1
|
Yang SY, Jin ML, Andriani L, Zhao Q, Ling YX, Lin CJ, Huang MY, Cai JY, Zhang YL, Hu X, Shao ZM, Zhang FL, Jin X, Cao AY, Li DQ. Kinesin-like protein KIFC2 stabilizes CDK4 to accelerate growth and confer resistance in HR+/HER2- breast cancer. J Clin Invest 2025; 135:e183531. [PMID: 40299549 PMCID: PMC12165803 DOI: 10.1172/jci183531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 04/23/2025] [Indexed: 05/01/2025] Open
Abstract
Hormone receptor-positive and human epidermal growth factor receptor 2-negative breast cancer (HR+/HER2- BC) is the most common subtype, with a high risk of long-term recurrence and metastasis. Endocrine therapy (ET) combined with cyclin-dependent kinase 4/6 (CDK4/6) inhibitors is a standard treatment for advanced/metastatic HR+/HER2- BC, but resistance remains a major clinical challenge. We report that kinesin family member C2 (KIFC2) was amplified in approximately 50% of patients with HR+/HER2- BC, and its high expression was associated with poor disease outcome, increased tumor protein p53 (TP53) somatic mutation, and active pyrimidine metabolism. Functional assays revealed that depletion of KIFC2 suppressed growth and enhanced sensitivity of HR+/HER2- BC cells to tamoxifen and CDK4/6 inhibitors. Mechanistically, KIFC2 stabilized CDK4 by enhancing its interaction with ubiquitin-specific peptidase 9 X-linked (USP9X). Importantly, reexpression of CDK4 in KIFC2-depleted cells partially rescued the decreased growth and increased sensitivity to tamoxifen and CDK4/6 inhibitors caused by KIFC2 depletion. Clinically, high KIFC2 mRNA expression was negatively associated with the survival rate of patients with HR+/HER2- BC who received adjuvant ET alone or in combination with CDK4/6 inhibitors. Collectively, these findings identify an important role for KIFC2 in HR+/HER2- BC growth and therapeutic resistance, and support its potential as a therapeutic target and predictive biomarker.
Collapse
Affiliation(s)
- Shao-Ying Yang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ming-Liang Jin
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Lisa Andriani
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Qian Zhao
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yun-Xiao Ling
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cai-Jin Lin
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Min-Ying Huang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia-Yang Cai
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Yin-Ling Zhang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xin Hu
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhi-Ming Shao
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Breast Cancer, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fang-Lin Zhang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi Jin
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - A Yong Cao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Da-Qiang Li
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Breast Cancer, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Zhang J, Liu R, Gao S, Chen W, Han X, Wang Z, Zhou H, Wang Y, Chen J, Ma Y, Liu K, Shen Z, Ding L, Li P, Hu X. A phase I dose-escalation and dose-expansion study of tibremciclib, a novel CDK4/6 inhibitor, monotherapy and in combination with fulvestrant in HR-positive/HER2-negative advanced breast cancer. ESMO Open 2025; 10:105121. [PMID: 40403388 DOI: 10.1016/j.esmoop.2025.105121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Tibremciclib (BPI-16350) is a novel potent selective inhibitor of cyclin-dependent kinase 4 and 6 (CDK4/6). This phase I, first-in-human study assessed the tibremciclib monotherapy and combined with fulvestrant in advanced breast cancer (ABC). PATIENTS AND METHODS This open-label, phase I study (NCT03791112) comprised dose escalation (phase Ia) and expansion (phase Ib). In phase Ia, Chinese patients with advanced solid tumors received tibremciclib monotherapy (50-500 mg orally once daily). Based on the tolerability and preliminary antitumor activity from phase Ia, two dose cohorts (300 or 400 mg orally once daily) were selected in phase Ib to combine with fulvestrant in patients with hormone receptor (HR)-positive/human epidermal growth receptor 2 (HER2)-negative ABC who failed endocrine therapy. The primary endpoints were safety and tolerability, and secondary endpoints included pharmacokinetics (PK) and antitumor activity. RESULTS In phase Ia, 24 patients with advanced tumors (23 ABC and 1 abdominal liposarcoma) received tibremciclib monotherapy, while in phase Ib, 78 patients with HR-positive/HER2-negative stage IV ABC received tibremciclib plus fulvestrant, with a median age of 56.0 (range 26-66) years and 53.5 (range 33-71) years, respectively. In phase Ia, one dose-limiting toxicity (grade 3 blood creatinine increased) occurred at 500 mg and the maximum tolerated dose was not reached. In phase Ib, the recommended phase II dose (RP2D) was tibremciclib 400 mg plus fulvestrant 500 mg. Frequently reported treatment-emergent adverse events of any grade were grade 1-2 blood creatinine increased, hypertriglyceridemia, and anemia. PK demonstrates dose proportionality, a half-life of ∼35.9-51.1 h, and no drug-drug interaction with fulvestrant. In phase Ia, one patient achieved partial response (PR), with a confirmed objective response rate (ORR) of 4.2% and a disease control rate (DCR) of 70.8%. In phase Ib, 42 patients achieved PR, with an ORR of 53.8% and a DCR of 87.2%. CONCLUSIONS Tibremciclib monotherapy or combined with fulvestrant was well tolerated with the RP2D set at 400 mg plus fulvestrant 500 mg. PK was consistent with dose proportionality. Both regimens showed preliminary antitumor activity in patients with HR-positive/HER2-negative ABC, supporting further evaluation in a phase III study (NCT05433480).
Collapse
Affiliation(s)
- J Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Phase I Clinical Trial Center, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - R Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Phase I Clinical Trial Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - S Gao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Phase I Clinical Trial Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - W Chen
- Department of Breast Oncology, Nanchang Third Hospital, Nanchang, China
| | - X Han
- Tumor Chemotherapy Department, Anhui Provincial Hospital, Hefei, China
| | - Z Wang
- Surgical Oncology, The First Affiliate Hospital of Bengbu Medical University, Bengbu, China
| | - H Zhou
- Drug Clinical Trial Institution, The First Affiliate Hospital of Bengbu Medical University, Bengbu, China
| | - Y Wang
- Department of Breast Oncology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - J Chen
- Medical Oncology, Yantai Yuhuangding Hospital, Yantai, China
| | - Y Ma
- Betta Pharmaceuticals Co., Ltd., Hangzhou, China
| | - K Liu
- Betta Pharmaceuticals Co., Ltd., Hangzhou, China
| | - Z Shen
- Betta Pharmaceuticals Co., Ltd., Hangzhou, China
| | - L Ding
- Betta Pharmaceuticals Co., Ltd., Hangzhou, China
| | - P Li
- Betta Pharmaceuticals Co., Ltd., Hangzhou, China
| | - X Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Zhang J, Xu X, Zhou Y, Su J, Wang J. A meta-analysis and systematic review of different cyclin-dependent kinase 4/6 inhibitors in breast cancer. Front Oncol 2025; 15:1472407. [PMID: 40104496 PMCID: PMC11913826 DOI: 10.3389/fonc.2025.1472407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 02/13/2025] [Indexed: 03/20/2025] Open
Abstract
Objective The objective of this study was to assess the effectiveness and safety of CDK4/6 inhibitors in the treatment of hormone receptor-positive (HR+) breast cancer by using meta-analysis. Methods To gather comprehensive and reliable data for our analysis, we systematically searched multiple databases for relevant studies. We utilized RevMan5.3 software to perform the meta-analysis. Results Following a rigorous screening and evaluation process, we ultimately included a total of 13 studies in our analysis. Our findings showed that compared to endocrine therapy alone, the combination of CDK4/6 inhibitors with endocrine therapy significantly increased both PFS [HR 0.54 (95%CI: 0.50, 0.58), P<0.00001], OS [HR 0.77 (95%CI: 0.50, 0.58), P<0.00001] and ORR [RR 1.39 (95% CI: 1.21, 1.60), P<0.00001). However, it was also found that CDK4/6 inhibitors caused adverse drug reactions related to the blood system and digestive system (P<0.0001). Conclusions Our meta-analysis demonstrates that the addition of CDK4/6 inhibitors to endocrine therapy can result in improved PFS and OS for HR+ breast cancer patients. Meanwhile, we recommend close monitoring and management of these potential side effects when utilizing these inhibitors in breast cancer treatment. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO, identifier CRD42023490499.
Collapse
Affiliation(s)
- Jialin Zhang
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Xinyu Xu
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Yeyue Zhou
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Jingyang Su
- Department of General internal medicine, Tongde Hospital Affiliated to Zhejiang Chinese Medical University (Tongde Hospital of Zhejiang Province), Hangzhou, China
| | - Jue Wang
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
| |
Collapse
|
4
|
Kindt CK, Ehmsen S, Traynor S, Policastro B, Nissen N, Jakobsen MK, Hundebøl MF, Johansen LE, Bak M, Arbajian E, Staaf J, Ditzel HJ, Alves CL. RET inhibition overcomes resistance to combined CDK4/6 inhibitor and endocrine therapy in ER+ breast cancer. Front Oncol 2025; 14:1497093. [PMID: 39931212 PMCID: PMC11808005 DOI: 10.3389/fonc.2024.1497093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/18/2024] [Indexed: 02/13/2025] Open
Abstract
Background Combined CDK4/6 inhibitor (CDK4/6i) and endocrine therapy significantly improve the outcome of patients with advanced estrogen receptor-positive (ER+) breast cancer. However, resistance to this treatment and disease progression remains a major clinical challenge. High expression of the receptor tyrosine kinase REarranged during Transfection (RET) has been associated with resistance to endocrine therapy in breast cancer, but the role of RET in CDK4/6i treatment response/resistance remains unexplored. Methods To identify gene expression alterations associated with resistance to combined endocrine therapy and CDK4/6i, we performed RNA sequencing of two ER+ breast cancer cell models resistant to this combined therapy. The functional role of RET was assessed by siRNA-mediated RET silencing and targeted inhibition with the FDA/EMA-approved RET-selective inhibitor selpercatinib in resistant breast cancer cells and patient-derived organoids (PDOs). RET silencing was evaluated mechanistically using global gene expression and pathway analysis. The clinical relevance of RET expression in ER+ breast cancer was investigated by gene array analysis of primary tumors treated with endocrine therapy and by immunohistochemical scoring of metastatic lesions from patients who received combined CDK4/6i and endocrine therapy. Results We show that RET is upregulated in ER+ breast cancer cell lines resistant to combined CDK4/6i and fulvestrant compared to isogenic cells resistant to fulvestrant alone. siRNA-mediated silence of RET in high RET-expressing, combined CDK4/6i- and fulvestrant-resistant cells reduced their growth partially by affecting cell cycle regulators of the G2-M phase and E2F targets. Notably, targeting RET with selpercatinib in combination with CDK4/6i inhibited the growth of CDK4/6i-resistant cell lines and resensitized ER+ breast cancer patient-derived organoids resistant to CDK4/6i. Finally, analysis of RET expression in ER+ breast cancer patients treated with endocrine therapy showed that high RET expression correlated with poor clinical outcomes. We further observed a shorter median survival to combined CDK4/6i and endocrine therapy in patients with RET-positive compared to RET-negative tumors, but this difference did not reach statistical significance. Conclusions Our findings show that RET is overexpressed in ER+ metastatic breast cancer resistant to combined CDK4/6i and endocrine therapy, rendering RET inhibition a promising therapeutic approach for patients who experience disease progression on combined CDK4/6i and endocrine therapy.
Collapse
Affiliation(s)
- Charlotte K. Kindt
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sidse Ehmsen
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Sofie Traynor
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Benedetta Policastro
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Nikoline Nissen
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Mie K. Jakobsen
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Monique F. Hundebøl
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lene E. Johansen
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Martin Bak
- Department of Pathology, Sydvestjysk Sygehus, Esbjerg, Denmark
| | - Elsa Arbajian
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Henrik J. Ditzel
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Carla L. Alves
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
5
|
Zheng M, Xu J, Yu S, Zhao Z, Zhang Y, Wei M. Utility of Multiparametric Breast MRI Radiomics to Predict Cyclin D1 and TGF-β1 Expression. J Comput Assist Tomogr 2025:00004728-990000000-00408. [PMID: 39794899 DOI: 10.1097/rct.0000000000001717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 11/26/2024] [Indexed: 01/13/2025]
Abstract
OBJECTIVE To develop a machine learning model that integrates clinical features and multisequence MRI radiomics for noninvasively predicting the expression status of prognostic-related factors cyclin D1 and TGF-β1 in breast cancer, providing additional information for the clinical development of personalized treatment plans. METHODS A total of 123 breast cancer patients confirmed by surgical pathology were retrospectively enrolled in our Hospital from January 2016 to July 2022. The patients were randomly divided into a training group (87 cases) and a validation group (36 cases). Preoperative routine and dynamic contrast-enhanced magnetic resonance imaging scans of the breast were performed for treatment subjects. The region of interest was manually outlined, and texture features were extracted using AK software. Subsequently, the LASSO algorithm was employed for dimensionality reduction and feature selection to establish the MRI radiomics labels. The diagnostic efficacy and clinical value were assessed through receiver operating characteristic (ROC) curve analysis and decision curve analysis (DCA). RESULTS In the cyclin D1 cohort, the area under the receiver operating characteristic (ROC) curve in the clinical prediction model training and validation groups was 0.738 and 0.656, respectively. The multisequence MRI radiomics prediction model achieved an AUC of 0.874 and 0.753 in these respective groups, while the combined prediction model yielded an AUC of 0.892 and 0.785. In the TGF-β1 cohort, the ROC AUC for the clinical prediction model was found to be 0.693 and 0.645 in the training and validation groups, respectively. For the multiseries MRI radiomics prediction model, it achieved an AUC of 0.875 and 0.760 in these respective groups; whereas for the combined prediction model, it reached an AUC of 0.904 and 0.833. Decision curve analysis (DCA) demonstrated that both cohorts indicated a higher clinical application value for the combined prediction model compared with both individual models-clinical prediction model alone or radiomics model. CONCLUSION The integration of clinical features and multisequence MRI radiomics in a combined modeling approach holds significant predictive value for the expression status of cyclin D1 and TGF-β1. The model provides a noninvasive, dynamic evaluation method that provides effective guidance for clinical treatment.
Collapse
Affiliation(s)
| | - Jiaqi Xu
- School of Medicine, Shaoxing University
| | - Shujie Yu
- School of Medicine, Shaoxing University
| | - Zhenhua Zhao
- Department of Radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| | - Yu Zhang
- Department of Radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| | - Mingzhu Wei
- Department of Radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| |
Collapse
|
6
|
Migliaccio I, Guarducci C, Malorni L. CDK4/6 Inhibitor Resistance in ER+ Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:475-493. [PMID: 39821039 DOI: 10.1007/978-3-031-70875-6_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
The cyclin-dependent kinases 4 and 6 inhibitors are the mainstay of treatment for patients with hormone receptor-positive and HER2-negative breast cancer. The ability of these drugs to improve the outcome of patients both in the metastatic and the early setting has been largely demonstrated. However, resistance, either de novo or acquired, represents a major clinical challenge. In the past years, efforts have been made to identify biomarkers that might help in a better selection of patients or to unravel the mechanisms leading to resistance in order to develop new therapeutic strategies to overcome it. Alterations of cell cycle-related genes and proteins are among the best characterized markers of resistance, and pathways impacting the cell cycle, including nuclear and growth factor receptors signaling, have been thoroughly investigated. Despite this, to date, cyclin-dependent kinases 4 and 6 inhibitors are administered based only on the hormone receptor and HER2 status of the tumor, and patients progressing on therapy are managed with currently available treatments. Here we summarize present knowledge on the cyclin-dependent kinases 4 and 6 inhibitors' mechanisms of action, efficacy data, and mechanisms of resistance.
Collapse
Affiliation(s)
- Ilenia Migliaccio
- Translational Research Unit, Hospital of Prato, AUSL Toscana Centro, Prato, Italy
| | - Cristina Guarducci
- Translational Research Unit, Hospital of Prato, AUSL Toscana Centro, Prato, Italy
| | - Luca Malorni
- Translational Research Unit, Hospital of Prato, AUSL Toscana Centro, Prato, Italy.
| |
Collapse
|
7
|
Gültürk I, Colak R, Kapar C, Guliyev M, Yıldırım C, Yilmaz M. Could macrocytosis predict survival In advanced breast cancer patients that were treated with CDK 4-6 inhibitors? Breast 2024; 78:103820. [PMID: 39405594 PMCID: PMC11752110 DOI: 10.1016/j.breast.2024.103820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/06/2024] [Accepted: 10/06/2024] [Indexed: 01/26/2025] Open
Abstract
INTRODUCTION Cyclin Dependent Kinase (CDK) 4-6 inhibitors are the recommended first-line treatment option for hormone-positive metastatic breast cancer (MBC). They show their effects by causing cell cycle arrest in G1-S phase. Neutropenia is the most common haematological side effect. In the literature, data on the association between CDK 4-6 inhibitors and macrocytosis are limited. We aimed to investigate the effect of macrocytosis on survival. METHODS We retrospectively analysed 133 patients with de novo hormone positive MBC using CDK 4-6 inhibitors in first line treatment. Mean Corpuscular Volume (MCV) > 100 was considered macrocytosis and patients were divided into two groups; MCV<100 and MCV >100. The association of macrocytosis with clinicopathological features, Progression Free Survival (PFS) and Overall Survival (OS) were evaluated. RESULTS 42 patients were receiving palbociclib and 81 patients were receiving ribociclib. Median OS was determined as 33 months and median PFS was determined as 22 months. Macrocytosis ever rate was 45.8 % during follow-up. Macrocytosis was observed in 4.2 % of the patients in the first month, 16.7 % in the third month, 41.6 % in the sixth month and 42.2 % in the twelfth month. ER receptor level, ki-67, macrocytosis at 6-12 months and macrocytosis-ever which were found to affect OS as a result of univariate Cox regression analysis, were evaluated with multivariate Cox regression models and it was observed that they had significant effect on PFS and OS. CONCLUSION Macrocytosis may be a useful biomarker for the prediction of PFS and OS in MBC patients receiving CDK 4-6 inhibitors.
Collapse
Affiliation(s)
- Ilkay Gültürk
- Istanbul Research and Training Hospital, Department of Medical Oncology, Istanbul, Turkey.
| | - Rumeysa Colak
- Bakirkoy Dr. Sadi Konuk Research and Training Hospital, Department of Medical Oncology, Istanbul, Turkey
| | - Caner Kapar
- Bakirkoy Dr. Sadi Konuk Research and Training Hospital, Department of Medical Oncology, Istanbul, Turkey
| | - Murad Guliyev
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department of Medical Oncology, Istanbul, Turkey
| | - Cigdem Yıldırım
- Istanbul Research and Training Hospital, Department of Medical Oncology, Istanbul, Turkey
| | - Mesut Yilmaz
- Bakirkoy Dr. Sadi Konuk Research and Training Hospital, Department of Medical Oncology, Istanbul, Turkey
| |
Collapse
|
8
|
Łupicka-Słowik A, Cossu F, Sieńczyk M. Palbociclib as an Antitumor Drug: A License to Kill. Molecules 2024; 29:5334. [PMID: 39598723 PMCID: PMC11596203 DOI: 10.3390/molecules29225334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
Neoplastic cells are characterized by uncontrolled cell divisions caused by cell cycle dysregulation. Key regulatory proteins governing the transition from the G1 to the S phase are the CDK4 and CDK6 kinases, which are controlled by D-type cyclins. The CDK4/6 kinases enable the use of these proteins as targets for anticancer therapy because they prevent the growth and the development of malignant cells by inhibiting their activity. This paper surveys the clinical trial results concerning palbociclib, the first in-class FDA-approved anticancer drug for hormone-dependent breast cancer. It discusses the therapeutic applications in breast cancer as well as in solid tumors and hematopoietic malignancies. Additionally, the paper presents an analysis of palbociclib resistance acquired during therapy and explores new approaches, such as modifications to palbociclib that enhance its desired activity or open up new therapeutic possibilities (PROTACs).
Collapse
Affiliation(s)
- Agnieszka Łupicka-Słowik
- Division of Organic and Medicinal Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland;
| | - Federica Cossu
- National Research Council, Institute of Biophysics (IBF-CNR), Milan Unit, Via Corti, 12, 20133 Milan, Italy;
| | - Marcin Sieńczyk
- Division of Organic and Medicinal Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland;
| |
Collapse
|
9
|
Ruckhäberle E, Schmidt M, Welt A, Harbeck N, Wöckel A, Gluz O, Park-Simon TW, Untch M, Lux MP. Palbociclib: Randomized Studies and Real-world Evidence as the Basis for Therapeutic Planning in Metastatic Breast Cancer. Geburtshilfe Frauenheilkd 2024; 84:813-836. [PMID: 39229634 PMCID: PMC11368471 DOI: 10.1055/a-2344-5269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/11/2024] [Indexed: 09/05/2024] Open
Abstract
Endocrine-based combination therapy with an inhibitor of the cyclin-dependent kinases 4 and 6 (CDK4/6 inhibitors) is currently the first-line therapy of choice for patients with hormone receptor-positive (HR+) and human epidermal growth factor receptor 2-negative (HER2-), locally advanced or metastatic breast cancer (mBC). The efficacy and safety of the treatment with palbociclib, the first CDK4/6 inhibitor approved for this indication, have been confirmed in large randomized controlled clinical trials (RCTs) with strictly defined patient cohorts. Since then, many relevant questions about CDK4/6 inhibition with palbociclib for mBC have been investigated in RCTs and real-world studies. Based on this evidence, palbociclib is widely used in clinical practice since many years because of its efficacy and good tolerability. The aim of this review is to summarize findings from RCTs and RWE considering clinically relevant aspects such as safety, tolerability, quality of life and efficacy with a focus on specific questions and patient characteristics. A critical discussion and review of the overall evidence for endocrine-based therapy with the CDK4/6 inhibitor palbociclib can contribute to support therapy decisions in daily clinical practice.
Collapse
Affiliation(s)
- Eugen Ruckhäberle
- Universitätsfrauenklinik Düsseldorf, Heinrich-Heine-Universität, CIO ABCD, Düsseldorf, Germany
| | - Marcus Schmidt
- Klinik und Poliklinik für Geburtshilfe und Frauengesundheit, Brustzentrum, Universitätsmedizin Mainz, Mainz, Germany
| | - Anja Welt
- Innere Klinik (Tumorforschung), Universitätsklinikum Essen, Essen, Germany
| | - Nadia Harbeck
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Brustzentrum, LMU Klinikum, München, Germany
| | - Achim Wöckel
- Frauenklinik und Poliklinik, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Oleg Gluz
- Ev. Krankenhaus Bethesda Mönchengladbach, Brustzentrum Niederrhein, Mönchengladbach, Germany
| | - Tjoung-Won Park-Simon
- Klinik für Frauenheilkunde und Geburtshilfe, Medizinische Hochschule Hannover, Hannover, Germany
| | - Michael Untch
- Fachabteilung Gynäkologie, Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Michael P. Lux
- Klinik für Gynäkologie und Geburtshilfe, Frauenklinik St. Louise, Paderborn, Frauenklinik St. Josefs-Krankenhaus, Salzkotten, St. Vincenz Kliniken Salzkotten + Paderborn, Paderborn, Germany
| |
Collapse
|
10
|
Nakamoto S, Shien T, Iwamoto T, Kubo S, Yamamoto M, Yamashita T, Kuwahara C, Ikeda M. Absolute lymphocyte count and neutrophil-to-lymphocyte ratio as predictors of CDK 4/6 inhibitor efficacy in advanced breast cancer. Sci Rep 2024; 14:9869. [PMID: 38684839 PMCID: PMC11059159 DOI: 10.1038/s41598-024-60101-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
Cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) are the standard agents for treating patients with estrogen receptor-positive and human epidermal growth factor receptor 2-negative advanced breast cancer (ER + HER2 - ABC). However, markers predicting the outcomes of CDK4/6i treatment have yet to be identified. This study was a single-center retrospective cohort study. We retrospectively evaluated 101 patients with ER + HER2 - ABC receiving CDK4/6i in combination with endocrine therapy at Fukuyama City Hospital between November 2017 and July 2021. We investigated the clinical outcomes and the safety of CDK4/6i treatment, and the absolute lymphocyte count (ALC) and neutrophil-to-lymphocyte ratio (NLR) as predictive markers for CDK4/6i. We defined the cut-off values as 1000/μL for ALC and 3 for NLR, and divided into "low" and "high" groups, respectively. We evaluated 43 and 58 patients who received abemaciclib and palbociclib, respectively. Patients with high ALC and low NLR had significantly longer overall survival than those with low ALC and high NLR (high vs. low; ALC: HR 0.29; 95% CI 0.12-0.70; NLR: HR 2.94; 95% CI 1.21-7.13). There was no significant difference in efficacy between abemaciclib and palbociclib and both had good safety profiles. We demonstrated that ALC and NLR might predict the outcomes of CDK4/6i treatment in patients with ER + HER2 - ABC.
Collapse
Affiliation(s)
- Shogo Nakamoto
- Department of Breast and Thyroid Surgery, Fukuyama City Hospital, 5-23-1 Zao, Fukuyama, 721-8511, Japan.
- Department of Breast and Endocrine Surgery, Okayama University Hospital, 2-5-1, Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan.
| | - Tadahiko Shien
- Department of Breast and Endocrine Surgery, Okayama University Hospital, 2-5-1, Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Takayuki Iwamoto
- Department of Breast and Endocrine Surgery, Okayama University Hospital, 2-5-1, Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Shinichiro Kubo
- Department of Breast and Thyroid Surgery, Fukuyama City Hospital, 5-23-1 Zao, Fukuyama, 721-8511, Japan
| | - Mari Yamamoto
- Department of Breast and Thyroid Surgery, Fukuyama City Hospital, 5-23-1 Zao, Fukuyama, 721-8511, Japan
| | - Tetsumasa Yamashita
- Department of Breast and Thyroid Surgery, Fukuyama City Hospital, 5-23-1 Zao, Fukuyama, 721-8511, Japan
| | - Chihiro Kuwahara
- Department of Breast and Thyroid Surgery, Fukuyama City Hospital, 5-23-1 Zao, Fukuyama, 721-8511, Japan
| | - Masahiko Ikeda
- Department of Breast and Thyroid Surgery, Fukuyama City Hospital, 5-23-1 Zao, Fukuyama, 721-8511, Japan
| |
Collapse
|
11
|
Wang J, Li B, Luo M, Huang J, Zhang K, Zheng S, Zhang S, Zhou J. Progression from ductal carcinoma in situ to invasive breast cancer: molecular features and clinical significance. Signal Transduct Target Ther 2024; 9:83. [PMID: 38570490 PMCID: PMC10991592 DOI: 10.1038/s41392-024-01779-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Ductal carcinoma in situ (DCIS) represents pre-invasive breast carcinoma. In untreated cases, 25-60% DCIS progress to invasive ductal carcinoma (IDC). The challenge lies in distinguishing between non-progressive and progressive DCIS, often resulting in over- or under-treatment in many cases. With increasing screen-detected DCIS in these years, the nature of DCIS has aroused worldwide attention. A deeper understanding of the biological nature of DCIS and the molecular journey of the DCIS-IDC transition is crucial for more effective clinical management. Here, we reviewed the key signaling pathways in breast cancer that may contribute to DCIS initiation and progression. We also explored the molecular features of DCIS and IDC, shedding light on the progression of DCIS through both inherent changes within tumor cells and alterations in the tumor microenvironment. In addition, valuable research tools utilized in studying DCIS including preclinical models and newer advanced technologies such as single-cell sequencing, spatial transcriptomics and artificial intelligence, have been systematically summarized. Further, we thoroughly discussed the clinical advancements in DCIS and IDC, including prognostic biomarkers and clinical managements, with the aim of facilitating more personalized treatment strategies in the future. Research on DCIS has already yielded significant insights into breast carcinogenesis and will continue to pave the way for practical clinical applications.
Collapse
Affiliation(s)
- Jing Wang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Baizhou Li
- Department of Pathology, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Meng Luo
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
- Department of Plastic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Huang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Kun Zhang
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu Zheng
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Suzhan Zhang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
| | - Jiaojiao Zhou
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
12
|
Fiste O, Mavrothalassitis E, Apostolidou K, Trika C, Liontos M, Koutsoukos K, Kaparelou M, Dimitrakakis C, Gavriatopoulou M, Dimopoulos MA, Zagouri F. Cardiovascular complications of ribociclib in breast cancer patients. Crit Rev Oncol Hematol 2024; 196:104296. [PMID: 38395242 DOI: 10.1016/j.critrevonc.2024.104296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors have unprecedentedly advanced hormone-dependent breast cancer treatment paradigm. In the metastatic setting, ribociclib has consistently demonstrated survival benefit in pre-, peri-, and postmenopausal patients, conjugating efficacy with health-related quality of life preservation. Accordingly, the emergence of cardiac and/or vascular adverse events related to this novel targeted agent is gaining significant interest. This narrative review provides an overview of the incidence and spectrum of cardiovascular toxicity, in both clinical trial framework and real-world evidence. The potential pathogenetic mechanism, along with the available diagnostic parameters including biomarkers, and proper management, are also summarized.
Collapse
Affiliation(s)
- Oraianthi Fiste
- Oncology Unit, Third Department of Internal Medicine and Laboratory, National and Kapodistrian University of Athens, Sotiria General Hospital, Athens 11527, Greece.
| | | | - Kleoniki Apostolidou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Alexandra Hospital, Athens 11528, Greece
| | - Chrysanthi Trika
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Alexandra Hospital, Athens 11528, Greece
| | - Michalis Liontos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Alexandra Hospital, Athens 11528, Greece
| | - Konstantinos Koutsoukos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Alexandra Hospital, Athens 11528, Greece
| | - Maria Kaparelou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Alexandra Hospital, Athens 11528, Greece
| | - Constantine Dimitrakakis
- First Department of Obstetrics and Gynecology, Alexandra University Hospital, National and Kapodistrian University of Athens, Athens 11528, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Alexandra Hospital, Athens 11528, Greece
| | - Meletios Athanasios Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Alexandra Hospital, Athens 11528, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Alexandra Hospital, Athens 11528, Greece
| |
Collapse
|
13
|
Rodrigues Alves N, Duarte AF, Ribeiro DF, Silva RS, Carvalho BA, Alpuim Costa D. Successful management of bilateral orbital metastases from invasive lobular breast cancer with abemaciclib and letrozole: a case report and literature review. Front Oncol 2024; 14:1286910. [PMID: 38322413 PMCID: PMC10845133 DOI: 10.3389/fonc.2024.1286910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/03/2024] [Indexed: 02/08/2024] Open
Abstract
Breast cancer is a significant global health concern, contributing to substantial morbidity and mortality among women. Hormone receptor-positive (HR+)/HER2-negative (HER2-) breast cancer constitutes a considerable proportion of cases, and significant advancements have been made in its management. CDK4/6 inhibitors (CDK4/6is) are a new targeted therapy that has demonstrated efficacy in adjuvant, advanced and metastatic settings. The propensity of lobular breast carcinomas for estrogen-rich sites, such as periocular tissues and orbital fat, may explain their tendency for orbital metastases. Current treatment strategies for these cases are predominantly palliative, and the prognosis remains poor. This article presents a unique case of a 51-year-old female with progressive right periorbital edema, pain, and limited ocular motility. An imaging work-up showed bilateral intra and extraconal orbital infiltration, which was biopsied. The histopathologic analysis disclosed mild chronic inflammatory infiltrate with thickened fibrous tissue and moderately differentiated lobular carcinoma cells, positive for GATA3 and CK7 markers, with 100% of tumor nuclei expressing estrogen receptors (ER+). A systemic evaluation showed a multicentric nodular formation in both breasts. Further diagnostic assessments unveiled an HR+/HER2- bilateral lobular breast carcinoma with synchronous bilateral orbital metastases. Systemic treatment was initiated with abemaciclib 150mg twice daily and letrozole 2.5mg once a day. However, this regimen was interrupted due to toxicity. After two weeks, treatment was resumed with a reduced abemaciclib dose (100mg twice daily) alongside letrozole, with a reasonable tolerance. Nearly two years after the initial diagnosis of inoperable metastatic cancer, the patient remains on the same systemic treatment regimen with no signs of invasive disease. This case report is the first of a patient presenting with bilateral orbital metastases from bilateral lobular breast cancer, showing an impressive and sustained response to a first-line treatment regimen combining abemaciclib and letrozole. A literature review on bilateral orbital metastases from breast cancer is also presented.
Collapse
Affiliation(s)
- Nuno Rodrigues Alves
- Department of Ophthalmology, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- Department of Ophthalmology, Unidade Local de Saúde de São José, Lisbon, Portugal
| | - Ana Filipa Duarte
- Department of Ophthalmology, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- Department of Ophthalmology, Unidade Local de Saúde de São José, Lisbon, Portugal
- Department of Ophthalmology, Hospital CUF Descobertas, Lisbon, Portugal
| | | | - Rita Sousa Silva
- Department of Ophthalmology, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- Department of Ophthalmology, Unidade Local de Saúde de São José, Lisbon, Portugal
- Department of Ophthalmology, Hospital Lusíadas de Lisboa, Lisbon, Portugal
| | - Bruno Almeida Carvalho
- Department of Ophthalmology, Hospital CUF Descobertas, Lisbon, Portugal
- Deparment of Ophthalmology, Clínica de São João de Deus, Lisbon, Portugal
| | - Diogo Alpuim Costa
- Department of Haematology and Oncology, CUF Oncologia, Lisbon, Portugal
- Department of Medical Oncology, Hospital de Cascais, Cascais, Portugal
- NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade NOVA de Lisboa (UNL), Lisbon, Portugal
- Department of Medical Oncology, AIM Cancer Center, Lisbon, Portugal
| |
Collapse
|
14
|
Nicolini A, Ferrari P. Targeted Therapies and Drug Resistance in Advanced Breast Cancer, Alternative Strategies and the Way beyond. Cancers (Basel) 2024; 16:466. [PMID: 38275906 PMCID: PMC10814066 DOI: 10.3390/cancers16020466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
"Targeted therapy" or "precision medicine" is a therapeutic strategy launched over two decades ago. It relies on drugs that inhibit key molecular mechanisms/pathways or genetic/epigenetic alterations that promote different cancer hallmarks. Many clinical trials, sponsored by multinational drug companies, have been carried out. During this time, research has increasingly uncovered the complexity of advanced breast cancer disease. Despite high expectations, patients have seen limited benefits from these clinical trials. Commonly, only a minority of trials are successful, and the few approved drugs are costly. The spread of this expensive therapeutic strategy has constrained the resources available for alternative research. Meanwhile, due to the high cost/benefit ratio, other therapeutic strategies have been proposed by researchers over time, though they are often not pursued due to a focus on precision medicine. Notable among these are drug repurposing and counteracting micrometastatic disease. The former provides an obvious answer to expensive targeted therapies, while the latter represents a new field to which efforts have recently been devoted, offering a "way beyond" the current research.
Collapse
Affiliation(s)
- Andrea Nicolini
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy
| | - Paola Ferrari
- Unit of Oncology, Department of Medical and Oncological Area, Azienda Ospedaliera—Universitaria Pisana, 56125 Pisa, Italy;
| |
Collapse
|
15
|
Moukas SI, Kasimir-Bauer S, Tewes M, Kolberg HC, Hoffmann O, Kimmig R, Keup C. Ratios of monocytes and neutrophils to lymphocytes in the blood predict benefit of CDK4/6 inhibitor treatment in metastatic breast cancer. Sci Rep 2023; 13:21262. [PMID: 38040730 PMCID: PMC10692150 DOI: 10.1038/s41598-023-47874-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/19/2023] [Indexed: 12/03/2023] Open
Abstract
Biomarkers to identify metastatic breast cancer (mBC) patients resistant to CDK4/6 inhibition (CDK4/6i) are currently missing. We evaluated the usefulness of the monocyte-to-lymphocyte ratio (MLR), the neutrophil-to-lymphocyte ratio (NLR) and the platelet-to-lymphocyte ratio (PLR) as predictive markers for de novo resistance to CDK4/6i. Various blood cell counts and MLR, NLR, PLR were recorded before treatment initiation (baseline) and four weeks later from 97 mBC patients receiving endocrine therapy (ET) alone or in combination with CDK4/6i. Binary blood cell count/ratios (mean = cut-off) were related to outcome using Cox regression. High MLR (p = 0.001) and high NLR (p = 0.01) at baseline significantly correlated with a shorter progression-free survival (PFS) in the CDK4/6i cohort, independent of any other clinical parameter as determined by multivariate Cox regression. Both, high MLR (p = 0.008) and high NLR (p = 0.043) as well as a decrease in PLR after four weeks of CDK4/6i first line treatment (p = 0.01) indicated a shorter overall survival. Moreover, decreasing PLR (p = 0.043) and increasing mean corpuscular volume (MCV; p = 0.011) within the first cycle of CDK4/6i correlated with a shorter PFS and decreasing MLR (p = 0.039) within the first cycle of first-line CDK4/6i was also correlated with shorter PFS. In summary, easily assessable blood cell parameter were shown to have predictive, monitoring and prognostic value and thus, could, in future, be used for individualized CDK4/6i therapy management. Most importantly, the imbalance of NLR and MLR at baseline might serve as predictive marker for de novo resistance to CDK4/6i in mBC patients.
Collapse
Affiliation(s)
- Stefanos Ioannis Moukas
- Department of Gynecology and Obstetrics, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Mitra Tewes
- Department of Medical Oncology, University Hospital Essen, 45147, Essen, Germany
- Department of Palliative Medicine, West German Cancer Center, University Hospital Essen, 45147, Essen, Germany
| | - Hans-Christian Kolberg
- Department of Gynecology and Obstetrics, Marienhospital Bottrop, 46236, Bottrop, Germany
| | - Oliver Hoffmann
- Department of Gynecology and Obstetrics, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Corinna Keup
- Department of Gynecology and Obstetrics, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
16
|
Soleimani M, Cheraqpour K, Koganti R, Djalilian AR. Cellular senescence and ophthalmic diseases: narrative review. Graefes Arch Clin Exp Ophthalmol 2023; 261:3067-3082. [PMID: 37079093 DOI: 10.1007/s00417-023-06070-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/21/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023] Open
Abstract
PURPOSE Cellular senescence is a state of permanent growth arrest whereby a cell reaches its replicative limit. However, senescence can also be triggered prematurely in certain stressors including radiation, oxidative stress, and chemotherapy. This stress-induced senescence has been studied in the context of promoting inflammation, tumor development, and several chronic degenerative diseases of aging. Emerging research has elucidated the role of senescence in various ocular diseases. METHODS The literature search was performed using PubMed with using the query (senescence OR aging) AND (eye disease OR ocular disease OR ophthalmic disease OR cornea OR glaucoma OR cataract OR retina) on October 20th, 2022. No time restriction was proposed. Articles were excluded if they were not referenced in English. RESULTS Overall, 51 articles regarding senescence and ocular diseases were found and summarized in this study. Several signaling pathways have been implicated in the development of senescence. Currently, senescence has been linked to various corneal and retinal pathologies, as well as cataract and glaucoma. Given the number of pathologies, senolytics, which are small molecules with the ability to selective targeting of senescent cells, can be used as therapeutic or prophylactic agents. CONCLUSIONS Senescence has been shown to underlie the pathogenesis of numerous ocular diseases. The overall literature on senescence and ocular disease is growing rapidly. There is an ongoing debate whether or not cellular senescence detected in experiments contributes in a significant way to diseases. Research on understanding the mechanism of senescence from ocular cells and tissues is just beginning. Multiple animal models are required to test potential senolytics. Currently, no studies exist to date which have demonstrated the benefits of senolytic therapies in human studies.
Collapse
Affiliation(s)
- Mohammad Soleimani
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Cornea Service, Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Illinois Eye and Ear Infirmary, 1855 W. Taylor Street, M/C 648, Chicago, IL, 60612, USA
| | - Kasra Cheraqpour
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA.
- Cornea Service, Stem Cell Therapy and Corneal Tissue Engineering Laboratory, Illinois Eye and Ear Infirmary, 1855 W. Taylor Street, M/C 648, Chicago, IL, 60612, USA.
| |
Collapse
|
17
|
da Silva FC, Brandão DC, Ferreira EA, Siqueira RP, Ferreira HSV, Da Silva Filho AA, Araújo TG. Tailoring Potential Natural Compounds for the Treatment of Luminal Breast Cancer. Pharmaceuticals (Basel) 2023; 16:1466. [PMID: 37895937 PMCID: PMC10610388 DOI: 10.3390/ph16101466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/24/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Breast cancer (BC) is the most diagnosed cancer worldwide, mainly affecting the epithelial cells from the mammary glands. When it expresses the estrogen receptor (ER), the tumor is called luminal BC, which is eligible for endocrine therapy with hormone signaling blockade. Hormone therapy is essential for the survival of patients, but therapeutic resistance has been shown to be worrying, significantly compromising the prognosis. In this context, the need to explore new compounds emerges, especially compounds of plant origin, since they are biologically active and particularly promising. Natural products are being continuously screened for treating cancer due to their chemical diversity, reduced toxicity, lower side effects, and low price. This review summarizes natural compounds for the treatment of luminal BC, emphasizing the activities of these compounds in ER-positive cells. Moreover, their potential as an alternative to endocrine resistance is explored, opening new opportunities for the design of optimized therapies.
Collapse
Affiliation(s)
- Fernanda Cardoso da Silva
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
| | - Douglas Cardoso Brandão
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
| | - Everton Allan Ferreira
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil; (E.A.F.); (A.A.D.S.F.)
| | - Raoni Pais Siqueira
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
| | - Helen Soares Valença Ferreira
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
| | - Ademar Alves Da Silva Filho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil; (E.A.F.); (A.A.D.S.F.)
| | - Thaise Gonçalves Araújo
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia 38405-302, MG, Brazil
| |
Collapse
|
18
|
Liu Y, Wu J, Ji Z, Chen L, Zou J, Zheng J, Lin W, Cai J, Chen Y, Zheng D, Chen Y, Li Z. Comparative efficacy and safety of different combinations of three CDK4/6 inhibitors with endocrine therapies in HR+/HER-2 - metastatic or advanced breast cancer patients: a network meta-analysis. BMC Cancer 2023; 23:816. [PMID: 37653504 PMCID: PMC10469949 DOI: 10.1186/s12885-023-11322-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 08/20/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND This network meta-analysis aimed to assess the comparative efficacy and safety of combinations involving three cyclin-dependent kinase 4/6 (CDK4/6) inhibitors and endocrine therapies (ETs) in patients with metastatic or advanced breast cancer (BC) who are hormone receptor-positive (HR+) and human epidermal growth factor receptor 2-negative (HER2-). METHODS We initially identified relevant studies from previous meta-analyses and then conducted a comprehensive search of PubMed, Embase, and the Cochrane Central Register of Controlled Trials (CENTRAL) databases to locate additional studies published between February 2020 and September 2021. Essential data were extracted, and a network meta-analysis was performed using R 4.1.1 software with a random-effects model. Furthermore, we assigned rankings to all available treatment combinations by calculating their cumulative probability. RESULTS Data analysis included ten reports from nine studies. Pooled results demonstrated that each treatment combination significantly reduced the hazard risk of progression-free survival (PFS) compared to treatment with an aromatase inhibitor (AI) or fulvestrant alone. However, there were no differences observed in PFS or overall survival (OS) among the different treatment combinations. Additionally, patients receiving palbociclib plus AI and abemaciclib plus AI or fulvestrant experienced more severe adverse events (AEs), with hazard ratios (HRs) of 10.83 (95% confidence interval [CI] = 2.3 to 52.51) and 4.8 (95%CI = 1.41 to 16.21), respectively. The HR for ribociclib plus AI was 9.45 (95%CI = 2.02 to 43.61), and the HR for palbociclib plus fulvestrant was 6.33 (95%CI = 1.03 to 39.86). Based on the ranking probabilities, palbociclib plus fulvestrant had the highest probability of achieving superior PFS (37.65%), followed by abemaciclib plus fulvestrant (28.76%). For OS, ribociclib plus fulvestrant ranked first (34.11%), with abemaciclib plus fulvestrant in second place (25.75%). In terms of safety, palbociclib plus AI (53.98%) or fulvestrant (51.37%) had the highest probabilities of being associated with adverse events. CONCLUSIONS Abemaciclib plus fulvestrant or ribociclib plus AI appear to be effective and relatively safe for the treatment of HR+/HER2- metastatic or advanced BC patients. However, given the reliance on limited evidence, our findings require further validation through additional studies.
Collapse
Affiliation(s)
- Yiyuan Liu
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou, University Medical College, Shantou, Guangdong, China
| | - Jinyao Wu
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou, University Medical College, Shantou, Guangdong, China
| | - Zeqi Ji
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou, University Medical College, Shantou, Guangdong, China
| | - Lingzhi Chen
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou, University Medical College, Shantou, Guangdong, China
| | - Juan Zou
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou, University Medical College, Shantou, Guangdong, China
| | - Jiehua Zheng
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou, University Medical College, Shantou, Guangdong, China
| | - Weixun Lin
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou, University Medical College, Shantou, Guangdong, China
| | - Jiehui Cai
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou, University Medical College, Shantou, Guangdong, China
| | - Yaokun Chen
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou, University Medical College, Shantou, Guangdong, China
| | - Daitian Zheng
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou, University Medical College, Shantou, Guangdong, China
| | - Yexi Chen
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou, University Medical College, Shantou, Guangdong, China.
| | - Zhiyang Li
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou, University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
19
|
Liang XB, Dai ZC, Zou R, Tang JX, Yao CW. The Therapeutic Potential of CDK4/6 Inhibitors, Novel Cancer Drugs, in Kidney Diseases. Int J Mol Sci 2023; 24:13558. [PMID: 37686364 PMCID: PMC10487876 DOI: 10.3390/ijms241713558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Inflammation is a crucial pathological feature in cancers and kidney diseases, playing a significant role in disease progression. Cyclin-dependent kinases CDK4 and CDK6 not only contribute to cell cycle progression but also participate in cell metabolism, immunogenicity and anti-tumor immune responses. Recently, CDK4/6 inhibitors have gained approval for investigational treatment of breast cancer and various other tumors. Kidney diseases and cancers commonly exhibit characteristic pathological features, such as the involvement of inflammatory cells and persistent chronic inflammation. Remarkably, CDK4/6 inhibitors have demonstrated impressive efficacy in treating non-cancerous conditions, including certain kidney diseases. Current studies have identified the renoprotective effect of CDK4/6 inhibitors, presenting a novel idea and potential direction for treating kidney diseases in the future. In this review, we briefly reviewed the cell cycle in mammals and the role of CDK4/6 in regulating it. We then provided an introduction to CDK4/6 inhibitors and their use in cancer treatment. Additionally, we emphasized the importance of these inhibitors in the treatment of kidney diseases. Collectively, growing evidence demonstrates that targeting CDK4 and CDK6 through CDK4/6 inhibitors might have therapeutic benefits in various cancers and kidney diseases and should be further explored in the future.
Collapse
Affiliation(s)
| | | | | | - Ji-Xin Tang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Diseases of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Cui-Wei Yao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Diseases of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| |
Collapse
|
20
|
Ji D, Luo Y, Wang J, Chen S, Lan B, Ma F, Xu B, Fan Y. CDK4/6 inhibitors, PI3K/mTOR inhibitors, and HDAC inhibitors as second-line treatments for hormone receptor-positive, HER2-negative advanced breast cancer: a network meta-analysis. BMC Cancer 2023; 23:805. [PMID: 37644396 PMCID: PMC10463765 DOI: 10.1186/s12885-023-11290-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND This study sought to compare the benefits and safety of agents including Cyclin-dependent kinase 4/6 (CDK4/6) inhibitors, phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) inhibitors, and histone deacetylase (HDAC) inhibitors as second-line treatments for these patients by conducting a comprehensive systematic review and network meta-analysis. METHODS The Medline, Embase and Cochrane Library databases were searched for randomized trials comparing CDK4/6 inhibitors, PI3K/mTOR inhibitors, or HDAC inhibitors vs. placebo with the addition of exemestane or fulvestrant as second-line treatments in patients with HR + advanced breast cancer up to December 16, 2021. Outcomes of interest were progression-free survival (PFS), overall response rate (ORR), overall survival (OS), clinical benefit rate (CBR), and grade 3-4 adverse drug events (ADEs). The present study was conducted according to the Cochrane Collaboration and PRISMA statements. The overall effect was pooled using the random effects model. RESULTS Seventeen studies with a total of 9,100 participants were included in the current study. Compared with placebo plus fulvestrant, PFS was significantly improved by CDK4/6 inhibitor plus fulvestrant, mTOR inhibitor plus fulvestrant, mTOR inhibitor plus exemestane, and PI3K inhibitor plus fulvestrant, but not HDAC inhibitor plus exemestane. While mTOR inhibitor plus exemestane was the best regimen (SUCRA value 89.5%), the mTOR inhibitor plus exemestane regimen induced more severe adverse events (SAEs) than the HDAC inhibitor plus exemestane regimen [OR, 95% CI: 2.40 (1.40-4.10)]. CONCLUSION mTOR inhibitor and CDK4/6 inhibitor-based regimens demonstrated superior clinical efficacy and comparable safety profiles as second-line treatment in patients with HR-positive, HER2-negative advanced breast cancer.
Collapse
Affiliation(s)
- Danyang Ji
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Yang Luo
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Jiayu Wang
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Shanshan Chen
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Bo Lan
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Fei Ma
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Binghe Xu
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China.
| | - Ying Fan
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
21
|
Ayyadurai VAS, Deonikar P. Attenuation of Aging-Related Oxidative Stress Pathways by Phytonutrients: A Computational Systems Biology Analysis. Nutrients 2023; 15:3762. [PMID: 37686794 PMCID: PMC10489992 DOI: 10.3390/nu15173762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Aging results from gradual accumulation of damage to the cellular functions caused by biochemical processes such as oxidative stress, inflammation-driven prolonged cellular senescence state, immune system malfunction, psychological stress, and epigenetic changes due to exposure to environmental toxins. Plant-derived bioactive molecules have been shown to ameliorate the damage from oxidative stress. This research seeks to uncover the mechanisms of action of how phytochemicals from fruit/berry/vegetable (FBV) juice powder mitigate oxidative stress. The study uses a computational systems biology approach to (1) identify biomolecular pathways of oxidative stress; (2) identify phytochemicals from FBV juice powder and their specific action on oxidative stress mechanisms; and (3) quantitatively estimate the effects of FBV juice powder bioactive compounds on oxidative stress. The compounds in FBV affected two oxidative stress molecular pathways: (1) reactive oxygen species (ROS) production and (2) antioxidant enzyme production. Six bioactive compounds including cyanidin, delphinidin, ellagic acid, kaempherol, malvidin, and rutin in FBV significantly lowered production of ROS and increased the production of antioxidant enzymes such as catalase, heme oxygenase-1, superoxide dismutase, and glutathione peroxidase. FBV juice powder provides a combination of bioactive compounds that attenuate aging by affecting multiple pathways of oxidative stress.
Collapse
Affiliation(s)
- V. A. Shiva Ayyadurai
- Systems Biology Group, CytoSolve Research Division, CytoSolve, Cambridge, MA 02138, USA;
| | | |
Collapse
|
22
|
Lee J, Lee YJ, Bae SJ, Baek SH, Kook Y, Cha YJ, Lee JW, Son BH, Ahn SH, Lee HJ, Gong G, Jeong J, Lee SB, Ahn SG. Ki-67, 21-Gene Recurrence Score, Endocrine Resistance, and Survival in Patients With Breast Cancer. JAMA Netw Open 2023; 6:e2330961. [PMID: 37647069 PMCID: PMC10469325 DOI: 10.1001/jamanetworkopen.2023.30961] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 07/20/2023] [Indexed: 09/01/2023] Open
Abstract
Importance Both high 21-gene recurrence score (RS) and high Ki-67 level are poor prognostic factors in patients with estrogen receptor (ER)-positive ERBB2-negative (ER+/ERBB-) breast cancer; however, a discrepancy between the 2 has been noted. Survival differences according to these 2 biomarkers are not well known. Objective To assess the associations between RS and Ki-67 expression and between Ki-67 expression and recurrence-free survival in patients with ER+/ERBB- breast cancer with low RS. Design, Setting, and Participants This cohort study included women treated for ER+/ERBB2- breast cancer who underwent the 21-gene RS test from March 2010 to December 2020 in 2 hospitals in Korea. Exposures Recurrence score and Ki-67 level. Main Outcomes and Measures A Cox proportional hazards regression model was used to examine the association of Ki-67 with recurrence-free survival (RFS), while a binary logistic regression model was used to examine the association between Ki-67 and secondary endocrine resistance. High Ki-67 expression was defined as 20% or greater, and low genomic risk as an RS of 25 or less. Secondary endocrine resistance was defined as breast cancer recurrence that occurred after at least 2 years of endocrine therapy and during or within the first year after completing 5 years of adjuvant endocrine therapy. Results A total of 2295 female patients were included (mean [SD] age, 49.8 [9.3] years), of whom 1948 (84.9%) were in the low genomic risk group and 1425 (62.1%) had low Ki-67 level. The median follow-up period was 40 months (range, 0-140 months). The RS and Ki-67 level had a moderate correlation (R = 0.455; P < .001). Of the patients with low Ki-67 level, 1341 (94.1%) had low RS, whereas 607 of 870 patients with high Ki-67 level (69.8%) had low RS. In patients with low RS, the RFS differed significantly according to Ki-67 level (low Ki-67, 98.5% vs high Ki-67, 96.5%; P = .002). Among the 1807 patients with low genomic risk who did not receive chemotherapy, high Ki-67 level was independently associated with recurrence (hazard ratio, 2.51; 95% CI, 1.27-4.96; P = .008). Recurrence after 3 years differed significantly according to Ki-67 level (low Ki-67, 98.7% vs high Ki-67, 95.7%; P = .003), whereas recurrence within 3 years did not differ (low Ki-67, 99.3% vs high Ki-67, 99.3%; P = .90). In addition, Ki-67 was associated with secondary endocrine resistance in patients with low RS who did not receive chemotherapy (odds ratio, 2.49; 95% CI, 1.13-5.50; P = .02). Conclusions and Relevance In this cohort study of patients with ER+/ERBB2- breast cancer, a moderate correlation was observed between Ki-67 and RS, and high Ki-67 level in patients with low genomic risk was associated with increased risk of secondary endocrine resistance.
Collapse
Affiliation(s)
- Janghee Lee
- Department of Surgery, Dongtan Sacred Heart Hospital, Hallym University, Dongtan, Republic of Korea
- Department of Medicine, Yonsei University Graduate School, Seoul, Republic of Korea
| | - Young-jin Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Soong June Bae
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Ho Baek
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoowon Kook
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoon Jin Cha
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Won Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Byung Ho Son
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sei Hyun Ahn
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hee Jin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joon Jeong
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sae Byul Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung Gwe Ahn
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
23
|
Elgohary S, El Tayebi HM. Inflammasomes in breast cancer: the ignition spark of progression and resistance? Expert Rev Mol Med 2023; 25:e22. [PMID: 37337426 DOI: 10.1017/erm.2023.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Inflammation and immune evasion are major key players in breast cancer (BC) progression. Recently, the FDA approved the use of anti-programmed death-ligand 1 antibody (anti-PD-L1) and phosphoinositide 3-kinase (PI3K) inhibitors against aggressive BC. Despite the paradigm shift in BC treatments, patients still suffer from resistance, recurrence and serious immune-related adverse events. These obstacles require unravelling of the hidden molecular contributors for such therapy failure hence yielding therapeutics that are at least as efficient yet safer. Inflammasome pathway is activated when the pattern recognition receptor senses danger signals (danger-associated molecular patterns) from damagedRdying cells or pathogen-associated molecular patterns found in microbes, leading to secretion of the active pro-inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18). It has been shown throughout numerous studies that inflammasome pathway enhanced invasion, metastasis, provoked BC progression and therapy resistance. Additionally, inflammasomes upregulated the proliferative index ki67 and enhanced PD-L1 expression leading to immunotherapy resistance. IL-1β contributed to significant decrease in oestrogen receptor levels and promoted BC chemo-resistance. High levels of IL-18 in sera of BC patients were associated with worst prognosis. Stimulation of purinergic receptors and modulation of adipokines in obese subjects activated inflammasomes that evoked radiotherapy resistance and BC progression. The micro RNA miR-223-3p attenuated the inflammasome over-expression leading to lowered tumour volume and lessened angiogenesis in BC. This review sheds the light on the molecular pathways of inflammasomes and their impacts in distinct BC subtypes. In addition, it highlights novel strategies in treatment and prevention of BC.
Collapse
Affiliation(s)
- Sawsan Elgohary
- Clinical Pharmacology and Pharmacogenomics Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Hend M El Tayebi
- Clinical Pharmacology and Pharmacogenomics Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| |
Collapse
|
24
|
Chen Q, Young L, Barsotti R. Mitochondria in cell senescence: A Friend or Foe? ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 136:35-91. [PMID: 37437984 DOI: 10.1016/bs.apcsb.2023.02.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Cell senescence denotes cell growth arrest in response to continuous replication or stresses damaging DNA or mitochondria. Mounting research suggests that cell senescence attributes to aging-associated failing organ function and diseases. Conversely, it participates in embryonic tissue maturation, wound healing, tissue regeneration, and tumor suppression. The acute or chronic properties and microenvironment may explain the double faces of senescence. Senescent cells display unique characteristics. In particular, its mitochondria become elongated with altered metabolomes and dynamics. Accordingly, mitochondria reform their function to produce more reactive oxygen species at the cost of low ATP production. Meanwhile, destructed mitochondrial unfolded protein responses further break the delicate proteostasis fostering mitochondrial dysfunction. Additionally, the release of mitochondrial damage-associated molecular patterns, mitochondrial Ca2+ overload, and altered NAD+ level intertwine other cellular organelle strengthening senescence. These findings further intrigue researchers to develop anti-senescence interventions. Applying mitochondrial-targeted antioxidants reduces cell senescence and mitigates aging by restoring mitochondrial function and attenuating oxidative stress. Metformin and caloric restriction also manifest senescent rescuing effects by increasing mitochondria efficiency and alleviating oxidative damage. On the other hand, Bcl2 family protein inhibitors eradicate senescent cells by inducing apoptosis to facilitate cancer chemotherapy. This review describes the different aspects of mitochondrial changes in senescence and highlights the recent progress of some anti-senescence strategies.
Collapse
Affiliation(s)
- Qian Chen
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States.
| | - Lindon Young
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| | - Robert Barsotti
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| |
Collapse
|
25
|
Zhou FH, Downton T, Freelander A, Hurwitz J, Caldon CE, Lim E. CDK4/6 inhibitor resistance in estrogen receptor positive breast cancer, a 2023 perspective. Front Cell Dev Biol 2023; 11:1148792. [PMID: 37035239 PMCID: PMC10073728 DOI: 10.3389/fcell.2023.1148792] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
CDK4/6 inhibitors have become game-changers in the treatment of estrogen receptor-positive (ER+) breast cancer, and in combination with endocrine therapy are the standard of care first-line treatment for ER+/HER2-negative advanced breast cancer. Although CDK4/6 inhibitors prolong survival for these patients, resistance is inevitable and there is currently no clear standard next-line treatment. There is an urgent unmet need to dissect the mechanisms which drive intrinsic and acquired resistance to CDK4/6 inhibitors and endocrine therapy to guide the subsequent therapeutic decisions. We will review the insights gained from preclinical studies and clinical cohorts into the diverse mechanisms of CDK4/6 inhibitor action and resistance, and highlight potential therapeutic strategies in the context of CDK4/6 inhibitor resistance.
Collapse
Affiliation(s)
- Fiona H. Zhou
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of NSW, Sydney, NSW, Australia
| | - Teesha Downton
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of NSW, Sydney, NSW, Australia
| | - Allegra Freelander
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of NSW, Sydney, NSW, Australia
| | - Joshua Hurwitz
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of NSW, Sydney, NSW, Australia
| | - C. Elizabeth Caldon
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of NSW, Sydney, NSW, Australia
| | - Elgene Lim
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of NSW, Sydney, NSW, Australia
| |
Collapse
|
26
|
Devlin MJ, Miller RE. Disparity in the era of personalized medicine for epithelial ovarian cancer. Ther Adv Med Oncol 2023; 15:17588359221148024. [PMID: 36643655 PMCID: PMC9837277 DOI: 10.1177/17588359221148024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023] Open
Abstract
The treatment of high-grade serous ovarian cancer and high-grade endometrioid ovarian cancer has seen significant improvements in recent years, with BRCA1/2 and homologous recombination status guiding a personalized approach which has resulted in improved patient outcomes. However, for other epithelial ovarian cancer subtypes, first-line treatment remains unchanged from the platinum-paclitaxel trials of the early 2000s. In this review, we explore novel therapeutic approaches being adopted in the treatment of clear cell, mucinous, carcinosarcoma and low-grade serous ovarian cancer and the biological rational behind them. We discuss why such disparities exist, the challenges faced in conducting dedicated trials in these rarer histologies and look towards new approaches being adopted to overcome them.
Collapse
Affiliation(s)
| | - Rowan E. Miller
- Department of Medical Oncology, St Bartholomew’s Hospital, London, UK,Department of Medical Oncology, University College London Hospital, London, UK
| |
Collapse
|
27
|
Abstract
Cellular senescence has become a subject of great interest within the ageing research field over the last 60 years, from the first observation in vitro by Leonard Hayflick and Paul Moorhead in 1961, to novel findings of phenotypic sub-types and senescence-like phenotype in post-mitotic cells. It has essential roles in wound healing, tumour suppression and the very first stages of human development, while causing widespread damage and dysfunction with age leading to a raft of age-related diseases. This chapter discusses these roles and their interlinking pathways, and how the observed accumulation of senescent cells with age has initiated a whole new field of ageing research, covering pathologies in the heart, liver, kidneys, muscles, brain and bone. This chapter will also examine how senescent cell accumulation presents in these different tissues, along with their roles in disease development. Finally, there is much focus on developing treatments for senescent cell accumulation in advanced age as a method of alleviating age-related disease. We will discuss here the various senolytic and senostatic treatment approaches and their successes and limitations, and the innovative new strategies being developed to address the differing effects of cellular senescence in ageing and disease.
Collapse
Affiliation(s)
- Rebecca Reed
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Satomi Miwa
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
28
|
Pan B, Hao Z, Xu Y, Wang Z, Yao R, Wang X, Ren C, Zhou Y, Sun Q, Huo L. Case report: 18F-FES PET/CT predicted treatment responses of second-line and third-line CDK4/6 inhibitors after disease progression on first-line CDK4/6 inhibitor in a HR+/HER2- metastatic breast cancer patient. Front Oncol 2022; 12:1095779. [PMID: 36620595 PMCID: PMC9816999 DOI: 10.3389/fonc.2022.1095779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/05/2022] [Indexed: 12/25/2022] Open
Abstract
Background Cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) has become the commonest first-line treatment of hormonal receptor positive and human epidermal growth factor receptor 2 negative (HR+/HER2-) metastatic breast cancer (MBC). However, therapy is quite individualized after progression of disease (PD) when CDK4/6i fails. Estrogen receptor (ER) status of metastatic lesions of bone, lung or liver might be different from the primary tumor and biopsy of metastatic lesions was invasive and not always available. Prediction of treatment response after PD of CDK4/6i remains unsolved. 18F-fluoroestradiol (FES) PET/CT could non-invasively reveal ER expression both in primary and metastatic breast cancer and recognize heterogeneity of ER status. Case presentation A 70-year-old woman with Parkinson's disease, osteoporosis and cardiovascular co-morbidity was diagnosed with HR+/HER2- breast cancer (pT2N2M0, stage IIIa). Three years later, she developed metastases in right lung and pleura with pleural effusion and received palbociclib + letrozole. After 8 months the disease progressed, and 18F-FES PET/CT revealed multiple ER-positive pleural lesions and ER-negative pulmonary nodules after PD and the progression-free survival (PFS) of first-line CDK4/6i was 8 months. Since most of the metastatic lesions were ER-positive, abemaciclib + fulvestrant were chosen as the second-line CDK4/6i treatment and the PFS was 15 months. Another 18F-FES PET/CT showed a new ER-positive pleural mass with multiple ER-negative pulmonary nodules. Since 18F-FES PET/CT revealed that the dominant lesions were still ER-positive, dalpiciclib + exemestane + fulvestrant were prescribed as the third-line CDK4/6i treatment. Currently the patient's disease had been stable for 2 months. Conclusion This case demonstrated that 18F-FES PET/CT could show ER heterogeneity non-invasively and reveal the treatment responses a predictive imaging tool of serial second- and third-line of CDK4/6i treatments when first-line CDK4/6i failed in HR+/HER2- MBC. So long as the dominant or newly-developed metastatic lesion was ER-positive on 18F-FES PET after first-line CDK4/6i, the patient might show certain therapeutic response towards endocrine-based treatment including second- and third-line of CDK4/6i, and thus increased the time to chemotherapy (TTC).
Collapse
Affiliation(s)
- Bo Pan
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhixin Hao
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China
| | - Ying Xu
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Wang
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ru Yao
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuefei Wang
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Ren
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China
| | - Yidong Zhou
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiang Sun
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,*Correspondence: Qiang Sun, ; Li Huo,
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China,*Correspondence: Qiang Sun, ; Li Huo,
| |
Collapse
|
29
|
Han L, Wang N, Li Y, Jiang S, Gu Y. A rapid reduction in tumor size by cyclin-dependent kinase inhibition in hormone receptor-positive postpartum breast cancer: a case report of two patients and a review of the literature. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1413. [PMID: 36660646 PMCID: PMC9843329 DOI: 10.21037/atm-22-5201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/07/2022] [Indexed: 12/29/2022]
Abstract
Background Postpartum breast cancer (PPBC) as an independent entity different from PABC. PPBC is defined as breast cancer (BC) diagnosed within 5 years after delivery in many relevant literatures and is associated with a poor prognosis and a decrease in overall survival. PPBC patients commonly present with inflammatory breast cancer (IBC) phenotype, multifocal lesions, and lymph node metastasis. Hormone receptor-positive (HR+) PPBC is an under-investigated subtype. In PPBC, the risk of death of HR+ subtype significantly increased two-fold, while that was only modestly increased for triple-negative breast cancer (TNBC) subtype, and was not significant in human epidermal growth factor receptor 2-positive (HER2+) subtype. HR+ PPBC is a subtype associating with enhanced signatures of cell cycle control, T-cell activation and exhaustion, decreased HR signaling, and altered P53 signaling. The recommended treatment for HR+ PPBC patients is still lacking. Cyclin-dependent kinase (CDK) 4/6 inhibitors are used as a novel treatment standard not only in pretreated patients but also in the first-line setting of HR+ metastatic breast cancer (MBC). However, there is no clinical case report on the application and efficacy of CDK4/6 inhibitors in HR+ PPBC patients. Case Description This article describes the clinical cases of two patients with advanced HR+ PPBC who were rapidly relieved after receiving leuprorelin combined with letrozole combined with dalpiciclib. We reviewed the related literature of PPBC, and found that HR+ PPBC has not been clinically classified as a BC subtype, and only some basic studies suggested that HR+ PPBC may be sensitive to CDK4/6 inhibitors. The purpose of this study is to provide the basis for the related research on the therapeutic effect of CDK4/6 inhibitors in HR+ PPBC through the report of clinical cases. Conclusions This article reports for the first time the good therapeutic effects of CDK4/6 inhibitors on HR+ PPBC patients. Based on our findings, we suggest that dalpiciclib combined with endocrine therapy can be considered as the first-line treatment for patients with advanced HR+ PPBC. Our case report provides new clinical evidence for the related research on the role of CDK4/6 inhibitors in HR+ PPBC therapy.
Collapse
Affiliation(s)
- Luhong Han
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Nan Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Li
- Department of Reproductive Medicine, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shan Jiang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanting Gu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
30
|
Grechukhina KS, Vorontsova KA, Filonenko DA, Tyutyunnik PS, Shchadrova VV, Zhukova LG. Antitumor response and quality of life: is there a need to sacrifice? Clinical observation: long-term and safe control of the disease using a combination of ribociclib with letrozole. Case report. JOURNAL OF MODERN ONCOLOGY 2022. [DOI: 10.26442/18151434.2022.3.201895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Metastatic luminal B HER2-negative breast cancer (HR+/HER2- mBC) occupies a leading place in the global structure of morbidity and mortality among women. The current gold standard of first-line treatment is the combination of CDK4/6 inhibitors with aromatase inhibitors, among which ribociclib with letrozole is distinguished. According to the MONALEESA-2 study, the addition of ribociclib to letrozole significantly increased the median overall survival to 63.9 months, reducing the risk of death by 24%. The safety profile of the combination is manageable, and the development of adverse events led to the interruption of therapy only in 7.5% of cases. A study of the actual clinical practice of CompLEEment-1 also confirmed the safety and effectiveness of the combination. Maintaining and improving the quality of life is one of the main tasks in the treatment of patients with HR+/HER2- mBC. According to the MONALEESA-2 study, the addition of ribociclib significantly affects the maintenance of quality of life and leads to a decrease in the intensity of pain syndrome. The published data allowed us to assign a combination of ribociclib and letrozole 4 points on the ESMO-MCBS scale. The safety of long-term use of the combination in the first line of treatment illustrated by clinical observation. The patient's progression-free survival during therapy was 40 months, which significantly exceeds the data of the MONALEESA-2 and CompLEEment-1 studies. The maximum effect (partial response according to RECIST 1.1 -40%) achieved after 24 weeks and persisted for 24 months. Clinically, the patient noted a decrease in the severity of the pain syndrome after 8 weeks of therapy. Against the background of therapy, it was possible to maintain the quality of life without sacrificing antitumor efficacy.
Collapse
|
31
|
Clinical management of metastatic hormone receptor-positive, HER2-negative breast cancer (MBC) after CDK 4/6 inhibitors: a retrospective single-institution study. Breast Cancer Res Treat 2022; 196:229-237. [PMID: 36045271 PMCID: PMC9829187 DOI: 10.1007/s10549-022-06713-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/14/2022] [Indexed: 01/12/2023]
Abstract
PURPOSE Cyclin-dependent kinase 4/6 inhibitors (CDK4/6is), in combination with endocrine therapy (ET), are standard either in the first (1L) or second-line (2L) setting for the treatment of hormone receptor (HR) positive, HER2-negative metastatic breast cancer (MBC). However, the optimal sequencing of treatments after progression on CDK4/6i remains unknown. We performed a single-institution analysis to identify treatments and outcomes after progression on a CDK4/6i. METHODS We identified patients with HR-positive, HER2-negative MBC prescribed a CDK4/6i in the 1L or 2L settings from December 2014 to February 2018 at Mayo Clinic in Rochester, Minnesota. Outcomes were collected through September 30, 2020. RESULTS Palbociclib, in combination with letrozole or fulvestrant, was the most prescribed CDK4/6i. The 1L and 2L CDK4/6i cohorts exhibited comparable overall survival (OS), but progression-free survival (PFS) was longer in the 1L than the 2L cohort [28.2 months (95% CI 19.6-34.9) vs 19.8 months (95% CI 15.7-29.6)]. The most common post-CDK4/6i treatments were PI3K/mTOR inhibitors (PI3K/mTORi), single-agent ET, or chemotherapy. PFS in the 1L CDK4/6i cohort following PI3K/mTORi was 8.5 months (95% CI 5.5 months-NE), single-agent ET was 6.0 months (95% CI 3.3-14.0 months), and chemotherapy PFS was 5.4 months (95% CI 3.3 months-NE). CONCLUSIONS Following progression on a CDK 4/6i, mPFS was short, with similar PFS times comparing chemotherapy and ET, with slightly longer PFS for targeted strategies (PI3K/mTOR). These results highlight a major need to better understand the mechanisms of CDK4/6i resistance and identify new therapeutic strategies for these patients.
Collapse
|
32
|
Ozyurt R, Ozpolat B. Molecular Mechanisms of Anti-Estrogen Therapy Resistance and Novel Targeted Therapies. Cancers (Basel) 2022; 14:5206. [PMID: 36358625 PMCID: PMC9655708 DOI: 10.3390/cancers14215206] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/05/2022] [Accepted: 10/20/2022] [Indexed: 07/29/2023] Open
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer in women, constituting one-third of all cancers in women, and it is the second leading cause of cancer-related deaths in the United States. Anti-estrogen therapies, such as selective estrogen receptor modulators, significantly improve survival in estrogen receptor-positive (ER+) BC patients, which represents about 70% of cases. However, about 60% of patients inevitably experience intrinsic or acquired resistance to anti-estrogen therapies, representing a major clinical problem that leads to relapse, metastasis, and patient deaths. The resistance mechanisms involve mutations of the direct targets of anti-estrogen therapies, compensatory survival pathways, as well as alterations in the expression of non-coding RNAs (e.g., microRNA) that regulate the activity of survival and signaling pathways. Although cyclin-dependent kinase 4/6 and phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) inhibitors have significantly improved survival, the efficacy of these therapies alone and in combination with anti-estrogen therapy for advanced ER+ BC, are not curative in advanced and metastatic disease. Therefore, understanding the molecular mechanisms causing treatment resistance is critical for developing highly effective therapies and improving patient survival. This review focuses on the key mechanisms that contribute to anti-estrogen therapy resistance and potential new treatment strategies alone and in combination with anti-estrogen drugs to improve the survival of BC patients.
Collapse
Affiliation(s)
- Rumeysa Ozyurt
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Houston Methodist Research Institute, Department of Nanomedicine, 6670 Bertner Ave, Houston, TX 77030, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Houston Methodist Research Institute, Department of Nanomedicine, 6670 Bertner Ave, Houston, TX 77030, USA
| |
Collapse
|
33
|
Salvador Bofill J, Moreno Anton F, Rodriguez Sanchez CA, Galve Calvo E, Hernando Melia C, Ciruelos Gil EM, Vidal M, Jiménez-Rodriguez B, De la Cruz Merino L, Martínez Jañez N, Villanueva Vazquez R, de Toro Salas R, Anton Torres A, Alvarez Lopez IM, Gavila Gregori J, Quiroga Garcia V, Vicente Rubio E, De la Haba-Rodriguez J, Gonzalez-Santiago S, Diaz Fernandez N, Barnadas Molins A, Cantos Sanchez de Ibargüen B, Delgado Mingorance JI, Bellet Ezquerra M, de Casa S, Gimeno A, Martin M. Safety and efficacy of ribociclib plus letrozole in patients with HR+, HER2- advanced breast cancer: Results from the Spanish sub-population of the phase 3b CompLEEment-1 trial. Breast 2022; 66:77-84. [PMID: 36206609 PMCID: PMC9535465 DOI: 10.1016/j.breast.2022.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/20/2022] [Accepted: 09/24/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Breast cancer is the most common malignancy and the second leading cause of cancer-related mortality in Spanish women. Ribociclib in combination with endocrine therapy (ET) has shown superiority in prolonging survival in patients with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer (ABC) vs. ET alone. METHODS CompLEEment-1 is a single-arm, open-label phase 3b trial evaluating ribociclib plus letrozole in a broad population of patients with HR+, HER2- ABC. The primary endpoints were safety and tolerability. Here we report data for Spanish patients enrolled in CompLEEment-1. RESULTS A total of 526 patients were evaluated (median follow-up: 26.97 months). Baseline characteristics showed a diverse population with a median age of 54 years. At study entry, 56.5% of patients had visceral metastases and 8.7% had received prior chemotherapy for advanced disease. Rates of all-grade and Grade ≥3 adverse events (AEs) were 99.0% and 76.2%, respectively; 21.3% of patients experienced a serious AE, and 15.8% of AEs led to treatment discontinuation. AEs of special interest of neutropenia, increased alanine aminotransferase, increased aspartate aminotransferase and QTcF prolongation occurred in 77.8%, 14.8%, 11.4% and 4.0% of patients, respectively. Patients aged >70 years experienced increased rates of all-grade and Grade ≥3 neutropenia and anemia. Efficacy results were consistent with the global study. CONCLUSIONS Results from Spanish patients enrolled in CompLEEment-1 are consistent with global data showing efficacy and a manageable safety profile for ribociclib plus letrozole treatment in patients with HR+, HER2- ABC, including populations of interest (NCT02941926). TRIAL REGISTRATION ClinicalTrials.gov NCT02941926.
Collapse
Affiliation(s)
- Javier Salvador Bofill
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), Sevilla, Spain,Corresponding author. Unidad de Oncología, Servicio de Oncología, Hospital Universitario Virgen del Rocío, Av. Manuel Siurot, s/n, 41013 Sevilla, Spain.
| | | | | | | | - Cristina Hernando Melia
- Servicio de Oncología, Hospital Clínico Universitario de Valencia e Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | | | - Maria Vidal
- Department of Medical Oncology, Hospital Clinic, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | - Vanesa Quiroga Garcia
- Departamento de Oncología, Badalona-Applied Research Group in Oncology (B-ARGO Group), Institut Català d’Oncologia, Badalona, Spain
| | | | - Juan De la Haba-Rodriguez
- Instituto Maimonides de Investigacion Biomedica (IMIBIC), Hospital Reina Sofía, Universidad de Córdoba, Spain
| | | | | | - Agusti Barnadas Molins
- Hospital Universitari Santa Creu i Sant Pau and CIBERONC Breast Cancer Programme, Department of Medicine, Universitat Autonoma Barcelona, Barcelona, Spain
| | | | | | - Meritxell Bellet Ezquerra
- Hospital Universitari Vall d’Hebron, Barcelona and Institut Oncològic Vall d’Hebron (VHIO), Barcelona, Spain
| | | | | | - Miguel Martin
- Hospital Universitario Gregorio Marañón, Madrid, Spain
| |
Collapse
|
34
|
Al-Qasem AJ, Alves CL, Ehmsen S, Tuttolomondo M, Terp MG, Johansen LE, Vever H, Hoeg LVA, Elias D, Bak M, Ditzel HJ. Co-targeting CDK2 and CDK4/6 overcomes resistance to aromatase and CDK4/6 inhibitors in ER+ breast cancer. NPJ Precis Oncol 2022; 6:68. [PMID: 36153348 PMCID: PMC9509389 DOI: 10.1038/s41698-022-00311-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 08/30/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractResistance to aromatase inhibitor (AI) treatment and combined CDK4/6 inhibitor (CDK4/6i) and endocrine therapy (ET) are crucial clinical challenges in treating estrogen receptor-positive (ER+) breast cancer. Understanding the resistance mechanisms and identifying reliable predictive biomarkers and novel treatment combinations to overcome resistance are urgently needed. Herein, we show that upregulation of CDK6, p-CDK2, and/or cyclin E1 is associated with adaptation and resistance to AI-monotherapy and combined CDK4/6i and ET in ER+ advanced breast cancer. Importantly, co-targeting CDK2 and CDK4/6 with ET synergistically impairs cellular growth, induces cell cycle arrest and apoptosis, and delays progression in AI-resistant and combined CDK4/6i and fulvestrant-resistant cell models and in an AI-resistant autocrine breast tumor in a postmenopausal xenograft model. Analysis of CDK6, p-CDK2, and/or cyclin E1 expression as a combined biomarker in metastatic lesions of ER+ advanced breast cancer patients treated with AI-monotherapy or combined CDK4/6i and ET revealed a correlation between high biomarker expression and shorter progression-free survival (PFS), and the biomarker combination was an independent prognostic factor in both patients cohorts. Our study supports the clinical development of therapeutic strategies co-targeting ER, CDK4/6 and CDK2 following progression on AI-monotherapy or combined CDK4/6i and ET to improve survival of patients exhibiting high tumor levels of CDK6, p-CDK2, and/or cyclin E1.
Collapse
|
35
|
Predictive biomarkers for molecularly targeted therapies and immunotherapies in breast cancer. Arch Pharm Res 2022; 45:597-617. [PMID: 35982262 DOI: 10.1007/s12272-022-01402-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 08/14/2022] [Indexed: 11/02/2022]
Abstract
Globally, breast cancer is the most common malignancy in women. Substantial efforts have been made to develop novel therapies, including targeted therapies and immunotherapies, for patients with breast cancer who do not respond to standard therapies. Consequently, new targeted therapies, such as cyclin-dependent kinase 4 and 6 inhibitors, poly (ADP-ribose) polymerase inhibitors, phosphoinositide 3-kinase inhibitor, and antibody-drug conjugates targeting human epidermal growth factor receptor 2 or trophoblast cell surface antigen-2, and immune checkpoint inhibitor targeting programmed cell death-1, have been developed and are now in clinical use. However, only some patients have benefited from these novel therapies; therefore, the identification and validation of reliable or more accurate biomarkers for predicting responses to these agents remain a major challenge. This review summarizes the currently available predictive biomarkers for breast cancer and describes recent efforts undertaken to identify potential predictive markers for molecularly targeted therapies and immune checkpoint inhibitors.
Collapse
|
36
|
Tatum JL, Kalen JD, Jacobs PM, Riffle LA, James A, Thang L, Sanders C, Hollingshead MG, Basuli F, Shi J, Doroshow JH. 3'-[ 18F]fluoro-3'-deoxythymidine ([ 18F]FLT) Positron Emission Tomography as an In Vivo Biomarker of inhibition of CDK 4/6-Rb pathway by Palbociclib in a patient derived bladder tumor. J Transl Med 2022; 20:375. [PMID: 35982453 PMCID: PMC9389794 DOI: 10.1186/s12967-022-03580-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/07/2022] [Indexed: 11/15/2022] Open
Abstract
Background Several new generation CDK4/6 inhibitors have been developed and approved for breast cancer therapy in combination with endocrine therapeutics. Application of these inhibitors either alone or in combination in other solid tumors has been proposed, but no imaging biomarkers of response have been reported in non-breast cancer animal models. The purpose of this study was to evaluate 3'-[18F]fluoro-3'-deoxythymidine ([18F]FLT) Positron Emission Tomography (PET) as in vivo biomarker of response to palbociclib in a non-breast cancer model. Methods Twenty-four NSG mice bearing patient derived xenografts (PDX) of a well-characterized bladder tumor were randomized into 4 treatment groups: vehicle (n = 6); palbociclib (n = 6); temozolomide (n = 6); and palbociclib plus temozolomide (n = 6) and treated with two cycles of therapy or vehicle. Tumor uptake of [18F]FLT was determined by micro-PET/CT at baseline, 3 days, and 9 days post initiation of therapy. Following the second cycle of therapy, the mice were maintained until their tumors reached a size requiring humane termination. Results [18F]FLT uptake decreased significantly in the palbociclib and combination arms (p = 0.0423 and 0.0106 respectively at day 3 and 0.0012 and 0.0031 at day 9) with stable tumor volume. In the temozolomide arm [18F]FLT uptake increased with day 9 uptake significantly different than baseline (p = 0.0418) and progressive tumor growth was observed during the treatment phase. All groups exhibited progressive disease after day 22, 10 days following cessation of therapy. Conclusion Significant decreases in [18F]FLT uptake as early as three days post initiation of therapy with palbociclib, alone or in combination with temozolomide, in this bladder cancer model correlates with an absence of tumor growth during therapy that persists until day 18 for the palbociclib group and day 22 for the combination group (6 days and 10 days) following cessation of therapy. These results support early modulation of [18F]FLT as an in vivo biomarker predictive of palbociclib therapy response in a non-breast cancer model. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03580-8.
Collapse
Affiliation(s)
- James L Tatum
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Joseph D Kalen
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Paula M Jacobs
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
| | - Lisa A Riffle
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Amy James
- Animal Research Technical Support, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Lai Thang
- Animal Research Technical Support, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Chelsea Sanders
- Animal Research Technical Support, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Melinda G Hollingshead
- Biological Testing Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, Frederick, MD, United States
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jianfeng Shi
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
37
|
Riegel K, Vijayarangakannan P, Kechagioglou P, Bogucka K, Rajalingam K. Recent advances in targeting protein kinases and pseudokinases in cancer biology. Front Cell Dev Biol 2022; 10:942500. [PMID: 35938171 PMCID: PMC9354965 DOI: 10.3389/fcell.2022.942500] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022] Open
Abstract
Kinases still remain the most favorable members of the druggable genome, and there are an increasing number of kinase inhibitors approved by the FDA to treat a variety of cancers. Here, we summarize recent developments in targeting kinases and pseudokinases with some examples. Targeting the cell cycle machinery garnered significant clinical success, however, a large section of the kinome remains understudied. We also review recent developments in the understanding of pseudokinases and discuss approaches on how to effectively target in cancer.
Collapse
Affiliation(s)
- Kristina Riegel
- Cell Biology Unit, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
| | | | - Petros Kechagioglou
- Cell Biology Unit, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
| | - Katarzyna Bogucka
- Cell Biology Unit, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
| | - Krishnaraj Rajalingam
- Cell Biology Unit, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
- *Correspondence: Krishnaraj Rajalingam,
| |
Collapse
|
38
|
Zhou H, Wang L, Liu S, Wang W. The role of phosphoinositide 3-kinases in immune-inflammatory responses: potential therapeutic targets for abdominal aortic aneurysm. Cell Cycle 2022; 21:2339-2364. [PMID: 35792922 DOI: 10.1080/15384101.2022.2094577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The pathogenesis of abdominal aortic aneurysm (AAA) includes inflammatory responses, matrix metalloproteinases (MMPs) degradation, VSMC apoptosis, oxidative stress, and angiogenesis, among which the inflammatory response plays a key role. At present, surgery is the only curing treatment, and no effective drug can delay AAA progression in clinical practice. Therefore, searching for a signaling pathway related to the immune-inflammatory response is an essential direction for developing drugs targeting AAA. Recent studies have confirmed that the PI3K family plays an important role in many inflammatory diseases and is involved in regulating various cellular functions, especially in the immune-inflammatory response. This review focuses on the role of each isoform of PI3K in each stage of AAA immune-inflammatory response, making available explorations for a deeper understanding of the mechanism of inflammation and immune response during the formation and development of AAA.
Collapse
Affiliation(s)
- Haiyang Zhou
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shuai Liu
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
39
|
Abstract
Cyclin-dependent kinase 4 (CDK4) and CDK6 are critical mediators of cellular transition into S phase and are important for the initiation, growth and survival of many cancer types. Pharmacological inhibitors of CDK4/6 have rapidly become a new standard of care for patients with advanced hormone receptor-positive breast cancer. As expected, CDK4/6 inhibitors arrest sensitive tumour cells in the G1 phase of the cell cycle. However, the effects of CDK4/6 inhibition are far more wide-reaching. New insights into their mechanisms of action have triggered identification of new therapeutic opportunities, including the development of novel combination regimens, expanded application to a broader range of cancers and use as supportive care to ameliorate the toxic effects of other therapies. Exploring these new opportunities in the clinic is an urgent priority, which in many cases has not been adequately addressed. Here, we provide a framework for conceptualizing the activity of CDK4/6 inhibitors in cancer and explain how this framework might shape the future clinical development of these agents. We also discuss the biological underpinnings of CDK4/6 inhibitor resistance, an increasingly common challenge in clinical oncology.
Collapse
Affiliation(s)
- Shom Goel
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| | - Johann S Bergholz
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jean J Zhao
- Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
40
|
Kellers F, Fernandez A, Konukiewitz B, Schindeldecker M, Tagscherer KE, Heintz A, Jesinghaus M, Roth W, Foersch S. Senescence-Associated Molecules and Tumor-Immune-Interactions as Prognostic Biomarkers in Colorectal Cancer. Front Med (Lausanne) 2022; 9:865230. [PMID: 35492321 PMCID: PMC9039237 DOI: 10.3389/fmed.2022.865230] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/08/2022] [Indexed: 11/20/2022] Open
Abstract
Background and Aims The initiation of cellular senescence in response to protumorigenic stimuli counteracts malignant progression in (pre)malignant cells. Besides arresting proliferation, cells entering this terminal differentiation state adopt a characteristic senescence-associated secretory phenotype (SASP) which initiates alterations to their microenvironment and effects immunosurveillance of tumorous lesions. However, some effects mediated by senescent cells contribute to disease progression. Currently, the exploration of senescent cells' impact on the tumor microenvironment and the evaluation of senescence as possible target in colorectal cancer (CRC) therapy demand reliable detection of cellular senescence in vivo. Therefore, specific immunohistochemical biomarkers are required. Our aim is to analyze the clinical implications of senescence detection in colorectal carcinoma and to investigate the interactions of senescent tumor cells and their immune microenvironment in vitro and in vivo. Methods Senescence was induced in CRC cell lines by low-dose-etoposide treatment and confirmed by Senescence-associated β-galactosidase (SA-β-GAL) staining and fluorescence activated cell sorting (FACS) analysis. Co-cultures of senescent cells and immune cells were established. Multiple cell viability assays, electron microscopy and live cell imaging were conducted. Immunohistochemical (IHC) markers of senescence and immune cell subtypes were studied in a cohort of CRC patients by analyzing a tissue micro array (TMA) and performing digital image analysis. Results were compared to disease-specific survival (DSS) and progression-free survival (PFS). Results Varying expression of senescence markers in tumor cells was associated with in- or decreased survival of CRC patients. Proximity analysis of p21-positive senescent tumor cells and cytotoxic T cells revealed a significantly better prognosis for patients in which these cell types have the possibility to directly interact. In vitro, NK-92 cells (mimicking natural killer T cells) or TALL-104 cells (mimicking both cytotoxic T cells and natural killer T cells) led to dose-dependent specific cytotoxicity in >75 % of the senescent CRC cells but <20 % of the proliferating control CRC cells. This immune cell-mediated senolysis seems to be facilitated via direct cell-cell contact inducing apoptosis and granule exocytosis. Conclusion Counteracting tumorigenesis, cellular senescence is of significant relevance in CRC. We show the dual role of senescence bearing both beneficial and malignancy-promoting potential in vivo. Absence as well as exceeding expression of senescence markers are associated with bad prognosis in CRC. The antitumorigenic potential of senescence induction is determined by tumor micromilieu and immune cell-mediated elimination of senescent cells.
Collapse
Affiliation(s)
- Franziska Kellers
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Aurélie Fernandez
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Björn Konukiewitz
- Institute of Pathology, Christian-Albrecht University of Kiel, Kiel, Germany
| | | | | | - Achim Heintz
- Department of General, Visceral and Vascular Surgery, Catholic Hospital Mainz, Mainz, Germany
| | - Moritz Jesinghaus
- Department of Pathology, University Hospital Marburg, Marburg, Germany
| | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
41
|
Kisla MM, Ates-Alagoz Z. Benzimidazoles Against Certain Breast Cancer Drug Targets: A Review. Mini Rev Med Chem 2022; 22:2463-2477. [PMID: 35345997 DOI: 10.2174/1389557522666220328161217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/01/2022] [Accepted: 02/09/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Benzimidazoles are widely used scaffolds against various types of cancer including breast cancer. To this end, anticancer agents must be developed using the knowledge of the specific targets of BC. OBJECTIVE In this study, we aim to review the compounds used against some of the biomolecular targets of breast cancer. To this end, we present information about the various targets, with their latest innovative studies. CONCLUSION Benzimidazole ring is an important building block that can target diverse cancer scenarios since it can structurally mimic biomolecules in the human body. Additionally, many studies imply the involvement of this moiety on a plethora of pathways and enzymes related to BC. Herein, our target-based collection of benzimidazole derivatives strongly suggests the utilization of benzimidazole derivatives against BC.
Collapse
Affiliation(s)
- Mehmet Murat Kisla
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Zeynep Ates-Alagoz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
42
|
Hortobagyi GN, Stemmer SM, Burris HA, Yap YS, Sonke GS, Hart L, Campone M, Petrakova K, Winer EP, Janni W, Conte P, Cameron DA, André F, Arteaga CL, Zarate JP, Chakravartty A, Taran T, Le Gac F, Serra P, O'Shaughnessy J. Overall Survival with Ribociclib plus Letrozole in Advanced Breast Cancer. N Engl J Med 2022; 386:942-950. [PMID: 35263519 DOI: 10.1056/nejmoa2114663] [Citation(s) in RCA: 382] [Impact Index Per Article: 127.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND In a previous analysis of this phase 3 trial, first-line ribociclib plus letrozole resulted in significantly longer progression-free survival than letrozole alone among postmenopausal patients with hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative advanced breast cancer. Whether overall survival would also be longer with ribociclib was not known. METHODS Here we report the results of the protocol-specified final analysis of overall survival, a key secondary end point. Patients were randomly assigned in a 1:1 ratio to receive either ribociclib or placebo in combination with letrozole. Overall survival was assessed with the use of a stratified log-rank test and summarized with the use of Kaplan-Meier methods after 400 deaths had occurred. A hierarchical testing strategy was used for the analysis of progression-free survival and overall survival to ensure the validity of the findings. RESULTS After a median follow-up of 6.6 years, 181 deaths had occurred among 334 patients (54.2%) in the ribociclib group and 219 among 334 (65.6%) in the placebo group. Ribociclib plus letrozole showed a significant overall survival benefit as compared with placebo plus letrozole. Median overall survival was 63.9 months (95% confidence interval [CI], 52.4 to 71.0) with ribociclib plus letrozole and 51.4 months (95% CI, 47.2 to 59.7) with placebo plus letrozole (hazard ratio for death, 0.76; 95% CI, 0.63 to 0.93; two-sided P = 0.008). No new safety signals were observed. CONCLUSIONS First-line therapy with ribociclib plus letrozole showed a significant overall survival benefit as compared with placebo plus letrozole in patients with HR-positive, HER2-negative advanced breast cancer. Median overall survival was more than 12 months longer with ribociclib than with placebo. (Funded by Novartis; MONALEESA-2 ClinicalTrials.gov number, NCT01958021.).
Collapse
Affiliation(s)
- Gabriel N Hortobagyi
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Salomon M Stemmer
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Howard A Burris
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Yoon-Sim Yap
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Gabe S Sonke
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Lowell Hart
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Mario Campone
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Katarina Petrakova
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Eric P Winer
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Wolfgang Janni
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Pierfranco Conte
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - David A Cameron
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Fabrice André
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Carlos L Arteaga
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Juan P Zarate
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Arunava Chakravartty
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Tetiana Taran
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Fabienne Le Gac
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Paolo Serra
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| | - Joyce O'Shaughnessy
- From the Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston (G.N.H.), and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center (C.L.A.), and Baylor University Medical Center, Texas Oncology, US Oncology (J.O.), Dallas - all in Texas; the Institute of Oncology, Davidoff Center, Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel (S.M.S.); Sarah Cannon Research Institute, Nashville (H.A.B.); the Department of Medical Oncology, National Cancer Centre Singapore, Singapore (Y.-S.Y.); the Department of Medical Oncology, Netherlands Cancer Institute and Borstkanker Onderzoek Groep Study Center, Amsterdam (G.S.S.); Florida Cancer Specialists, Sarah Cannon Research Institute, Fort Myers (L.H.); the Department of Medical Oncology, Institut de Cancérologie de l'Ouest-René Gauducheau, Saint-Herblain (M.C.), and the Department of Medical Oncology, Institut Gustave Roussy, Medical School, Université Paris-Saclay, Villejuif (F.A.) - both in France; the Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic (K.P.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston (E.P.W.); the Department of Gynecology, University of Ulm, Ulm, Germany (W.J.); the Department of Surgery, Oncology, and Gastroenterology, University of Padua, and the Division of Medical Oncology 2, Istituto Oncologico Veneto, IRCCS, Padua, Italy (P.C.); the Edinburgh Cancer Research Centre, Institute of Genomics and Cancer, University of Edinburgh, Edinburgh (D.A.C.); Novartis Pharmaceuticals, East Hanover, NJ (J.P.Z., A.C.); and Novartis Pharma, Basel, Switzerland (T.T., F.L.G., P.S.)
| |
Collapse
|
43
|
Cetin B, Wabl CA, Gumusay O. CDK4/6 inhibitors: mechanisms of resistance and potential biomarkers of responsiveness in breast cancer. Future Oncol 2022; 18:1143-1157. [PMID: 35137602 DOI: 10.2217/fon-2021-0842] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hormone receptor (HR)-positive, HER2-negative tumors represent the most common form of metastatic breast cancer (MBC), and endocrine therapy has been the mainstay treatment for several decades. Recently, a novel drug class called CDK4/6 inhibitors in combination with endocrine therapy have remarkably improved the outcome of patients with HR-positive, HER2-negative MBC by targeting the cell cycle machinery and overcoming aspects of endocrine resistance. Several potential cell-cycle-specific and nonspecific mechanisms of resistance to CDK4/6 inhibitors have been reported in recent studies. This review discusses potential resistance mechanisms to CDK4/6 inhibitors, the use of biomarkers to guide treatment for HR-positive, HER2-negative MBC and possible approaches to overcome resistance to CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Bulent Cetin
- Department of Internal Medicine, Division of Medical Oncology, Suleyman Demirel University Faculty of Medicine, Isparta, 32260, Turkey
| | - Chiara A Wabl
- University of California, San Francisco School of Medicine, San Francisco, CA 94143, USA
| | - Ozge Gumusay
- University of California Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94143, USA
| |
Collapse
|
44
|
Barone I, Caruso A, Gelsomino L, Giordano C, Bonofiglio D, Catalano S, Andò S. Obesity and endocrine therapy resistance in breast cancer: Mechanistic insights and perspectives. Obes Rev 2022; 23:e13358. [PMID: 34559450 PMCID: PMC9285685 DOI: 10.1111/obr.13358] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/07/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022]
Abstract
The incidence of obesity, a recognized risk factor for various metabolic and chronic diseases, including numerous types of cancers, has risen dramatically over the recent decades worldwide. To date, convincing research in this area has painted a complex picture about the adverse impact of high body adiposity on breast cancer onset and progression. However, an emerging but overlooked issue of clinical significance is the limited efficacy of the conventional endocrine therapies with selective estrogen receptor modulators (SERMs) or degraders (SERDs) and aromatase inhibitors (AIs) in patients affected by breast cancer and obesity. The mechanisms behind the interplay between obesity and endocrine therapy resistance are likely to be multifactorial. Therefore, what have we actually learned during these years and which are the main challenges in the field? In this review, we will critically discuss the epidemiological evidence linking obesity to endocrine therapeutic responses and we will outline the molecular players involved in this harmful connection. Given the escalating global epidemic of obesity, advances in understanding this critical node will offer new precision medicine-based therapeutic interventions and more appropriate dosing schedule for treating patients affected by obesity and with breast tumors resistant to endocrine therapies.
Collapse
Affiliation(s)
- Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Amanda Caruso
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| |
Collapse
|
45
|
Mandigo AC, Shafi AA, McCann JJ, Yuan W, Laufer TS, Bogdan D, Gallagher L, Dylgjeri E, Semenova G, Vasilevskaya IA, Schiewer MJ, McNair CM, de Bono JS, Knudsen KE. Novel Oncogenic Transcription Factor Cooperation in RB-Deficient Cancer. Cancer Res 2022; 82:221-234. [PMID: 34625422 PMCID: PMC9397633 DOI: 10.1158/0008-5472.can-21-1159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/14/2021] [Accepted: 09/09/2021] [Indexed: 01/07/2023]
Abstract
The retinoblastoma tumor suppressor (RB) is a critical regulator of E2F-dependent transcription, controlling a multitude of protumorigenic networks including but not limited to cell-cycle control. Here, genome-wide assessment of E2F1 function after RB loss in isogenic models of prostate cancer revealed unexpected repositioning and cooperation with oncogenic transcription factors, including the major driver of disease progression, the androgen receptor (AR). Further investigation revealed that observed AR/E2F1 cooperation elicited novel transcriptional networks that promote cancer phenotypes, especially as related to evasion of cell death. These observations were reflected in assessment of human disease, indicating the clinical relevance of the AR/E2F1 cooperome in prostate cancer. Together, these studies reveal new mechanisms by which RB loss induces cancer progression and highlight the importance of understanding the targets of E2F1 function. SIGNIFICANCE: This study identifies that RB loss in prostate cancer drives cooperation between AR and E2F1 as coregulators of transcription, which is linked to the progression of advanced disease.
Collapse
Affiliation(s)
- Amy C Mandigo
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ayesha A Shafi
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jennifer J McCann
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Wei Yuan
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | - Talya S Laufer
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Denisa Bogdan
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | - Lewis Gallagher
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | - Emanuela Dylgjeri
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Galina Semenova
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Irina A Vasilevskaya
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Matthew J Schiewer
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Chris M McNair
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Johann S de Bono
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | - Karen E Knudsen
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Department of Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
46
|
Sharma M, Mittapelly N, Banala VT, Urandur S, Gautam S, Marwaha D, Rai N, Singh N, Gupta A, Mitra K, Mishra PR. Amalgamated Microneedle Array Bearing Ribociclib-Loaded Transfersomes Eradicates Breast Cancer via CD44 Targeting. Biomacromolecules 2022; 23:661-675. [PMID: 34978424 DOI: 10.1021/acs.biomac.1c01076] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
HR+/HER2- metastatic breast cancer (MBC) is one of the most common and life-threatening conditions diagnosed in women. The endocrine therapy using an orally active CDK4/6 inhibitor, ribociclib (RB), is the most intriguing approach for treating HR+/HER2- MBC. However, the repeated three to six cycles of multiple dosing and non-targeted distribution of RB led to severe neutropenia; hepatobiliary, gastrointestinal, and renal toxicities, and QT interval prolongation. Here, a novel organic solvent-free HA-PVA-PVP (hyaluronic acid-polyvinyl alcohol-polyvinyl pyrrolidone) composed of a microneedle (MN) array is formulated to deliver RB, integrated with amphiphilic conjugated polymer (HA-GMS)-anchored ultradeformable transfersomes. This unique MN array efficiently crafts microchannels in the skin, allowing HA-RB-Ts to internalize into the tumor cells through lymphatic and systemic absorption and interact with CD44 both spatially and temporally with an amplification of drug release time up to 6-folds. The pharmacokinetic and tissue distribution studies portray drug concentrations within the therapeutic window as long as 48 h, facilitating thrice-a-week frequency with the lower dose, and rule out severe toxicities, with a significant reduction in 8.3-fold RB concentration in vital organs that ultimately enhances the survival rate. Thus, the novel MN system pursues a unique embeddable feature and offers an effective, self-administrable, biodegradable, and chronic treatment option for patients requiring long-term cancer treatments.
Collapse
Affiliation(s)
- Madhu Sharma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Knudsen ES, Schultz E, Hamilton D, Attwood K, Edge S, O’Connor T, Levine E, Witkiewicz AK. OUP accepted manuscript. Oncologist 2022; 27:646-654. [PMID: 35666660 PMCID: PMC9355808 DOI: 10.1093/oncolo/oyac089] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/18/2022] [Indexed: 11/25/2022] Open
Abstract
Background A study was initiated at Roswell Park Comprehensive Cancer Center to capture the real-world experience related to the use of CDK4/6 inhibitors (Ciclibs) for the treatment of metastatic hormone receptor-positive and HER2-negative breast cancer (HR+/HER2-). Patients and Methods A total of 222 patients were evaluated who received CDK4/6 inhibitors in the period from 2015 to 2021. Detailed clinical and demographic information was obtained on each patient and used to define clinical and demographic features associated with progression-free survival on CDK4/6 inhibitor-based therapies. Results In this real-world analysis, the majority of patients received palbociclib as the CDK4/6 inhibitor with letrozole or fulvestrant as the predominant endocrine therapies. The median progression-free survival (PFS) in the letrozole (27.6 months) and fulvestrant (17.2 months) groups were comparable to that observed in clinical trials. As expected, age at start of the treatment and menopausal status influenced endocrine therapy utilization but were not associated with PFS. Patients with recurrent disease had shorter PFS (P = .0024) than those presenting with de novo metastasis. The presence of visceral metastasis trended toward shorter PFS (P = .051). Similarly, prior endocrine therapy (P = .003) or chemotherapy (P = .036) was associated with shorter PFS. Body mass index was not associated with PFS or with dose interruption and/or modification. While the number of minorities in this analysis is limited (n = 26), these patients as a group had statistically shorter PFS on treatment (P = .002). Conclusions The real-world progression-free survival with CDK4/6 inhibitors mimics that observed in the clinical trial. A number of clinical and demographic features were associated with PFS on CDK4/6 inhibitor-based therapy. Further studies are ongoing to validate these findings incorporating additional cancer centers.
Collapse
Affiliation(s)
- Erik S Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Emily Schultz
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Deanna Hamilton
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kris Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Stephen Edge
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Tracey O’Connor
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ellis Levine
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Agnieszka K Witkiewicz
- Corresponding author: Agnieszka K. Witkiewicz, MD, Roswell Park Comprehensive Cancer Center, Elm and Carlton St, Buffalo, NY 14203, USA.
| |
Collapse
|
48
|
Varna D, Geromichalou E, Papachristou E, Papi R, Hatzidimitriou AG, Panteris E, Psomas G, Geromichalos GD, Aslanidis P, Choli-Papadopoulou T, Angaridis PA. Biocompatible silver(I) complexes with heterocyclic thioamide ligands for selective killing of cancer cells and high antimicrobial activity - A combined in vitro and in silico study. J Inorg Biochem 2021; 228:111695. [PMID: 35007963 DOI: 10.1016/j.jinorgbio.2021.111695] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/02/2021] [Accepted: 12/23/2021] [Indexed: 12/11/2022]
Abstract
A series of heteroleptic Ag(I) complexes bearing 4,6-dimethyl-2-pyrimidinethiol (dmp2SH), i.e., [AgCl(dmp2SH)(PPh3)2] (1), [Ag(dmp2SH)(PPh3)2]NO3 (2), [Ag(dmp2SΗ)(xantphos)]NO3 (3), [Ag(μ-dmp2S)(PPh3)]2 (4), [Ag(dmp2S)(xantphos)] (5), [Ag(μ-dmp2S)(DPEphos)]2 (6) (xantphos = 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene and DPEPhos = bis[(2-diphenylphosphino)phenyl]ether) were synthesized. The complexes display systematic variation of particular structural characteristics which were proved to have a significant impact on their in vitro cytotoxicity and antimicrobial properties. A moderate-to-high potential for bacteria growth inhibition was observed for all complexes, with 2, 3 and 5 being particularly effective against Gram-(+) bacteria (IC50 = 1.6-4.5 μM). The three complexes exhibit high in vitro cytotoxicity against HeLa and MCF-7 cancer cells (IC50 = 0.32-3.00 μΜ), suggesting the importance of coordination unsaturation and cationic charge for effective bioactivity. A very low cytotoxicity against HDFa normal cells was observed, revealing a high degree of selectivity (selectivity index ~10) and, hence, biocompatibility. Fluorescence microscopy using 2 showed effective targeting on the membrane of the HeLa cancer cells, subsequently inducing cell death. Binding of the complexes to serum albumin proteins is reasonably strong for potential uptake and subsequent release to target sites. A moderate in vitro antioxidant capacity for free radicals scavenging was observed and a low potential to destroy the double-strand structure of calf-thymus DNA by intercalation, suggesting likely implication of these properties in the bioactivity mechanisms of these complexes. Further insight into possible mechanisms of bioactivity was obtained by molecular modeling calculations, by exploring their ability to act as potential inhibitors of DNA-gyrase, human estrogen receptor alpha, human cyclin-dependent kinase 6, and human papillomavirus E6 oncoprotein.
Collapse
Affiliation(s)
- Despoina Varna
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Elena Geromichalou
- Laboratory of Pharmacology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Athens 11527, Greece
| | - Eleni Papachristou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Rigini Papi
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Antonios G Hatzidimitriou
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Emmanuel Panteris
- Laboratory of Botany, Department of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - George Psomas
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - George D Geromichalos
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Paraskevas Aslanidis
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Theodora Choli-Papadopoulou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece.
| | - Panagiotis A Angaridis
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece.
| |
Collapse
|
49
|
Integrating CDK4/6 inhibitors in the treatment of patients with early breast cancer. Breast 2021; 62 Suppl 1:S70-S79. [PMID: 34930649 PMCID: PMC9097805 DOI: 10.1016/j.breast.2021.12.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/07/2021] [Accepted: 12/12/2021] [Indexed: 12/13/2022] Open
Abstract
CDK4/6 inhibitors have an established role in the treatment of hormone receptor positive HER2-negative advanced breast cancer. All studies conducted in metastatic breast cancer showed a benefit in delaying progression when added to standard endocrine therapy, regardless of therapy line, pretreatment, menopausal status, site of metastasis, CDK4/6 inhibitor used and associated endocrine therapy. A benefit in overall survival has also been demonstrated. In early breast cancer, only the MonarchE study has shown an improved invasive disease-free survival with abemaciclib taken for 2 years, whereas the Penelope-B did not meet the primary endpoint and the PALLAS study was terminated early for futility. Studies conducted in the neoadjuvant setting might help to explain the discordant results. CDK4/6 inhibitors increase PFS in advanced breast cancer in all subgroups. 2-years abemaciclib added to endocrine therapy improves invasive disease-free survival in high-risk breast cancer. Palbociclib did not improve invasive disease-free survival in early breast cancer.
Collapse
|
50
|
Saatci O, Huynh-Dam KT, Sahin O. Endocrine resistance in breast cancer: from molecular mechanisms to therapeutic strategies. J Mol Med (Berl) 2021; 99:1691-1710. [PMID: 34623477 PMCID: PMC8611518 DOI: 10.1007/s00109-021-02136-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/20/2021] [Accepted: 09/06/2021] [Indexed: 12/31/2022]
Abstract
Estrogen receptor-positive (ER +) breast cancer accounts for approximately 75% of all breast cancers. Endocrine therapies, including selective ER modulators (SERMs), aromatase inhibitors (AIs), and selective ER down-regulators (SERDs) provide substantial clinical benefit by reducing the risk of disease recurrence and mortality. However, resistance to endocrine therapies represents a major challenge, limiting the success of ER + breast cancer treatment. Mechanisms of endocrine resistance involve alterations in ER signaling via modulation of ER (e.g., ER downregulation, ESR1 mutations or fusions); alterations in ER coactivators/corepressors, transcription factors (TFs), nuclear receptors and epigenetic modulators; regulation of signaling pathways; modulation of cell cycle regulators; stress signaling; and alterations in tumor microenvironment, nutrient stress, and metabolic regulation. Current therapeutic strategies to improve outcome of endocrine-resistant patients in clinics include inhibitors against mechanistic target of rapamycin (mTOR), cyclin-dependent kinase (CDK) 4/6, and the phosphoinositide 3-kinase (PI3K) subunit, p110α. Preclinical studies reveal novel therapeutic targets, some of which are currently tested in clinical trials as single agents or in combination with endocrine therapies, such as ER partial agonists, ER proteolysis targeting chimeras (PROTACs), next-generation SERDs, AKT inhibitors, epidermal growth factor receptor 1 and 2 (EGFR/HER2) dual inhibitors, HER2 targeting antibody-drug conjugates (ADCs) and histone deacetylase (HDAC) inhibitors. In this review, we summarize the established and emerging mechanisms of endocrine resistance, alterations during metastatic recurrence, and discuss the approved therapies and ongoing clinical trials testing the combination of novel targeted therapies with endocrine therapy in endocrine-resistant ER + breast cancer patients.
Collapse
Affiliation(s)
- Ozge Saatci
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, 715, Sumter Street, CLS609D, Columbia, SC, 29208, USA
| | - Kim-Tuyen Huynh-Dam
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, 715, Sumter Street, CLS609D, Columbia, SC, 29208, USA
| | - Ozgur Sahin
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, 715, Sumter Street, CLS609D, Columbia, SC, 29208, USA.
| |
Collapse
|