1
|
-M Fan TW, Yan J, Goncalves CFL, Islam JMM, Lin P, Kaddah MMY, Higashi RM, Lane AN, Wang X, Zhu C. Patient-derived organotypic tissue cultures as a platform to evaluate metabolic reprogramming in breast cancer patients. J Biol Chem 2025:108495. [PMID: 40209948 DOI: 10.1016/j.jbc.2025.108495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025] Open
Abstract
Patient-derived organotypic tissue cultures (PD-OTC) are unique models for probing cancer metabolism and therapeutic responses. They retain patient tissue architectures/ microenvironments that are difficult to recapitulate while affording comparison of cancer (CA) versus matched non-cancer (NC) tissue responses to treatments. We have developed a long-term culturing method for fresh and cryopreserved PD-OTC of breast cancer patients bearing invasive ductal carcinoma. Five PD-OTC came from patients with treatment-naïve primary ER+/PR+/HER2- tumors while one came from a patient with neoadjuvant therapy for locally metastatic ERlow/PR-/HER2- tumor. They all exhibited tissue outgrowth in one month with some CA OTC harboring isolatable organoids and fibroblasts. We interrogated reprogrammed metabolism in CA versus paired NC OTC with dual 2H7-glucose/13C5,15N2-Gln tracers coupled with Stable Isotope-Resolved Metabolomic analysis. We noted variable activation of glycolysis, cataplerotic/anaplerotic Krebs cycle including reductive carboxylation, the pentose phosphate pathway, riboneogenesis, gluconeogenesis (GNG), de novo and salvage synthesis of purine/pyrimidine nucleotides, and ADP-ribosylation in CA PD-OTC. Altered metabolic activities were in part accountable by expression changes in key enzymes measured by Reverse Phase Protein Array profiling. Notably, Gln-fueled GNG products were preferentially diverted to support purine nucleotide synthesis. When blocking this novel process with an inhibitor of phosphoenolpyruvate carboxykinase (3-mercaptopicolinic acid or 3-MPA), metastatic, ERlow/PR-/HER2- CA OTC displayed compromised cellularity, reduced outgrowth, and disrupted growth/survival-supporting metabolism but the matched NC OTC did not. Thus, our PD-OTC culturing method not only promoted understanding of actual patient's tumor metabolism to uncover viable metabolic targets but also enabled target testing and elucidation of therapeutic efficacy.
Collapse
Affiliation(s)
- Teresa W -M Fan
- Center for Environmental and Systems Biochemistry (CESB), Department of Toxicology and Cancer Biology, and Markey Cancer Center, University of Kentucky.
| | - Jing Yan
- F. Joseph Halcomb III, M.D. Department of Biomedical Engineering, University of Kentucky
| | | | - Jahid M M Islam
- Center for Environmental and Systems Biochemistry (CESB), Department of Toxicology and Cancer Biology, and Markey Cancer Center, University of Kentucky
| | - Penghui Lin
- Center for Environmental and Systems Biochemistry (CESB), Department of Toxicology and Cancer Biology, and Markey Cancer Center, University of Kentucky
| | - Mohamed M Y Kaddah
- Center for Environmental and Systems Biochemistry (CESB), Department of Toxicology and Cancer Biology, and Markey Cancer Center, University of Kentucky; Pharmaceutical and Fermentation Industries Development Center, City of Scientific Research and Technological Applications, New Borg El-Arab 21934, Alexandria, Egypt
| | - Richard M Higashi
- Center for Environmental and Systems Biochemistry (CESB), Department of Toxicology and Cancer Biology, and Markey Cancer Center, University of Kentucky
| | - Andrew N Lane
- Center for Environmental and Systems Biochemistry (CESB), Department of Toxicology and Cancer Biology, and Markey Cancer Center, University of Kentucky
| | | | - Caigang Zhu
- F. Joseph Halcomb III, M.D. Department of Biomedical Engineering, University of Kentucky.
| |
Collapse
|
2
|
Zapatero-Solana E, Ding Y, Pulliam N, de Dios A, Ortiz-Ruiz MJ, Lallena MJ. Models of Early Resistance to CDK4/6 Inhibitors Unveil Potential Therapeutic Treatment Sequencing. Int J Mol Sci 2025; 26:2643. [PMID: 40141282 PMCID: PMC11942273 DOI: 10.3390/ijms26062643] [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: 02/04/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND CDK4/6 inhibitors (CDK4/6i) combined with hormone therapies have demonstrated clinical benefit in HR+, HER2- breast cancer patients. However, the onset of resistance remains a concern and highlights a need for therapeutic strategies to improve outcomes. The objective of this study was to develop an in vitro model to better understand the mechanisms of resistance to CDK4/6i + hormone therapies and identify therapeutic strategies with potential to overcome this resistance. METHODS The HR+, HER2- T47D breast cancer cell line genetically modified with a Geminin-Venus reporter construct was treated with CDK4/6i (abemaciclib or palbociclib) in combination with 4-hydroxytamoxifen (tamoxifen). Resistant cells were identified by cell sorting for Geminin (%GEM+), a marker of the S/G2/M phases of the cell cycle, and confirmed by treatment with tamoxifen plus the CDK4/6i used to drive resistance. In resistant cells, following treatment with CDK4/6i + ET (tamoxifen or fulvestrant), the effects on cell proliferation (%GEM+) and viability, gene expression, and protein analysis to evaluate CDK4/6-cyclin D complex composition were examined. RESULTS Palbociclib + tamoxifen-resistant (PTxR) cells treated with abemaciclib + ET showed decreased %GEM+, %Ki67, and colony formation ability, compared to abemaciclib + tamoxifen-resistant (ATxR) cells treated with palbociclib + ET. Additionally, PTxR cells showed increased CDK4-p21 interaction, compared to ATxR. The CDK6 levels were greater in ATxR cells compared to PTxR cells, associated with CDK4/6i resistance. Additionally, abemaciclib + fulvestrant continued to robustly decrease pRb levels in PTxR models compared to palbociclib + fulvestrant in ATxR models. Transcriptome analysis revealed a depression of the cell cycle and E2F- and Rb-related genes in PTxR cells following treatment with abemaciclib + ET, not present in ATxR cells treated with palbociclib + ET. Both resistant models showed increased EGFR-related gene expression. CONCLUSION Taken together, we describe CDK4/6i-dependent mechanisms resulting in early-onset resistance to CDK4/6i + ET, using clinically relevant drug concentrations, in preclinical breast cancer cell models. The characterization of these preclinical models post progression on CDK4/6 inhibitor + ET treatment highlights the potential that the specific sequencing of CDK4/6 inhibitors could offer to overcome acquired resistance to CDK4/6i + ET. Abemaciclib + fulvestrant is currently under clinical investigation in patients with HR+, HER2- breast cancer and progression on prior CDK4/6i + ET (NCT05169567, postMONARCH).
Collapse
Affiliation(s)
| | - Yan Ding
- Eli Lilly and Company, Indianapolis, IN 46285, USA; (Y.D.); (N.P.); (A.d.D.)
| | - Nicholas Pulliam
- Eli Lilly and Company, Indianapolis, IN 46285, USA; (Y.D.); (N.P.); (A.d.D.)
| | - Alfonso de Dios
- Eli Lilly and Company, Indianapolis, IN 46285, USA; (Y.D.); (N.P.); (A.d.D.)
| | | | | |
Collapse
|
3
|
Carbone FP, Ancona P, Volinia S, Terrazzan A, Bianchi N. Druggable Molecular Networks in BRCA1/BRCA2-Mutated Breast Cancer. BIOLOGY 2025; 14:253. [PMID: 40136510 PMCID: PMC11940086 DOI: 10.3390/biology14030253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025]
Abstract
Mutations in the tumor suppressor genes BRCA1 and BRCA2 are associated with the triple-negative breast cancer phenotype, particularly aggressive and hard-to-treat tumors lacking estrogen, progesterone, and human epidermal growth factor receptor 2. This research aimed to understand the metabolic and genetic links behind BRCA1 and BRCA2 mutations and investigate their relationship with effective therapies. Using the Cytoscape software, two networks were generated through a bibliographic analysis of articles retrieved from the PubMed-NCBI database. We identified 98 genes deregulated by BRCA mutations, and 24 were modulated by therapies. In particular, BIRC5, SIRT1, MYC, EZH2, and CSN2 are influenced by BRCA1, while BCL2, BAX, and BRIP1 are influenced by BRCA2 mutation. Moreover, the study evaluated the efficacy of several promising therapies, targeting only BRCA1/BRCA2-mutated cells. In this context, CDDO-Imidazolide was shown to increase ROS levels and induce DNA damage. Similarly, resveratrol decreased the expression of the anti-apoptotic gene BIRC5 while it increased SIRT1 both in vitro and in vivo. Other specific drugs were found to induce apoptosis selectively in BRCA-mutated cells or block cell growth when the mutation occurs, i.e., 3-deazaneplanocin A, genistein or daidzein, and PARP inhibitors. Finally, over-representation analysis on the genes highlights ferroptosis and proteoglycan pathways as potential drug targets for more effective treatments.
Collapse
Affiliation(s)
- Francesca Pia Carbone
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
| | - Pietro Ancona
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
| | - Stefano Volinia
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
- Laboratory for Technologies of Advanced Therapies (LTTA), 44121 Ferrara, Italy
| | - Anna Terrazzan
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
- Laboratory for Technologies of Advanced Therapies (LTTA), 44121 Ferrara, Italy
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
| |
Collapse
|
4
|
Bhagwat SV, Mur C, Vandekopple M, Zhao B, Shen W, Marugán C, Capen A, Kindler L, Stephens JR, Huber L, Castanares MA, Garcia-Tapia D, Cohen JD, Bastian J, Mattioni B, Yuen E, Baker TK, Rodriguez Cruz V, Fei D, Manro JR, Pulliam N, Dowless MS, Ortiz Ruiz MJ, Yu C, Puca L, Klippel A, Bacchion F, Ismail-Khan R, Rodrik-Outmezguine V, Peng SB, Lallena MJ, Gong X, de Dios A. Imlunestrant Is an Oral, Brain-Penetrant Selective Estrogen Receptor Degrader with Potent Antitumor Activity in ESR1 Wild-Type and Mutant Breast Cancer. Cancer Res 2025; 85:777-790. [PMID: 39652577 PMCID: PMC11831106 DOI: 10.1158/0008-5472.can-24-2608] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/01/2024] [Accepted: 12/05/2024] [Indexed: 02/18/2025]
Abstract
Targeting of the estrogen receptor (ER) by antiestrogens is the standard of care for patients with ER+ HER2- advanced/metastatic breast cancer. Although antiestrogens that degrade ERα (fulvestrant) or block estrogen production (aromatase inhibitors) have improved patient outcomes, clinically important challenges remain related to drug administration, limited bioavailability, lack of brain exposure, and acquired resistance due to ESR1 mutations. These limitations indicate a need for more robust ER-targeted therapies. Here, we discovered and characterized imlunestrant, a next-generation potent, brain-penetrant oral selective ER degrader. Imlunestrant degraded ERα and decreased ERα-mediated gene expression both in vitro and in vivo. Cell proliferation and tumor growth in ESR1 wild-type (WT) and mutant models were significantly inhibited by imlunestrant. Combining imlunestrant with abemaciclib (CDK4/6 inhibitor), alpelisib (PI3K inhibitor), or everolimus (mTOR inhibitor) further enhanced tumor growth inhibition, regardless of ESR1 mutational status. In an ER+ breast cancer intracranial tumor model, imlunestrant prolonged survival compared with vehicle or alternative selective ER degrader therapies. Together, these findings support the potential of imlunestrant to degrade ERα and suppress the growth of ESR1-WT and mutant breast cancer, including brain metastatic tumors. Significance: Imlunestrant, a next-generation, brain-penetrant oral ERα degrader, displays potent activity in ESR1 wild-type and mutant breast cancer, enhances combination activity with standard-of-care agents, and inhibits growth of ER+ intracranial tumors.
Collapse
Affiliation(s)
| | | | | | - Baohui Zhao
- Eli Lilly and Company, Indianapolis, Indiana
| | - Weihua Shen
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | | | | | | | | | | | | | | | | | - Eunice Yuen
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | | | | | | | | | | | - Chunping Yu
- Lilly (China) Research and Development Co., Ltd., Shang Hai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Hao W, Jialong Z, Jiuzhi Y, Yang Y, Chongning L, Jincai L. ADP-ribosylation, a multifaceted modification: Functions and mechanisms in aging and aging-related diseases. Ageing Res Rev 2024; 98:102347. [PMID: 38815933 DOI: 10.1016/j.arr.2024.102347] [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: 02/02/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/01/2024]
Abstract
Aging, a complex biological process, plays key roles the development of multiple disorders referred as aging-related diseases involving cardiovascular diseases, stroke, neurodegenerative diseases, cancers, lipid metabolism-related diseases. ADP-ribosylation is a reversible modification onto proteins and nucleic acids to alter their structures and/or functions. Growing evidence support the importance of ADP-ribosylation and ADP-ribosylation-associated enzymes in aging and age-related diseases. In this review, we summarized ADP-ribosylation-associated proteins including ADP-ribosyl transferases, the ADP-ribosyl hydrolyses and ADP-ribose binding domains. Furthermore, we outlined the latest knowledge about regulation of ADP-ribosylation in the pathogenesis and progression of main aging-related diseases, organism aging and cellular senescence, and we also speculated the underlying mechanisms to better disclose this novel molecular network. Moreover, we discussed current issues and provided an outlook for future research, aiming to revealing the unknown bio-properties of ADP-ribosylation, and establishing a novel therapeutic perspective in aging-related diseases and health aging via targeting ADP-ribosylation.
Collapse
Affiliation(s)
- Wu Hao
- College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhao Jialong
- College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yuan Jiuzhi
- College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yu Yang
- College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Lv Chongning
- College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China; Liaoning Provincial Key Laboratory of TCM Resources Conservation and Development, Shenyang Pharmaceutical University, Shenyang, China
| | - Lu Jincai
- College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China; Liaoning Provincial Key Laboratory of TCM Resources Conservation and Development, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
6
|
Hany D, Zoetemelk M, Bhattacharya K, Nowak-Sliwinska P, Picard D. Network-informed discovery of multidrug combinations for ERα+/HER2-/PI3Kα-mutant breast cancer. Cell Mol Life Sci 2023; 80:80. [PMID: 36869202 PMCID: PMC10032341 DOI: 10.1007/s00018-023-04730-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/20/2023] [Accepted: 02/19/2023] [Indexed: 03/05/2023]
Abstract
Breast cancer is a persistent threat to women worldwide. A large proportion of breast cancers are dependent on the estrogen receptor α (ERα) for tumor progression. Therefore, targeting ERα with antagonists, such as tamoxifen, or estrogen deprivation by aromatase inhibitors remain standard therapies for ERα + breast cancer. The clinical benefits of monotherapy are often counterbalanced by off-target toxicity and development of resistance. Combinations of more than two drugs might be of great therapeutic value to prevent resistance, and to reduce doses, and hence, decrease toxicity. We mined data from the literature and public repositories to construct a network of potential drug targets for synergistic multidrug combinations. With 9 drugs, we performed a phenotypic combinatorial screen with ERα + breast cancer cell lines. We identified two optimized low-dose combinations of 3 and 4 drugs of high therapeutic relevance to the frequent ERα + /HER2-/PI3Kα-mutant subtype of breast cancer. The 3-drug combination targets ERα in combination with PI3Kα and cyclin-dependent kinase inhibitor 1 (p21). In addition, the 4-drug combination contains an inhibitor for poly (ADP-ribose) polymerase 1 (PARP1), which showed benefits in long-term treatments. Moreover, we validated the efficacy of the combinations in tamoxifen-resistant cell lines, patient-derived organoids, and xenograft experiments. Thus, we propose multidrug combinations that have the potential to overcome the standard issues of current monotherapies.
Collapse
Affiliation(s)
- Dina Hany
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, 1211, Genève 4, Switzerland
- On leave from: Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, 21311, Egypt
| | - Marloes Zoetemelk
- Groupe de Pharmacologie Moléculaire, Section des Sciences Pharmaceutiques, Université de Genève, Genève, Switzerland
- Institut des Sciences Pharmaceutiques de Suisse Occidentale, Université de Genève, Genève, Switzerland
- Centre de Recherche Translationnelle en Onco-hématologie, Université de Genève, Genève, Switzerland
| | - Kaushik Bhattacharya
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, 1211, Genève 4, Switzerland
| | - Patrycja Nowak-Sliwinska
- Groupe de Pharmacologie Moléculaire, Section des Sciences Pharmaceutiques, Université de Genève, Genève, Switzerland
- Institut des Sciences Pharmaceutiques de Suisse Occidentale, Université de Genève, Genève, Switzerland
- Centre de Recherche Translationnelle en Onco-hématologie, Université de Genève, Genève, Switzerland
| | - Didier Picard
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, 1211, Genève 4, Switzerland.
| |
Collapse
|
7
|
Zhu T, Zheng JY, Huang LL, Wang YH, Yao DF, Dai HB. Human PARP1 substrates and regulators of its catalytic activity: An updated overview. Front Pharmacol 2023; 14:1137151. [PMID: 36909172 PMCID: PMC9995695 DOI: 10.3389/fphar.2023.1137151] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) is a key DNA damage sensor that is recruited to damaged sites after DNA strand breaks to initiate DNA repair. This is achieved by catalyzing attachment of ADP-ribose moieties, which are donated from NAD+, on the amino acid residues of itself or other acceptor proteins. PARP inhibitors (PARPi) that inhibit PARP catalytic activity and induce PARP trapping are commonly used for treating BRCA1/2-deficient breast and ovarian cancers through synergistic lethality. Unfortunately, resistance to PARPi frequently occurs. In this review, we present the novel substrates and regulators of the PARP1-catalyzed poly (ADP-ribosyl)ation (PARylatison) that have been identified in the last 3 years. The overall aim is the presentation of protein interactions of potential therapeutic intervention for overcoming the resistance to PARPi.
Collapse
Affiliation(s)
- Tao Zhu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ju-Yan Zheng
- Institute of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Ling Huang
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan-Hong Wang
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Di-Fei Yao
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hai-Bin Dai
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
8
|
Liu J, Wang Q, Kang Y, Xu S, Pang D. Unconventional protein post-translational modifications: the helmsmen in breast cancer. Cell Biosci 2022; 12:22. [PMID: 35216622 PMCID: PMC8881842 DOI: 10.1186/s13578-022-00756-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/07/2022] [Indexed: 01/10/2023] Open
Abstract
AbstractBreast cancer is the most prevalent malignant tumor and a leading cause of mortality among females worldwide. The tumorigenesis and progression of breast cancer involve complex pathophysiological processes, which may be mediated by post-translational modifications (PTMs) of proteins, stimulated by various genes and signaling pathways. Studies into PTMs have long been dominated by the investigation of protein phosphorylation and histone epigenetic modifications. However, with great advances in proteomic techniques, several other PTMs, such as acetylation, glycosylation, sumoylation, methylation, ubiquitination, citrullination, and palmitoylation have been confirmed in breast cancer. Nevertheless, the mechanisms, effects, and inhibitors of these unconventional PTMs (particularly, the non-histone modifications other than phosphorylation) received comparatively little attention. Therefore, in this review, we illustrate the functions of these PTMs and highlight their impact on the oncogenesis and progression of breast cancer. Identification of novel potential therapeutic drugs targeting PTMs and development of biological markers for the detection of breast cancer would be significantly valuable for the efficient selection of therapeutic regimens and prediction of disease prognosis in patients with breast cancer.
Collapse
|
9
|
Asghari A, Wall K, Gill M, Vecchio ND, Allahbakhsh F, Wu J, Deng N, Zheng WJ, Wu H, Umetani M, Maroufy V. A novel group of genes that cause endocrine resistance in breast cancer identified by dynamic gene expression analysis. Oncotarget 2022; 13:600-613. [PMID: 35401937 PMCID: PMC8986262 DOI: 10.18632/oncotarget.28225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/25/2022] [Indexed: 11/25/2022] Open
Abstract
Breast cancer (BC) is the most common type of cancer diagnosed in women. Among female cancer deaths, BC is the second leading cause of death worldwide. For estrogen receptor-positive (ER-positive) breast cancers, endocrine therapy is an effective therapeutic approach. However, in many cases, an ER-positive tumor becomes unresponsive to endocrine therapy, and tumor regrowth occurs after treatment. While some genetic mutations contribute to resistance in some patients, the underlying causes of resistance to endocrine therapy are mostly undetermined. In this study, we utilized a recently developed statistical approach to investigate the dynamic behavior of gene expression during the development of endocrine resistance and identified a novel group of genes whose time course expression significantly change during cell modelling of endocrine resistant BC development. Expression of a subset of these genes was also differentially expressed in microarray analysis of endocrine-resistant and endocrine-sensitive tumor samples. Surprisingly, a subset of those genes was also differentially genes expressed in triple-negative breast cancer (TNBC) as compared with ER-positive BC. The findings suggest shared genetic mechanisms may underlie the development of endocrine resistant BC and TNBC. Our findings identify 34 novel genes for further study as potential therapeutic targets for treatment of endocrine-resistant BC and TNBC.
Collapse
Affiliation(s)
- Arvand Asghari
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
- These authors contributed equally to this work
| | - Katherine Wall
- Department of Biostatistics and Data Science, School of Public Health, UTHealth, Houston, TX 77030, USA
- These authors contributed equally to this work
| | - Michael Gill
- Department of Biostatistics and Data Science, School of Public Health, UTHealth, Houston, TX 77030, USA
| | - Natascha Del Vecchio
- Chicago Center for HIV Elimination, University of Chicago, Chicago, IL 60637, USA
| | - Farnaz Allahbakhsh
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Jacky Wu
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Nan Deng
- Clinical Cancer Prevention Department, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - W. Jim Zheng
- School of Biomedical Informatics, UTHealth, Houston, TX 77030, USA
| | - Hulin Wu
- Department of Biostatistics and Data Science, School of Public Health, UTHealth, Houston, TX 77030, USA
| | - Michihisa Umetani
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
- Health Research Institute, University of Houston, Houston, TX 77204, USA
| | - Vahed Maroufy
- Department of Biostatistics and Data Science, School of Public Health, UTHealth, Houston, TX 77030, USA
| |
Collapse
|
10
|
Tolaney SM, Toi M, Neven P, Sohn J, Grischke EM, Llombart-Cussac A, Soliman H, Wang H, Wijayawardana S, Jansen VM, Litchfield LM, Sledge GW. Clinical Significance of PIK3CA and ESR1 Mutations in circulating tumor DNA: Analysis from the MONARCH 2 Study of Abemaciclib Plus Fulvestrant. Clin Cancer Res 2022; 28:1500-1506. [PMID: 35121623 DOI: 10.1158/1078-0432.ccr-21-3276] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/17/2021] [Accepted: 01/31/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND PIK3CA and ESR1 mutations have been implicated in resistance to endocrine therapy (ET) in HR+, HER2- advanced breast cancer (ABC). Inhibition of CDK4&6 has been hypothesized as a therapeutic strategy to overcome endocrine resistance in patients with PIK3CA- or ESR1-mutant breast cancers. The objective of this exploratory analysis was to assess efficacy of abemaciclib plus fulvestrant in patients with or without PIK3CA or ESR1 mutations in MONARCH 2. PATIENTS AND METHODS MONARCH 2 was a global, randomized, double-blind Phase 3 trial of abemaciclib plus fulvestrant in women with HR+, HER2- ABC that had progressed on ET. Patients were randomized 2:1 to receive abemaciclib plus fulvestrant or placebo plus fulvestrant. Exploratory analyses assessed progression-free survival (PFS) and overall survival (OS), and other endpoints, in patients with or without PIK3CA or ESR1 mutations detectable in baseline ctDNA. RESULTS Abemaciclib plus fulvestrant improved PFS compared to placebo plus fulvestrant in both PIK3CA-wild-type and PIK3CA-mutant subgroups, as well as both ESR1-wild-type and ESR1-mutant subgroups. Additional endpoints, including OS, were also improved following treatment with abemaciclib plus fulvestrant regardless of PIK3CA or ESR1 mutation status. CONCLUSION Abemaciclib plus fulvestrant was effective regardless of PIK3CA or ESR1 mutation status, with benefit in both PFS and OS, with a numerically greater improvement in median PFS relative to placebo plus fulvestrant for PIK3CA or ESR1-mutant tumors compared to the respective wild-type subgroups, in women with HR+, HER2- ABC that had progressed on ET.
Collapse
Affiliation(s)
- Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute
| | | | | | - Joohyuk Sohn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine
| | - Eva-Maria Grischke
- gynecology, Universitӓts Frauenklinik Tubingen, Eberhard Karls University
| | | | - Hatem Soliman
- Department of Breast Oncology, Moffitt Cancer Center
| | | | | | | | | | | |
Collapse
|
11
|
Kupczyk P, Simiczyjew A, Marczuk J, Dratkiewicz E, Beberok A, Rok J, Pieniazek M, Biecek P, Nevozhay D, Slowikowski B, Chodaczek G, Wrzesniok D, Nowak D, Donizy P. PARP1 as a Marker of an Aggressive Clinical Phenotype in Cutaneous Melanoma-A Clinical and an In Vitro Study. Cells 2021; 10:286. [PMID: 33572647 PMCID: PMC7911865 DOI: 10.3390/cells10020286] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 01/12/2023] Open
Abstract
(1) Background: Poly(ADP-ribose) polymerase 1) (PARP1) is a pleiotropic enzyme involved in several cellular processes, e.g., DNA damage repair, regulation of mitosis, and immune response. Little is known about the role of PARP1 in melanoma development and progression. We aimed to investigate the prognostic significance of PARP1 expression in cutaneous melanoma through evaluation of mRNA and protein levels of PARP1 in normal melanocytes and melanoma cell lines, as well as in patients' tissue material from surgical resections. (2) Methods: An in vitro model was based on two types of normal human melanocytes (HEMn-DP and HEMn-LP) and four melanoma cell lines (A375, WM1341D, Hs294T, and WM9). PARP1 mRNA gene expression was estimated using real-time polymerase chain reaction (RT-PCR), whereas the protein level of PARP1 was evaluated by fluorescence confocal microscopy and then confirmed by Western Blotting analysis. The expression of PARP1 was also assessed by immunohistochemistry in formalin-fixed paraffin-embedded tissues of 128 primary cutaneous melanoma patients and correlated with follow-up and clinicopathologic features. (3) Results: The in vitro study showed that melanoma cells exhibited significantly higher PARP1 expression at mRNA and protein levels than normal melanocytes. High PARP1 expression was also associated with the invasiveness of tumor cells. Elevated nuclear PARP1 expression in patients without nodal metastases strongly correlated with significantly shorter disease-free survival (p = 0.0015) and revealed a trend with shorter cancer-specific overall survival (p = 0.05). High PARP1 immunoreactivity in the lymph node-negative group of patients was significantly associated with higher Breslow tumor thickness, presence of ulceration, and a higher mitotic index (p = 0.0016, p = 0.023, and p < 0.001, respectively). In patients with nodal metastases, high PARP1 expression significantly correlated with the presence of microsatellitosis (p = 0.034), but we did not confirm the prognostic significance of PARP1 expression in these patients. In the entire analyzed group of patients (with and without nodal metastases at the time of diagnosis), PARP1 expression was associated with a high mitotic index (p = 0.001) and the presence of ulceration (p = 0.036). Moreover, in patients with elevated PARP1 expression, melanoma was more frequently located in the skin of the head and neck region (p = 0.015). In multivariate analysis, high PARP1 expression was an independent unfavorable prognosticator in lymph node-negative cutaneous melanoma patients. (4) Conclusions: In vitro molecular biology approaches demonstrated enhanced PARP1 expression in cutaneous melanoma. These results were confirmed by the immunohistochemical study with clinical parameter analysis, which showed that a high level of PARP1 correlated with unfavorable clinical outcome. These observations raise the potential role of PARP1 inhibitor-based therapy in cutaneous melanoma.
Collapse
Affiliation(s)
- Piotr Kupczyk
- Department of Pathomorphology, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Aleksandra Simiczyjew
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.S.); (E.D.); (D.N.)
| | - Jakub Marczuk
- Department of Dermatology, Research and Development Center, Regional Specialized Hospital, 51-124 Wroclaw, Poland;
| | - Ewelina Dratkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.S.); (E.D.); (D.N.)
| | - Artur Beberok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.B.); (J.R.); (D.W.)
| | - Jakub Rok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.B.); (J.R.); (D.W.)
| | - Malgorzata Pieniazek
- Department of Clinical Oncology, Tadeusz Koszarowski Regional Oncology Centre, 45-061 Opole, Poland;
| | - Przemyslaw Biecek
- Faculty of Mathemathics and Information Science, Warsaw University of Technology, 00-662 Warsaw, Poland;
| | - Dmitry Nevozhay
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- School of Biomedicine, Far Eastern Federal University, 690950 Vladivostok, Russia
| | - Bartosz Slowikowski
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| | - Grzegorz Chodaczek
- Laboratory of Bioimaging, Łukasiewicz Research Network—PORT Polish Center for Technology Development, 54-066 Wroclaw, Poland;
| | - Dorota Wrzesniok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.B.); (J.R.); (D.W.)
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (A.S.); (E.D.); (D.N.)
| | - Piotr Donizy
- Department of Pathomorphology and Oncological Cytology, Wroclaw Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
12
|
Conrad LB, Lin KY, Nandu T, Gibson BA, Lea JS, Kraus WL. ADP-Ribosylation Levels and Patterns Correlate with Gene Expression and Clinical Outcomes in Ovarian Cancers. Mol Cancer Ther 2019; 19:282-291. [PMID: 31594824 DOI: 10.1158/1535-7163.mct-19-0569] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/13/2019] [Accepted: 09/20/2019] [Indexed: 12/31/2022]
Abstract
Inhibitors of nuclear PARP enzymes (e.g., PARP-1) have improved clinical outcomes in ovarian cancer, especially in patients with BRCA1/2 gene mutations or additional homologous recombination (HR) DNA repair pathway deficiencies. These defects serve as biomarkers for response to PARP inhibitors (PARPi). We sought to identify an additional biomarker that could predict responses to both conventional chemotherapy and PARPi in ovarian cancers. We focused on cellular ADP-ribosylation (ADPRylation), which is catalyzed by PARP enzymes and detected by detection reagents we developed previously. We determined molecular phenotypes of 34 high-grade serous ovarian cancers and associated them with clinical outcomes. We used the levels and patterns of ADPRylation and PARP-1 to distribute ovarian cancers into distinct molecular phenotypes, which exhibit dramatically different gene expression profiles. In addition, the levels and patterns of ADPRylation, PARP-1 protein, and gene expression correlated with clinical outcomes in response to platinum-based chemotherapy, with cancers exhibiting the highest levels of ADPRylation having the best outcomes independent of BRCA1/2 status. Finally, in cell culture-based assays using patient-derived ovarian cancer cell lines, ADPRylation levels correlated with sensitivity to the PARPi, Olaparib, with cell lines exhibiting high levels of ADPRylation having greater sensitivity to Olaparib. Collectively, our study demonstrates that ovarian cancers exhibit a wide range of ADPRylation levels, which correlate with therapeutic responses and clinical outcomes. These results suggest ADPRylation may be a useful biomarker for PARPi sensitivity in ovarian cancers, independent of BRCA1/2 or homologous recombination deficiency status.
Collapse
Affiliation(s)
- Lesley B Conrad
- Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas.,Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas.,Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ken Y Lin
- Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas.,Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas.,Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tulip Nandu
- Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas.,Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bryan A Gibson
- Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas.,Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jayanthi S Lea
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - W Lee Kraus
- Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas. .,Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|