51
|
Zhang Y, Xing Z, Liu T, Tang M, Mi L, Zhu J, Wu W, Wei T. Targeted therapy and drug resistance in thyroid cancer. Eur J Med Chem 2022; 238:114500. [DOI: 10.1016/j.ejmech.2022.114500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/24/2022]
|
52
|
Cai J, Jacob S, Kurupi R, Dalton KM, Coon C, Greninger P, Egan RK, Stein GT, Murchie E, McClanaghan J, Adachi Y, Hirade K, Dozmorov M, Glod J, Boikos SA, Ebi H, Hao H, Caponigro G, Benes CH, Faber AC. High-risk neuroblastoma with NF1 loss of function is targetable using SHP2 inhibition. Cell Rep 2022; 40:111095. [PMID: 35905710 DOI: 10.1016/j.celrep.2022.111095] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/04/2021] [Accepted: 06/23/2022] [Indexed: 12/19/2022] Open
Abstract
Reoccurring/high-risk neuroblastoma (NB) tumors have the enrichment of non-RAS/RAF mutations along the mitogen-activated protein kinase (MAPK) signaling pathway, suggesting that activation of MEK/ERK is critical for their survival. However, based on preclinical data, MEK inhibitors are unlikely to be active in NB and have demonstrated dose-limiting toxicities that limit their use. Here, we explore an alternative way to target the MAPK pathway in high-risk NB. We find that NB models are among the most sensitive among over 900 tumor-derived cell lines to the allosteric SHP2 inhibitor SHP099. Sensitivity to SHP099 in NB is greater in models with loss or low expression of the RAS GTPase activation protein (GAP) neurofibromin 1 (NF1). Furthermore, NF1 is lower in advanced and relapsed NB and NF1 loss is enriched in high-risk NB tumors regardless of MYCN status. SHP2 inhibition consistently blocks tumor growth in high-risk NB mouse models, revealing a new drug target in relapsed NB.
Collapse
Affiliation(s)
- Jinyang Cai
- Philips Institute for Oral Health Research, School of Dentistry, and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Sheeba Jacob
- Philips Institute for Oral Health Research, School of Dentistry, and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Richard Kurupi
- Philips Institute for Oral Health Research, School of Dentistry, and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Krista M Dalton
- Philips Institute for Oral Health Research, School of Dentistry, and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Colin Coon
- Philips Institute for Oral Health Research, School of Dentistry, and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Patricia Greninger
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Regina K Egan
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Giovanna T Stein
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Ellen Murchie
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Joseph McClanaghan
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Yuta Adachi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Aichi 464-8681, Japan
| | - Kentaro Hirade
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Aichi 464-8681, Japan
| | - Mikhail Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - John Glod
- National Cancer Institute, Pediatric Branch, Oncology, Bethesda, MD, USA
| | - Sosipatros A Boikos
- Department of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Aichi 464-8681, Japan
| | - Huaixiang Hao
- Novartis Institute for Biological Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Giordano Caponigro
- Novartis Institute for Biological Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Cyril H Benes
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Anthony C Faber
- Philips Institute for Oral Health Research, School of Dentistry, and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
53
|
Dahmani C, Corre E, Dandou S, Mangé A, Radulescu O, Coopman PJ, Cuq P, Larive RM. La résistance aux inhibiteurs de BRAF. Med Sci (Paris) 2022; 38:570-578. [DOI: 10.1051/medsci/2022083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
La voie de signalisation MAPK/ERK est une voie centrale de la signalisation intracellulaire. Sa dérégulation participe à la transformation et la progression tumorales. Dans plusieurs cancers, la découverte de mutations activatrices de BRAF, à l’origine de l’activation de cette voie, a ouvert de nouvelles perspectives thérapeutiques avec le développement d’inhibiteurs spécifiques de la protéine. Selon les cancers, ces inhibiteurs ont cependant montré soit une efficacité insuffisante, due à la résistance primaire des cellules tumorales, soit une efficacité transitoire, due à l’apparition d’une résistance acquise. Dans cette revue, nous revenons sur les découvertes qui ont conduit au développement de ces inhibiteurs de BRAF. Nous détaillons également les mécanismes moléculaires et cellulaires de la résistance à ces inhibiteurs observée dans différents types de cancers. Comprendre ces mécanismes est en effet primordial pour développer des stratégies thérapeutiques qui soient plus efficaces.
Collapse
|
54
|
Redox Homeostasis in Thyroid Cancer: Implications in Na +/I - Symporter (NIS) Regulation. Int J Mol Sci 2022; 23:ijms23116129. [PMID: 35682803 PMCID: PMC9181215 DOI: 10.3390/ijms23116129] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/17/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
Radioiodine therapy (RAI) is a standard and effective therapeutic approach for differentiated thyroid cancers (DTCs) based on the unique capacity for iodide uptake and accumulation of the thyroid gland through the Na+/I− symporter (NIS). However, around 5–15% of DTC patients may become refractory to radioiodine, which is associated with a worse prognosis. The loss of RAI avidity due to thyroid cancers is attributed to cell dedifferentiation, resulting in NIS repression by transcriptional and post-transcriptional mechanisms. Targeting the signaling pathways potentially involved in this process to induce de novo iodide uptake in refractory tumors is the rationale of “redifferentiation strategies”. Oxidative stress (OS) results from the imbalance between ROS production and depuration that favors a pro-oxidative environment, resulting from increased ROS production, decreased antioxidant defenses, or both. NIS expression and function are regulated by the cellular redox state in cancer and non-cancer contexts. In addition, OS has been implicated in thyroid tumorigenesis and thyroid cancer cell dedifferentiation. Here, we review the main aspects of redox homeostasis in thyrocytes and discuss potential ROS-dependent mechanisms involved in NIS repression in thyroid cancer.
Collapse
|
55
|
Oh JM, Rajendran RL, Gangadaran P, Hong CM, Jeong JH, Lee J, Ahn BC. Targeting GLI1 Transcription Factor for Restoring Iodine Avidity with Redifferentiation in Radioactive-Iodine Refractory Thyroid Cancers. Cancers (Basel) 2022; 14:cancers14071782. [PMID: 35406554 PMCID: PMC8997411 DOI: 10.3390/cancers14071782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Thyroid cancers have an excellent prognosis by standard therapy of surgery followed by radioactive-iodine therapy. However, metastatic thyroid cancers do not response to radioactive-iodine therapy by losing iodine avidity. Therefore, reversing iodine avidity to metastatic thyroid cancers gives a new chance of applying radioactive-iodine therapy to the cancers. In the current study, we found that GLI1 knockdown can revert iodine non-avid thyroid cancers to iodine avid cancers by increasing expression of thyroid-specific proteins. Restoration of iodine avidity in thyroid cancers makes the cancers sensitive to radioactive-iodine therapy again. Therefore, the GLI1 can be a potential therapeutic target of radioactive-iodine resistant thyroid cancers. Abstract Radioactive-iodine (RAI) therapy is the mainstay for patients with recurrent and metastatic thyroid cancer. However, many patients exhibit dedifferentiation characteristics along with lack of sodium iodide symporter (NIS) functionality, low expression of thyroid-specific proteins, and poor RAI uptake, leading to poor prognosis. Previous studies have demonstrated the effect of GLI family zinc finger 1 (GLI1) inhibition on tumor growth and apoptosis. In this study, we investigated the role of GLI1 in the context of redifferentiation and improvement in the efficacy of RAI therapy for thyroid cancer. We evaluated GLI1 expression in several thyroid cancer cell lines and selected TPC-1 and SW1736 cell lines showing the high expression of GLI. We performed GLI1 knockdown and evaluated the changes of thyroid-specific proteins expression, RAI uptake and I-131-mediated cytotoxicity. The effect of GANT61 (GLI1 inhibitor) on endogenous NIS expression was also assessed. Endogenous NIS expression upregulated by inhibiting GLI1, in addition, increased expression level in plasma membrane. Also, GLI1 knockdown increased expression of thyroid-specific proteins. Restoration of thyroid-specific proteins increased RAI uptake and I-131-mediated cytotoxic effect. Treatment with GANT61 also increased expression of endogenous NIS. Targeting GLI1 can be a potential strategy with redifferentiation for restoring RAI avidity in dedifferentiated thyroid cancers.
Collapse
Affiliation(s)
- Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; zzu--@hanmail.net
| | - Ju Hye Jeong
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; zzu--@hanmail.net
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; zzu--@hanmail.net
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; zzu--@hanmail.net
- Correspondence: ; Tel.: +82-53-420-5583
| |
Collapse
|
56
|
Systematic review of randomised clinical trials and observational studies for patients with RAS wild-type or BRAF V600E-mutant metastatic and/or unresectable colorectal cancer. Crit Rev Oncol Hematol 2022; 173:103646. [PMID: 35344913 DOI: 10.1016/j.critrevonc.2022.103646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 02/07/2023] Open
Abstract
Approximately 8-10% of metastatic colorectal cancer (mCRC) tumours harbour BRAFV600E mutations. Eleven randomised controlled trials (RCTs) and 24 non-RCTs were identified. Seven studies evaluated BRAF inhibitors. Single-agent BRAF inhibitors had minimal efficacy, whereas BRAF inhibitor plus anti-EGFR therapy improved outcomes. In BEACON CRC, overall survival (OS) was significantly longer for patients receiving encorafenib plus cetuximab ± binimetinib when compared with irinotecan/FOLFIRI plus cetuximab as second- and third-line therapy. Seven prospective non-RCTs reported worse OS and progression-free survival (PFS) for patients with BRAFV600E-mutant vs BRAF wild-type mCRC. Eight RCTs reported that PFS and OS were generally shorter for patients with BRAFV600E-mutant mCRC vs those with KRAS or RAS wild-type mCRC. Patients with BRAFV600E-mutant mCRC have worse outcomes with conventional therapy vs patients with BRAF wild-type tumours. BRAF inhibitors in conjunction with anti-EGFR therapy improves outcomes for patients with BRAFV600E-mutant mCRC vs conventional therapy or a BRAF inhibitor alone.
Collapse
|
57
|
Kelley MB, Geddes TJ, Ochiai M, Lampl NM, Kothmann WW, Fierstein SR, Kent V, DeCicco-Skinner K. Loss of Tpl2 activates compensatory signaling and resistance to EGFR/MET dual inhibition in v-RAS transduced keratinocytes. PLoS One 2022; 17:e0266017. [PMID: 35325006 PMCID: PMC8947257 DOI: 10.1371/journal.pone.0266017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 03/11/2022] [Indexed: 11/18/2022] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second most common form of skin cancer in the United States, affecting one million people per year. Patients with aggressive disease have limited treatment options and high mortality, highlighting the need to identify new biomarkers linked to poor clinical outcome. HRAS mutations are found in skin papillomas and cSCCs and increase in frequency when MAP3K family members are inhibited, suggesting a link between blockade of mitogen-activated protein kinase (MAPK) signaling and initiation of RAS-primed cells. Tpl2, a MAP3K gene, can serve as a tumor suppressor gene in cSCC. We have previously shown that upon Tpl2 ablation, mice have heightened sensitivity to aberrant RAS signaling. Tpl2-/- mice display significantly higher numbers of papillomas and cSCCs in two-stage chemical carcinogenesis studies and increased tumorigenicity of keratinocytes expressing oncogenic v-rasHa in nude mouse skin grafts. In part, this is mediated through increased mesenchymal-epithelial transition factor (MET) receptor activity. Epidermal Growth Factor Receptor (EGFR) is reported to be an essential factor for MET-driven carcinogenesis and MET activation may confer resistance to EGFR therapies, suggesting that the concurrent use of both an EGFR inhibitor and a MET inhibitor may show promise in advanced cSCCs. In this study we assessed whether normal or Ras-transformed Tpl2-/- keratinocytes have aberrant EGFR signaling and whether concomitant treatment with EGFR/MET tyrosine kinase inhibitors was more effective than single agents in reducing growth and angiogenic potential of Ras-transformed keratinocytes. Tpl2-/- keratinocytes exhibited increased HER-2 and STAT-3 under basal conditions and elevated p-MET and p-EGFR when transduced with oncogenic RAS. Inhibition of MET by Capmatinib increased p-EGFR in Tpl2-/- keratinocytes and papillomas, and inhibition of EGFR by Gefitinib increased HER2 and HER3 signaling in both genotypes. Treatment of keratinocytes with EGFR and MET inhibitors, in combination, significantly enhanced endothelial tube formation, MMP-9 activity and activation of other RTKs, with more pronounced effects when Tpl2 was ablated. These data indicate that Tpl2 cross-talks with both EGFR and MET signaling pathways. Upon inhibition of EGFR/MET signaling, a myriad of escape mechanisms exists in keratinocytes to overcome targeted drug effects.
Collapse
Affiliation(s)
- Mary B. Kelley
- Department of Biology, American University, Washington, DC, United States of America
| | - Taylor J. Geddes
- Department of Biology, American University, Washington, DC, United States of America
| | - Maria Ochiai
- Department of Biology, American University, Washington, DC, United States of America
| | - Noah M. Lampl
- Department of Biology, American University, Washington, DC, United States of America
| | - W. Wade Kothmann
- Department of Biology, American University, Washington, DC, United States of America
| | - Sara R. Fierstein
- Department of Biology, American University, Washington, DC, United States of America
| | - Victoria Kent
- Department of Biology, American University, Washington, DC, United States of America
| | | |
Collapse
|
58
|
Silaghi H, Lozovanu V, Georgescu CE, Pop C, Nasui BA, Cătoi AF, Silaghi CA. State of the Art in the Current Management and Future Directions of Targeted Therapy for Differentiated Thyroid Cancer. Int J Mol Sci 2022; 23:ijms23073470. [PMID: 35408830 PMCID: PMC8998761 DOI: 10.3390/ijms23073470] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 02/06/2023] Open
Abstract
Two-thirds of differentiated thyroid cancer (DTC) patients with distant metastases would be classified as radioactive iodine-refractory (RAIR-DTC), evolving into a poor outcome. Recent advances underlying DTC molecular mechanisms have shifted the therapy focus from the standard approach to targeting specific genetic dysregulations. Lenvatinib and sorafenib are first-line, multitargeted tyrosine kinase inhibitors (TKIs) approved to treat advanced, progressive RAIR-DTC. However, other anti-angiogenic drugs, including single targeted TKIs, are currently being evaluated as alternative or salvage therapy after the failure of first-line TKIs. Combinatorial therapy of mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signalling cascade inhibitors has become a highly advocated strategy to improve the low efficiency of the single agent treatment. Recent studies pointed out targetable alternative pathways to overcome the resistance to MAPK and PI3K pathways’ inhibitors. Because radioiodine resistance originates in DTC loss of differentiation, redifferentiation therapies are currently being explored for efficacy. The present review will summarize the conventional management of DTC, the first-line and alternative TKIs in RAIR-DTC, and the approaches that seek to overcome the resistance to MAPK and PI3K pathways’ inhibitors. We also aim to emphasize the latest achievements in the research of redifferentiation therapy, immunotherapy, and agents targeting gene rearrangements in advanced DTC.
Collapse
Affiliation(s)
- Horatiu Silaghi
- Department of Surgery V, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania;
| | - Vera Lozovanu
- County Clinical Emergency Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania;
| | - Carmen Emanuela Georgescu
- Department of Endocrinology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.E.G.); (C.A.S.)
| | - Cristina Pop
- Department of Pharmacology, Physiology, and Pathophysiology, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 6A Louis Pasteur Street, 400349 Cluj-Napoca, Romania
- Correspondence:
| | - Bogdana Adriana Nasui
- Department of Community Health, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 6 Louis Pasteur Street, 400349 Cluj-Napoca, Romania;
| | - Adriana Florinela Cătoi
- Department of Pathophysiology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania;
| | - Cristina Alina Silaghi
- Department of Endocrinology, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.E.G.); (C.A.S.)
| |
Collapse
|
59
|
Mooz J, Riegel K, PS H, Sadanandam A, Marini F, Klein M, Werner U, Roth W, Wilken-Schmitz A, Tegeder I, Rajalingam K. ARAF suppresses ERBB3 expression and metastasis in a subset of lung cancers. SCIENCE ADVANCES 2022; 8:eabk1538. [PMID: 35302851 PMCID: PMC8932670 DOI: 10.1126/sciadv.abk1538] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 01/27/2022] [Indexed: 06/14/2023]
Abstract
RAF kinases are highly conserved serine/threonine kinases, and among the three RAF isoforms (ARAF, BRAF, and CRAF), the pathophysiological relevance of ARAF is not well defined. Here, we show that patients with lung cancer exhibit low expression of ARAF, which is associated with lymph node metastasis and poor patient survival. We uncover that depletion of ARAF promotes anchorage-independent growth and metastasis through activation of AKT signaling in a subset of lung cancer cells. We identified that loss of ARAF was associated with an increase in ERBB3 expression in a kinase-independent manner. ARAF suppressed the promoter activity of ERBB3, and reconstitution of ARAF in ARAF-depleted cells led to the reversal of enhanced ERBB3-AKT signaling. Furthermore, ARAF inhibited neuregulin 1 (hNRG1)-mediated AKT activation through controlling ERBB3 expression via the transcription factor KLF5. Our results disclose a critical dual role for ARAF kinase in the negative regulation of ERBB3-AKT signaling, thereby suppressing tumor metastasis.
Collapse
Affiliation(s)
- Juliane Mooz
- Cell Biology Unit, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Kristina Riegel
- Cell Biology Unit, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Hari PS
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Anguraj Sadanandam
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Federico Marini
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Klein
- Institute of Immunology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | | | - Wilfried Roth
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Annett Wilken-Schmitz
- Institute of Clinical Pharmacology, Goethe-University, Medical Faculty, Frankfurt (Main), Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University, Medical Faculty, Frankfurt (Main), Germany
| | - Krishnaraj Rajalingam
- Cell Biology Unit, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- University Cancer Center Mainz, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
60
|
Tchekmedyian V, Dunn L, Sherman E, Baxi SS, Grewal RK, Larson SM, Pentlow KS, Haque S, Tuttle RM, Sabra MM, Fish S, Boucai L, Walters J, Ghossein RA, Seshan VE, Knauf JA, Pfister DG, Fagin JA, Ho AL. Enhancing Radioiodine Incorporation in BRAF-Mutant, Radioiodine-Refractory Thyroid Cancers with Vemurafenib and the Anti-ErbB3 Monoclonal Antibody CDX-3379: Results of a Pilot Clinical Trial. Thyroid 2022; 32:273-282. [PMID: 35045748 PMCID: PMC9206492 DOI: 10.1089/thy.2021.0565] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Oncogenic activation of mitogen-activated protein kinase (MAPK) signaling is associated with radioiodine refractory (RAIR) thyroid cancer. Preclinical models suggest that activation of the receptor tyrosine kinase erbB-3 (HER3) mitigates the MAPK pathway inhibition achieved by BRAF inhibitors in BRAFV600E mutant thyroid cancers. We hypothesized that combined inhibition of BRAF and HER3 using vemurafenib and the human monoclonal antibody CDX-3379, respectively, would potently inhibit MAPK activation and restore radioactive iodine (RAI) avidity in patients with BRAF-mutant RAIR thyroid cancer. Methods: Patients with BRAFV600E RAIR thyroid cancer were evaluated by thyrogen-stimulated iodine-124 (124I) positron emission tomography-computed tomography (PET/CT) at baseline and after 5 weeks of treatment with oral vemurafenib 960 mg twice daily alone for 1 week, followed by vemurafenib in combination with 1000 mg of intravenous CDX-3379 every 2 weeks. Patients with adequate 124I uptake on the second PET/CT then received therapeutic radioactive iodine (131I) with vemurafenb+CDX-3379. All therapy was discontinued two days later. Treatment response was monitored by serum thyroglobulin measurements and imaging. The primary endpoints were safety and tolerability of vemurafenib+CDX-3379, as well as the proportion of patients after vemurafenb+CDX-3379 therapy with enhanced RAI incorporation warranting therapeutic 131I. Results: Seven patients were enrolled; six were evaluable for the primary endpoints. No grade 3 or 4 toxicities related to CDX-3379 were observed. Five patients had increased RAI uptake after treatment; in 4 patients this increased uptake warranted therapeutic 131I. At 6 months, 2 patients achieved partial response after 131I and 2 progression of disease. Next-generation sequencing of 5 patients showed that all had co-occurring telomerase reverse transcriptase promoter alterations. A deleterious mutation in the SWItch/Sucrose Non-Fermentable (SWI/SNF) gene ARID2 was discovered in the patient without enhanced RAI avidity after therapy and an RAI-resistant tumor from another patient that was sampled off-study. Conclusions: The endpoints for success were met, providing preliminary evidence of vemurafenib+CDX-3379 safety and efficacy for enhancing RAI uptake. Preclinical data and genomic profiling in this small cohort suggest SWI/SNF gene mutations should be investigated as potential markers of resistance to redifferentiation strategies. Further evaluation of vemurafenib+CDX-3379 as a redifferentiation therapy in a larger trial is warranted (ClinicalTrials.gov: NCT02456701).
Collapse
Affiliation(s)
| | - Lara Dunn
- Department of Medicine, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Eric Sherman
- Department of Medicine, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | | | | | | | | | - Sofia Haque
- Department of Radiology, New York, New York, USA
| | - R. Michael Tuttle
- Department of Medicine, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Mona M. Sabra
- Department of Medicine, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Stephanie Fish
- Department of Medicine, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Laura Boucai
- Department of Medicine, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | | | | | | | - Jeffrey A. Knauf
- Department of Human Oncology and Pathogenesis Program; Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - David G. Pfister
- Department of Medicine, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - James A. Fagin
- Department of Medicine, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
- Department of Human Oncology and Pathogenesis Program; Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alan L. Ho
- Department of Medicine, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
- Address correspondence to: Alan L. Ho, MD, PhD, Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, USA
| |
Collapse
|
61
|
SGSM2 inhibits thyroid cancer progression by activating RAP1 and enhancing competitive RAS inhibition. Cell Death Dis 2022; 13:218. [PMID: 35264562 PMCID: PMC8907342 DOI: 10.1038/s41419-022-04598-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 01/17/2022] [Accepted: 01/28/2022] [Indexed: 11/25/2022]
Abstract
Thyroid cancer (TC) is one of the most common malignancies involving the head and neck, and its incidences are increasing every year. Small G protein signaling modulators 2 (SGSM2) belongs to a newly identified protein group that contributes to numerous cancer progression. However, its role in TC remains unknown. The aim of this study was to explore the functions and underlying molecular mechanism of SGSM2 in the progression of thyroid tumorigenesis. Here, we demonstrated that SGSM2 expression was markedly decreased in TC, and that lower SGSM2 expression was potentially related to worse patient prognosis. Meanwhile, the SGSM2 levels were not directly correlated with BRAF or RAS mutations in TC. Based on our functional analysis, ectopic SGSM2 expression strongly prevented cell proliferation, migration, invasion, and tumorigenic activity in TC cells that harbored wild type RAS. Mechanistically, we demonstrated that SGSM2 interacted with Small G protein Ras-associated protein 1(RAP1) and augmented its activity. Activated RAP1 then competitively suppressed RAS activation and thereby downregulated output of MAPK/ERK and PI3K/Akt networks, which are primary contributors of TC. In summary, the present study reports a tumor suppressive role of SGSM2 in TC. Moreover, we revealed the underlying molecular mechanism, thus providing a potential therapeutic target for TCs that harbor wild type RAS. A schematic model of SGSM2 suppressing the progression of RASWT thyroid cancer.![]()
Collapse
|
62
|
Hou P, Wang YA. Conquering oncogenic KRAS and its bypass mechanisms. Theranostics 2022; 12:5691-5709. [PMID: 35966590 PMCID: PMC9373815 DOI: 10.7150/thno.71260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/05/2022] [Indexed: 11/19/2022] Open
Abstract
Aberrant activation of KRAS signaling is common in cancer, which has catalyzed heroic drug development efforts to target KRAS directly or its downstream signaling effectors. Recent works have yielded novel small molecule drugs with promising preclinical and clinical activities. Yet, no matter how a cancer is addicted to a specific target - cancer's genetic and biological plasticity fashions a variety of resistance mechanisms as a fait accompli, limiting clinical benefit of targeted interventions. Knowledge of these mechanisms may inform combination strategies to attack both oncogenic KRAS and subsequent bypass mechanisms.
Collapse
Affiliation(s)
- Pingping Hou
- Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103, USA.,Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey 07103, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA.,Lead contact
| | - Y Alan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
63
|
Lee M, Untch BR, Xu B, Ghossein R, Han C, Kuo F, Valero C, Nadeem Z, Patel N, Makarov V, Dogan S, Wong RJ, Sherman EJ, Ho AL, Chan TA, Fagin JA, Morris LGT. Genomic and Transcriptomic Correlates of Thyroid Carcinoma Evolution after BRAF Inhibitor Therapy. Mol Cancer Res 2022; 20:45-55. [PMID: 34635506 PMCID: PMC8738128 DOI: 10.1158/1541-7786.mcr-21-0442] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/12/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Targeted inhibition of BRAF V600E achieves tumor control in a subset of advanced thyroid tumors. Nearly all tumors develop resistance, and some have been observed to subsequently undergo dedifferentiation. The molecular alterations associated with thyroid cancer dedifferentiation in the setting of BRAF inhibition are unknown. We analyzed targeted next-generation sequencing data from 639 advanced, recurrent and/or metastatic thyroid carcinomas, including 15 tumors that were treated with BRAF inhibitor drugs and had tissue sampled during or posttreatment, 8 of which had matched pretherapy samples. Pre- and posttherapy tissues from one additional patient were profiled with whole-exome sequencing and RNA expression profiling. Mutations in genes comprising the SWI/SNF chromatin remodeling complex and the PI3K-AKT-mTOR, MAPK, and JAK-STAT pathways all increased in prevalence across more dedifferentiated thyroid cancer histologies. Of 7 thyroid cancers that dedifferentiated after BRAF inhibition, 6 had mutations in these pathways. These mutations were mostly absent from matched pretreatment samples and were rarely detected in tumors that did not dedifferentiate. Additional analyses in one of the vemurafenib-treated tumors before and after anaplastic transformation revealed the emergence of an oncogenic PIK3CA mutation, activation of ERK signaling, dedifferentiation, and development of an immunosuppressive tumor microenvironment. These findings validate earlier preclinical data implicating these genetic pathways in resistance to BRAF inhibitors, and suggest that genetic alterations mediating acquired drug resistance may also promote thyroid tumor dedifferentiation. IMPLICATIONS: The possibility that thyroid cancer dedifferentiation may be attributed to selective pressure applied by BRAF inhibitor-targeted therapy should be investigated further.
Collapse
Affiliation(s)
- Mark Lee
- Weill Cornell Medicine, New York, New York
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian R Untch
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bin Xu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronald Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Catherine Han
- Weill Cornell Medicine, New York, New York
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fengshen Kuo
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cristina Valero
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zaineb Nadeem
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neal Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio
| | - Snjezana Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard J Wong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric J Sherman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Timothy A Chan
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio
| | - James A Fagin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Luc G T Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
64
|
HER3 Augmentation via Blockade of EGFR/AKT Signaling Enhances Anticancer Activity of HER3-Targeting Patritumab Deruxtecan in EGFR-Mutated Non–Small Cell Lung Cancer. Clin Cancer Res 2021; 28:390-403. [DOI: 10.1158/1078-0432.ccr-21-3359] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/04/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022]
|
65
|
Rocca A, Kholodenko BN. Can Systems Biology Advance Clinical Precision Oncology? Cancers (Basel) 2021; 13:6312. [PMID: 34944932 PMCID: PMC8699328 DOI: 10.3390/cancers13246312] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 12/10/2021] [Indexed: 12/13/2022] Open
Abstract
Precision oncology is perceived as a way forward to treat individual cancer patients. However, knowing particular cancer mutations is not enough for optimal therapeutic treatment, because cancer genotype-phenotype relationships are nonlinear and dynamic. Systems biology studies the biological processes at the systems' level, using an array of techniques, ranging from statistical methods to network reconstruction and analysis, to mathematical modeling. Its goal is to reconstruct the complex and often counterintuitive dynamic behavior of biological systems and quantitatively predict their responses to environmental perturbations. In this paper, we review the impact of systems biology on precision oncology. We show examples of how the analysis of signal transduction networks allows to dissect resistance to targeted therapies and inform the choice of combinations of targeted drugs based on tumor molecular alterations. Patient-specific biomarkers based on dynamical models of signaling networks can have a greater prognostic value than conventional biomarkers. These examples support systems biology models as valuable tools to advance clinical and translational oncological research.
Collapse
Affiliation(s)
- Andrea Rocca
- Hygiene and Public Health, Local Health Unit of Romagna, 47121 Forlì, Italy
| | - Boris N. Kholodenko
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
66
|
Schreck KC, Morin A, Zhao G, Allen AN, Flannery P, Glantz M, Green AL, Jones C, Jones KL, Kilburn LB, Nazemi KJ, Samuel D, Sanford B, Solomon DA, Wang J, Pratilas CA, Nicolaides T, Mulcahy Levy JM. Deconvoluting Mechanisms of Acquired Resistance to RAF Inhibitors in BRAF V600E-Mutant Human Glioma. Clin Cancer Res 2021; 27:6197-6208. [PMID: 34433654 PMCID: PMC8595717 DOI: 10.1158/1078-0432.ccr-21-2660] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/12/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Selective RAF-targeted therapy is effective in some patients with BRAFV600E-mutated glioma, though emergent and adaptive resistance occurs through ill-defined mechanisms. EXPERIMENTAL DESIGN Paired pre-/post- RAF inhibitor (RAFi)-treated glioma samples (N = 15) were obtained and queried for treatment-emergent genomic alterations using DNA and RNA sequencing (RNA-seq). Functional validation of putative resistance mechanisms was performed using established and patient-derived BRAFV600E-mutant glioma cell lines. RESULTS Analysis of 15 tissue sample pairs identified 13 alterations conferring putative resistance were identified among nine paired samples (including mutations involving ERRFI1, BAP1, ANKHD1, and MAP2K1). We performed functional validation of mechanisms of resistance, including loss of NF1, PTEN, or CBL, in BRAFV600E-mutant glioma lines, and demonstrate they are capable of conferring resistance in vitro. Knockdown of CBL resulted in increased EGFR expression and phosphorylation, a possible mechanism for maintaining ERK signaling within the cell. Combination therapy with a MEKi or EGFR inhibitor was able to overcome resistance to BRAFi, in NF1 knockdown and CBL knockdown, respectively. Restoration of wild-type PTEN in B76 cells (PTEN-/-) restored sensitivity to BRAFi. We identified and validated CRAF upregulation as a mechanism of resistance in one resistant sample. RNA-seq analysis identified two emergent expression patterns in resistant samples, consistent with expression patterns of known glioma subtypes. CONCLUSIONS Resistance mechanisms to BRAFi in glioma are varied and may predict effective precision combinations of targeted therapy, highlighting the importance of a personalized approach.
Collapse
Affiliation(s)
- Karisa C Schreck
- Department of Neurology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Neurosurgery, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Andrew Morin
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, Colorado
| | - Guisheng Zhao
- Department of Pediatrics, NYU Langone Health, New York, New York
| | - Amy N Allen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Pediatrics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Patrick Flannery
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, Colorado
| | - Michael Glantz
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania
- Department of Oncology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Adam L Green
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, Colorado
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado
| | - Chris Jones
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | | | - Lindsay B Kilburn
- Division of Oncology and the Brain Tumor Institute, Children's National Hospital, Washington, DC
| | - Kellie J Nazemi
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon
| | - David Samuel
- Department of Hematology-Oncology, Valley Children's Healthcare, Madera, California
| | - Bridget Sanford
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - David A Solomon
- Department of Pathology, University of California, San Francisco, California
| | - Jiawan Wang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Pediatrics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Christine A Pratilas
- Department of Neurology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Pediatrics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | | | - Jean M Mulcahy Levy
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado.
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, Colorado
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
67
|
Brandenburg T, Muchalla P, Theurer S, Schmid KW, Führer D. Therapeutic Effect of Combined Dabrafenib and Trametinib Treatment of BRAF V600E-Mutated Primary Squamous Cell Carcinoma of the Thyroid: A Case Report. Eur Thyroid J 2021; 10:511-516. [PMID: 34956922 PMCID: PMC8647138 DOI: 10.1159/000518055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/18/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Primary squamous cell carcinoma (PSCC) of the thyroid is an exceptionally rare malignancy accounting for <1% of all primary thyroid cancers. Therapy is multimodal including surgery, radiotherapy, and chemotherapy but with no consensus for management and therapy. Here, we describe a case of a male patient who presented with a BRAF V600E-mutated PSCC of the thyroid gland showing response to combined dabrafenib and trametinib therapy over a period of >12 months. CASE PRESENTATION A 78-year-old male patient presented with a 3-week history of dysphonia and dyspnoea. Laryngoscopy revealed a mechanical obstruction by a right-sided, subglottical mass, which on cervical ultrasound was highly suggestive of anaplastic thyroid carcinoma. Additional workup including esophagogastroduodenoscopy showed compression of the oesophagus but no oesophageal infiltration by the tumour. Immunohistochemistry displayed CK19-positive cells indicating epithelial origin of the tumour. CK5/6 and P40 immunohistochemistry confirmed the morphological impression of squamous cell differentiation while staining with thyroid markers TTF-1 and TPO was negative and PAX8 showed a nuclear positive signal. Based on immunohistopathology, presence of TP53 and BRAF V600E mutations, and exclusion of metastatic squamous cell carcinoma of other origin, the diagnosis of a PSCC of the thyroid was established. As an individualized treatment concept, we decided to advocate combined BRAF V600E targeting by the multikinase inhibitors dabrafenib and trametinib. This led to drastic improvement in patient's quality of life without severe side effects over a period of >12 months. CONCLUSION In this case, molecular diagnosis allowed a highly individualized treatment concept with combined dabrafenib and trametinib therapy.
Collapse
Affiliation(s)
- Tim Brandenburg
- Department of Endocrinology, Diabetes and Metabolism, Endocrine Tumour Center at West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Endocrine Tumour Center at West German Cancer Center, Member of ENDO-ERN and EURACAN, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- *Tim Brandenburg,
| | - Philipp Muchalla
- Department of Endocrinology, Diabetes and Metabolism, Endocrine Tumour Center at West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Endocrine Tumour Center at West German Cancer Center, Member of ENDO-ERN and EURACAN, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sarah Theurer
- Endocrine Tumour Center at West German Cancer Center, Member of ENDO-ERN and EURACAN, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- University Duisburg-Essen, Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kurt Werner Schmid
- Endocrine Tumour Center at West German Cancer Center, Member of ENDO-ERN and EURACAN, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- University Duisburg-Essen, Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dagmar Führer
- Department of Endocrinology, Diabetes and Metabolism, Endocrine Tumour Center at West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Endocrine Tumour Center at West German Cancer Center, Member of ENDO-ERN and EURACAN, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
68
|
Bagheri-Yarmand R, Busaidy NL, McBeath E, Danysh BP, Evans KW, Moss TJ, Akcakanat A, Ng PKS, Knippler CM, Golden JA, Williams MD, Multani AS, Cabanillas ME, Shaw KR, Meric-Bernstam F, Shah MH, Ringel MD, Hofmann MC. RAC1 Alterations Induce Acquired Dabrafenib Resistance in Association with Anaplastic Transformation in a Papillary Thyroid Cancer Patient. Cancers (Basel) 2021; 13:4950. [PMID: 34638434 PMCID: PMC8507731 DOI: 10.3390/cancers13194950] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 11/19/2022] Open
Abstract
BRAF-activating mutations are the most frequent driver mutations in papillary thyroid cancer (PTC). Targeted inhibitors such as dabrafenib have been used in advanced BRAF-mutated PTC; however, acquired resistance to the drug is common and little is known about other effectors that may play integral roles in this resistance. In addition, the induction of PTC dedifferentiation into highly aggressive KRAS-driven anaplastic thyroid cancer (ATC) has been reported. We detected a novel RAC1 (P34R) mutation acquired during dabrafenib treatment in a progressive metastatic lesion with ATC phenotype. To identify a potential functional link between this novel mutation and tumor dedifferentiation, we developed a cell line derived from the metastatic lesion and compared its behavior to isogenic cell lines and primary tumor samples. Our data demonstrated that RAC1 mutations induce changes in cell morphology, reorganization of F-actin almost exclusively at the cell cortex, and changes in cell adhesion properties. We also established that RAC1 amplification, with or without mutation, is sufficient to drive cell proliferation and resistance to BRAF inhibition. Further, we identified polyploidy of chromosome 7, which harbors RAC1, in both the metastatic lesion and its derived cell line. Copy number amplification and overexpression of other genes located on this chromosome, such as TWIST1, EGFR, and MET were also detected, which might also lead to dabrafenib resistance. Our study suggests that polyploidy leading to increased expression of specific genes, particularly those located on chromosome 7, should be considered when analyzing aggressive thyroid tumor samples and in further treatments.
Collapse
Affiliation(s)
- Rozita Bagheri-Yarmand
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.B.-Y.); (N.L.B.); (E.M.); (B.P.D.); (J.A.G.); (M.E.C.)
| | - Naifa L. Busaidy
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.B.-Y.); (N.L.B.); (E.M.); (B.P.D.); (J.A.G.); (M.E.C.)
| | - Elena McBeath
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.B.-Y.); (N.L.B.); (E.M.); (B.P.D.); (J.A.G.); (M.E.C.)
| | - Brian P. Danysh
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.B.-Y.); (N.L.B.); (E.M.); (B.P.D.); (J.A.G.); (M.E.C.)
| | - Kurt W. Evans
- Department of Investigative Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.W.E.); (A.A.); (P.K.S.N.); (K.R.S.); (F.M.-B.)
| | - Tyler J. Moss
- Bioinformatics & Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Argun Akcakanat
- Department of Investigative Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.W.E.); (A.A.); (P.K.S.N.); (K.R.S.); (F.M.-B.)
| | - Patrick K. S. Ng
- Department of Investigative Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.W.E.); (A.A.); (P.K.S.N.); (K.R.S.); (F.M.-B.)
| | - Christina M. Knippler
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA; (C.M.K.); (M.D.R.)
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, GA 30322, USA
| | - Jalyn A. Golden
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.B.-Y.); (N.L.B.); (E.M.); (B.P.D.); (J.A.G.); (M.E.C.)
| | - Michelle D. Williams
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Asha S. Multani
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Maria E. Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.B.-Y.); (N.L.B.); (E.M.); (B.P.D.); (J.A.G.); (M.E.C.)
| | - Kenna R. Shaw
- Department of Investigative Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.W.E.); (A.A.); (P.K.S.N.); (K.R.S.); (F.M.-B.)
| | - Funda Meric-Bernstam
- Department of Investigative Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.W.E.); (A.A.); (P.K.S.N.); (K.R.S.); (F.M.-B.)
| | - Manisha H. Shah
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Matthew D. Ringel
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA; (C.M.K.); (M.D.R.)
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Marie Claude Hofmann
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (R.B.-Y.); (N.L.B.); (E.M.); (B.P.D.); (J.A.G.); (M.E.C.)
| |
Collapse
|
69
|
Precision Medicine for Colorectal Cancer with Liquid Biopsy and Immunotherapy. Cancers (Basel) 2021; 13:cancers13194803. [PMID: 34638288 PMCID: PMC8507967 DOI: 10.3390/cancers13194803] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary There are some challenges to improve the clinical outcome of colorectal cancers (CRCs) by implementing new technologies, such as early detection of recurrence/relapse and selection of appropriate drugs based on the genomic profiles of tumors. For example, the genomic characteristics of tumors can be analyzed by blood-based tests, namely ‘liquid biopsies’, which are minimally-invasive and can be performed repeatedly during the treatment course. Hence, liquid biopsies are considered to hold great promise to fill these gaps in clinical routines. In this review, we addressed clinical usefulness of liquid biopsies in the clinical management of CRC patients, including cancer screening, detection of minimal residual disease, selection of appropriate molecular-targeted drugs, monitoring of the treatment responsiveness, and very early detection of recurrence/relapse of the disease. Furthermore, we discussed the possibility of adoptive T cell therapies and a future personalized immunotherapy based on tumor genome information. Abstract In the field of colorectal cancer (CRC) treatment, diagnostic modalities and chemotherapy regimens have progressed remarkably in the last two decades. However, it is still difficult to identify minimal residual disease (MRD) necessary for early detection of recurrence/relapse of tumors and to select and provide appropriate drugs timely before a tumor becomes multi-drug-resistant and more aggressive. We consider the leveraging of in-depth genomic profiles of tumors as a significant breakthrough to further improve the overall prognosis of CRC patients. With the recent technological advances in methodologies and bioinformatics, the genomic profiles can be analyzed profoundly without delay by blood-based tests—‘liquid biopsies’. From a clinical point of view, a minimally-invasive liquid biopsy is thought to be a promising method and can be implemented in routine clinical settings in order to meet unmet clinical needs. In this review, we highlighted clinical usefulness of liquid biopsies in the clinical management of CRC patients, including cancer screening, detection of MRD, selection of appropriate molecular-targeted drugs, monitoring of the treatment responsiveness, and very early detection of recurrence/relapse of the disease. In addition, we addressed a possibility of adoptive T cell therapies and a future personalized immunotherapy based on tumor genome information.
Collapse
|
70
|
Valvo V, Iesato A, Kavanagh TR, Priolo C, Zsengeller Z, Pontecorvi A, Stillman IE, Burke SD, Liu X, Nucera C. Fine-Tuning Lipid Metabolism by Targeting Mitochondria-Associated Acetyl-CoA-Carboxylase 2 in BRAFV600E Papillary Thyroid Carcinoma. Thyroid 2021; 31:1335-1358. [PMID: 33107403 PMCID: PMC8558082 DOI: 10.1089/thy.2020.0311] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background: BRAFV600E acts as an ATP-dependent cytosolic kinase. BRAFV600E inhibitors are widely available, but resistance to them is widely reported in the clinic. Lipid metabolism (fatty acids) is fundamental for energy and to control cell stress. Whether and how BRAFV600E impacts lipid metabolism regulation in papillary thyroid carcinoma (PTC) is still unknown. Acetyl-CoA carboxylase (ACC) is a rate-limiting enzyme for de novo lipid synthesis and inhibition of fatty acid oxidation (FAO). ACC1 and ACC2 genes encode distinct isoforms of ACC. The aim of our study was to determine the relationship between BRAFV600E and ACC in PTC. Methods: We performed RNA-seq and DNA copy number analyses in PTC and normal thyroid (NT) in The Cancer Genome Atlas samples. Validations were performed by using assays on PTC-derived cell lines of differing BRAF status and a xenograft mouse model derived from a heterozygous BRAFWT/V600E PTC-derived cell line with knockdown (sh) of ACC1 or ACC2. Results:ACC2 mRNA expression was significantly downregulated in BRAFV600E-PTC vs. BRAFWT-PTC or NT clinical samples. ACC2 protein levels were downregulated in BRAFV600E-PTC cell lines vs. the BRAFWT/WT PTC cell line. Vemurafenib increased ACC2 (and to a lesser extent ACC1) mRNA levels in PTC-derived cell lines in a BRAFV600E allelic dose-dependent manner. BRAFV600E inhibition increased de novo lipid synthesis rates, and decreased FAO due to oxygen consumption rate (OCR), and extracellular acidification rate (ECAR), after addition of palmitate. Only shACC2 significantly increased OCR rates due to FAO, while it decreased ECAR in BRAFV600E PTC-derived cells vs. controls. BRAFV600E inhibition synergized with shACC2 to increase intracellular reactive oxygen species production, leading to increased cell proliferation and, ultimately, vemurafenib resistance. Mice implanted with a BRAFWT/V600E PTC-derived cell line with shACC2 showed significantly increased tumor growth after vemurafenib treatment, while vehicle-treated controls, or shGFP control cells treated with vemurafenib showed stable tumor growth. Conclusions: These findings suggest a potential link between BRAFV600E and lipid metabolism regulation in PTC. BRAFV600E downregulates ACC2 levels, which deregulates de novo lipid synthesis, FAO due to OCR, and ECAR rates. ShACC2 may contribute to vemurafenib resistance and increased tumor growth. ACC2 rescue may represent a novel molecular strategy for overcoming resistance to BRAFV600E inhibitors in refractory PTC.
Collapse
Affiliation(s)
- Veronica Valvo
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Department of Pathology, Cancer Research Institute (CRI), Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Asumi Iesato
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Department of Pathology, Cancer Research Institute (CRI), Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Taylor R. Kavanagh
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carmen Priolo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Alfredo Pontecorvi
- Department of Medicine, Agostino Gemelli Medical School, UCSC, Rome, Italy
| | - Isaac E. Stillman
- Department of Pathology; Harvard Medical School, Boston, Massachusetts, USA
| | - Suzanne D. Burke
- Department of Medicine; Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaowen Liu
- Department of Emergency Medicine; Harvard Medical School, Boston, Massachusetts, USA
| | - Carmelo Nucera
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Department of Pathology, Cancer Research Institute (CRI), Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Center for Vascular Biology Research (CVBR); Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Address correspondence to: Carmelo Nucera, MD, PhD, Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Department of Pathology, Cancer Research Institute (CRI) Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Office: RN270K, 99 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
71
|
Davidson CD, Bolf EL, Gillis NE, Cozzens LM, Tomczak JA, Carr FE. Thyroid Hormone Receptor Beta Inhibits PI3K-Akt-mTOR Signaling Axis in Anaplastic Thyroid Cancer via Genomic Mechanisms. J Endocr Soc 2021; 5:bvab102. [PMID: 34258492 PMCID: PMC8271203 DOI: 10.1210/jendso/bvab102] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Indexed: 01/21/2023] Open
Abstract
Thyroid cancer is the most common endocrine malignancy, and the global incidence has increased rapidly over the past few decades. Anaplastic thyroid cancer (ATC) is highly aggressive, dedifferentiated, and patients have a median survival of fewer than 6 months. Oncogenic alterations in ATC include aberrant phosphoinositide 3 kinase (PI3K) signaling through receptor tyrosine kinase (RTK) amplification, loss of phosphoinositide phosphatase expression and function, and protein kinase B (Akt) amplification. Furthermore, the loss of expression of the tumor suppressor thyroid hormone receptor beta (TRβ) is strongly associated with ATC. TRβ is known to suppress PI3K in follicular thyroid cancer and breast cancer by binding to the PI3K regulatory subunit p85α. However, the role of TRβ in suppressing PI3K signaling in ATC is not completely delineated. Here we report that TRβ indeed suppresses PI3K signaling in ATC cell lines through unreported genomic mechanisms, including a decrease in RTK expression and an increase in phosphoinositide and Akt phosphatase expression. Furthermore, the reintroduction and activation of TRβ in ATC cell lines enables an increase in the efficacy of the competitive PI3K inhibitors LY294002 and buparlisib on cell viability, migration, and suppression of PI3K signaling. These findings not only uncover additional tumor suppressor mechanisms of TRβ but shed light on the implication of TRβ status and activation on inhibitor efficacy in ATC tumors.
Collapse
Affiliation(s)
- Cole D Davidson
- Department of Pharmacology, Larner College of Medicine, Burlington, Vermont 05405, USA
- University of Vermont Cancer Center, Burlington, Vermont 05401, USA
| | - Eric L Bolf
- Department of Pharmacology, Larner College of Medicine, Burlington, Vermont 05405, USA
- University of Vermont Cancer Center, Burlington, Vermont 05401, USA
| | - Noelle E Gillis
- Department of Pharmacology, Larner College of Medicine, Burlington, Vermont 05405, USA
- University of Vermont Cancer Center, Burlington, Vermont 05401, USA
| | - Lauren M Cozzens
- Department of Pharmacology, Larner College of Medicine, Burlington, Vermont 05405, USA
| | - Jennifer A Tomczak
- Department of Pharmacology, Larner College of Medicine, Burlington, Vermont 05405, USA
| | - Frances E Carr
- Department of Pharmacology, Larner College of Medicine, Burlington, Vermont 05405, USA
- University of Vermont Cancer Center, Burlington, Vermont 05401, USA
| |
Collapse
|
72
|
Jin Y, Liu B, Younis MH, Huang G, Liu J, Cai W, Wei W. Next-Generation Molecular Imaging of Thyroid Cancer. Cancers (Basel) 2021; 13:3188. [PMID: 34202358 PMCID: PMC8268517 DOI: 10.3390/cancers13133188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/20/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
An essential aspect of thyroid cancer (TC) management is personalized and precision medicine. Functional imaging of TC with radioiodine and [18F]FDG has been frequently used in disease evaluation for several decades now. Recently, advances in molecular imaging have led to the development of novel tracers based on aptamer, peptide, antibody, nanobody, antibody fragment, and nanoparticle platforms. The emerging targets-including HER2, CD54, SHP2, CD33, and more-are promising targets for clinical translation soon. The significance of these tracers may be realized by outlining the way they support the management of TC. The provided examples focus on where preclinical investigations can be translated. Furthermore, advances in the molecular imaging of TC may inspire the development of novel therapeutic or theranostic tracers. In this review, we summarize TC-targeting probes which include transporter-based and immuno-based imaging moieties. We summarize the most recent evidence in this field and outline how these emerging strategies may potentially optimize clinical practice.
Collapse
Affiliation(s)
- Yuchen Jin
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
- Department of Nuclear Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200233, China
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Beibei Liu
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People’s Hospital Affiliatede to Shanghai Jiao Tong University, Shanghai 200233, China;
| | - Muhsin H. Younis
- Departments of Radiology and Medical Physics, University of Wisconsin–Madison, Madison, WI 53705-2275, USA;
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin–Madison, Madison, WI 53705-2275, USA;
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
| | - Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd., Shanghai 200127, China; (Y.J.); (G.H.); (J.L.)
| |
Collapse
|
73
|
Jiménez-Mora E, Gallego B, Díaz-Gago S, Lasa M, Baquero P, Chiloeches A. V600EBRAF Inhibition Induces Cytoprotective Autophagy through AMPK in Thyroid Cancer Cells. Int J Mol Sci 2021; 22:ijms22116033. [PMID: 34204950 PMCID: PMC8199856 DOI: 10.3390/ijms22116033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/17/2022] Open
Abstract
The dysregulation of autophagy is important in the development of many cancers, including thyroid cancer, where V600EBRAF is a main oncogene. Here, we analyse the effect of V600EBRAF inhibition on autophagy, the mechanisms involved in this regulation and the role of autophagy in cell survival of thyroid cancer cells. We reveal that the inhibition of V600EBRAF activity with its specific inhibitor PLX4720 or the depletion of its expression by siRNA induces autophagy in thyroid tumour cells. We show that V600EBRAF downregulation increases LKB1-AMPK signalling and decreases mTOR activity through a MEK/ERK-dependent mechanism. Moreover, we demonstrate that PLX4720 activates ULK1 and increases autophagy through the activation of the AMPK-ULK1 pathway, but not by the inhibition of mTOR. In addition, we find that autophagy blockade decreases cell viability and sensitize thyroid cancer cells to V600EBRAF inhibition by PLX4720 treatment. Finally, we generate a thyroid xenograft model to demonstrate that autophagy inhibition synergistically enhances the anti-proliferative and pro-apoptotic effects of V600EBRAF inhibition in vivo. Collectively, we uncover a new role of AMPK in mediating the induction of cytoprotective autophagy by V600EBRAF inhibition. In addition, these data establish a rationale for designing an integrated therapy targeting V600EBRAF and the LKB1-AMPK-ULK1-autophagy axis for the treatment of V600EBRAF-positive thyroid tumours.
Collapse
Affiliation(s)
- Eva Jiménez-Mora
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
| | - Beatriz Gallego
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
| | - Sergio Díaz-Gago
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
| | - Marina Lasa
- Departamento de Bioquímica-Instituto de Investigaciones Biomédicas “Alberto Sols”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28029 Madrid, Spain;
| | - Pablo Baquero
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
| | - Antonio Chiloeches
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
- Correspondence:
| |
Collapse
|
74
|
Wang Y, Hu Z, Ma W, Niu Y, Su J, Zhang L, Zhao P. Signal transducer and activator of transcription 3 inhibition alleviates resistance to BRAF inhibition in anaplastic thyroid cancer. Invest New Drugs 2021; 39:764-774. [PMID: 33245464 DOI: 10.1007/s10637-020-01024-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022]
Abstract
Anaplastic thyroid cancer (ATC) is a rare type of thyroid cancer (TC) with no effective therapeutic strategy. Although surgery, chemotherapy and radiation are all available for ATC treatment, the median survival for ATC patients is less than 6 months. In this study, we aimed to study on resistant mechanisms to B-Raf proto-oncogene serine/threonine kinase (BRAF) inhibitor and identify effective combinational therapy for ATC patients. TC cells were treated with Vemurafenib and cell apoptosis and viability were analyzed by flow cytometry and MTT assay. Monolayer and sphere cells were isolated from ATC cells to detect the mRNA level of stem cell markers and differentiation markers by RT-PCR. Phosphor-STAT3 level in sphere and monolayer cells was tested by Western blotting. The xenotransplantation animal model has established to analyze the anti-tumor effect of Vemurafenib and Stattic combinational therapy. Undifferentiated TC cells were resistant to Vemurafenib treatment. Sphere cells isolated from ATC showed no significant change in cell viability and apoptosis upon Vemurafenib treatment, and expressed a high level of stem cell marker and phosphor-STAT3. STAT3 inhibition enhanced the tumorigenic capacity and increased Vemurafenib sensitivity in ATC cell lines. Stattic significantly enhanced anti-tumor effect of Vemurafenib in mouse model. Our findings demonstrate that the combinational therapy of Vemurafenib and Stattic is an effective therapeutic treatment for ATC patients.
Collapse
Affiliation(s)
- Ying Wang
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Zhigang Hu
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Weiyuan Ma
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China
| | - Yong Niu
- Quyang People's Hospital, Taihang Road, Quyang County, Baoding, 071000, Hebei, China
| | - Jingwei Su
- Quyang People's Hospital, Taihang Road, Quyang County, Baoding, 071000, Hebei, China
| | - Lingxiang Zhang
- Xingtai Ninth Hospital, No.163 Jiankang East Road, Julu County, Xingtai, 054000, Hebei, China
| | - Pengxin Zhao
- The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
75
|
BRAF Inhibitors Induce Feedback Activation of RAS Pathway in Thyroid Cancer Cells. Int J Mol Sci 2021; 22:ijms22115744. [PMID: 34072194 PMCID: PMC8198461 DOI: 10.3390/ijms22115744] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022] Open
Abstract
BRAFV600E is the most frequent oncogenic mutation identified in papillary thyroid cancer (PTC). In PTC patients who do not respond to standard treatment, BRAF inhibitors are currently tested as alternative strategies. However, as observed for other targeted therapies, patients eventually develop drug resistance. The mechanisms of BRAF inhibitors response are still poorly understood in a thyroid cancer (TC) context. In this study, we investigated in BRAFV600E mutated TC cell lines the effects of Vemurafenib and Dabrafenib, two BRAF inhibitors currently used in a clinical setting. We assessed cell proliferation, and the expression and activity of the thyroid function related transporter NIS following the treatment with BRAF inhibitors. In addition, we investigated the global gene expression by microarray, the relevant modulated biological processes by gene set enrichment analysis (GSEA), and TC specific gene signatures related to MAPK pathway activation, thyroid differentiation, and transcriptional profile associated with BRAFV600E or RAS mutation. We found that both inhibitors induce antiproliferative and redifferentiative effects on TC cells, as well as a rewiring of the MAPK pathway related to RAS signaling. Our results suggest a possible mechanism of drug response to the BRAF inhibitors Vemurafenib or Dabrafenib, supporting very recent findings in TC patients treated with targeted therapies.
Collapse
|
76
|
Kholodenko BN, Rauch N, Kolch W, Rukhlenko OS. A systematic analysis of signaling reactivation and drug resistance. Cell Rep 2021; 35:109157. [PMID: 34038718 PMCID: PMC8202068 DOI: 10.1016/j.celrep.2021.109157] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/24/2021] [Accepted: 04/29/2021] [Indexed: 01/07/2023] Open
Abstract
Increasing evidence suggests that the reactivation of initially inhibited signaling pathways causes drug resistance. Here, we analyze how network topologies affect signaling responses to drug treatment. Network-dependent drug resistance is commonly attributed to negative and positive feedback loops. However, feedback loops by themselves cannot completely reactivate steady-state signaling. Newly synthesized negative feedback regulators can induce a transient overshoot but cannot fully restore output signaling. Complete signaling reactivation can only occur when at least two routes, an activating and inhibitory, connect an inhibited upstream protein to a downstream output. Irrespective of the network topology, drug-induced overexpression or increase in target dimerization can restore or even paradoxically increase downstream pathway activity. Kinase dimerization cooperates with inhibitor-mediated alleviation of negative feedback. Our findings inform drug development by considering network context and optimizing the design drug combinations. As an example, we predict and experimentally confirm specific combinations of RAF inhibitors that block mutant NRAS signaling. Kholodenko et al. uncover signaling network circuitries and molecular mechanisms necessary and sufficient for complete reactivation or overshoot of steady-state signaling after kinase inhibitor treatment. The two means to revive signaling output fully are through network topology or reactivation of the kinase activity of the primary drug target. Blocking RAF dimer activity by a combination of type I½ and type II RAF inhibitors efficiently blocks mutant NRAS-driven ERK signaling.
Collapse
Affiliation(s)
- Boris N Kholodenko
- Systems Biology Ireland, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland; Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
| | - Nora Rauch
- Systems Biology Ireland, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Walter Kolch
- Systems Biology Ireland, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Oleksii S Rukhlenko
- Systems Biology Ireland, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
77
|
Kulkarni O, Sugier PE, Guibon J, Boland-Augé A, Lonjou C, Bacq-Daian D, Olaso R, Rubino C, Souchard V, Rachedi F, Lence-Anta JJ, Ortiz RM, Xhaard C, Laurent-Puig P, Mulot C, Guizard AV, Schvartz C, Boutron-Ruault MC, Ostroumova E, Kesminiene A, Deleuze JF, Guénel P, De Vathaire F, Truong T, Lesueur F. Gene network and biological pathways associated with susceptibility to differentiated thyroid carcinoma. Sci Rep 2021; 11:8932. [PMID: 33903625 PMCID: PMC8076215 DOI: 10.1038/s41598-021-88253-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/09/2021] [Indexed: 12/11/2022] Open
Abstract
Variants identified in earlier genome-wide association studies (GWAS) on differentiated thyroid carcinoma (DTC) explain about 10% of the overall estimated genetic contribution and could not provide complete insights into biological mechanisms involved in DTC susceptibility. Integrating systems biology information from model organisms, genome-wide expression data from tumor and matched normal tissue and GWAS data could help identifying DTC-associated genes, and pathways or functional networks in which they are involved. We performed data mining of GWAS data of the EPITHYR consortium (1551 cases and 1957 controls) using various pathways and protein-protein interaction (PPI) annotation databases and gene expression data from The Cancer Genome Atlas. We identified eight DTC-associated genes at known loci 2q35 (DIRC3), 8p12 (NRG1), 9q22 (FOXE1, TRMO, HEMGN, ANP32B, NANS) and 14q13 (MBIP). Using the EW_dmGWAS approach we found that gene networks related to glycogenolysis, glycogen metabolism, insulin metabolism and signal transduction pathways associated with muscle contraction were overrepresented with association signals (false discovery rate adjusted p-value < 0.05). Additionally, suggestive association of 21 KEGG and 75 REACTOME pathways with DTC indicate a link between DTC susceptibility and functions related to metabolism of cholesterol, amino sugar and nucleotide sugar metabolism, steroid biosynthesis, and downregulation of ERBB2 signaling pathways. Together, our results provide novel insights into biological mechanisms contributing to DTC risk.
Collapse
Affiliation(s)
- Om Kulkarni
- Inserm, U900, Institut Curie, PSL University, Mines ParisTech, 75248, Paris, France
| | | | - Julie Guibon
- Inserm, U900, Institut Curie, PSL University, Mines ParisTech, 75248, Paris, France
- Université Paris-Saclay, UVSQ, Gustave Roussy, Inserm, CESP, 94807, Villejuif, France
| | - Anne Boland-Augé
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057, Evry, France
| | - Christine Lonjou
- Inserm, U900, Institut Curie, PSL University, Mines ParisTech, 75248, Paris, France
| | - Delphine Bacq-Daian
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057, Evry, France
| | - Robert Olaso
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057, Evry, France
| | - Carole Rubino
- Université Paris-Saclay, UVSQ, Gustave Roussy, Inserm, CESP, 94807, Villejuif, France
| | - Vincent Souchard
- Université Paris-Saclay, UVSQ, Gustave Roussy, Inserm, CESP, 94807, Villejuif, France
| | - Frédérique Rachedi
- Centre Hospitalier Territorial de Polynésie Française, CHTPF, Pirae, Tahiti, 98713, Papeete, French Polynesia
| | | | - Rosa Maria Ortiz
- Instituto Nacional de Oncologia y de Radiobiologia, INOR, La Havana, Cuba
| | - Constance Xhaard
- Université Paris-Saclay, UVSQ, Gustave Roussy, Inserm, CESP, 94807, Villejuif, France
- University of Lorraine, INSERM CIC 1433, Nancy CHRU, Inserm U1116, FCRIN, INI-CRCT, 54000, Nancy, France
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, EPIGENETEC, 75006, Paris, France
| | - Claire Mulot
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, EPIGENETEC, 75006, Paris, France
| | - Anne-Valérie Guizard
- Registre Général des Tumeurs du Calvados, Centre François Baclesse, 14000, Caen, France
- Inserm U1086-UCNB, Cancers and Prevention, 14000, Caen, France
| | - Claire Schvartz
- Registre des Cancers Thyroïdiens, Institut Jean Godinot, 51100, Reims, France
| | | | - Evgenia Ostroumova
- Environment and Radiation Section, International Agency for Research on Cancer, 69008, Lyon, France
| | - Ausrele Kesminiene
- Environment and Radiation Section, International Agency for Research on Cancer, 69008, Lyon, France
| | - Jean-François Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057, Evry, France
| | - Pascal Guénel
- Université Paris-Saclay, UVSQ, Gustave Roussy, Inserm, CESP, 94807, Villejuif, France
| | - Florent De Vathaire
- Université Paris-Saclay, UVSQ, Gustave Roussy, Inserm, CESP, 94807, Villejuif, France
| | - Thérèse Truong
- Université Paris-Saclay, UVSQ, Gustave Roussy, Inserm, CESP, 94807, Villejuif, France
| | - Fabienne Lesueur
- Inserm, U900, Institut Curie, PSL University, Mines ParisTech, 75248, Paris, France.
| |
Collapse
|
78
|
Oh JM, Ahn BC. Molecular mechanisms of radioactive iodine refractoriness in differentiated thyroid cancer: Impaired sodium iodide symporter (NIS) expression owing to altered signaling pathway activity and intracellular localization of NIS. Theranostics 2021; 11:6251-6277. [PMID: 33995657 PMCID: PMC8120202 DOI: 10.7150/thno.57689] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/22/2021] [Indexed: 12/16/2022] Open
Abstract
The advanced, metastatic differentiated thyroid cancers (DTCs) have a poor prognosis mainly owing to radioactive iodine (RAI) refractoriness caused by decreased expression of sodium iodide symporter (NIS), diminished targeting of NIS to the cell membrane, or both, thereby decreasing the efficacy of RAI therapy. Genetic aberrations (such as BRAF, RAS, and RET/PTC rearrangements) have been reported to be prominently responsible for the onset, progression, and dedifferentiation of DTCs, mainly through the activation of mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/AKT signaling pathways. Eventually, these alterations result in a lack of NIS and disabling of RAI uptake, leading to the development of resistance to RAI therapy. Over the past decade, promising approaches with various targets have been reported to restore NIS expression and RAI uptake in preclinical studies. In this review, we summarized comprehensive molecular mechanisms underlying the dedifferentiation in RAI-refractory DTCs and reviews strategies for restoring RAI avidity by tackling the mechanisms.
Collapse
|
79
|
Haikala HM, Jänne PA. Thirty Years of HER3: From Basic Biology to Therapeutic Interventions. Clin Cancer Res 2021; 27:3528-3539. [PMID: 33608318 DOI: 10.1158/1078-0432.ccr-20-4465] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/13/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
HER3 is a pseudokinase member of the EGFR family having a role in both tumor progression and drug resistance. Although HER3 was discovered more than 30 years ago, no therapeutic interventions have reached clinical approval to date. Because the evidence of the importance of HER3 is accumulating, increased amounts of preclinical and clinical trials with HER3-targeting agents are emerging. In this review article, we discuss the most recent HER3 biology in tumorigenic events and drug resistance and provide an overview of the current and emerging strategies to target HER3.
Collapse
Affiliation(s)
- Heidi M Haikala
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
80
|
Han Y, Yu X, Yin Y, Lv Z, Jia C, Liao Y, Sun H, Liu T, Cong L, Fei Z, Fu D, Cong X, Qu S. Identification of Potential BRAF Inhibitor Joint Therapy Targets in PTC based on WGCAN and DCGA. J Cancer 2021; 12:1779-1791. [PMID: 33613767 PMCID: PMC7890315 DOI: 10.7150/jca.51551] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/28/2020] [Indexed: 01/08/2023] Open
Abstract
As the most common mutation in papillary thyroid cancer (PTC), B-type Raf kinase V600E mutation (BRAFV600E ) has become an important target for the clinical treatment of PTC. However, the clinical application still faces the problem of resistance to BRAF inhibitors (BRAFi). Therefore, exploring BRAFV600E-associated prognostic factors to providing potential joint targets is important for combined targeted therapy with BRAFi. In this study, we combined transcript data and clinical information from 199 BRAF wild-type (BRAFWT ) patients and 283 BRAFV600E mutant patients collected from The Cancer Genome Atlas (TCGA), and screened 455 BRAFV600E- associated genes through differential analysis and weighted gene co-expression network analysis. Based on these BRAFV600E -associated genes, we performed functional enrichment analysis and co-expression differential analysis and constructed a core co-expression network. Next, genes in the differential co-expression network were used to predict drugs for therapy in the crowd extracted expression of differential signatures (CREEDS) database, and the key genes were selected based on the hub co-expression network through survival analyses and receiver operating characteristic (ROC) curve analyses. Finally, we obtained eight BRAFV600E -associated biomarkers with both prognostic and diagnostic values as potential BRAFi joint targets, including FN1, MET, SLC34A2, NGEF, TBC1D2, PLCD3, PROS1, and NECTIN4. Among these genes, FN1, MET, PROS1, and TBC1D2 were validated through GEO database. Two novel biomarkers, PROS1 and TBC1D2, were further validated by qRT-PCR experiment. Besides, we obtained four potential targeted drugs that could be used in combination with BRAFi to treat PTC, including MET inhibitor, ERBB3 inhibitor, anti-NaPi2b antibody-drug conjugate, and carboplatin through literature review. The study provided potential drug targets for combination therapy with BRAFi for PTC to overcome the drug resistance for BRAFi.
Collapse
Affiliation(s)
- YaLi Han
- Shanghai Center for Thyroid Disease, Shanghai Tenth People's Hospital, Shanghai, China
| | - XiaQing Yu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - YuZhen Yin
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhongwei Lv
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - ChengYou Jia
- Shanghai Center for Thyroid Disease, Shanghai Tenth People's Hospital, Shanghai, China
| | - Yina Liao
- Shanghai Center for Thyroid Disease, Shanghai Tenth People's Hospital, Shanghai, China
| | - Hongyan Sun
- Department of biobank, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Tie Liu
- Department of biobank, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Lele Cong
- Department of biobank, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - ZhaoLiang Fei
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Da Fu
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xianling Cong
- Department of biobank, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Shen Qu
- Shanghai Center for Thyroid Disease, Shanghai Tenth People's Hospital, Shanghai, China
| |
Collapse
|
81
|
Su X, Li P, Han B, Jia H, Liang Q, Wang H, Gu M, Cai J, Li S, Zhou Y, Yi X, Wei W. Vitamin C sensitizes BRAF V600E thyroid cancer to PLX4032 via inhibiting the feedback activation of MAPK/ERK signal by PLX4032. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:34. [PMID: 33468157 PMCID: PMC7816401 DOI: 10.1186/s13046-021-01831-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/05/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND BRAFV600E mutation is the most common mutation in thyroid cancer. It strongly activates MAPK/ERK pathway and indicates an invasive subtype of thyroid cancer. PLX4032 is a selective oral inhibitor of the BRAFV600 kinase although with limited effect in treating this panel of thyroid cancer, due to the feedback activation of MAPK/ERK as well as PI3K/AKT pathways. It was investigated that Vitamin C plays a positive role in inhibiting these pathways in thyroid cancer. However, whether Vitamin C could enhance the antitumor effect of PLX4032 remains largely unclear. METHODS The antitumor efficacy of combination therapy with PLX4032 and Vitamin C on BRAFMT thyroid cancer cell was assessed by the MTT assay, EdU assay and colony formation, Chou-Talalay way was employed to analyze the synergistic effect. Flow cytometry were employed to assess cells' apoptosis and cell cycle arrest in response to combination therapy. Xenograft models were used to test its in vivo antitumor activity. Western blot and IHC were applied to investigate the mechanism underlying synergistic effect. RESULTS PLX4032 or Vitamin C monotherapy was mildly effective in treating BRAFMT thyroid cancer cell and xenografts model. The combination therapy significantly inhibited cancer cell proliferation and tumor growth in nude mice, and induced cell apoptosis and cell cycle arrest compared to either monotherapy. PLX4032 monotherapy induced feedback activation of MAPK/ERK as well as PI3K/AKT pathway; while combination therapy significantly relieved this feedback. CONCLUSION Vitamin C promotes the antitumor effect of PLX4032 in BRAFMT thyroid cancer cell and xenografts model via relieving the feedback activation of MAPK/ERK as well as PI3K/AKT pathway. PLX4032/Vitamin C combination may be a potential therapeutic approach to treat BRAFMT thyroid cancer.
Collapse
Affiliation(s)
- Xi Su
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, ShenZhen Peking University-The Hong Kong University of Science and Technology Medical Centre, No.1120, LianHua Road, FuTian district, Shenzhen, 518036, China
| | - Peng Li
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, ShenZhen Peking University-The Hong Kong University of Science and Technology Medical Centre, No.1120, LianHua Road, FuTian district, Shenzhen, 518036, China
| | - Bin Han
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, ShenZhen Peking University-The Hong Kong University of Science and Technology Medical Centre, No.1120, LianHua Road, FuTian district, Shenzhen, 518036, China
| | - Hao Jia
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, ShenZhen Peking University-The Hong Kong University of Science and Technology Medical Centre, No.1120, LianHua Road, FuTian district, Shenzhen, 518036, China
| | - Qingzhuang Liang
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, ShenZhen Peking University-The Hong Kong University of Science and Technology Medical Centre, No.1120, LianHua Road, FuTian district, Shenzhen, 518036, China
| | - Haichao Wang
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, ShenZhen Peking University-The Hong Kong University of Science and Technology Medical Centre, No.1120, LianHua Road, FuTian district, Shenzhen, 518036, China
| | - Mengwei Gu
- Department of Merchandising, Walmart (China) Investment Co., Ltd, Shenzhen, China
| | - Jiaxuan Cai
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, ShenZhen Peking University-The Hong Kong University of Science and Technology Medical Centre, No.1120, LianHua Road, FuTian district, Shenzhen, 518036, China
| | - Shaolei Li
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, ShenZhen Peking University-The Hong Kong University of Science and Technology Medical Centre, No.1120, LianHua Road, FuTian district, Shenzhen, 518036, China
| | - Yaqi Zhou
- Department of Otorhinolaryngology, Peking University Shenzhen Hospital, ShenZhen Peking University-The Hong Kong University of Science and Technology Medical Centre, Shenzhen, China
| | - Xin Yi
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, ShenZhen Peking University-The Hong Kong University of Science and Technology Medical Centre, No.1120, LianHua Road, FuTian district, Shenzhen, 518036, China
| | - Wei Wei
- Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, ShenZhen Peking University-The Hong Kong University of Science and Technology Medical Centre, No.1120, LianHua Road, FuTian district, Shenzhen, 518036, China.
| |
Collapse
|
82
|
Ringel MD. New Horizons: Emerging Therapies and Targets in Thyroid Cancer. J Clin Endocrinol Metab 2021; 106:e382-e388. [PMID: 32977343 PMCID: PMC7765632 DOI: 10.1210/clinem/dgaa687] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/23/2020] [Indexed: 12/21/2022]
Abstract
The treatment of patients with progressive metastatic follicular cell-derived and medullary thyroid cancers that do not respond to standard therapeutic modalities presents a therapeutic challenge. As a deeper understanding of the molecular drivers for these tumors has occurred and more potent and specific compounds are developed, the number of Food and Drug Administration (FDA)-approved treatments for thyroid cancer has expanded. In addition, with the advent of disease-agnostic target-directed FDA approvals an ever-broadening number of therapeutic options are available for clinicians and patients. However, to date, complete remissions are rare, the average durations of response are relatively modest, and toxicities are common. These factors accentuate the need for further understanding of the mechanisms of resistance that result in treatment failures, the development of biomarkers that can improve patient selection for treatment earlier in the disease process, and the continued need for new therapeutic strategies. In this article, recent approvals relevant to thyroid cancer will be discussed along with selected new potential avenues that might be exploited for future therapies.
Collapse
Affiliation(s)
- Matthew D Ringel
- Division of Endocrinology, Diabetes, and Metabolism and Cancer Biology Program, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio
- Correspondence and Reprint Requests: Matthew D. Ringel, MD, McCampbell Hall South, Room 565, 1581 Dodd Drive, Columbus, OH 43210, USA. E-mail:
| |
Collapse
|
83
|
Fullmer T, Cabanillas ME, Zafereo M. Novel Therapeutics in Radioactive Iodine-Resistant Thyroid Cancer. Front Endocrinol (Lausanne) 2021; 12:720723. [PMID: 34335481 PMCID: PMC8321684 DOI: 10.3389/fendo.2021.720723] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 06/24/2021] [Indexed: 01/18/2023] Open
Abstract
Iodine-resistant cancers account for the vast majority of thyroid related mortality and, until recently, there were limited therapeutic options. However, over the last decade our understanding of the molecular foundation of thyroid function and carcinogenesis has driven the development of many novel therapeutics. These include FDA approved tyrosine kinase inhibitors and small molecular inhibitors of VEGFR, BRAF, MEK, NTRK and RET, which collectively have significantly changed the prognostic outlook for this patient population. Some therapeutics can re-sensitize de-differentiated cancers to iodine, allowing for radioactive iodine treatment and improved disease control. Remarkably, there is now an FDA approved treatment for BRAF-mutated patients with anaplastic thyroid cancer, previously considered invariably and rapidly fatal. The treatment landscape for iodine-resistant thyroid cancer is changing rapidly with many new targets, therapeutics, clinical trials, and approved treatments. We provide an up-to-date review of novel therapeutic options in the treatment of iodine-resistant thyroid cancer.
Collapse
Affiliation(s)
- Tanner Fullmer
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria E. Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mark Zafereo
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Mark Zafereo,
| |
Collapse
|
84
|
Saqcena M, Leandro-Garcia LJ, Maag JLV, Tchekmedyian V, Krishnamoorthy GP, Tamarapu PP, Tiedje V, Reuter V, Knauf JA, de Stanchina E, Xu B, Liao XH, Refetoff S, Ghossein R, Chi P, Ho AL, Koche RP, Fagin JA. SWI/SNF Complex Mutations Promote Thyroid Tumor Progression and Insensitivity to Redifferentiation Therapies. Cancer Discov 2020; 11:1158-1175. [PMID: 33318036 DOI: 10.1158/2159-8290.cd-20-0735] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/16/2020] [Accepted: 12/09/2020] [Indexed: 12/21/2022]
Abstract
Mutations of subunits of the SWI/SNF chromatin remodeling complexes occur commonly in cancers of different lineages, including advanced thyroid cancers. Here we show that thyroid-specific loss of Arid1a, Arid2, or Smarcb1 in mouse BRAFV600E-mutant tumors promotes disease progression and decreased survival, associated with lesion-specific effects on chromatin accessibility and differentiation. As compared with normal thyrocytes, BRAFV600E-mutant mouse papillary thyroid cancers have decreased lineage transcription factor expression and accessibility to their target DNA binding sites, leading to impairment of thyroid-differentiated gene expression and radioiodine incorporation, which is rescued by MAPK inhibition. Loss of individual SWI/SNF subunits in BRAF tumors leads to a repressive chromatin state that cannot be reversed by MAPK pathway blockade, rendering them insensitive to its redifferentiation effects. Our results show that SWI/SNF complexes are central to the maintenance of differentiated function in thyroid cancers, and their loss confers radioiodine refractoriness and resistance to MAPK inhibitor-based redifferentiation therapies. SIGNIFICANCE: Reprogramming cancer differentiation confers therapeutic benefit in various disease contexts. Oncogenic BRAF silences genes required for radioiodine responsiveness in thyroid cancer. Mutations in SWI/SNF genes result in loss of chromatin accessibility at thyroid lineage specification genes in BRAF-mutant thyroid tumors, rendering them insensitive to the redifferentiation effects of MAPK blockade.This article is highlighted in the In This Issue feature, p. 995.
Collapse
Affiliation(s)
- Mahesh Saqcena
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Jesper L V Maag
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vatche Tchekmedyian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gnana P Krishnamoorthy
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Prasanna P Tamarapu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vera Tiedje
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vincent Reuter
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jeffrey A Knauf
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bin Xu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Samuel Refetoff
- Departments of Medicine and Pediatrics and the Committee on Genetics, The University of Chicago, Chicago, Illinois
| | - Ronald Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ping Chi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James A Fagin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
85
|
Ney GM, McKay L, Koschmann C, Mody R, Li Q. The Emerging Role of Ras Pathway Signaling in Pediatric Cancer. Cancer Res 2020; 80:5155-5163. [PMID: 32907837 PMCID: PMC10081825 DOI: 10.1158/0008-5472.can-20-0916] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/04/2020] [Accepted: 09/02/2020] [Indexed: 11/16/2022]
Abstract
As genomic sequencing has become more widely available, the high prevalence of Ras pathway mutations in pediatric diseases has begun to emerge. Germline Ras-activating mutations have been known to contribute to cancer predisposition in a group of disorders known as the RASopathies, and now large pediatric sequencing studies have identified frequent somatic Ras pathway alterations across a diverse group of pediatric malignancies. These include glial brain tumors, relapsed high-risk neuroblastoma, embryonal rhabdomyosarcoma, acute myeloid leukemia, and relapsed acute lymphoblastic leukemia, and their prognostic impact is becoming increasingly better understood. Clinically, there has been success in targeting the Ras pathway in pediatric diseases, including the use of MEK inhibitors in plexiform neurofibromas associated with neurofibromatosis type 1 and the use of Ras pathway inhibitors in low-grade gliomas. Given the importance of this pathway in pediatric cancer, it is imperative that future studies strive to better understand the functional significance of these mutations, including their role in tumor growth and treatment resistance and how they can be better targeted to improve outcomes.
Collapse
Affiliation(s)
- Gina M Ney
- Department of Pediatrics, University of Michigan, Ann Arbor, MI.
| | - Laura McKay
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Carl Koschmann
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Rajen Mody
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Qing Li
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI.
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
86
|
Chen H, Smaill JB, Liu T, Ding K, Lu X. Small-Molecule Inhibitors Directly Targeting KRAS as Anticancer Therapeutics. J Med Chem 2020; 63:14404-14424. [PMID: 33225706 DOI: 10.1021/acs.jmedchem.0c01312] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
KRAS, the most frequently mutated oncogene, plays a predominant role in driving initiation and progression of cancers. Decades of effort to target KRAS using small molecules has been unsuccessful, causing KRAS to be considered an "undruggable" cancer target. However, this view began to change recently, as drug discovery techniques have developed several KRAS G12C allosteric inhibitors that are currently being evaluated in clinical trials. Herein we provide an in-depth analysis of the structure and binding pockets of KRAS, medicinal chemistry optimization processes, and the biological characterization of small-molecule inhibitors that directly target KRAS, including covalent allosteric inhibitors specific for the G12C mutant, GTP-competitive inhibitors targeting the nucleotide-binding site, and protein-protein interaction inhibitors that bind in the switch I/II pocket or the A59 site. Additionally, we propose potential challenges faced by these new classes of KRAS inhibitors under clinical evaluation.
Collapse
Affiliation(s)
- Hao Chen
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Jeff B Smaill
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Tongzheng Liu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Xiaoyun Lu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| |
Collapse
|
87
|
Wang J, Pollard K, Calizo A, Pratilas CA. Activation of Receptor Tyrosine Kinases Mediates Acquired Resistance to MEK Inhibition in Malignant Peripheral Nerve Sheath Tumors. Cancer Res 2020; 81:747-762. [PMID: 33203698 DOI: 10.1158/0008-5472.can-20-1992] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/08/2020] [Accepted: 11/12/2020] [Indexed: 11/16/2022]
Abstract
Malignant peripheral nerve sheath tumors often arise in patients with neurofibromatosis type 1 and are among the most treatment-refractory types of sarcoma. Overall survival in patients with relapsed disease remains poor, and thus novel therapeutic approaches are needed. NF1 is essential for negative regulation of RAS activity and is altered in about 90% of malignant peripheral nerve sheath tumors (MPNST). A complex interplay of upstream signaling and parallel RAS-driven pathways characterizes NF1-driven tumorigenesis, and inhibiting more than one RAS effector pathway is therefore necessary. To devise potential combination therapeutic strategies, we identified actionable alterations in signaling that underlie adaptive and acquired resistance to MEK inhibitor (MEKi). Using a series of proteomic, biochemical, and genetic approaches in an in vitro model of MEKi resistance provided a rationale for combination therapies. HGF/MET signaling was elevated in the MEKi-resistant model. HGF overexpression conferred resistance to MEKi in parental cells. Depletion of HGF or MET restored sensitivity of MEKi-resistant cells to MEKi. Finally, a combination of MEK and MET inhibition demonstrated activity in models of MPNST and may therefore be effective in patients with MPNST harboring genetic alterations in NF1. SIGNIFICANCE: This study demonstrates that MEKi plus MET inhibitor may delay or prevent a novel mechanism of acquired MEKi resistance, with clinical implications for MPNST patients harboring NF1 alterations.
Collapse
Affiliation(s)
- Jiawan Wang
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kai Pollard
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ana Calizo
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine A Pratilas
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
88
|
Kim D, Xue JY, Lito P. Targeting KRAS(G12C): From Inhibitory Mechanism to Modulation of Antitumor Effects in Patients. Cell 2020; 183:850-859. [PMID: 33065029 PMCID: PMC7669705 DOI: 10.1016/j.cell.2020.09.044] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/24/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023]
Abstract
KRAS mutations are among the most common genetic alterations in lung, colorectal, and pancreatic cancers. Direct inhibition of KRAS oncoproteins has been a long-standing pursuit in precision oncology, one established shortly after the discovery of RAS mutations in human cancer cells nearly 40 years ago. Recent advances in medicinal chemistry have established inhibitors targeting KRAS(G12C), a mutation found in ∼13% of lung adenocarcinomas and, at a lower frequency, in other cancers. Preclinical studies describing their discovery and mechanism of action, coupled with emerging clinical data from patients treated with these drugs, have sparked a renewed enthusiasm in the study of KRAS and its therapeutic potential. Here, we discuss how these advances are reshaping the fundamental aspects of KRAS oncoprotein biology and the strides being made toward improving patient outcomes in the clinic.
Collapse
Affiliation(s)
- Dongsung Kim
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Jenny Yaohua Xue
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Piro Lito
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
89
|
Tabatabaeian H, Peiling Yang S, Tay Y. Non-Coding RNAs: Uncharted Mediators of Thyroid Cancer Pathogenesis. Cancers (Basel) 2020; 12:E3264. [PMID: 33158279 PMCID: PMC7694276 DOI: 10.3390/cancers12113264] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 12/24/2022] Open
Abstract
Thyroid cancer is the most prevalent malignancy of the endocrine system and the ninth most common cancer globally. Despite the advances in the management of thyroid cancer, there are critical issues with the diagnosis and treatment of thyroid cancer that result in the poor overall survival of undifferentiated and metastatic thyroid cancer patients. Recent studies have revealed the role of different non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) that are dysregulated during thyroid cancer development or the acquisition of resistance to therapeutics, and may play key roles in treatment failure and poor prognosis of the thyroid cancer patients. Here, we systematically review the emerging roles and molecular mechanisms of ncRNAs that regulate thyroid tumorigenesis and drug response. We then propose the potential clinical implications of ncRNAs as novel diagnostic and prognostic biomarkers for thyroid cancer.
Collapse
Affiliation(s)
- Hossein Tabatabaeian
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
| | - Samantha Peiling Yang
- Endocrinology Division, Department of Medicine, National University Hospital, Singapore 119228, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yvonne Tay
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
90
|
Gerosa L, Chidley C, Fröhlich F, Sanchez G, Lim SK, Muhlich J, Chen JY, Vallabhaneni S, Baker GJ, Schapiro D, Atanasova MI, Chylek LA, Shi T, Yi L, Nicora CD, Claas A, Ng TSC, Kohler RH, Lauffenburger DA, Weissleder R, Miller MA, Qian WJ, Wiley HS, Sorger PK. Receptor-Driven ERK Pulses Reconfigure MAPK Signaling and Enable Persistence of Drug-Adapted BRAF-Mutant Melanoma Cells. Cell Syst 2020; 11:478-494.e9. [PMID: 33113355 DOI: 10.1016/j.cels.2020.10.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 07/21/2020] [Accepted: 10/03/2020] [Indexed: 02/06/2023]
Abstract
Targeted inhibition of oncogenic pathways can be highly effective in halting the rapid growth of tumors but often leads to the emergence of slowly dividing persister cells, which constitute a reservoir for the selection of drug-resistant clones. In BRAFV600E melanomas, RAF and MEK inhibitors efficiently block oncogenic signaling, but persister cells emerge. Here, we show that persister cells escape drug-induced cell-cycle arrest via brief, sporadic ERK pulses generated by transmembrane receptors and growth factors operating in an autocrine/paracrine manner. Quantitative proteomics and computational modeling show that ERK pulsing is enabled by rewiring of mitogen-activated protein kinase (MAPK) signaling: from an oncogenic BRAFV600E monomer-driven configuration that is drug sensitive to a receptor-driven configuration that involves Ras-GTP and RAF dimers and is highly resistant to RAF and MEK inhibitors. Altogether, this work shows that pulsatile MAPK activation by factors in the microenvironment generates a persistent population of melanoma cells that rewires MAPK signaling to sustain non-genetic drug resistance.
Collapse
Affiliation(s)
- Luca Gerosa
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher Chidley
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Fabian Fröhlich
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Gabriela Sanchez
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sang Kyun Lim
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Jeremy Muhlich
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Jia-Yun Chen
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sreeram Vallabhaneni
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Gregory J Baker
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Denis Schapiro
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Mariya I Atanasova
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Lily A Chylek
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Tujin Shi
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Lian Yi
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Carrie D Nicora
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Allison Claas
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Thomas S C Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA
| | - Rainer H Kohler
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - H Steven Wiley
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
91
|
Abstract
INTRODUCTION Fine-needle aspiration (FNA) is an important diagnostic tool for the evaluation of thyroid nodules. However, in almost ¼ of all nodules submitted to FNA cytology is indeterminate. Since the majority of genetic alterations in thyroid cancer have been identified, the use of molecular testing platforms has been endorsed by American Thyroid Association for management of indeterminate nodules. EVIDENCE ACQUISITION Several commercial tests were based on mRNA expression of FNA samples (Afirma®, Veracyte, South San Francisco, CA, USA) while others detect DNA alterations (ThyroSeq, UPMC, Pittsburgh, PA, USA). Noncommercial tests detect limited number of point mutations or re-arrangements (gene panels). Literature study included a Pubmed research for adult original studies from 2003 to 2020, focusing on terms such as "molecular tests," "nodules with indeterminate AUS/FLUS and FN/SFN cytology." EVIDENCE SYNTHESIS Gene expression profile tests serve as "rule out" tests due to their high negative predictive value and perform better in a setting of low cancer pretest probability. Genetic alteration platforms display high positive predictive value and serve as rather "rule in" tests but their diagnostic accuracy is hampered either because a small proportion of nodules does not harbor any of these alterations targeted (gene panels) or because commonly identified RAS mutations can also be found in benign nodules. CONCLUSIONS Next generation sequencing development and incorporation of other genetic markers such as miRNA can improve diagnostic accuracy of molecular tests.
Collapse
Affiliation(s)
- Olga Karapanou
- 401Hellenic Army General Military Hospital of Athens, Athens, Greece -
| |
Collapse
|
92
|
Wang J, Pollard K, Allen AN, Tomar T, Pijnenburg D, Yao Z, Rodriguez FJ, Pratilas CA. Combined Inhibition of SHP2 and MEK Is Effective in Models of NF1-Deficient Malignant Peripheral Nerve Sheath Tumors. Cancer Res 2020; 80:5367-5379. [PMID: 33032988 DOI: 10.1158/0008-5472.can-20-1365] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/18/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022]
Abstract
Loss of the RAS GTPase-activating protein (RAS-GAP) NF1 drives aberrant activation of RAS/MEK/ERK signaling and other effector pathways in the majority of malignant peripheral nerve sheath tumors (MPNST). These dysregulated pathways represent potential targets for therapeutic intervention. However, studies of novel single agents including MEK inhibitors (MEKi) have demonstrated limited efficacy both preclinically and clinically, with little advancement in overall patient survival. By interrogation of kinome activity through an unbiased screen and targeted evaluation of the signaling response to MEK inhibition, we have identified global activation of upstream receptor tyrosine kinases (RTK) that converges on activation of RAS as a mechanism to limit sensitivity to MEK inhibition. As no direct inhibitors of pan-RAS were available, an inhibitor of the protein tyrosine phosphatase SHP2, a critical mediator of RAS signal transduction downstream of multiple RTK, represented an alternate strategy. The combination of MEKi plus SHP099 was superior to MEKi alone in models of NF1-MPNST, including those with acquired resistance to MEKi. Our findings have immediate translational implications and may inform future clinical trials for patients with MPNST harboring alterations in NF1. SIGNIFICANCE: Combined inhibition of MEK and SHP2 is effective in models of NF1-MPNST, both those naïve to and those resistant to MEKi, as well as in the MPNST precursor lesion plexiform neurofibroma.
Collapse
Affiliation(s)
- Jiawan Wang
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kai Pollard
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Amy N Allen
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tushar Tomar
- PamGene International BV, 's-Hertogenbosch, the Netherlands
| | | | - Zhan Yao
- Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fausto J Rodriguez
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine A Pratilas
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
93
|
Hernando J, Ros J, Arroyo A, Capdevila J. Clinical and Translational Challenges in Thyroid Cancer. Curr Med Chem 2020; 27:4806-4822. [PMID: 32056516 DOI: 10.2174/0929867327666200214125712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/23/2020] [Accepted: 01/31/2020] [Indexed: 01/15/2023]
Abstract
Thyroid cancer is the most common endocrine malignancy and it accounts for 1% of all newly diagnosed tumors. Approximately 10% of patients with differentiated thyroid carcinomas (DTC) and 30% with medullary thyroid carcinoma (MTC) could not be cured with locoregional treatment and could develop metastatic disease. In addition, one of the most aggressive solid tumors can arise from the thyroid gland, the anaplastic thyroid carcinoma, with a median overall survival of less than 6 months. Currently, only four drugs are approved for the treatment of DTC and MTC and several unmet needs are focusing the scientific discussions, including the resistant setting, the off-target side effects that may reduce the efficacy and the molecular knowledge-based combinations. In this review, we aimed to discuss the current molecular landscape and treatment of thyroid cancers, and the ongoing clinical and translational research lines focusing on new drugs and drug combinations to improve the inhibition of driver mutations, such as BRAF and RET, and how systemic therapies that improved outcomes of other cancer types, like immunotherapy and peptide receptor radionuclide therapy, may play a role in the future management of advanced thyroid cancers.
Collapse
Affiliation(s)
- Jorge Hernando
- Medical Oncology Department, Gastrointestinal and Endocrine Tumor Unit, Vall d´Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), Pg Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - Javier Ros
- Medical Oncology Department, Gastrointestinal and Endocrine Tumor Unit, Vall d´Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), Pg Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - Alvaro Arroyo
- Medical Oncology Department, Gastrointestinal and Endocrine Tumor Unit, Vall d´Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), Pg Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - Jaume Capdevila
- Medical Oncology Department, Gastrointestinal and Endocrine Tumor Unit, Vall d´Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), Pg Vall d´Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
94
|
Li Y, Su X, Feng C, Liu S, Guan H, Sun Y, He N, Ji M, Hou P. CYP2S1 is a synthetic lethal target in BRAF V600E-driven thyroid cancers. Signal Transduct Target Ther 2020; 5:191. [PMID: 32913191 PMCID: PMC7483764 DOI: 10.1038/s41392-020-00231-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 06/11/2020] [Accepted: 06/19/2020] [Indexed: 12/19/2022] Open
Abstract
BRAFV600E is the most common genetic alteration and has become a major therapeutic target in thyroid cancers; however, intrinsic feedback mechanism limited clinical use of BRAFV600E specific inhibitors. Synthetic lethal is a kind of interaction between two genes, where only simultaneously perturbing both of the genes can lead to lethality. Here, we identified CYP2S1 as a synthetic lethal partner of BRAFV600E in thyroid cancers. First, we found that CYP2S1 was highly expressed in papillary thyroid cancers (PTCs) compared to normal thyroid tissues, particularly in conventional PTCs (CPTCs) and tall-cell PTCs (TCPTCs), and its expression was positively associated with BRAFV600E mutation. CYP2S1 knockdown selectively inhibited cell proliferation, migration, invasion and tumorigenic potential in nude mice, and promoted cell apoptosis in BRAFV600E mutated thyroid cancer cells, but not in BRAF wild-type ones. Mechanistically, BRAFV600E-mediated MAPK/ERK cascade upregulated CYP2S1 expression by an AHR-dependent pathway, while CYP2S1 in turn enhanced transcriptional activity of AHR through its metabolites. This AHR/CYP2S1 feedback loop strongly amplified oncogenic role of BRAFV600E in thyroid cancer cells, thereby causing synthetic lethal interaction between CYP2S1 and BRAFV600E. Finally, we demonstrated CYP2S1 as a potential therapeutic target in both BRAFV600E-drived xenograft and transgenic mouse models by targetedly delivering CYP2S1-specific siRNA. Altogether, our data demonstrate CYP2S1 as a synthetic lethal partner of BRAFV600E in thyroid cancers, and indicate that targeting CYP2S1 will provide a new therapeutic strategy for BRAFV600E mutated thyroid cancers.
Collapse
Affiliation(s)
- Yiqi Li
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Xi Su
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Chao Feng
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Siyu Liu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Haixia Guan
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of China Medical University, Shenyang, 110001, P.R. China
| | - Yue Sun
- Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Nongyue He
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, P.R. China.
| | - Meiju Ji
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China.
| | - Peng Hou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China. .,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China.
| |
Collapse
|
95
|
Cabanillas ME, Dadu R, Iyer P, Wanland KB, Busaidy NL, Ying A, Gule-Monroe M, Wang JR, Zafereo M, Hofmann MC. Acquired Secondary RAS Mutation in BRAF V600E-Mutated Thyroid Cancer Patients Treated with BRAF Inhibitors. Thyroid 2020; 30:1288-1296. [PMID: 32216548 PMCID: PMC7869871 DOI: 10.1089/thy.2019.0514] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background: The BRAFV600E mutation is the most common driver mutation in papillary thyroid cancer (PTC) and anaplastic thyroid cancer (ATC). This mutation is considered actionable and, for BRAFV600E-mutated ATC, a BRAF inhibitor (dabrafenib) in combination with an MEK inhibitor (trametinib) is FDA approved. BRAF inhibitors have also shown efficacy in BRAFV600E-mutated PTC. However, as with all targeted therapies, resistance to these drugs eventually develops. It is essential that we understand the mechanisms of resistance to the BRAF inhibitors in thyroid cancer to develop future strategies to effectively treat these patients and improve survival. Patients: Herein, we describe four patients with thyroid cancer treated with selective BRAF inhibitors, who developed a RAS mutation in addition to the BRAFV600E mutation at progression. Results: Patients 1 and 3 acquired a KRASG12V mutation in the progressive tumor, patient 2 acquired a NRASQ61K mutation in a progressive lymph node, and patient 4 acquired NRASG13D mutation on liquid biopsy performed at the time of radiographic disease progression. Conclusion: Similar to the melanoma experience, the emergence of RAS mutations appears to act as a mechanism of resistance to BRAF inhibitors in thyroid cancers.
Collapse
Affiliation(s)
- Maria E. Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Address correspondence to: Maria E. Cabanillas, MD, Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Ramona Dadu
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Pryianka Iyer
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kacey B. Wanland
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Naifa L. Busaidy
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anita Ying
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Maria Gule-Monroe
- Department of Diagnostic Radiology, and The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer R. Wang
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mark Zafereo
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marie-Claude Hofmann
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
96
|
Coordinated dysregulation of cancer progression by the HER family and p21-activated kinases. Cancer Metastasis Rev 2020; 39:583-601. [PMID: 32820388 DOI: 10.1007/s10555-020-09922-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/20/2022]
Abstract
Most epithelial cancer types are polygenic in nature and are driven by coordinated dysregulation of multiple regulatory pathways, genes, and protein modifications. The process of coordinated regulation of cancer promoting pathways in response to extrinsic and intrinsic signals facilitates the dysregulation of several pathways with complementary functions, contributing to the hallmarks of cancer. Dysregulation and hyperactivation of cell surface human epidermal growth factor receptors (HERs) and cytoskeleton remodeling by p21-activated kinases (PAKs) are two prominent interconnected aspects of oncogenesis. We briefly discuss the discoveries and significant advances in the area of coordinated regulation of HERs and PAKs in the development and progression of breast and other epithelial cancers. We also discuss how initial studies involving heregulin signaling via HER3-HER2 axis and HER2-overexpressing breast cancer cells not only discovered a mechanistic role of PAK1 in breast cancer pathobiology but also acted as a bridge in generating a broader cancer research interest in other PAK family members and cancer types and catalyzed establishing the role of PAKs in human cancer, at-large. In addition, growth factor stimulation of the PAK pathway also helped to recognize new facets of PAKs, connecting the PAK pathway to oncogenesis, nuclear signaling, gene expression, mitotic progression, DNA damage response, among other phenotypic responses, and shaped the field of PAK cancer research. Finally, we recount some of the current limitations of HER- and PAK-directed therapeutics in counteracting acquired therapeutic resistance and discuss how cancer's as a polygenic disease may be best targeted with a polygenic approach.
Collapse
|
97
|
The Possible Role of Cancer Stem Cells in the Resistance to Kinase Inhibitors of Advanced Thyroid Cancer. Cancers (Basel) 2020; 12:cancers12082249. [PMID: 32796774 PMCID: PMC7465706 DOI: 10.3390/cancers12082249] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/03/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023] Open
Abstract
Target therapy with various kinase inhibitors (KIs) has been extended to patients with advanced thyroid cancer, but only a subset of these compounds has displayed efficacy in clinical use. However, after an initial response to KIs, dramatic disease progression occurs in most cases. With the discovery of cancer stem cells (CSCs), it is possible to postulate that thyroid cancer resistance to KI therapies, both intrinsic and acquired, may be sustained by this cell subtype. Indeed, CSCs have been considered as the main drivers of metastatic activity and therapeutic resistance, because of their ability to generate heterogeneous secondary cell populations and survive treatment by remaining in a quiescent state. Hence, despite the impressive progress in understanding of the molecular basis of thyroid tumorigenesis, drug resistance is still the major challenge in advanced thyroid cancer management. In this view, definition of the role of CSCs in thyroid cancer resistance may be crucial to identifying new therapeutic targets and preventing resistance to anti-cancer treatments and tumor relapse. The aim of this review is to elucidate the possible role of CSCs in the development of resistance of advanced thyroid cancer to current anti-cancer therapies and their potential implications in the management of these patients.
Collapse
|
98
|
San Román Gil M, Pozas J, Molina-Cerrillo J, Gómez J, Pian H, Pozas M, Carrato A, Grande E, Alonso-Gordoa T. Current and Future Role of Tyrosine Kinases Inhibition in Thyroid Cancer: From Biology to Therapy. Int J Mol Sci 2020; 21:E4951. [PMID: 32668761 PMCID: PMC7403957 DOI: 10.3390/ijms21144951] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022] Open
Abstract
Thyroid cancer represents a heterogenous disease whose incidence has increased in the last decades. Although three main different subtypes have been described, molecular characterization is progressively being included in the diagnostic and therapeutic algorithm of these patients. In fact, thyroid cancer is a landmark in the oncological approach to solid tumors as it harbors key genetic alterations driving tumor progression that have been demonstrated to be potential actionable targets. Within this promising and rapid changing scenario, current efforts are directed to improve tumor characterization for an accurate guidance in the therapeutic management. In this sense, it is strongly recommended to perform tissue genotyping to patients that are going to be considered for systemic therapy in order to select the adequate treatment, according to recent clinical trials data. Overall, the aim of this article is to provide a comprehensive review on the molecular biology of thyroid cancer focusing on the key role of tyrosine kinases. Additionally, from a clinical point of view, we provide a thorough perspective, current and future, in the treatment landscape of this tumor.
Collapse
MESH Headings
- Adenocarcinoma, Follicular/enzymology
- Adenocarcinoma, Follicular/genetics
- Adenocarcinoma, Follicular/therapy
- Adenoma, Oxyphilic/enzymology
- Adenoma, Oxyphilic/genetics
- Adenoma, Oxyphilic/therapy
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents, Immunological/therapeutic use
- Carcinoma, Medullary/enzymology
- Carcinoma, Medullary/genetics
- Carcinoma, Medullary/therapy
- Carcinoma, Papillary/enzymology
- Carcinoma, Papillary/genetics
- Carcinoma, Papillary/therapy
- Clinical Trials as Topic
- Combined Modality Therapy
- Disease Management
- Forecasting
- Genes, Neoplasm
- Humans
- Immune Checkpoint Inhibitors/therapeutic use
- Immunoconjugates/therapeutic use
- Immunotherapy
- Iodine Radioisotopes/therapeutic use
- Molecular Targeted Therapy
- Multicenter Studies as Topic
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Oncogene Proteins, Fusion/genetics
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Protein-Tyrosine Kinases/genetics
- Randomized Controlled Trials as Topic
- Thyroid Neoplasms/enzymology
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/therapy
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- María San Román Gil
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain; (M.S.R.G.); (J.P.); (M.P.); (A.C.); (T.A.-G.)
| | - Javier Pozas
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain; (M.S.R.G.); (J.P.); (M.P.); (A.C.); (T.A.-G.)
| | - Javier Molina-Cerrillo
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain; (M.S.R.G.); (J.P.); (M.P.); (A.C.); (T.A.-G.)
- The Ramon y Cajal Health Research Institute (IRYCIS), CIBERONC, 28034 Madrid, Spain
- Medicine School, Alcalá University, 28805 Madrid, Spain; (J.G.); (H.P.)
| | - Joaquín Gómez
- Medicine School, Alcalá University, 28805 Madrid, Spain; (J.G.); (H.P.)
- General Surgery Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Héctor Pian
- Medicine School, Alcalá University, 28805 Madrid, Spain; (J.G.); (H.P.)
- Pathology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Miguel Pozas
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain; (M.S.R.G.); (J.P.); (M.P.); (A.C.); (T.A.-G.)
| | - Alfredo Carrato
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain; (M.S.R.G.); (J.P.); (M.P.); (A.C.); (T.A.-G.)
- The Ramon y Cajal Health Research Institute (IRYCIS), CIBERONC, 28034 Madrid, Spain
- Medicine School, Alcalá University, 28805 Madrid, Spain; (J.G.); (H.P.)
| | - Enrique Grande
- Medical Oncology Department, MD Anderson Cancer Center, 28033 Madrid, Spain;
| | - Teresa Alonso-Gordoa
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain; (M.S.R.G.); (J.P.); (M.P.); (A.C.); (T.A.-G.)
- The Ramon y Cajal Health Research Institute (IRYCIS), CIBERONC, 28034 Madrid, Spain
- Medicine School, Alcalá University, 28805 Madrid, Spain; (J.G.); (H.P.)
| |
Collapse
|
99
|
Affiliation(s)
- Jenny Y Xue
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY.,Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY
| | - Piro Lito
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
100
|
Kumar R, George B, Campbell MR, Verma N, Paul AM, Melo-Alvim C, Ribeiro L, Pillai MR, da Costa LM, Moasser MM. HER family in cancer progression: From discovery to 2020 and beyond. Adv Cancer Res 2020; 147:109-160. [PMID: 32593399 DOI: 10.1016/bs.acr.2020.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The human epidermal growth factor receptor (HER) family of receptor tyrosine kinases (RTKs) are among the first layer of molecules that receive, interpret, and transduce signals leading to distinct cancer cell phenotypes. Since the discovery of the tooth-lid factor-later characterized as the epidermal growth factor (EGF)-and its high-affinity binding EGF receptor, HER kinases have emerged as one of the commonly upregulated or hyperactivated or mutated kinases in epithelial tumors, thus allowing HER1-3 family members to regulate several hallmarks of cancer development and progression. Each member of the HER family exhibits shared and unique structural features to engage multiple receptor activation modes, leading to a range of overlapping and distinct phenotypes. EGFR, the founding HER family member, provided the roadmap for the development of the cell surface RTK-directed targeted cancer therapy by serving as a prototype/precursor for the currently used HER-directed cancer drugs. We herein provide a brief account of the discoveries, defining moments, and historical context of the HER family and guidepost advances in basic, translational, and clinical research that solidified a prominent position of the HER family in cancer research and treatment. We also discuss the significance of HER3 pseudokinase in cancer biology; its unique structural features that drive transregulation among HER1-3, leading to a superior proximal signaling response; and potential role of HER3 as a shared effector of acquired therapeutic resistance against diverse oncology drugs. Finally, we also narrate some of the current drawbacks of HER-directed therapies and provide insights into postulated advances in HER biology with extensive implications of these therapies in cancer research and treatment.
Collapse
Affiliation(s)
- Rakesh Kumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India; Department of Medicine, Division of Hematology & Oncology, Rutgers New Jersey Medical School, Newark, NJ, United States; Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Bijesh George
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| | - Marcia R Campbell
- Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States
| | - Nandini Verma
- Advanced Centre for Treatment, Research and Education in Cancer, Mumbai, India
| | - Aswathy Mary Paul
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| | - Cecília Melo-Alvim
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Leonor Ribeiro
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - M Radhakrishna Pillai
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| | - Luis Marques da Costa
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Mark M Moasser
- Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States.
| |
Collapse
|