1
|
van Ravensteijn SG, Amir AL, Tauriello DVF, van Herpen CML, Boers-Sonderen MJ, Wesseling YJW, van Brussel AGC, Keizer DM, Verheul HMW, Bol KF. Exploring the relation between TGF-β pathway activity and response to checkpoint inhibition in patients with metastatic melanoma. Clin Exp Immunol 2025; 219:uxae108. [PMID: 39668127 PMCID: PMC11773812 DOI: 10.1093/cei/uxae108] [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: 06/25/2024] [Revised: 09/23/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024] Open
Abstract
INTRODUCTION Immune checkpoint inhibition (ICI) is highly effective for the treatment of melanoma, but intrinsic resistance is present in a subgroup of patients. TGF-β pathway activity may play a role in this resistance by preventing T-cells from entering the tumor microenvironment, causing immune escape. We investigated the association of TGF-β signal transduction pathway activity with resistance to ICI treatment in advanced melanoma. Furthermore, other pathway activities were analyzed to better understand their potential role in ICI resistance. METHOD The activity of 8 signaling pathways (TGF-β, Hedgehog, MAPK, AR, NOTCH, PI3K, JAK/STAT1-2, and NFkB) was analyzed from tumor tissue from patients with advanced melanoma. Pathway activity scores (PAS) were explored for associations with survival and response to ICI in 34 patients (19 non-responders and 15 responders). A second, independent method to investigate the predictive value of TGF-β pathway activation was conducted by determining levels of phosphorylated SMAD2. RESULTS The mean TGF-β PAS of responders vs non-responders was 53.9 vs 56.8 (P = 0.265). No significant relation with progression-free survival was detected for TGF-β activity (P = 0.078). No association between pSMAD2 staining and treatment response or survival was identified. In contrast, Hedgehog scores of responders versus non-responders were 35.7 vs 41.6 (P = 0.038). High Hedgehog PAS was the sole significant predictor of resistance to ICI (OR 0.88, P = 0.033) and worse progression-free survival (HR 1-1.1, P = 0.012). CONCLUSION TGF-β pathway activation showed no significant relation with treatment response to ICI or survival in patients with advanced melanoma. Hedgehog PAS was identified as a possible biomarker associated with both treatment response and survival.
Collapse
Affiliation(s)
| | - Avital L Amir
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Daniele V F Tauriello
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Carla M L van Herpen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marye J Boers-Sonderen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | | | | | - Henk M W Verheul
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Kalijn F Bol
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
2
|
van Weelden WJ, Birkendahl PB, Lalisang RI, Inthout J, Kruitwagen RFPM, Romano A, Pijnenborg JMA. Author reply. BJOG 2024; 131:382-383. [PMID: 37814390 DOI: 10.1111/1471-0528.17676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 07/12/2023] [Indexed: 10/11/2023]
Affiliation(s)
- Willem Jan van Weelden
- Department of Obstetrics and Gynaecology, Radboud Institute for Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Obstetrics and Gynaecology, Canisius Wilhelmina Ziekenhuis, Nijmegen, The Netherlands
| | - Philine B Birkendahl
- Department of Obstetrics and Gynaecology, Radboud Institute for Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Roy I Lalisang
- Division of Medical Oncology, Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands
- GROW-School of Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Joanna Inthout
- Section Biostatistics, Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Roy F P M Kruitwagen
- GROW-School of Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Andrea Romano
- GROW-School of Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Johanna M A Pijnenborg
- Department of Obstetrics and Gynaecology, Radboud Institute for Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
3
|
van der Ploeg P, Hendrikse CSE, Thijs AMJ, Westgeest HM, Smedts HPM, Vos MC, Jalving M, Lok CAR, Boere IA, van Ham MAPC, Ottevanger PB, Westermann AM, Mom CH, Lalisang RI, Lambrechts S, Bekkers RLM, Piek JMJ. Phenotype-guided targeted therapy based on functional signal transduction pathway activity in recurrent ovarian cancer patients: The STAPOVER study protocol. Heliyon 2024; 10:e23170. [PMID: 38187310 PMCID: PMC10770441 DOI: 10.1016/j.heliyon.2023.e23170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 11/15/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024] Open
Abstract
Objective Ovarian cancer is the fifth cause of cancer-related death among women. The benefit of targeted therapy for ovarian cancer patients is limited even if treatment is stratified by molecular signature. There remains a high unmet need for alternative diagnostics that better predict targeted therapy, as current diagnostics are generally inaccurate predictors. Quantitative assessment of functional signal transduction pathway (STP) activity from mRNA measurements of target genes is an alternative approach. Therefore, we aim to identify aberrantly activated STPs in tumour tissue of patients with recurrent ovarian cancer and start phenotype-guided targeted therapy to improve survival without compromising quality of life. Study design Patients with recurrent ovarian cancer and either 1) have platinum-resistant disease, 2) refrain from standard therapy or 3) are asymptomatic and not yet eligible for standard therapy will be included in this multi-centre prospective cohort study with multiple stepwise executed treatment arms. Targeted therapy will be available for patients with aberrantly high functional activity of the oestrogen receptor, androgen receptor, phosphoinositide 3-kinase or Hedgehog STP. The primary endpoint of this study is the progression-free survival (PFS) ratio (PFS2/PFS1 ratio) according to RECIST 1.1 determined by the PFS on matched targeted therapy (PFS2) compared to PFS on prior therapy (PFS1). Secondary endpoints include among others best overall response, overall survival, side effects, health-related quality of life and cost-effectiveness. Conclusion The results of this study will show the clinical applicability of STP activity in selecting recurrent ovarian cancer patients for effective therapies.
Collapse
Affiliation(s)
- Phyllis van der Ploeg
- Department of Obstetrics and Gynaecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Cynthia SE. Hendrikse
- Department of Obstetrics and Gynaecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Anna MJ. Thijs
- Department of Internal Medicine and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
| | - Hans M. Westgeest
- Department of Internal Medicine, Amphia Hospital, Breda, the Netherlands
| | - Huberdina PM. Smedts
- Department of Obstetrics and Gynaecology, Amphia Hospital, Breda, the Netherlands
| | - M Caroline Vos
- Department of Obstetrics and Gynaecology, Elisabeth-Tweesteden Hospital, Tilburg, the Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University Medical Centre Groningen, Groningen, the Netherlands
| | - Christianne AR. Lok
- Department of Gynaecologic Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Ingrid A. Boere
- Department of Medical Oncology, Erasmus Medical Centre Cancer Institute, Rotterdam, the Netherlands
| | - Maaike APC. van Ham
- Department of Obstetrics and Gynaecology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | | | - Anneke M. Westermann
- Department of Oncology, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Constantijne H. Mom
- Department of Obstetrics and Gynaecology, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Roy I. Lalisang
- Department of Medical Oncology, Maastricht University Medical Centre +, Maastricht, the Netherlands
| | - Sandrina Lambrechts
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre +, Maastricht, the Netherlands
| | - Ruud LM. Bekkers
- Department of Obstetrics and Gynaecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Jurgen MJ. Piek
- Department of Obstetrics and Gynaecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
| |
Collapse
|
4
|
Verschoor N, Smid M, Jager A, Sleijfer S, Wilting SM, Martens JWM. Integrative whole-genome and transcriptome analysis of HER2-amplified metastatic breast cancer. Breast Cancer Res 2023; 25:145. [PMID: 37968696 PMCID: PMC10648326 DOI: 10.1186/s13058-023-01743-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/06/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND In breast cancer, the advent of anti-HER2 therapies has made HER2+ tumors a highly relevant subgroup. However, the exact characteristics which prohibit clinical response to anti-HER2 therapies and drive disease progression are not yet fully known. Integrative whole-genome and transcriptomic sequencing data from both primary and metastatic HER2-positive breast cancer will enhance our understanding of underlying biological processes. METHODS Here, we used WGS and RNA sequencing data of 700 metastatic breast tumors, of which 68 being HER2+, to search for specific genomic features of HER2+ disease and therapy resistance. Furthermore, we integrated results with transcriptomic data to associate tumors exhibiting a HER2+-specific gene expression profile with ERBB2 mutation status, prior therapy and relevant gene expression signatures. RESULTS Overall genomic profiles of primary and metastatic HER2+ breast cancers were similar, and no specific acquired genomics traits connected to prior anti-HER2 treatment were observed. However, specific genomic features were predictive of progression-free survival on post-biopsy anti-HER2 treatment. Furthermore, a HER2-driven expression profile grouped HER2-amplified tumors with ERBB2-mutated cases and cases without HER2 alterations. The latter were reported as ER positive in primary disease, but the metastatic biopsy showed low ESR1 expression and upregulation of the MAPK pathway, suggesting transformation to ER independence. CONCLUSIONS In summary, although the quantity of variants increased throughout HER2-positive breast cancer progression, the genomic composition remained largely consistent, thus yielding no new major processes beside those already operational in primary disease. Our results suggest that integrated genomic and transcriptomic analyses may be key in establishing therapeutic options.
Collapse
Affiliation(s)
- Noortje Verschoor
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| | - Marcel Smid
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Saskia M Wilting
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| |
Collapse
|
5
|
Banerjee S, Giannone G, Clamp AR, Ennis DP, Glasspool RM, Herbertson R, Krell J, Riisnaes R, Mirza HB, Cheng Z, McDermott J, Green C, Kristeleit RS, George A, Gourley C, Lewsley LA, Rai D, Banerji U, Hinsley S, McNeish IA. Efficacy and Safety of Weekly Paclitaxel Plus Vistusertib vs Paclitaxel Alone in Patients With Platinum-Resistant Ovarian High-Grade Serous Carcinoma: The OCTOPUS Multicenter, Phase 2, Randomized Clinical Trial. JAMA Oncol 2023; 9:675-682. [PMID: 36928279 PMCID: PMC10020933 DOI: 10.1001/jamaoncol.2022.7966] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/05/2022] [Indexed: 03/18/2023]
Abstract
Importance Patients with platinum-resistant or refractory ovarian high-grade serous carcinoma (PR-HGSC) have a poor prognosis and few therapeutic options. Preclinical studies support targeting PI3K/AKT/mTOR signaling in this setting, and a phase 1 study of the dual mTORC1/mTORC2 inhibitor vistusertib with weekly paclitaxel showed activity. Objective To evaluate whether the addition of vistusertib to weekly paclitaxel improves clinical outcomes in patients with PR-HGSC. Design, Setting, and Participants This phase 2, double-blind, placebo-controlled multicenter randomized clinical trial recruited patients from UK cancer centers between January 2016 and March 2018. Patients with PR-HGSC of ovarian, fallopian tube, or primary peritoneal origin and with measurable or evaluable disease (Response Evaluation Criteria in Solid Tumors version 1.1 and/or Gynecological Cancer Intergroup cancer antigen 125 criteria) were eligible. There were no restrictions on number of lines of prior therapy. Data analysis was performed from May 2019 to January 2022. Interventions Patients were randomized (1:1) to weekly paclitaxel (80 mg/m2 days 1, 8, and 15 of a 28-day cycle) plus oral vistusertib (50 mg twice daily) or placebo. Main Outcomes and Measures The primary end point was progression-free survival in the intention-to-treat population. Secondary end points included response rate, overall survival, and quality of life. Results A total of 140 patients (median [range] age, 63 [36-86] years; 17.9% with platinum-refractory disease; 53.6% with ≥3 prior therapies) were randomized. In the paclitaxel plus vistusertib vs paclitaxel plus placebo groups, there was no difference in progression-free survival (median, 4.5 vs 4.1 months; hazard ratio [HR], 0.84; 80% CI, 0.67-1.07; 1-sided P = .18), overall survival (median, 9.7 vs 11.1 months; HR, 1.21; 80% CI, 0.91-1.60) or response rate (odds ratio, 0.86; 80% CI, 0.55-1.36). Grade 3 to 4 adverse events were 41.2% (weekly paclitaxel plus vistusertib) vs 36.7% (weekly paclitaxel plus placebo), and there was no difference in quality of life. Conclusions and Relevance In this randomized clinical trial of weekly paclitaxel and dual mTORC1/2 inhibition in patients with PR-HGSC, vistusertib did not improve clinical activity of weekly paclitaxel. Trial Registration isrctn.org Identifier: ISRCTN16426935.
Collapse
Affiliation(s)
- Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies, Institute of Cancer Research, London, United Kingdom
| | - Gaia Giannone
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Andrew R. Clamp
- The Christie NHS Foundation Trust and University of Manchester, Manchester, United Kingdom
| | - Darren P. Ennis
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, United Kingdom
| | | | - Rebecca Herbertson
- Sussex Cancer Centre, Royal Sussex County Hospital, Brighton, United Kingdom
| | - Jonathan Krell
- Medical Oncology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Ruth Riisnaes
- Division of Cancer Therapeutics, Institute of Cancer Research, London, United Kingdom
| | - Hasan B. Mirza
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Zhao Cheng
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Jacqueline McDermott
- Department of Histopathology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Clare Green
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Rebecca S. Kristeleit
- Research Department of Oncology, UCL Cancer Institute, University College London, London, United Kingdom
- Now with Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Angela George
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Charlie Gourley
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Liz-Anne Lewsley
- CRUK Glasgow Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Debbie Rai
- CRUK Glasgow Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Udai Banerji
- Division of Cancer Therapeutics, Institute of Cancer Research, London, United Kingdom
| | - Samantha Hinsley
- CRUK Glasgow Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Iain A. McNeish
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, United Kingdom
- Medical Oncology, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
6
|
El-Khouly OA, Henen MA, El-Sayed MAA, El-Messery SM. Design, synthesis and computational study of new benzofuran hybrids as dual PI3K/VEGFR2 inhibitors targeting cancer. Sci Rep 2022; 12:17104. [PMID: 36224254 PMCID: PMC9556824 DOI: 10.1038/s41598-022-21277-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/26/2022] [Indexed: 01/04/2023] Open
Abstract
Design and synthesis of a new series of benzofuran derivatives has been performed. 1H-NMR, 13C-NMR, elemental analysis, and IR were used to confirm the structures of the produced compounds. Hepatocellular carcinoma (HePG2), mammary gland breast cancer (MCF-7), epithelioid carcinoma cervical cancer (Hela), and human prostate cancer are used to test anticancer activity (PC3). In compared to DOX (4.17-8.87 µM), Compound 8 demonstrated the highest activity against HePG and PC3 cell lines, with an IC50 range of 11-17 µM. Compound 8 inhibited PI3K and VEGFR-2 with IC50 values of 2.21 and 68 nM, respectively, compared to 6.18 nM for compound LY294002 and 31.2 nM for compound sorafenib as PI3K and VEGFR-2 reference inhibitors, selectively. The molecular docking and binding affinity of the generated compounds were estimated and studied computationally utilizing molecular operating environment software as a PI3K and VEGFR-2 inhibitor (MOE). In conclusion, compound 8 exhibited significant action against hepatocellular and cervical cancer cell lines. Mechanistic study showed that it had a dual inhibitory effect against PI3K and VEGFR-2.
Collapse
Affiliation(s)
- Omar A. El-Khouly
- grid.10251.370000000103426662Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt ,grid.10251.370000000103426662Faculty of Pharmacy, New Mansoura University, P.O. Box 35712, New Mansoura, Egypt
| | - Morkos A. Henen
- grid.10251.370000000103426662Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt ,grid.241116.10000000107903411Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, USA
| | - Magda A.-A. El-Sayed
- grid.10251.370000000103426662Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt ,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University, P.O. Box 34518, New Damietta, Egypt
| | - Shahenda M. El-Messery
- grid.10251.370000000103426662Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt ,grid.10251.370000000103426662Faculty of Pharmacy, New Mansoura University, P.O. Box 35712, New Mansoura, Egypt
| |
Collapse
|
7
|
Liu C, Hu S, Xu X, Zhang Y, Wang B, Song S, Yang Z. Evaluation of tumour heterogeneity by 18F-fluoroestradiol PET as a predictive measure in breast cancer patients receiving palbociclib combined with endocrine treatment. BREAST CANCER RESEARCH : BCR 2022; 24:57. [PMID: 36028895 PMCID: PMC9419349 DOI: 10.1186/s13058-022-01555-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 08/16/2022] [Indexed: 01/26/2023]
Abstract
Background Predictive biomarkers are needed to identify oestrogen receptor-positive, human epidermal growth factor receptor 2-negative (ER + /HER2-) metastatic breast cancer (MBC) patients who would likely benefit from cyclin-dependent kinase 4 and 6 inhibitors combined with endocrine therapy. Therefore, we performed an exploratory study to evaluate the tumour heterogeneity parameters based on 16α-18F-fluoro-17β-oestradiol (18F-FES)-PET imaging as a potential marker to predict progression-free survival (PFS) in MBC patients receiving palbociclib combined with endocrine therapy. Methods Fifty-six ER + MBC patients underwent 18F-FES-PET/CT before the initiation of palbociclib. 18F-FES uptake was quantified and expressed as the standardized uptake value (SUV). Interlesional heterogeneity was qualitatively identified according to the presence or absence of 18F-FES-negative lesions. Intralesional heterogeneity was measured by the SUV-based heterogeneity index (HI = SUVmax/SUVmean). Association with survival was evaluated using the Cox proportional hazards model. Results A total of 551 metastatic lesions were found in 56 patients: 507 lesions were identified as 18F-FES-positive, 38 lesions were distributed across 10 patients without 18F-FES uptake, and the remaining 6 were liver lesions. Forty-three patients obtained a clinical benefit, and 13 developed progressive disease (PD) within 24 weeks. Nine out of 10 patients with an 18F-FES-negative site developed PD, and the median PFS was only 2.4 months. Among 46 patients with only 18F-FES-positive lesions, only four patients had PD, and the median PFS was 23.6 months. There were statistically significant differences between the two groups (P < 0.001). For the subgroup of patients with only 18F-FES-positive lesions, low FES-HI patients experienced substantially longer PFS times than those with high FES-HI (26.5 months vs. 16.5 months, P = 0.004). Conclusions 18F-FES-PET may provide a promising method for identifying and selecting candidate ER + /HER2- MBC patients who would most likely benefit from palbociclib combined with endocrine treatment and could serve as a predictive marker for treatment response. Trial registration NCT04992156, Date of registration: August 5, 2021 (retrospectively registered).
Collapse
Affiliation(s)
- Cheng Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Shanghai Institute of Medical Imaging, Fudan University, Shanghai, 200032, China.,Center for Biomedical Imaging, Fudan University, Shanghai, 200032, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032, China
| | - Shihui Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaoping Xu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Center for Biomedical Imaging, Fudan University, Shanghai, 200032, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032, China
| | - Yongping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Center for Biomedical Imaging, Fudan University, Shanghai, 200032, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032, China
| | - Biyun Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Institute of Medical Imaging, Fudan University, Shanghai, 200032, China. .,Center for Biomedical Imaging, Fudan University, Shanghai, 200032, China. .,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032, China.
| | - Zhongyi Yang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, No.270, Dong'an Road, Xuhui District, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Center for Biomedical Imaging, Fudan University, Shanghai, 200032, China. .,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032, China.
| |
Collapse
|
8
|
Qiao D, Qin X, Yang H, Liu X, Liu L, Liu S, Jia Z. Estradiol mediates the interaction of LINC01541 and miR-429 to promote angiogenesis of G1/G2 endometrioid adenocarcinoma in-vitro: A pilot study. Front Oncol 2022; 12:951573. [PMID: 35992774 PMCID: PMC9389109 DOI: 10.3389/fonc.2022.951573] [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: 05/24/2022] [Accepted: 07/18/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundEndometrioid adenocarcinoma (EAC) is the most common subtype of endometrial cancer (EC) and is an estrogen-related cancer. In this study, we sought to investigate the expressions and mechanism of action of 17β-estradiol (E2) and long noncoding RNA (lncRNA) LINC01541 in G1/G2 EAC samples.MethodsThe expressions of estrogen receptor β (ESR2), LINC01541, miR-200s, and VEGFA were evaluated using real-time PCR in human EAC tissues (n = 8) and adjacent normal tissues (n = 8). Two EC cell lines (Ishikawa and RL95-2) were selected for validation in vitro. Bioinformatics analyses and luciferase reporter analyses were performed to verify potential binding sites. qRT-PCR, Western blot, and CCK-8 were used to identify the regulatory mechanisms of related genes in cell biological behavior.ResultsCompared with adjacent normal tissues, LINC01541 and miR-200s family (except miR-200c) were highly expressed in EAC tissues (n=8), while ESR2 and VEGFA were lowly expressed in EAC tissues (* P < 0.05; ** P < 0.01). In vitro: E2 inhibited the expression of LINC01541 and miR-429 in both cell lines, and estrogen antagonist (PHTPP) could reverse this effect, in addition, PHTPP could promote the proliferation of these two cancer cells, cell transfection LINC01541 also had this effect after overexpression of plasmid and miR-429 mimic. E2 promotes the expression of VEGFA in both cell lines, and PHTPP can also reverse this effect. LINC01541 interacts with miR-429 to promote the expression of each other, and both inhibit the synthesis of VEGFA in EAC cells after overexpression. Through the double validation of bioinformatics analysis and dual fluorescein reporter gene, it was confirmed that miR-429 targets the regulation of VEGFA expression (* P < 0.05; ** P < 0.01).ConclusionE2 promotes the synthesis of VEGFA by altering the expression levels of LINC01541 and miR-429 in EAC, thereby affecting the angiogenesis process of EAC. Also, E2-mediated LINC01541/miR-429 expression may affect cell migration in EAC. In addition, we identified a reciprocal promotion between LINC01541 and miR-429.
Collapse
Affiliation(s)
- Dan Qiao
- Department of Gynecology, Dalian Medical University, Dalian, China
| | - Xiaoduo Qin
- Department of Gynecology, Dalian Medical University, Dalian, China
| | - Haiyan Yang
- Department of Gynecology, Dalian Medical University, Dalian, China
| | - Xuantong Liu
- Department of Gynecology, Changzhou No. 2 People’s Hospital, Changzhou, China
| | - Libing Liu
- Department of Gynecology, Changzhou No. 2 People’s Hospital, Changzhou, China
| | - Sufen Liu
- Department of Gynecology, Changzhou No. 2 People’s Hospital, Changzhou, China
- *Correspondence: Sufen Liu, ; Zhongzhi Jia,
| | - Zhongzhi Jia
- Department of Interventional Radiology, Changzhou No. 2 People’s Hospital, Changzhou, China
- *Correspondence: Sufen Liu, ; Zhongzhi Jia,
| |
Collapse
|
9
|
Matrix Metalloproteinase-1 (MMP1) Upregulation through Promoter Hypomethylation Enhances Tamoxifen Resistance in Breast Cancer. Cancers (Basel) 2022; 14:cancers14051232. [PMID: 35267540 PMCID: PMC8909089 DOI: 10.3390/cancers14051232] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Cancer recurrence caused by tamoxifen resistance hampers chemotherapy in breast cancer patients. The reasons behind the resistance were investigated by screening epigenetically regulated genes through analysis of methylation data from tamoxifen-resistant MCF-7 cells. MMP1 locus was found to be hypomethylated at a promoter CpG site and its expression was upregulated in the cell line, which was verified by the drug-resistant tumor tissues from breast cancer patients (n = 28). Downregulating MMP1 using a short hairpin RNA inhibited the growth of resistant cells and increased sensitivity to tamoxifen in vitro as well as in a xenografted mouse model in vivo. This study suggests that MMP1 is potentially a target gene to control tamoxifen resistance in breast cancer. Abstract Background: Tamoxifen (tam) is widely used to treat estrogen-positive breast cancer. However, cancer recurrence after chemotherapy remains a major obstacle to achieve good patient prognoses. In this study, we aimed to identify genes responsible for epigenetic regulation of tam resistance in breast cancer. Methods: Methylation microarray data were analyzed to screen highly hypomethylated genes in tam resistant (tamR) breast cancer cells. Quantitative RT-PCR, Western blot analysis, and immunohistochemical staining were used to quantify expression levels of genes in cultured cells and cancer tissues. Effects of matrix metalloproteinase-1 (MMP1) expression on cancer cell growth and drug resistance were examined through colony formation assays and flow cytometry. Xenografted mice were generated to investigate the effects of MMP1 on drug resistance in vivo. Results: MMP1 was found to be hypomethylated and overexpressed in tamR MCF-7 (MCF-7/tamR) cells and in tamR breast cancer tissues. Methylation was found to be inversely associated with MMP1 expression level in breast cancer tissues, and patients with lower MMP1 expression exhibited a better prognosis for survival. Downregulating MMP1 using shRNA induced tam sensitivity in MCF-7/tamR cells along with increased apoptosis. The xenografted MCF-7/tamR cells that stably expressed short hairpin RNA (shRNA) against MMP1 exhibited retarded tumor growth compared to that in cells expressing the control shRNA, which was further suppressed by tam. Conclusions: MMP1 can be upregulated through promoter hypomethylation in tamR breast cancer, functioning as a resistance driver gene. MMP1 can be a potential target to suppress tamR to achieve better prognoses of breast cancer patients.
Collapse
|
10
|
van Weelden WJ, Lalisang RI, Bulten J, Lindemann K, van Beekhuizen HJ, Trum H, Boll D, Werner HM, van Lonkhuijzen LR, Yigit R, Forsse D, Witteveen PO, Galaal K, van Ginkel A, Bignotti E, Weinberger V, Sweegers S, Kroep JR, Cabrera S, Snijders MP, Inda MA, Eriksson AGZ, Krakstad C, Romano A, van de Stolpe A, Pijnenborg JM, Pijnenborg JMA. Impact of hormonal biomarkers on response to hormonal therapy in advanced and recurrent endometrial cancer. Am J Obstet Gynecol 2021; 225:407.e1-407.e16. [PMID: 34019887 DOI: 10.1016/j.ajog.2021.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/02/2021] [Accepted: 05/08/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Approximately 20% of women with endometrial cancer have advanced-stage disease or suffer from a recurrence. For these women, prognosis is poor, and palliative treatment options include hormonal therapy and chemotherapy. Lack of predictive biomarkers and suboptimal use of existing markers for response to hormonal therapy have resulted in overall limited efficacy. OBJECTIVE This study aimed to improve the efficacy of hormonal therapy by relating immunohistochemical expression of estrogen and progesterone receptors and estrogen receptor pathway activity scores to response to hormonal therapy. STUDY DESIGN Patients with advanced or recurrent endometrial cancer and available biopsies taken before the start of hormonal therapy were identified in 16 centers within the European Network for Individualized Treatment in Endometrial Cancer and the Dutch Gynecologic Oncology Group. Tumor tissue was analyzed for estrogen and progesterone receptor expressions and estrogen receptor pathway activity using a quantitative polymerase chain reaction-based messenger RNA model to measure the activity of estrogen receptor-related target genes in tumor RNA. The primary endpoint was response rate defined as complete and partial response using the Response Evaluation Criteria in Solid Tumors. The secondary endpoints were clinical benefit rate and progression-free survival. RESULTS Pretreatment biopsies with sufficient endometrial cancer tissue and complete response evaluation were available in 81 of 105 eligible cases. Here, 22 of 81 patients (27.2%) with a response had estrogen and progesterone receptor expressions of >50%, resulting in a response rate of 32.3% (95% confidence interval, 20.9-43.7) for an estrogen receptor expression of >50% and 50.0% (95% confidence interval, 35.2-64.8) for a progesterone receptor expression of >50%. Clinical benefit rate was 56.9% for an estrogen receptor expression of >50% (95% confidence interval, 44.9-68.9) and 75.0% (95% confidence interval, 62.2-87.8) for a progesterone receptor expression of >50%. The application of the estrogen receptor pathway test to cases with a progesterone receptor expression of >50% resulted in a response rate of 57.6% (95% confidence interval, 42.1-73.1). After 2 years of follow-up, 34.3% of cases (95% confidence interval, 20-48) with a progesterone receptor expression of >50% and 35.8% of cases (95% confidence interval, 20-52) with an estrogen receptor pathway activity score of >15 had not progressed. CONCLUSION The prediction of response to hormonal treatment in endometrial cancer improves substantially with a 50% cutoff level for progesterone receptor immunohistochemical expression and by applying a sequential test algorithm using progesterone receptor immunohistochemical expression and estrogen receptor pathway activity scores. However, results need to be validated in the prospective Prediction of Response to Hormonal Therapy in Advanced and Recurrent Endometrial Cancer (PROMOTE) study.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Johanna M A Pijnenborg
- Department of Obstetrics and Gynaecology, Radboud Institute of Health Sciences, Radboud university medical center, Nijmegen, the Netherlands
| |
Collapse
|
11
|
Lassche G, Tada Y, van Herpen CML, Jonker MA, Nagao T, Saotome T, Hirai H, Saigusa N, Takahashi H, Ojiri H, van Engen-Van Grunsven ACH, Schalken JA, Fushimi C, Verhaegh GW. Predictive and Prognostic Biomarker Identification in a Large Cohort of Androgen Receptor-Positive Salivary Duct Carcinoma Patients Scheduled for Combined Androgen Blockade. Cancers (Basel) 2021; 13:cancers13143527. [PMID: 34298742 PMCID: PMC8307921 DOI: 10.3390/cancers13143527] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 12/19/2022] Open
Abstract
Patients suffering from recurrent or metastatic (R/M) salivary duct carcinoma (SDC) are often treated with combined androgen blockade (CAB). However, CAB frequently fails, resulting in a worse prognosis. Therefore, biomarkers that can predict treatment failure are urgently needed. mRNA from 76 R/M androgen receptor (AR)-positive SDC patients treated with leuprorelin acetate combined with bicalutamide was extracted from pre-treatment tumor specimens. AR, Notch, MAPK, TGFβ, estrogen receptor (ER), Hedgehog (HH), and PI3K signaling pathway activity scores (PAS) were determined based on the expression levels of target genes. Additionally, 5-alpha reductase type 1 (SRD5A1) expression was determined. These markers were related to clinical benefit (complete/partial response or stable disease ≥6 months) and progression-free and overall survival (PFS/OS). SRD5A1 expression had the highest general predictive value for clinical benefit and positive predictive value (PPV: 85.7%). AR PAS had the highest negative predictive value (NPV: 93.3%). The fitting of a multivariable model led to the identification of SRD5A1, TGFβ, and Notch PAS as the most predictive combination. High AR, high Notch, high ER, low HH PAS, and high SRD5A1 expression were also of prognostic importance regarding PFS and SRD5A1 expression levels for OS. AR, Notch PAS, and SRD5A1 expression have the potential to predict the clinical benefit of CAB treatment in SDC patients. SRD5A1 expression can identify patients that will and AR PAS patients that will not experience clinical benefit (85.7% and 93.3% for PPV and NPV, respectively). The predictive potential of SRD5A1 expression forms a rational basis for including SRD5A1-inhibitors in SDC patients' treatment.
Collapse
Affiliation(s)
- Gerben Lassche
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands;
| | - Yuichiro Tada
- Department of Head and Neck Oncology and Surgery, International University of Health and Welfare, Mita Hospital, Tokyo 108-8329, Japan; (Y.T.); (C.F.)
| | - Carla M. L. van Herpen
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands;
- Correspondence: ; Tel.: +31-24-3667251
| | - Marianne A. Jonker
- Department of Health Evidence, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands;
| | - Toshitaka Nagao
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo 160-0023, Japan; (T.N.); (H.H.); (N.S.)
| | - Takashi Saotome
- Division of Medical Oncology, Matsudo City General Hospital, Chiba 270-2296, Japan;
| | - Hideaki Hirai
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo 160-0023, Japan; (T.N.); (H.H.); (N.S.)
| | - Natsuki Saigusa
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo 160-0023, Japan; (T.N.); (H.H.); (N.S.)
| | - Hideaki Takahashi
- Department of Otorhinolaryngology, Head and Neck Surgery, School of Medicine, Yokohama City University, Kanagawa 236-0004, Japan;
| | - Hiroya Ojiri
- Department of Radiology, The Jikei University School of Medicine, Tokyo 105-8461, Japan;
| | | | - Jack A. Schalken
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands; (J.A.S.); (G.W.V.)
| | - Chihiro Fushimi
- Department of Head and Neck Oncology and Surgery, International University of Health and Welfare, Mita Hospital, Tokyo 108-8329, Japan; (Y.T.); (C.F.)
| | - Gerald W. Verhaegh
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands; (J.A.S.); (G.W.V.)
| |
Collapse
|
12
|
van der Ploeg P, Uittenboogaard A, Thijs AMJ, Westgeest HM, Boere IA, Lambrechts S, van de Stolpe A, Bekkers RLM, Piek JMJ. The effectiveness of monotherapy with PI3K/AKT/mTOR pathway inhibitors in ovarian cancer: A meta-analysis. Gynecol Oncol 2021; 163:433-444. [PMID: 34253390 DOI: 10.1016/j.ygyno.2021.07.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To determine the clinical benefit of monotherapy with PI3K/AKT/mTOR inhibitors in patients diagnosed with advanced or recurrent ovarian cancer and to investigate the predictive value of current PI3K/AKT/mTOR biomarkers on therapy response. METHODS A systematic search was conducted in PubMed, Embase and the Cochrane Library for articles reporting on treatment with PI3K/AKT/mTOR inhibitors in ovarian cancer. The primary endpoint was defined as the clinical benefit rate (CBR), including the proportion of patients with complete (CR) and partial response (PR) and stable disease (SD). Secondary endpoints included the overall response rate (ORR, including CR and PR) and drug-related grade 3 and 4 adverse events. RESULTS We included 233 patients from 19 studies and observed a pooled CBR of 32% (95% CI 20-44%) and ORR of 3% (95% CI 0-6%) in advanced or recurrent ovarian cancer patients treated with PI3K/AKT/mTOR inhibitors. Subgroup analysis tended to favor the studies who selected patients based on current PI3K/AKT/mTOR biomarker criteria (e.g. genomic alterations or loss of PTEN protein expression), but the difference in CBR was not statistically significant from studies with unselected populations (respectively, CBR of 42% (95% CI 23-62%) and 27% (95% CI 14-42%), P = 0.217). To better reflect true patient benefit, we excluded SD <6 months as a beneficial outcome which resulted in a pooled CBR of 7% (95% CI 2-13%). The overall proportion of patients with drug-related grade 3 and 4 adverse events was 36%. CONCLUSIONS The efficacy of monotherapy with PI3K/AKT/mTOR inhibitors in advanced recurrent ovarian cancer patients is limited to a small subgroup and selection of patients with the use of current biomarkers did not improved the CBR significantly. Given the toxicity profile, we suggest that current treatment with PI3K/AKT/mTOR inhibitors should not be initiated unless in clinical trials. Furthermore, improved biomarkers to measure functional PI3K/AKT/mTOR pathway activity are needed to optimize patient selection.
Collapse
Affiliation(s)
- Phyllis van der Ploeg
- Department of Obstetrics and Gynecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands.
| | - Aniek Uittenboogaard
- Department of Obstetrics and Gynecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
| | - Anna M J Thijs
- Department of Medical Oncology, Catharina Hospital, Eindhoven, the Netherlands
| | | | - Ingrid A Boere
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Sandrina Lambrechts
- Department of Obstetrics and Gynecology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | | | - Ruud L M Bekkers
- Department of Obstetrics and Gynecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Jurgen M J Piek
- Department of Obstetrics and Gynecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
| |
Collapse
|
13
|
Chen J, Wan R, Li Q, Rao Z, Wang Y, Zhang L, Teichmann AT. Utilizing the Hippo pathway as a therapeutic target for combating endocrine-resistant breast cancer. Cancer Cell Int 2021; 21:306. [PMID: 34112175 PMCID: PMC8194146 DOI: 10.1186/s12935-021-01999-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/27/2021] [Indexed: 12/14/2022] Open
Abstract
Drug resistance is always a great obstacle in any endocrine therapy of breast cancer. Although the combination of endocrine therapy and targeted therapy has been shown to significantly improve prognosis, refractory endocrine resistance is still common. Dysregulation of the Hippo pathway is often related to the occurrence and the development of many tumors. Targeted therapies of this pathway have played important roles in the study of triple negative breast cancer (TNBC). Targeting the Hippo pathway in combination with chemotherapy or other targeted therapies has been shown to significantly improve specific antitumor effects and reduce cancer antidrug resistance. Further exploration has shown that the Hippo pathway is closely related to endocrine resistance, and it plays a "co-correlation point" role in numerous pathways involving endocrine resistance, including related pathways in breast cancer stem cells (BCSCs). Agents and miRNAs targeting the components of the Hippo pathway are expected to significantly enhance the sensitivity of breast cancer cells to endocrine therapy. This review initially explains the possible mechanism of the Hippo pathway in combating endocrine resistance, and it concludes by recommending endocrine therapy in combination with therapies targeting the Hippo pathway in the study of endocrine-resistant breast cancers.
Collapse
Affiliation(s)
- Jing Chen
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China.,Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Runlan Wan
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Qinqin Li
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China.,Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Zhenghuan Rao
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China.,Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yanlin Wang
- North Sichuan Medical College, Nanchong, 637000, China
| | - Lei Zhang
- Department of Gynaecology, The Second People's Hospital of Yibin, Yibin, 644000, China
| | - Alexander Tobias Teichmann
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China. .,Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
14
|
Functional estrogen receptor signaling pathway activity in high-grade serous ovarian carcinoma as compared to estrogen receptor protein expression by immunohistochemistry. Cell Oncol (Dordr) 2021; 44:951-957. [PMID: 33723801 PMCID: PMC8338831 DOI: 10.1007/s13402-021-00600-5] [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] [Accepted: 03/02/2021] [Indexed: 01/25/2023] Open
Abstract
Purpose Anti-estrogen therapy may be used as a palliative treatment option in high-grade serous ovarian carcinomas (HGSC). However, clinical implementation is limited as the use of estrogen receptor (ER) protein expression by immunohistochemistry remains insufficient in predicting therapy response. To determine the accuracy of ER protein expression as a marker for ER signaling pathway activity, we aimed to correlate ER protein expression to functional ER signaling pathway activity in HGSC. Methods Immunohistochemical ER protein expression was visually scored using total percentages of stained tumor cells and histoscores. Subsequently, mRNA was extracted, and RT-qPCR analysis was performed. Functional ER pathway activity was assessed by a computational Bayesian model inferring ER signaling pathway activity from mRNA levels of ER-specific target genes. Results Our analysis of 29 HGSCs shows that neither total percentage of ER protein expression, nor ER histoscores are significantly correlated to ER signaling pathway activity (respectively, p = 0.473 and p = 0.606). Classification of HGSC into three groups based on ER histoscores 0–100 (n = 6), 101–200 (n = 15) and 201–300 (n = 8) resulted in comparable mean ER signaling pathway activity among the groups (p = 0.356). Several samples in the higher ER histoscore groups had low ER signaling pathway activity, indicating that nuclear ER protein expression is not sufficient to describe transcriptional ER activation. Conclusion Positive immunohistochemical ER staining is not always indicative of an active ER signaling pathway and is, therefore, a poor predictor of anti-estrogen response. Further research is needed to prove the predictive value of ER signaling pathway activity regarding anti-estrogen sensitivity in HGSC patients. Supplementary Information The online version contains supplementary material available at 10.1007/s13402-021-00600-5.
Collapse
|
15
|
van de Stolpe A, Verhaegh W, Blay JY, Ma CX, Pauwels P, Pegram M, Prenen H, De Ruysscher D, Saba NF, Slovin SF, Willard-Gallo K, Husain H. RNA Based Approaches to Profile Oncogenic Pathways From Low Quantity Samples to Drive Precision Oncology Strategies. Front Genet 2021; 11:598118. [PMID: 33613616 PMCID: PMC7893109 DOI: 10.3389/fgene.2020.598118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/07/2020] [Indexed: 12/31/2022] Open
Abstract
Precision treatment of cancer requires knowledge on active tumor driving signal transduction pathways to select the optimal effective targeted treatment. Currently only a subset of patients derive clinical benefit from mutation based targeted treatment, due to intrinsic and acquired drug resistance mechanisms. Phenotypic assays to identify the tumor driving pathway based on protein analysis are difficult to multiplex on routine pathology samples. In contrast, the transcriptome contains information on signaling pathway activity and can complement genomic analyses. Here we present the validation and clinical application of a new knowledge-based mRNA-based diagnostic assay platform (OncoSignal) for measuring activity of relevant signaling pathways simultaneously and quantitatively with high resolution in tissue samples and circulating tumor cells, specifically with very small specimen quantities. The approach uses mRNA levels of a pathway's direct target genes, selected based on literature for multiple proof points, and used as evidence that a pathway is functionally activated. Using these validated target genes, a Bayesian network model has been built and calibrated on mRNA measurements of samples with known pathway status, which is used next to calculate a pathway activity score on individual test samples. Translation to RT-qPCR assays enables broad clinical diagnostic applications, including small analytes. A large number of cancer samples have been analyzed across a variety of cancer histologies and benchmarked across normal controls. Assays have been used to characterize cell types in the cancer cell microenvironment, including immune cells in which activated and immunotolerant states can be distinguished. Results support the expectation that the assays provide information on cancer driving signaling pathways which is difficult to derive from next generation DNA sequencing analysis. Current clinical oncology applications have been complementary to genomic mutation analysis to improve precision medicine: (1) prediction of response and resistance to various therapies, especially targeted therapy and immunotherapy; (2) assessment and monitoring of therapy efficacy; (3) prediction of invasive cancer cell behavior and prognosis; (4) measurement of circulating tumor cells. Preclinical oncology applications lie in a better understanding of cancer behavior across cancer types, and in development of a pathophysiology-based cancer classification for development of novel therapies and precision medicine.
Collapse
Affiliation(s)
| | | | - Jean-Yves Blay
- Medical Oncology, Université Claude Bernard Lyon 1, Lyon, France
- Centre Léon Bérard, Lyon, France
| | - Cynthia X. Ma
- Medicine, Division of Oncology, Section of Medical Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Patrick Pauwels
- Molecular Pathology, Centre for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
| | - Mark Pegram
- Stanford University School of Medicine, Clinical Research, Stanford Cancer Institute, Stanford, CA, United States
| | - Hans Prenen
- Oncology Department, Head of Phase I – Early Clinical Trials Unit, Clinical Trial Management Program, Oncology Department, Antwerp University Hospital, Antwerp, Belgium
| | - Dirk De Ruysscher
- Oncology-Radiotherapy, Maastro/Maastricht University Medical Center, Maastricht, Netherlands
| | - Nabil F. Saba
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, United States
- Head and Neck Medical Oncology Program, Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | | | | | - Hatim Husain
- University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
16
|
El-Khouly OA, Henen MA, El-Sayed MAA, Shabaan MI, El-Messery SM. Synthesis, anticancer and antimicrobial evaluation of new benzofuran based derivatives: PI3K inhibition, quorum sensing and molecular modeling study. Bioorg Med Chem 2020; 31:115976. [PMID: 33388654 DOI: 10.1016/j.bmc.2020.115976] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 12/23/2022]
Abstract
A new series of benzofuran derivatives has been designed and synthesized. The structures of the synthesized compounds have been confirmed by the use of 1H NMR, 13C NMR, 2D 1H-1H NOESY NMR, and IR. Anticancer activity is evaluated against Hepatocellular carcinoma (HePG2), mammary gland breast cancer (MCF-7), Epitheliod carcinoma cervix cancer (Hela) and human prostate cancer (PC3). Compounds 8, 9, and 11 showed the highest activity towards the four cell lines with an IC50 range of 8.49-16.72 µM, 6.55-13.14 µM and 4-8.99 µM respectively in comparison to DOX (4.17-8.87 µM). Phosphatidylinositol-3-kinases (PI3K) inhibition was evaluated against the most active anticancer compounds 8, 9 and 11. Compounds 8, 9 and 11 showed good inhibitory activity against PI3Kα with IC50 values 4.1, 7.8, and 20.5 µM, respectively in comparison to 6.18 µM for the reference compound LY294002. In addition, activity of compounds 8 and 9 on cell cycle arrest and induction of apoptosis in different phases of MCF-7 cells were assessed and detected pre-G1 apoptosis and cell growth arrest at G2/M. Also, both extrinsic and intrinsic apoptosis in MCF-7 cells induced by compounds 8 and 9. Molecular docking, binding affinity surface mapping, and contact preference of the synthesized compounds 8, 9 and 11 against PI3K were estimated and studied computationally using molecular operating environment software (MOE) and showed good interaction with essential residues for inhibition Val851. In addition, antimicrobial activity was evaluated against gram positive isolates as Staphylococcus aureus and Bacillus cereus, gram negative isolate as Escherichia coli, Pseudomonas aeruginosa and antifungal potential against Candida albicans. Compound 17 showed outstanding anti Gram-positive activity with MIC values 8 and 256 µg/mL in Staphylococcus aureus and Bacillus cereus respectively. Also, compounds 15, 17, 18 and 21 showed good anti Gram-negative activity with MIC value 512 µg/mL for all compounds. In addition, the state-of-art quorum sensing (QS) inhibiting effects were detected using Chromobacterium violaceum and compounds 7, 9, 10, 11, and 12 showed good QS inhibition (3, 3, 5, 2, and 7 mm).
Collapse
Affiliation(s)
- Omar A El-Khouly
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt
| | - Morkos A Henen
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt; Department of Biochemistry & Molecular Genetics, University of Colorado, Denver, USA.
| | - Magda A-A El-Sayed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt; Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Horus University, P.O. Box 34518, New Damietta, Egypt
| | - Mona I Shabaan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt
| | - Shahenda M El-Messery
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, P.O. Box 35516, Mansoura, Egypt.
| |
Collapse
|
17
|
Bouwman W, Verhaegh W, Holtzer L, van de Stolpe A. Measurement of Cellular Immune Response to Viral Infection and Vaccination. Front Immunol 2020; 11:575074. [PMID: 33193365 PMCID: PMC7604353 DOI: 10.3389/fimmu.2020.575074] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022] Open
Abstract
Combined cellular and humoral host immune response determine the clinical course of a viral infection and effectiveness of vaccination, but currently the cellular immune response cannot be measured on simple blood samples. As functional activity of immune cells is determined by coordinated activity of signaling pathways, we developed mRNA-based JAK-STAT signaling pathway activity assays to quantitatively measure the cellular immune response on Affymetrix expression microarray data of various types of blood samples from virally infected patients (influenza, RSV, dengue, yellow fever, rotavirus) or vaccinated individuals, and to determine vaccine immunogenicity. JAK-STAT1/2 pathway activity was increased in blood samples of patients with viral, but not bacterial, infection and was higher in influenza compared to RSV-infected patients, reflecting known differences in immunogenicity. High JAK-STAT3 pathway activity was associated with more severe RSV infection. In contrast to inactivated influenza virus vaccine, live yellow fever vaccine did induce JAK-STAT1/2 pathway activity in blood samples, indicating superior immunogenicity. Normal (healthy) JAK-STAT1/2 pathway activity was established, enabling assay interpretation without the need for a reference sample. The JAK-STAT pathway assays enable measurement of cellular immune response for prognosis, therapy stratification, vaccine development, and clinical testing.
Collapse
|