1
|
Hossain MA. A comprehensive review of targeting RAF kinase in cancer. Eur J Pharmacol 2025; 986:177142. [PMID: 39577552 DOI: 10.1016/j.ejphar.2024.177142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024]
Abstract
RAF kinases, particularly the BRAF isoform, play a crucial role in the MAPK/ERK signaling pathway, regulating key cellular processes such as proliferation, differentiation, and survival. Dysregulation of this pathway often caused by mutations in the BRAF gene or alterations in upstream regulators like Ras and receptor tyrosine kinases contributes significantly to cancer development. Mutations, such as BRAF-V600E, are present in a variety of malignancies, with the highest prevalence in melanoma. Targeted therapies against RAF kinases have achieved substantial success, especially in BRAF-V600E-mutant melanomas, where inhibitors like vemurafenib and dabrafenib have demonstrated remarkable efficacy, leading to improved patient outcomes. These inhibitors have also shown clinical benefits in cancers such as thyroid and colorectal carcinoma, although to a lesser extent. Despite these successes, therapeutic resistance remains a major hurdle. Resistance mechanisms, including RAF dimerization, feedback reactivation of the MAPK pathway, and paradoxical activation of ERK signaling, often lead to diminished efficacy over time, resulting in disease progression or even secondary malignancies. In response, current research is focusing on novel therapeutic strategies, including combination therapies that target multiple components of the pathway simultaneously, such as MEK inhibitors used in tandem with RAF inhibitors. Additionally, next-generation RAF inhibitors are being developed to address resistance and enhance therapeutic specificity. This review discusses the clinical advancements in RAF-targeted therapies, with a focus on ongoing efforts to overcome therapeutic resistance and enhance outcomes for cancer patients. It also underscores the persistent challenges in effectively targeting RAF kinase in oncology.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
2
|
Loos B, Salas-Bastos A, Nordin A, Debbache J, Stierli S, Cheng PF, Rufli S, Wyss C, Levesque MP, Dummer R, Wong WWL, Pascolo S, Cantù C, Sommer L. TGFβ signaling sensitizes MEKi-resistant human melanoma to targeted therapy-induced apoptosis. Cell Death Dis 2024; 15:925. [PMID: 39709491 DOI: 10.1038/s41419-024-07305-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
The TGFβ signaling pathway is known for its pleiotropic functions in a plethora of biological processes. In melanoma, TGFβ signaling promotes invasiveness and metastasis formation. However, its involvement in the response to therapy is controversial. While several studies have linked TGFβ signaling to elevated resistance to targeted therapy in melanoma, separate findings have indicated a favorable treatment response through TGFβ-mediated increase of cell death. We now found that the outcome of TGFβ signaling in the context of targeted therapy is dose dependent. Unlike low doses, high levels of TGFβ signal activation induce apoptosis upon simultaneous MAPK pathway inhibition, even in targeted therapy resistant melanoma cell lines. Using transcriptomic analyses, combined with genomic target identification of the critical TGFβ signaling effector SMAD4, we demonstrate that parallel activation of TGFβ signaling and MAPK pathway inhibition causes a complete switch of TGFβ target genes from promoting pro-invasive processes to fueling pro-apoptotic pathways. Investigations of underlying mechanisms identified a novel apoptosis-inducing gene signature. Functional validation of signature members highlighted a central role of the pro-apoptotic BCL2 family member BCL2L11 (BIM) in mediating apoptosis in this condition. Using a modified, synthetic version of the TGFB1 mRNA for intra-tumoral injections, we additionally showcase a potential therapeutic application of this treatment combination.
Collapse
Affiliation(s)
- Benjamin Loos
- University of Zürich, Institute of Anatomy, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Adrian Salas-Bastos
- University of Zürich, Institute of Anatomy, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Anna Nordin
- Wallenberg Centre for Molecular Medicine, Linköping University, 58185, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology; Faculty of Medicine and Health Sciences, Linköping University, 58185, Linköping, Sweden
| | - Julien Debbache
- University of Zürich, Institute of Anatomy, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Salome Stierli
- University of Zürich, Institute of Anatomy, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Phil F Cheng
- University of Zürich Hospital, University of Zürich, Department of Dermatology, Raemistrasse 100, 8091, Zürich, Switzerland
| | - Stefanie Rufli
- University of Zurich, Institute of Experimental Immunology, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Conrad Wyss
- University of Zürich Hospital, University of Zürich, Department of Dermatology, Raemistrasse 100, 8091, Zürich, Switzerland
| | - Mitchell P Levesque
- University of Zürich Hospital, University of Zürich, Department of Dermatology, Raemistrasse 100, 8091, Zürich, Switzerland
| | - Reinhard Dummer
- University of Zürich Hospital, University of Zürich, Department of Dermatology, Raemistrasse 100, 8091, Zürich, Switzerland
| | - Wendy Wei-Lynn Wong
- University of Zurich, Institute of Experimental Immunology, Winterthurerstrasse 190, 8057, Zürich, Switzerland
- Department of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | - Steve Pascolo
- University of Zürich Hospital, University of Zürich, Department of Dermatology, Raemistrasse 100, 8091, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | - Claudio Cantù
- Wallenberg Centre for Molecular Medicine, Linköping University, 58185, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology; Faculty of Medicine and Health Sciences, Linköping University, 58185, Linköping, Sweden
| | - Lukas Sommer
- University of Zürich, Institute of Anatomy, Winterthurerstrasse 190, 8057, Zürich, Switzerland.
| |
Collapse
|
3
|
Kot M, Simiczyjew A, Wądzyńska J, Ziętek M, Matkowski R, Nowak D. Characterization of two melanoma cell lines resistant to BRAF/MEK inhibitors (vemurafenib and cobimetinib). Cell Commun Signal 2024; 22:410. [PMID: 39175042 PMCID: PMC11342534 DOI: 10.1186/s12964-024-01788-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND BRAF (v-raf murine sarcoma viral oncogene homolog B1)/MEK (mitogen-activated protein kinase kinase) inhibitors are used for melanoma treatment. Unfortunately, patients treated with this combined therapy develop resistance to treatment quite quickly, but the mechanisms underlying this phenomenon are not yet fully understood. Here, we report and characterize two melanoma cell lines (WM9 and Hs294T) resistant to BRAF (vemurafenib) and MEK (cobimetinib) inhibitors. METHODS Cell viability was assessed via the XTT test. The level of selected proteins as well as activation of signaling pathways were evaluated using Western blotting. The expression of the chosen genes was assessed by RT-PCR. The distribution of cell cycle phases was analyzed by flow cytometry, and confocal microscopy was used to take photos of spheroids. The composition of cytokines secreted by cells was determined using a human cytokine array. RESULTS The resistant cells had increased survival and activation of ERK kinase in the presence of BRAF/MEK inhibitors. The IC50 values for these cells were over 1000 times higher than for controls. Resistant cells also exhibited elevated activation of AKT, p38, and JNK signaling pathways with increased expression of EGFR, ErbB2, MET, and PDGFRβ receptors as well as reduced expression of ErbB3 receptor. Furthermore, these cells demonstrated increased expression of genes encoding proteins involved in drug transport and metabolism. Resistant cells also exhibited features of epithelial-mesenchymal transition and cancer stem cells as well as reduced proliferation rate and elevated cytokine secretion. CONCLUSIONS In summary, this work describes BRAF/MEK-inhibitor-resistant melanoma cells, allowing for better understanding the underlying mechanisms of resistance. The results may thus contribute to the development of new, more effective therapeutic strategies.
Collapse
Affiliation(s)
- Magdalena Kot
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Aleksandra Simiczyjew
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland.
| | - Justyna Wądzyńska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Marcin Ziętek
- Department of Oncology, Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
- Lower Silesian Oncology, Pulmonology, and Hematology Center, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
| | - Rafał Matkowski
- Department of Oncology, Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
- Lower Silesian Oncology, Pulmonology, and Hematology Center, Plac Hirszfelda 12, Wroclaw, 53-413, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw, 50-383, Poland
| |
Collapse
|
4
|
Fateeva A, Eddy K, Chen S. Current State of Melanoma Therapy and Next Steps: Battling Therapeutic Resistance. Cancers (Basel) 2024; 16:1571. [PMID: 38672652 PMCID: PMC11049326 DOI: 10.3390/cancers16081571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Melanoma is the most aggressive and deadly form of skin cancer due to its high propensity to metastasize to distant organs. Significant progress has been made in the last few decades in melanoma therapeutics, most notably in targeted therapy and immunotherapy. These approaches have greatly improved treatment response outcomes; however, they remain limited in their abilities to hinder disease progression due, in part, to the onset of acquired resistance. In parallel, intrinsic resistance to therapy remains an issue to be resolved. In this review, we summarize currently available therapeutic options for melanoma treatment and focus on possible mechanisms that drive therapeutic resistance. A better understanding of therapy resistance will provide improved rational strategies to overcome these obstacles.
Collapse
Affiliation(s)
- Anna Fateeva
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA; (A.F.); (K.E.)
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854, USA
| | - Kevinn Eddy
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA; (A.F.); (K.E.)
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854, USA
| | - Suzie Chen
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA; (A.F.); (K.E.)
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ 08854, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- U.S. Department of Veterans Affairs, New Jersey Health Care System, East Orange, NJ 07018, USA
| |
Collapse
|
5
|
Li S, Zhong H, Wang Z, Chen J, Huang Z, Zou T, You J. Dietary protein restriction regulates skeletal muscle fiber metabolic characteristics associated with the FGF21-ERK1/2 pathway. iScience 2024; 27:109249. [PMID: 38450157 PMCID: PMC10915561 DOI: 10.1016/j.isci.2024.109249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/10/2023] [Accepted: 02/13/2024] [Indexed: 03/08/2024] Open
Abstract
Under conditions of dietary amino acid balance, decreasing the dietary crude protein (CP) level in pigs has a beneficial effect on meat quality. To further elucidate the mechanism, we explored the alteration of muscle fiber characteristics and key regulators related to myogenesis in the skeletal muscle of pigs fed a protein restricted diet. Compared to pigs fed a normal protein diet, dietary protein restriction significantly increased the slow-twitch muscle fiber proportion in skeletal muscle, succinic dehydrogenase (SDH) activity, the concentrations of ascorbate, biotin, palmitoleic acid, and the ratio of s-adenosylhomocysteine (SAM) to s-adenosylhomocysteine (SAH), but the fast-twitch muscle fiber proportion, lactate dehydrogenase (LDH) activity, the concentrations of ATP, glucose-6-phosphate, SAM, and SAH in skeletal muscle, and the ratio of serum triiodothyronine (T3) to tetraiodothyronine (T4) were decreased. In conclusion, we demonstrated that dietary protein restriction induced skeletal muscle fiber remodeling association the regulation of FGF21-ERK1/2-mTORC1 signaling in weaned piglets.
Collapse
Affiliation(s)
- Shuo Li
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China
| | - Haopeng Zhong
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China
| | - Zirui Wang
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China
| | - Jun Chen
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China
| | - Zhouyin Huang
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China
| | - Tiande Zou
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China
| | - Jinming You
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China
| |
Collapse
|
6
|
Kozyra P, Pitucha M. Revisiting the Role of B-RAF Kinase as a Therapeutic Target in Melanoma. Curr Med Chem 2024; 31:2003-2020. [PMID: 37855341 DOI: 10.2174/0109298673258495231011065225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/19/2023] [Accepted: 08/17/2023] [Indexed: 10/20/2023]
Abstract
Malignant melanoma is the rarest but most aggressive and deadly skin cancer. Melanoma is the result of a malignant transformation of melanocytes, which leads to their uncontrolled proliferation. Mutations in the mitogen-activated protein kinase (MAPK) pathway, which are crucial for the control of cellular processes, such as apoptosis, division, growth, differentiation, and migration, are one of its most common causes. BRAF kinase, as one of the known targets of this pathway, has been known for many years as a prominent molecular target in melanoma therapy, and the following mini-review outlines the state-of-the-art knowledge regarding its structure, mutations and mechanisms.
Collapse
Affiliation(s)
- Paweł Kozyra
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, Lublin, PL, 20093, Poland
| | - Monika Pitucha
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, Lublin, PL-20093, Poland
| |
Collapse
|
7
|
Huang F, Cai F, Dahabieh MS, Gunawardena K, Talebi A, Dehairs J, El-Turk F, Park JY, Li M, Goncalves C, Gagnon N, Su J, LaPierre JH, Gaub P, Joyal JS, Mitchell JJ, Swinnen JV, Miller WH, del Rincón SV. Peroxisome disruption alters lipid metabolism and potentiates antitumor response with MAPK-targeted therapy in melanoma. J Clin Invest 2023; 133:e166644. [PMID: 37616051 PMCID: PMC10575734 DOI: 10.1172/jci166644] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 08/22/2023] [Indexed: 08/25/2023] Open
Abstract
Melanomas reprogram their metabolism to rapidly adapt to therapy-induced stress conditions, allowing them to persist and ultimately develop resistance. We report that a subpopulation of melanoma cells tolerate MAPK pathway inhibitors (MAPKis) through a concerted metabolic reprogramming mediated by peroxisomes and UDP-glucose ceramide glycosyltransferase (UGCG). Compromising peroxisome biogenesis, by repressing PEX3 expression, potentiated the proapoptotic effects of MAPKis via an induction of ceramides, an effect limited by UGCG-mediated ceramide metabolism. Cotargeting PEX3 and UGCG selectively eliminated a subset of metabolically active, drug-tolerant CD36+ melanoma persister cells, thereby sensitizing melanoma to MAPKis and delaying resistance. Increased levels of peroxisomal genes and UGCG were found in patient-derived MAPKi-relapsed melanomas, and simultaneously inhibiting PEX3 and UGCG restored MAPKi sensitivity in multiple models of therapy resistance. Finally, combination therapy consisting of a newly identified inhibitor of the PEX3-PEX19 interaction, a UGCG inhibitor, and MAPKis demonstrated potent antitumor activity in preclinical melanoma models, thus representing a promising approach for melanoma treatment.
Collapse
Affiliation(s)
- Fan Huang
- Lady Davis Institute
- Department of Experimental Medicine, and
| | - Feiyang Cai
- Lady Davis Institute
- Department of Experimental Medicine, and
| | | | | | - Ali Talebi
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Farah El-Turk
- McGill University Health Centre, Montreal, Quebec, Canada
- Centre Hospitalier Universitaire Sainte Justine, Montreal, Quebec, Canada
| | - Jae Yeon Park
- McGill University Health Centre, Montreal, Quebec, Canada
| | - Mengqi Li
- Lady Davis Institute
- Department of Experimental Medicine, and
| | | | | | | | | | - Perrine Gaub
- Centre de Recherche, CHU St. Justine, Montréal, Quebec, Canada
| | | | | | - Johannes V. Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Wilson H. Miller
- Lady Davis Institute
- Department of Experimental Medicine, and
- Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Sonia V. del Rincón
- Lady Davis Institute
- Department of Experimental Medicine, and
- Department of Oncology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
8
|
Redondo-Muñoz M, Rodriguez-Baena FJ, Aldaz P, Caballé-Mestres A, Moncho-Amor V, Otaegi-Ugartemendia M, Carrasco-Garcia E, Olias-Arjona A, Lasheras-Otero I, Santamaria E, Bocanegra A, Chocarro L, Grier A, Dzieciatkowska M M, Bigas C, Martin J, Urdiroz-Urricelqui U, Marzo F, Santamaria E, Kochan G, Escors D, Larrayoz IM, Heyn H, D'Alessandro A, Attolini CSO, Matheu A, Wellbrock C, Benitah SA, Sanchez-Laorden B, Arozarena I. Metabolic rewiring induced by ranolazine improves melanoma responses to targeted therapy and immunotherapy. Nat Metab 2023; 5:1544-1562. [PMID: 37563469 PMCID: PMC10513932 DOI: 10.1038/s42255-023-00861-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 07/07/2023] [Indexed: 08/12/2023]
Abstract
Resistance of melanoma to targeted therapy and immunotherapy is linked to metabolic rewiring. Here, we show that increased fatty acid oxidation (FAO) during prolonged BRAF inhibitor (BRAFi) treatment contributes to acquired therapy resistance in mice. Targeting FAO using the US Food and Drug Administration-approved and European Medicines Agency-approved anti-anginal drug ranolazine (RANO) delays tumour recurrence with acquired BRAFi resistance. Single-cell RNA-sequencing analysis reveals that RANO diminishes the abundance of the therapy-resistant NGFRhi neural crest stem cell subpopulation. Moreover, by rewiring the methionine salvage pathway, RANO enhances melanoma immunogenicity through increased antigen presentation and interferon signalling. Combination of RANO with anti-PD-L1 antibodies strongly improves survival by increasing antitumour immune responses. Altogether, we show that RANO increases the efficacy of targeted melanoma therapy through its effects on FAO and the methionine salvage pathway. Importantly, our study suggests that RANO could sensitize BRAFi-resistant tumours to immunotherapy. Since RANO has very mild side-effects, it might constitute a therapeutic option to improve the two main strategies currently used to treat metastatic melanoma.
Collapse
Grants
- P30 CA046934 NCI NIH HHS
- Ministry of Economy and Competitiveness | Instituto de Salud Carlos III (Institute of Health Carlos III)
- Departamento de Salud del Gobierno de Navarra, Spain (Grant Ref. No: GºNa 71/17)
- Marta Redondo-Muñoz is funded by a PhD studentship from the Department of Industry of the Government of Navarra, Spain. MRM acknowledges funding from the Grupo Español Multidisciplinar de Melanoma
- The University of Colorado School of Medicine Metabolomics Core is supported in part by the University of Colorado Cancer Center award from the National Cancer Institute P30CA046934
- David Escors Acknowledges funding from The Spanish Association against Cancer (AECC), PROYE16001ESCO), Biomedicine Project Grant from the Department of Health of the Government of Navarre-FEDER funds (BMED 050-2019, 51-2021) ; Strategic projects from the Department of Industry, Government of Navarre (AGATA, Ref. 0011-1411-2020-000013; LINTERNA, Ref. 0011-1411-2020-000033; DESCARTHES, 0011-1411-2019-000058).
- Research in the S.A.B. laboratory is supported partially by the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (Grant agreement No. 787041), the Government of Cataluña (SGR grant), the Government of Spain (MINECO), the La Marató/TV3 Foundation, the Foundation Lilliane Bettencourt, the Spanish Association for Cancer Research (AECC) and The Worldwide Cancer Research Foundation (WCRF)
- Work in B.S-L´s lab is funded by:PID2019-106852-RBI00 funded by MCIN/AEI/ 10.13039/501100011033, the Melanoma Research Alliance (https://doi.org/10.48050/pc.gr.91574 to B.S-L) and the FERO Foundation.
Collapse
Affiliation(s)
- Marta Redondo-Muñoz
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | | | - Paula Aldaz
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | - Adriá Caballé-Mestres
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Verónica Moncho-Amor
- Cellular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Madrid, Spain
| | | | - Estefania Carrasco-Garcia
- Cellular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Madrid, Spain
| | - Ana Olias-Arjona
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | - Irene Lasheras-Otero
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | - Eva Santamaria
- Hepatology Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Bocanegra
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Luisa Chocarro
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Abby Grier
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Monika Dzieciatkowska M
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Claudia Bigas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Josefina Martin
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Uxue Urdiroz-Urricelqui
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Florencio Marzo
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | - Enrique Santamaria
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Grazyna Kochan
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - David Escors
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Ignacio Marcos Larrayoz
- Biomarkers and Molecular Signaling Group, Center for Biomedical Research of La Rioja (CIBIR), Foundation Rioja Salud, Logroño, Spain
- Unidad Predepartamental de Enfermería, Universidad de La Rioja (UR), Logroño, Spain
| | - Holger Heyn
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Angelo D'Alessandro
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Camille Stephan-Otto Attolini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Ander Matheu
- Cellular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Claudia Wellbrock
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
- Department of Health Sciences, Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| | | | - Imanol Arozarena
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.
- Health Research Institute of Navarre (IdiSNA), Pamplona, Spain.
| |
Collapse
|
9
|
Lasheras-Otero I, Feliu I, Maillo A, Moreno H, Redondo-Muñoz M, Aldaz P, Bocanegra A, Olias-Arjona A, Lecanda F, Fernandez-Irigoyen J, Santamaria E, Larrayoz IM, Gomez-Cabrero D, Wellbrock C, Vicent S, Arozarena I. The Regulators of Peroxisomal Acyl-Carnitine Shuttle CROT and CRAT Promote Metastasis in Melanoma. J Invest Dermatol 2023; 143:305-316.e5. [PMID: 36058299 DOI: 10.1016/j.jid.2022.08.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/26/2022] [Accepted: 08/03/2022] [Indexed: 01/25/2023]
Abstract
Circulating tumor cells are the key link between a primary tumor and distant metastases, but once in the bloodstream, loss of adhesion induces cell death. To identify the mechanisms relevant for melanoma circulating tumor cell survival, we performed RNA sequencing and discovered that detached melanoma cells and isolated melanoma circulating tumor cells rewire lipid metabolism by upregulating fatty acid (FA) transport and FA beta-oxidation‒related genes. In patients with melanoma, high expression of FA transporters and FA beta-oxidation enzymes significantly correlates with reduced progression-free and overall survival. Among the highest expressed regulators in melanoma circulating tumor cells were the carnitine transferases carnitine O-octanoyltransferase and carnitine acetyltransferase, which control the shuttle of peroxisome-derived medium-chain FAs toward mitochondria to fuel mitochondrial FA beta-oxidation. Knockdown of carnitine O-octanoyltransferase or carnitine acetyltransferase and short-term treatment with peroxisomal or mitochondrial FA beta-oxidation inhibitors thioridazine or ranolazine suppressed melanoma metastasis in mice. Carnitine O-octanoyltransferase and carnitine acetyltransferase depletion could be rescued by medium-chain FA supplementation, indicating that the peroxisomal supply of FAs is crucial for the survival of nonadherent melanoma cells. Our study identifies targeting the FA-based cross-talk between peroxisomes and mitochondria as a potential therapeutic opportunity to challenge melanoma progression. Moreover, the discovery of the antimetastatic activity of the Food and Drug Administration‒approved drug ranolazine carries translational potential.
Collapse
Affiliation(s)
- Irene Lasheras-Otero
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Iker Feliu
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Program in Solid Tumors, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Alberto Maillo
- Translational Bioinformatics Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain
| | - Haritz Moreno
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Program in Solid Tumors, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Marta Redondo-Muñoz
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Paula Aldaz
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Ana Bocanegra
- Oncoimmunology Group, Navarrabiomed, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain
| | - Ana Olias-Arjona
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Fernando Lecanda
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Program in Solid Tumors, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain; Center for Biomedical Research Network on Cancer (CIBERONC), Madrid, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Joaquin Fernandez-Irigoyen
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Proteomics Platform, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain
| | - Enrique Santamaria
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Clinical Neuroproteomics Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain
| | - Ignacio M Larrayoz
- Biomarkers and Molecular Signaling Group, Center for Biomedical Research of La Rioja (CIBIR), Foundation Rioja Salud, Logroño, Spain; Pre-departmental Nursing Unit, University of La Rioja (UR), Logroño, La Rioja, Spain
| | - David Gomez-Cabrero
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Translational Bioinformatics Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain; Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Claudia Wellbrock
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain
| | - Silvestre Vicent
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Program in Solid Tumors, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain; Center for Biomedical Research Network on Cancer (CIBERONC), Madrid, Spain
| | - Imanol Arozarena
- Cancer Signaling Unit, Navarrabiomed, University Hospital of Navarra (HUN), Public University of Navarra (UPNA), Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
| |
Collapse
|
10
|
Chaudhary KR, Kinslow CJ, Cheng H, Silva JM, Yu J, Wang TJ, Hei TK, Halmos B, Cheng SK. Smurf2 inhibition enhances chemotherapy and radiation sensitivity in non-small-cell lung cancer. Sci Rep 2022; 12:10140. [PMID: 35710591 PMCID: PMC9203496 DOI: 10.1038/s41598-022-14448-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 06/06/2022] [Indexed: 12/21/2022] Open
Abstract
Lung cancer has been the most common cancer worldwide for several decades. The outcomes of patients with locally advanced lung cancer remain dismal, and only a minority of patients survive more than 5 years. However, tumor therapeutic resistance mechanisms are poorly studied. Identification of therapeutic resistance pathways in lung cancer in order to increase the sensitivity of lung tumor cells to therapeutic agents is a crucial but challenging need. To identify novel genes that modulate the response to platinum-based therapy, we performed a genome-wide high-throughput ribonucleic acid interference (RNAi) screen via transfection of human lung cancer (PC9) cells with a viral short hairpin RNA (shRNA) library. We further validated a potential target via 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and clonogenic survival assays on PC9 and A549 lung tumor cells transfected with small interfering RNAs (siRNAs) to successfully downregulate protein expression and then treated with increasing doses of cisplatin or X-ray radiation. We determined protein expression by immunohistochemistry (IHC) after chemoradiotherapy and analyzed gene expression-based survival outcomes in two cohorts of human non-small-cell lung cancer (NSCLC) patients. The screen identified several targets involved in epithelial-to-mesenchymal transition (EMT), including Smurf1, Smurf2, YAP1, and CEBPZ, and glycolytic pathway proteins, including PFKFB3. Furthermore, we found that the small molecule proteasome inhibitor bortezomib significantly downregulated Smurf2 in lung cancer cells. The addition of bortezomib in combination with cisplatin and radiation therapy in PC9 and A549 cells led to an increase in deoxyribonucleic acid (DNA) double-strand breaks with increased numbers of γ-H2AX-positive cells and upregulation of apoptosis. Finally, we found that Smurf2 protein expression was upregulated in situ after treatment with cisplatin and radiation therapy in a relevant cohort of patients with stage III NSCLC. Additionally, Smurf2 gene expression was the strongest predictor of survival in patients with squamous NSCLC after chemotherapy or chemoradiotherapy. We successfully identified and validated Smurf2 as both a common modulator of resistance and an actionable target in lung cancer. These results suggest the urgent need to investigate clinical Smurf2 inhibition via bortezomib in combination with cisplatin and radiation for patients with locally advanced NSCLC.
Collapse
Affiliation(s)
- Kunal R Chaudhary
- Department of Radiation Oncology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Connor J Kinslow
- Department of Radiation Oncology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Haiying Cheng
- Department of Oncology, Albert Einstein College of Medicine of Yeshiva University/Montefiore Medical Center, Bronx, NY, USA
| | - Jose M Silva
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Tony J Wang
- Department of Radiation Oncology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Tom K Hei
- Department of Radiation Oncology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Balazs Halmos
- Department of Oncology, Albert Einstein College of Medicine of Yeshiva University/Montefiore Medical Center, Bronx, NY, USA.,Division of Hematology/Oncology, Herbert Irving Comprehensive Cancer Center, New York Presbyterian Hospital-Columbia University Medical Center, New York, NY, USA
| | - Simon K Cheng
- Department of Radiation Oncology, Columbia University College of Physicians and Surgeons, New York, NY, USA. .,Department of Radiation Oncology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
11
|
Zhang M, Zhang YY, Chen Y, Wang J, Wang Q, Lu H. TGF-β Signaling and Resistance to Cancer Therapy. Front Cell Dev Biol 2021; 9:786728. [PMID: 34917620 PMCID: PMC8669610 DOI: 10.3389/fcell.2021.786728] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
The transforming growth factor β (TGF-β) pathway, which is well studied for its ability to inhibit cell proliferation in early stages of tumorigenesis while promoting epithelial-mesenchymal transition and invasion in advanced cancer, is considered to act as a double-edged sword in cancer. Multiple inhibitors have been developed to target TGF-β signaling, but results from clinical trials were inconsistent, suggesting that the functions of TGF-β in human cancers are not yet fully explored. Multiple drug resistance is a major challenge in cancer therapy; emerging evidence indicates that TGF-β signaling may be a key factor in cancer resistance to chemotherapy, targeted therapy and immunotherapy. Finally, combining anti-TGF-β therapy with other cancer therapy is an attractive venue to be explored for the treatment of therapy-resistant cancer.
Collapse
Affiliation(s)
- Maoduo Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ying Yi Zhang
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Yongze Chen
- College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jia Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hezhe Lu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Singh N, Bhatnagar S. Machine Learning for Prediction of Drug Targets in Microbe Associated Cardiovascular Diseases by Incorporating Host-pathogen Interaction Network Parameters. Mol Inform 2021; 41:e2100115. [PMID: 34676983 DOI: 10.1002/minf.202100115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/01/2021] [Indexed: 12/20/2022]
Abstract
Host-pathogen interactions play a crucial role in invasion, infection, and induction of immune response in humans. In this work, four machine learning algorithms, namely Logistic regression, K-nearest neighbor, Support Vector Machine, and Random Forest were implemented for the classification of drug targets. The algorithms were trained using 3400 hosts and 3800 pathogen drug and non-drug target proteins as learning instances. For each protein, 68 pathogen and 73 host features were computed that included sequence, structure, biological and host-pathogen network centrality characteristics. The Random Forest classifier model achieved the best accuracy after 10-fold cross-validation. 99 % accuracy was achieved with a ROC-AUC score of 0.99±0.01 for both pathogen and host training sets. The Eigenvector Centrality of host-pathogen interactions and host-host interactions was the top feature in performing classification of pathogen and host targets respectively. Other features important for classification were the presence of catalytic and binding sites, low instability/aliphatic index, and cellular location. The Random Forest classifier was then used for prediction of drug targets involved in Microbe Associated Cardiovascular Diseases. 331 host and 743 pathogen proteins were predicted as drug targets by the random forest model and can be validated experimentally for therapeutic intervention in Microbe Associated Cardiovascular Diseases.
Collapse
Affiliation(s)
- Nirupma Singh
- Department of Biotechnology, Netaji Subhas Institute of Technology, Dwarka, New Delhi, 110078, India
| | - Sonika Bhatnagar
- Department of Biotechnology, Netaji Subhas Institute of Technology, Dwarka, New Delhi, 110078, India.,Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology Dwarka, New Delhi, 110078, India
| |
Collapse
|
13
|
Wang Z, Huang Y, Chu F, Liao K, Cui Z, Chen J, Tang S. Integrated Analysis of DNA methylation and transcriptome profile to identify key features of age-related macular degeneration. Bioengineered 2021; 12:7061-7078. [PMID: 34569899 PMCID: PMC8806579 DOI: 10.1080/21655979.2021.1976502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Age-related macular degeneration (AMD) is a common vision-threatening disease. The current study sought to integrate DNA methylation with transcriptome profile to explore key features in AMD. Gene expression data were obtained from the Gene Expression Omnibus (GEO, accession ID: GSE135092) and DNA methylation data were obtained from the ArrayExpress repository (E-MTAB-7183). A total of 456 differentially expressed genes (DEGs) and 4827 intragenic differentially methylated CpGs (DMCs) were identified between AMD and controls. DEGs and DMCs were intersected and 19 epigenetically induced (EI) genes and 15 epigenetically suppressed (ES) genes were identified. Immune cell infiltration analysis was performed to estimate the abundance of different types of immune cell in each sample. Enrichment scores of inflammatory response and tumor necrosis factor-alpha (TNFα) signaling via nuclear factor kappa B (NF-κb) were positively correlated with abundance of activated memory CD4 T cells and M1 macrophages. Subsequently, two significant random forest classifiers were constructed based on DNA methylation and transcriptome data. SMAD2 and NGFR were selected as key genes through functional epigenetic modules (FEM) analysis. Expression level of SMAD2, NGFR and their integrating proteins was validated in hydrogen peroxide (H2O2) and TNFα co-treated retinal pigment epithelium (RPE) in vitro. The findings of the current study showed that local inflammation and systemic inflammatory host response play key roles in pathogenesis of AMD. SMAD2 and NGFR provide new insight in understanding the molecular mechanism and are potential therapeutic targets for development of AMD therapy.
Collapse
Affiliation(s)
- Zhijie Wang
- Aier School of Ophthalmology, Central South University, Changsha, China.,Aier Eye Institute, Changsha, China
| | - Yinhua Huang
- Aier School of Ophthalmology, Central South University, Changsha, China.,Aier Eye Institute, Changsha, China
| | - Feixue Chu
- Hangzhou Xihu Zhijiang Eye Hospital, Hangzhou, China
| | - Kai Liao
- Aier School of Ophthalmology, Central South University, Changsha, China.,Aier Eye Institute, Changsha, China
| | | | - Jiansu Chen
- Aier School of Ophthalmology, Central South University, Changsha, China.,Aier Eye Institute, Changsha, China.,Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China.,Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Shibo Tang
- Aier School of Ophthalmology, Central South University, Changsha, China.,Aier Eye Institute, Changsha, China.,Cas Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
14
|
Vergani E, Frigerio S, Dugo M, Devecchi A, Feltrin E, De Cecco L, Vallacchi V, Cossa M, Di Guardo L, Manoukian S, Peissel B, Ferrari A, Gallino G, Maurichi A, Rivoltini L, Sensi M, Rodolfo M. Genetic Variants and Somatic Alterations Associated with MITF-E318K Germline Mutation in Melanoma Patients. Genes (Basel) 2021; 12:1440. [PMID: 34573422 PMCID: PMC8469310 DOI: 10.3390/genes12091440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022] Open
Abstract
The MITF-E318K variant has been implicated in genetic predisposition to cutaneous melanoma. We addressed the occurrence of MITF-E318K and its association with germline status of CDKN2A and MC1R genes in a hospital-based series of 248 melanoma patients including cohorts of multiple, familial, pediatric, sporadic and melanoma associated with other tumors. Seven MITF-E318K carriers were identified, spanning every group except the pediatric patients. Three carriers showed mutated CDKN2A, five displayed MC1R variants, while the sporadic carrier revealed no variants. Germline/tumor whole exome sequencing for this carrier revealed germline variants of unknown significance in ATM and FANCI genes and, in four BRAF-V600E metastases, somatic loss of the MITF wild-type allele, amplification of MITF-E318K and deletion of a 9p21.3 chromosomal region including CDKN2A and MTAP. In silico analysis of tumors from MITF-E318K melanoma carriers in the TCGA Pan-Cancer-Atlas dataset confirmed the association with BRAF mutation and 9p21.3 deletion revealing a common genetic pattern. MTAP was the gene deleted at homozygous level in the highest number of patients. These results support the utility of both germline and tumor genome analysis to define tumor groups providing enhanced information for clinical strategies and highlight the importance of melanoma prevention programs for MITF-E318K patients.
Collapse
Affiliation(s)
- Elisabetta Vergani
- Unit of Immunotherapy, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; (E.V.); (S.F.); (V.V.); (L.R.)
| | - Simona Frigerio
- Unit of Immunotherapy, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; (E.V.); (S.F.); (V.V.); (L.R.)
| | - Matteo Dugo
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy; (M.D.); (A.D.); (L.D.C.); (M.S.)
| | - Andrea Devecchi
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy; (M.D.); (A.D.); (L.D.C.); (M.S.)
| | - Erika Feltrin
- CRIBI Biotechnology Center, Via Bassi 58/B, 35131 Padua, Italy;
| | - Loris De Cecco
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy; (M.D.); (A.D.); (L.D.C.); (M.S.)
| | - Viviana Vallacchi
- Unit of Immunotherapy, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; (E.V.); (S.F.); (V.V.); (L.R.)
| | - Mara Cossa
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy;
| | - Lorenza Di Guardo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy;
| | - Siranoush Manoukian
- Unit of Medical Genetics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; (S.M.); (B.P.)
| | - Bernard Peissel
- Unit of Medical Genetics, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; (S.M.); (B.P.)
| | - Andrea Ferrari
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy;
| | - Gianfrancesco Gallino
- Melanoma and Sarcoma Surgery Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; (G.G.); (A.M.)
| | - Andrea Maurichi
- Melanoma and Sarcoma Surgery Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; (G.G.); (A.M.)
| | - Licia Rivoltini
- Unit of Immunotherapy, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; (E.V.); (S.F.); (V.V.); (L.R.)
| | - Marialuisa Sensi
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy; (M.D.); (A.D.); (L.D.C.); (M.S.)
| | - Monica Rodolfo
- Unit of Immunotherapy, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy; (E.V.); (S.F.); (V.V.); (L.R.)
| |
Collapse
|
15
|
Huang F, Santinon F, Flores González RE, del Rincón SV. Melanoma Plasticity: Promoter of Metastasis and Resistance to Therapy. Front Oncol 2021; 11:756001. [PMID: 34604096 PMCID: PMC8481945 DOI: 10.3389/fonc.2021.756001] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022] Open
Abstract
Melanoma is the deadliest form of skin cancer. Although targeted therapies and immunotherapies have revolutionized the treatment of metastatic melanoma, most patients are not cured. Therapy resistance remains a significant clinical challenge. Melanoma comprises phenotypically distinct subpopulations of cells, exhibiting distinct gene signatures leading to tumor heterogeneity and favoring therapeutic resistance. Cellular plasticity in melanoma is referred to as phenotype switching. Regardless of their genomic classification, melanomas switch from a proliferative and differentiated phenotype to an invasive, dedifferentiated and often therapy-resistant state. In this review we discuss potential mechanisms underpinning melanoma phenotype switching, how this cellular plasticity contributes to resistance to both targeted therapies and immunotherapies. Finally, we highlight novel strategies to target plasticity and their potential clinical impact in melanoma.
Collapse
Affiliation(s)
- Fan Huang
- Lady Davis Institute, McGill University, Montréal, QC, Canada
- Department of Experimental Medicine, McGill University, Montréal, QC, Canada
| | - François Santinon
- Lady Davis Institute, McGill University, Montréal, QC, Canada
- Department of Experimental Medicine, McGill University, Montréal, QC, Canada
| | - Raúl Ernesto Flores González
- Lady Davis Institute, McGill University, Montréal, QC, Canada
- Department of Experimental Medicine, McGill University, Montréal, QC, Canada
| | - Sonia V. del Rincón
- Lady Davis Institute, McGill University, Montréal, QC, Canada
- Department of Experimental Medicine, McGill University, Montréal, QC, Canada
- Department of Oncology, McGill University, Montréal, QC, Canada
| |
Collapse
|
16
|
Oba J, Woodman SE. The genetic and epigenetic basis of distinct melanoma types. J Dermatol 2021; 48:925-939. [PMID: 34008215 DOI: 10.1111/1346-8138.15957] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022]
Abstract
Melanoma represents the deadliest skin cancer. Recent therapeutic developments, including targeted and immune therapies have revolutionized clinical management and improved patient outcome. This progress was achieved by rigorous molecular and functional studies followed by robust clinical trials. The identification of key genomic alterations and gene expression profiles have propelled the understanding of distinct characteristics within melanoma subtypes. The aim of this review is to summarize and highlight the main genetic and epigenetic findings of melanomas and highlight their pathological and therapeutic importance.
Collapse
Affiliation(s)
- Junna Oba
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Scott E Woodman
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
17
|
Chen X, Li Y, Yao T, Jia R. Benefits of Zebrafish Xenograft Models in Cancer Research. Front Cell Dev Biol 2021; 9:616551. [PMID: 33644052 PMCID: PMC7905065 DOI: 10.3389/fcell.2021.616551] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
As a promising in vivo tool for cancer research, zebrafish have been widely applied in various tumor studies. The zebrafish xenograft model is a low-cost, high-throughput tool for cancer research that can be established quickly and requires only a small sample size, which makes it favorite among researchers. Zebrafish patient-derived xenograft (zPDX) models provide promising evidence for short-term clinical treatment. In this review, we discuss the characteristics and advantages of zebrafish, such as their transparent and translucent features, the use of vascular fluorescence imaging, the establishment of metastatic and intracranial orthotopic models, individual pharmacokinetics measurements, and tumor microenvironment. Furthermore, we introduce how these characteristics and advantages are applied other in tumor studies. Finally, we discuss the future direction of the use of zebrafish in tumor studies and provide new ideas for the application of it.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Tengteng Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
18
|
Ballotti R, Cheli Y, Bertolotto C. The complex relationship between MITF and the immune system: a Melanoma ImmunoTherapy (response) Factor? Mol Cancer 2020; 19:170. [PMID: 33276788 PMCID: PMC7718690 DOI: 10.1186/s12943-020-01290-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/29/2020] [Indexed: 12/18/2022] Open
Abstract
The clinical benefit of immune checkpoint inhibitory therapy (ICT) in advanced melanomas is limited by primary and acquired resistance. The molecular determinants of the resistance have been extensively studied, but these discoveries have not yet been translated into therapeutic benefits. As such, a paradigm shift in melanoma treatment, to surmount the therapeutic impasses linked to the resistance, is an important ongoing challenge.This review outlines the multifaceted interplay between microphthalmia-associated transcription factor (MITF), a major determinant of the biology of melanoma cells, and the immune system. In melanomas, MITF functions downstream oncogenic pathways and microenvironment stimuli that restrain the immune responses. We highlight how MITF, by controlling differentiation and genome integrity, may regulate melanoma-specific antigen expression by interfering with the endolysosomal pathway, KARS1, and antigen processing and presentation. MITF also modulates the expression of coinhibitory receptors, i.e., PD-L1 and HVEM, and the production of an inflammatory secretome, which directly affects the infiltration and/or activation of the immune cells.Furthermore, MITF is also a key determinant of melanoma cell plasticity and tumor heterogeneity, which are undoubtedly one of the major hurdles for an effective immunotherapy. Finally, we briefly discuss the role of MITF in kidney cancer, where it also plays a key role, and in immune cells, establishing MITF as a central mediator in the regulation of immune responses in melanoma and other cancers.We propose that a better understanding of MITF and immune system intersections could help in the tailoring of current ICT in melanomas and pave the way for clinical benefits and long-lasting responses.
Collapse
Affiliation(s)
- Robert Ballotti
- Université Côte d'Azur, Nice, France
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020 and Equipe labellisée ARC 2019, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Yann Cheli
- Université Côte d'Azur, Nice, France
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020 and Equipe labellisée ARC 2019, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Corine Bertolotto
- Université Côte d'Azur, Nice, France.
- Inserm, Biology and Pathologies of melanocytes, team1, Equipe labellisée Ligue 2020 and Equipe labellisée ARC 2019, Centre Méditerranéen de Médecine Moléculaire, Nice, France.
| |
Collapse
|
19
|
Guo Y, Li X, He Z, Ma D, Zhang Z, Wang W, Xiong J, Kuang X, Wang J. HDAC3 Silencing Enhances Acute B Lymphoblastic Leukaemia Cells Sensitivity to MG-132 by Inhibiting the JAK/Signal Transducer and Activator of Transcription 3 Signaling Pathway. Chemotherapy 2020; 65:85-100. [PMID: 32966974 DOI: 10.1159/000500713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/30/2019] [Indexed: 11/19/2022]
Abstract
PURPOSE HDAC3, which is associated with smurf2, has been shown to be associated with poor prognosis in B-ALL. This study examined the efficacy of targeting HDAC3 combined with MG-132 as a possible therapeutic strategy for B-ALL patients. METHODS Real-time PCR and western blot were used to measure the expression of smurf2 and HDAC3 from B-ALL patients bone marrow samples. Sup-B15 and CCRF-SB cells were treated with MG-132, small interfering RNA of smurf2 or HDAC3. A plasmid designed to up-regulate smurf2 expression was transfected into B-ALL cells. Flow cytometry and western blot were used to measure variation due to these treatments in terms of apoptosis and cell cycle arrest. RESULTS Expression of Smurf2 and HDAC3 mRNA were inversely related in B-ALL patients. Up-regulation of smurf2 or MG-132 influenced HDAC3, further inhibiting the JAK/signal transducer and activator of transcription 3 (STAT3) signal pathway and inducing apoptosis in B-ALL cells. When we treated Sup-B15 and CCRF-SB cells with siHDAC3 and MG-132 for 24 h, silencing HDAC3 enhanced the apoptosis rate induced by MG-132 in B-ALL cells and further inhibited the JAK/STAT3 pathway. Furthermore, MG-132 was observed to cause G2/M phase arrest in B-ALL cells and inhibited the JAK/STAT3 pathway, leading to apoptosis. CONCLUSIONS Silencing of HDAC3 enhanced the sensitivity of B-ALL cells to MG-132. The combination of targeting HDAC3 and MG-132 may provide a new avenue for clinical treatment of acute B lymphocytic leukaemia and improve the poor survival of leukaemia patients.
Collapse
Affiliation(s)
- Yongling Guo
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China.,Department of Hematology, Guiyang Hospital of Guizhou Aviation Industry Group, Guiyang, China
| | - Xinyao Li
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Zhengchang He
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Dan Ma
- Key Laboratory of Hematological Disease Diagnostic Treat Centre of Guizhou Province, Guiyang, China
| | - Zhaoyuan Zhang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Weili Wang
- College of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jie Xiong
- Key Laboratory of Hematological Disease Diagnostic Treat Centre of Guizhou Province, Guiyang, China
| | - Xinyi Kuang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Jishi Wang
- Key Laboratory of Hematological Disease Diagnostic Treat Centre of Guizhou Province, Guiyang, China,
| |
Collapse
|
20
|
Tangella LP, Clark ME, Gray ES. Resistance mechanisms to targeted therapy in BRAF-mutant melanoma - A mini review. Biochim Biophys Acta Gen Subj 2020; 1865:129736. [PMID: 32956754 DOI: 10.1016/j.bbagen.2020.129736] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/28/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The introduction of targeted therapies for the treatment of BRAF-mutant melanomas have improved survival rates in a significant proportion of patients. Nonetheless, the emergence of resistance to treatment remains inevitable in most patients. SCOPE OF REVIEW Here, we review known and emerging molecular mechanisms that underlay the development of resistance to MAPK inhibition in melanoma cells and the potential strategies to overcome these mechanisms. MAJOR CONCLUSIONS Multiple genetic and non-genetic mechanisms contribute to treatment failure, commonly leading to the reactivation of the MAPK pathway. A variety of resistance mechanisms are enabled by the underlying heterogeneity and plasticity of melanoma cells. Moreover, it has become apparent that resistance to targeted therapy is underpinned by early functional adaptations involving the rewiring of cell states and metabolic pathways. GENERAL SIGNIFICANCE The evidence presented suggest that the use of a combinatorial treatment approach would delay the emergence of resistance and improve patient outcomes.
Collapse
Affiliation(s)
- Lokeswari P Tangella
- School of Medical and Health Sciences, Edith Cowan University, Perth 6027, Western Australia, Australia
| | - Michael E Clark
- School of Medical and Health Sciences, Edith Cowan University, Perth 6027, Western Australia, Australia
| | - Elin S Gray
- School of Medical and Health Sciences, Edith Cowan University, Perth 6027, Western Australia, Australia.
| |
Collapse
|
21
|
Targen S, Kaya T, Avci ME, Gunes D, Keskus AG, Konu O. ZenoFishDb v1.1: A Database for Xenotransplantation Studies in Zebrafish. Zebrafish 2020; 17:305-318. [PMID: 32931381 DOI: 10.1089/zeb.2020.1869] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Rapidly accumulating literature has proven feasibility of the zebrafish xenograft models in cancer research. Nevertheless, online databases for searching the current zebrafish xenograft literature are in great demand. Herein, we have developed a manually curated database, called ZenoFishDb v1.1 (https://konulab.shinyapps.io/zenofishdb), based on R Shiny platform aiming to provide searchable information on ever increasing collection of zebrafish studies for cancer cell line transplantation and patient-derived xenografts (PDXs). ZenoFishDb v1.1 user interface contains four modules: DataTable, Visualization, PDX Details, and PDX Charts. The DataTable and Visualization pages represent xenograft study details, including injected cell lines, PDX injections, molecular modifications of cell lines, zebrafish strains, as well as technical aspects of the xenotransplantation procedures in table, bar, and/or pie chart formats. The PDX Details module provides comprehensive information on the patient details in table format and can be searched and visualized. Overall, ZenoFishDb v1.1 enables researchers to effectively search, list, and visualize different technical and biological attributes of zebrafish xenotransplantation studies particularly focusing on the new trends that make use of reporters, RNA interference, overexpression, or mutant gene constructs of transplanted cancer cells, stem cells, and PDXs, as well as distinguished host modifications.
Collapse
Affiliation(s)
- Seniye Targen
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Tuğberk Kaya
- Interdisciplinary Program in Neuroscience, Bilkent University, Ankara, Turkey.,Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - M Ender Avci
- Izmir Biomedicine and Genome Center, Dokuz Eylul University, Izmir, Turkey
| | - Damla Gunes
- Interdisciplinary Program in Neuroscience, Bilkent University, Ankara, Turkey
| | - Ayse Gokce Keskus
- Interdisciplinary Program in Neuroscience, Bilkent University, Ankara, Turkey
| | - Ozlen Konu
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey.,Interdisciplinary Program in Neuroscience, Bilkent University, Ankara, Turkey.,UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| |
Collapse
|
22
|
Fazio M, Ablain J, Chuan Y, Langenau DM, Zon LI. Zebrafish patient avatars in cancer biology and precision cancer therapy. Nat Rev Cancer 2020; 20:263-273. [PMID: 32251397 PMCID: PMC8011456 DOI: 10.1038/s41568-020-0252-3] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2020] [Indexed: 01/05/2023]
Abstract
In precision oncology, two major strategies are being pursued for predicting clinically relevant tumour behaviours, such as treatment response and emergence of drug resistance: inference based on genomic, transcriptomic, epigenomic and/or proteomic analysis of patient samples, and phenotypic assays in personalized cancer avatars. The latter approach has historically relied on in vivo mouse xenografts and in vitro organoids or 2D cell cultures. Recent progress in rapid combinatorial genetic modelling, the development of a genetically immunocompromised strain for xenotransplantation of human patient samples in adult zebrafish and the first clinical trial using xenotransplantation in zebrafish larvae for phenotypic testing of drug response bring this tiny vertebrate to the forefront of the precision medicine arena. In this Review, we discuss advances in transgenic and transplantation-based zebrafish cancer avatars, and how these models compare with and complement mouse xenografts and human organoids. We also outline the unique opportunities that these different models present for prediction studies and current challenges they face for future clinical deployment.
Collapse
Affiliation(s)
- Maurizio Fazio
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Julien Ablain
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Yan Chuan
- Molecular Pathology Unit, Cancer Center, Massachusetts General Hospital Research Institute, Charlestown, MA, USA
| | - David M Langenau
- Molecular Pathology Unit, Cancer Center, Massachusetts General Hospital Research Institute, Charlestown, MA, USA
| | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
23
|
Dissecting Mechanisms of Melanoma Resistance to BRAF and MEK Inhibitors Revealed Genetic and Non-Genetic Patient- and Drug-Specific Alterations and Remarkable Phenotypic Plasticity. Cells 2020; 9:cells9010142. [PMID: 31936151 PMCID: PMC7017165 DOI: 10.3390/cells9010142] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/29/2019] [Accepted: 01/03/2020] [Indexed: 12/14/2022] Open
Abstract
The clinical benefit of MAPK pathway inhibition in BRAF-mutant melanoma patients is limited by the development of acquired resistance. Using drug-naïve cell lines derived from tumor specimens, we established a preclinical model of melanoma resistance to vemurafenib or trametinib to provide insight into resistance mechanisms. Dissecting the mechanisms accompanying the development of resistance, we have shown that (i) most of genetic and non-genetic alterations are triggered in a cell line- and/or drug-specific manner; (ii) several changes previously assigned to the development of resistance are induced as the immediate response to the extent measurable at the bulk levels; (iii) reprogramming observed in cross-resistance experiments and growth factor-dependence restricted by the drug presence indicate that phenotypic plasticity of melanoma cells largely contributes to the sustained resistance. Whole-exome sequencing revealed novel genetic alterations, including a frameshift variant of RBMX found exclusively in phospho-AKThigh resistant cell lines. There was no similar pattern of phenotypic alterations among eleven resistant cell lines, including expression/activity of crucial regulators, such as MITF, AXL, SOX, and NGFR, which suggests that patient-to-patient variability is richer and more nuanced than previously described. This diversity should be considered during the development of new strategies to circumvent the acquired resistance to targeted therapies.
Collapse
|
24
|
Boumahdi S, de Sauvage FJ. The great escape: tumour cell plasticity in resistance to targeted therapy. Nat Rev Drug Discov 2020; 19:39-56. [PMID: 31601994 DOI: 10.1038/s41573-019-0044-1] [Citation(s) in RCA: 455] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2019] [Indexed: 01/05/2023]
Abstract
The success of targeted therapies in cancer treatment has been impeded by various mechanisms of resistance. Besides the acquisition of resistance-conferring genetic mutations, reversible mechanisms that lead to drug tolerance have emerged. Plasticity in tumour cells drives their transformation towards a phenotypic state that no longer depends on the drug-targeted pathway. These drug-refractory cells constitute a pool of slow-cycling cells that can either regain drug sensitivity upon treatment discontinuation or acquire permanent resistance to therapy and drive relapse. In the past few years, cell plasticity has emerged as a mode of targeted therapy evasion in various cancers, ranging from prostate and lung adenocarcinoma to melanoma and basal cell carcinoma. Our understanding of the mechanisms that control this phenotypic switch has also expanded, revealing the crucial role of reprogramming factors and chromatin remodelling. Further deciphering the molecular basis of tumour cell plasticity has the potential to contribute to new therapeutic strategies which, combined with existing anticancer treatments, could lead to deeper and longer-lasting clinical responses.
Collapse
Affiliation(s)
- Soufiane Boumahdi
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA
| | | |
Collapse
|
25
|
Ferguson J, Wilcock DJ, McEntegart S, Badrock AP, Levesque M, Dummer R, Wellbrock C, Smith MP. Osteoblasts contribute to a protective niche that supports melanoma cell proliferation and survival. Pigment Cell Melanoma Res 2020; 33:74-85. [PMID: 31323160 PMCID: PMC6972519 DOI: 10.1111/pcmr.12812] [Citation(s) in RCA: 5] [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: 12/03/2018] [Revised: 06/17/2019] [Accepted: 07/04/2019] [Indexed: 12/19/2022]
Abstract
Melanoma is the deadliest form of skin cancer; a primary driver of this high level of morbidity is the propensity of melanoma cells to metastasize. When malignant tumours develop distant metastatic lesions the new local tissue niche is known to impact on the biology of the cancer cells. However, little is known about how different metastatic tissue sites impact on frontline targeted therapies. Intriguingly, melanoma bone lesions have significantly lower response to BRAF or MEK inhibitor therapies. Here, we have investigated how the cellular niche of the bone can support melanoma cells by stimulating growth and survival via paracrine signalling between osteoblasts and cancer cells. Melanoma cells can enhance the differentiation of osteoblasts leading to increased production of secreted ligands, including RANKL. Differentiated osteoblasts in turn can support melanoma cell proliferation and survival via the secretion of RANKL that elevates the levels of the transcription factor MITF, even in the presence of BRAF inhibitor. By blocking RANKL signalling, either via neutralizing antibodies, genetic alterations or the RANKL receptor inhibitor SPD304, the survival advantage provided by osteoblasts could be overcome.
Collapse
Affiliation(s)
- Jennifer Ferguson
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Daniel J. Wilcock
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Sophie McEntegart
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Andrew P. Badrock
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Mitch Levesque
- Department of Dermatology, Universitäts Spital ZürichUniversity of ZürichZurichSwitzerland
| | - Reinhard Dummer
- Department of Dermatology, Universitäts Spital ZürichUniversity of ZürichZurichSwitzerland
| | - Claudia Wellbrock
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Michael P. Smith
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| |
Collapse
|
26
|
Yao Y, Wang L, Wang X. Modeling of Solid-Tumor Microenvironment in Zebrafish (Danio Rerio) Larvae. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:413-428. [PMID: 32130712 DOI: 10.1007/978-3-030-34025-4_22] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The zebrafish larvae have emerged as a powerful model for studying tumorigenesis in vivo, with remarkable conservation with mammals in genetics, molecular and cell biology. Zebrafish tumor models bear the significant advantages of optical clarity in comparison to that in the mammalian models, allowing noninvasive investigation of the tumor cell and its microenvironment at single-cell resolution. Here we review recent progressions in the field of zebrafish models of solid tumor diseases in two main categories: the genetically engineered tumor models in which all cells in the tumor microenvironment are zebrafish cells, and xenograft tumor models in which the tumor microenvironment is composed of zebrafish cells and cells from other species. Notably, the zebrafish patient-derived xenograft (zPDX) models can be used for personalized drug assessment on primary tumor biopsies, including the pancreatic cancer. For the future studies, a series of high throughput drug screenings on the library of transgenic zebrafish models of solid tumor are expected to provide systematic database of oncogenic mutation, cell-of-origin, and leading compounds; and the humanization of zebrafish in genetics and cellular composition will make it more practical hosts for zPDX modeling. Together, zebrafish tumor model systems are unique and convenient in vivo platforms, with great potential to serve as valuable tools for cancer researches.
Collapse
Affiliation(s)
- Yuxiao Yao
- Cancer Metabolism Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lei Wang
- Cancer Metabolism Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xu Wang
- Cancer Metabolism Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
27
|
Bai X, Fisher DE, Flaherty KT. Cell-state dynamics and therapeutic resistance in melanoma from the perspective of MITF and IFNγ pathways. Nat Rev Clin Oncol 2019; 16:549-562. [PMID: 30967646 PMCID: PMC7185899 DOI: 10.1038/s41571-019-0204-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Targeted therapy and immunotherapy have greatly improved the prognosis of patients with metastatic melanoma, but resistance to these therapeutic modalities limits the percentage of patients with long-lasting responses. Accumulating evidence indicates that a persisting subpopulation of melanoma cells contributes to resistance to targeted therapy or immunotherapy, even in patients who initially have a therapeutic response; however, the root mechanism of resistance remains elusive. To address this problem, we propose a new model, in which dynamic fluctuations of protein expression at the single-cell level and longitudinal reshaping of the cellular state at the cell-population level explain the whole process of therapeutic resistance development. Conceptually, we focused on two different pivotal signalling pathways (mediated by microphthalmia-associated transcription factor (MITF) and IFNγ) to construct the evolving trajectories of melanoma and described each of the cell states. Accordingly, the development of therapeutic resistance could be divided into three main phases: early survival of cell populations, reversal of senescence, and the establishment of new homeostatic states and development of irreversible resistance. On the basis of existing data, we propose future directions in both translational research and the design of therapeutic strategies that incorporate this emerging understanding of resistance.
Collapse
Affiliation(s)
- Xue Bai
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - David E Fisher
- Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Adaptive Responses as Mechanisms of Resistance to BRAF Inhibitors in Melanoma. Cancers (Basel) 2019; 11:cancers11081176. [PMID: 31416288 PMCID: PMC6721815 DOI: 10.3390/cancers11081176] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022] Open
Abstract
: The introduction of v-raf murine sarcoma viral oncogene homolog B (BRAF) inhibitors in melanoma patients with BRAF (V600E) mutations has demonstrated significant clinical benefits. However, rarely do tumours regress completely. Frequently, the reason for this is that therapies targeting specific oncogenic mutations induce a number of intrinsic compensatory mechanisms, also known as adaptive responses or feedback loops, that enhance the pro-survival and pro-proliferative capacity of a proportion of the original tumour population, thereby resulting in tumour progression. In this review we will summarize the known adaptive responses that limit BRAF mutant therapy and discuss potential novel combinatorial therapies to overcome resistance.
Collapse
|
29
|
Plasticity of Drug-Naïve and Vemurafenib- or Trametinib-Resistant Melanoma Cells in Execution of Differentiation/Pigmentation Program. JOURNAL OF ONCOLOGY 2019; 2019:1697913. [PMID: 31354817 PMCID: PMC6636509 DOI: 10.1155/2019/1697913] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/09/2019] [Indexed: 12/13/2022]
Abstract
Melanoma plasticity creates a plethora of opportunities for cancer cells to escape treatment. Thus, therapies must target all cancer cell subpopulations bearing the potential to contribute to disease. The role of the differentiation/pigmentation program in intrinsic and acquired drug resistance is largely uncharacterized. MITF level and expression of MITF-dependent pigmentation-related genes, MLANA, PMEL, TYR, and DCT, in drug-naïve and vemurafenib- or trametinib-treated patient-derived melanoma cell lines and their drug-resistant counterparts were analysed and referred to genomic alterations. Variability in execution of pigmentation/differentiation program was detected in patient-derived melanoma cell lines. Acute treatment with vemurafenib or trametinib enhanced expression of pigmentation-related genes in MITF-Mhigh melanoma cells, partially as the consequence of transcriptional reprograming. During development of resistance, changes in pigmentation program were not unidirectional, but also not universal as expression of different pigmentation-related genes was diversely affected. In selected resistant cell lines, differentiation/pigmentation was promoted and might be considered as one of drug-tolerant phenotypes. In other resistant lines, dedifferentiation was induced. Upon drug withdrawal ("drug holiday"), the dedifferentiation process in resistant cells either was enhanced but reversed by drug reexposure suggesting involvement of epigenetic mechanisms or was irreversible. The irreversible dedifferentiation might be connected with homozygous loss-of-function mutation in MC1R, as MC1RR151C +/+ variant was found exclusively in drug-naïve MITF-Mlow dedifferentiated cells and drug-resistant cells derived from MITFhigh/MC1RWT cells undergoing irreversible dedifferentiation. MC1RR151C +/+ variant might be further investigated as a parameter potentially impacting melanoma patient stratification and aiding in treatment decision.
Collapse
|
30
|
Arozarena I, Wellbrock C. Phenotype plasticity as enabler of melanoma progression and therapy resistance. Nat Rev Cancer 2019; 19:377-391. [PMID: 31209265 DOI: 10.1038/s41568-019-0154-4] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/13/2019] [Indexed: 12/31/2022]
Abstract
Malignant melanoma is notorious for its inter- and intratumour heterogeneity, based on transcriptionally distinct melanoma cell phenotypes. It is thought that these distinct phenotypes are plastic in nature and that their transcriptional reprogramming enables heterogeneous tumours both to undergo different stages of melanoma progression and to adjust to drug exposure during treatment. Recent advances in genomic technologies and the rapidly expanding availability of large gene expression datasets have allowed for a refined definition of the gene signatures that characterize these phenotypes and have revealed that phenotype plasticity plays a major role in the resistance to both targeted therapy and immunotherapy. In this Review we discuss the definition of melanoma phenotypes through particular transcriptional states and reveal the prognostic relevance of the related gene expression signatures. We review how the establishment of phenotypes is controlled and which roles phenotype plasticity plays in melanoma development and therapy. Because phenotype plasticity in melanoma bears a great resemblance to epithelial-mesenchymal transition, the lessons learned from melanoma will also benefit our understanding of other cancer types.
Collapse
Affiliation(s)
- Imanol Arozarena
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| | - Claudia Wellbrock
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
31
|
Tuncer E, Calçada RR, Zingg D, Varum S, Cheng P, Freiberger SN, Deng CX, Kleiter I, Levesque MP, Dummer R, Sommer L. SMAD signaling promotes melanoma metastasis independently of phenotype switching. J Clin Invest 2019; 129:2702-2716. [PMID: 31039140 DOI: 10.1172/jci94295] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The development of metastatic melanoma is thought to require the dynamic shifting of neoplastic cells between proliferative and invasive phenotypes. Contrary to this conventional "phenotype switching" model, we now show that disease progression can involve malignant melanoma cells simultaneously displaying proliferative and invasive properties. Using a genetic mouse model of melanoma in combination with in vitro analyses of melanoma cell lines, we found that conditional deletion of the downstream signaling molecule Smad4, which abrogates all canonical TGF-β signaling, indeed inhibits both tumor growth and metastasis. Conditional deletion of the inhibitory signaling factor Smad7, however, generated cells that are both highly invasive and proliferative, indicating that invasiveness is compatible with a high proliferation rate. In fact, conditional Smad7 deletion led to sustained melanoma growth and at the same time promoted massive metastasis formation, a result consistent with data indicating that low SMAD7 levels in patient tumors are associated with a poor survival. Our findings reveal that modulation of SMAD7 levels can overcome the need for phenotype switching during tumor progression and may thus represent a novel therapeutic target in metastatic disease.
Collapse
Affiliation(s)
- Eylul Tuncer
- Stem Cell Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Raquel R Calçada
- Stem Cell Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Daniel Zingg
- Stem Cell Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Sandra Varum
- Stem Cell Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Phil Cheng
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | | | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Ingo Kleiter
- Department of Neurology, Ruhr-University Bochum, Bochum, Germany and Marianne-Strauß-Klinik, Behandlungszentrum Kempfenhausen für Multiple Sklerose Kranke gGmbH, Berg, Germany
| | | | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Lukas Sommer
- Stem Cell Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Mo X, Preston S, Zaidi MR. Macroenvironment-gene-microenvironment interactions in ultraviolet radiation-induced melanomagenesis. Adv Cancer Res 2019; 144:1-54. [PMID: 31349897 DOI: 10.1016/bs.acr.2019.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cutaneous malignant melanoma is one of the few major cancers that continue to exhibit a positive rate of increase in the developed world. A wealth of epidemiological data has undisputedly implicated ultraviolet radiation (UVR) from sunlight and artificial sources as the major risk factor for melanomagenesis. However, the molecular mechanisms of this cause-and-effect relationship remain murky and understudied. Recent efforts on multiple fronts have brought unprecedented expansion of our knowledge base on this subject and it is now clear that melanoma is caused by a complex interaction between genetic predisposition and environmental exposure, primarily to UVR. Here we provide an overview of the effects of the macroenvironment (UVR) on the skin microenvironment and melanocyte-specific intrinsic (mostly genetic) landscape, which conspire to produce one of the deadliest malignancies.
Collapse
Affiliation(s)
- Xuan Mo
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Sarah Preston
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - M Raza Zaidi
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.
| |
Collapse
|
33
|
Many ways to resistance: How melanoma cells evade targeted therapies. Biochim Biophys Acta Rev Cancer 2019; 1871:313-322. [DOI: 10.1016/j.bbcan.2019.02.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/20/2019] [Accepted: 02/13/2019] [Indexed: 12/25/2022]
|
34
|
Rabbie R, Ferguson P, Molina‐Aguilar C, Adams DJ, Robles‐Espinoza CD. Melanoma subtypes: genomic profiles, prognostic molecular markers and therapeutic possibilities. J Pathol 2019; 247:539-551. [PMID: 30511391 PMCID: PMC6492003 DOI: 10.1002/path.5213] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 12/24/2022]
Abstract
Melanoma is characterised by its ability to metastasise at early stages of tumour development. Current clinico-pathologic staging based on the American Joint Committee on Cancer criteria is used to guide surveillance and management in early-stage disease, but its ability to predict clinical outcome has limitations. Herein we review the genomics of melanoma subtypes including cutaneous, acral, uveal and mucosal, with a focus on the prognostic and predictive significance of key molecular aberrations. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Roy Rabbie
- Experimental Cancer GeneticsThe Wellcome Sanger InstituteHinxtonUK
- Cambridge Cancer CentreCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Peter Ferguson
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred HospitalSydneyAustralia
- Melanoma Institute Australia, The University of SydneySydneyAustralia
| | - Christian Molina‐Aguilar
- Laboratorio Internacional de Investigación sobre el Genoma HumanoUniversidad Nacional Autónoma de MéxicoSantiago de QuerétaroMexico
| | - David J Adams
- Experimental Cancer GeneticsThe Wellcome Sanger InstituteHinxtonUK
| | - Carla D Robles‐Espinoza
- Experimental Cancer GeneticsThe Wellcome Sanger InstituteHinxtonUK
- Laboratorio Internacional de Investigación sobre el Genoma HumanoUniversidad Nacional Autónoma de MéxicoSantiago de QuerétaroMexico
| |
Collapse
|
35
|
Liu Y, Cui S, Li W, Zhao Y, Yan X, Xu J. PAX3 is a biomarker and prognostic factor in melanoma: Database mining. Oncol Lett 2019; 17:4985-4993. [PMID: 31186709 PMCID: PMC6507366 DOI: 10.3892/ol.2019.10155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 02/14/2019] [Indexed: 11/24/2022] Open
Abstract
Paired box 3 (PAX3) is a transcription factor and critical regulator of pigment cell development during embryonic development. However, while there have been several studies on PAX3, its expression patterns and precise role remain to be clarified. The present study is an in-depth computational study of tumor-associated gene information, with specific emphasis on the expression of PAX3 in melanoma, using Oncomine along with an investigation of corresponding expression profiles in an array of cancer cell lines through Cancer Cell Line Encyclopedia analysis. Based on Kaplan-Meier analysis, the prognostic value of high PAX3 expression in tissues from patients with melanoma compared with normal tissues was assessed. PAX3 was more highly expressed in male patients with melanoma compared with female patients with melanoma. Using Oncomine and Coexpedia analysis, it was demonstrated that PAX3 expression was clearly associated with SRY-box 10 expression. The survival analysis results revealed that high PAX3 mRNA expression was associated with worse survival rates in patients with melanoma. These results suggested that PAX3 may be a biomarker and essential prognostic factor for melanoma, and provided an important theoretical basis for the development of melanoma treatments.
Collapse
Affiliation(s)
- Yong Liu
- Department of Dermatology, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi 710003, P.R. China
| | - Shengnan Cui
- Department of Hematology, The Second Affiliated Hospital, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, P.R. China
| | - Wenbin Li
- Department of Dermatology, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi 710003, P.R. China
| | - Yiding Zhao
- Department of Dermatology, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi 710003, P.R. China
| | - Xiaoning Yan
- Department of Dermatology, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi 710003, P.R. China
| | - Jianqin Xu
- Department of Dermatology, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi 710003, P.R. China
| |
Collapse
|
36
|
Smith MP, Rana S, Ferguson J, Rowling EJ, Flaherty KT, Wargo JA, Marais R, Wellbrock C. A PAX3/BRN2 rheostat controls the dynamics of BRAF mediated MITF regulation in MITF high /AXL low melanoma. Pigment Cell Melanoma Res 2019; 32:280-291. [PMID: 30277012 PMCID: PMC6392120 DOI: 10.1111/pcmr.12741] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 12/14/2022]
Abstract
The BRAF kinase and the MAPK pathway are targets of current melanoma therapies. However, MAPK pathway inhibition results in dynamic changes of downstream targets that can counteract inhibitor-action not only in during treatment, but also in acquired resistant tumours. One such dynamic change involves the expression of the transcription factor MITF, a crucial regulator of cell survival and proliferation in untreated as well as drug-addicted acquired resistant melanoma. Tight control over MITF expression levels is required for optimal melanoma growth, and while it is well established that the MAPK pathway regulates MITF expression, the actual mechanism is insufficiently understood. We reveal here, how BRAF through action on the transcription factors BRN2 and PAX3 executes control over the regulation of MITF expression in a manner that allows for considerable plasticity. This plasticity provides robustness to the BRAF mediated MITF regulation and explains the dynamics in MITF expression that are observed in patients in response to MAPK inhibitor therapy.
Collapse
Affiliation(s)
- Michael P. Smith
- Manchester Cancer Research Centre, Faculty of Biology, Medicine & Health, Division of Cancer SciencesThe University of ManchesterManchesterUK
| | - Sareena Rana
- Division of Molecular PathologyThe Institute of Cancer ResearchLondonUK
| | - Jennifer Ferguson
- Manchester Cancer Research Centre, Faculty of Biology, Medicine & Health, Division of Cancer SciencesThe University of ManchesterManchesterUK
| | - Emily J. Rowling
- Manchester Cancer Research Centre, Faculty of Biology, Medicine & Health, Division of Cancer SciencesThe University of ManchesterManchesterUK
| | | | - Jennifer A. Wargo
- Divison of Surgical OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTexas
| | - Richard Marais
- Molecular Oncology GroupCancer Research UK Manchester Institute, The University of Manchester, Astra Zeneca Logistics CentreMacclesfieldUK
| | - Claudia Wellbrock
- Manchester Cancer Research Centre, Faculty of Biology, Medicine & Health, Division of Cancer SciencesThe University of ManchesterManchesterUK
| |
Collapse
|
37
|
Ooshima A, Park J, Kim SJ. Phosphorylation status at Smad3 linker region modulates transforming growth factor-β-induced epithelial-mesenchymal transition and cancer progression. Cancer Sci 2019; 110:481-488. [PMID: 30589983 PMCID: PMC6361575 DOI: 10.1111/cas.13922] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/19/2018] [Accepted: 12/23/2018] [Indexed: 01/09/2023] Open
Abstract
Smad3, a major transcription factor in transforming growth factor-β (TGF-β) signaling, plays critical roles in both tumor-suppressive and pro-oncogenic functions. Upon TGF-β stimulation, the C-terminal tail of Smad3 undergoes phosphorylation that is essential for canonical TGF-β signaling. The Smad3 linker region contains serine/threonine phosphorylation sites and can be phosphorylated by intracellular kinases, such as the MAPK family, cyclin-dependent kinase (CDK) family and glycogen synthase kinase-3β (GSK-3β). Previous reports based on cell culture studies by us and others showed that mutation of Smad3 linker phosphorylation sites dramatically intensifies TGF-β responses as well as growth-inhibitory function and epithelial-mesenchymal transition (EMT), suggesting that Smad3 linker phosphorylation suppresses TGF-β transcriptional activities. However, recent discoveries of Smad3-interacting molecules that preferentially bind phosphorylated Smad3 linker serine/threonine residues have shown a multitude of signal transductions that either enhance or suppress TGF-β responses associated with Smad3 turnover or cancer progression. This review aims at providing new insight into the perplexing mechanisms of TGF-β signaling affected by Smad3 linker phosphorylation and further attempts to gain insight into elimination and protection of TGF-β-mediated oncogenic and growth-suppressive signals, respectively.
Collapse
Affiliation(s)
- Akira Ooshima
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea
| | - Jinah Park
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea
| | - Seong-Jin Kim
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea.,Graduate School of Convergence Science and Technology, Seoul National University, Suwon, Korea
| |
Collapse
|
38
|
Koganti P, Levy-Cohen G, Blank M. Smurfs in Protein Homeostasis, Signaling, and Cancer. Front Oncol 2018; 8:295. [PMID: 30116722 PMCID: PMC6082930 DOI: 10.3389/fonc.2018.00295] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022] Open
Abstract
Protein ubiquitination is an evolutionary conserved highly-orchestrated enzymatic cascade essential for normal cellular functions and homeostasis maintenance. This pathway relies on a defined set of cellular enzymes, among them, substrate-specific E3 ubiquitin ligases (E3s). These ligases are the most critical players, as they define the spatiotemporal nature of ubiquitination and confer specificity to this cascade. Smurf1 and Smurf2 (Smurfs) are the C2-WW-HECT-domain E3 ubiquitin ligases, which recently emerged as important determinants of pivotal cellular processes. These processes include cell proliferation and differentiation, chromatin organization and dynamics, DNA damage response and genomic integrity maintenance, gene expression, cell stemness, migration, and invasion. All these processes are intimately connected and profoundly altered in cancer. Initially, Smurf proteins were identified as negative regulators of the bone morphogenetic protein (BMP) and the transforming growth factor beta (TGF-β) signaling pathways. However, recent studies have extended the scope of Smurfs' biological functions beyond the BMP/TGF-β signaling regulation. Here, we provide a critical literature overview and updates on the regulatory roles of Smurfs in molecular and cell biology, with an emphasis on cancer. We also highlight the studies demonstrating the impact of Smurf proteins on tumor cell sensitivity to anticancer therapies. Further in-depth analyses of Smurfs' biological functions and influences on molecular pathways could provide novel therapeutic targets and paradigms for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Praveen Koganti
- Laboratory of Molecular and Cellular Cancer Biology, Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Gal Levy-Cohen
- Laboratory of Molecular and Cellular Cancer Biology, Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Michael Blank
- Laboratory of Molecular and Cellular Cancer Biology, Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
39
|
Pan J, Huang C, Chen G, Cai Z, Zhang Z. MicroRNA-451 blockade promotes osteoblastic differentiation and skeletal anabolic effects by promoting YWHAZ-mediated RUNX2 protein stabilization. MEDCHEMCOMM 2018; 9:1359-1368. [PMID: 30151091 PMCID: PMC6097111 DOI: 10.1039/c8md00187a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 06/28/2018] [Indexed: 12/12/2022]
Abstract
Background: senile osteoporosis researchers are now seeking to promote osteoblastogenesis and resultant bone formation to directly counteract age-related bone loss. Targeting microRNA (miRNA) activity in adult osteoblasts may be a successful therapeutic strategy for age-related bone loss. We investigated the mechanism(s) by which miRNAs negatively regulate osteoblastogenesis and bone formation in vitro and in vivo. Methods: we performed a miRNA microarray screen followed by PCR validation in adult bone marrow-derived mesenchymal stem cells during the proliferation-to-mineralization transition to identify downregulated miRNAs, most notably miR-451. Primary human calvarial pre-osteoblasts were isolated and transfected with miR-451's agomir or antagomir for in vitro assays. Bioinformatics analysis and in vitro experiments verified YWHAZ as a miR-451 target gene. We next investigated the effects of YWHAZ knockdown on osteoblastic differentiation. To examine the effects of miR-451's antagomir in vivo, we injected ovariectomized (OVX) or sham-operated mice with miR-451's antagomir over a period of six weeks. We isolated stromal cells from murine bone marrow on week six for further ex vivo experimentation. Results: miR-451's antagomir stimulated pre-osteoblast differentiation into a more differentiated, mineralized phenotype. This phenotype was associated with upregulated RUNX2, ALP, and COL1A1 protein expression. miR-451's antagomir derepresses YWHAZ expression, thereby enhancing RUNX2 protein stability and promoting osteoblastic differentiation. When injected in vivo, miR-451's antagomir promotes osteoblastogenesis and mineralization, reversed OVX-induced bone loss, and increased bone strength in OVX and sham-operated mice. Conclusions: miR-451 suppresses osteoblastogenesis in vitro and in vivo. miR-451 inhibition may serve as an effective anabolic therapeutic strategy in senile osteoporosis patients.
Collapse
Affiliation(s)
- Jieen Pan
- Department of Orthopaedics , The Second Hospital of Jiaxing , No. 1518, Huancheng Road Nanhu District , Jiaxing 314000 , Zhejiang Province , P.R. China .
| | - Chenglong Huang
- Department of Orthopaedics , The Second Hospital of Jiaxing , No. 1518, Huancheng Road Nanhu District , Jiaxing 314000 , Zhejiang Province , P.R. China .
| | - Gang Chen
- Department of Orthopaedics , The Second Hospital of Jiaxing , No. 1518, Huancheng Road Nanhu District , Jiaxing 314000 , Zhejiang Province , P.R. China .
| | - Zhenhai Cai
- Department of Orthopaedics , The Second Hospital of Jiaxing , No. 1518, Huancheng Road Nanhu District , Jiaxing 314000 , Zhejiang Province , P.R. China .
| | - Zhongwei Zhang
- Department of Orthopaedics , The Second Hospital of Jiaxing , No. 1518, Huancheng Road Nanhu District , Jiaxing 314000 , Zhejiang Province , P.R. China .
| |
Collapse
|
40
|
Rambow F, Rogiers A, Marin-Bejar O, Aibar S, Femel J, Dewaele M, Karras P, Brown D, Chang YH, Debiec-Rychter M, Adriaens C, Radaelli E, Wolter P, Bechter O, Dummer R, Levesque M, Piris A, Frederick DT, Boland G, Flaherty KT, van den Oord J, Voet T, Aerts S, Lund AW, Marine JC. Toward Minimal Residual Disease-Directed Therapy in Melanoma. Cell 2018; 174:843-855.e19. [PMID: 30017245 DOI: 10.1016/j.cell.2018.06.025] [Citation(s) in RCA: 447] [Impact Index Per Article: 63.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 04/13/2018] [Accepted: 06/12/2018] [Indexed: 01/05/2023]
Abstract
Many patients with advanced cancers achieve dramatic responses to a panoply of therapeutics yet retain minimal residual disease (MRD), which ultimately results in relapse. To gain insights into the biology of MRD, we applied single-cell RNA sequencing to malignant cells isolated from BRAF mutant patient-derived xenograft melanoma cohorts exposed to concurrent RAF/MEK-inhibition. We identified distinct drug-tolerant transcriptional states, varying combinations of which co-occurred within MRDs from PDXs and biopsies of patients on treatment. One of these exhibited a neural crest stem cell (NCSC) transcriptional program largely driven by the nuclear receptor RXRG. An RXR antagonist mitigated accumulation of NCSCs in MRD and delayed the development of resistance. These data identify NCSCs as key drivers of resistance and illustrate the therapeutic potential of MRD-directed therapy. They also highlight how gene regulatory network architecture reprogramming may be therapeutically exploited to limit cellular heterogeneity, a key driver of disease progression and therapy resistance.
Collapse
Affiliation(s)
- Florian Rambow
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Aljosja Rogiers
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Oskar Marin-Bejar
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sara Aibar
- Laboratory of Computational Biology, VIB Center for Brain & Disease Research, KU Leuven, Leuven, Belgium; Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Julia Femel
- Department of Cell, Developmental, and Cancer Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Michael Dewaele
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Panagiotis Karras
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Daniel Brown
- Laboratory of reproductive genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR, USA
| | - Maria Debiec-Rychter
- Laboratory for Genetics of Malignant Disorders, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Carmen Adriaens
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Enrico Radaelli
- Comparative Pathology Core, University of Pennsylvania, Department of Pathobiology, Philadelphia, PA, USA
| | - Pascal Wolter
- Department of General Medical Oncology, UZ Leuven, Leuven, Belgium
| | - Oliver Bechter
- Department of General Medical Oncology, UZ Leuven, Leuven, Belgium
| | - Reinhard Dummer
- Department of Dermatology, University of Zürich Hospital, Zürich, Switzerland
| | - Mitchell Levesque
- Department of Dermatology, University of Zürich Hospital, Zürich, Switzerland
| | - Adriano Piris
- Department of Surgical Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Dennie T Frederick
- Department of Surgical Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Genevieve Boland
- Department of Surgical Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Keith T Flaherty
- Department of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Joost van den Oord
- Laboratory of Translational Cell and Tissue Research, Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Thierry Voet
- Laboratory of reproductive genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Stein Aerts
- Laboratory of Computational Biology, VIB Center for Brain & Disease Research, KU Leuven, Leuven, Belgium; Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Amanda W Lund
- Department of Cell, Developmental, and Cancer Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
41
|
Miskolczi Z, Smith MP, Rowling EJ, Ferguson J, Barriuso J, Wellbrock C. Collagen abundance controls melanoma phenotypes through lineage-specific microenvironment sensing. Oncogene 2018; 37:3166-3182. [PMID: 29545604 PMCID: PMC5992128 DOI: 10.1038/s41388-018-0209-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/16/2018] [Accepted: 02/13/2018] [Indexed: 01/15/2023]
Abstract
Despite the general focus on an invasive and de-differentiated phenotype as main driver of cancer metastasis, in melanoma patients many metastatic lesions display a high degree of pigmentation, indicative for a differentiated phenotype. Indeed, studies in mice and fish show that melanoma cells switch to a differentiated phenotype at secondary sites, possibly because in melanoma differentiation is closely linked to proliferation through the lineage-specific transcriptional master regulator MITF. Importantly, while a lot of effort has gone into identifying factors that induce the de-differentiated/invasive phenotype, it is not well understood how the switch to the differentiated/proliferative phenotype is controlled. We identify collagen as a contributor to this switch. We demonstrate that collagen stiffness induces melanoma differentiation through a YAP/PAX3/MITF axis and show that in melanoma patients increased collagen abundance correlates with nuclear YAP localization. However, the interrogation of large patient datasets revealed that in the context of the tumour microenvironment, YAP function is more complex. In the absence of fibroblasts, YAP/PAX3-mediated transcription prevails, but in the presence of fibroblasts tumour growth factor-β suppresses YAP/PAX3-mediated MITF expression and induces YAP/TEAD/SMAD-driven transcription and a de-differentiated phenotype. Intriguingly, while high collagen expression is correlated with poorer patient survival, the worst prognosis is seen in patients with high collagen expression, who also express MITF target genes such as the differentiation markers TRPM1, TYR and TYRP1, as well as CDK4. In summary, we reveal a distinct lineage-specific route of YAP signalling that contributes to the regulation of melanoma pigmentation and uncovers a set of potential biomarkers predictive for poor survival.
Collapse
Affiliation(s)
- Zsofia Miskolczi
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, School of Medical Sciences, Division of Cancer Sciences, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Michael P Smith
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, School of Medical Sciences, Division of Cancer Sciences, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Emily J Rowling
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, School of Medical Sciences, Division of Cancer Sciences, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Jennifer Ferguson
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, School of Medical Sciences, Division of Cancer Sciences, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Jorge Barriuso
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, School of Medical Sciences, Division of Cancer Sciences, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Claudia Wellbrock
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, School of Medical Sciences, Division of Cancer Sciences, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
42
|
Ahmed F, Haass NK. Microenvironment-Driven Dynamic Heterogeneity and Phenotypic Plasticity as a Mechanism of Melanoma Therapy Resistance. Front Oncol 2018; 8:173. [PMID: 29881716 PMCID: PMC5976798 DOI: 10.3389/fonc.2018.00173] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/03/2018] [Indexed: 12/11/2022] Open
Abstract
Drug resistance constitutes a major challenge in designing melanoma therapies. Microenvironment-driven tumor heterogeneity and plasticity play a key role in this phenomenon. Melanoma is highly heterogeneous with diverse genomic alterations and expression of different biological markers. In addition, melanoma cells are highly plastic and capable of adapting quickly to changing microenvironmental conditions. These contribute to variations in therapy response and durability between individual melanoma patients. In response to changing microenvironmental conditions, like hypoxia and nutrient starvation, proliferative melanoma cells can switch to an invasive slow-cycling state. Cells in this state are more aggressive and metastatic, and show increased intrinsic drug resistance. During continuous treatment, slow-cycling cells are enriched within the tumor and give rise to a new proliferative subpopulation with increased drug resistance, by exerting their stem cell-like behavior and phenotypic plasticity. In melanoma, the proliferative and invasive states are defined by high and low microphthalmia-associated transcription factor (MITF) expression, respectively. It has been observed that in MITFhigh melanomas, inhibition of MITF increases the efficacy of targeted therapies and delays the acquisition of drug resistance. Contrarily, MITF is downregulated in melanomas with acquired drug resistance. According to the phenotype switching theory, the gene expression profile of the MITFlow state is predominantly regulated by WNT5A, AXL, and NF-κB signaling. Thus, different combinations of therapies should be effective in treating different phases of melanoma, such as the combination of targeted therapies with inhibitors of MITF expression during the initial treatment phase, but with inhibitors of WNT5A/AXL/NF-κB signaling during relapse.
Collapse
Affiliation(s)
- Farzana Ahmed
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Nikolas K. Haass
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
- Discipline of Dermatology, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
43
|
Kundu A, Khouri MG, Aryana S, Firestone GL. 1-Benzyl-indole-3-carbinol is a highly potent new small molecule inhibitor of Wnt/β-catenin signaling in melanoma cells that coordinately inhibits cell proliferation and disrupts expression of microphthalmia-associated transcription factor isoform-M. Carcinogenesis 2017; 38:1207-1217. [PMID: 29028954 DOI: 10.1093/carcin/bgx103] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 09/19/2017] [Indexed: 11/15/2022] Open
Abstract
1-Benzyl-indole-3-carbinol (1-benzyl-I3C), a synthetic analogue of the crucifer-derived natural phytochemical I3C, displayed significantly wider sensitivity and anti-proliferative potency in melanoma cells than the natural compound. Unlike I3C, which targets mainly oncogenic BRAF-expressing cells, 1-benzyl-I3C effectively inhibited proliferation of melanoma cells with a more extensive range of mutational profiles, including those expressing wild-type BRAF. In both cultured melanoma cell lines and in vivo in melanoma cell-derived tumor xenografts, 1-benzyl-I3C disrupted canonical Wnt/β-catenin signaling that resulted in the downregulation of β-catenin protein levels with a concomitant increase in levels of the β-catenin destruction complex components such as glycogen synthase kinase-3β (GSK-3β) and Axin. Concurrent with the inhibition of Wnt/β-catenin signaling, 1-benzyl-I3C strongly downregulated expression of the melanoma master regulator, microphthalmia-associated transcription factor isoform-M (MITF-M) by inhibiting promoter activity through the consensus lymphoid enhancer factor-1 (LEF-1)/T-cell transcription factor (TCF) DNA-binding site. Chromatin immunoprecipitation revealed that 1-benzyl-I3C downregulated interactions of endogenous LEF-1 with the MITF-M promoter. 1-Benzyl-I3C ablated Wnt-activated LEF-1-dependent reporter gene activity in a TOP FLASH assay that was rescued by expression of a constitutively active form of the Wnt co-receptor low-density lipoprotein receptor-related protein (LRP6), indicating that 1-benzyl-I3C disrupts Wnt/β-catenin signaling at or upstream of LRP6. In oncogenic BRAF-expressing melanoma cells, combinations of 1-benzyl-I3C and Vemurafenib, a clinically employed BRAF inhibitor, showed strong anti-proliferative effects. Taken together, our observations demonstrate that 1-benzyl-I3C represents a new and highly potent indolecarbinol-based small molecule inhibitor of Wnt/β-catenin signaling that has intriguing translational potential, alone or in combination with other anti-cancer agents, to treat human melanoma.
Collapse
Affiliation(s)
- Aishwarya Kundu
- Department of Molecular and Cell Biology and the Cancer Research Laboratory, University of California at Berkeley, USA
| | - Michelle G Khouri
- Department of Molecular and Cell Biology and the Cancer Research Laboratory, University of California at Berkeley, USA
| | - Sheila Aryana
- Department of Molecular and Cell Biology and the Cancer Research Laboratory, University of California at Berkeley, USA
| | | |
Collapse
|
44
|
Astone M, Dankert EN, Alam SK, Hoeppner LH. Fishing for cures: The alLURE of using zebrafish to develop precision oncology therapies. NPJ Precis Oncol 2017; 1. [PMID: 29376139 PMCID: PMC5784449 DOI: 10.1038/s41698-017-0043-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Zebrafish have proven to be a valuable model to study human cancer biology with the ultimate aim of developing new therapies. Danio rerio are amenable to in vivo imaging, high-throughput drug screening, mutagenesis, and transgenesis, and they share histological and genetic similarities with Homo sapiens. The significance of zebrafish in the field of precision oncology is rapidly emerging. Indeed, modeling cancer in zebrafish has already been used to identify tumor biomarkers, define therapeutic targets and provide an in vivo platform for drug discovery. New zebrafish studies are starting to pave the way to direct individualized clinical applications. Patient-derived cancer cell xenograft models have demonstrated the feasibility of using zebrafish as a real-time avatar of prognosis and drug response to identify the most ideal therapy for an individual patient. Genetic cancer modeling in zebrafish, now facilitated by rapidly evolving genome editing techniques, represents another innovative approach to recapitulate human oncogenesis and develop individualized treatments. Utilizing zebrafish to design customizable precision therapies will improve the clinical outcome of patients afflicted with cancer.
Collapse
Affiliation(s)
- Matteo Astone
- The Hormel Institute, University of Minnesota, Austin, MN, 55912
| | - Erin N Dankert
- The Hormel Institute, University of Minnesota, Austin, MN, 55912
| | - Sk Kayum Alam
- The Hormel Institute, University of Minnesota, Austin, MN, 55912
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, 55912
| |
Collapse
|
45
|
Arozarena I, Wellbrock C. Overcoming resistance to BRAF inhibitors. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:387. [PMID: 29114545 PMCID: PMC5653517 DOI: 10.21037/atm.2017.06.09] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022]
Abstract
The discovery of activating mutations in the serine/threonine (S/T) kinase BRAF followed by a wave of follow-up research manifested that the MAPK-pathway plays a critical role in melanoma initiation and progression. BRAF and MEK inhibitors produce an unparalleled response rate in melanoma, but it is now clear that most responses are transient, and while some patients show long lasting responses the majority progress within 1 year. In accordance with the key role played by the MAPK-pathway in BRAF mutant melanomas, disease progression is mostly due to the appearance of drug-resistance mechanisms leading to restoration of MAPK-pathway activity. In the present article we will review the development, application and clinical effects of BRAF and MEK inhibitors both, as single agent and in combination in the context of targeted therapy in melanoma. We will then describe the most prominent mechanisms of resistance found in patients progressed on these targeted therapies. Finally we will discuss strategies for further optimizing the use of MAPK inhibitors and will describe the potential of alternative combination therapies to either delay the onset of resistance to MAPK inhibitors or directly target specific mechanisms of resistance to BRAF/MEK inhibitors.
Collapse
Affiliation(s)
- Imanol Arozarena
- Navarrabiomed-Fundación Miguel Servet-Idisna, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Claudia Wellbrock
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
46
|
Smith MP, Rowling EJ, Miskolczi Z, Ferguson J, Spoerri L, Haass NK, Sloss O, McEntegart S, Arozarena I, von Kriegsheim A, Rodriguez J, Brunton H, Kmarashev J, Levesque MP, Dummer R, Frederick DT, Andrews MC, Cooper ZA, Flaherty KT, Wargo JA, Wellbrock C. Targeting endothelin receptor signalling overcomes heterogeneity driven therapy failure. EMBO Mol Med 2017; 9:1011-1029. [PMID: 28606996 PMCID: PMC5538298 DOI: 10.15252/emmm.201607156] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 05/04/2017] [Accepted: 05/05/2017] [Indexed: 11/17/2022] Open
Abstract
Approaches to prolong responses to BRAF targeting drugs in melanoma patients are challenged by phenotype heterogeneity. Melanomas of a "MITF-high" phenotype usually respond well to BRAF inhibitor therapy, but these melanomas also contain subpopulations of the de novo resistance "AXL-high" phenotype. > 50% of melanomas progress with enriched "AXL-high" populations, and because AXL is linked to de-differentiation and invasiveness avoiding an "AXL-high relapse" is desirable. We discovered that phenotype heterogeneity is supported during the response phase of BRAF inhibitor therapy due to MITF-induced expression of endothelin 1 (EDN1). EDN1 expression is enhanced in tumours of patients on treatment and confers drug resistance through ERK re-activation in a paracrine manner. Most importantly, EDN1 not only supports MITF-high populations through the endothelin receptor B (EDNRB), but also AXL-high populations through EDNRA, making it a master regulator of phenotype heterogeneity. Endothelin receptor antagonists suppress AXL-high-expressing cells and sensitize to BRAF inhibition, suggesting that targeting EDN1 signalling could improve BRAF inhibitor responses without selecting for AXL-high cells.
Collapse
Affiliation(s)
- Michael P Smith
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Emily J Rowling
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Zsofia Miskolczi
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Jennifer Ferguson
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Loredana Spoerri
- Translational Research Institute, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Nikolas K Haass
- Translational Research Institute, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
- Discipline of Dermatology, University of Sydney, Sydney, NSW, Australia
| | - Olivia Sloss
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Sophie McEntegart
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Imanol Arozarena
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Navarrabiomed-Fundación Miguel Servet-Idisna, Pamplona, Spain
| | | | - Javier Rodriguez
- Systems Biology Ireland, School of Medicine, UCD, Dublin 4, Ireland
| | - Holly Brunton
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Jivko Kmarashev
- Department of Dermatology, Universitätsspital Zürich, University of Zurich, Zurich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, Universitätsspital Zürich, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, Universitätsspital Zürich, University of Zurich, Zurich, Switzerland
| | - Dennie T Frederick
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Miles C Andrews
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zachary A Cooper
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keith T Flaherty
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Jennifer A Wargo
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Claudia Wellbrock
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
47
|
Najem A, Krayem M, Salès F, Hussein N, Badran B, Robert C, Awada A, Journe F, Ghanem GE. P53 and MITF/Bcl-2 identified as key pathways in the acquired resistance of NRAS-mutant melanoma to MEK inhibition. Eur J Cancer 2017; 83:154-165. [PMID: 28738256 DOI: 10.1016/j.ejca.2017.06.033] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/19/2017] [Accepted: 06/27/2017] [Indexed: 01/03/2023]
Abstract
Activating mutations in Neuroblastoma RAS viral oncogene homolog (NRAS) are found in 15-30% of melanomas and are associated with a poor prognosis. Although MAP kinase kinase (MEK) inhibitors used as single agents showed a limited clinical benefit in patients with NRAS-mutant melanoma due to their rather cytostatic effect and high toxicity, their combination with other inhibitors of pathways known to cooperate with MEK inhibition may maximise their antitumour activity. Similarly, in a context where p53 is largely inactivated in melanoma, hyperexpression of Microphthalmia associated transcription factor (MITF) and its downstream anti-apoptotic targets may be the cause of the restraint cytotoxic effects of MEK inhibitors. Indeed, drug combinations targeting both mutant BRAF and MITF or one of its important targets Bcl-2 were effective in mutant BRAF melanoma but had no effect on acquired resistance. Therefore, we aimed to further investigate the downstream MITF targets that can explain this anti-apoptotic effect and to evaluate in parallel the effect of p53 reactivation on the promotion of apoptosis under MEK inhibition in a panel of Q61NRAS-mutant melanoma cells. First, we showed that MEK inhibition (pimasertib) led to a significant inhibition of cell proliferation but with a limited effect on apoptosis that could be explained by the systematic MITF upregulation. Mimicking the MITF effect via cyclic adenosine monophosphate activation conferred resistance to MEK inhibition and upregulated Bcl-2 expression. In addition, acquired resistance to MEK inhibition was associated with a strong activation of the anti-apoptotic signalling MITF/Bcl-2. More importantly, selective Bcl-2 inhibition by ABT-199 or Bcl-2 knockout using CRISPR/Cas9 system annihilated the acquired resistance and restored the sensitivity of NRAS-mutant melanoma cells to MEK inhibition. Strikingly and similarly, direct p53 reactivation (PRIMA-1Met, APR-246) also broke resistance and synergised with MEK inhibition to induce massive apoptosis in NRAS-mutant melanoma cells with wild-type or mutant p53. Hence, our data identify MITF/Bcl-2 as a key mechanism underlying resistance of NRAS-mutant melanoma cells to apoptosis by MEK inhibitors and paves the way for a promising drug combination that could prevent or reverse anti-MEK resistance in this group of patients.
Collapse
Affiliation(s)
- Ahmad Najem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Mohammad Krayem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - François Salès
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium; Department of Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Nader Hussein
- Department of Biochemistry, Lebanese University, Beirut, Lebanon
| | - Bassam Badran
- Department of Biochemistry, Lebanese University, Beirut, Lebanon
| | | | - Ahmad Awada
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Fabrice Journe
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium; Service d'Anatomie Humaine et d'Oncologie Expérimentale, Université de Mons, Mons, Belgium
| | - Ghanem E Ghanem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
48
|
Arozarena I, Wellbrock C. Targeting invasive properties of melanoma cells. FEBS J 2017; 284:2148-2162. [PMID: 28196297 DOI: 10.1111/febs.14040] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/26/2017] [Accepted: 02/10/2017] [Indexed: 02/11/2024]
Abstract
Melanoma is a skin cancer notorious for its metastatic potential. As an initial step of the metastatic cascade, melanoma cells part from the primary tumour and invade the surrounding tissue, which is crucial for their dissemination and the formation of distant secondary tumours. Over the last two decades, our understanding of both, general and melanoma specific mechanisms of invasion has significantly improved, but to date no efficient therapeutic strategy tackling the invasive properties of melanoma cells has reached the clinic. In this review, we assess the major contributions towards the understanding of the molecular biology of melanoma cell invasion with a focus on melanoma specific traits. These traits are based on the neural crest origin of melanoma cells and explain their intrinsic invasive nature. A particular emphasis is given not only to lineage specific signalling mediated by TGFβ, and noncanonical and canonical WNT signalling, but also to the role of PDE5A and RHO-GTPases in modulating modes of melanoma cell invasion. We discuss existing caveats in the current understanding of the metastatic properties of melanoma cells, as well as the relevance of the 'phenotype switch' model and 'co-operativity' between different phenotypes in heterogeneous tumours. At the centre of these phenotypes is the lineage commitment factor microphthalmia-associated transcription factor, one of the most crucial regulators of the balance between de-differentiation (neural crest specific gene expression) and differentiation (melanocyte specific gene expression) that defines invasive and noninvasive melanoma cell phenotypes. Finally, we provide insight into the current evidence linking resistance to targeted therapies to invasive properties of melanoma cells.
Collapse
Affiliation(s)
- Imanol Arozarena
- Cancer Signalling Group, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Claudia Wellbrock
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| |
Collapse
|
49
|
Young HL, Rowling EJ, Bugatti M, Giurisato E, Luheshi N, Arozarena I, Acosta JC, Kamarashev J, Frederick DT, Cooper ZA, Reuben A, Gil J, Flaherty KT, Wargo JA, Vermi W, Smith MP, Wellbrock C, Hurlstone A. An adaptive signaling network in melanoma inflammatory niches confers tolerance to MAPK signaling inhibition. J Exp Med 2017; 214:1691-1710. [PMID: 28450382 PMCID: PMC5460994 DOI: 10.1084/jem.20160855] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 12/16/2016] [Accepted: 03/10/2017] [Indexed: 12/22/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) pathway antagonists induce profound clinical responses in advanced cutaneous melanoma, but complete remissions are frustrated by the development of acquired resistance. Before resistance emerges, adaptive responses establish a mutation-independent drug tolerance. Antagonizing these adaptive responses could improve drug effects, thereby thwarting the emergence of acquired resistance. In this study, we reveal that inflammatory niches consisting of tumor-associated macrophages and fibroblasts contribute to treatment tolerance through a cytokine-signaling network that involves macrophage-derived IL-1β and fibroblast-derived CXCR2 ligands. Fibroblasts require IL-1β to produce CXCR2 ligands, and loss of host IL-1R signaling in vivo reduces melanoma growth. In tumors from patients on treatment, signaling from inflammatory niches is amplified in the presence of MAPK inhibitors. Signaling from inflammatory niches counteracts combined BRAF/MEK (MAPK/extracellular signal-regulated kinase kinase) inhibitor treatment, and consequently, inhibiting IL-1R or CXCR2 signaling in vivo enhanced the efficacy of MAPK inhibitors. We conclude that melanoma inflammatory niches adapt to and confer drug tolerance toward BRAF and MEK inhibitors early during treatment.
Collapse
Affiliation(s)
- Helen L Young
- Manchester Cancer Research Centre, Faculty of Biology, Medicine, and Health, School of Medical Sciences, Division of Molecular and Clinical Cancer Studies, The University of Manchester, Manchester M13 9PT, England, UK
| | - Emily J Rowling
- Manchester Cancer Research Centre, Faculty of Biology, Medicine, and Health, School of Medical Sciences, Division of Molecular and Clinical Cancer Studies, The University of Manchester, Manchester M13 9PT, England, UK
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, 25123 Brescia, Italy
| | - Emanuele Giurisato
- Manchester Cancer Research Centre, Faculty of Biology, Medicine, and Health, School of Medical Sciences, Division of Molecular and Clinical Cancer Studies, The University of Manchester, Manchester M13 9PT, England, UK
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Nadia Luheshi
- Division of Oncology, MedImmune Ltd, Cambridge CB21 6GH, England, UK
| | - Imanol Arozarena
- Manchester Cancer Research Centre, Faculty of Biology, Medicine, and Health, School of Medical Sciences, Division of Molecular and Clinical Cancer Studies, The University of Manchester, Manchester M13 9PT, England, UK
| | - Juan-Carlos Acosta
- Edinburgh Cancer Research Centre, Medical Research Council Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh EH4 2XR, Scotland, UK
| | - Jivko Kamarashev
- Department of Dermatology, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Dennie T Frederick
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA 02114
| | - Zachary A Cooper
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Alexandre Reuben
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Jesus Gil
- Medical Research Council London Institute of Medical Sciences, London W12 0NN, England, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, England, UK
| | - Keith T Flaherty
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA 02114
| | - Jennifer A Wargo
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - William Vermi
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, 25123 Brescia, Italy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael P Smith
- Manchester Cancer Research Centre, Faculty of Biology, Medicine, and Health, School of Medical Sciences, Division of Molecular and Clinical Cancer Studies, The University of Manchester, Manchester M13 9PT, England, UK
| | - Claudia Wellbrock
- Manchester Cancer Research Centre, Faculty of Biology, Medicine, and Health, School of Medical Sciences, Division of Molecular and Clinical Cancer Studies, The University of Manchester, Manchester M13 9PT, England, UK
| | - Adam Hurlstone
- Manchester Cancer Research Centre, Faculty of Biology, Medicine, and Health, School of Medical Sciences, Division of Molecular and Clinical Cancer Studies, The University of Manchester, Manchester M13 9PT, England, UK
| |
Collapse
|
50
|
The Slow Cycling Phenotype: A Growing Problem for Treatment Resistance in Melanoma. Mol Cancer Ther 2017; 16:1002-1009. [DOI: 10.1158/1535-7163.mct-16-0535] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/27/2016] [Accepted: 01/20/2017] [Indexed: 11/16/2022]
|