, Winnipeg, MB
| | - K J Jerzak
- Division of Medical Oncology, Sunnybrook Odette Cancer Centre, Toronto, ON
- Sunnybrook Research Institute, Toronto, ON
| |
Collapse
|
106
|
Vogeser M. From therapeutic drug monitoring to total drug monitoring and drug-omics. ACTA ACUST UNITED AC 2020; 59:287-290. [DOI: 10.1515/cclm-2020-0339] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/11/2020] [Indexed: 11/15/2022]
Abstract
Abstract
In view of the role of pharmacotherapy in medicine, on the one hand, and the powerful technical possibilities that are now available on the other hand, therapeutic drug monitoring is a surprisingly neglected area of laboratory medicine. In this viewpoint article, an “omics approach” to pharmacovigilance and drug monitoring is proposed and discussed. A realistic goal for laboratory medicine in the 21st century should indeed be to enable clinicians to check whether the right drug is present in the right patient with an appropriate blood concentration for each compound.
Collapse
Affiliation(s)
- Michael Vogeser
- Institute of Laboratory Medicine , Hospital of the University of Munich (LMU) , Marchioninistr. 15 , 81377 Munich , Germany
| |
Collapse
|
107
|
Iorfida M, Mazza M, Munzone E. Fulvestrant in Combination with CDK4/6 Inhibitors for HER2- Metastatic Breast Cancers: Current Perspectives. BREAST CANCER-TARGETS AND THERAPY 2020; 12:45-56. [PMID: 32256106 PMCID: PMC7090187 DOI: 10.2147/bctt.s196240] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/08/2020] [Indexed: 11/23/2022]
Abstract
The development of CDK 4/6 inhibitors has dramatically changed the therapeutic management of hormone receptor-positive (HR+) and HER2 negative metastatic breast cancer (MBC). In combination with fulvestrant, palbociclib, ribociclib and abemaciclib have each been approved for HR+/HER2- MBC following the results of randomized Phase III studies (PALOMA-3, MONALEESA-3, MONARCH-2) and shown a significant advantage in PFS. Data from clinical trials support the combination with aromatase inhibitors in the first line setting and with fulvestrant in the second line. Each agent is well tolerated, and most of the toxicities observed with this class of drugs are generally easily manageable and free from particular complications. The latest evidence from MONARCH-2 and MONALEESA-3 trials shows benefits in terms of overall survival (OS), suggesting an option of using fulvestrant in combination with CDK 4/6 inhibitors in the first line setting. Additional research is needed to determine optimal treatment sequencing, understand the mechanisms of resistance, and develop novel therapeutic strategies to overcome clinical resistance and further improve the outcomes of patients with HR+/HER- MBC. Key questions in the field include the further impact on progression-free survival, overall survival, and the role of continuing CDK 4/6 blockade beyond progression. The purpose of this review is to describe the clinical relevance of fulvestrant in combination with CDK 4/6 inhibitors in HR+/HER2- MBC patients, as well as to discuss the current controversies and evolving research areas.
Collapse
Affiliation(s)
- Monica Iorfida
- Division of Medical Senology, European Institute of Oncology, IRCCS, Milano 20141, Italy
| | - Manuelita Mazza
- Division of Medical Senology, European Institute of Oncology, IRCCS, Milano 20141, Italy
| | - Elisabetta Munzone
- Division of Medical Senology, European Institute of Oncology, IRCCS, Milano 20141, Italy
| |
Collapse
|
108
|
Steinebach C, Ng YLD, Sosič I, Lee CS, Chen S, Lindner S, Vu LP, Bricelj A, Haschemi R, Monschke M, Steinwarz E, Wagner KG, Bendas G, Luo J, Gütschow M, Krönke J. Systematic exploration of different E3 ubiquitin ligases: an approach towards potent and selective CDK6 degraders. Chem Sci 2020; 11:3474-3486. [PMID: 33133483 PMCID: PMC7552917 DOI: 10.1039/d0sc00167h] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 02/28/2020] [Indexed: 12/18/2022] Open
Abstract
Cyclin-dependent kinase 6 (CDK6) is an important regulator of the cell cycle. Together with CDK4, it phosphorylates and inactivates retinoblastoma (Rb) protein.
Cyclin-dependent kinase 6 (CDK6) is an important regulator of the cell cycle. Together with CDK4, it phosphorylates and inactivates retinoblastoma (Rb) protein. In tumour cells, CDK6 is frequently upregulated and CDK4/6 kinase inhibitors like palbociclib possess high activity in breast cancer and other malignancies. Besides its crucial catalytic function, kinase-independent roles of CDK6 have been described. Therefore, targeted degradation of CDK6 may be advantageous over kinase inhibition. Proteolysis targeting chimeras (PROTACs) structurally based on the cereblon (CRBN) ligand thalidomide have recently been described to degrade the targets CDK4/6. However, CRBN-based PROTACs have several limitations including the remaining activity of immunomodulatory drugs (IMiDs) on Ikaros transcription factors as well as CRBN inactivation as a resistance mechanism in cancer. Here, we systematically explored the chemical space of CDK4/6 PROTACs by addressing different E3 ligases and connecting their respective small-molecule binders via various linkers to palbociclib. The spectrum of CDK6-specific PROTACs was extended to von Hippel Lindau (VHL) and cellular inhibitor of apoptosis protein 1 (cIAP1) that are essential for most cancer cells and therefore less likely to be inactivated. Our VHL-based PROTAC series included compounds that were either specific for CDK6 or exhibited dual activity against CDK4 and CDK6. IAP-based PROTACs caused a combined degradation of CDK4/6 and IAPs resulting in synergistic effects on cancer cell growth. Our new degraders showed potent and long-lasting degrading activity in human and mouse cells and inhibited proliferation of several leukemia, myeloma and breast cancer cell lines. In conclusion, we show that VHL- and IAP-based PROTACs are an attractive approach for targeted degradation of CDK4/6 in cancer.
Collapse
Affiliation(s)
- Christian Steinebach
- Pharmaceutical Institute , Department of Pharmaceutical & Medicinal Chemistry , University of Bonn , An der Immenburg 4 , 53121 Bonn , Germany .
| | - Yuen Lam Dora Ng
- Department of Internal Medicine III , University Hospital Ulm , Albert-Einstein-Allee 23 , 89081 Ulm , Germany .
| | - Izidor Sosič
- Faculty of Pharmacy , University of Ljubljana , Aškerčeva cesta 7 , 1000 Ljubljana , Slovenia
| | - Chih-Shia Lee
- Laboratory of Cancer Biology and Genetics , Center for Cancer Research , National Cancer Institute , Bethesda , MD 20892 , USA
| | - Sirui Chen
- Department of Internal Medicine III , University Hospital Ulm , Albert-Einstein-Allee 23 , 89081 Ulm , Germany .
| | - Stefanie Lindner
- Department of Internal Medicine III , University Hospital Ulm , Albert-Einstein-Allee 23 , 89081 Ulm , Germany .
| | - Lan Phuong Vu
- Pharmaceutical Institute , Department of Pharmaceutical & Medicinal Chemistry , University of Bonn , An der Immenburg 4 , 53121 Bonn , Germany .
| | - Aleša Bricelj
- Faculty of Pharmacy , University of Ljubljana , Aškerčeva cesta 7 , 1000 Ljubljana , Slovenia
| | - Reza Haschemi
- Pharmaceutical Institute , Department of Pharmaceutical & Cell Biological Chemistry , University of Bonn , An der Immenburg 4 , 53121 Bonn , Germany
| | - Marius Monschke
- Pharmaceutical Institute , Pharmaceutical Technology , University of Bonn , Gerhard-Domagk-Straße 3 , 53121 Bonn , Germany
| | - Elisabeth Steinwarz
- Pharmaceutical Institute , Department of Pharmaceutical & Cell Biological Chemistry , University of Bonn , An der Immenburg 4 , 53121 Bonn , Germany
| | - Karl G Wagner
- Pharmaceutical Institute , Pharmaceutical Technology , University of Bonn , Gerhard-Domagk-Straße 3 , 53121 Bonn , Germany
| | - Gerd Bendas
- Pharmaceutical Institute , Department of Pharmaceutical & Cell Biological Chemistry , University of Bonn , An der Immenburg 4 , 53121 Bonn , Germany
| | - Ji Luo
- Laboratory of Cancer Biology and Genetics , Center for Cancer Research , National Cancer Institute , Bethesda , MD 20892 , USA
| | - Michael Gütschow
- Pharmaceutical Institute , Department of Pharmaceutical & Medicinal Chemistry , University of Bonn , An der Immenburg 4 , 53121 Bonn , Germany .
| | - Jan Krönke
- Department of Internal Medicine III , University Hospital Ulm , Albert-Einstein-Allee 23 , 89081 Ulm , Germany .
| |
Collapse
|
109
|
Toda H, Seki N, Kurozumi S, Shinden Y, Yamada Y, Nohata N, Moriya S, Idichi T, Maemura K, Fujii T, Horiguchi J, Kijima Y, Natsugoe S. RNA-sequence-based microRNA expression signature in breast cancer: tumor-suppressive miR-101-5p regulates molecular pathogenesis. Mol Oncol 2020; 14:426-446. [PMID: 31755218 PMCID: PMC6998431 DOI: 10.1002/1878-0261.12602] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/05/2019] [Accepted: 11/19/2019] [Indexed: 12/24/2022] Open
Abstract
Aberrantly expressed microRNA (miRNA) are known to disrupt intracellular RNA networks in cancer cells. Exploring miRNA-dependent molecular networks is a major challenge in cancer research. In this study, we performed RNA-sequencing of breast cancer (BrCa) clinical specimens to identify tumor-suppressive miRNA in BrCa. In total, 64 miRNA were identified as candidate tumor-suppressive miRNA in BrCa cells. Analysis of our BrCa signature revealed that several miRNA duplexes (guide strand/passenger strand) derived from pre-miRNA were downregulated in BrCa tissues (e.g. miR-99a-5p/-3p, miR-101-5p/-3p, miR-126-5p/-3p, miR-143-5p/-3p, and miR-144-5p/-3p). Among these miRNA, we focused on miR-101-5p, the passenger strand of pre-miR-101, and investigated its tumor-suppressive roles and oncogenic targets in BrCa cells. Low expression of miR-101-5p predicted poor prognosis in patients with BrCa (overall survival rate: P = 0.0316). Ectopic expression of miR-101-5p attenuated aggressive phenotypes, e.g. proliferation, migration, and invasion, in BrCa cells. Finally, we identified seven putative oncogenic genes (i.e. High Mobility Group Box 3, Epithelial splicing regulatory protein 1, GINS complex subunit 1 (GINS1), Tumor Protein D52, Serine/Arginine-Rich Splicing Factor Kinase 1, Vang-like protein 1, and Mago Homolog B) regulated by miR-101-5p in BrCa cells. The expression of these target genes was associated with the molecular pathogenesis of BrCa. Furthermore, we explored the oncogenic roles of GINS1, whose function had not been previously elucidated, in BrCa cells. Aberrant expression of GINS1 mRNA and protein was observed in BrCa clinical specimens, and high GINS1 expression significantly predicted poor prognosis in patients with BrCa (overall survival rate: P = 0.0126). Knockdown of GINS1 inhibited the malignant features of BrCa cells. Thus, identification of tumor-suppressive miRNA and molecular networks controlled by these miRNA in BrCa cells may be an effective strategy for elucidation of the molecular pathogenesis of this disease.
Collapse
Affiliation(s)
- Hiroko Toda
- Department of Digestive Surgery, Breast and Thyroid SurgeryGraduate School of Medical and Dental SciencesKagoshima UniversityJapan
| | - Naohiko Seki
- Department of Functional GenomicsChiba University Graduate School of MedicineJapan
| | - Sasagu Kurozumi
- Department of General Surgical ScienceGunma University Graduate School of MedicineJapan
| | - Yoshiaki Shinden
- Department of Digestive Surgery, Breast and Thyroid SurgeryGraduate School of Medical and Dental SciencesKagoshima UniversityJapan
| | - Yasutaka Yamada
- Department of Functional GenomicsChiba University Graduate School of MedicineJapan
| | | | - Shogo Moriya
- Department of Biochemistry and GeneticsChiba University Graduate School of MedicineJapan
| | - Tetsuya Idichi
- Department of Digestive Surgery, Breast and Thyroid SurgeryGraduate School of Medical and Dental SciencesKagoshima UniversityJapan
| | - Kosei Maemura
- Department of Digestive Surgery, Breast and Thyroid SurgeryGraduate School of Medical and Dental SciencesKagoshima UniversityJapan
| | - Takaaki Fujii
- Department of General Surgical ScienceGunma University Graduate School of MedicineJapan
| | - Jun Horiguchi
- Department of Breast SurgeryInternational University of Health and WelfareChibaJapan
| | - Yuko Kijima
- Department of Digestive Surgery, Breast and Thyroid SurgeryGraduate School of Medical and Dental SciencesKagoshima UniversityJapan
- Department of Breast SurgeryFujita Health UniversityAichiJapan
| | - Shoji Natsugoe
- Department of Digestive Surgery, Breast and Thyroid SurgeryGraduate School of Medical and Dental SciencesKagoshima UniversityJapan
| |
Collapse
|
110
|
Zhang J, Wang Q, Wang Q, Cao J, Sun J, Zhu Z. Mechanisms of resistance to estrogen receptor modulators in ER+/HER2- advanced breast cancer. Cell Mol Life Sci 2020; 77:559-572. [PMID: 31471681 PMCID: PMC11105043 DOI: 10.1007/s00018-019-03281-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023]
Abstract
Endocrine therapy represents a mainstay adjuvant treatment of estrogen receptor-positive (ER+) breast cancer in clinical practice with an overall survival (OS) benefit. However, the emergence of resistance is inevitable over time and is present in one-third of the ER+ breast tumors. Several mechanisms of endocrine resistance in ER+/HER2- advanced breast cancers, through ERα itself, receptor tyrosine signaling, or cell cycle pathway, have been identified to be pivotal in endocrine therapy. The epigenetic alterations also contribute to ensuring tumor cells' escape from endocrine therapies. The strategy of combined hormone therapy with targeted pharmaceutical compounds has shown an improvement of progression-free survival or OS in clinical practice, including three different classes of drugs: CDK4/6 inhibitors, selective inhibitor of PI3Kα and mTOR inhibitors. Many therapeutic targets of cell cycle pathway and cell signaling and their combination strategies have recently entered clinical trials. This review focuses on Cyclin D-CDK4/6-RB axis, PI3K pathway and HDACs. Additionally, genomic evolution is complex in tumors exposed to hormonal therapy. We highlight the genomic alterations present in ESR1 and PIK3CA genes to elucidate adaptive mechanisms of endocrine resistance, and discuss how these mutations may inform novel combinations to improve clinical outcomes in the future.
Collapse
Affiliation(s)
- Jin Zhang
- Tianjin Key Laboratory of Protein Science, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qianying Wang
- Tianjin Key Laboratory of Protein Science, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qing Wang
- Tianjin Key Laboratory of Protein Science, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jiangran Cao
- Tianjin Key Laboratory of Protein Science, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jiafu Sun
- Tianjin Key Laboratory of Protein Science, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Zhengmao Zhu
- Tianjin Key Laboratory of Protein Science, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
111
|
Pozdeyev N, Rose MM, Bowles DW, Schweppe RE. Molecular therapeutics for anaplastic thyroid cancer. Semin Cancer Biol 2020; 61:23-29. [PMID: 31991166 DOI: 10.1016/j.semcancer.2020.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 02/07/2023]
Abstract
Anaplastic thyroid cancer (ATC) represents one of the most lethal human cancers and although this tumor type is rare, ATC accounts for the majority of deaths from thyroid cancer. Due to the rarity of ATC, a comprehensive genomic characterization of this tumor type has been challenging, and thus the development of new therapies has been lacking. To date, there is only one mutation-driven targeted therapy for BRAF-mutant ATC. Recent genomic studies have used next generation sequencing to define the genetic landscape of ATC in order to identify new therapeutic targets. Together, these studies have confirmed the role of oncogenic mutations of MAPK pathway as key drivers of differentiated thyroid cancer (BRAF, RAS), and that additional genetic alterations in the PI3K pathway, TP53, and the TERT promoter are necessary for anaplastic transformation. Recent novel findings have linked the high mutational burden associated with ATC with mutations in the Mismatch Repair (MMR) pathway and overactivity of the AID/APOBEC family of cytidine deaminases. Additional novel mutations include cell cycle genes, SWI/SNF chromatin remodeling complex, and histone modification genes. Mutations in RAC1 were also identified in ATC, which have important implications for BRAF-directed therapies. In this review, we summarize these novel findings and the new genetic landscape of ATC. We further discuss the development of therapies targeting these pathways that are being tested in clinical and preclinical studies.
Collapse
Affiliation(s)
- Nikita Pozdeyev
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, Aurora, CO, USA; Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, Aurora, CO, USA
| | - Madison M Rose
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, Aurora, CO, USA
| | - Daniel W Bowles
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rebecca E Schweppe
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, Aurora, CO, USA.
| |
Collapse
|
112
|
Sharma AV, Reddin G, Forrestal B, Barac A. Cardiovascular Disease Risk in Survivors of Breast Cancer. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2019; 21:79. [PMID: 31820123 DOI: 10.1007/s11936-019-0788-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Early detection and improved treatment in breast cancer have resulted in an increased number of survivors. Cardiovascular disease now remains an important cause for morbidity and mortality in this population. There is a growing gap in the knowledge about the optimal long-term cardiovascular management of this population. FINDINGS Breast cancer and cardiovascular disease share a number of common risk factors. Different breast cancer treatment modalities, including anthracyclines, radiation, and hormonal therapy, can act in synergy with preexisting and/or new cardiovascular risk factors to result in significant cardiovascular disease. We summarize the recent evidence about cardiovascular effects of breast cancer therapy and recommendations for their diagnosis and management during the cancer treatment continuum into survivorship. We also present current research initiatives and how they inform clinical care.
Collapse
Affiliation(s)
| | - Gemma Reddin
- MedStar Georgetown University Hospital, Washington, DC, USA
| | - Brian Forrestal
- MedStar Washington Hospital Center, Washington, DC, USA.,MedStar Georgetown University Hospital, Washington, DC, USA
| | - Ana Barac
- MedStar Washington Hospital Center, Washington, DC, USA. .,MedStar Georgetown University Hospital, Washington, DC, USA. .,MedStar Heart and Vascular Institute, Georgetown University, Washington, DC, USA.
| |
Collapse
|
113
|
Wander SA, Spring LM, Bardia A. Genetics to epigenetics: targeting histone deacetylases in hormone receptor-positive metastatic breast cancer. Lancet Oncol 2019; 20:746-748. [DOI: 10.1016/s1470-2045(19)30279-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/13/2022]
|
114
|
Abstract
Homology modeling is a computational approach to generate three-dimensional structures of protein targets when experimental data about similar proteins are available. Although experimental methods such as X-ray crystallography and nuclear magnetic resonance spectroscopy successfully solved the structures of nearly 150,000 macromolecules, there is still a gap in our structural knowledge. We can fulfill this gap with computational methodologies. Our goal in this chapter is to explain how to perform homology modeling of protein targets for drug development. We choose as a homology modeling tool the program MODELLER. To illustrate its use, we describe how to model the structure of human cyclin-dependent kinase 3 using MODELLER. We explain the modeling procedure of CDK3 apoenzyme and the structure of this enzyme in complex with roscovitine.
Collapse
Affiliation(s)
- Gabriela Bitencourt-Ferreira
- Escola de Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, RS, Brazil
| | - Walter Filgueira de Azevedo
- Escola de Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, RS, Brazil.
| |
Collapse
|