1
|
González-Conde M, Yáñez C, Abuín C, Keup C, Lago-Lestón R, Aybar M, Pedrouzo L, Palacios P, Curiel T, Cueva J, Rodríguez C, Carmona M, Cortegoso A, García-Caballero T, Muinelo-Romay L, Kasimir-Bauer S, López-López R, Costa C. Gene expression analysis in circulating tumour cells to determine resistance to CDK4/6 inhibitors plus endocrine therapy in HR + /HER2- metastatic breast cancer patients. J Transl Med 2025; 23:400. [PMID: 40186268 PMCID: PMC11971781 DOI: 10.1186/s12967-025-06374-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/08/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Metastatic breast cancer (BC) is the main cause of cancer-related mortality in women worldwide. HR + /HER2- BC patients are treated with endocrine therapy (ET), but therapeutic resistance is common. The combination of cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) with ET was approved for metastatic BC patients and extended the median progression-free survival to 24 months. This therapy is not always effective, and in every patient, resistance ultimately occurs, but the underlying resistance mechanisms remain unclear. To address this gap, we explored circulating tumour cells (CTCs) as biomarkers to assess treatment response and resistance in metastatic HR + /HER2- BC patients receiving CDK4/6i plus ET. METHODS In total, 53 HR + /HER2- metastatic BC patients who received a CDK4/6i plus ET as first-line treatment were analysed, including samples from internal and external validation cohorts. CTCs were isolated using the negative enrichment approach RosetteSep (STEMCELL Technologies) or positive immunomagnetic selection targeting EpCAM, EGFR, and HER2 (AdnaTest EMT-2/StemCell Select™, QIAGEN). RNA was extracted from CTCs and PBMCs for nCounter analysis (Pancancer pathways panel) in a discovery phase. Subsequent validation was performed by RT-qPCR. RESULTS CTC gene expression analysis revealed that non responder patients (those who experienced disease progression before 180 days) exhibited elevated PRKCB (p-value: 0.011), MAPK3 (p-value: 0.006) and STAT3 (p-value: 0.008) expression, while responders showed increased CDK6 (p-value: 0.011) and CCND1 (p-value: 0.035) expression at baseline. CTC transcriptional characterization revealed a gene expression signature (STAT3highPRKCBhighCDK6low) that accurately classified HR + /HER2- metastatic BC patients who responded to CDK4/6i plus ET, regardless of the CTC isolation method (AUC > 0.8). CTC characterization at progression also identified biomarkers linked to therapy resistance, including the epigenetic regulators EZH2 and HDAC6 and the cell cycle regulator CDC7, which could guide the selection of subsequent therapy lines. The expression of the CDK4 and STAT3 genes in CTCs was associated with progression-free survival and overall survival, respectively. Likewise, the presence of ≥ one CTC after one cycle of therapy predicts a worse prognosis. CONCLUSIONS CTC gene expression provides information about treatment outcomes in HR + /HER2- metastatic BC patients receiving CDK4/6i plus ET and could guide personalized strategies and improve prognosis.
Collapse
MESH Headings
- Humans
- Neoplastic Cells, Circulating/metabolism
- Neoplastic Cells, Circulating/pathology
- Neoplastic Cells, Circulating/drug effects
- Breast Neoplasms/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/blood
- Female
- Cyclin-Dependent Kinase 6/antagonists & inhibitors
- Cyclin-Dependent Kinase 6/metabolism
- Cyclin-Dependent Kinase 4/antagonists & inhibitors
- Cyclin-Dependent Kinase 4/metabolism
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Receptor, ErbB-2/metabolism
- Neoplasm Metastasis
- Middle Aged
- Gene Expression Regulation, Neoplastic/drug effects
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Aged
- Gene Expression Profiling
- Adult
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Antineoplastic Agents, Hormonal/therapeutic use
Collapse
Affiliation(s)
- Miriam González-Conde
- Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Celso Yáñez
- Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Carmen Abuín
- Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Corinna Keup
- Department of Gynaecology and Obstetrics, University Hospital Essen, 45147, Essen, Germany
| | - Ramón Lago-Lestón
- Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Liquid Biopsy Analysis Unit, Oncomet, Santiago de Compostela, Spain
| | - Maribel Aybar
- Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Lucía Pedrouzo
- Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Patricia Palacios
- Department of Oncology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain
| | - Teresa Curiel
- Department of Oncology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain
| | - Juan Cueva
- Department of Oncology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain
| | - Carmela Rodríguez
- Department of Oncology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain
| | - Marta Carmona
- Department of Oncology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain
| | - Alexandra Cortegoso
- Department of Oncology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain
| | - Tomás García-Caballero
- Department of Morphological Sciences, University of Santiago de Compostela. Health Research Institute of Santiago, Santiago de Compostela, Spain
| | - Laura Muinelo-Romay
- Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, Madrid, Spain
- Liquid Biopsy Analysis Unit, Oncomet, Santiago de Compostela, Spain
| | - Sabine Kasimir-Bauer
- Department of Gynaecology and Obstetrics, University Hospital Essen, 45147, Essen, Germany
| | - Rafael López-López
- Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.
- Department of Oncology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain.
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, Madrid, Spain.
| | - Clotilde Costa
- Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, Madrid, Spain.
| |
Collapse
|
2
|
Liu ZY, Liu RT, Cheng WH, Zhang BY, Zhang XY, Zhou Y, Ye XQ, Zhou CY, Wang XJ, Sun Q, Ji J. Neratinib derivative 7A induces apoptosis in colon cancer cells via the p53 pathway. Bioorg Med Chem Lett 2025; 117:130069. [PMID: 39674380 DOI: 10.1016/j.bmcl.2024.130069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/16/2024]
Abstract
Colorectal cancer remains a significant health threat, with its incidence continuously rising, underscoring the urgent need for the development of new therapeutic agents. In our previous research, we identified 7A, a derivative of Neratinib, as having pronounced antitumor activity. However, its specific effects and mechanisms in colorectal cancer have not been thoroughly investigated. Therefore, this study employed in vivo and in vitro experiments, utilizing techniques such as RNA sequencing, Western blotting, and PCR, to provide a comprehensive analysis of 7A's mechanism of action in colorectal cancer. The results indicate that 7A induces DNA damage and activates the P53 pathway, thereby promoting apoptosis in colorectal cancer cells. Additionally, 7A treatment significantly reduced angiogenesis and tumor weight. Our findings suggest that 7A, a Neratinib derivative, holds promise as a novel candidate for colorectal cancer therapy.
Collapse
Affiliation(s)
- Zhi-Yu Liu
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Ruo-Tong Liu
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Wen-Hao Cheng
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Bo-Yu Zhang
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Xing-Yu Zhang
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Ying Zhou
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Xiao-Qing Ye
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Chun-Yun Zhou
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China
| | - Xiu-Jun Wang
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China.
| | - Qian Sun
- The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University. Lianyungang 222000, China.
| | - Jing Ji
- College of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, China; College of Pharmacy and Chemistry and Chemical Engineering, Taizhou University, Taizhou, 225 300, China.
| |
Collapse
|
3
|
Miniati M, Conversano C, Palagini L, Buccianelli B, Fabrini M, Mancino M, Laliscia C, Marazziti D, Paiar F, Gemignani A. Bipolar Disorder Treatments and Ovarian Cancer: A Systematic Review. CLINICAL NEUROPSYCHIATRY 2020; 17:300-313. [PMID: 34909008 PMCID: PMC8629050 DOI: 10.36131/cnfioritieditore20200508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
OBJECTIVE We reviewed literature on drugs for bipolar disorders (BD), utilized in ovarian cancer (OC). METHOD We adhered to the Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) guidelines in completion of this systematic review. RESULTS We identified 73 papers. Thirty-two studies were finally included. BD is rarely diagnosed in OC patients. Limited finding from case reports is available. Drugs used to treat BD (mainly lithium and valproic acid) have been extensively studied in add-on to chemotherapy for treatment-resistant OC cells or in animal models, with promising results in vitro but not in vivo. CONCLUSIONS The clinical underestimation of BD in OC has leaded to the almost complete absence of evidences for a soundly based clinical guidance in this field. There is a urgent need for a systematic multi-disciplinary approach to OC.
Collapse
Affiliation(s)
- Mario Miniati
- Department of Clinical and Experimental Medicine, University of Pisa, 57 Via Roma, Italy,(E-MAIL:)
| | - Ciro Conversano
- Department of Surgical Pathology, Medical, Molecular and Critical Area, University of Pisa, 57 Via Roma, Pisa, Italy,(E-MAIL:)
| | - Laura Palagini
- Corresponding author Laura Palagini, M.D., Ph.D. Department of Clinical and Experimental Medicine, University of Pisa 57 Via Roma, Pisa, Italy E-mail:
| | | | - Mariagrazia Fabrini
- Department of Radiotherapy, University of Pisa, 57 Via Roma, Pisa, Italy,(E-MAIL:)
| | - Maricia Mancino
- Department of Radiotherapy, University of Pisa, 57 Via Roma, Pisa, Italy,(E-MAIL:)
| | - Concetta Laliscia
- Department of Radiotherapy, University of Pisa, 57 Via Roma, Pisa, Italy,(E-MAIL:)
| | | | - Fabiola Paiar
- Department of Radiotherapy, University of Pisa, 57 Via Roma, Pisa, Italy,(E-MAIL:)
| | - Angelo Gemignani
- Department of Surgical Pathology, Medical, Molecular and Critical Area, University of Pisa, 57 Via Roma, Pisa, Italy,(E-MAIL:)
| |
Collapse
|
4
|
Knudsen ES, Shapiro GI, Keyomarsi K. Selective CDK4/6 Inhibitors: Biologic Outcomes, Determinants of Sensitivity, Mechanisms of Resistance, Combinatorial Approaches, and Pharmacodynamic Biomarkers. Am Soc Clin Oncol Educ Book 2020; 40:115-126. [PMID: 32421454 PMCID: PMC7306922 DOI: 10.1200/edbk_281085] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
CDK4/6 inhibitors are now part of the standard armamentarium for hormone receptor-positive breast cancer. In this article, we review the biologic outcomes imposed by these drugs on cancer cells, determinants of response, mechanisms of intrinsic and acquired resistance, as well as combinatorial approaches emanating from mechanistic studies that may allow use of these agents to extend beyond breast cancer. In addition, we will address tumor-, imaging-, and blood-based pharmacodynamic biomarkers that can inform rationally designed trials as clinical development continues.
Collapse
Affiliation(s)
- Erik S. Knudsen
- Center for Personalized Medicine and Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY
| | - Geoffrey I. Shapiro
- Early Drug Development Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Khandan Keyomarsi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
5
|
Dent P, Booth L, Roberts JL, Poklepovic A, Hancock JF. Fingolimod Augments Monomethylfumarate Killing of GBM Cells. Front Oncol 2020; 10:22. [PMID: 32047722 PMCID: PMC6997152 DOI: 10.3389/fonc.2020.00022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/08/2020] [Indexed: 12/26/2022] Open
Abstract
Previously we demonstrated that the multiple sclerosis drug dimethyl fumarate (DMF) and its plasma breakdown product MMF could interact with chemotherapeutic agents to kill both GBM cells and activated microglia. The trial NCT02337426 demonstrated the safety of DMF in newly diagnosed GBM patients when combined with the standard of care Stupp protocol. We hypothesized that another multiple sclerosis drug, fingolimod (FTY720) would synergize with MMF to kill GBM cells. MMF and fingolimod interacted in a greater than additive fashion to kill PDX GBM isolates. MMF and fingolimod radiosensitized glioma cells and enhanced the lethality of temozolomide. Exposure to [MMF + fingolimod] activated an ATM-dependent toxic autophagy pathway, enhanced protective endoplasmic reticulum stress signaling, and inactivated protective PI3K, STAT, and YAP function. The drug combination reduced the expression of protective c-FLIP-s, MCL-1, BCL-XL, and in parallel caused cell-surface clustering of the death receptor CD95. Knock down of CD95 or over-expression of c-FLIP-s or BCL-XL suppressed killing. Fingolimod and MMF interacted in a greater than additive fashion to rapidly enhance reactive oxygen species production and over-expression of either thioredoxin or super-oxide dismutase two significantly reduced the drug-induced phosphorylation of ATM, autophagosome formation and [MMF + fingolimod] lethality. In contrast, the production of ROS was only marginally reduced in cells lacking ATM, CD95, or Beclin1. Collectively, our data demonstrate that the primary generation of ROS by [MMF + fingolimod] plays a key role, via the induction of toxic autophagy and death receptor signaling, in the killing of GBM cells.
Collapse
Affiliation(s)
- Paul Dent
- Departments of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Laurence Booth
- Departments of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Jane L Roberts
- Departments of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Andrew Poklepovic
- Departments of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - John F Hancock
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, United States
| |
Collapse
|
6
|
Booth L, Roberts JL, West C, Von Hoff D, Dent P. GZ17-6.02 initiates DNA damage causing autophagosome-dependent HDAC degradation resulting in enhanced anti-PD1 checkpoint inhibitory antibody efficacy. J Cell Physiol 2020; 235:8098-8113. [PMID: 31951027 DOI: 10.1002/jcp.29464] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/08/2020] [Indexed: 12/20/2022]
Abstract
Our studies examined the molecular mechanisms by which the novel cancer therapeutic GZ17-6.02 (NCT03775525) killed GI tumor cells. TZ17-6.02 activated ATM which was responsible for increased phosphorylation of nuclear γH2AX and AMPKα T172. ATM-AMPK signaling was responsible for the subsequent inactivation of mTORC1 and mTORC2, dephosphorylation of ULK1 S757, and increased phosphorylation of ULK1 S317 and of ATG13 S318, which collectively caused enhanced autophagosome formation. GZ17-6.02 interacted with 5-fluorouracil in an additive to greater than additive fashion to kill all of the tested GI tumor cell types. This was associated with greater ATM activation and a greater mammalian target of rapamycin inactivation and autophagosome induction. As a result, autophagy-dependent degradation of multiple histone deacetylase (HDAC) proteins and chaperone proteins occurred. Loss of HDAC expression was causal in reduced expression of programed death ligand 1 (PD-L1), ornithine decarboxylase, and indole amine 2,3-dioxygenase (IDO1) and in the elevated expression of major histocompatibility complex Class IA (MHCA). Treatment with GZ17-6.02 also resulted in enhanced efficacy of a subsequently administered anti-PD1 checkpoint inhibitory antibody. Thus, the primary mode of GZ17-6.02 action is to induce a DNA damage response concomitant with ATM activation, that triggers a series of interconnected molecular events that result in tumor cell death and enhanced immunogenicity.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Jane L Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | | | - Daniel Von Hoff
- Translational Genomics Research Institute (TGEN), Phoenix, Arizona
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
7
|
Booth L, Poklepovic A, Dent P. Not the comfy chair! Cancer drugs that act against multiple active sites. Expert Opin Ther Targets 2019; 23:893-901. [PMID: 31709855 DOI: 10.1080/14728222.2019.1691526] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Discoveries of novel signal transduction pathways in the 1990s stimulated drug companies to develop small molecule tyrosine kinase and serine / threonine kinase inhibitors which were based on catalytic site inhibition. All kinases bind ATP and catalyze phosphate transfer and, therefore, inhibitors that block ATP binding and its metabolism would be predicted to have a known on-target specificity but were also likely to have many unknown or unrecognized targets due to similarities in all ATP binding pockets. This on-target off-target biology of kinase inhibitors, which exhibit a "signal" in the clinic, means that therapeutically valuable agents are acting through unknown biological processes to mediate their anti-tumor effects.Areas covered: This perspective discusses drug therapies whose actions cannot be explained by their actions on the original targeted kinase; it concludes with a methodology to screen for changes in cell signaling via in-cell western immunoblotting.Expert opinion: Most malignancies do not depend on survival signaling from one specific mutated proto-oncogene, especially for previously treated malignancies where multiple clonal variants of the primary tumor have evolved. Hence, the concept of a highly "personalized medicine" approach fails because it is unlikely that a specific therapy will kill all clonal variants of the tumor.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
8
|
Preclinical Characteristics of the Irreversible Pan-HER Kinase Inhibitor Neratinib Compared with Lapatinib: Implications for the Treatment of HER2-Positive and HER2-Mutated Breast Cancer. Cancers (Basel) 2019; 11:cancers11060737. [PMID: 31141894 PMCID: PMC6628314 DOI: 10.3390/cancers11060737] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 12/13/2022] Open
Abstract
An estimated 15–20% of breast cancers overexpress human epidermal growth factor receptor 2 (HER2/ERBB2/neu). Two small-molecule tyrosine kinase inhibitors (TKIs), lapatinib and neratinib, have been approved for the treatment of HER2-positive (HER2+) breast cancer. Lapatinib, a reversible epidermal growth factor receptor (EGFR/ERBB1/HER1) and HER2 TKI, is used for the treatment of advanced HER2+ breast cancer in combination with capecitabine, in combination with trastuzumab in patients with hormone receptor-negative metastatic breast cancer, and in combination with an aromatase inhibitor for the first-line treatment of HER2+ breast cancer. Neratinib, a next-generation, irreversible pan-HER TKI, is used in the US for extended adjuvant treatment of adult patients with early-stage HER2+ breast cancer following 1 year of trastuzumab. In Europe, neratinib is used in the extended adjuvant treatment of adult patients with early-stage hormone receptor-positive HER2+ breast cancer who are less than 1 year from the completion of prior adjuvant trastuzumab-based therapy. Preclinical studies have shown that these agents have distinct properties that may impact their clinical activity. This review describes the preclinical characterization of lapatinib and neratinib, with a focus on the differences between these two agents that may have implications for patient management.
Collapse
|
9
|
Booth L, Roberts JL, Sander C, Lalani AS, Kirkwood JM, Hancock JF, Poklepovic A, Dent P. Neratinib and entinostat combine to rapidly reduce the expression of K-RAS, N-RAS, Gα q and Gα 11 and kill uveal melanoma cells. Cancer Biol Ther 2018; 20:700-710. [PMID: 30571927 DOI: 10.1080/15384047.2018.1551747] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
There is no efficacious standard of care therapy for uveal melanoma. Unlike cutaneous disease, uveal melanoma does not exhibit RAS mutations but instead contains mutations with ~90% penetrance in either Gαq or Gα11. Previously we demonstrated that neratinib caused ERBB1/2/4 and RAS internalization into autolysosomes which resulted in their proteolytic degradation. In PDX isolates of uveal melanoma, neratinib caused the internalization and degradation of Gαq and Gα11 in parallel with ERBB1 breakdown. These effects were enhanced by the HDAC inhibitor entinostat. Similar data were obtained using GFP/RFP tagged forms of K-RAS V12. Down regulation of Gαq and Gα11 expression and RAS-GFP/RFP fluorescence required Beclin1 and ATG5. The [neratinib + entinostat] combination engaged multiple pathways to mediate killing. One was from ROS-dependent activation of ATM via AMPK-ULK1-ATG13-Beclin1/ATG5. Another pathway was from CD95 via caspase 8-RIP1/RIP3. A third was from reduced expression of HSP70, HSP90, HDAC6 and phosphorylation of eIF2α. Downstream of the mitochondrion both caspase 9 and AIF played roles in tumor cell execution. Knock down of ATM/AMPK/ULK-1 prevented ATG13 phosphorylation and degradation of RAS and Gα proteins. Over-expression of activated mTOR prevented ATG13 phosphorylation and suppressed killing. Knock down of eIF2α maintained BCL-XL and MCL-1 expression. Within 6h, [neratinib + entinostat] reduced the expression of the immunology biomarkers PD-L1, ODC, IDO-1 and enhanced MHCA levels. Our data demonstrate that [neratinib + entinostat] down-regulates oncogenic RAS and the two key oncogenic drivers present in most uveal melanoma patients and causes a multifactorial form of killing via mitochondrial dysfunction and toxic autophagy. Abbreviations: ERK: extracellular regulated kinase; PI3K: phosphatidyl inositol 3 kinase; ca: constitutively active; dn: dominant negative; ER: endoplasmic reticulum; AIF: apoptosis inducing factor; AMPK: AMP-dependent protein kinase; mTOR: mammalian target of rapamycin; JAK: Janus Kinase; STAT: Signal Transducers and Activators of Transcription; MAPK: mitogen activated protein kinase; PTEN: phosphatase and tensin homologue on chromosome ten; ROS: reactive oxygen species; CMV: empty vector plasmid or virus; si: small interfering; SCR: scrambled; IP: immunoprecipitation; VEH: vehicle; HDAC: histone deacetylase.
Collapse
Affiliation(s)
- Laurence Booth
- a Departments of Biochemistry and Molecular Biology , Virginia Commonwealth University , Richmond , VA , USA
| | - Jane L Roberts
- a Departments of Biochemistry and Molecular Biology , Virginia Commonwealth University , Richmond , VA , USA
| | - Cindy Sander
- b Melanoma and Skin Cancer Program, Hillman Cancer Research Pavilion Laboratory , University of Pittsburgh Cancer Institute , Pittsburgh , PA , USA
| | | | - John M Kirkwood
- b Melanoma and Skin Cancer Program, Hillman Cancer Research Pavilion Laboratory , University of Pittsburgh Cancer Institute , Pittsburgh , PA , USA
| | - John F Hancock
- d Department of Integrative Biology and Pharmacology , University of Texas Health Science Center , Houston , TX , USA
| | - Andrew Poklepovic
- e Departments of Medicine , Virginia Commonwealth University , Richmond , VA , USA
| | - Paul Dent
- a Departments of Biochemistry and Molecular Biology , Virginia Commonwealth University , Richmond , VA , USA
| |
Collapse
|