1
|
Choi YJ, Yang MK, Kim N, Khwarg SI, Choung H, Kim JE. Expression of nuclear receptors and glucose metabolic pathway proteins in sebaceous carcinoma: Androgen receptor and monocarboxylate transporter 1 have a key role in disease progression. Oncol Lett 2024; 28:593. [PMID: 39421321 PMCID: PMC11484244 DOI: 10.3892/ol.2024.14726] [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: 05/15/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Standard systemic treatments are not consistently effective for treating unresectable or advanced sebaceous carcinoma (SC). The present study investigated the pathogenic roles of nuclear receptors (NRs), glucose metabolic dysregulation and immune checkpoint proteins in SC as prognostic markers or therapeutic targets. Patients with pathologically confirmed SC between January 2002 and December 2019 at three university hospitals in South Korea were included in the present study. Immunohistochemistry was performed on paraffin-embedded tumor tissues for glucocorticoid receptors (GR), androgen receptors (AR), estrogen receptors (ER), progesterone receptors (PR), glucose transporter 1 (GLUT1), monocarboxylate transporters (MCT1 and MCT4), CD147, phosphorylated adenosine monophosphate-activated protein kinase (pAMPK) and the immune checkpoint protein, programmed cell death-ligand 1 (PD-L1). The results were semi-quantitatively assessed and the associations of these proteins with various clinicopathological parameters were determined. A total of 39 cases of SC comprising 19 periocular and 20 extraocular tumors were enrolled. NRs were frequently detected in the tumor nuclei, with GR having the highest frequency (89.7%), followed by AR, ER (both 51.3%) and PR (41.0%). Regarding glucose metabolism, CD147, GLUT1 and MCT1 were highly expressed at 100, 89.7 and 87.2%, respectively, whereas MCT4 and pAMPK expression levels were relatively low at 38.5 and 35.9%, respectively. Membranous expression of PD-L1 was detected in five cases (12.8%), four of which were extraocular. In the multivariate analysis, advanced stage, low AR positivity and high MCT1 expression were independent poor prognostic factors for metastasis-free survival (all P<0.05). The present results suggested that hormonal and metabolic dysregulation may be associated with the pathogenesis of SC, and that AR and MCT1 in particular may serve as prognostic indicators and potential therapeutic targets. Additionally, ~10% of SC cases exhibited PD-L1 expression within the druggable range, and these patients are expected to benefit from treatment with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Youn Joo Choi
- Department of Ophthalmology, Kangdong Sacred Heart Hospital, Hallym University Medical Center, Seoul 05355, Republic of Korea
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Min Kyu Yang
- Department of Ophthalmology, Asan Medical Center, Ulsan University College of Medicine, Seoul 05505, Republic of Korea
| | - Namju Kim
- Department of Ophthalmology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Gyeonggi 13620, Republic of Korea
| | - Sang In Khwarg
- Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hokyung Choung
- Department of Ophthalmology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul 07061, Republic of Korea
| | - Ji Eun Kim
- Department of Pathology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul 07061, Republic of Korea
| |
Collapse
|
2
|
Liu R, Zou Z, Zhang Z, He H, Xi M, Liang Y, Ye J, Dai Q, Wu Y, Tan H, Zhong W, Wang Z, Liang Y. Evaluation of glucocorticoid-related genes reveals GPD1 as a therapeutic target and regulator of sphingosine 1-phosphate metabolism in CRPC. Cancer Lett 2024; 605:217286. [PMID: 39413958 DOI: 10.1016/j.canlet.2024.217286] [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: 09/21/2023] [Revised: 09/08/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024]
Abstract
Prostate cancer (PCa) is an androgen-dependent disease, with castration-resistant prostate cancer (CRPC) being an advanced stage that no longer responds to androgen deprivation therapy (ADT). Mounting evidence suggests that glucocorticoid receptors (GR) confer resistance to ADT in CRPC patients by bypassing androgen receptor (AR) blockade. GR, as a novel therapeutic target in CRPC, has attracted substantial attention worldwide. This study utilized bioinformatic analysis of publicly available CRPC single-cell data to develop a consensus glucocorticoid-related signature (Glu-sig) that can serve as an independent predictor for relapse-free survival. Our results revealed that the signature demonstrated consistent and robust performance across seven publicly accessible datasets and an internal cohort. Furthermore, our findings demonstrated that glycerol-3-phosphate dehydrogenase 1 (GPD1) in Glu-sig can significantly promote CRPC progression by mediating the cell cycle pathway. Additionally, GPD1 was shown to be regulated by GR, with the GR antagonist mifepristone enhancing the anti-tumorigenic effects of GPD1 in CRPC cells. Mechanistically, targeting GPD1 induced the production of sphingosine 1-phosphate (S1P) and enhanced histone acetylation, thereby inducing the transcription of p21 that involved in cell cycle regulation. In conclusion, Glu-sig could serve as a robust and promising tool to improve the clinical outcomes of PCa patients, and modulating the GR/GPD1 axis that promotes tumor growth may be a promising approach for delaying CRPC progression.
Collapse
Affiliation(s)
- Ren Liu
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihao Zou
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China; Guangzhou Laboratory, Guangzhou, China
| | - Zhengrong Zhang
- Department of Urology, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Huichan He
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Ming Xi
- Department of Urology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Yingke Liang
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jianheng Ye
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qishan Dai
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongding Wu
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huijing Tan
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Weide Zhong
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China; Guangzhou Laboratory, Guangzhou, China; Macau Institute of Systems Engineering, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China.
| | - Zongren Wang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Yuxiang Liang
- Department of Urology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Borazanci EH, Bahary N, Chung V, Huyck TK, Kio EA, Chiorean EG, Skeel RT, Alese OB, Cardin DB, Fountzilas C, Hanna WT, Leal AD, Lee V, Noonan AM, Philip PA, Wainberg ZA, Pashova H, Mann G, Oberstein PE. Relacorilant plus nab-paclitaxel for the treatment of metastatic pancreatic ductal adenocarcinoma: results from the open-label RELIANT study. Oncologist 2024; 29:957-965. [PMID: 39191530 PMCID: PMC11546724 DOI: 10.1093/oncolo/oyae210] [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: 03/29/2024] [Accepted: 06/15/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Modulation of glucocorticoid receptor (GR) activity in tumor cells enhances chemotherapy efficacy. We evaluated the selective GR modulator relacorilant plus nab-paclitaxel in patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) who had received at least 2 prior therapy lines. PATIENTS AND METHODS In this open-label, single-arm, phase III study, patients received once-daily oral relacorilant (100 mg, titrated to 150 mg in 25 mg increments/cycle) and nab-paclitaxel (80 mg/m2) on days 1, 8, and 15 of 28-day cycles. The primary efficacy endpoint was objective response rate (ORR) by blinded independent central review. Progression-free survival (PFS), overall survival (OS), target gene modulation, and safety were also assessed. RESULTS Of 43 patients enrolled, 31 were evaluable for ORR (12 did not reach first postbaseline radiographic assessment). An interim analysis to assess whether ORR was ≥10% showed no confirmed responses and the study was discontinued. Two (6.5%) patients attained unconfirmed partial responses and 15 (48.4%) had stable disease. Fourteen of 31 (45.2%) patients had reductions in target lesion size, despite prior nab-paclitaxel exposure in 12 of the 14. Median PFS and OS were 2.4 months (95% CI, 1.4-4.2) and 3.9 months (95% CI, 2.8-4.9), respectively. The most common adverse events were fatigue and nausea. RNA analysis confirmed that relacorilant plus nab-paclitaxel suppressed 8 cortisol target genes of interest. CONCLUSION Relacorilant plus nab-paclitaxel showed modest antitumor activity in heavily pretreated patients with mPDAC, with no new safety signals. Studies of this combination in other indications with a high unmet medical need are ongoing.
Collapse
Affiliation(s)
| | - Nathan Bahary
- Allegheny Health Network Cancer Institute, Pittsburgh, PA, United States
| | - Vincent Chung
- City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | | | - Ebenezer A Kio
- Goshen Center for Cancer Care, Goshen, IN, United States
| | | | - Roland T Skeel
- University of Toledo Medical Center, Toledo, OH, United States
| | - Olatunji B Alese
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Dana B Cardin
- Vanderbilt-Ingram Cancer Center, Nashville, TN, United States
| | | | - Wahid T Hanna
- University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Alexis D Leal
- University of Colorado School of Medicine, Aurora, CO, United States
| | - Valerie Lee
- Johns Hopkins University School of Medicine, Baltimore, MA, United States
| | - Anne M Noonan
- The Ohio State University, Columbus, OH, United States
| | - Philip A Philip
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Zev A Wainberg
- University of California School of Medicine, Los Angeles, CA, United States
| | - Hristina Pashova
- Corcept Therapeutics Incorporated, Menlo Park, CA, United States
| | - Grace Mann
- Corcept Therapeutics Incorporated, Menlo Park, CA, United States
| | | |
Collapse
|
4
|
Reznik E, Torjani A. Mechanisms of stress-attributed breast cancer incidence and progression. Cancer Causes Control 2024; 35:1413-1432. [PMID: 39012513 DOI: 10.1007/s10552-024-01884-2] [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: 03/01/2024] [Accepted: 04/19/2024] [Indexed: 07/17/2024]
Abstract
Breast cancer is the most commonly diagnosed cancer and the second leading cause of cancer deaths in women, with psychosocial stress commonly cited by patients as one of its causes. While there is conflicting epidemiological evidence investigating the association between psychosocial stress and breast cancer incidence and progression, there is reason to believe that interventions aimed at reducing stress pharmacologically or psychologically may improve breast cancer outcomes. The aim of this review is to discuss the molecular and biological mechanisms of stress-attributed breast cancer incidence and progression, including the induction of the hypothalamic-pituitary-adrenal (HPA) axis and the sympathetic nervous system (SNS), as well as decreased immune function and stress hormone-induced resistance to chemotherapy. Moreover, these mechanisms have been cited as potential therapeutic targets of pharmacologic and psychological interventions that may improve the care, well-being and survival of breast cancer patients. Further research is recommended to investigate whether interventions in the primary care setting for women with risk factors for breast cancer development may lead to a decreased incidence of invasive breast tumors.
Collapse
Affiliation(s)
- Elizabeth Reznik
- Department of Internal Medicine, NewYork-Presbyterian Weill Cornell, New York, NY, USA.
| | - Ava Torjani
- Department of Ophthalmology, Keck School of Medicine of USC, Los Angeles, CA, USA
| |
Collapse
|
5
|
Chen CT, Khanna V, Kummar S, Abdul-Karim RM, Sommerhalder D, Tolcher AW, Ueno NT, Davis SL, Orr DW, Hamilton E, Patel MR, Spira AI, Jauhari S, Florou V, Duff M, Xu R, Wang J, Barkund SR, Zhou H, Pankov A, Kong W, Jahchan NS, Jackson EL, Sun JD, Junttila MR, Multani PS, Daemen A, Chow Maneval E, Munster PN. ORIC-101, a Glucocorticoid Receptor Antagonist, in Combination with Nab-Paclitaxel in Patients with Advanced Solid Tumors. CANCER RESEARCH COMMUNICATIONS 2024; 4:2415-2426. [PMID: 39177285 PMCID: PMC11396014 DOI: 10.1158/2767-9764.crc-24-0115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/22/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
PURPOSE In preclinical models, glucocorticoid receptor (GR) signaling drives resistance to taxane chemotherapy in multiple solid tumors via upregulation of antiapoptotic pathways. ORIC-101 is a potent and selective GR antagonist that was investigated in combination with taxane chemotherapy as an anticancer regimen preclinically and in a phase 1 clinical trial. PATIENTS AND METHODS The ability of ORIC-101 to reverse taxane resistance was assessed in cell lines and xenograft models, and a phase 1 study (NCT03928314) was conducted in patients with advanced solid tumors to determine the dose, safety, and antitumor activity of ORIC-101 with nab-paclitaxel. RESULTS ORIC-101 reversed chemoprotection induced by glucocorticoids in vitro and achieved tumor regressions when combined with paclitaxel in both taxane-naïve and -resistant xenograft models. In the phase 1 study, 21 patients were treated in dose escalation and 62 patients were treated in dose expansion. All patients in dose expansion had previously progressed on a taxane-based regimen. In dose escalation, five objective responses were observed. A preplanned futility analysis in dose expansion showed a 3.2% (95% confidence interval, 0.4-11.2) objective response rate with a median progression-free survival of 2 months (95% confidence interval, 1.8-2.8) across all four cohorts, leading to study termination. Pharmacodynamic analysis of tissue and plasma showed GR pathway downregulation in most patients in cycle 1. CONCLUSIONS ORIC-101 with nab-paclitaxel showed limited clinical activity in taxane-resistant solid tumors. Despite clear inhibition of GR pathway signaling, the insufficient clinical signal underscores the challenges of targeting a single resistance pathway when multiple mechanisms of resistance may be in play. SIGNIFICANCE Glucocorticoid receptor (GR) upregulation is a mechanism of resistance to taxane chemotherapy in preclinical cancer models. ORIC-101 is a small molecule GR inhibitor. In this phase 1 study, ORIC-101 plus nab-paclitaxel did not show meaningful clinical benefit in patients who previously progressed on taxanes despite successful GR pathway downregulation.
Collapse
Affiliation(s)
- Christopher T. Chen
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Palo Alto, California.
| | - Vishesh Khanna
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Palo Alto, California.
| | - Shivaani Kummar
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Palo Alto, California.
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon.
| | | | | | | | - Naoto T. Ueno
- University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | | | | | | - Manish R. Patel
- Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, Florida.
| | | | - Shekeab Jauhari
- Florida Cancer Specialists/Sarah Cannon Research Institute, Lake Mary, Florida.
| | - Vaia Florou
- 1Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | - Maureen Duff
- ORIC Pharmaceuticals, South San Francisco, California.
| | - Rongda Xu
- ORIC Pharmaceuticals, South San Francisco, California.
| | - Jian Wang
- ORIC Pharmaceuticals, South San Francisco, California.
| | | | - Haiying Zhou
- ORIC Pharmaceuticals, South San Francisco, California.
| | | | - Wayne Kong
- ORIC Pharmaceuticals, South San Francisco, California.
| | | | | | | | | | | | | | | | - Pamela N. Munster
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California.
| |
Collapse
|
6
|
Giudice E, Salutari V, Sassu CM, Ghizzoni V, Carbone MV, Vertechy L, Fagotti A, Scambia G, Marchetti C. Relacorilant in recurrent ovarian cancer: clinical evidence and future perspectives. Expert Rev Anticancer Ther 2024; 24:649-655. [PMID: 38861580 DOI: 10.1080/14737140.2024.2362178] [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: 02/27/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024]
Abstract
INTRODUCTION Relacorilant (CORT125134, Corcept Therapeutics) is a selective glucocorticoid receptor modulator, which reverses the glucocorticoid-mediated anti-apoptotic effects and restores the taxane chemosensitivity in epithelial ovarian cancer cells. Given those preclinical findings, relacorilant is currently under investigation in clinical trials in combination with nab-paclitaxel for the platinum-resistant ovarian cancer setting. AREAS COVERED Already published preclinical and clinical evidence of relacorilant antitumor activity was analyzed and discussed. Ongoing clinical trials registered on clincaltrials.gov were also reported. The review aimed to summarize the status of relacorilant, the mechanism of action, the published and ongoing trials, and its safety and efficacy. EXPERT OPINION Relacorilant combined with nab-paclitaxel, may represent a promising strategy for the treatment of platinum-resistant ovarian cancer patients. After preliminary positive results in terms of clinical efficacy, a randomized phase III trial is ongoing to confirm the findings from the published phase II study.
Collapse
Affiliation(s)
- Elena Giudice
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Vanda Salutari
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carolina Maria Sassu
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Viola Ghizzoni
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Maria Vittoria Carbone
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Laura Vertechy
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anna Fagotti
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Scambia
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudia Marchetti
- Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institute of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
7
|
Elia A, Pataccini G, Saldain L, Ambrosio L, Lanari C, Rojas P. Antiprogestins for breast cancer treatment: We are almost ready. J Steroid Biochem Mol Biol 2024; 241:106515. [PMID: 38554981 DOI: 10.1016/j.jsbmb.2024.106515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/05/2024] [Accepted: 03/20/2024] [Indexed: 04/02/2024]
Abstract
The development of antiprogestins was initially a gynecological purpose. However, since mifepristone was developed, its application for breast cancer treatment was immediately proposed. Later, new compounds with lower antiglucocorticoid and antiandrogenic effects were developed to be applied to different pathologies, including breast cancer. We describe herein the studies performed in the breast cancer field with special focus on those reported in recent years, ranging from preclinical biological models to those carried out in patients. We highlight the potential use of antiprogestins in breast cancer prevention in women with BRCA1 mutations, and their use for breast cancer treatment, emphasizing the need to elucidate which patients will respond. In this sense, the PR isoform ratio has emerged as a possible tool to predict antiprogestin responsiveness. The effects of combined treatments of antiprogestins together with other drugs currently used in the clinic, such as tamoxifen, CDK4/CDK6 inhibitors or pembrolizumab in preclinical models is discussed since it is in this scenario that antiprogestins will be probably introduced. Finally, we explain how transcriptomic or proteomic studies, that were carried out in different luminal breast cancer models and in breast cancer samples that responded or were predicted to respond to the antiprogestin therapy, show a decrease in proliferative pathways. Deregulated pathways intrinsic of each model are discussed, as well as how these analyses may contribute to a better understanding of the mechanisms involved.
Collapse
Affiliation(s)
- Andrés Elia
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME CONICET), Buenos Aires, Argentina
| | - Gabriela Pataccini
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME CONICET), Buenos Aires, Argentina
| | - Leo Saldain
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME CONICET), Buenos Aires, Argentina
| | - Luisa Ambrosio
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME CONICET), Buenos Aires, Argentina
| | - Claudia Lanari
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME CONICET), Buenos Aires, Argentina
| | - Paola Rojas
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME CONICET), Buenos Aires, Argentina.
| |
Collapse
|
8
|
Olawaiye AB, Kim JW, Bagameri A, Bishop E, Chudecka-Głaz A, Devaux A, Gladieff L, Gordinier ME, Korach J, McCollum ME, Mileshkin L, Monk BJ, Nicum S, Nogueira-Rodrigues A, Oaknin A, O'Malley DM, Orlando M, Dreiling L, Tudor IC, Lorusso D. Clinical Trial Protocol for ROSELLA: a phase 3 study of relacorilant in combination with nab-paclitaxel versus nab-paclitaxel monotherapy in advanced platinum-resistant ovarian cancer. J Gynecol Oncol 2024; 35:e111. [PMID: 39032926 PMCID: PMC11262895 DOI: 10.3802/jgo.2024.35.e111] [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: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Ovarian cancer has the highest mortality among gynecologic cancers, primarily because it typically is diagnosed at a late stage and because of the development of chemoresistance in recurrent disease. Improving outcomes in women with platinum-resistant ovarian cancer is a substantial unmet need. Activation of the glucocorticoid receptor (GR) by cortisol has been shown to suppress the apoptotic pathways used by cytotoxic agents, limiting their efficacy. Selective GR modulation may be able to counteract cortisol's antiapoptotic effects, enhancing chemotherapy's efficacy. A previous phase 2 study has shown that adding intermittently dosed relacorilant, a selective GR modulator, to nab-paclitaxel improved outcomes, including progression-free survival (PFS) and overall survival (OS), with minimal added toxicity, in women with recurrent platinum-resistant ovarian cancer. The ROSELLA study aims to confirm and expand on these findings in a larger population. METHODS ROSELLA is a phase 3, randomized, 2-arm, open-label, global multicenter study in women with recurrent, platinum-resistant, high-grade serous epithelial ovarian, primary peritoneal, or fallopian tube cancer. Eligible participants have received 1 to 3 lines of prior systemic anticancer therapy, including ≥1 prior line of platinum therapy and prior treatment with bevacizumab, with documented progressive disease or intolerance to the most recent therapy. There is no biomarker-based requirement for participant selection. Participants are randomized 1:1 to receive intermittently dosed relacorilant in combination with nab-paclitaxel or nab-paclitaxel monotherapy. The study's primary efficacy endpoint is PFS as assessed by blinded independent central review. Secondary efficacy endpoints include OS, investigator-assessed PFS, objective response rate, best overall response, duration of response, clinical benefit rate at 24 weeks, and cancer antigen 125 response. The study is also evaluating safety and patient-reported outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05257408; European Union Drug Regulating Authorities Clinical Trials Database Identifier: 2022-000662-18.
Collapse
Affiliation(s)
- Alexander B Olawaiye
- University of Pittsburgh School of Medicine and Magee-Womens Hospital, Gynecologic Oncology Group, Pittsburgh, PA, USA.
| | - Jae-Weon Kim
- Department of Obstetrics and Gynecology, Seoul National University, Seoul, Korea
| | | | - Erin Bishop
- Medical College of Wisconsin, Gynecologic Oncology Group, Milwaukee, WI, USA
| | - Anita Chudecka-Głaz
- Pomeranian Medical University, Polish Gynecologic Oncology Group, Szczecin, Poland
| | - Alix Devaux
- Oncology Department of Grand Hôpital de Charleroi, Charleroi, Belgium
| | - Laurence Gladieff
- Institut Claudius Regaud-Institut Universitaire du Cancer de Toulouse Oncopole, Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens, Toulouse, France
| | | | - Jacob Korach
- Sheba Medical Center, School of Medicine, Tel Aviv University, Israeli Society of Gynecologic Oncology, Tel Aviv, Israel
| | | | - Linda Mileshkin
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Bradley J Monk
- Gynecologic Oncology Group Foundation; Florida Cancer Specialists and Research Institute, West Palm Beach, FL, USA
| | - Shibani Nicum
- University College London Cancer Institute, National Cancer Research Institute, London, UK
| | | | - Ana Oaknin
- Medical Oncology Service, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - David M O'Malley
- The Ohio State University and the James Cancer Center, Gynecologic Oncology Group, Columbus, OH, USA
| | - Mauro Orlando
- Instituto Alexander Fleming, Buenos Aires, Argentina
| | | | - Iulia C Tudor
- Corcept Therapeutics Incorporated, Menlo Park, CA, USA
| | - Domenica Lorusso
- Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico and Catholic University of the Sacred Heart, Multicentre Italian Trials in Ovarian Cancer and Gynecologic Malignancies, Rome, Italy
| |
Collapse
|
9
|
Taya M, Hou X, Veneris JT, Kazi N, Larson MC, Maurer MJ, Heinzen EP, Chen H, Lastra R, Oberg AL, Weroha SJ, Fleming GF, Conzen SD. Investigation of selective glucocorticoid receptor modulation in high-grade serous ovarian cancer PDX models. J Gynecol Oncol 2024; 36:36.e4. [PMID: 38909640 DOI: 10.3802/jgo.2025.36.e4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/18/2024] [Accepted: 05/07/2024] [Indexed: 06/25/2024] Open
Abstract
OBJECTIVE In ovarian cancer (OvCa), tumor cell high glucocorticoid receptor (GR) has been associated with poor patient prognosis. In vitro, GR activation inhibits chemotherapy-induced OvCa cell death in association with transcriptional upregulation of genes encoding anti-apoptotic proteins. A recent randomized phase II study demonstrated improvement in progression-free survival (PFS) for heavily pre-treated OvCa patients randomized to receive therapy with a selective GR modulator (SGRM) plus chemotherapy compared to chemotherapy alone. We hypothesized that SGRM therapy would improve carboplatin response in OvCa patient-derived xenograft (PDX). METHODS Six high-grade serous (HGS) OvCa PDX models expressing GR mRNA (NR3C1) and protein were treated with chemotherapy +/- SGRM. Tumor size was measured longitudinally by peritoneal transcutaneous ultrasonography. RESULTS One of the 6 GR-positive PDX models showed a significant improvement in PFS with the addition of a SGRM. Interestingly, the single model with an improved PFS was least carboplatin sensitive. Possible explanations for the modest SGRM activity include the high carboplatin sensitivity of 5 of the PDX tumors and the potential that SGRMs activate the tumor invasive immune cells in patients (absent from immunocompromised mice). The level of tumor GR protein expression alone appears insufficient for predicting SGRM response. CONCLUSION The significant improvement in PFS shown in 1 of the 6 models after treatment with a SGRM plus chemotherapy underscores the need to determine predictive biomarkers for SGRM therapy in HGS OvCa and to better identify patient subgroups that are most likely to benefit from adding GR modulation to chemotherapy.
Collapse
Affiliation(s)
- Manisha Taya
- Division of Hematology and Oncology, UT Southwestern, Dallas, TX, USA
| | - Xiaonan Hou
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Jennifer T Veneris
- Department of Medicine, Section of Hematology and Oncology, The University of Chicago, Chicago, IL, USA
| | - Nina Kazi
- Division of Hematology and Oncology, UT Southwestern, Dallas, TX, USA
| | - Melissa C Larson
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Matthew J Maurer
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Ethan P Heinzen
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | - Hao Chen
- Department of Pathology, UT Southwestern, Dallas, TX, USA
| | - Ricardo Lastra
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Ann L Oberg
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - S John Weroha
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Gini F Fleming
- Department of Medicine, Section of Hematology and Oncology, The University of Chicago, Chicago, IL, USA
| | - Suzanne D Conzen
- Division of Hematology and Oncology, UT Southwestern, Dallas, TX, USA.
| |
Collapse
|
10
|
Thakur D, Sengupta D, Mahapatra E, Das S, Sarkar R, Mukherjee S. Glucocorticoid receptor: a harmonizer of cellular plasticity in breast cancer-directs the road towards therapy resistance, metastatic progression and recurrence. Cancer Metastasis Rev 2024; 43:481-499. [PMID: 38170347 DOI: 10.1007/s10555-023-10163-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
Recent therapeutic advances have significantly uplifted the quality of life in breast cancer patients, yet several impediments block the road to disease-free survival. This involves unresponsiveness towards administered therapy, epithelial to mesenchymal transition, and metastatic progression with the eventual appearance of recurrent disease. Attainment of such characteristics is a huge adaptive challenge to which tumour cells respond by acquiring diverse phenotypically plastic states. Several signalling networks and mediators are involved in such a process. Glucocorticoid receptor being a mediator of stress response imparts prognostic significance in the context of breast carcinoma. Involvement of the glucocorticoid receptor in the signalling cascade of breast cancer phenotypic plasticity needs further elucidation. This review attempted to shed light on the inter-regulatory interactions of the glucocorticoid receptor with the mediators of the plasticity program in breast cancer; which may provide a hint for strategizing therapeutics against the glucocorticoid/glucocorticoid receptor axis so as to modulate phenotypic plasticity in breast carcinoma.
Collapse
Affiliation(s)
- Debanjan Thakur
- Department of Environmental Carcinogenesis and Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, India
| | - Debomita Sengupta
- Department of Environmental Carcinogenesis and Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, India
| | - Elizabeth Mahapatra
- Department of Environmental Carcinogenesis and Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, India
| | - Salini Das
- Department of Environmental Carcinogenesis and Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, India
| | - Ruma Sarkar
- B. D. Patel Institute of Paramedical Sciences, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Gujarat, 388421, India
| | - Sutapa Mukherjee
- Department of Environmental Carcinogenesis and Toxicology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, India.
| |
Collapse
|
11
|
Elía A, Saldain L, Lovisi S, Martínez Vazquez P, Burruchaga J, Lamb CA, Lüthy IA, Diez F, Homer NZM, Andrew R, Rojas P, Lanari C. Steroid profile in patients with breast cancer and in mice treated with mifepristone. Endocr Relat Cancer 2024; 31:e230238. [PMID: 37962553 PMCID: PMC10762537 DOI: 10.1530/erc-23-0238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/13/2023] [Indexed: 11/15/2023]
Abstract
Progesterone receptors (PRs) are biomarkers used as prognostic and predictive factors in breast cancer, but they are still not used as therapeutic targets. We have proposed that the ratio between PR isoforms A and B (PRA and PRB) predicts antiprogestin responsiveness. The MIPRA trial confirmed the benefit of 200 mg mifepristone, administered to patients with tumors with a high PRA/PRB ratio, but dose-ranging has not been conducted. The aim of this study was to establish the plasma mifepristone levels of patients from the MIPRA trial, along with the resultant steroid profiles, and compare these with those observed in mifepristone-treated mice using therapeutic schemes able to induce the regression of experimental mammary carcinomas with high PRA/PRB ratios: 6 mg pellets implanted subcutaneously, or daily doses of 12 mg/kg body weight. The plasma levels of mifepristone and other 19 plasma steroids were measured by liquid chromatography-tandem mass spectometry. In mifepristone-treated mice, plasma levels were lower than those registered in mifepristone-treated patients (i.e. day 7 after treatment initiation, pellet-treated mice: 8.4 ± 3.9 ng/mL; mifepristone-treated patients: 300.3 ± 31.7 ng/mL (mean ± s.d.; P < 0.001)). The increase in corticoid related steroids observed in patients was not observed in mifepristone-treated mice. The increase in progesterone levels was the most significant side effect detected in mifepristone-treated mice after 14 or 21 days of treatment, probably due to an ovarian compensatory effect not observed in postmenopausal patients. We conclude that in future clinical trials using mifepristone, the possibility of lowering the standard daily dose of 200 mg should be considered.
Collapse
Affiliation(s)
- Andres Elía
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Argentina
| | - Leo Saldain
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Argentina
| | - Silvia Lovisi
- Hospital de Agudos “Magdalena V de Martínez”, General Pacheco, Argentina
| | | | - Javier Burruchaga
- Hospital de Agudos “Magdalena V de Martínez”, General Pacheco, Argentina
| | - Caroline A Lamb
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Argentina
| | | | - Federico Diez
- University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, UK
| | - Natalie Z M Homer
- University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, UK
| | - Ruth Andrew
- University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, UK
| | - Paola Rojas
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Argentina
| | - Claudia Lanari
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Argentina
| |
Collapse
|
12
|
Bakhshi P, Ho JQ, Zanganeh S. Sex-specific outcomes in cancer therapy: the central role of hormones. FRONTIERS IN MEDICAL TECHNOLOGY 2024; 6:1320690. [PMID: 38362126 PMCID: PMC10867131 DOI: 10.3389/fmedt.2024.1320690] [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: 10/12/2023] [Accepted: 01/08/2024] [Indexed: 02/17/2024] Open
Abstract
Sex hormones play a pivotal role in modulating various physiological processes, with emerging evidence underscoring their influence on cancer progression and treatment outcomes. This review delves into the intricate relationship between sex hormones and cancer, elucidating the underlying biological mechanisms and their clinical implications. We explore the multifaceted roles of estrogen, androgens, and progesterone, highlighting their respective influence on specific cancers such as breast, ovarian, endometrial, and prostate. Special attention is given to estrogen receptor-positive (ER+) and estrogen receptor-negative (ER-) tumors, androgen receptor signaling, and the dual role of progesterone in both promoting and inhibiting cancer progression. Clinical observations reveal varied treatment responses contingent upon hormonal levels, with certain therapies like tamoxifen, aromatase inhibitors, and anti-androgens demonstrating notable success. However, disparities in treatment outcomes between males and females in hormone-sensitive cancers necessitate further exploration. Therapeutically, the utilization of hormone replacement therapy (HRT) during cancer treatments presents both potential risks and benefits. The promise of personalized therapies, tailored to an individual's hormonal profile, offers a novel approach to optimizing therapeutic outcomes. Concurrently, the burgeoning exploration of new drugs and interventions targeting hormonal pathways heralds a future of more effective and precise treatments for hormone-sensitive cancers. This review underscores the pressing need for a deeper understanding of sex hormones in cancer therapy and the ensuing implications for future therapeutic innovations.
Collapse
Affiliation(s)
- Parisa Bakhshi
- Research and Development, MetasFree Biopharmaceutical Company, Mansfield, MA, United States
| | - Jim Q. Ho
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Steven Zanganeh
- Research and Development, MetasFree Biopharmaceutical Company, Mansfield, MA, United States
| |
Collapse
|
13
|
Alghamdi SS, Alshafi RA, Huwaizi S, Suliman RS, Mohammed AE, Alehaideb ZI, Alturki AY, Alghashem SA, Rahman I. Exploring in vitro and in silico Biological Activities of Calligonum Comosum and Rumex Vesicarius: Implications on Anticancer and Antibacterial Therapeutics. Saudi Pharm J 2023; 31:101794. [PMID: 37822695 PMCID: PMC10562755 DOI: 10.1016/j.jsps.2023.101794] [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: 05/11/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023] Open
Abstract
Introduction The adverse effects of clinically used anti-cancer medication and the rise in resistive micro-organisms have limited therapeutic options. Multiple anti-cancer drugs are derived from medicinal herbs which also have shown anti-bacterial effects. This study aimed to identify the optimal extraction solvent for detecting the cytotoxic and anti-bacterial effects of Calligonum comosum (C. Comosum) and Rumex vesicarius (R. Vesicarius) extracts. Additionally, the study aimed to identify active metabolites and assess their potential as future drug candidates for anti-cancer and anti-bacterial therapeutics. Methods Leaves from both plants were extracted using ethanol, ethyl acetate, chloroform, and water. The cytotoxic effects of the extracts were tested on liver, colon, and breast cancer cell lines. Apoptosis was assessed using High Content Imaging (HCI) and the ApoTox triplex Glo assay. The anti-bacterial effects were determined using agar-well diffusion. Liquid chromatography-mass spectrometry (LC-MS) was used to tentatively identify the secondary metabolites. In silico computational studies were conducted to determine the metabolites' mode of action, safety, and pharmacokinetic properties. Results The ethanolic extract of C. Comosum exhibited potent cytotoxicity on breast cancer cell lines, with IC50 values of 54.97 μg/mL and 58 μg/mL for KAIMRC2 and MDA-MB-231, respectively. It also induced apoptosis in colon and breast cancer cell lines. All tested extracts of C. Comosum and R. Vesicarius demonstrated anti-bacterial activity against Staphylococcus aureus and Escherichia coli. Seven active metabolites were identified, one of which is Kaempferol 3-O-Glucoside-7-O-Rhamnoside, which showed strong (predicted) anti-cancer activity. Kaempferol 3-O-Glucoside-7-O-Rhamnoside and Quercetin-3-O-Glucuronide also exhibited potential anti-bacterial effects on gram-positive and negative bacteria. Conclusion Ethanol extraction of C. Comosum solubilizes active metabolites with potential therapeutic applications in cancer treatment and bacterial infections. Kaempferol 3-O-Glucoside-7-O-Rhamnoside, in particular, shows promise as a dual therapeutic drug candidate for further research and development to improve its efficacy, safety, and pharmacokinetic profile.
Collapse
Affiliation(s)
- Sahar S. Alghamdi
- College of Pharmacy (COP), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- King Abdulaziz Medical City, Ministry of the National Guard – Health Affairs, Riyadh 11426, Saudi Arabia
| | - Raghad A. Alshafi
- College of Pharmacy (COP), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Sarah Huwaizi
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Rasha S. Suliman
- Pharmacy Department, Fatima College of Health Sciences (FCHS), Abu Dhabi, United Arab Emirates
| | - Afrah E. Mohammed
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University (PNU), P.O. Box 16 84428, Riyadh 11671, Saudi Arabia
| | - Zeyad I. Alehaideb
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Allulu Y. Alturki
- College of Pharmacy (COP), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Sara A. Alghashem
- College of Pharmacy (COP), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Ishrat Rahman
- Department of Basic Dental Sciences, College of Dentistry, Princess Nourah bint Abdulrahman University (PNU), P.O. Box 84428, Riyadh 11671, Saudi Arabia
| |
Collapse
|
14
|
Colombo N, Van Gorp T, Matulonis UA, Oaknin A, Grisham RN, Fleming GF, Olawaiye AB, Nguyen DD, Greenstein AE, Custodio JM, Pashova HI, Tudor IC, Lorusso D. Relacorilant + Nab-Paclitaxel in Patients With Recurrent, Platinum-Resistant Ovarian Cancer: A Three-Arm, Randomized, Controlled, Open-Label Phase II Study. J Clin Oncol 2023; 41:4779-4789. [PMID: 37364223 PMCID: PMC10602497 DOI: 10.1200/jco.22.02624] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 04/21/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
PURPOSE Despite therapeutic advances, outcomes for patients with platinum-resistant/refractory ovarian cancer remain poor. Selective glucocorticoid receptor modulation with relacorilant may restore chemosensitivity and enhance chemotherapy efficacy. METHODS This three-arm, randomized, controlled, open-label phase II study (ClinicalTrials.gov identifier: NCT03776812) enrolled women with recurrent, platinum-resistant/refractory, high-grade serous or endometrioid epithelial ovarian, primary peritoneal, or fallopian tube cancer, or ovarian carcinosarcoma treated with ≤4 prior chemotherapeutic regimens. Patients were randomly assigned 1:1:1 to (1) nab-paclitaxel (80 mg/m2) + intermittent relacorilant (150 mg the day before, of, and after nab-paclitaxel); (2) nab-paclitaxel (80 mg/m2) + continuous relacorilant (100 mg once daily); or (3) nab-paclitaxel monotherapy (100 mg/m2). Nab-paclitaxel was administered on days 1, 8, and 15 of each 28-day cycle. The primary end point was progression-free survival (PFS) by investigator assessment; objective response rate (ORR), duration of response (DOR), overall survival (OS), and safety were secondary end points. RESULTS A total of 178 women were randomly assigned. Intermittent relacorilant + nab-paclitaxel improved PFS (hazard ratio [HR], 0.66; log-rank test P = .038; median follow-up, 11.1 months) and DOR (HR, 0.36; P = .006) versus nab-paclitaxel monotherapy, while ORR was similar across arms. At the preplanned OS analysis (median follow-up, 22.5 months), the OS HR was 0.67 (P = .066) for the intermittent arm versus nab-paclitaxel monotherapy. Continuous relacorilant + nab-paclitaxel showed numerically improved median PFS but did not result in significant improvement over nab-paclitaxel monotherapy. Adverse events were comparable across study arms, with neutropenia, anemia, peripheral neuropathy, and fatigue/asthenia being the most common grade ≥3 adverse events. CONCLUSION Intermittent relacorilant + nab-paclitaxel improved PFS, DOR, and OS compared with nab-paclitaxel monotherapy. On the basis of protocol-prespecified Hochberg step-up multiplicity adjustment, the primary end point did not reach statistical significance (P < .025). A phase III evaluation of this regimen is underway (ClinicalTrials.gov identifier: NCT05257408).
Collapse
Affiliation(s)
- Nicoletta Colombo
- Gynecologic Oncology Program, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Medicine and Surgery, University Milan-Bicocca, Milan, Italy
| | - Toon Van Gorp
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University Hospital Leuven, Leuven Cancer Institute, Leuven, Belgium
| | | | - Ana Oaknin
- Gynaecologic Cancer Programme, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Rachel N. Grisham
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, NY
| | | | - Alexander B. Olawaiye
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | | | | | | | | | - Domenica Lorusso
- Fondazione Policlinico Universitario Gemelli IRCCS, Catholic University of Sacred Heart, Rome, Italy
| |
Collapse
|
15
|
Johnson KCC, Goldstein D, Tharakan J, Quiroga D, Kassem M, Grimm M, Miah A, Vargo C, Berger M, Sudheendra P, Pariser A, Gatti-Mays ME, Williams N, Stover D, Sardesai S, Wesolowski R, Ramaswamy B, Tozbikian G, Schnell PM, Cherian MA. The Immunomodulatory Effects of Dexamethasone on Neoadjuvant Chemotherapy for Triple-Negative Breast Cancer. Oncol Ther 2023; 11:361-374. [PMID: 37354381 PMCID: PMC10447758 DOI: 10.1007/s40487-023-00235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 05/26/2023] [Indexed: 06/26/2023] Open
Abstract
INTRODUCTION The immunomodulatory impact of corticosteroids and concurrent chemotherapy is poorly understood within triple-negative breast cancer (TNBC). On a biochemical level, steroids have been linked to the signaling of chemotherapy-resistant pathways. However, on a clinical level, steroids play an essential role in chemotherapy tolerance through the prevention of chemotherapy-induced nausea and vomiting (CINV) and hypersensitivity reactions. Given these conflicting roles, we wanted to evaluate this interplay more rigorously in the context of early-stage TNBC. METHODS We performed a retrospective analysis of patients with operable TNBC who received neoadjuvant chemotherapy (NAC) between January 2012 and November 2018, with the primary goal of examining the dose-dependent relationship between pathological complete response (pCR) rates and corticosteroid use. Secondary endpoints included the impact of steroid dosing on overall survival (OS) and recurrence-free survival (RFS), along with a breakdown in pCR rates based on steroid doses provided during each chemotherapy phase. Further adjusted analyses were performed based on patient age, diabetic status, and anatomical stage. Finally, we explored the relationship between tumor-infiltrating lymphocytes (TILs) seen on tissue samples at baseline and dexamethasone doses in terms of pCR rates. RESULTS In total, of the 174 patients screened within this study period, 116 met full eligibility criteria. Of these eligible patients, all were female, with a median age of 51.5 years (27.0 to 74.0) and a mean body mass index (BMI) of 29.7 [standard deviation (SD) 7.04]. The majority were nondiabetic (80.2%). For cancer stage, 69.8% (n = 81) had stage 2 breast cancer. We found no statistically significant association between pCR rates and dexamethasone use, both in terms of the total dose (p = 0.55) and mean dose per NAC cycle (p = 0.74). Similarly, no difference was noted when adjusting for diabetic status, metformin use, or age at diagnosis, regardless of the total steroid dose provided (p = 0.72) or mean dose per cycle (p = 0.49). No meaningful changes to pCR rate were seen with higher mean or higher total steroid doses during the paclitaxel (T) phase (adjusted p = 0.16 and p = 0.76, respectively) or doxorubicin and cyclophosphamide (AC) phase (adjusted p = 0.83 and p = 0.77, respectively). Furthermore, we found no clinically significant association between dexamethasone dose and either RFS (p = 0.45) or OS (p = 0.89). Of the 56 patients who had available pre-treatment biopsy tissue samples, 27 achieved pCR, with higher TILs at baseline being associated with higher pCR rates, regardless of the mean dexamethasone dose used. CONCLUSION Our findings demonstrate that dexamethasone has no clinically significant impact on pCR, RFS, or OS when given concurrently with NAC in patients with curative TNBC, regardless of diabetic status.
Collapse
Affiliation(s)
- Kai Conrad Cecil Johnson
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | | | - Jasmin Tharakan
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Dionisia Quiroga
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Mahmoud Kassem
- Department of Surgery, Mercy Health West Hospital, Cincinnati, OH, USA
| | - Michael Grimm
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Abdul Miah
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Craig Vargo
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Michael Berger
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Preeti Sudheendra
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Ashley Pariser
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Margaret E Gatti-Mays
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Nicole Williams
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Daniel Stover
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Sagar Sardesai
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Robert Wesolowski
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Gary Tozbikian
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Patrick M Schnell
- Division of Biostatistics, The Ohio State University College of Public Health, Columbus, OH, USA
| | - Mathew A Cherian
- Division of Medical Oncology, Wexner Medical Center, The OH State University Comprehensive Cancer Center-James Cancer Hospital and Solove Research Institute, Biomedical Research Tower, Room 888, 460 W 12th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
16
|
Gu W, Zheng H, Canessa CM. Phosphatases maintain low catalytic activity of SGK1: DNA damage resets the balance in favor of phosphorylation. J Biol Chem 2023; 299:104941. [PMID: 37343701 PMCID: PMC10372406 DOI: 10.1016/j.jbc.2023.104941] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/23/2023] Open
Abstract
The serum- and glucocorticoid-induced kinase 1 (SGK1) promotes cell survival under stress conditions and facilitates the emergence of drug resistance in cancer. The underlying mechanisms of these observations are not fully understood. In this study, we found that SGK1 activity is suppressed by the action of the S/T phosphatases PP5 and PP2A, which constantly dephosphorylate SGK1. Using newly developed anti-phospho SGK1 antibodies and inhibitors of phosphatases, we determined that the high degree of dephosphorylation is caused by two factors: the tendency of SGK1 to unfold, which makes it dependent on Hsp90 chaperone complexes composed of four proteins, Hsp90/CDC37/PP5/SGK1, and where the phosphatase PP5 persistently dephosphorylates SGK1 within the complex. SGK1 binding to PP2A regulatory subunits B55γ and B55δ brings PP2A catalytic subunit close to exposed SGK1 phosphoresidues. A further association of phosphorylated pS37-FAM122A-an endogenous inhibitor of PP2A-to the holoenzyme diminishes dephosphorylation of SGK1 mediated by PP2A. Our study also reveals that genotoxic stress can reverse the dominant impact of phosphatases over kinases by activating the DNA-dependent protein kinase, which enhances mTORC2 activity directed to SGK1. Thus, our results provide insight into a molecular pathway that enables SGK1 to gain phosphorylation and catalytic activity and promote cell survival, potentially diminishing the efficacy of cancer treatments. As the DNA damage response operates in many cancer cells and is further induced by chemotherapies, the findings of this study could have significant implications for the development of novel cancer therapies targeting SGK1.
Collapse
Affiliation(s)
- Wenxue Gu
- School of Medicine, Tsinghua University, Beijing, China
| | - Hongyan Zheng
- School of Medicine, Tsinghua University, Beijing, China
| | - Cecilia M Canessa
- School of Medicine, Tsinghua University, Beijing, China; Cellular and Molecular Physiology, School of Medicine, Yale University, New Haven, USA.
| |
Collapse
|
17
|
Lang JD, Nguyen TVV, Levin MK, Blas PE, Williams HL, Rodriguez ESR, Briones N, Mueller C, Selleck W, Moore S, Zismann VL, Hendricks WPD, Espina V, O'Shaughnessy J. Pilot clinical trial and phenotypic analysis in chemotherapy-pretreated, metastatic triple-negative breast cancer patients treated with oral TAK-228 and TAK-117 (PIKTOR) to increase DNA damage repair deficiency followed by cisplatin and nab paclitaxel. Biomark Res 2023; 11:73. [PMID: 37491309 PMCID: PMC10369813 DOI: 10.1186/s40364-023-00511-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/04/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND A subset of triple-negative breast cancers (TNBCs) have homologous recombination deficiency with upregulation of compensatory DNA repair pathways. PIKTOR, a combination of TAK-228 (TORC1/2 inhibitor) and TAK-117 (PI3Kα inhibitor), is hypothesized to increase genomic instability and increase DNA damage repair (DDR) deficiency, leading to increased sensitivity to DNA-damaging chemotherapy and to immune checkpoint blockade inhibitors. METHODS 10 metastatic TNBC patients received 4 mg TAK-228 and 200 mg TAK-117 (PIKTOR) orally each day for 3 days followed by 4 days off, weekly, until disease progression (PD), followed by intravenous cisplatin 75 mg/m2 plus nab paclitaxel 220 mg/m2 every 3 weeks for up to 6 cycles. Patients received subsequent treatment with pembrolizumab and/or chemotherapy. Primary endpoints were objective response rate with cisplatin/nab paclitaxel and safety. Biopsies of a metastatic lesion were collected prior to and at PD on PIKTOR. Whole exome and RNA-sequencing and reverse phase protein arrays (RPPA) were used to phenotype tumors pre- and post-PIKTOR for alterations in DDR, proliferation, and immune response. RESULTS With cisplatin/nab paclitaxel (cis/nab pac) therapy post PIKTOR, 3 patients had clinical benefit (1 partial response (PR) and 2 stable disease (SD) ≥ 6 months) and continued to have durable benefit in progression-free survival with pembrolizumab post-cis/nab pac for 1.2, 2, and 3.6 years. Their post-PIKTOR metastatic tissue displayed decreased mismatch repair (MMR), increased tumor mutation burden, and significantly lower levels of 53BP1, DAG Lipase β, GCN2, AKT Ser473, and PKCzeta Thr410/403 compared to pre-PIKTOR tumor tissue. CONCLUSIONS Priming patients' chemotherapy-pretreated metastatic TNBC with PIKTOR led to very prolonged response/disease control with subsequent cis/nab pac, followed by pembrolizumab, in 3 of 10 treated patients. Our multi-omics approach revealed a higher number of genomic alterations, reductions in MMR, and alterations in immune and stress response pathways post-PIKTOR in patients who had durable responses. TRIAL REGISTRATION This clinical trial was registered on June 21, 2017, at ClinicalTrials.gov using identifier NCT03193853.
Collapse
Affiliation(s)
- Jessica D Lang
- The Translational Genomics Research Institute (TGen), Integrated Cancer Genomics Division, Phoenix, AZ, 85004, USA
- Department of Pathology and Laboratory Medicine, Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Tuong Vi V Nguyen
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 22030, USA
| | - Maren K Levin
- Baylor Scott & White Research Institute, Dallas, TX, 75246, USA
| | - Page E Blas
- Baylor Scott & White Research Institute, Dallas, TX, 75246, USA
| | | | | | - Natalia Briones
- The Translational Genomics Research Institute (TGen), Integrated Cancer Genomics Division, Phoenix, AZ, 85004, USA
| | - Claudius Mueller
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 22030, USA
| | - William Selleck
- The Translational Genomics Research Institute (TGen), Integrated Cancer Genomics Division, Phoenix, AZ, 85004, USA
| | - Sarah Moore
- The Translational Genomics Research Institute (TGen), Integrated Cancer Genomics Division, Phoenix, AZ, 85004, USA
| | - Victoria L Zismann
- The Translational Genomics Research Institute (TGen), Integrated Cancer Genomics Division, Phoenix, AZ, 85004, USA
| | - William P D Hendricks
- The Translational Genomics Research Institute (TGen), Integrated Cancer Genomics Division, Phoenix, AZ, 85004, USA
| | - Virginia Espina
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 22030, USA
| | - Joyce O'Shaughnessy
- Baylor University Medical Center, Texas Oncology, 3410 Worth Street, Suite 400, Dallas, TX, 75246, USA.
| |
Collapse
|
18
|
Zhang J, Miki Y, Iwabuchi E, Xu J, Kanai A, Sagara Y, Ohi Y, Rai Y, Yamaguchi R, Tanaka M, Ishida T, Suzuki T, Sasano H. Induction of SGK1 via glucocorticoid-influenced clinical outcome of triple-negative breast cancer patients. Breast Cancer Res Treat 2023:10.1007/s10549-023-06990-4. [PMID: 37286891 DOI: 10.1007/s10549-023-06990-4] [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: 04/11/2023] [Accepted: 05/24/2023] [Indexed: 06/09/2023]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is a highly heterogeneous and aggressive breast malignancy. Glucocorticoid (GC)-glucocorticoid receptor (GR) pathway plays a pivotal role in the cellular responses to various stresses including chemotherapy. Serum- and glucocorticoid-induced kinase-1 (SGK1) is known as an important downstream effector molecule in the GR signaling pathway, we attempted to explore its clinicopathological and functional significance in TNBC in which GR is expressed. METHODS We first immunolocalized GR and SGK1 and correlated the results with clinicopathological variables and clinical outcome in 131 TNBC patients. We also evaluated the effects of SGK1 on the cell proliferation and migration in TNBC cell lines with administration of dexamethasone (DEX) to further clarify the significance of SGK1. RESULTS The status of SGK1 in carcinoma cells was significantly associated with adverse clinical outcome in TNBC patients examined and was significantly associated with lymph node metastasis, pathological stage, and lymphatic invasion of the patients. In particular, SGK1 immunoreactivity was significantly associated with an increased risk of recurrence in GR-positive TNBC patients. Subsequent in vitro studies also demonstrated that DEX promoted TNBC cell migration and the silencing of gene expression did inhibit the cell proliferation and migration of TNBC cells under DEX treatment. CONCLUSIONS To the best of our knowledge, this is the first study to explore an association between SGK1 and clinicopathological variables and clinical outcome of TNBC patients. SGK1 status was significantly positively correlated with adverse clinical outcome of TNBC patients and promoted carcinoma cell proliferation and migration of carcinoma cells.
Collapse
Affiliation(s)
- Junjia Zhang
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Miki
- Department of Nursing, Faculty of Medical Science and Welfare, Tohoku Bunka Gakuen University, Sendai, Japan.
| | - Erina Iwabuchi
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junyao Xu
- The Cancer Hospital of the University of Chinese Academy of Sciences Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Ayako Kanai
- Department of Breast Surgery, Hachinohe City Hospital, Aomori, Japan
| | - Yasuaki Sagara
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Kagoshima, Japan
| | - Yasuyo Ohi
- Department of Pathology, Sagara Hospital, Kagoshima, Japan
| | - Yoshiaki Rai
- Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Kagoshima, Japan
| | - Rin Yamaguchi
- Department of Pathology, Nagasaki University Hospital, Nagasaki, Japan
| | - Maki Tanaka
- JCHO Kurume General Hospital, Fukuoka, Japan
| | - Takanori Ishida
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Suzuki
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
19
|
Yan J, Chen Y, Luo M, Hu X, Li H, Liu Q, Zou Z. Chronic stress in solid tumor development: from mechanisms to interventions. J Biomed Sci 2023; 30:8. [PMID: 36707854 PMCID: PMC9883141 DOI: 10.1186/s12929-023-00903-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/17/2023] [Indexed: 01/29/2023] Open
Abstract
Chronic stress results in disturbances of body hormones through the neuroendocrine system. Cancer patients often experience recurrent anxiety and restlessness during disease progression and treatment, which aggravates disease progression and hinders treatment effects. Recent studies have shown that chronic stress-regulated neuroendocrine systems secret hormones to activate many signaling pathways related to tumor development in tumor cells. The activated neuroendocrine system acts not only on tumor cells but also modulates the survival and metabolic changes of surrounding non-cancerous cells. Current clinical evidences also suggest that chronic stress affects the outcome of cancer treatment. However, in clinic, there is lack of effective treatment for chronic stress in cancer patients. In this review, we discuss the main mechanisms by which chronic stress regulates the tumor microenvironment, including functional regulation of tumor cells by stress hormones (stem cell-like properties, metastasis, angiogenesis, DNA damage accumulation, and apoptotic resistance), metabolic reprogramming and immune escape, and peritumor neuromodulation. Based on the current clinical treatment framework for cancer and chronic stress, we also summarize pharmacological and non-pharmacological therapeutic approaches to provide some directions for cancer therapy.
Collapse
Affiliation(s)
- Jiajing Yan
- grid.263785.d0000 0004 0368 7397MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631 China
| | - Yibing Chen
- grid.207374.50000 0001 2189 3846Department of Gynecology and Obstetrics, First Affiliated Hospital, Genetic and Prenatal Diagnosis Center, Zhengzhou University, Zhengzhou, 450001 China
| | - Minhua Luo
- grid.263785.d0000 0004 0368 7397MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631 China
| | - Xinyu Hu
- grid.263785.d0000 0004 0368 7397MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631 China
| | - Hongsheng Li
- grid.410737.60000 0000 8653 1072Department of Breast Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095 China
| | - Quentin Liu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510631 China ,grid.411971.b0000 0000 9558 1426Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044 Liaoning China
| | - Zhengzhi Zou
- grid.263785.d0000 0004 0368 7397MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631 China ,grid.263785.d0000 0004 0368 7397Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, 510631 China
| |
Collapse
|
20
|
Blanco-Nistal MM, Fernández-Fernández JA. Glucocorticoid Effect in Cancer Patients. Methods Mol Biol 2023; 2704:339-352. [PMID: 37642855 DOI: 10.1007/978-1-0716-3385-4_21] [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: 08/31/2023]
Abstract
The use of glucocorticoids is very varied in the context of cancer patients and includes the treatment of symptoms related to cancer, but also the management of the most common side effects of antitumor treatments or adverse events related to the immune system. There is a quantity of experimental evidence demonstrating that cancer cells are immunogenic. However, the effective activation of anticancer T cell responses closely depends on an efficient antigen presentation carried out by professional antigen-presenting cells such as dendritic cells (DCs). The classic strategies to improve the medical management of inflammation are aimed at exacerbating the host's immune response. Although successful in treating a number of diseases, these drugs have limited efficacy and variable responses can lead to unpredictable results. The ideal therapy should reduce inflammation without inducing immunosuppression and remains a challenge for healthcare personnel.
Collapse
|
21
|
Mitre-Aguilar IB, Moreno-Mitre D, Melendez-Zajgla J, Maldonado V, Jacobo-Herrera NJ, Ramirez-Gonzalez V, Mendoza-Almanza G. The Role of Glucocorticoids in Breast Cancer Therapy. Curr Oncol 2022; 30:298-314. [PMID: 36661673 PMCID: PMC9858160 DOI: 10.3390/curroncol30010024] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Glucocorticoids (GCs) are anti-inflammatory and immunosuppressive steroid molecules secreted by the adrenal gland and regulated by the hypothalamic-pituitary-adrenal (HPA) axis. GCs present a circadian release pattern under normal conditions; they increase their release under stress conditions. Their mechanism of action can be via the receptor-independent or receptor-dependent pathway. The receptor-dependent pathway translocates to the nucleus, where the ligand-receptor complex binds to specific sequences in the DNA to modulate the transcription of specific genes. The glucocorticoid receptor (GR) and its endogenous ligand cortisol (CORT) in humans, and corticosterone in rodents or its exogenous ligand, dexamethasone (DEX), have been extensively studied in breast cancer. Its clinical utility in oncology has mainly focused on using DEX as an antiemetic to prevent chemotherapy-induced nausea and vomiting. In this review, we compile the results reported in the literature in recent years, highlighting current trends and unresolved controversies in this field. Specifically, in breast cancer, GR is considered a marker of poor prognosis, and a therapeutic target for the triple-negative breast cancer (TNBC) subtype, and efforts are being made to develop better GR antagonists with fewer side effects. It is necessary to know the type of breast cancer to differentiate the treatment for estrogen receptor (ER)-positive, ER-negative, and TNBC, to implement therapies that include the use of GCs.
Collapse
Affiliation(s)
- Irma B. Mitre-Aguilar
- Unidad de Bioquimica, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran (INCMNSZ), Mexico City 14080, Mexico
| | - Daniel Moreno-Mitre
- Centro de Desarrollo de Destrezas Médicas (CEDDEM), Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran (INCMNSZ), Mexico City 14080, Mexico
| | - Jorge Melendez-Zajgla
- Laboratorio de Genomica Funcional del Cancer, Instituto Nacional de Medicina Genomica (INMEGEN), Mexico City 14610, Mexico
| | - Vilma Maldonado
- Laboratorio de Epigenetica, Instituto Nacional de Medicina Genomica (INMEGEN), Mexico City 14610, Mexico
| | - Nadia J. Jacobo-Herrera
- Unidad de Bioquimica, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran (INCMNSZ), Mexico City 14080, Mexico
| | - Victoria Ramirez-Gonzalez
- Departamento de Cirugía-Experimental, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran (INCMNSZ), Mexico City 14080, Mexico
| | - Gretel Mendoza-Almanza
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Laboratorio de Epigenetica, Instituto Nacional de Medicina Genomica (INMEGEN), Mexico City 14610, Mexico
| |
Collapse
|
22
|
Gradowski Farias da Costa do Nascimento T, de Oliveira Thomazini ME, de França Junior N, de Castro Poncio L, Fonseca AS, de Figueiredo BC, Weber SH, Herai RH, de Noronha L, Cavalli LR, Feltes BC, Elifio-Esposito S. Systems biology network reveals the correlation between COX-2 expression and Ch 7q copy number alterations in Ch 11q-deleted pediatric neuroblastoma tumors. Genes Cancer 2022; 13:60-71. [PMCID: PMC9718587 DOI: 10.18632/genesandcancer.225] [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: 06/02/2022] [Accepted: 11/18/2022] [Indexed: 12/05/2022] Open
Abstract
Tumor-associated inflammation and chromosomal aberrations can play crucial roles in cancer development and progression. In neuroblastoma (NB), the enzyme cyclooxygenase-2 (COX-2) is associated with copy number alterations on the long arm of chromosome 11 (Ch 11q), defining an aggressive disease subset. This retrospective study included formalin-fixed paraffin-embedded tumor samples collected from nine patients during diagnosis at the pediatric Pequeno Principe Hospital, Curitiba, PR, Brazil, and post-chemotherapy (CT). COX-2 expression was evaluated using immunohistochemistry and correlated with the genome profile of paired pre- and post-CT samples, determined by array comparative genomic hybridization. A systems biology approach elucidated the PTGS2 network interaction. The results showed positive correlations between pre-CT Ch 7q gain and COX-2 expression (ρ = 0.825; p-value = 0.006) and negative correlations between Ch 7q gain and Ch 11q deletion (ρ = −0.919; p-value = 0.0005). Three samples showed Ch 11q deletion and Ch 7q gain. Network analysis identified a direct connection between CAV-1 (Ch 7q) and COX-2 in NB tumors and highlighted the connection between amplified genes in Ch 7q and deleted ones in 11q. The identification of hub-bottleneck-switch genes provides new biological insights into this connection between NB, tumorigenesis, and inflammation.
Collapse
Affiliation(s)
| | - Mateus Eduardo de Oliveira Thomazini
- 1Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil,2Biotechnology Undergraduate Program. School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
| | - Nilton de França Junior
- 1Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
| | | | - Aline Simoneti Fonseca
- 3Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | | | - Saulo Henrique Weber
- 4Graduate Program in Animal Science, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
| | - Roberto Hirochi Herai
- 1Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil,5Research Department, Instituto Buko Kaesemodel (IBK), Curitiba, Paraná, Brazil
| | - Lucia de Noronha
- 1Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
| | - Luciane R. Cavalli
- 3Research Institute Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil,6Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - Bruno César Feltes
- 7Institute of Informatics, Department of Theoretical Informatics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil,8Institute of Biosciences, Department of Biophysics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Selene Elifio-Esposito
- 1Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil,Correspondence to:Selene Elifio-Esposito, email:
| |
Collapse
|
23
|
Badawy AB. Tryptophan metabolism and disposition in cancer biology and immunotherapy. Biosci Rep 2022; 42:BSR20221682. [PMID: 36286592 PMCID: PMC9653095 DOI: 10.1042/bsr20221682] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 08/31/2023] Open
Abstract
Tumours utilise tryptophan (Trp) and its metabolites to promote their growth and evade host defences. They recruit Trp through up-regulation of Trp transporters, and up-regulate key enzymes of Trp degradation and down-regulate others. Thus, Trp 2,3-dioxygenase (TDO2), indoleamine 2,3-dioxygenase 1 (IDO1), IDO2, N'-formylkynurenine formamidase (FAMID) and Kyn aminotransferase 1 (KAT1) are all up-regulated in many cancer types, whereas Kyn monooxygenase (KMO), kynureninase (KYNU), 2-amino-3-carboxymuconic acid-6-semialdehyde decarboxylase (ACMSD) and quinolinate phosphoribosyltransferase (QPRT) are up-regulated in a few, but down-regulated in many, cancers. This results in accumulation of the aryl hydrocarbon receptor (AhR) ligand kynurenic acid and in depriving the host of NAD+ by blocking its synthesis from quinolinic acid. The host loses more NAD+ by up-regulation of the NAD+-consuming poly (ADP-ribose) polymerases (PARPs) and the protein acetylaters SIRTs. The nicotinamide arising from PARP and SIRT activation can be recycled in tumours to NAD+ by the up-regulated key enzymes of the salvage pathway. Up-regulation of the Trp transporters SLC1A5 and SLC7A5 is associated mostly with that of TDO2 = FAMID > KAT1 > IDO2 > IDO1. Tumours down-regulate enzymes of serotonin synthesis, thereby removing competition for Trp from the serotonin pathway. Strategies for combating tumoral immune escape could involve inhibition of Trp transport into tumours, inhibition of TDO and IDOs, inhibition of FAMID, inhibition of KAT and KYNU, inhibition of NMPRT and NMNAT, inhibition of the AhR, IL-4I1, PARPs and SIRTs, and by decreasing plasma free Trp availability to tumours by albumin infusion or antilipolytic agents and inhibition of glucocorticoid induction of TDO by glucocorticoid antagonism.
Collapse
Affiliation(s)
- Abdulla A.-B. Badawy
- Formerly School of Health Sciences, Cardiff Metropolitan University, Western Avenue, Cardiff CF5 2YB, Wales, U.K
| |
Collapse
|
24
|
Munster PN, Greenstein AE, Fleming GF, Borazanci E, Sharma MR, Custodio JM, Tudor IC, Pashova HI, Shepherd SP, Grauer A, Sachdev JC. Overcoming Taxane Resistance: Preclinical and Phase 1 Studies of Relacorilant, a Selective Glucocorticoid Receptor Modulator, with Nab-Paclitaxel in Solid Tumors. Clin Cancer Res 2022; 28:3214-3224. [PMID: 35583817 PMCID: PMC9662918 DOI: 10.1158/1078-0432.ccr-21-4363] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/01/2022] [Accepted: 05/16/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Chemotherapy resistance remains a major problem in many solid tumors, including breast, ovarian, and pancreatic cancer. Glucocorticoids are one potential driver of chemotherapy resistance as they can mediate tumor progression via induction of cell-survival pathways. We investigated whether combining the selective glucocorticoid receptor (GR) modulator relacorilant with taxanes can enhance antitumor activity. PATIENTS AND METHODS The effect of relacorilant on paclitaxel efficacy was assessed in OVCAR5 cells in vitro and in the MIA PaCa-2 xenograft. A phase 1 study of patients with advanced solid tumors was conducted to determine the recommended phase 2 dose of relacorilant + nab-paclitaxel. RESULTS In OVCAR5 cells, relacorilant reversed the deleterious effects of glucocorticoids on paclitaxel efficacy (P < 0.001). Compared with paclitaxel alone, relacorilant + paclitaxel reduced tumor growth and slowed time to progression in xenograft models (both P < 0.0001). In the heavily pretreated phase 1 population [median (range) of prior regimens: 3 (1-8), prior taxane in 75.3% (55/73)], 33% (19/57) of response-evaluable patients achieved durable disease control (≥16 weeks) with relacorilant + nab-paclitaxel and 28.6% (12/42) experienced longer duration of benefit than on prior taxane (up to 6.4×). The most common dose-limiting toxicity of the combination was neutropenia, which was manageable with prophylactic G-CSF. Clinical benefit with relacorilant + nab-paclitaxel was also associated with GR-regulated transcript-level changes in a panel of GR-controlled genes. CONCLUSIONS The observed preclinical, clinical, and GR-specific pharmacodynamic responses demonstrate that selective GR modulation with relacorilant combined with nab-paclitaxel may promote chemotherapy response and is tolerable. Further evaluation of this combination in tumor types responsive to taxanes is ongoing.
Collapse
Affiliation(s)
- Pamela N. Munster
- Department of Medicine (Hematology/Oncology), University of California San Francisco, San Francisco, California.,Corresponding Author: Pamela N. Munster, University of California, San Francisco, Box 1711, San Francisco, CA 94143. Phone: 415-502-3414; E-mail:
| | | | - Gini F. Fleming
- Department of Medicine, the University of Chicago, Chicago, Illinois
| | | | - Manish R. Sharma
- Department of Medicine, the University of Chicago, Chicago, Illinois
| | | | | | | | | | | | | |
Collapse
|
25
|
Han GH, Yun H, Kim J, Chung JY, Kim JH, Cho H. Overexpression of glucocorticoid receptor promotes the poor progression and induces cisplatin resistance through p38 MAP kinase in cervical cancer patients. Am J Cancer Res 2022; 12:3437-3454. [PMID: 35968326 PMCID: PMC9360232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/20/2022] [Indexed: 06/15/2023] Open
Abstract
Glucocorticoid receptor (GR) is activated by synthetic glucocorticoid or endogenous cortisol which were released by the physical and psychosocial stress, and recent studies reported that it is involved in tumor initiation and metastasis in various solid cancers. However, role of GR in cervical cancer has not been elucidated yet. Therefore, here we aim to unveil the role of GR in cervical cancer with cervical cancer clinical specimen and cervical cancer cell lines. We found that overexpression of GR was associated with poor prognosis in cervical cancer patients. Also, GR knockdown in cervical cancer cell lines showed diminished proliferation, invasion and EMT properties. Besides, we found that GR was positively associated with FoxP3 expression, and combination of GR and FoxP3 overexpression revealed as more reliable biomarker for poor prognosis and poor response to chemotherapy of cervical cancer patient than GR alone. Moreover, FACS-based Annexin-V/PI double staining and cleavage of poly ADP ribose polymerase (PARP) showed that siGR enhanced cisplatin-induced apoptosis, which was mediated by p38 MAP kinase. Collectively, our findings established that the combination of high GR and FoxP3 was associated with cervical cancer progression and platinum resistance, suggesting a potential predictive biomarker for clinical management in patients with cervical cancer.
Collapse
Affiliation(s)
- Gwan Hee Han
- Department of Obstetrics and Gynecology, Kyung Hee University Hospital at GangdongSeoul 05278, Republic of Korea
| | - Hee Yun
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of MedicineSeoul 06299, Republic of Korea
| | - Julie Kim
- Weill Cornell Medical College1300 York Ave, New York, NY 10065, USA
| | - Joon-Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Yonsei University College of MedicineSeoul 06299, Republic of Korea
- Institute of Women’s Life Medical Science, Yonsei University College of MedicineSeoul 03722, Republic of Korea
| | - Hanbyoul Cho
- Department of Obstetrics and Gynecology, Yonsei University College of MedicineSeoul 06299, Republic of Korea
- Institute of Women’s Life Medical Science, Yonsei University College of MedicineSeoul 03722, Republic of Korea
| |
Collapse
|
26
|
Zhong C, Lu Y, Li Y, Xie H, Zhou G, Jia L. Similarities and differences between embryonic implantation and CTC invasion: Exploring the roles of abortifacients in cancer metastasis chemoprevention. Eur J Med Chem 2022; 237:114416. [DOI: 10.1016/j.ejmech.2022.114416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/18/2022] [Accepted: 04/24/2022] [Indexed: 11/03/2022]
|
27
|
Li K, Zong D, Sun J, Chen D, Ma M, Jia L. Rewiring of the Endocrine Network in Triple-Negative Breast Cancer. Front Oncol 2022; 12:830894. [PMID: 35847875 PMCID: PMC9280148 DOI: 10.3389/fonc.2022.830894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/31/2022] [Indexed: 12/19/2022] Open
Abstract
The immunohistochemical definition of estrogen/progesterone receptors dictates endocrine feasibility in the treatment course of breast cancer. Characterized by the deficiency of estrogen receptor α, ERα-negative breast cancers are dissociated from any endocrine regimens in the routine clinical setting, triple-negative breast cancer in particular. However, the stereotype was challenged by triple-negative breast cancers’ retained sensitivity and vulnerability to endocrine agents. The interplay of hormone action and the carcinogenic signaling program previously underscored was gradually recognized along with the increasing investigation. In parallel, the overlooked endocrine-responsiveness in ERα-negative breast cancers attracted attention and supplied fresh insight into the therapeutic strategy in an ERα-independent manner. This review elaborates on the genomic and non-genomic steroid hormone actions and endocrine-related signals in triple-negative breast cancers attached to the hormone insensitivity label. We also shed light on the non-canonical mechanism detected in common hormone agents to showcase their pleiotropic effects.
Collapse
Affiliation(s)
- Kaixuan Li
- Department of Integrated Traditional Chinese and Western Medicine Oncology, China-Japan Friendship Hospital, Beijing, China
- Beijing University of Chinese medicine, Beijing, China
| | | | - Jianrong Sun
- School of Clinical Medicine. Beijing University of Chinese Medicine, Beijing, China
| | - Danxiang Chen
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Minkai Ma
- Department of Integrated Traditional Chinese and Western Medicine Oncology, The Fourth Central Hospital, Baoding, China
| | - Liqun Jia
- Department of Integrated Traditional Chinese and Western Medicine Oncology, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Liqun Jia,
| |
Collapse
|
28
|
De Francesco EM, Cirillo F, Vella V, Belfiore A, Maggiolini M, Lappano R. Triple-negative breast cancer drug resistance, durable efficacy, and cure: How advanced biological insights and emerging drug modalities could transform progress. Expert Opin Ther Targets 2022; 26:513-535. [PMID: 35761781 DOI: 10.1080/14728222.2022.2094762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is a heterogeneous disease characterized by the lack of estrogen receptor (ER), progesterone receptor (PR) and epidermal growth factor receptor 2 (HER2) and often associated with poor survival outcomes. The backbone of current treatments for TNBC relies on chemotherapy; however, resistance to cytotoxic agents is a commonly encountered hurdle to overcome. AREAS COVERED : Current understanding on the mechanisms involved in TNBC chemoresistance is evaluated and novel potential actionable targets and recently explored modalities for carrying and delivering chemotherapeutics are highlighted. EXPERT OPINION : A comprehensive identification of both genomic and functional TNBC signatures is required for a more definite categorization of the patients in order to prevent insensitivity to chemotherapy and therefore realize the full potential of precision-medicine approaches. In this scenario, cell-line-derived xenografts (CDX), patient-derived xenografts (PDX), patient-derived orthotopic xenografts (PDOX) and patient-derived organoids (PDO) are indispensable experimental models for evaluating the efficacy of drug candidates and predicting the therapeutic response. The combination of increasingly sensitive "omics" technologies, computational algorithms and innovative drug modalities may accelerate the successful translation of novel candidate TNBC targets from basic research to clinical settings, thus contributing to reach optimal clinical output, with lower side effects and reduced resistance.
Collapse
Affiliation(s)
- Ernestina Marianna De Francesco
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Veronica Vella
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
29
|
High glucocorticoid receptor expression in the sarcomatous versus carcinomatous elements of Mullerian carcinosarcomas. Gynecol Oncol Rep 2022; 41:100987. [PMID: 35519002 PMCID: PMC9065886 DOI: 10.1016/j.gore.2022.100987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 11/23/2022] Open
|
30
|
Paul U, Banerjee S. The functional significance and cross-talk of non-coding RNAs in triple negative and quadruple negative breast cancer. Mol Biol Rep 2022; 49:6899-6918. [PMID: 35235157 DOI: 10.1007/s11033-022-07288-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/18/2022] [Indexed: 12/13/2022]
Abstract
One of the leading causes of cancer-related deaths worldwide is breast cancer, among which triple-negative breast cancer (TNBC) is the most malignant and lethal subtype. This cancer accounts for 10-20% of all breast cancer deaths. Proliferation, tumorigenesis, and prognosis of TNBC are affected when the androgen receptor (AR) is not expressed, and it is classified as quadruple negative breast cancer (QNBC). Non-coding RNAs, such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), play a significant role in tumorigenesis by virtue of their oncogenic and tumor-suppressive properties. To regulate tumorigenesis, miRNAs interact with their target mRNAs and modulate their expression, whereas lncRNAs can either act alone or interact with miRNAs or other molecules through various signaling pathways. Conversely, circRNAs regulate tumorigenesis by acting as miRNA sponges predominantly. Recently, non-coding RNAs were studied comprehensively for their roles in tumor proliferation, progression, and metastasis. As a result of existing studies and research progress, non-coding RNAs have been implicated in TNBC, necessitating their use as biomarkers for future diagnostic applications. In this review, the non-coding RNAs are explicitly implicated in the regulation of breast cancer, and their cross-talk between TNBC and QNBC is also discussed.
Collapse
Affiliation(s)
- Utpalendu Paul
- School of Bio Science and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Satarupa Banerjee
- School of Bio Science and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
31
|
Rozen EJ, Shohet JM. Systematic review of the receptor tyrosine kinase superfamily in neuroblastoma pathophysiology. Cancer Metastasis Rev 2022; 41:33-52. [PMID: 34716856 PMCID: PMC8924100 DOI: 10.1007/s10555-021-10001-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Neuroblastoma is a devastating disease accounting for 15% of all childhood cancer deaths. Yet, our understanding of key molecular drivers such as receptor tyrosine kinases (RTKs) in this pathology remains poorly clarified. Here, we provide a systematic analysis of the RTK superfamily in the context of neuroblastoma pathogenesis. METHODS Statistical correlations for all RTK family members' expression to neuroblastoma patient survival across 10 independent patient cohorts were annotated, synthesized, and ranked using the R2: Genomics Analysis and Visualization Platform. Gene expression of selected members across different cancer cell lines was further analyzed in the Cancer Cell Line Encyclopedia, part of the Cancer Dependency Map portal (depmap portal ( http://depmap.org )). Finally, we provide a detailed literature review for highly ranked candidates. RESULTS Our analysis defined two subsets of RTKs showing robust associations with either better or worse survival, constituting potential novel players in neuroblastoma pathophysiology, diagnosis, and therapy. We review the available literature regarding the oncogenic functions of these RTKs, their roles in neuroblastoma pathophysiology, and potential utility as therapeutic targets. CONCLUSIONS Our systematic analysis and review of the RTK superfamily in neuroblastoma pathogenesis provides a new resource to guide the research community towards focused efforts investigating signaling pathways that contribute to neuroblastoma tumor establishment, growth, and/or aggressiveness and targeting these druggable molecules in novel therapeutic strategies.
Collapse
Affiliation(s)
- Esteban Javier Rozen
- Department of Pediatrics, UMass Chan Medical School, Lazare Research Building LRB603, 364 Plantation Street, Worcester, MA, 01605, USA.
| | - Jason Matthew Shohet
- Division of Hematology/Oncology, Department of Pediatrics, UMass Chan Medical School, Lazare Research Building LRB603, 364 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
32
|
Lee J, Troike K, Fodor R, Lathia JD. Unexplored Functions of Sex Hormones in Glioblastoma Cancer Stem Cells. Endocrinology 2022; 163:bqac002. [PMID: 35023543 PMCID: PMC8807164 DOI: 10.1210/endocr/bqac002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Indexed: 01/14/2023]
Abstract
Biological sex impacts a wide array of molecular and cellular functions that impact organismal development and can influence disease trajectory in a variety of pathophysiological states. In nonreproductive cancers, epidemiological sex differences have been observed in a series of tumors, and recent work has identified previously unappreciated sex differences in molecular genetics and immune response. However, the extent of these sex differences in terms of drivers of tumor growth and therapeutic response is less clear. In glioblastoma (GBM), the most common primary malignant brain tumor, there is a male bias in incidence and outcome, and key genetic and epigenetic differences, as well as differences in immune response driven by immune-suppressive myeloid populations, have recently been revealed. GBM is a prototypic tumor in which cellular heterogeneity is driven by populations of therapeutically resistant cancer stem cells (CSCs) that underlie tumor growth and recurrence. There is emerging evidence that GBM CSCs may show a sex difference, with male tumor cells showing enhanced self-renewal, but how sex differences impact CSC function is not clear. In this mini-review, we focus on how sex hormones may impact CSCs in GBM and implications for other cancers with a pronounced CSC population. We also explore opportunities to leverage new models to better understand the contribution of sex hormones vs sex chromosomes to CSC function. With the rising interest in sex differences in cancer, there is an immediate need to understand the extent to which sex differences impact tumor growth, including effects on CSC function.
Collapse
Affiliation(s)
- Juyeun Lee
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic
| | - Katie Troike
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University
| | - R’ay Fodor
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University
| | - Justin D Lathia
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic
- Case Comprehensive Cancer Center
| |
Collapse
|
33
|
Aspros KGM, Carter JM, Hoskin TL, Suman VJ, Subramaniam M, Emch MJ, Ye Z, Sun Z, Sinnwell JP, Thompson KJ, Tang X, Rodman EPB, Wang X, Nelson AW, Chernukhin I, Hamdan FH, Bruinsma ES, Carroll JS, Fernandez-Zapico ME, Johnsen SA, Kalari KR, Huang H, Leon-Ferre RA, Couch FJ, Ingle JN, Goetz MP, Hawse JR. Estrogen receptor beta repurposes EZH2 to suppress oncogenic NFκB/p65 signaling in triple negative breast cancer. NPJ Breast Cancer 2022; 8:20. [PMID: 35177654 PMCID: PMC8854734 DOI: 10.1038/s41523-022-00387-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 01/21/2022] [Indexed: 12/14/2022] Open
Abstract
Triple Negative Breast Cancer (TNBC) accounts for 15-20% of all breast cancer cases, yet is responsible for a disproportionately high percentage of breast cancer mortalities. Thus, there is an urgent need to identify novel biomarkers and therapeutic targets based on the molecular events driving TNBC pathobiology. Estrogen receptor beta (ERβ) is known to elicit anti-cancer effects in TNBC, however its mechanisms of action remain elusive. Here, we report the expression profiles of ERβ and its association with clinicopathological features and patient outcomes in the largest cohort of TNBC to date. In this cohort, ERβ was expressed in approximately 18% of TNBCs, and expression of ERβ was associated with favorable clinicopathological features, but correlated with different overall survival outcomes according to menopausal status. Mechanistically, ERβ formed a co-repressor complex involving enhancer of zeste homologue 2/polycomb repressive complex 2 (EZH2/PRC2) that functioned to suppress oncogenic NFκB/RELA (p65) activity. Importantly, p65 was shown to be required for formation of this complex and for ERβ-mediated suppression of TNBC. Our findings indicate that ERβ+ tumors exhibit different characteristics compared to ERβ- tumors and demonstrate that ERβ functions as a molecular switch for EZH2, repurposing it for tumor suppressive activities and repression of oncogenic p65 signaling.
Collapse
Affiliation(s)
- Kirsten G M Aspros
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Tanya L Hoskin
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Vera J Suman
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Malayannan Subramaniam
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Michael J Emch
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Zhenqing Ye
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Zhifu Sun
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jason P Sinnwell
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kevin J Thompson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xiaojia Tang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Esther P B Rodman
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xiyin Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Adam W Nelson
- Cancer Research UK Cambridge Research Institute, University of Cambridge, Cambridge, UK
| | - Igor Chernukhin
- Cancer Research UK Cambridge Research Institute, University of Cambridge, Cambridge, UK
| | - Feda H Hamdan
- Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Elizabeth S Bruinsma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jason S Carroll
- Cancer Research UK Cambridge Research Institute, University of Cambridge, Cambridge, UK
| | - Martin E Fernandez-Zapico
- Shulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Steven A Johnsen
- Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Krishna R Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Urology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Fergus J Couch
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Matthew P Goetz
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
34
|
Filipe A, Katopodis P, Chudasama D, Kerslake R, Jeyaneethi J, Anikin V, Silva E, Kyrou I, Randeva HS, Sisu C, Hall M, Karteris E. Differential Expression of RAD51AP1 in Ovarian Cancer: Effects of siRNA In Vitro. J Pers Med 2022; 12:jpm12020201. [PMID: 35207688 PMCID: PMC8876735 DOI: 10.3390/jpm12020201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/08/2022] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
Background: DNA double strand breaks can affect genome integrity potentially leading to cancer. RAD51-associated protein 1 (RAD51AP1), an accessory protein to RAD51, is critical for homologous recombination, a key DNA damage response pathway. Emerging studies indicate a novel role for RAD51AP1 in carcinogenesis. Here we provide additional insight into the role of RAD51AP1 in ovarian cancer (OvCa). Methods: Gene expression and patient phenotype data were obtained from TCGA and GTEX project consortia for bioinformatics analysis. Immunohistochemistry of OvCa tissue microarray was undertaken. Functional analyses were performed in a SKOV3 OvCa cell line with down-regulation of RAD51AP1 using siRNA. Results: RAD51AP1 is overexpressed at gene level in primary and recurrent OvCa compared to controls. At protein level, RAD51AP1 was up-regulated in low grade serous tumors compared to high grade OvCa. There was higher expression of RAD51AP1 in OvCa metastatic to lymph nodes compared to primary cancer samples. Gene enrichment analyses identified 12 differentially expressed genes (DEGs) related to OvCa, eight of which are also common in tissue from patients with type 2 diabetes mellitus (T2DM). Conclusions: RAD51AP1 is overexpressed in OvCa, Given the link between OvCa and T2DM, the eight-gene signature shows potential for predictive value.
Collapse
Affiliation(s)
- Alice Filipe
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
| | - Periklis Katopodis
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield UB9 6JH, UK;
| | - Dimple Chudasama
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
| | - Rachel Kerslake
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield UB9 6JH, UK;
| | - Jeyarooban Jeyaneethi
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield UB9 6JH, UK;
| | - Vladimir Anikin
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield UB9 6JH, UK;
- Department of Oncology and Reconstructive Surgery, Sechenov First Moscow State, Medical University, 119146 Moscow, Russia
| | - Elisabete Silva
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (I.K.); (H.S.R.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry CV1 5FB, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- Department of Food Science & Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (I.K.); (H.S.R.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Cristina Sisu
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
| | - Marcia Hall
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
- Mount Vernon Cancer Centre, Northwood, London HA6 2RN, UK
- Correspondence: (M.H.); (E.K.)
| | - Emmanouil Karteris
- Department of Life Sciences, Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (A.F.); (P.K.); (D.C.); (R.K.); (J.J.); (E.S.); (C.S.)
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, Harefield UB9 6JH, UK;
- Correspondence: (M.H.); (E.K.)
| |
Collapse
|
35
|
Manjili MH, Khazaie K. Pattern recognition of tumor dormancy and relapse beyond cell-intrinsic and cell-extrinsic pathways. Semin Cancer Biol 2022; 78:1-4. [PMID: 34990835 DOI: 10.1016/j.semcancer.2021.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this thematic issue, several mechanisms of tumor dormancy and relapse are discussed. The reviews suggest mutual interactions and communications between malignant cells and other cells in their niche during tumor dormancy. Nevertheless, a complete understanding of tumor dormancy remains elusive. This is because we are getting lost in details of cell-intrinsic and cell-extrinsic molecular pathways without being able to discover the pattern of tumor dormancy. Here, we discuss some conceptual frameworks and methodological approaches that facilitate pattern recognition of tumor dormancy, and propose that settling on certain biological scale such as mitochondria would be the key to discover the pattern of tumor dormancy and relapse.
Collapse
Affiliation(s)
- Masoud H Manjili
- Department of Microbiology & Immunology, VCU School of Medicine, Massey Cancer Center, 401 College Street, Box 980035, Richmond, VA, 23298, United States.
| | | |
Collapse
|
36
|
Butz H, Patócs A. Mechanisms behind context-dependent role of glucocorticoids in breast cancer progression. Cancer Metastasis Rev 2022; 41:803-832. [PMID: 35761157 PMCID: PMC9758252 DOI: 10.1007/s10555-022-10047-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/09/2022] [Indexed: 02/08/2023]
Abstract
Glucocorticoids (GCs), mostly dexamethasone (dex), are routinely administered as adjuvant therapy to manage side effects in breast cancer. However, recently, it has been revealed that dex triggers different effects and correlates with opposite outcomes depending on the breast cancer molecular subtype. This has raised new concerns regarding the generalized use of GC and suggested that the context-dependent effects of GCs can be taken into potential consideration during treatment design. Based on this, attention has recently been drawn to the role of the glucocorticoid receptor (GR) in development and progression of breast cancer. Therefore, in this comprehensive review, we aimed to summarize the different mechanisms behind different context-dependent GC actions in breast cancer by applying a multilevel examination, starting from the association of variants of the GR-encoding gene to expression at the mRNA and protein level of the receptor, and its interactions with other factors influencing GC action in breast cancer. The role of GCs in chemosensitivity and chemoresistance observed during breast cancer therapy is discussed. In addition, experiences using GC targeting therapeutic options (already used and investigated in preclinical and clinical trials), such as classic GC dexamethasone, selective glucocorticoid receptor agonists and modulators, the GC antagonist mifepristone, and GR coregulators, are also summarized. Evidence presented can aid a better understanding of the biology of context-dependent GC action that can lead to further advances in the personalized therapy of breast cancer by the evaluation of GR along with the conventional estrogen receptor (ER) and progesterone receptor (PR) in the routine diagnostic procedure.
Collapse
Affiliation(s)
- Henriett Butz
- Department of Molecular Genetics and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary.
- Hereditary Tumours Research Group, Hungarian Academy of Sciences-Semmelweis University, Budapest, Hungary.
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary.
| | - Attila Patócs
- Department of Molecular Genetics and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
- Hereditary Tumours Research Group, Hungarian Academy of Sciences-Semmelweis University, Budapest, Hungary
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
37
|
Zhao CC, Zhang CG, Sun X, Guo Q, Liu J, Liu Y, Hao YN, Feng G, Yang L, Liu H, Liu J. Paclitaxel-based supramolecular hydrogel loaded with mifepristone for the inhibition of breast cancer metastasis. Cancer Sci 2021; 113:733-743. [PMID: 34859546 PMCID: PMC8819302 DOI: 10.1111/cas.15230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 12/16/2022] Open
Abstract
Breast cancer is the leading cause of cancer death among women and almost all of the breast cancer-caused mortality is related to metastasis. It has been reported that glucocorticoid facilitates the metastasis of breast cancer in mice, and mifepristone can antagonize the effect of glucocorticoid. Paclitaxel is one of the important drugs in the treatment of breast cancer. Mifepristone combined with paclitaxel could be an effective strategy for inhibiting breast cancer metastasis. However, their inherent defects, in terms of short blood circulation half-life and lack of tumor targeting, not only limit their effectiveness but also cause adverse reactions. Therefore, our aim is to explore a novel protocol against breast cancer metastasis, further optimize its therapeutic efficacy by a nanodelivery system, and explore its mechanism. Herein, a paclitaxel-conjugated and mifepristone-loaded hydrogel (PM-nano) was prepared by self-assembly. Its characterizations were studied. The antimetastatic effect was evaluated in vitro and in vivo and its mechanism was also explored by western blot assay. The resultant PM-nano was developed with favorable water solubility and good biocompatibility. Moreover, PM-nano displayed increased cell uptake properties and stimulated drug release in the tumor micro-acidic environment. The PM-nano was more effective in inhibiting the proliferation and metastasis of breast cancer than other groups in vitro and in vivo. The PM-nano might inhibit metastasis through glucocorticoid receptor/receptor tyrosine kinase-like orphan receptor 1 and MMPs. Taken together, PM-nano showed superior antimetastatic effects against breast cancer and excellent biocompatibility in vitro and in vivo, providing a new option for limiting metastasis.
Collapse
Affiliation(s)
- Cui-Cui Zhao
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chuan-Gui Zhang
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xuan Sun
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qingxiang Guo
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jinjian Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yan Liu
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ya-Nan Hao
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Guowei Feng
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lijun Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hong Liu
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
38
|
Eckerling A, Ricon-Becker I, Sorski L, Sandbank E, Ben-Eliyahu S. Stress and cancer: mechanisms, significance and future directions. Nat Rev Cancer 2021; 21:767-785. [PMID: 34508247 DOI: 10.1038/s41568-021-00395-5] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/23/2021] [Indexed: 02/08/2023]
Abstract
The notion that stress and cancer are interlinked has dominated lay discourse for decades. More recent animal studies indicate that stress can substantially facilitate cancer progression through modulating most hallmarks of cancer, and molecular and systemic mechanisms mediating these effects have been elucidated. However, available clinical evidence for such deleterious effects is inconsistent, as epidemiological and stress-reducing clinical interventions have yielded mixed effects on cancer mortality. In this Review, we describe and discuss specific mediating mechanisms identified by preclinical research, and parallel clinical findings. We explain the discrepancy between preclinical and clinical outcomes, through pointing to experimental strengths leveraged by animal studies and through discussing methodological and conceptual obstacles that prevent clinical studies from reflecting the impacts of stress. We suggest approaches to circumvent such obstacles, based on targeting critical phases of cancer progression that are more likely to be stress-sensitive; pharmacologically limiting adrenergic-inflammatory responses triggered by medical procedures; and focusing on more vulnerable populations, employing personalized pharmacological and psychosocial approaches. Recent clinical trials support our hypothesis that psychological and/or pharmacological inhibition of excess adrenergic and/or inflammatory stress signalling, especially alongside cancer treatments, could save lives.
Collapse
Affiliation(s)
- Anabel Eckerling
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Itay Ricon-Becker
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Liat Sorski
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Elad Sandbank
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shamgar Ben-Eliyahu
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
39
|
Kumar S, Freelander A, Lim E. Type 1 Nuclear Receptor Activity in Breast Cancer: Translating Preclinical Insights to the Clinic. Cancers (Basel) 2021; 13:4972. [PMID: 34638457 PMCID: PMC8507977 DOI: 10.3390/cancers13194972] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/30/2022] Open
Abstract
The nuclear receptor (NR) family of transcription factors is intimately associated with the development, progression and treatment of breast cancer. They are used diagnostically and prognostically, and crosstalk between nuclear receptor pathways and growth factor signalling has been demonstrated in all major subtypes of breast cancer. The majority of breast cancers are driven by estrogen receptor α (ER), and anti-estrogenic therapies remain the backbone of treatment, leading to clinically impactful improvements in patient outcomes. This serves as a blueprint for the development of therapies targeting other nuclear receptors. More recently, pivotal findings into modulating the progesterone (PR) and androgen receptors (AR), with accompanying mechanistic insights into NR crosstalk and interactions with other proliferative pathways, have led to clinical trials in all of the major breast cancer subtypes. A growing body of evidence now supports targeting other Type 1 nuclear receptors such as the glucocorticoid receptor (GR), as well as Type 2 NRs such as the vitamin D receptor (VDR). Here, we reviewed the existing preclinical insights into nuclear receptor activity in breast cancer, with a focus on Type 1 NRs. We also discussed the potential to translate these findings into improving patient outcomes.
Collapse
Affiliation(s)
- Sanjeev Kumar
- Faculty of Medicine, St Vincent’s Clinical School, University of New South Wales, Darlinghurst 2010, Australia; (A.F.); (E.L.)
- Garvan Institute of Medical Research, University of New South Wales, Darlinghurst 2010, Australia
| | - Allegra Freelander
- Faculty of Medicine, St Vincent’s Clinical School, University of New South Wales, Darlinghurst 2010, Australia; (A.F.); (E.L.)
- Garvan Institute of Medical Research, University of New South Wales, Darlinghurst 2010, Australia
| | - Elgene Lim
- Faculty of Medicine, St Vincent’s Clinical School, University of New South Wales, Darlinghurst 2010, Australia; (A.F.); (E.L.)
- Garvan Institute of Medical Research, University of New South Wales, Darlinghurst 2010, Australia
| |
Collapse
|
40
|
Kowalczyk W, Waliszczak G, Jach R, Dulińska-Litewka J. Steroid Receptors in Breast Cancer: Understanding of Molecular Function as a Basis for Effective Therapy Development. Cancers (Basel) 2021; 13:4779. [PMID: 34638264 PMCID: PMC8507808 DOI: 10.3390/cancers13194779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/18/2021] [Accepted: 09/20/2021] [Indexed: 12/21/2022] Open
Abstract
Breast cancer remains one of the most important health problems worldwide. The family of steroid receptors (SRs), which comprise estrogen (ER), progesterone (PR), androgen (AR), glucocorticoid (GR) and mineralocorticoid (MR) receptors, along with a receptor for a secosteroid-vitamin D, play a crucial role in the pathogenesis of the disease. They function predominantly as nuclear receptors to regulate gene expression, however, their full spectrum of action reaches far beyond this basic mechanism. SRs are involved in a vast variety of interactions with other proteins, including extensive crosstalk with each other. How they affect the biology of a breast cell depends on such factors as post-translational modifications, expression of coregulators, or which SR isoform is predominantly synthesized in a given cellular context. Although ER has been successfully utilized as a breast cancer therapy target for years, research on therapeutic application of other SRs is still ongoing. Designing effective hormone therapies requires thorough understanding of the molecular function of the SRs. Over the past decades, huge amount of data was obtained in multiple studies exploring this field, therefore in this review we attempt to summarize the current knowledge in a comprehensive way.
Collapse
Affiliation(s)
- Wojciech Kowalczyk
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Kraków, Poland; (W.K.); (G.W.)
| | - Grzegorz Waliszczak
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Kraków, Poland; (W.K.); (G.W.)
| | - Robert Jach
- Department of Gynecology and Obstetrics, Jagiellonian University Medical College, 23 Kopernika St., 31-501 Kraków, Poland;
| | - Joanna Dulińska-Litewka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Kraków, Poland; (W.K.); (G.W.)
| |
Collapse
|
41
|
Kamaraju S, Fowler AM, Weil E, Wisinski KB, Truong TH, Lehr M, Chaudhary LN, Cheng YC, Chitambar CR, Rui H, Yee D, Lange C. Leveraging Antiprogestins in the Treatment of Metastatic Breast Cancer. Endocrinology 2021; 162:6178343. [PMID: 33735382 DOI: 10.1210/endocr/bqab060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Indexed: 12/20/2022]
Abstract
Although incurable, the prognosis for patients with metastatic breast cancer (MBC) has considerably improved with the approvals of multiple targeted and cytotoxic therapies. For hormone receptor-positive (HR+), ie, estrogen receptor and progesterone receptor positive (ER+/PgR+) and human epidermal growth factor receptor-2 negative (ie, ERBB2 gene nonamplified or HER2-) MBC, current approved treatment options include palliative endocrine therapy (ET), cyclin-dependent kinase (CDK 4/6) inhibitors, mTOR inhibitors, and PI3 kinase inhibitors. Most treatments target ER+ disease regardless of PgR status. Although the presence of PgR is crucial for ER+ cell proliferation in both normal and malignant mammary tissue, currently, there are no approved treatments that specifically target PgR. Recent literature has demonstrated the potential of antiprogestins in the treatment of MBC both in preclinical and clinical studies. Antiprogestins, including selective PgR modulators (SPRMs) that act as PgR antagonists, are a promising class of therapeutics for overcoming endocrine resistance in patients who develop activating estrogen receptor 1 (ESR1) and phosphatidylinositol 3-kinase (PI3K) gene mutations after prior endocrine therapy. Herein, we summarize the role of PgR and antiprogestins in the treatment of MBC. Other aspects on the use of functional imaging, clinical trials incorporating novel antiprogestins, and potential treatment combinations to overcome endocrine resistance will be briefly discussed.
Collapse
Affiliation(s)
- Sailaja Kamaraju
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amy M Fowler
- Division of Hematology-Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Division of Hematology-Oncology Medical College of Wisconsin, Cancer Center, 4th Fl Administrative Offices, Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Elizabeth Weil
- Froedtert Health, Cancer Center, Milwaukee, WI 53226, USA
| | - Kari B Wisinski
- Division of Hematology-Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Division of Hematology-Oncology Medical College of Wisconsin, Cancer Center, 4th Fl Administrative Offices, Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Thu H Truong
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Martin Lehr
- Context Therapeutics, Philadelphia, PA 19104, USA
| | - Lubna N Chaudhary
- Division of Hematology-Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Division of Hematology-Oncology Medical College of Wisconsin, Cancer Center, 4th Fl Administrative Offices, Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Yee Chung Cheng
- Division of Hematology-Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Division of Hematology-Oncology Medical College of Wisconsin, Cancer Center, 4th Fl Administrative Offices, Watertown Plank Road, Milwaukee, WI 53226, USA
| | | | - Hallgeir Rui
- Pathology and Laboratory Medicine, Medical College of Wisconsin , Milwaukee, WI 53226, USA
| | - Douglas Yee
- Division of Hematology-Oncology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Carol Lange
- Division of Hematology-Oncology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
42
|
Zhang ZQ, Wang X, Xue BH, Zhao Y, Xie F, Wang SD, Xue C, Wang Y, Zhang YS, Qian LJ. Chronic stress promotes glioma cell proliferation via the PI3K/Akt signaling pathway. Oncol Rep 2021; 46:202. [PMID: 34296295 PMCID: PMC8329913 DOI: 10.3892/or.2021.8153] [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] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/10/2021] [Indexed: 01/05/2023] Open
Abstract
High malignancy and high mortality of glioma render it urgent to elucidate the underlying mechanisms of glioma carcinogenesis and explore novel targets for therapy. Epidemiologic and clinical studies have revealed that chronic stress promotes the progression of various solid tumors and is correlated with poor prognosis; however, findings reporting the involvement of chronic stress in glioma are rare. In the present study, a chronic restraint animal model and a chronic stress cell model were established to explore the effects of chronic stress on glioma and its molecular mechanisms. The results revealed that chronic stress promoted glioma growth in vivo, and the serum levels of the stress hormones glucocorticoid (GC) and noradrenaline (NE) were significantly increased. In addition, GC and NE were verified to accelerate the proliferation of glioma cells in vitro. Mechanistically, the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway was revealed to be activated under stress conditions, and inhibition of the expression of p-Akt could restrain the stress hormone-induced glioma cell proliferation. In addition, our data indicated that the GC receptor (GR) and β-adrenergic receptors (ADRBs) were both required for the biological functions of GC and NE in glioma cells. In conclusion, these results indicated that chronic stress and the stress hormones GC and NE activated PI3K/Akt signaling through binding to GR and ADRBs, thereby promoting glioma cell growth. Our findings may provide potential therapeutic targets and pave the way for the development of new strategies to protect patients with glioma from the detrimental effects of stress on tumor progression.
Collapse
Affiliation(s)
- Zi-Qian Zhang
- Laboratory Animal Center, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Xue Wang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medicine Sciences, Beijing 100850, P.R. China
| | - Bing-Hua Xue
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medicine Sciences, Beijing 100850, P.R. China
| | - Yun Zhao
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medicine Sciences, Beijing 100850, P.R. China
| | - Fang Xie
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medicine Sciences, Beijing 100850, P.R. China
| | - Shi-Da Wang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medicine Sciences, Beijing 100850, P.R. China
| | - Cong Xue
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medicine Sciences, Beijing 100850, P.R. China
| | - Ying Wang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medicine Sciences, Beijing 100850, P.R. China
| | - Yan-Shu Zhang
- Laboratory Animal Center, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Ling-Jia Qian
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medicine Sciences, Beijing 100850, P.R. China
| |
Collapse
|
43
|
Greenstein AE, Hunt HJ. Glucocorticoid receptor antagonism promotes apoptosis in solid tumor cells. Oncotarget 2021; 12:1243-1255. [PMID: 34194622 PMCID: PMC8238250 DOI: 10.18632/oncotarget.27989] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/02/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Resistance to antiproliferative chemotherapies remains a significant challenge in the care of patients with solid tumors. Glucocorticoids, including endogenous cortisol, have been shown to induce pro-survival pathways in epithelial tumor cells. While pro-apoptotic effects of glucocorticoid receptor (GR) antagonism have been demonstrated under select conditions, the breadth and nature of these effects have not been fully established. Materials and Methods: To guide studies in cancer patients, relacorilant, an investigational selective GR modulator (SGRM) that antagonizes cortisol activity, was assessed in various tumor types, with multiple cytotoxic combination partners, and in the presence of physiological cortisol concentrations. Results: In the MIA PaCa-2 cell line, paclitaxel-driven apoptosis was blunted by cortisol and restored by relacorilant. In the OVCAR5 cell line, relacorilant improved the efficacy of paclitaxel and the potency of platinum agents. A screen to identify optimal combination partners for relacorilant showed that microtubule-targeted agents consistently benefited from combination with relacorilant. These findings were confirmed in xenograft models, including MIA PaCa-2, HeLa, and a cholangiocarcinoma patient-derived xenograft. In vivo, tumor-cell apoptosis was increased when relacorilant was added to paclitaxel in multiple models. Conclusions: These observations support recently reported findings of clinical benefit when relacorilant is added to paclitaxel-containing therapy in patients with ovarian and pancreatic cancers and provide a new rationale for combining relacorilant with additional cytotoxic agents.
Collapse
|
44
|
Noureddine LM, Trédan O, Hussein N, Badran B, Le Romancer M, Poulard C. Glucocorticoid Receptor: A Multifaceted Actor in Breast Cancer. Int J Mol Sci 2021; 22:ijms22094446. [PMID: 33923160 PMCID: PMC8123001 DOI: 10.3390/ijms22094446] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) is one of the most common cancers in women worldwide. Even though the role of estrogen receptor alpha (ERα) is extensively documented in the development of breast tumors, other members of the nuclear receptor family have emerged as important players. Synthetic glucocorticoids (GCs) such as dexamethasone (dex) are commonly used in BC for their antiemetic, anti-inflammatory, as well as energy and appetite stimulating properties, and to manage the side effects of chemotherapy. However, dex triggers different effects depending on the BC subtype. The glucocorticoid receptor (GR) is also an important marker in BC, as high GR expression is correlated with a poor and good prognosis in ERα-negative and ERα-positive BCs, respectively. Indeed, though it drives the expression of pro-tumorigenic genes in ERα-negative BCs and is involved in resistance to chemotherapy and metastasis formation, dex inhibits estrogen-mediated cell proliferation in ERα-positive BCs. Recently, a new natural ligand for GR called OCDO was identified. OCDO is a cholesterol metabolite with oncogenic properties, triggering mammary cell proliferation in vitro and in vivo. In this review, we summarize recent data on GR signaling and its involvement in tumoral breast tissue, via its different ligands.
Collapse
Affiliation(s)
- Lara Malik Noureddine
- Université de Lyon, F-69000 Lyon, France; (L.M.N.); (O.T.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences, Lebanese University, Hadat-Beirut 90656, Lebanon; (N.H.); (B.B.)
| | - Olivier Trédan
- Université de Lyon, F-69000 Lyon, France; (L.M.N.); (O.T.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- Centre Leon Bérard, Oncology Department, F-69000 Lyon, France
| | - Nader Hussein
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences, Lebanese University, Hadat-Beirut 90656, Lebanon; (N.H.); (B.B.)
| | - Bassam Badran
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences, Lebanese University, Hadat-Beirut 90656, Lebanon; (N.H.); (B.B.)
| | - Muriel Le Romancer
- Université de Lyon, F-69000 Lyon, France; (L.M.N.); (O.T.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Coralie Poulard
- Université de Lyon, F-69000 Lyon, France; (L.M.N.); (O.T.); (M.L.R.)
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
- Correspondence: ; Tel.: +33-478-786-663; Fax: +33-478-782-720
| |
Collapse
|
45
|
Wang H, Jiang Y. SRp20: A potential therapeutic target for human tumors. Pathol Res Pract 2021; 224:153444. [PMID: 34126370 DOI: 10.1016/j.prp.2021.153444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/11/2021] [Accepted: 04/11/2021] [Indexed: 12/12/2022]
Abstract
As an important member of SR protein family, SRp20 plays a crucial role in alternative splicing. It not only participates in cell cycle regulation, export of mRNA, cleaving of primary microRNAs, homologous recombination-mediated DNA repair, cellular senescence and apoptosis, but also gets involved in the integrity and pluripotency of genome. Alternative splicing maintains a strict balance in the body to ensure the normal physiological function of cells. Once the balance is broken, diseases, even tumors, will follow. Through the analysis of SRp20-related articles, we found that Alzheimer's disease, glaucoma, bipolar disorder and other diseases have a certain relationship with SRp20. More importantly, SRp20 is closely related to the occurrence, proliferation, invasion and metastasis of various tumors, as well as chemotherapy resistance. Some SRp20 inhibitors have shown significant anticancer efficacy, suggesting a potential therapeutic strategy for tumors.
Collapse
Affiliation(s)
- Han Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yanxia Jiang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
46
|
Hegde M, Joshi MB. Comprehensive analysis of regulation of DNA methyltransferase isoforms in human breast tumors. J Cancer Res Clin Oncol 2021; 147:937-971. [PMID: 33604794 PMCID: PMC7954751 DOI: 10.1007/s00432-021-03519-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/10/2021] [Indexed: 12/14/2022]
Abstract
Significant reprogramming of epigenome is widely described during pathogenesis of breast cancer. Transformation of normal cell to hyperplastic cell and to neoplastic phenotype is associated with aberrant DNA (de)methylation, which, through promoter and enhancer methylation changes, activates oncogenes and silence tumor suppressor genes in variety of tumors including breast. DNA methylation, one of the major epigenetic mechanisms is catalyzed by evolutionarily conserved isoforms namely, DNMT1, DNMT3A and DNMT3B in humans. Over the years, studies have demonstrated intricate and complex regulation of DNMT isoforms at transcriptional, translational and post-translational levels. The recent findings of allosteric regulation of DNMT isoforms and regulation by other interacting chromatin modifying proteins emphasizes functional integrity and their contribution for the development of breast cancer and progression. DNMT isoforms are regulated by several intrinsic and extrinsic parameters. In the present review, we have extensively performed bioinformatics analysis of expression of DNMT isoforms along with their transcriptional and post-transcriptional regulators such as transcription factors, interacting proteins, hormones, cytokines and dietary elements along with their significance during pathogenesis of breast tumors. Our review manuscript provides a comprehensive understanding of key factors regulating DNMT isoforms in breast tumor pathology and documents unsolved issues.
Collapse
Affiliation(s)
- Mangala Hegde
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Planetarium Complex, Manipal, 576104, India
| | - Manjunath B Joshi
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Planetarium Complex, Manipal, 576104, India.
| |
Collapse
|
47
|
Prognostic Significance of Glucocorticoid Receptor Expression in Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2021; 13:cancers13071649. [PMID: 33916028 PMCID: PMC8037088 DOI: 10.3390/cancers13071649] [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] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/17/2021] [Accepted: 03/24/2021] [Indexed: 01/30/2023] Open
Abstract
Simple Summary In solid tumours, emerging evidence indicates that signalling through the glucocorticoid receptor (GR) can encourage the growth and spread of tumours and so drugs targeting this receptor are in development for use in cancer treatment. For these reasons, GR may be useful in anticipating a patient’s outcome upon their cancer diagnosis or to predict their tumours response to drugs targeting this receptor. In this review we aim to ascertain whether GR expression in tumours affects cancer patient survival. Overall, GR expression did not affect patient survival when assessing all cancer types. However, we found that in certain cancer subtypes such as gynaecological cancers (endometrial and ovarian) and early stage, untreated triple negative breast cancers, high GR expression is linked with cancer progression and therefore a poorer patient prognosis. Further studies are needed to uncover the exact role of GR in specific tumour (sub)types in order to provide the correct patients with GR targeting therapies. Abstract In solid malignancies, the glucocorticoid receptor (GR) signalling axis is associated with tumour progression and GR antagonists are in clinical development. Therefore, GR expression may be a useful potential prognostic or predictive biomarker for GR antagonist therapy in cancer. The aim of this review is to investigate if GR expression in tumours is predictive of overall survival or progression free survival. Twenty-five studies were identified through systematic searches of three databases and a meta-analysis conducted using a random effects model, quantifying statistical heterogeneity. Subgroup analysis was conducted for cancer types and publication bias was assessed via funnel plots. There was high heterogeneity in meta-analysis of the studies in all cancer types, which found no association between high GR expression with overall survival (pooled unadjusted HR 1.16, 95% CI (0.89–1.50), n = 2814; pooled adjusted HR 1.02, 95% CI (0.77–1.37), n = 2355) or progression-free survival (pooled unadjusted HR 1.12, 95% CI (0.88–1.42), n = 3365; pooled adjusted HR 1.04, 95% CI (0.6–1.81), n = 582) across all cancer types. However, subgroup meta-analyses showed that high GR expression in gynaecological cancers (endometrial and ovarian) (unadjusted HR 1.83, 95% CI (1.31–2.56), n = 664) and early stage, untreated triple negative breast cancers (TNBCs) (unadjusted HR 1.73, 95% CI (1.35–2.23), n = 687) is associated with disease progression. GR expression in late stage, chemotherapy treated TNBC was not prognostic (unadjusted HR 0.76, 95% CI (0.44, 1.32), n = 287). In conclusion, high GR expression is associated with an increased risk of disease progression in gynaecological and early stage, untreated TNBC. Additional studies are required to elucidate the tumour specific function of the GR receptor in order to ensure GR antagonists target the correct patient groups.
Collapse
|
48
|
Li Z, Yin H, Shen Y, Ren M, Xu X. The influence of phenolic environmental estrogen on the transcriptome of uterine leiomyoma cells: A whole transcriptome profiling-based analysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 211:111945. [PMID: 33516137 DOI: 10.1016/j.ecoenv.2021.111945] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 01/09/2021] [Accepted: 01/13/2021] [Indexed: 06/12/2023]
Abstract
OBJECTIVE The study aimed to recognize potential molecular targets and signal pathways whereby phenolic environmental estrogen promotes the proliferation of uterine leiomyoma cells. METHODS Primary cultured cell lines of uterine leiomyoma were treated with 0.1% DMSO, 10.0μmol/L Bisphenol A (BPA), and 32.0μmol/L Nonylphenol (NP) for 48 h before RNA-seq was performed. Those genes affected by BPA and NP were identified. Then, Gene Ontology (GO) enrichment, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, and Protein-protein Interaction (PPI) analysis were performed. Quantitative real-time polymerase chain reaction (q-PCR) and western blot were used to verify the differentially expressed gene and protein. RESULTS Compared to with the control group, 739 differentially expressed genes were identified in both the BPA group and the NP group. GO enrichment analysis showed that the most enriched GO terms were connective tissue development and G1/S transition of mitotic cell cycle, and extracellular matrix. The results of KEGG enrichment analysis showed that differentially expressed mRNA were enriched mainly in three primary pathways, including environmental information processing, human diseases, and cellular processes. The cell cycle, PI3K-Akt signaling pathway are significantly enriched. The q-PCR and western blot verified the cell cycle associated genes and proteins were upregulated in both BPA group and NP group. Both BPA and NP activated the PI3K-AKT signaling pathway. CONCLUSION Phenolic environmental estrogens may promote the proliferation and cell cycle progression of uterine leiomyoma cells through rapid non-genomic ER signaling, which leads to disordered cell cycle regulation and accelerates the transition of the cell cycle from G0/G1 phase to S phase. In addition, as an external stimulant, phenolic estrogen promotes the upregulation of inflammatory factors in uterine leiomyomas.
Collapse
Affiliation(s)
- Zemin Li
- Department of Obstetrics and Gynaecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Han Yin
- Department of Obstetrics and Gynaecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Yang Shen
- Department of Obstetrics and Gynaecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Mulan Ren
- Department of Obstetrics and Gynaecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Xiaolan Xu
- Xinghua City People's Hospital, Xinghua 225700, China.
| |
Collapse
|
49
|
Custodio JM, Donaldson KM, Hunt HJ. An In Vitro and In Vivo Evaluation of the Effect of Relacorilant on the Activity of Cytochrome P450 Drug Metabolizing Enzymes. J Clin Pharmacol 2021; 61:244-253. [PMID: 32869328 PMCID: PMC7818505 DOI: 10.1002/jcph.1731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/10/2020] [Indexed: 11/06/2022]
Abstract
Relacorilant is a selective modulator of the glucocorticoid receptor in development for the treatment of several serious diseases. The widely used cocktail method was employed to assess relacorilant's effect on various cytochrome P450 (CYP) drug metabolizing enzymes in vitro and in vivo. Inhibition of CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2B6, CYP2C8, CYP3A4, and CYP3A5 as well as induction of CYP1A2, CYP2B6, and CYP3A4 were assessed in vitro (relacorilant concentrations up to 10 µM). A clinical study in healthy subjects (n = 27) evaluated the inhibition of CYP3A4, CYP2C8, and CYP2C9 in vivo by administering single doses of probe CYP substrates (midazolam, pioglitazone, and tolbutamide) alone and in combination with relacorilant (350 mg). Pharmacokinetic sampling was conducted, and safety was assessed throughout the study. Pharmacokinetic parameters were evaluated using 90% confidence intervals of the geometric least squares mean ratios of test (probe substrate with relacorilant) vs reference (probe substrate alone) using boundaries of 80% to 125%. In vitro, relacorilant inhibited CYP3A4, CYP2C8, and CYP2C9 but did not meaningfully affect the activity of the other CYP enzymes evaluated. Consistent with the in vitro data, relacorilant was shown to be a strong CYP3A inhibitor in vivo (>8-fold increase in midazolam area under the concentration versus time curve from time zero to the last quantifiable concentration and area under the concentration versus time curve from time zero extrapolated to infinity). Coadministration of relacorilant with drugs highly dependent on CYP3A for clearance is expected to increase the concentrations of these drugs. Importantly, clinical evaluation of relacorilant showed no inhibition of CYP2C8 or CYP2C9 in vivo. Accordingly, drugs that are substrates of only CYP2C8 and/or CYP2C9 can be coadministered with relacorilant without dose adjustment.
Collapse
|
50
|
Malavia N, Kuche K, Ghadi R, Jain S. A bird's eye view of the advanced approaches and strategies for overshadowing triple negative breast cancer. J Control Release 2020; 330:72-100. [PMID: 33321156 DOI: 10.1016/j.jconrel.2020.12.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Triple negative breast cancer (TNBC) is one of the most aggressive form of breast cancer. It is characterized by the absence of estrogen, progesterone and human epidermal growth factor receptors. The main issue with TNBC is that it exhibits poor prognosis, high risk of relapse, short progression-free survival and low overall survival in patients. This is because the conventional therapy used for managing TNBC has issues pertaining to poor bioavailability, lower cellular uptake, increased off-target effects and development of resistance. To overcome such pitfalls, several other approaches are explored. In this context, the present manuscript showcases three of the most widely used approaches which are (i) nanotechnology-based approach; (ii) gene therapy approach and (iii) Phytochemical-based approach. The ultimate focus is to present and explain the insightful reports based on these approaches. Further, the review also expounds on the identified molecular targets and novel targeting ligands which are explored for managing TNBC effectively. Thus, in a nutshell, the review tries to highlight these existing treatment approaches which might inspire for future development of novel therapies with a potential of overshadowing TNBC.
Collapse
Affiliation(s)
- Nilesh Malavia
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India
| | - Kaushik Kuche
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India
| | - Rohan Ghadi
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India.
| |
Collapse
|