1
|
Caksa S, Baqai U, Aplin AE. The future of targeted kinase inhibitors in melanoma. Pharmacol Ther 2022; 239:108200. [PMID: 35513054 PMCID: PMC10187889 DOI: 10.1016/j.pharmthera.2022.108200] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 12/13/2022]
Abstract
Melanoma is a cancer of the pigment-producing cells of the body and its incidence is rising. Targeted inhibitors that act against kinases in the MAPK pathway are approved for BRAF-mutant metastatic cutaneous melanoma and increase patients' survival. Response to these therapies is limited by drug resistance and is less durable than with immune checkpoint inhibition. Conversely, rare melanoma subtypes have few therapeutic options for advanced disease and MAPK pathway targeting agents show minimal anti-tumor effects. Nevertheless, there is a future for targeted kinase inhibitors in melanoma: in new applications such as adjuvant or neoadjuvant therapy and in novel combinations with immunotherapies or other targeted therapies. Pre-clinical studies continue to identify tumor dependencies and their corresponding actionable drug targets, paving the way for rational targeted kinase inhibitor combinations as a personalized medicine approach for melanoma.
Collapse
Affiliation(s)
- Signe Caksa
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Usman Baqai
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
2
|
Gandullo-Sánchez L, Ocaña A, Pandiella A. HER3 in cancer: from the bench to the bedside. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:310. [PMID: 36271429 PMCID: PMC9585794 DOI: 10.1186/s13046-022-02515-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/07/2022] [Indexed: 11/15/2022]
Abstract
The HER3 protein, that belongs to the ErbB/HER receptor tyrosine kinase (RTK) family, is expressed in several types of tumors. That fact, together with the role of HER3 in promoting cell proliferation, implicate that targeting HER3 may have therapeutic relevance. Furthermore, expression and activation of HER3 has been linked to resistance to drugs that target other HER receptors such as agents that act on EGFR or HER2. In addition, HER3 has been associated to resistance to some chemotherapeutic drugs. Because of those circumstances, efforts to develop and test agents targeting HER3 have been carried out. Two types of agents targeting HER3 have been developed. The most abundant are antibodies or engineered antibody derivatives that specifically recognize the extracellular region of HER3. In addition, the use of aptamers specifically interacting with HER3, vaccines or HER3-targeting siRNAs have also been developed. Here we discuss the state of the art of the preclinical and clinical development of drugs aimed at targeting HER3 with therapeutic purposes.
Collapse
Affiliation(s)
- Lucía Gandullo-Sánchez
- grid.428472.f0000 0004 1794 2467Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - Alberto Ocaña
- grid.411068.a0000 0001 0671 5785Hospital Clínico San Carlos and CIBERONC, 28040 Madrid, Spain
| | - Atanasio Pandiella
- grid.428472.f0000 0004 1794 2467Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| |
Collapse
|
3
|
Sankarasubramanian S, Prabhakar P, Narasimhan MK. Genetic insights into cardiac tumors: a comprehensive review. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:164. [PMID: 35972566 DOI: 10.1007/s12032-022-01761-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022]
Abstract
Cardiac neoplasms are rare, however, also a curable form of the disease once detected early. In recent years the viscus tumors have gained their highlights, due to the advancement in techniques like echocardiography both 2D and 3D, MRI, etc. These cardiac tumors are divided based on their benign and malignant nature and also as well as primary and secondary cardiac tumors. Largely the primary cardiac tumors are often than secondary cardiac tumors. The secondary tumor happens anywhere in the body involving the heart. The most common malignant tumors are sarcoma, some are angiosarcomas, fibromas, rhabdosarcoma, and leiomyosarcoma. The primary sarcoma affects both men and women at an equal rate with non-specific symptoms. These conditions led to high demand in genomic testing that helps in spot the mutation that leads to the particular type of cardiac neoplasm and it additionally helps to screen the mutated sequence and stop it from being inherited. Recent studies on cardiac tumors have revealed many genes that are involved in tumorigenesis and technologies have enabled the right screening of the tumor location within the heart and their histopathological studies were also studied. This review principally focuses on the understanding of the various forms of cardiac tumors, genetic variants involved and their influence, genetic testing, and different diagnostic approaches in cardiac tumors.
Collapse
Affiliation(s)
- Sivaramasundaram Sankarasubramanian
- Department of Genetic Engineering, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Prathiksha Prabhakar
- Department of Genetic Engineering, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Manoj Kumar Narasimhan
- Department of Genetic Engineering, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chennai, Tamil Nadu, 603203, India.
| |
Collapse
|
4
|
Leu JI, Murphy ME, George DL. Targeting ErbB3 and Cellular NADPH/NADP + Abundance Sensitizes Cutaneous Melanomas to Ferroptosis Inducers. ACS Chem Biol 2022; 17:1038-1044. [PMID: 35420772 PMCID: PMC9208338 DOI: 10.1021/acschembio.2c00113] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Melanoma is a serious health challenge. Ferroptosis is a regulated form of oxidative cell death that shows varied efficacy in melanoma. We aimed to better understand the molecular basis for this differential ferroptosis sensitivity. We find that elevated expression of ErbB3 (V-Erb-B2 Avian Erythroblastic Leukemia Viral Oncogene Homologue 3) associates with ferroptosis resistance and that ErbB3 knockdown sensitizes to ferroptosis inducers. ErbB3 depletion also promotes a marked reduction in the cellular ratio of GSH/GSSG (reduced/oxidized glutathione) and that of NADPH/NADP+ (reduced/oxidized nicotinamide adenine dinucleotide phosphate), together with an increase in the abundance of the lipid peroxidation product malondialdehyde (MDA). We identify several small molecule inhibitors targeting ErbB3 signaling pathways that also reduce the NADPH/NADP+ and GSH/GSSG ratios, concomitantly sensitizing the melanomas to ferroptosis activators. These findings point to a previously unrecognized role of ErbB3 in ferroptosis sensitivity and provide new insight into pathways that regulate this cell death process.
Collapse
Affiliation(s)
- Julia I. Leu
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Maureen E Murphy
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Donna L George
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
5
|
Beauchamp RL, Erdin S, Witt L, Jordan JT, Plotkin SR, Gusella JF, Ramesh V. mTOR kinase inhibition disrupts neuregulin 1-ERBB3 autocrine signaling and sensitizes NF2-deficient meningioma cellular models to IGF1R inhibition. J Biol Chem 2021; 296:100157. [PMID: 33273014 PMCID: PMC7949095 DOI: 10.1074/jbc.ra120.014960] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/23/2020] [Accepted: 12/03/2020] [Indexed: 12/16/2022] Open
Abstract
Meningiomas (MNs), arising from the arachnoid/meningeal layer, are nonresponsive to chemotherapies, with ∼50% showing loss of the Neurofibromatosis 2 (NF2) tumor suppressor gene. Previously, we established NF2 loss activates mechanistic target of rapamycin complex 1 (mTORC1) and mechanistic target of rapamycin complex 2 (mTORC2) signaling, leading to clinical trials for NF2 and MN. Recently our omics studies identified activated ephrin (EPH) receptor and Src family kinases upon NF2 loss. Here, we report increased expression of several ligands in NF2-null human arachnoidal cells (ACs) and the MN cell line Ben-Men-1, particularly neuregulin-1/heregulin (NRG1), and confirm increased NRG1 secretion and activation of V-ERB-B avian erythroblastic leukemia viral oncogene homolog 3 (ERBB3) receptor kinase. Conditioned-medium from NF2-null ACs or exogenous NRG1 stimulated ERBB3, EPHA2, and mTORC1/2 signaling, suggesting pathway crosstalk. NF2-null cells treated with an ERBB3-neutralizing antibody partially downregulated mTOR pathway activation but showed no effect on viability. mTORC1/2 inhibitor treatment decreased NRG1 expression and downregulated ERBB3 while re-activating pAkt T308, suggesting a mechanism independent of NRG1-ERBB3 but likely involving activation of another upstream receptor kinase. Transcriptomics after mTORC1/2 inhibition confirmed decreased ERBB3/ERBB4 while revealing increased expression of insulin-like growth factor receptor 1 (IGF1R). Drug treatment co-targeting mTORC1/2 and IGF1R/insulin receptor attenuated pAkt T308 and showed synergistic effects on viability. Our findings indicate potential autocrine signaling where NF2 loss leads to secretion/activation of NRG1-ERBB3 signaling. mTORC1/2 inhibition downregulates NRG1-ERBB3, while upregulating pAkt T308 through an adaptive response involving IGF1R/insulin receptor and co-targeting these pathways may prove effective for treatment of NF2-deficient MN.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/pharmacology
- Autocrine Communication/genetics
- Benzamides/pharmacology
- Benzoxazoles/pharmacology
- Cell Line, Tumor
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Gene Expression Regulation
- Humans
- Lapatinib/pharmacology
- Meningeal Neoplasms/genetics
- Meningeal Neoplasms/metabolism
- Meningeal Neoplasms/pathology
- Meningioma/genetics
- Meningioma/metabolism
- Meningioma/pathology
- Morpholines/pharmacology
- Neuregulin-1/antagonists & inhibitors
- Neuregulin-1/genetics
- Neuregulin-1/metabolism
- Neurofibromin 2/deficiency
- Neurofibromin 2/genetics
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Pyrazoles/pharmacology
- Pyrimidines/pharmacology
- Receptor, EphA2/genetics
- Receptor, EphA2/metabolism
- Receptor, ErbB-3/antagonists & inhibitors
- Receptor, ErbB-3/genetics
- Receptor, ErbB-3/metabolism
- Receptor, IGF Type 1/antagonists & inhibitors
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Signal Transduction
- Sirolimus/pharmacology
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- Transcriptome
- Triazines/pharmacology
Collapse
Affiliation(s)
- Roberta L Beauchamp
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Serkan Erdin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Luke Witt
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Justin T Jordan
- Department of Neurology and Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Scott R Plotkin
- Department of Neurology and Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - James F Gusella
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Vijaya Ramesh
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
6
|
Louveau B, Jouenne F, Têtu P, Sadoux A, Gruber A, Lopes E, Delyon J, Serror K, Marco O, Da Meda L, Ndiaye A, Lermine A, Dumaz N, Battistella M, Baroudjian B, Lebbe C, Mourah S. A Melanoma-Tailored Next-Generation Sequencing Panel Coupled with a Comprehensive Analysis to Improve Routine Melanoma Genotyping. Target Oncol 2020; 15:759-771. [PMID: 33151472 DOI: 10.1007/s11523-020-00764-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Tumor molecular deciphering is crucial in clinical management. Pan-cancer next-generation sequencing panels have moved towards exhaustive molecular characterization. However, because of treatment resistance and the growing emergence of pharmacological targets, tumor-specific customized panels are needed to guide therapeutic strategies. OBJECTIVE The objective of this study was to present such a customized next-generation sequencing panel in melanoma. METHODS Melanoma patients with somatic molecular profiling performed as part of routine care were included. High-throughput sequencing was performed with a melanoma tailored next-generation sequencing panel of 64 genes involved in molecular classification, prognosis, theranostic, and therapeutic resistance. Single nucleotide variants and copy number variations were screened, and a comprehensive molecular analysis identified clinically relevant alterations. RESULTS Four hundred and twenty-one melanoma cases were analyzed (before any treatment initiation for 94.8% of patients). After bioinformatic prioritization, we uncovered 561 single nucleotide variants, 164 copy number variations, and four splice-site mutations. At least one alteration was detected in 368 (87.4%) lesions, with BRAF, NRAS, CDKN2A, CCND1, and MET as the most frequently altered genes. Among patients with BRAFV600 mutated melanoma, 44.5% (77 of 173) harbored at least one concurrent alteration driving potential resistance to mitogen-activated protein kinase inhibitors. In patients with RAS hotspot mutated lesions and in patients with neither BRAFV600 nor RAS hotspot mutations, alterations constituting potential pharmacological targets were found in 56.9% (66 of 116) and 47.7% (63 of 132) of cases, respectively. CONCLUSIONS Our tailored next-generation sequencing assay coupled with a comprehensive analysis may improve therapeutic management in a significant number of patients with melanoma. Updating such a panel and implementing multi-omic approaches will further enhance patients' clinical management.
Collapse
Affiliation(s)
- Baptiste Louveau
- Department of Pharmacology and Solid Tumor Genomics, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, 1 Avenue Claude Vellefaux, 75475, Paris Cedex 10, France.,Université de Paris, Paris, France.,INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Paris, France
| | - Fanélie Jouenne
- Department of Pharmacology and Solid Tumor Genomics, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, 1 Avenue Claude Vellefaux, 75475, Paris Cedex 10, France.,Université de Paris, Paris, France.,INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Paris, France
| | - Pauline Têtu
- Department of Dermatology, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Aurélie Sadoux
- Department of Pharmacology and Solid Tumor Genomics, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, 1 Avenue Claude Vellefaux, 75475, Paris Cedex 10, France
| | - Aurélia Gruber
- Department of Pharmacology and Solid Tumor Genomics, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, 1 Avenue Claude Vellefaux, 75475, Paris Cedex 10, France
| | - Eddie Lopes
- Department of Pharmacology and Solid Tumor Genomics, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, 1 Avenue Claude Vellefaux, 75475, Paris Cedex 10, France
| | - Julie Delyon
- Université de Paris, Paris, France.,INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Paris, France.,Department of Dermatology, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Kevin Serror
- Department of Plastic, Reconstructive and Esthetic Surgery, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Oren Marco
- Department of Plastic, Reconstructive and Esthetic Surgery, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Laetitia Da Meda
- Department of Dermatology, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Aminata Ndiaye
- MOABI-APHP Bioinformatics Platform-WIND-DSI, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alban Lermine
- MOABI-APHP Bioinformatics Platform-WIND-DSI, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nicolas Dumaz
- INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Paris, France
| | - Maxime Battistella
- Université de Paris, Paris, France.,INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Paris, France.,Department of Pathology, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Barouyr Baroudjian
- Department of Dermatology, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Céleste Lebbe
- Université de Paris, Paris, France.,INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Paris, France.,Department of Dermatology, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Samia Mourah
- Department of Pharmacology and Solid Tumor Genomics, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, 1 Avenue Claude Vellefaux, 75475, Paris Cedex 10, France. .,Université de Paris, Paris, France. .,INSERM UMR-S 976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Paris, France.
| |
Collapse
|
7
|
Targeted therapies in melanoma beyond BRAF: targeting NRAS-mutated and KIT-mutated melanoma. Curr Opin Oncol 2020; 32:79-84. [PMID: 31833955 DOI: 10.1097/cco.0000000000000606] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW Melanoma treatment have been revolutionized since 2010 by the development of immune checkpoint inhibitors, and, for BRAF-mutated melanoma, targeted therapies based on BRAF and MEK inhibitors, which is a model of effective targeted therapy in cancer. However, patients with BRAF wild type cannot benefit for such treatments. In this review, we will focus on the current clinical development of targeted therapies beyond BRAF, in NRAS-mutated and KIT-altered melanoma. RECENT FINDINGS In NRAS-mutated melanoma, targeted therapies based on MEK inhibition are being developed as monotherapy or in combination with MAPK, PI3K or CDK4/6 inhibitor. Targeted therapies of KIT-altered melanoma patients is based in KIT inhibitor (mostly imatinib, nilotinib), although for both melanoma subtypes, results are for now disappointing as compared with BRAF and MEK inhibitors in BRAF-mutated melanoma. SUMMARY Combined therapeutic targeted strategies are awaited in NRAS-mutated and KIT-altered melanoma and could provide additional benefit.
Collapse
|
8
|
Xu Y, Mu Y, Wang L, Zhang X. Detailed Analysis of Molecular Mechanisms in Primary and Metastatic Melanoma. J Comput Biol 2020; 27:9-19. [PMID: 31424282 DOI: 10.1089/cmb.2019.0197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yangchun Xu
- Department of Dermatology and Second Hospital of Jilin University, Changchun, China
| | - Yan Mu
- Department of Dermatology and Second Hospital of Jilin University, Changchun, China
| | - Ling Wang
- Department of Gynecology, Second Hospital of Jilin University, Changchun, China
| | - Xuan Zhang
- Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
9
|
ErbB3 Phosphorylation as Central Event in Adaptive Resistance to Targeted Therapy in Metastatic Melanoma: Early Detection in CTCs during Therapy and Insights into Regulation by Autocrine Neuregulin. Cancers (Basel) 2019; 11:cancers11101425. [PMID: 31557826 PMCID: PMC6826737 DOI: 10.3390/cancers11101425] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/20/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022] Open
Abstract
In recent years the introduction of target therapies with BRAF and MEK inhibitors (MAPKi) and of immunotherapy with anti-CTLA-4 and anti-PD-1 monoclonal antibodies have dramatically improved survival of metastatic melanoma patients. Despite these changes drug resistance remains a major hurdle. Several mechanisms are at the basis of drug resistance. Particular attention has been devoted over the last years to unravel mechanisms at the basis of adaptive/non genetic resistance occurring in BRAF mutated melanomas upon treatment with to MAPKi. In this paper we focus on the involvement of activation of ErbB3 receptor following early exposure of melanoma cells to BRAF or MEK inhibitors, and the following induction of PI3K/AKT pathway. Although different mechanisms have been invoked in the past at the basis of this activation we show here with a combination of approaches that autocrine production of neuregulin by melanoma cells is a major factor responsible for ErbB3 phosphorylation and downstream AKT activation. Interestingly the kinetic of neuregulin production and of the ensuing ErbB3 phosphorylation is different in different melanoma cell lines which underscores the high degree of tumor heterogeneity. Moreover, heterogeneity is further highlighted by the evidence that in different cell lines neuregulin upregulation can occur at the transcriptional or at the post-transcritpional level. Finally we complement our study by showing with a liquid biopsy assay that circulating tumor cells (CTCs) from melanoma patients undergo upregulation of ErbB3 phosphorylation in vivo shortly after initiation of therapy.
Collapse
|
10
|
Appenzeller S, Gesierich A, Thiem A, Hufnagel A, Jessen C, Kneitz H, Regensburger M, Schmidt C, Zirkenbach V, Bischler T, Schilling B, Siedel C, Goebeler ME, Houben R, Schrama D, Gehrig A, Rost S, Maurus K, Bargou R, Rosenwald A, Schartl M, Goebeler M, Meierjohann S. The identification of patient-specific mutations reveals dual pathway activation in most patients with melanoma and activated receptor tyrosine kinases in BRAF/NRAS wild-type melanomas. Cancer 2018; 125:586-600. [PMID: 30561760 DOI: 10.1002/cncr.31843] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/23/2018] [Accepted: 10/02/2018] [Indexed: 11/07/2022]
Abstract
BACKGROUND Increasing knowledge of cancer genomes has triggered the development of specific targeted inhibitors, thus providing a valuable therapeutic pool. METHODS In this report, the authors analyze the presence of targetable alterations in 136 tumor samples from 92 patients with melanoma using a comprehensive approach based on targeted DNA sequencing and supported by RNA and protein analysis. Three topics of high clinical relevance are addressed: the identification of rare, activating alterations; the detection of patient-specific, co-occurring single nucleotide variants (SNVs) and copy number variations (CNVs) in parallel pathways; and the presence of cancer-relevant germline mutations. RESULTS The analysis of patient-matched blood and tumor samples was done with a custom-designed gene panel that was enriched for genes from clinically targetable pathways. To detect alterations with high therapeutic relevance for patients with unknown driver mutations, genes that are untypical for melanoma also were included. Among all patients, CNVs were identified in one-third of samples and contained amplifications of druggable kinases, such as CDK4, ERBB2, and KIT. Considering SNVs and CNVs, 60% of patients with metastases exhibited co-occurring activations of at least 2 pathways, thus providing a rationale for individualized combination therapies. Unexpectedly, 9% of patients carry potentially protumorigenic germline mutations frequently affecting receptor tyrosine kinases. Remarkably two-thirds of BRAF/NRAS wild-type melanomas harbor activating mutations or CNVs in receptor tyrosine kinases. CONCLUSIONS The results indicate that the integrated analysis of SNVs, CNVs, and germline mutations reveals new druggable targets for combination tumor therapy.
Collapse
Affiliation(s)
- Silke Appenzeller
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Anja Gesierich
- Department of Dermatology, Venereology, and Allergology and Skin Cancer Center, University Hospital Würzburg, Würzburg, Germany
| | - Alexander Thiem
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany.,Department of Dermatology, Venereology, and Allergology and Skin Cancer Center, University Hospital Würzburg, Würzburg, Germany
| | - Anita Hufnagel
- Department of Physiological Chemistry, University of Würzburg, Würzburg, Germany
| | - Christina Jessen
- Department of Physiological Chemistry, University of Würzburg, Würzburg, Germany
| | - Hermann Kneitz
- Department of Dermatology, Venereology, and Allergology and Skin Cancer Center, University Hospital Würzburg, Würzburg, Germany
| | - Martina Regensburger
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany.,Department of Physiological Chemistry, University of Würzburg, Würzburg, Germany
| | - Cornelia Schmidt
- Department of Physiological Chemistry, University of Würzburg, Würzburg, Germany
| | - Vanessa Zirkenbach
- Department of Physiological Chemistry, University of Würzburg, Würzburg, Germany
| | - Thorsten Bischler
- Core Unit Systems Medicine, University of Würzburg, Würzburg, Germany
| | - Bastian Schilling
- Department of Dermatology, Venereology, and Allergology and Skin Cancer Center, University Hospital Würzburg, Würzburg, Germany
| | - Claudia Siedel
- Department of Dermatology, Venereology, and Allergology and Skin Cancer Center, University Hospital Würzburg, Würzburg, Germany
| | | | - Roland Houben
- Department of Dermatology, Venereology, and Allergology and Skin Cancer Center, University Hospital Würzburg, Würzburg, Germany
| | - David Schrama
- Department of Dermatology, Venereology, and Allergology and Skin Cancer Center, University Hospital Würzburg, Würzburg, Germany
| | - Andrea Gehrig
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Simone Rost
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Katja Maurus
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Ralf Bargou
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | | | - Manfred Schartl
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany.,Department of Physiological Chemistry, University of Würzburg, Würzburg, Germany.,Texas A&M Institute for Advanced Studies and Department of Biology, Texas A&M University, College Station, Texas
| | - Matthias Goebeler
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany.,Department of Dermatology, Venereology, and Allergology and Skin Cancer Center, University Hospital Würzburg, Würzburg, Germany
| | - Svenja Meierjohann
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany.,Department of Physiological Chemistry, University of Würzburg, Würzburg, Germany
| |
Collapse
|
11
|
Capparelli C, Purwin TJ, Heilman SA, Chervoneva I, McCue PA, Berger AC, Davies MA, Gershenwald JE, Krepler C, Aplin AE. ErbB3 Targeting Enhances the Effects of MEK Inhibitor in Wild-Type BRAF/NRAS Melanoma. Cancer Res 2018; 78:5680-5693. [PMID: 30115691 DOI: 10.1158/0008-5472.can-18-1001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/23/2018] [Accepted: 08/02/2018] [Indexed: 12/12/2022]
Abstract
MEK-ERK1/2 signaling is elevated in melanomas that are wild-type for both BRAF and NRAS (WT/WT), but patients are insensitive to MEK inhibitors. Stromal-derived growth factors may mediate resistance to targeted inhibitors, and optimizing the use of targeted inhibitors for patients with WT/WT melanoma is a clinical unmet need. Here, we studied adaptive responses to MEK inhibition in WT/WT cutaneous melanoma. The Cancer Genome Atlas data set and tumor microarray studies of WT/WT melanomas showed that high levels of neuregulin-1 (NRG1) were associated with stromal content and ErbB3 signaling. Of growth factors implicated in resistance to targeted inhibitors, NRG1 was effective at mediating resistance to MEK inhibitors in patient-derived WT/WT melanoma cells. Furthermore, ErbB3/ErbB2 signaling was adaptively upregulated following MEK inhibition. Patient-derived cancer-associated fibroblast studies demonstrated that stromal-derived NRG1 activated ErbB3/ErbB2 signaling and enhanced resistance to a MEK inhibitor. ErbB3- and ErbB2-neutralizing antibodies blocked the protective effects of NRG1 in vitro and cooperated with the MEK inhibitor to delay tumor growth in both cell line and patient-derived xenograft models. These results highlight tumor microenvironment regulation of targeted inhibitor resistance in WT/WT melanoma and provide a rationale for combining MEK inhibitors with anti-ErbB3/ErbB2 antibodies in patients with WT/WT cutaneous melanoma, for whom there are no effective targeted therapy options.Significance: This work suggests a mechanism by which NRG1 regulates the sensitivity of WT NRAS/BRAF melanomas to MEK inhibitors and provides a rationale for combining MEK inhibitors with anti-ErbB2/ErbB3 antibodies in these tumors. Cancer Res; 78(19); 5680-93. ©2018 AACR.
Collapse
Affiliation(s)
- Claudia Capparelli
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Timothy J Purwin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Shea A Heilman
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Inna Chervoneva
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Peter A McCue
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Adam C Berger
- Department of Surgery, Division of General Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Clemens Krepler
- The Wistar Institute, Molecular and Cellular Oncogenesis Program, Melanoma Research Center, Philadelphia, Pennsylvania
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. .,Sidney Kimmel Cancer Center at Jefferson, Philadelphia, Pennsylvania
| |
Collapse
|
12
|
Khalil HS, Langdon SP, Goltsov A, Soininen T, Harrison DJ, Bown J, Deeni YY. A novel mechanism of action of HER2 targeted immunotherapy is explained by inhibition of NRF2 function in ovarian cancer cells. Oncotarget 2018; 7:75874-75901. [PMID: 27713148 PMCID: PMC5342785 DOI: 10.18632/oncotarget.12425] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/21/2016] [Indexed: 12/16/2022] Open
Abstract
Nuclear erythroid related factor-2 (NRF2) is known to promote cancer therapeutic detoxification and crosstalk with growth promoting pathways. HER2 receptor tyrosine kinase is frequently overexpressed in cancers leading to uncontrolled receptor activation and signaling. A combination of HER2 targeting monoclonal antibodies shows greater anticancer efficacy than the single targeting antibodies, however, its mechanism of action is largely unclear. Here we report novel actions of anti-HER2 drugs, Trastuzumab and Pertuzumab, involving NRF2. HER2 targeting by antibodies inhibited growth in association with persistent generation of reactive oxygen species (ROS), glutathione (GSH) depletion, reduction in NRF2 levels and inhibition of NRF2 function in ovarian cancer cell lines. The combination of antibodies produced more potent effects than single antibody alone; downregulated NRF2 substrates by repressing the Antioxidant Response (AR) pathway with concomitant transcriptional inhibition of NRF2. We showed the antibody combination produced increased methylation at the NRF2 promoter consistent with repression of NRF2 antioxidant function, as HDAC and methylation inhibitors reversed such produced transcriptional effects. These findings demonstrate a novel mechanism and role for NRF2 in mediating the response of cancer cells to the combination of Trastuzumab and Pertuzumab and reinforce the importance of NRF2 in drug resistance and as a key anticancer target.
Collapse
Affiliation(s)
- Hilal S Khalil
- Division of Science, School of Science, Engineering and Technology, Abertay University, Dundee, DD1 1HG, United Kingdom
| | - Simon P Langdon
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, United Kingdom
| | - Alexey Goltsov
- Division of Science, School of Science, Engineering and Technology, Abertay University, Dundee, DD1 1HG, United Kingdom
| | - Tero Soininen
- Division of Science, School of Science, Engineering and Technology, Abertay University, Dundee, DD1 1HG, United Kingdom
| | - David J Harrison
- School of Medicine, University of St Andrews, St Andrews, KY16 9TF, United Kingdom
| | - James Bown
- Division of Computing and Mathematics, School of Arts, Media, and Computer Games, Abertay University, Dundee, DD1 1HG, United Kingdom
| | - Yusuf Y Deeni
- Division of Science, School of Science, Engineering and Technology, Abertay University, Dundee, DD1 1HG, United Kingdom
| |
Collapse
|
13
|
Mota JM, Collier KA, Barros Costa RL, Taxter T, Kalyan A, Leite CA, Chae YK, Giles FJ, Carneiro BA. A comprehensive review of heregulins, HER3, and HER4 as potential therapeutic targets in cancer. Oncotarget 2017; 8:89284-89306. [PMID: 29179520 PMCID: PMC5687690 DOI: 10.18632/oncotarget.18467] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 04/17/2017] [Indexed: 12/30/2022] Open
Abstract
Heregulins (HRGs) bind to the receptors HER3 or HER4, induce receptor dimerization, and trigger downstream signaling that leads to tumor progression and resistance to targeted therapies. Increased expression of HRGs has been associated with worse clinical prognosis; therefore, attempts to block HRG-dependent tumor growth have been pursued. This manuscript summarizes the function and signaling of HRGs and review the preclinical evidence of its involvement in carcinogenesis, prognosis, and treatment resistance in several malignancies such as colorectal cancer, non-small cell lung cancer, ovarian cancer, and breast cancer. Agents in preclinical development and clinical trials of novel therapeutics targeting HRG-dependent signaling are also discussed, including anti-HER3 and -HER4 antibodies, anti-metalloproteinase agents, and HRG fusion proteins. Although several trials have indicated an acceptable safety profile, translating preclinical findings into clinical practice remains a challenge in this field, possibly due to the complexity of downstream signaling and patterns of HRG, HER3 and HER4 expression in different cancer subtypes. Improving patient selection through biomarkers and understanding the resistance mechanisms may translate into significant clinical benefits in the near future.
Collapse
Affiliation(s)
- Jose Mauricio Mota
- Instituto do Câncer do Estado de São Paulo, Division of Oncology, Universidade de São Paulo, São Paulo, Brazil
| | - Katharine Ann Collier
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ricardo Lima Barros Costa
- Developmental Therapeutics Program, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Timothy Taxter
- Developmental Therapeutics Program, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Aparna Kalyan
- Developmental Therapeutics Program, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Caio A. Leite
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Young Kwang Chae
- Developmental Therapeutics Program, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Francis J. Giles
- Developmental Therapeutics Program, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Benedito A. Carneiro
- Developmental Therapeutics Program, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
14
|
Boespflug A, Caramel J, Dalle S, Thomas L. Treatment of NRAS-mutated advanced or metastatic melanoma: rationale, current trials and evidence to date. Ther Adv Med Oncol 2017; 9:481-492. [PMID: 28717400 PMCID: PMC5502949 DOI: 10.1177/1758834017708160] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 04/13/2017] [Indexed: 12/19/2022] Open
Abstract
The disease course of BRAF (v-raf murine sarcoma viral oncogene homolog B1)-mutant melanoma has been drastically improved by the arrival of targeted therapies. NRAS (neuroblastoma RAS viral oncogene homolog)-mutated melanoma represents 15–25% of all metastatic melanoma patients. It currently does not have an approved targeted therapy. Metastatic patients receive immune-based therapies as first-line treatments, then cytotoxic chemotherapy like carboplatin/paclitaxel (C/P), dacarbazine (DTIC) or temozolomide (TMZ) as a second-line treatment. We will review current preclinical and clinical developments in NRAS-mutated melanoma, and analyze ongoing clinical trials that are evaluating the benefit of different targeted and immune-based therapies, either tested as single agents or in combination, in NRAS-mutant melanoma.
Collapse
Affiliation(s)
| | - Julie Caramel
- Cancer Research Center of Lyon, Claude Bernard Lyon-1 University, INSERM1052, CNRS 5286, Lyon, France
| | | | - Luc Thomas
- Service de Dermatologie, CH Lyon Sud, 165 Chemin du Grand Revoyet, 69495 Pierre Bénite, Cedex, France
| |
Collapse
|
15
|
Kerketta R, Halász ÁM, Steinkamp MP, Wilson BS, Edwards JS. Effect of Spatial Inhomogeneities on the Membrane Surface on Receptor Dimerization and Signal Initiation. Front Cell Dev Biol 2016; 4:81. [PMID: 27570763 PMCID: PMC4981600 DOI: 10.3389/fcell.2016.00081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 07/25/2016] [Indexed: 11/23/2022] Open
Abstract
Important signal transduction pathways originate on the plasma membrane, where microdomains may transiently entrap diffusing receptors. This results in a non-random distribution of receptors even in the resting state, which can be visualized as “clusters” by high resolution imaging methods. Here, we explore how spatial in-homogeneities in the plasma membrane might influence the dimerization and phosphorylation status of ErbB2 and ErbB3, two receptor tyrosine kinases that preferentially heterodimerize and are often co-expressed in cancer. This theoretical study is based upon spatial stochastic simulations of the two-dimensional membrane landscape, where variables include differential distributions and overlap of transient confinement zones (“domains”) for the two receptor species. The in silico model is parameterized and validated using data from single particle tracking experiments. We report key differences in signaling output based on the degree of overlap between domains and the relative retention of receptors in such domains, expressed as escape probability. Results predict that a high overlap of domains, which favors transient co-confinement of both receptor species, will enhance the rate of hetero-interactions. Where domains do not overlap, simulations confirm expectations that homo-interactions are favored. Since ErbB3 is uniquely dependent on ErbB2 interactions for activation of its catalytic activity, variations in domain overlap or escape probability markedly alter the predicted patterns and time course of ErbB3 and ErbB2 phosphorylation. Taken together, these results implicate membrane domain organization as an important modulator of signal initiation, motivating the design of novel experimental approaches to measure these important parameters across a wider range of receptor systems.
Collapse
Affiliation(s)
- Romica Kerketta
- Department of Pathology, University of New Mexico Health Sciences Center Albuquerque, NM, USA
| | - Ádám M Halász
- Department of Mathematics and Mary Babb Randolph Cancer Center, West Virginia University Morgantown, WV, USA
| | - Mara P Steinkamp
- Department of Pathology, University of New Mexico Health Sciences CenterAlbuquerque, NM, USA; Cancer Center, University of New Mexico Health Sciences CenterAlbuquerque, NM, USA
| | - Bridget S Wilson
- Department of Pathology, University of New Mexico Health Sciences CenterAlbuquerque, NM, USA; Cancer Center, University of New Mexico Health Sciences CenterAlbuquerque, NM, USA
| | - Jeremy S Edwards
- Cancer Center, University of New Mexico Health Sciences CenterAlbuquerque, NM, USA; Department of Chemical and Biological Engineering, University of New MexicoAlbuquerque, NM, USA; Department of Chemistry and Chemical Biology, University of New MexicoAlbuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New MexicoAlbuquerque, NM, USA
| |
Collapse
|
16
|
Xiao Z, Carrasco RA, Schifferli K, Kinneer K, Tammali R, Chen H, Rothstein R, Wetzel L, Yang C, Chowdhury P, Tsui P, Steiner P, Jallal B, Herbst R, Hollingsworth RE, Tice DA. A Potent HER3 Monoclonal Antibody That Blocks Both Ligand-Dependent and -Independent Activities: Differential Impacts of PTEN Status on Tumor Response. Mol Cancer Ther 2016; 15:689-701. [PMID: 26880266 DOI: 10.1158/1535-7163.mct-15-0555] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 02/09/2016] [Indexed: 11/16/2022]
Abstract
HER3/ERBB3 is a kinase-deficient member of the EGFR family receptor tyrosine kinases (RTK) that is broadly expressed and activated in human cancers. HER3 is a compelling cancer target due to its important role in activation of the oncogenic PI3K/AKT pathway. It has also been demonstrated to confer tumor resistance to a variety of cancer therapies, especially targeted drugs against EGFR and HER2. HER3 can be activated by its ligand (heregulin/HRG), which induces HER3 heterodimerization with EGFR, HER2, or other RTKs. Alternatively, HER3 can be activated in a ligand-independent manner through heterodimerization with HER2 in HER2-amplified cells. We developed a fully human mAb against HER3 (KTN3379) that efficiently suppressed HER3 activity in both ligand-dependent and independent settings. Correspondingly, KTN3379 inhibited tumor growth in divergent tumor models driven by either ligand-dependent or independent mechanisms in vitro and in vivo Most intriguingly, while investigating the mechanistic underpinnings of tumor response to KTN3379, we discovered an interesting dichotomy in that PTEN loss, a frequently occurring oncogenic lesion in a broad range of cancer types, substantially blunted the tumor response in HER2-amplified cancer, but not in the ligand-driven cancer. To our knowledge, this represents the first study ascertaining the impact of PTEN loss on the antitumor efficacy of a HER3 mAb. KTN3379 is currently undergoing a phase Ib clinical trial in patients with advanced solid tumors. Our current study may help us optimize patient selection schemes for KTN3379 to maximize its clinical benefits. Mol Cancer Ther; 15(4); 689-701. ©2016 AACR.
Collapse
Affiliation(s)
- Zhan Xiao
- Oncology Research, MedImmune, Inc., One MedImmune Way, Gaithersburg, MD.
| | - Rosa A Carrasco
- Oncology Research, MedImmune, Inc., One MedImmune Way, Gaithersburg, MD
| | - Kevin Schifferli
- Oncology Research, MedImmune, Inc., One MedImmune Way, Gaithersburg, MD
| | - Krista Kinneer
- Oncology Research, MedImmune, Inc., One MedImmune Way, Gaithersburg, MD
| | - Ravinder Tammali
- Oncology Research, MedImmune, Inc., One MedImmune Way, Gaithersburg, MD
| | - Hong Chen
- Oncology Research, MedImmune, Inc., One MedImmune Way, Gaithersburg, MD
| | - Ray Rothstein
- Oncology Research, MedImmune, Inc., One MedImmune Way, Gaithersburg, MD
| | - Leslie Wetzel
- Oncology Research, MedImmune, Inc., One MedImmune Way, Gaithersburg, MD
| | - Chunning Yang
- Antibody Development and Protein Engineering, MedImmune, Inc., Gaithersburg, Maryland
| | - Partha Chowdhury
- Antibody Development and Protein Engineering, MedImmune, Inc., Gaithersburg, Maryland
| | - Ping Tsui
- Antibody Development and Protein Engineering, MedImmune, Inc., Gaithersburg, Maryland
| | - Philipp Steiner
- Oncology Research, MedImmune, Inc., One MedImmune Way, Gaithersburg, MD
| | - Bahija Jallal
- Oncology Research, MedImmune, Inc., One MedImmune Way, Gaithersburg, MD
| | - Ronald Herbst
- Oncology Research, MedImmune, Inc., One MedImmune Way, Gaithersburg, MD
| | | | - David A Tice
- Oncology Research, MedImmune, Inc., One MedImmune Way, Gaithersburg, MD
| |
Collapse
|
17
|
Capparelli C, Rosenbaum S, Berger AC, Aplin AE. Fibroblast-derived neuregulin 1 promotes compensatory ErbB3 receptor signaling in mutant BRAF melanoma. J Biol Chem 2015; 290:24267-77. [PMID: 26269601 DOI: 10.1074/jbc.m115.657270] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Indexed: 01/07/2023] Open
Abstract
Rapidly accelerated fibrosarcoma (RAF) inhibitors are first-line treatments for patients harboring V600E/K mutant BRAF melanoma. Although RAF inhibitors produce high response rates, the degree of tumor regression is heterogeneous. Compensatory/adaptive responses to targeted inhibitors are frequently initiated by the activation of growth factor receptor tyrosine kinases, including ErbB3, and factors from the tumor microenvironment may play an important role. We have shown previously that mutant v-raf murine sarcoma viral oncogene homolog B1 (BRAF) melanoma cells have enhanced activation of ErbB3 following RAF inhibition. However, the source of neuregulin 1 (NRG1), the ligand for ErbB3, is unknown. In this study, we demonstrate that NRG1 is highly expressed by dermal fibroblasts and cancer-associated fibroblasts (CAFs) isolated from mutant BRAF melanomas. Conditioned medium from fibroblasts and CAFs enhanced ErbB3 pathway activation and limited RAF inhibitor cytotoxicity in V600 mutant BRAF-harboring melanomas. Targeting the ErbB3/ErbB2 pathway partially reversed the protective effects of fibroblast/CAF-derived NRG1 on cell growth properties of RAF inhibitor-treated melanoma cells. These findings support the idea that NRG1, acting in a paracrine manner, promotes resistance to RAF inhibitors and emphasize that targeting the ErbB3/ErbB2 pathway will likely improve the efficacy of RAF inhibitors for mutant BRAF melanoma patients.
Collapse
Affiliation(s)
| | - Sheera Rosenbaum
- From the Department of Cancer Biology, Sidney Kimmel Cancer Center, and
| | - Adam C Berger
- Department of Surgery, Division of General Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Andrew E Aplin
- From the Department of Cancer Biology, Sidney Kimmel Cancer Center, and
| |
Collapse
|