1
|
Gao Y, Zheng H. Role of mitochondria and potential of mitochondria-targeted therapy in BRAF mutant cancer: A review. Crit Rev Oncol Hematol 2024; 203:104484. [PMID: 39197669 DOI: 10.1016/j.critrevonc.2024.104484] [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: 05/23/2024] [Revised: 07/28/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024] Open
Abstract
The classical mitogen-activated protein kinase (MAPK) signaling pathway, the Ras/Raf/MEK (mitogen-activated protein kinase/ERK kinase)/ERK protein kinase cascade, is a conserved cascade that regulates cell growth, differentiation, and proliferation. The significance of BRAF in cancer was established with the discovery of cancer-activating mutations in BRAF in several human tumors in 2002. Currently, BRAF is recognized as a driver mutation that affects cancer phenotypes in different ways, making it an important therapeutic target for cancer. BRAF-selective inhibitors have shown promise in clinical trials involving patients with metastatic melanoma. However, resistance mechanisms to BRAF inhibitors therapy have resulted in short-lived therapeutic responses. Further in-depth research is imperative to explore resistance mechanisms that oppose the effectiveness of BRAF inhibitors. Metabolic reprogramming has emerging role in BRAF-mutant cancers. In particular, mitochondrial metabolism and its closely related signaling pathways mediated by mitochondria have become recognized as potential new targets for treating BRAF-mutant cancers. This review, examines the progress in understanding BRAF mutations in cancer, the clinicopathological correlation of BRAF inhibitors, and recent advances in mitochondrial metabolism, mitochondrial dynamics and mitochondrial mediated death in BRAF-mutant cancer. This review will inform future cancer research and lay the foundation for novel treatment combinations of BRAF-mutant cancers.
Collapse
Affiliation(s)
- Yanyan Gao
- Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hua Zheng
- Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
2
|
Loftus AW, Zarei M, Kakish H, Hajihassani O, Hue JJ, Boutros C, Graor HJ, Nakazzi F, Bahlibi T, Winter JM, Rothermel LD. Therapeutic implications of the metabolic changes associated with BRAF inhibition in melanoma. Cancer Treat Rev 2024; 129:102795. [PMID: 38972133 PMCID: PMC11361048 DOI: 10.1016/j.ctrv.2024.102795] [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: 04/28/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/09/2024]
Abstract
Melanoma metabolism can be reprogrammed by activating BRAF mutations. These mutations are present in up to 50% of cutaneous melanomas, with the most common being V600E. BRAF mutations augment glycolysis to promote macromolecular synthesis and proliferation. Prior to the development of targeted anti-BRAF therapies, these mutations were associated with accelerated clinical disease in the metastatic setting. Combination BRAF and MEK inhibition is a first line treatment option for locally advanced or metastatic melanoma harboring targetable BRAF mutations. This therapy shows excellent response rates but these responses are not durable, with almost all patients developing resistance. When BRAF mutated melanoma cells are inhibited with targeted therapies the metabolism of those cells also changes. These cells rely less on glycolysis for energy production, and instead shift to a mitochondrial phenotype with upregulated TCA cycle activity and oxidative phosphorylation. An increased dependence on glutamine utilization is exhibited to support TCA cycle substrates in this metabolic rewiring of BRAF mutated melanoma. Herein we describe the relevant core metabolic pathways modulated by BRAF inhibition. These adaptive pathways represent vulnerabilities that could be targeted to overcome resistance to BRAF inhibitors. This review evaluates current and future therapeutic strategies that target metabolic reprogramming in melanoma cells, particularly in response to BRAF inhibition.
Collapse
Affiliation(s)
- Alexander W Loftus
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - Mehrdad Zarei
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Hanna Kakish
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - Omid Hajihassani
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Jonathan J Hue
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - Christina Boutros
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - Hallie J Graor
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Faith Nakazzi
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Tsegaw Bahlibi
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Jordan M Winter
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, 11100 Euclid Ave., Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Luke D Rothermel
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, 11100 Euclid Ave., Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
3
|
Uslu C, Kapan E, Lyakhovich A. Cancer resistance and metastasis are maintained through oxidative phosphorylation. Cancer Lett 2024; 587:216705. [PMID: 38373691 DOI: 10.1016/j.canlet.2024.216705] [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: 10/24/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/21/2024]
Abstract
Malignant tumors have increased energy requirements due to growth, differentiation or response to stress. A significant number of studies in recent years have described upregulation of mitochondrial genes responsible for oxidative phosphorylation (OXPHOS) in some tumors. Although OXPHOS is replaced by glycolysis in some tumors (Warburg effect), both processes can occur simultaneously during the evolution of the same malignancies. In particular, chemoresistant and/or cancer stem cells appear to find a way to activate OXPHOS and metastasize. In this paper, we discuss recent work showing upregulation of OXPHOS in chemoresistant tumors and cell models. In addition, we show an inverse correlation of OXPHOS gene expression with the survival time of cancer patients after chemotherapy and discuss combination therapies for resistant tumors.
Collapse
Affiliation(s)
- Cemile Uslu
- Sabanci University, Molecular Biology, Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Turkey
| | - Eda Kapan
- Sabanci University, Molecular Biology, Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Turkey
| | - Alex Lyakhovich
- Sabanci University, Molecular Biology, Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Turkey.
| |
Collapse
|
4
|
Yang Y, An Y, Ren M, Wang H, Bai J, Du W, Kong D. The mechanisms of action of mitochondrial targeting agents in cancer: inhibiting oxidative phosphorylation and inducing apoptosis. Front Pharmacol 2023; 14:1243613. [PMID: 37954849 PMCID: PMC10635426 DOI: 10.3389/fphar.2023.1243613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/12/2023] [Indexed: 11/14/2023] Open
Abstract
The tumor microenvironment affects the structure and metabolic function of mitochondria in tumor cells. This process involves changes in metabolic activity, an increase in the amount of reactive oxygen species (ROS) in tumor cells compared to normal cells, the production of more intracellular free radicals, and the activation of oxidative pathways. From a practical perspective, it is advantageous to develop drugs that target mitochondria for the treatment of malignant tumors. Such drugs can enhance the selectivity of treatments for specific cell groups, minimize toxic effects on normal tissues, and improve combinational treatments. Mitochondrial targeting agents typically rely on small molecule medications (such as synthetic small molecules agents, active ingredients of plants, mitochondrial inhibitors or autophagy inhibitors, and others), modified mitochondrial delivery system agents (such as lipophilic cation modification or combining other molecules to form targeted mitochondrial agents), and a few mitochondrial complex inhibitors. This article will review these compounds in three main areas: oxidative phosphorylation (OXPHOS), changes in ROS levels, and endogenous oxidative and apoptotic processes.
Collapse
Affiliation(s)
- Yi Yang
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yahui An
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Mingli Ren
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haijiao Wang
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Bai
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenli Du
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Dezhi Kong
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
5
|
Song L, Wei X, Zhang X, Lu Y. Combining single-cell and transcriptomic analysis revealed the immunomodulatory effect of GOT2 on a glutamine-dependent manner in cutaneous melanoma. Front Pharmacol 2023; 14:1241454. [PMID: 37693904 PMCID: PMC10483140 DOI: 10.3389/fphar.2023.1241454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023] Open
Abstract
Background: Reprogramming in glutamine metabolism is a hallmark of cancers, while its role in cutaneous melanoma has not been studied at great length. Methods: Here, we constructed a glutamine metabolism-related prognostic signature in cutaneous melanoma with a variety of bioinformatics methods according to the glutamine metabolism regulatory molecules. Moreover, experimental verification was carried out for the key gene. Results: We have identified two subgroups of cutaneous melanoma patients, each with different prognoses, immune characteristics, and genetic mutations. GOT2 was the most concerned key gene among the model genes. We verified its role in promoting tumor cell proliferation by CCK-8 and clone formation assays. Conclusion: Our study cast new light on the prognosis of cutaneous melanoma, and the internal mechanism regulating glutamine metabolism of GOT2 may provide a new avenue for treating the cutaneous melanoma disease precisely.
Collapse
Affiliation(s)
- Lebin Song
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiyi Wei
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xi Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Lu
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
6
|
Gopalkrishnan A, Wang N, Cruz-Rangel S, Kassab AY, Shiva S, Kurukulasuriya C, Monga SP, DeBerardinis RJ, Kiselyov K, Duvvuri U. Lysosomal mitochondrial interaction promotes tumor growth in squamous cell carcinoma of the head and neck. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.25.546311. [PMID: 37425842 PMCID: PMC10326999 DOI: 10.1101/2023.06.25.546311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Tumor growth and proliferation are regulated by numerous mechanisms. Communication between intracellular organelles has recently been shown to regulate cellular proliferation and fitness. The way lysosomes and mitochondria communicate with each other (lysosomal/mitochondrial interaction) is emerging as a major determinant of tumor proliferation and growth. About 30% of squamous carcinomas (including squamous cell carcinoma of the head and neck, SCCHN) overexpress TMEM16A, a calcium-activated chloride channel, which promotes cellular growth and negatively correlates with patient survival. TMEM16A has recently been shown to drive lysosomal biogenesis, but its impact on mitochondrial function is unclear. Here, we show that (1) patients with high TMEM16A SCCHN display increased mitochondrial content specifically complex I; (2) In vitro and in vivo models uniquely depend on mitochondrial complex I activity for growth and survival; (3) β-catenin/NRF2 signaling is a critical linchpin that drives mitochondrial biogenesis, and (4) mitochondrial complex I and lysosomal function are codependent for proliferation. Taken together, our data demonstrate that LMI drives tumor proliferation and facilitates a functional interaction between lysosomes and mitochondria. Therefore, inhibition of LMI may serve as a therapeutic strategy for patients with SCCHN.
Collapse
|
7
|
Bai R, Cui J. Mitochondrial immune regulation and anti-tumor immunotherapy strategies targeting mitochondria. Cancer Lett 2023; 564:216223. [PMID: 37172686 DOI: 10.1016/j.canlet.2023.216223] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/25/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Cancer cells adapt to increasing energy and biosynthetic demands by reprogramming their metabolic pathways. Mitochondria are important organelles for the metabolic reprogramming of tumor cells. In addition to supplying energy, they play crucial roles in the survival, immune evasion, tumor progression, and treatment resistance of the hypoxic tumor microenvironment (TME) in cancer cells. With the development of the life sciences, scientists have gained an in-depth understanding of immunity, metabolism, and cancer, and numerous studies have emphasized that mitochondria are essential for tumor immune escape and the regulation of immune cell metabolism and activation. Moreover, recent evidence suggests that targeting the mitochondria-related pathway with anticancer drugs can initiate the killing of cancer cells by increasing the ability of cancer cells to be recognized by immune cells, tumor antigen presentation ability, and the anti-tumor function of immune cells. This review discusses the effects of mitochondrial morphology and function on the phenotype and function of immune cells under normal and TME conditions, the effects of mitochondrial changes in tumors and microenvironments on tumor immune escape and immune cell function, and finally focuses on the recent research progress and future challenges of novel anti-tumor immunotherapy strategies targeting mitochondria.
Collapse
Affiliation(s)
- Rilan Bai
- Cancer Center, the First Hospital of Jilin University, Changchun, 130021, China
| | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
8
|
He P, Feng J, Xia X, Sun Y, He J, Guan T, Peng Y, Zhang X, Liu M, Pang X, Chen Y. Discovery of a Potent and Oral Available Complex I OXPHOS Inhibitor That Abrogates Tumor Growth and Circumvents MEKi Resistance. J Med Chem 2023; 66:6047-6069. [PMID: 37130350 DOI: 10.1021/acs.jmedchem.2c01844] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Targeting oxidative phosphorylation (OXPHOS) has emerged as a promising therapeutic strategy for cancer therapy. Here, we discovered a 1H-1,2,3-triazole derivative HP661 as a highly potent and orally available OXPHOS inhibitor that effectively blocked the activity of mitochondrial complex I. HP661 specifically compromised the mitochondrial oxygen consumption of high-OXPHOS lung cancer cells but not that of low-OXPHOS lung cancer cells or normal cells in the low nanomolar range. Notably, mitogen-activated protein kinase kinase (MEK) inhibitor (trametinib)-resistant lung cancer cells with high levels of OXPHOS also showed marked sensitivity to HP661, as indicated by decreased clonogenic growth and increased cell apoptosis upon treatment. In a mouse model of high-OXPHOS lung cancer, HP661 treatment not only significantly suppressed tumor growth but also augmented the therapeutic efficacy of trametinib by impairing tumor mitochondrial respiration. In summary, we identified HP661 as a highly effective OXPHOS inhibitor to abrogate the growth of high OXPHOS-dependent tumors and conquer high OXPHOS-mediated drug resistance.
Collapse
Affiliation(s)
- Peng He
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Juanjuan Feng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
- Joint Center for Translational Medicine, Southern Medical University Affiliated Fengxian Hospital, Shanghai 201499, China
| | - Xinting Xia
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yue Sun
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jia He
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Tian Guan
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yangrui Peng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xueli Zhang
- Joint Center for Translational Medicine, Southern Medical University Affiliated Fengxian Hospital, Shanghai 201499, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiufeng Pang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
9
|
Cadassou O, Petter Jordheim L. OXPHOS inhibitors, metabolism and targeted therapies in cancer. Biochem Pharmacol 2023; 211:115531. [PMID: 37019188 DOI: 10.1016/j.bcp.2023.115531] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023]
Abstract
More and more studies highlight the complex metabolic characteristics and plasticity of cancer cells. To address these specificities and explore the associated vulnerabilities, new metabolism-targeting therapeutic strategies are being developed. It is more and more accepted that cancer cells do not produce their energy only from aerobic glycolysis, as some subtypes strongly rely on mitochondrial respiration (OXPHOS). This review focuses on classical and promising OXPHOS inhibitors (OXPHOSi), unravelling their interest and modes of actions in cancer, particularly in combination with other strategies. Indeed, in monotherapy, OXPHOSi display limited efficiency as they mostly trigger cell death in cancer cell subtypes that strongly depend on mitochondrial respiration and are not able to shift to other metabolic pathways to produce energy. Nevertheless, they remain very interesting in combination with conventional therapeutic strategies such as chemotherapy and radiotherapy, increasing their anti-tumoral actions. In addition, OXPHOSi can be included in even more innovative strategies such as combinations with other metabolic drugs or immunotherapies.
Collapse
|
10
|
Chouhan S, Sawant M, Weimholt C, Luo J, Sprung RW, Terrado M, Mueller DM, Earp HS, Mahajan NP. TNK2/ACK1-mediated phosphorylation of ATP5F1A (ATP synthase F1 subunit alpha) selectively augments survival of prostate cancer while engendering mitochondrial vulnerability. Autophagy 2023; 19:1000-1025. [PMID: 35895804 PMCID: PMC9980697 DOI: 10.1080/15548627.2022.2103961] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 11/02/2022] Open
Abstract
The challenge of rapid macromolecular synthesis enforces the energy-hungry cancer cell mitochondria to switch their metabolic phenotypes, accomplished by activation of oncogenic tyrosine kinases. Precisely how kinase activity is directly exploited by cancer cell mitochondria to meet high-energy demand, remains to be deciphered. Here we show that a non-receptor tyrosine kinase, TNK2/ACK1 (tyrosine kinase non receptor 2), phosphorylated ATP5F1A (ATP synthase F1 subunit alpha) at Tyr243 and Tyr246 (Tyr200 and 203 in the mature protein, respectively) that not only increased the stability of complex V, but also increased mitochondrial energy output in cancer cells. Further, phospho-ATP5F1A (p-Y-ATP5F1A) prevented its binding to its physiological inhibitor, ATP5IF1 (ATP synthase inhibitory factor subunit 1), causing sustained mitochondrial activity to promote cancer cell growth. TNK2 inhibitor, (R)-9b reversed this process and induced mitophagy-based autophagy to mitigate prostate tumor growth while sparing normal prostate cells. Further, depletion of p-Y-ATP5F1A was needed for (R)-9b-mediated mitophagic response and tumor growth. Moreover, Tnk2 transgenic mice displayed increased p-Y-ATP5F1A and loss of mitophagy and exhibited formation of prostatic intraepithelial neoplasia (PINs). Consistent with these data, a marked increase in p-Y-ATP5F1A was seen as prostate cancer progressed to the malignant stage. Overall, this study uncovered the molecular intricacy of tyrosine kinase-mediated mitochondrial energy regulation as a distinct cancer cell mitochondrial vulnerability and provided evidence that TNK2 inhibitors can act as "mitocans" to induce cancer-specific mitophagy.Abbreviations: ATP5F1A: ATP synthase F1 subunit alpha; ATP5IF1: ATP synthase inhibitory factor subunit 1; CRPC: castration-resistant prostate cancer; DNM1L: dynamin 1 like; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; Mdivi-1: mitochondrial division inhibitor 1; Mut-ATP5F1A: Y243,246A mutant of ATP5F1A; OXPHOS: oxidative phosphorylation; PC: prostate cancer; PINK1: PTEN induced kinase 1; p-Y-ATP5F1A: phosphorylated tyrosine 243 and 246 on ATP5F1A; TNK2/ACK1: tyrosine kinase non receptor 2; Ub: ubiquitin; WT: wild type.
Collapse
Affiliation(s)
- Surbhi Chouhan
- Department of Surgery, Cancer Research Building, St. Louis, MO, USA
- Division of Urologic Surgery Washington University, St. Louis, MO, USA
| | - Mithila Sawant
- Department of Surgery, Cancer Research Building, St. Louis, MO, USA
- Division of Urologic Surgery Washington University, St. Louis, MO, USA
| | - Cody Weimholt
- Department of Pathology & Immunology Washington University, St. Louis, MO, USA
| | - Jingqin Luo
- Division of Public Health Sciences, Washington University, St. Louis, MO, USA
| | - Robert W. Sprung
- Department of Surgery, Cancer Research Building, St. Louis, MO, USA
| | - Mailyn Terrado
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA
| | - David M. Mueller
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA
| | - H. Shelton Earp
- Lineberger Comprehensive Cancer Center, Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Nupam P. Mahajan
- Department of Surgery, Cancer Research Building, St. Louis, MO, USA
- Division of Urologic Surgery Washington University, St. Louis, MO, USA
- Siteman Cancer Center Washington University, St. Louis, MO, USA
| |
Collapse
|
11
|
Liu Y, Sun Y, Guo Y, Shi X, Chen X, Feng W, Wu LL, Zhang J, Yu S, Wang Y, Shi Y. An Overview: The Diversified Role of Mitochondria in Cancer Metabolism. Int J Biol Sci 2023; 19:897-915. [PMID: 36778129 PMCID: PMC9910000 DOI: 10.7150/ijbs.81609] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/04/2023] [Indexed: 02/04/2023] Open
Abstract
Mitochondria are intracellular organelles involved in energy production, cell metabolism and cell signaling. They are essential not only in the process of ATP synthesis, lipid metabolism and nucleic acid metabolism, but also in tumor development and metastasis. Mutations in mtDNA are commonly found in cancer cells to promote the rewiring of bioenergetics and biosynthesis, various metabolites especially oncometabolites in mitochondria regulate tumor metabolism and progression. And mutation of enzymes in the TCA cycle leads to the unusual accumulation of certain metabolites and oncometabolites. Mitochondria have been demonstrated as the target for cancer treatment. Cancer cells rely on two main energy resources: oxidative phosphorylation (OXPHOS) and glycolysis. By manipulating OXPHOS genes or adjusting the metabolites production in mitochondria, tumor growth can be restrained. For example, enhanced complex I activity increases NAD+/NADH to prevent metastasis and progression of cancers. In this review, we discussed mitochondrial function in cancer cell metabolism and specially explored the unique role of mitochondria in cancer stem cells and the tumor microenvironment. Targeting the OXPHOS pathway and mitochondria-related metabolism emerging as a potential therapeutic strategy for various cancers.
Collapse
Affiliation(s)
- Yu'e Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yihong Sun
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yadong Guo
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoyun Shi
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Xi Chen
- Xi Chen, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Wenfeng Feng
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Lei-Lei Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 200433, Shanghai, China
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 39216, Jackson, Mississippi, USA
| | - Shibo Yu
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yufeng Shi
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China.,Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai 200092, China
| |
Collapse
|
12
|
Wu C, Liu Y, Liu W, Zou T, Lu S, Zhu C, He L, Chen J, Fang L, Zou L, Wang P, Fan L, Wang H, You H, Chen J, Fang J, Jiang C, Shi Y. NNMT-DNMT1 Axis is Essential for Maintaining Cancer Cell Sensitivity to Oxidative Phosphorylation Inhibition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 10:e2202642. [PMID: 36382559 PMCID: PMC9811437 DOI: 10.1002/advs.202202642] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 10/06/2022] [Indexed: 06/16/2023]
Abstract
Lacking a clear understanding of the molecular mechanism determining cancer cell sensitivity to oxidative phosphorylation (OXPHOS) inhibition limits the development of OXPHOS-targeting cancer treatment. Here, cancer cell lines sensitive or resistant to OXPHOS inhibition are identified by screening. OXPHOS inhibition-sensitive cancer cells possess increased OXPHOS activity and silenced nicotinamide N-methyltransferase (NNMT) expression. NNMT expression negatively correlates with OXPHOS inhibition sensitivity and functionally downregulates the intracellular levels of S-adenosyl methionine (SAM). Expression of DNA methyltransferase 1 (DNMT1), a SAM consumer, positively correlates with OXPHOS inhibition sensitivity. NNMT overexpression and DNMT1 inhibition render OXPHOS inhibition-sensitive cancer cells resistant. Importantly, treatments of OXPHOS inhibitors (Gboxin and Berberine) hamper the growth of mouse tumor xenografts by OXPHOS inhibition sensitive but not resistant cancer cells. What's more, the retrospective study of 62 tumor samples from a clinical trial demonstrates that administration of Berberine reduces the tumor recurrence rate of NNMTlow /DNMT1high but not NNMThigh /DNMT1low colorectal adenomas (CRAs). These results thus reveal a critical role of the NNMT-DNMT1 axis in determining cancer cell reliance on mitochondrial OXPHOS and suggest that NNMT and DNMT1 are faithful biomarkers for OXPHOS-targeting cancer therapies.
Collapse
Affiliation(s)
- Changqing Wu
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Yu'e Liu
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Wenju Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Tianhui Zou
- State Key Laboratory for Oncogenes and Related GenesKey Laboratory of Gastroenterology & HepatologyMinistry of HealthDivision of Gastroenterology and HepatologyShanghai Institute of Digestive DiseaseRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
| | - Shaojuan Lu
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Chengjie Zhu
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Le He
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Jie Chen
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Lan Fang
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Lin Zou
- Clinical Research UnitChildren's Hospital of Shanghai Jiaotong UniversityShanghai200062China
| | - Ping Wang
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Lihong Fan
- Department of Respiratory MedicineShanghai Tenth People's HospitalTongji University School of MedicineShanghai200072China
| | - Hongxiang Wang
- Department of NeurosurgeryChanghai HospitalNaval Medical UniversityNO.168 Changhai RoadShanghai200433China
| | - Han You
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamen361005China
| | - Juxiang Chen
- Department of NeurosurgeryChanghai HospitalNaval Medical UniversityNO.168 Changhai RoadShanghai200433China
| | - Jing‐Yuan Fang
- State Key Laboratory for Oncogenes and Related GenesKey Laboratory of Gastroenterology & HepatologyMinistry of HealthDivision of Gastroenterology and HepatologyShanghai Institute of Digestive DiseaseRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
| | - Cizhong Jiang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Yufeng Shi
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
- Clinical Center for Brain and Spinal Cord ResearchTongji UniversityShanghai200092China
| |
Collapse
|
13
|
Karz A, Dimitrova M, Kleffman K, Alvarez-Breckenridge C, Atkins MB, Boire A, Bosenberg M, Brastianos P, Cahill DP, Chen Q, Ferguson S, Forsyth P, Glitza Oliva IC, Goldberg SB, Holmen SL, Knisely JPS, Merlino G, Nguyen DX, Pacold ME, Perez-Guijarro E, Smalley KSM, Tawbi HA, Wen PY, Davies MA, Kluger HM, Mehnert JM, Hernando E. Melanoma central nervous system metastases: An update to approaches, challenges, and opportunities. Pigment Cell Melanoma Res 2022; 35:554-572. [PMID: 35912544 PMCID: PMC10171356 DOI: 10.1111/pcmr.13059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/29/2022] [Indexed: 01/27/2023]
Abstract
Brain metastases are the most common brain malignancy. This review discusses the studies presented at the third annual meeting of the Melanoma Research Foundation in the context of other recent reports on the biology and treatment of melanoma brain metastases (MBM). Although symptomatic MBM patients were historically excluded from immunotherapy trials, efforts from clinicians and patient advocates have resulted in more inclusive and even dedicated clinical trials for MBM patients. The results of checkpoint inhibitor trials were discussed in conversation with current standards of care for MBM patients, including steroids, radiotherapy, and targeted therapy. Advances in the basic scientific understanding of MBM, including the role of astrocytes and metabolic adaptations to the brain microenvironment, are exposing new vulnerabilities which could be exploited for therapeutic purposes. Technical advances including single-cell omics and multiplex imaging are expanding our understanding of the MBM ecosystem and its response to therapy. This unprecedented level of spatial and temporal resolution is expected to dramatically advance the field in the coming years and render novel treatment approaches that might improve MBM patient outcomes.
Collapse
Affiliation(s)
- Alcida Karz
- Department of Pathology, NYU Grossman School of Medicine, New York, USA.,Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, USA
| | - Maya Dimitrova
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, USA.,Department of Medicine, NYU Grossman School of Medicine, New York, USA
| | - Kevin Kleffman
- Department of Pathology, NYU Grossman School of Medicine, New York, USA.,Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, USA
| | | | - Michael B Atkins
- Georgetown-Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Adrienne Boire
- Human Oncology and Pathogenesis Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Marcus Bosenberg
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research NCI, NIH, USA
| | - Priscilla Brastianos
- MGH Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel P Cahill
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Qing Chen
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Sherise Ferguson
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Peter Forsyth
- Department of Neuro-Oncology and Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Isabella C Glitza Oliva
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sarah B Goldberg
- Department of Medicine (Medical Oncology), Yale School of Medicine, New Haven, Connecticut, USA
| | - Sheri L Holmen
- Huntsman Cancer Institute and Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| | - Jonathan P S Knisely
- Meyer Cancer Center and Department of Radiation Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research NCI, NIH, USA
| | - Don X Nguyen
- Department of Pathology, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michael E Pacold
- Department of Radiation Oncology, NYU Langone Health and NYU Grossman School of Medicine, New York, New York, USA
| | - Eva Perez-Guijarro
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research NCI, NIH, USA
| | - Keiran S M Smalley
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, United States, Boston, Massachusetts, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Harriet M Kluger
- Department of Medicine (Medical Oncology), Yale School of Medicine, New Haven, Connecticut, USA
| | - Janice M Mehnert
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, USA.,Department of Medicine, NYU Grossman School of Medicine, New York, USA
| | - Eva Hernando
- Department of Pathology, NYU Grossman School of Medicine, New York, USA.,Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, USA
| |
Collapse
|
14
|
Beck TC, Arhontoulis DC, Morningstar JE, Hyams N, Stoddard A, Springs K, Mukherjee R, Helke K, Guo L, Moore K, Gensemer C, Biggs R, Petrucci T, Kwon J, Stayer K, Koren N, Harvey A, Holman H, Dunne J, Fulmer D, Vohra A, Mai L, Dooley S, Weninger J, Vaena S, Romeo M, Muise-Helmericks RC, Mei Y, Norris RA. Cellular and Molecular Mechanisms of MEK1 Inhibitor-Induced Cardiotoxicity. JACC CardioOncol 2022; 4:535-548. [PMID: 36444237 PMCID: PMC9700254 DOI: 10.1016/j.jaccao.2022.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Trametinib is a MEK1 (mitogen-activated extracellular signal-related kinase kinase 1) inhibitor used in the treatment of BRAF (rapid accelerated fibrosarcoma B-type)-mutated metastatic melanoma. Roughly 11% of patients develop cardiomyopathy following long-term trametinib exposure. Although described clinically, the molecular landscape of trametinib cardiotoxicity has not been characterized. Objectives The aim of this study was to test the hypothesis that trametinib promotes widespread transcriptomic and cellular changes consistent with oxidative stress and impairs cardiac function. Methods Mice were treated with trametinib (1 mg/kg/d). Echocardiography was performed pre- and post-treatment. Gross, histopathologic, and biochemical assessments were performed to probe for molecular and cellular changes. Human cardiac organoids were used as an in vitro measurement of cardiotoxicity and recovery. Results Long-term administration of trametinib was associated with significant reductions in survival and left ventricular ejection fraction. Histologic analyses of the heart revealed myocardial vacuolization and calcification in 28% of animals. Bulk RNA sequencing identified 435 differentially expressed genes and 116 differential signaling pathways following trametinib treatment. Upstream gene analysis predicted interleukin-6 as a regulator of 17 relevant differentially expressed genes, suggestive of PI3K/AKT and JAK/STAT activation, which was subsequently validated. Trametinib hearts displayed elevated markers of oxidative stress, myofibrillar degeneration, an 11-fold down-regulation of the apelin receptor, and connexin-43 mislocalization. To confirm the direct cardiotoxic effects of trametinib, human cardiac organoids were treated for 6 days, followed by a 6-day media-only recovery. Trametinib-treated organoids exhibited reductions in diameter and contractility, followed by partial recovery with removal of treatment. Conclusions These data describe pathologic changes observed in trametinib cardiotoxicity, supporting the exploration of drug holidays and alternative pharmacologic strategies for disease prevention.
Collapse
Affiliation(s)
- Tyler C. Beck
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Dimitrios C. Arhontoulis
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Jordan E. Morningstar
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Nathaniel Hyams
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Andrew Stoddard
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kendra Springs
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Rupak Mukherjee
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kris Helke
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lilong Guo
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kelsey Moore
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Cortney Gensemer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Rachel Biggs
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Taylor Petrucci
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jennie Kwon
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kristina Stayer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Natalie Koren
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Andrew Harvey
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Heather Holman
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jaclyn Dunne
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Diana Fulmer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ayesha Vohra
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Le Mai
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sarah Dooley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Julianna Weninger
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Silvia Vaena
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Martin Romeo
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Robin C. Muise-Helmericks
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ying Mei
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Russell A. Norris
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
15
|
Cheng X, Dai C, Wen Y, Wang X, Bo X, He S, Peng S. NeRD: a multichannel neural network to predict cellular response of drugs by integrating multidimensional data. BMC Med 2022; 20:368. [PMID: 36244991 PMCID: PMC9575288 DOI: 10.1186/s12916-022-02549-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Considering the heterogeneity of tumors, it is a key issue in precision medicine to predict the drug response of each individual. The accumulation of various types of drug informatics and multi-omics data facilitates the development of efficient models for drug response prediction. However, the selection of high-quality data sources and the design of suitable methods remain a challenge. METHODS In this paper, we design NeRD, a multidimensional data integration model based on the PRISM drug response database, to predict the cellular response of drugs. Four feature extractors, including drug structure extractor (DSE), molecular fingerprint extractor (MFE), miRNA expression extractor (mEE), and copy number extractor (CNE), are designed for different types and dimensions of data. A fully connected network is used to fuse all features and make predictions. RESULTS Experimental results demonstrate the effective integration of the global and local structural features of drugs, as well as the features of cell lines from different omics data. For all metrics tested on the PRISM database, NeRD surpassed previous approaches. We also verified that NeRD has strong reliability in the prediction results of new samples. Moreover, unlike other algorithms, when the amount of training data was reduced, NeRD maintained stable performance. CONCLUSIONS NeRD's feature fusion provides a new idea for drug response prediction, which is of great significance for precise cancer treatment.
Collapse
Affiliation(s)
- Xiaoxiao Cheng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Chong Dai
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.,Department of Biotechnology, Beijing Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Yuqi Wen
- Department of Biotechnology, Beijing Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Xiaoqi Wang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Xiaochen Bo
- Department of Biotechnology, Beijing Institute of Health Service and Transfusion Medicine, Beijing, China.
| | - Song He
- Department of Biotechnology, Beijing Institute of Health Service and Transfusion Medicine, Beijing, China.
| | - Shaoliang Peng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China. .,The State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China.
| |
Collapse
|
16
|
Feng J, Lian Z, Xia X, Lu Y, Hu K, Zhang Y, Liu Y, Hu L, Yuan K, Sun Z, Pang X. Targeting metabolic vulnerability in mitochondria conquers MEK inhibitor resistance in KRAS-mutant lung cancer. Acta Pharm Sin B 2022; 13:1145-1163. [PMID: 36970205 PMCID: PMC10031260 DOI: 10.1016/j.apsb.2022.10.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/18/2022] [Accepted: 09/23/2022] [Indexed: 11/01/2022] Open
Abstract
MEK is a canonical effector of mutant KRAS; however, MEK inhibitors fail to yield satisfactory clinical outcomes in KRAS-mutant cancers. Here, we identified mitochondrial oxidative phosphorylation (OXPHOS) induction as a profound metabolic alteration to confer KRAS-mutant non-small cell lung cancer (NSCLC) resistance to the clinical MEK inhibitor trametinib. Metabolic flux analysis demonstrated that pyruvate metabolism and fatty acid oxidation were markedly enhanced and coordinately powered the OXPHOS system in resistant cells after trametinib treatment, satisfying their energy demand and protecting them from apoptosis. As molecular events in this process, the pyruvate dehydrogenase complex (PDHc) and carnitine palmitoyl transferase IA (CPTIA), two rate-limiting enzymes that control the metabolic flux of pyruvate and palmitic acid to mitochondrial respiration were activated through phosphorylation and transcriptional regulation. Importantly, the co-administration of trametinib and IACS-010759, a clinical mitochondrial complex I inhibitor that blocks OXPHOS, significantly impeded tumor growth and prolonged mouse survival. Overall, our findings reveal that MEK inhibitor therapy creates a metabolic vulnerability in the mitochondria and further develop an effective combinatorial strategy to circumvent MEK inhibitors resistance in KRAS-driven NSCLC.
Collapse
|
17
|
Gallez B. The Role of Imaging Biomarkers to Guide Pharmacological Interventions Targeting Tumor Hypoxia. Front Pharmacol 2022; 13:853568. [PMID: 35910347 PMCID: PMC9335493 DOI: 10.3389/fphar.2022.853568] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is a common feature of solid tumors that contributes to angiogenesis, invasiveness, metastasis, altered metabolism and genomic instability. As hypoxia is a major actor in tumor progression and resistance to radiotherapy, chemotherapy and immunotherapy, multiple approaches have emerged to target tumor hypoxia. It includes among others pharmacological interventions designed to alleviate tumor hypoxia at the time of radiation therapy, prodrugs that are selectively activated in hypoxic cells or inhibitors of molecular targets involved in hypoxic cell survival (i.e., hypoxia inducible factors HIFs, PI3K/AKT/mTOR pathway, unfolded protein response). While numerous strategies were successful in pre-clinical models, their translation in the clinical practice has been disappointing so far. This therapeutic failure often results from the absence of appropriate stratification of patients that could benefit from targeted interventions. Companion diagnostics may help at different levels of the research and development, and in matching a patient to a specific intervention targeting hypoxia. In this review, we discuss the relative merits of the existing hypoxia biomarkers, their current status and the challenges for their future validation as companion diagnostics adapted to the nature of the intervention.
Collapse
Affiliation(s)
- Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
18
|
Arast Y, Heidary M, Tanbakousazan F, Behnamipour S, Vazirizadeh A, Pourahmad J. Selective Toxicity of Cistanche tubulosa Root Extract on Cancerous Skin Mitochondria isolated from animal Model of Melanoma. Cutan Ocul Toxicol 2022; 41:243-249. [PMID: 35796072 DOI: 10.1080/15569527.2022.2096628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
INTRODUCTION As a major public health issue, the skin cancer is a leading reason of death and has resulted in significant financial and human losses globally. Numerous environmental and internal variables may both drive and exacerbate the pathophysiology of the skin cancer. Marine herbs and animals, including marine sponges, cucumbers, and squirts, have been shown to have cytotoxic consequences on cancerous cells in prior research. PURPOSE melanoma mitochondria obtained from skin of melanoma animal model are studied in this research to see whether extracts from Cistanche tubulosa, a plant endemic to the northern coasts of the Persian Gulf, has a cytotoxic impact on them. MATERIAL AND METHOD In this study, the mitochondria isolated from melanoma cells via differential centrifugation and treated with various concentrations (1250, 2500 and 5000 µg/ml) of methanolic extract of C. tubulosa. Then MTT, ROS, MMP decline, mitochondrial swelling, cytochrome c release and flow cytometry assays were performed on them. RESULTS The results of MTT assay showed that the IC50 of C. tubulosa extract is 2500 μg/ml and C. tubulosa extract induced a selectively significant (P < 0.05) concentration-dependent decrease in the SDH activity in cancerous skin mitochondria. The ROS results also showed that all concentrations of C. tubulosa extracts significantly increased ROS production, MMP decline and the release of cytochrome c in cancer groups mitochondria. The swelling of mitochondria isolated from the cancer group was significantly increased compared to the control group. In addition, the results of apoptosis assay showed that addition of root extract of Cistanche tubulosa on melanoma cells increased apoptosis, while it had no effect on control non tumor cells. DISCUSSION AND CONCLUSION Based on these results, the presence of potentially bioactive compounds in C. tubulosa make this Persian Gulf coastal herb a strong candidate for further molecular studies and clinical research in the field of melanoma cancer therapy.
Collapse
Affiliation(s)
- Yalda Arast
- Research center of Environmental Pollutants, Qom University of Medical Sciences, Qom, Iran
| | - Mobina Heidary
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences Tehran, Iran
| | - Farahnaz Tanbakousazan
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences Tehran, Iran
| | - Somaye Behnamipour
- Research center of Environmental Pollutants, Qom University of Medical Sciences, Qom, Iran
| | - Amir Vazirizadeh
- Persian Gulf Research Institute, Marine Biology and Fishery Sciences Department, Persian Gulf University, Bandar Abbas. Iran
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences Tehran, Iran
| |
Collapse
|
19
|
Wang J, Wu H, Zhao Q, Zou Y, Ding D, Yin H, Xu H. Aggregation-Induced Emission Photosensitizer Synergizes Photodynamic Therapy and the Inhibition of the NF-κB Signaling Pathway to Overcome Hypoxia in Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2022; 14:29613-29625. [PMID: 35729075 DOI: 10.1021/acsami.2c06063] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Triple-negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer, and TNBC patients often develop resistance to endocrine or molecular targeted therapy. Thus, a search for effective treatments is urgently required. Photodynamic therapy (PDT) has been verified to be a successful therapy for cancer. However, this treatment is oxygen-consuming, thus considerably limiting the PDT outcomes. The present study introduced a multistage drug delivery system to alleviate hypoxia and enhance PDT efficiency. Specifically, aggregation-induced emission luminogen (AIEgen) TPE-Py was first introduced to achieve PDT properties, and natural naphthohydroquinone dimer Rubioncolin C (RC), a blocker of mitochondria-associated oxidative phosphorylation (OXPHOS) and an NF-κB inhibitor, was applied to suppress the O2 consumption of OXPHOS and mitigate hypoxia thereafter. Enhanced PDT efficiency was validated by in vitro and in vivo TNBC models. In terms of the mechanism, AIEgen-based PDT synergized with RC could induce a fatal burst of reactive oxygen species (ROS) and ROS-mediated apoptosis. Moreover, this combination promoted the effectiveness of PDT by inhibiting the NF-κB signaling pathway. All of these results demonstrated that the administration system not only achieved a synergistic anti-TNBC effect but also expanded the clinical application of AIEgen-based PDT by overcoming hypoxia and inhibiting the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jia Wang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Haisi Wu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Qianqian Zhao
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Yifan Zou
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education and College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Haitao Yin
- Department of Radiotherapy, Xuzhou Central Hospital, Xuzhou 221009, P. R. China
| | - Huae Xu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
| |
Collapse
|
20
|
Mudassar F, Shen H, Cook KM, Hau E. Improving the synergistic combination of programmed death‐1/programmed death ligand‐1 blockade and radiotherapy by targeting the hypoxic tumour microenvironment. J Med Imaging Radiat Oncol 2022; 66:560-574. [PMID: 35466515 PMCID: PMC9322583 DOI: 10.1111/1754-9485.13416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/05/2022] [Accepted: 04/10/2022] [Indexed: 11/28/2022]
Abstract
Immune checkpoint inhibition with PD‐1/PD‐L1 blockade is a promising area in the field of anti‐cancer therapy. Although clinical data have revealed success of PD‐1/PD‐L1 blockade as monotherapy or in combination with CTLA‐4 or chemotherapy, the combination with radiotherapy could further boost anti‐tumour immunity and enhance clinical outcomes due to the immunostimulatory effects of radiation. However, the synergistic combination of PD‐1/PD‐L1 blockade and radiotherapy can be challenged by the complex nature of the tumour microenvironment (TME), including the presence of tumour hypoxia. Hypoxia is a major barrier to the effectiveness of both radiotherapy and PD‐1/PD‐L1 blockade immunotherapy. Thus, targeting the hypoxic TME is an attractive strategy to enhance the efficacy of the combination. Addition of compounds that directly or indirectly reduce hypoxia, to the combination of PD‐1/PD‐L1 inhibitors and radiotherapy may optimize the success of the combination and improve therapeutic outcomes. In this review, we will discuss the synergistic combination of PD‐1/PD‐L1 blockade and radiotherapy and highlight the role of hypoxic TME in impeding the success of both therapies. In addition, we will address the potential approaches for targeting tumour hypoxia and how exploiting these strategies could benefit the combination of PD‐1/PD‐L1 blockade and radiotherapy.
Collapse
Affiliation(s)
- Faiqa Mudassar
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research The Westmead Institute for Medical Research Sydney New South Wales Australia
- Sydney Medical School The University of Sydney Sydney New South Wales Australia
| | - Han Shen
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research The Westmead Institute for Medical Research Sydney New South Wales Australia
- Sydney Medical School The University of Sydney Sydney New South Wales Australia
| | - Kristina M Cook
- Sydney Medical School The University of Sydney Sydney New South Wales Australia
- Charles Perkins Centre The University of Sydney Sydney New South Wales Australia
| | - Eric Hau
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research The Westmead Institute for Medical Research Sydney New South Wales Australia
- Sydney Medical School The University of Sydney Sydney New South Wales Australia
- Department of Radiation Oncology, Crown Princess Mary Cancer Centre Westmead Hospital Sydney New South Wales Australia
- Blacktown Hematology and Cancer Centre Blacktown Hospital Sydney New South Wales Australia
| |
Collapse
|
21
|
Kalyanaraman B. Exploiting the tumor immune microenvironment and immunometabolism using mitochondria-targeted drugs: Challenges and opportunities in racial disparity and cancer outcome research. FASEB J 2022; 36:e22226. [PMID: 35233843 PMCID: PMC9242412 DOI: 10.1096/fj.202101862r] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 12/16/2022]
Abstract
Black and Hispanic cancer patients have a higher incidence of cancer mortality. Many factors (e.g., socioeconomic differences, insufficient access to healthcare) contribute to racial disparity. Emerging research implicates biological disparity in cancer outcomes. Studies show distinct differences in the tumor immune microenvironment (TIME) in Black cancer patients. Studies also have linked altered mitochondrial metabolism to changes in immune cell activation in TIME. Recent publications revealed a novel immunomodulatory role for triphenylphosphonium-based mitochondrial-targeted drugs (MTDs). These are synthetically modified, naturally occurring molecules (e.g., honokiol, magnolol, metformin) or FDA-approved small molecule drugs (e.g., atovaquone, hydroxyurea). Modifications involve conjugating the parent molecule via an alkyl linker chain to a triphenylphosphonium moiety. These modified molecules (e.g., Mito-honokiol, Mito-magnolol, Mito-metformin, Mito-atovaquone, Mito-hydroxyurea) accumulate in tumor cell mitochondria more effectively than in normal cells and inhibit mitochondrial respiration, induce reactive oxygen species, activate AMPK and redox transcription factors, and inhibit cancer cell proliferation. Besides these intrinsic effects of MTDs in redox signaling and proliferation in tumors, MTDs induced extrinsic effects in the TIME of mouse xenografts. MTD treatment inhibited tumor-suppressive immune cells, myeloid-derived suppressor cells, and regulatory T cells, and activated T cells and antitumor immune effects. One key biological disparity in Black cancer patients was related to altered mitochondrial oxidative metabolism; MTDs targeting vulnerabilities in tumor cells and the TIME may help us understand this biological disparity. Clinical trials should include an appropriate number of Black and Hispanic cancer patients and should validate the intratumoral, antihypoxic effects of MTDs with imaging.
Collapse
Affiliation(s)
- Balaraman Kalyanaraman
- Department of BiophysicsMedical College of WisconsinMilwaukeeWisconsinUSA
- Center for Disease Prevention ResearchMedical College of WisconsinMilwaukeeWisconsinUSA
| |
Collapse
|
22
|
Ghilardi C, Moreira-Barbosa C, Brunelli L, Ostano P, Panini N, Lupi M, Anastasia A, Fiordaliso F, Salio M, Formenti L, Russo M, Arrigoni E, Chiaradonna F, Chiorino G, Draetta G, Marszalek JR, Vellano CP, Pastorelli R, Bani M, Decio A, Giavazzi R. PGC1α/β Expression Predicts Therapeutic Response to Oxidative Phosphorylation Inhibition in Ovarian Cancer. Cancer Res 2022; 82:1423-1434. [PMID: 35131872 PMCID: PMC9359716 DOI: 10.1158/0008-5472.can-21-1223] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/11/2021] [Accepted: 02/02/2022] [Indexed: 01/07/2023]
Abstract
Ovarian cancer is the deadliest gynecologic cancer, and novel therapeutic options are crucial to improve overall survival. Here we provide evidence that impairment of oxidative phosphorylation (OXPHOS) can help control ovarian cancer progression, and this benefit correlates with expression of the two mitochondrial master regulators PGC1α and PGC1β. In orthotopic patient-derived ovarian cancer xenografts (OC-PDX), concomitant high expression of PGC1α and PGC1β (PGC1α/β) fostered a unique transcriptional signature, leading to increased mitochondrial abundance, enhanced tricarboxylic acid cycling, and elevated cellular respiration that ultimately conferred vulnerability to OXPHOS inhibition. Treatment with the respiratory chain complex I inhibitor IACS-010759 caused mitochondrial swelling and ATP depletion that consequently delayed malignant progression and prolonged the lifespan of high PGC1α/β-expressing OC-PDX-bearing mice. Conversely, low PGC1α/β OC-PDXs were not affected by IACS-010759, thus pinpointing a selective antitumor effect of OXPHOS inhibition. The clinical relevance of these findings was substantiated by analysis of ovarian cancer patient datasets, which showed that 25% of all cases displayed high PGC1α/β expression along with an activated mitochondrial gene program. This study endorses the use of OXPHOS inhibitors to manage ovarian cancer and identifies the high expression of both PGC1α and β as biomarkers to refine the selection of patients likely to benefit most from this therapy. SIGNIFICANCE OXPHOS inhibition in ovarian cancer can exploit the metabolic vulnerabilities conferred by high PGC1α/β expression and offers an effective approach to manage patients on the basis of PGC1α/β expression.
Collapse
Affiliation(s)
- Carmen Ghilardi
- Laboratory of Cancer Metastasis Therapeutics, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.,Corresponding Author: Carmen Ghilardi, Department of Oncology, Laboratory of Cancer Metastasis Therapeutics; Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy. Phone: 39-02-39014226; Fax: 39-02-39014734; E-mail:
| | - Catarina Moreira-Barbosa
- Laboratory of Cancer Metastasis Therapeutics, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.,Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Laura Brunelli
- Laboratory of Mass Spectrometry, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Paola Ostano
- Cancer Genomics Laboratory, Fondazione Edo ed Elvo Tempia Valenta, Biella, Italy
| | - Nicolò Panini
- Laboratory of Anticancer Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Monica Lupi
- Laboratory of Anticancer Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Alessia Anastasia
- Laboratory of Cancer Metastasis Therapeutics, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Fabio Fiordaliso
- Laboratory of Cardiovascular Clinical Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Monica Salio
- Laboratory of Cardiovascular Clinical Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Laura Formenti
- Laboratory of Cancer Metastasis Therapeutics, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.,Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Massimo Russo
- Laboratory of Cancer Metastasis Therapeutics, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Edoardo Arrigoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | | | - Giovanna Chiorino
- Cancer Genomics Laboratory, Fondazione Edo ed Elvo Tempia Valenta, Biella, Italy
| | - Giulio Draetta
- Institute for Applied Cancer Science, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, Texas.,TRACTION Platform, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph R. Marszalek
- TRACTION Platform, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher P. Vellano
- TRACTION Platform, Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roberta Pastorelli
- Laboratory of Mass Spectrometry, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - MariaRosa Bani
- Laboratory of Cancer Metastasis Therapeutics, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Alessandra Decio
- Laboratory of Cancer Metastasis Therapeutics, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Raffaella Giavazzi
- Laboratory of Cancer Metastasis Therapeutics, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
23
|
Xue D, Xu Y, Kyani A, Roy J, Dai L, Sun D, Neamati N. Multiparameter Optimization of Oxidative Phosphorylation Inhibitors for the Treatment of Pancreatic Cancer. J Med Chem 2022; 65:3404-3419. [PMID: 35167303 DOI: 10.1021/acs.jmedchem.1c01934] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Targeting oxidative phosphorylation (OXPHOS) complexes is an emerging strategy to disrupt the metabolism of select cancer subtypes and to overcome resistance to targeted therapies. Here, we describe our lead optimization campaign on a series of benzene-1,4-disulfonamides as novel OXPHOS complex I inhibitors. This effort led to the discovery of compound 23 (DX3-213B) as one of the most potent complex I inhibitors reported to date. DX3-213B disrupts adenosine triphosphate (ATP) generation, inhibits complex I function, and results in the growth inhibition of pancreatic cancer cells in the low nanomolar range. Importantly, the oral administration of DX3-213B resulted in significant in vivo efficacy in a pancreatic cancer syngeneic model without obvious toxicity. Our data clearly demonstrate that OXPHOS inhibition can be a safe and efficacious strategy to treat pancreatic cancer.
Collapse
|
24
|
de Groot E, Varghese S, Tan L, Knighton B, Sobieski M, Nguyen N, Park YS, Powell R, Lorenzi PL, Zheng B, Stephan C, Gopal YNV. Combined inhibition of HMGCoA reductase and mitochondrial complex I induces tumor regression of BRAF inhibitor-resistant melanomas. Cancer Metab 2022; 10:6. [PMID: 35193687 PMCID: PMC8862475 DOI: 10.1186/s40170-022-00281-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/10/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Primary and posttreatment resistance to BRAFV600 mutation-targeting inhibitors leads to disease relapse in a majority of melanoma patients. In many instances, this resistance is promoted by upregulation of mitochondrial oxidative phosphorylation (OxPhos) in melanoma cells. We recently showed that a novel electron transport chain (ETC) complex I inhibitor, IACS-010759 (IACS), abolished OxPhos and significantly inhibited tumor growth of high-OxPhos, BRAF inhibitor (BRAFi)-resistant human melanomas. However, the inhibition was not uniform across different high OxPhos melanomas, and combination with BRAFi did not improve efficacy. METHODS We performed a high-throughput unbiased combinatorial drug screen of clinically relevant small molecules to identify the most potent combination agent with IACS for inhibiting the growth of high-OxPhos, BRAFi-resistant melanomas. We performed bioenergetics and carbon-13 metabolite tracing to delineate the metabolic basis of sensitization of melanomas to the combination treatment. We performed xenograft tumor growth studies and Reverse-Phase Protein Array (RPPA)-based functional proteomics analysis of tumors from mice fed with regular or high-fat diet to evaluate in vivo molecular basis of sensitization to the combination treatment. RESULTS A combinatorial drug screen and subsequent validation studies identified Atorvastatin (STN), a hydroxymethylglutaryl-coenzyme A reductase inhibitor (HMGCRi), as the most potent treatment combination with IACS to inhibit in vitro cell growth and induce tumor regression or stasis of some BRAFi-resistant melanomas. Bioenergetics analysis revealed a dependence on fatty acid metabolism in melanomas that responded to the combination treatment. RPPA analysis and carbon-13 tracing analysis in these melanoma cells showed that IACS treatment decreased metabolic fuel utilization for fatty acid metabolism, but increased substrate availability for activation of the mevalonate pathway by HMGCR, creating a dependence on this pathway. Functional proteomic analysis showed that IACS treatment inhibited MAPK but activated AKT pathway. Combination treatment with STN counteracted AKT activation. CONCLUSIONS STN and other clinically approved HMGCRi could be promising combinatorial agents for improving the efficacy of ETC inhibitors like IACS in BRAFi-resistant melanomas.
Collapse
Affiliation(s)
- Evelyn de Groot
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Sruthy Varghese
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Barbara Knighton
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Mary Sobieski
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Nghi Nguyen
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Yong Sung Park
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Reid Powell
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Bin Zheng
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Clifford Stephan
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Y N Vashisht Gopal
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
25
|
Greene J, Segaran A, Lord S. Targeting OXPHOS and the electronic transport chain in cancer; molecular and therapeutic implications. Semin Cancer Biol 2022; 86:851-859. [PMID: 35122973 DOI: 10.1016/j.semcancer.2022.02.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 12/11/2022]
Abstract
Oxidative phosphorylation (OXPHOS) takes place in mitochondria and is the process whereby cells use carbon fuels and oxygen to generate ATP. Formerly OXPHOS was thought to be reduced in tumours and that glycolysis was the critical pathway for generation of ATP but it is now clear that OXPHOS, at least in many tumour types, plays a critical role in delivering the bioenergetic and macromolecular anabolic requirements of cancer cells. There is now great interest in targeting the OXPHOS and the electron transport chain for cancer therapy and in this review article we describe current therapeutic approaches and challenges.
Collapse
Affiliation(s)
- John Greene
- Department of Oncology, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Ashvina Segaran
- Ludwig Institute for Cancer Research, University of Oxford, Old Road Campus Research Building, Oxford, United Kingdom
| | - Simon Lord
- Department of Oncology, University of Oxford, Churchill Hospital, Oxford, United Kingdom.
| |
Collapse
|
26
|
Kanakkanthara A, Hou X, Ekstrom TL, Zanfagnin V, Huehls AM, Kelly RL, Ding H, Larson MC, Vasmatzis G, Oberg AL, Kaufmann SH, Mansfield AS, John Weroha S, Karnitz LM. Repurposing Ceritinib Induces DNA Damage and Enhances PARP Inhibitor Responses in High-Grade Serous Ovarian Carcinoma. Cancer Res 2022; 82:307-319. [PMID: 34810199 PMCID: PMC8770599 DOI: 10.1158/0008-5472.can-21-0732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 08/05/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022]
Abstract
PARP inhibitors (PARPi) have activity in homologous recombination (HR) repair-deficient, high-grade serous ovarian cancers (HGSOC). However, even responsive tumors develop PARPi resistance, highlighting the need to delay or prevent the appearance of PARPi resistance. Here, we showed that the ALK kinase inhibitor ceritinib synergizes with PARPis by inhibiting complex I of the mitochondrial electron transport chain, which increases production of reactive oxygen species (ROS) and subsequent induction of oxidative DNA damage that is repaired in a PARP-dependent manner. In addition, combined treatment with ceritinib and PARPi synergized in HGSOC cell lines irrespective of HR status, and a combination of ceritinib with the PARPi olaparib induced tumor regression more effectively than olaparib alone in HGSOC patient-derived xenograft (PDX) models. Notably, the ceritinib and olaparib combination was most effective in PDX models with preexisting PARPi sensitivity and was well tolerated. These findings unveil suppression of mitochondrial respiration, accumulation of ROS, and subsequent induction of DNA damage as novel effects of ceritinib. They also suggest that the ceritinib and PARPi combination warrants further investigation as a means to enhance PARPi activity in HGSOC, particularly in tumors with preexisting HR defects. SIGNIFICANCE: The kinase inhibitor ceritinib synergizes with PARPi to induce tumor regression in ovarian cancer models, suggesting that ceritinib combined with PARPi may be an effective strategy for treating ovarian cancer.
Collapse
Affiliation(s)
- Arun Kanakkanthara
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA,To whom correspondence should be addressed: Larry M. Karnitz, Department of Oncology, Gonda 19-300, Mayo Clinic, 200 First Street SW, Rochester, MN 55905. Telephone: 507-284-3124; .; S. John Weroha, Department of Oncology, Guggenheim 13-01C, Mayo Clinic, 200 First Street SW, Rochester, MN 55905. Telephone: 507-284-3731; ; Arun Kanakkanthara, Department of Oncology, Gonda 19-300, Mayo Clinic, 200 First Street SW, Rochester, MN 55905. Telephone: 507-266-0268;
| | - Xiaonan Hou
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | | - Rebecca L. Kelly
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Husheng Ding
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Melissa C. Larson
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, Minnesota, USA
| | - George Vasmatzis
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ann L. Oberg
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Scott H. Kaufmann
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - S. John Weroha
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA,To whom correspondence should be addressed: Larry M. Karnitz, Department of Oncology, Gonda 19-300, Mayo Clinic, 200 First Street SW, Rochester, MN 55905. Telephone: 507-284-3124; .; S. John Weroha, Department of Oncology, Guggenheim 13-01C, Mayo Clinic, 200 First Street SW, Rochester, MN 55905. Telephone: 507-284-3731; ; Arun Kanakkanthara, Department of Oncology, Gonda 19-300, Mayo Clinic, 200 First Street SW, Rochester, MN 55905. Telephone: 507-266-0268;
| | - Larry M. Karnitz
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA,To whom correspondence should be addressed: Larry M. Karnitz, Department of Oncology, Gonda 19-300, Mayo Clinic, 200 First Street SW, Rochester, MN 55905. Telephone: 507-284-3124; .; S. John Weroha, Department of Oncology, Guggenheim 13-01C, Mayo Clinic, 200 First Street SW, Rochester, MN 55905. Telephone: 507-284-3731; ; Arun Kanakkanthara, Department of Oncology, Gonda 19-300, Mayo Clinic, 200 First Street SW, Rochester, MN 55905. Telephone: 507-266-0268;
| |
Collapse
|
27
|
Lemberg KM, Gori SS, Tsukamoto T, Rais R, Slusher BS. Clinical development of metabolic inhibitors for oncology. J Clin Invest 2022; 132:e148550. [PMID: 34981784 PMCID: PMC8718137 DOI: 10.1172/jci148550] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Metabolic inhibitors have been used in oncology for decades, dating back to antimetabolites developed in the 1940s. In the past 25 years, there has been increased recognition of metabolic derangements in tumor cells leading to a resurgence of interest in targeting metabolism. More recently there has been recognition that drugs targeting tumor metabolism also affect the often acidic, hypoxic, immunosuppressive tumor microenvironment (TME) and non-tumor cell populations within it, including immune cells. Here we review small-molecule metabolic inhibitors currently in clinical development for oncology applications. For each agent, we evaluate the preclinical studies demonstrating antitumor and TME effects and review ongoing clinical trials. The goal of this Review is to provide an overview of the landscape of metabolic inhibitors in clinical development for oncology.
Collapse
Affiliation(s)
- Kathryn M. Lemberg
- Johns Hopkins Drug Discovery
- Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center
| | | | - Takashi Tsukamoto
- Johns Hopkins Drug Discovery
- Department of Neurology
- Department of Pharmacology and Molecular Sciences
| | - Rana Rais
- Johns Hopkins Drug Discovery
- Department of Neurology
- Department of Pharmacology and Molecular Sciences
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery
- Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center
- Department of Neurology
- Department of Pharmacology and Molecular Sciences
- Department of Medicine, and
- Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Nguyen TTT, Shang E, Westhoff MA, Karpel-Massler G, Siegelin MD. Methodological Approaches for Assessing Metabolomic Changes in Glioblastomas. Methods Mol Biol 2022; 2445:305-328. [PMID: 34973000 DOI: 10.1007/978-1-0716-2071-7_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Glioblastoma (GBM), a highly malignant primary brain tumor, inevitably leads to death. In the last decade, a variety of novel molecular characteristics of GBMs were unraveled. The identification of the mutation in the IDH1 and less commonly IDH2 gene was surprising and ever since has nurtured research in the field of GBM metabolism. While initially thought that mutated IDH1 were to act as a loss of function mutation it became clear that it conferred the production of an oncometabolite that in turn substantially reprograms GBM metabolism. While mutated IDH1 represents truly the tip of the iceberg, there are numerous other related observations in GBM that are of significant interest to the field, including the notion that oxidative metabolism appears to play a more critical role than believed earlier. Metabolic zoning is another important hallmark of GBM since it was found that the infiltrative margin that drives GBM progression reveals enrichment of fatty acid derivatives. Consistently, fatty acid metabolism appears to be a novel therapeutic target for GBM. How metabolism in GBM intersects is another pivotal issue that appears to be important for its progression and response and resistance to therapies. In this review, we will summarize some of the most relevant findings related to GBM metabolism and cell death and how these observations are influencing the field. We will provide current approaches that are applied in the field to measure metabolomic changes in GBM models, including the detection of unlabeled and labeled metabolites as well as extracellular flux analysis.
Collapse
Affiliation(s)
- Trang T T Nguyen
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Enyuan Shang
- Department of Biological Sciences, Bronx Community College, City University of New York, Bronx, NY, USA
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | | | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
29
|
Saberian C, Sperduto P, Davies MA. Targeted therapy strategies for melanoma brain metastasis. Neurooncol Adv 2021; 3:v75-v85. [PMID: 34859235 PMCID: PMC8633745 DOI: 10.1093/noajnl/vdab131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Melanoma is the most aggressive of the common forms of skin cancer. Metastasis to the central nervous system is one of the most common and deadly complications of this disease. Historically, melanoma patients with brain metastases had a median survival of less than 6 months. However, outcomes of melanoma patients have markedly improved over the last decade due to new therapeutic approaches, including immune and targeted therapies. Targeted therapies leverage the high rate of driver mutations in this disease, which result in the activation of multiple key signaling pathways. The RAS-RAF-MEK-ERK pathway is activated in the majority of cutaneous melanomas, most commonly by point mutations in the Braf serine-threonine kinase. While most early targeted therapy studies excluded melanoma patients with brain metastases, subsequent studies have shown that BRAF inhibitors, now generally given concurrently with MEK inhibitors, achieve high rates of tumor response and disease control in Braf-mutant melanoma brain metastases (MBMs). Unfortunately, the duration of these responses is generally relatively short- and shorter than is observed in extracranial metastases. This review will summarize current data regarding the safety and efficacy of targeted therapies for MBMs and discuss rational combinatorial strategies that may improve outcomes further.
Collapse
Affiliation(s)
- Chantal Saberian
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Paul Sperduto
- Minneapolis Radiation Oncology, Minneapolis, Minnesota, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
30
|
Stine ZE, Schug ZT, Salvino JM, Dang CV. Targeting cancer metabolism in the era of precision oncology. Nat Rev Drug Discov 2021; 21:141-162. [PMID: 34862480 PMCID: PMC8641543 DOI: 10.1038/s41573-021-00339-6] [Citation(s) in RCA: 493] [Impact Index Per Article: 164.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2021] [Indexed: 12/23/2022]
Abstract
One hundred years have passed since Warburg discovered alterations in cancer metabolism, more than 70 years since Sidney Farber introduced anti-folates that transformed the treatment of childhood leukaemia, and 20 years since metabolism was linked to oncogenes. However, progress in targeting cancer metabolism therapeutically in the past decade has been limited. Only a few metabolism-based drugs for cancer have been successfully developed, some of which are in - or en route to - clinical trials. Strategies for targeting the intrinsic metabolism of cancer cells often did not account for the metabolism of non-cancer stromal and immune cells, which have pivotal roles in tumour progression and maintenance. By considering immune cell metabolism and the clinical manifestations of inborn errors of metabolism, it may be possible to isolate undesirable off-tumour, on-target effects of metabolic drugs during their development. Hence, the conceptual framework for drug design must consider the metabolic vulnerabilities of non-cancer cells in the tumour immune microenvironment, as well as those of cancer cells. In this Review, we cover the recent developments, notable milestones and setbacks in targeting cancer metabolism, and discuss the way forward for the field.
Collapse
Affiliation(s)
| | | | | | - Chi V Dang
- The Wistar Institute Philadelphia, Philadelphia, PA, USA. .,Ludwig Institute for Cancer Research New York, New York, NY, USA.
| |
Collapse
|
31
|
Metabolic Plasticity in Melanoma Progression and Response to Oncogene Targeted Therapies. Cancers (Basel) 2021; 13:cancers13225810. [PMID: 34830962 PMCID: PMC8616485 DOI: 10.3390/cancers13225810] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Targeted anti-cancer therapies have revolutionised melanoma patient care; however, cures remain uncommon due to acquired drug resistance that results in disease relapse. Recent insights from the clinic and experimental settings have identified a key role for metabolic plasticity, defined as the flexibility to utilise different nutrients and process them in different ways, in both disease progression and response to targeted therapies. Here, we discuss how this plasticity creates a moving target with important implications for identifying new combination therapies. Abstract Resistance to therapy continues to be a barrier to curative treatments in melanoma. Recent insights from the clinic and experimental settings have highlighted a range of non-genetic adaptive mechanisms that contribute to therapy resistance and disease relapse, including transcriptional, post-transcriptional and metabolic reprogramming. A growing body of evidence highlights the inherent plasticity of melanoma metabolism, evidenced by reversible metabolome alterations and flexibility in fuel usage that occur during metastasis and response to anti-cancer therapies. Here, we discuss how the inherent metabolic plasticity of melanoma cells facilitates both disease progression and acquisition of anti-cancer therapy resistance. In particular, we discuss in detail the different metabolic changes that occur during the three major phases of the targeted therapy response—the early response, drug tolerance and acquired resistance. We also discuss how non-genetic programs, including transcription and translation, control this process. The prevalence and diverse array of these non-genetic resistance mechanisms poses a new challenge to the field that requires innovative strategies to monitor and counteract these adaptive processes in the quest to prevent therapy resistance.
Collapse
|
32
|
Anderson NM, Qin X, Finan JM, Lam A, Athoe J, Missiaen R, Skuli N, Kennedy A, Saini AS, Tao T, Zhu S, Nissim I, Look AT, Qing G, Simon MC, Feng H. Metabolic Enzyme DLST Promotes Tumor Aggression and Reveals a Vulnerability to OXPHOS Inhibition in High-Risk Neuroblastoma. Cancer Res 2021; 81:4417-4430. [PMID: 34233924 DOI: 10.1158/0008-5472.can-20-2153] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 03/13/2021] [Accepted: 06/25/2021] [Indexed: 11/16/2022]
Abstract
High-risk neuroblastoma remains therapeutically challenging to treat, and the mechanisms promoting disease aggression are poorly understood. Here, we show that elevated expression of dihydrolipoamide S-succinyltransferase (DLST) predicts poor treatment outcome and aggressive disease in patients with neuroblastoma. DLST is an E2 component of the α-ketoglutarate (αKG) dehydrogenase complex, which governs the entry of glutamine into the tricarboxylic acid cycle (TCA) for oxidative decarboxylation. During this irreversible step, αKG is converted into succinyl-CoA, producing NADH for oxidative phosphorylation (OXPHOS). Utilizing a zebrafish model of MYCN-driven neuroblastoma, we demonstrate that even modest increases in DLST expression promote tumor aggression, while monoallelic dlst loss impedes disease initiation and progression. DLST depletion in human MYCN-amplified neuroblastoma cells minimally affected glutamine anaplerosis and did not alter TCA cycle metabolites other than αKG. However, DLST loss significantly suppressed NADH production and impaired OXPHOS, leading to growth arrest and apoptosis of neuroblastoma cells. In addition, multiple inhibitors targeting the electron transport chain, including the potent IACS-010759 that is currently in clinical testing for other cancers, efficiently reduced neuroblastoma proliferation in vitro. IACS-010759 also suppressed tumor growth in zebrafish and mouse xenograft models of high-risk neuroblastoma. Together, these results demonstrate that DLST promotes neuroblastoma aggression and unveils OXPHOS as an essential contributor to high-risk neuroblastoma. SIGNIFICANCE: These findings demonstrate a novel role for DLST in neuroblastoma aggression and identify the OXPHOS inhibitor IACS-010759 as a potential therapeutic strategy for this deadly disease.
Collapse
Affiliation(s)
- Nicole M Anderson
- Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania.,Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaodan Qin
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, The Center for Cancer Research, Boston University School of Medicine, Boston, Massachusetts
| | - Jennifer M Finan
- Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Andrew Lam
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, The Center for Cancer Research, Boston University School of Medicine, Boston, Massachusetts
| | - Jacob Athoe
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, The Center for Cancer Research, Boston University School of Medicine, Boston, Massachusetts
| | - Rindert Missiaen
- Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Nicolas Skuli
- Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Annie Kennedy
- Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Amandeep S Saini
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, The Center for Cancer Research, Boston University School of Medicine, Boston, Massachusetts
| | - Ting Tao
- National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shizhen Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Cancer Center, Rochester, Minnesota
| | - Itzhak Nissim
- Division of Genetics and Metabolism, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pediatrics, Biochemistry, and Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - A Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Guoliang Qing
- Frontier Science Center for Immunology & Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania. .,Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hui Feng
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, The Center for Cancer Research, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
33
|
Many Distinct Ways Lead to Drug Resistance in BRAF- and NRAS-Mutated Melanomas. Life (Basel) 2021; 11:life11050424. [PMID: 34063141 PMCID: PMC8148104 DOI: 10.3390/life11050424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/23/2021] [Accepted: 04/30/2021] [Indexed: 11/17/2022] Open
Abstract
Advanced melanoma is a relentless tumor with a high metastatic potential. The combat of melanoma by using the targeted therapy is impeded because several major driver mutations fuel its growth (predominantly BRAF and NRAS). Both these mutated oncogenes strongly activate the MAPK (MEK/ERK) pathway. Therefore, specific inhibitors of these oncoproteins or MAPK pathway components or their combination have been used for tumor eradication. After a good initial response, resistant cells develop almost universally and need the drug for further expansion. Multiple mechanisms, sometimes very distant from the MAPK pathway, are responsible for the development of resistance. Here, we review many of the mechanisms causing resistance and leading to the dismal final outcome of mutated BRAF and NRAS therapy. Very heterogeneous events lead to drug resistance. Due to this, each individual mechanism would be in fact needed to be determined for a personalized therapy to treat patients more efficiently and causally according to molecular findings. This procedure is practically impossible in the clinic. Other approaches are therefore needed, such as combined treatment with more drugs simultaneously from the beginning of the therapy. This could eradicate tumor cells more rapidly and greatly diminish the possibility of emerging mechanisms that allow the evolution of drug resistance.
Collapse
|
34
|
Targeting Mitochondrial Metabolism in Clear Cell Carcinoma of the Ovaries. Int J Mol Sci 2021; 22:ijms22094750. [PMID: 33947138 PMCID: PMC8124918 DOI: 10.3390/ijms22094750] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
Ovarian clear cell carcinoma (OCCC) is a rare but chemorefractory tumor. About 50% of all OCCC patients have inactivating mutations of ARID1A, a member of the SWI/SNF chromatin-remodeling complex. Members of the SWI/SNF remodeling have emerged as regulators of the energetic metabolism of mammalian cells; however, the role of ARID1A as a modulator of the mitochondrial metabolism in OCCCs is yet to be defined. Here, we show that ARID1A loss results in increased mitochondrial metabolism and renders ARID1A-mutated cells increasingly and selectively dependent on it. The increase in mitochondrial activity following ARID1A loss is associated with increase in c-Myc expression and increased mitochondrial number and reduction of their size consistent with a higher mitochondrial cristae/outer membrane ratio. Significantly, preclinical testing of the complex I mitochondrial inhibitor IACS-010759 showed it extends overall survival in a preclinical model of ARID1A-mutated OCCC. These findings provide for the targeting mitochondrial activity in ARID1A-mutated OCCCs.
Collapse
|
35
|
Kosaisawe N, Sparta B, Pargett M, Teragawa CK, Albeck JG. Transient phases of OXPHOS inhibitor resistance reveal underlying metabolic heterogeneity in single cells. Cell Metab 2021; 33:649-665.e8. [PMID: 33561427 PMCID: PMC8005262 DOI: 10.1016/j.cmet.2021.01.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/13/2020] [Accepted: 01/13/2021] [Indexed: 12/16/2022]
Abstract
Cell-to-cell heterogeneity in metabolism plays an unknown role in physiology and pharmacology. To functionally characterize cellular variability in metabolism, we treated cells with inhibitors of oxidative phosphorylation (OXPHOS) and monitored their responses with live-cell reporters for ATP, ADP/ATP, or activity of the energy-sensing kinase AMPK. Across multiple OXPHOS inhibitors and cell types, we identified a subpopulation of cells resistant to activation of AMPK and reduction of ADP/ATP ratio. This resistant state persists transiently for at least several hours and can be inherited during cell divisions. OXPHOS inhibition suppresses the mTORC1 and ERK growth signaling pathways in sensitive cells, but not in resistant cells. Resistance is linked to a multi-factorial combination of increased glucose uptake, reduced protein biosynthesis, and G0/G1 cell-cycle status. Our results reveal dynamic fluctuations in cellular energetic balance and provide a basis for measuring and predicting the distribution of cellular responses to OXPHOS inhibition.
Collapse
Affiliation(s)
- Nont Kosaisawe
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Breanne Sparta
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Michael Pargett
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Carolyn K Teragawa
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - John G Albeck
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
36
|
Fischer GM, Guerrieri RA, Hu Q, Joon AY, Kumar S, Haydu LE, McQuade JL, Vashisht Gopal YN, Knighton B, Deng W, Hudgens CW, Lazar AJ, Tetzlaff MT, Davies MA. Clinical, molecular, metabolic, and immune features associated with oxidative phosphorylation in melanoma brain metastases. Neurooncol Adv 2021; 3:vdaa177. [PMID: 33575655 PMCID: PMC7865080 DOI: 10.1093/noajnl/vdaa177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Recently, we showed that melanoma brain metastases (MBMs) are characterized by increased utilization of the oxidative phosphorylation (OXPHOS) metabolic pathway compared to melanoma extracranial metastases (ECMs). MBM growth was inhibited by a potent direct OXPHOS inhibitor, but observed toxicities support the need to identify alternative therapeutic strategies. Thus, we explored the features associated with OXPHOS to improve our understanding of the pathogenesis and potential therapeutic vulnerabilities of MBMs. Methods We applied an OXPHOS gene signature to our cohort of surgically resected MBMs that had undergone RNA-sequencing (RNA-seq) (n = 88). Clustering by curated gene sets identified MBMs with significant enrichment (High-OXPHOS; n = 21) and depletion (Low-OXPHOS; n = 25) of OXPHOS genes. Clinical data, RNA-seq analysis, and immunohistochemistry were utilized to identify significant clinical, molecular, metabolic, and immune associations with OXPHOS in MBMs. Preclinical models were used to further compare melanomas with High- and Low-OXPHOS and for functional validation. Results High-OXPHOS MBMs were associated with shorter survival from craniotomy compared to Low-OXPHOS MBMs. High-OXPHOS MBMs exhibited an increase in glutamine metabolism, and treatment with the glutaminase inhibitor CB839 improved survival in mice with MAPKi-resistant, High-OXPHOS intracranial xenografts. High-OXPHOS MBMs also exhibited a transcriptional signature of deficient immune activation, which was reversed in B16-F10 intracranial tumors with metformin treatment, an OXPHOS inhibitor. Conclusions OXPHOS is associated with distinct clinical, molecular, metabolic, and immune phenotypes in MBMs. These associations suggest rational therapeutic strategies for further testing to improve outcomes in MBM patients.
Collapse
Affiliation(s)
- Grant M Fischer
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Renato A Guerrieri
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Qianghua Hu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Aron Y Joon
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Swaminathan Kumar
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lauren E Haydu
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer L McQuade
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Y N Vashisht Gopal
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Barbara Knighton
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wanleng Deng
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Courtney W Hudgens
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alexander J Lazar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael T Tetzlaff
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael A Davies
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
37
|
Varghese S, Pramanik S, Williams LJ, Hodges HR, Hudgens CW, Fischer GM, Luo CK, Knighton B, Tan L, Lorenzi PL, Mackinnon AL, McQuade JL, Hailemichael Y, Roszik J, Peng W, Vashisht Gopal YN. The Glutaminase Inhibitor CB-839 (Telaglenastat) Enhances the Antimelanoma Activity of T-Cell-Mediated Immunotherapies. Mol Cancer Ther 2020; 20:500-511. [PMID: 33361272 DOI: 10.1158/1535-7163.mct-20-0430] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/09/2020] [Accepted: 12/16/2020] [Indexed: 01/02/2023]
Abstract
Immune-checkpoint inhibitors and adoptive tumor-infiltrating lymphocyte (TIL) therapies have profoundly improved the survival of patients with melanoma. However, a majority of patients do not respond to these agents, and many responders experience disease relapse. Although numerous innovative treatments are being explored to offset the limitations of these agents, novel therapeutic combinations with immunotherapies have the potential to improve patient responses. In this study, we evaluated the antimelanoma activity of immunotherapy combinations with Telaglenastat (CB-839), a potent glutaminase inhibitor (GLSi) that has favorable systemic tolerance. In in vitro TIL:tumor coculture studies, CB-839 treatment improved the cytotoxic activity of autologous TILs on patient-derived melanoma cells. CB-839 treatment decreased the conversion of glutamine to alpha-ketoglutarate (αKGA) more potently in tumor cells versus TILs in these cocultures. These results suggest that CB-839 may improve immune function in a tumor microenvironment by differentially altering tumor and immune cell metabolism. In vivo CB-839 treatment activated melanoma antigen-specific T cells and improved their tumor killing activity in an immune-competent mouse model of adoptive T-cell therapy. Additionally, the combination of CB-839 with anti-PD1 or anti-CTLA4 antibodies increased tumor infiltration by effector T cells and improved the antitumor activity of these checkpoint inhibitors in a high mutation burden mouse melanoma model. Responsiveness to these treatments was also accompanied by an increase of interferon gamma (IFNγ)-associated gene expression in the tumors. Together, these results provide a strong rationale for combining CB-839 with immune therapies to improve efficacy of these treatments against melanoma.
Collapse
Affiliation(s)
- Sruthy Varghese
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX.,Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Snigdha Pramanik
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Leila J Williams
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Hannah R Hodges
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX.,Department of Chemical Engineering, University of Texas, Austin, TX
| | - Courtney W Hudgens
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Grant M Fischer
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Catherine K Luo
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX.,Department of Cellular and Molecular Biology, John Hopkins University, Baltimore, MD
| | - Barbara Knighton
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | | | - Jennifer L McQuade
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Yared Hailemichael
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Jason Roszik
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston, Houston, TX
| | - Y N Vashisht Gopal
- Department of Melanoma Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX. .,Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
38
|
Steindl A, Berghoff AS. Brain metastases in metastatic cancer: a review of recent advances in systemic therapies. Expert Rev Anticancer Ther 2020; 21:325-339. [PMID: 33196341 DOI: 10.1080/14737140.2021.1851200] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Brain metastases (BM) are a frequent complication of metastatic cancer. Due to the wider availability and application of screening procedures, an increasing fraction of patients are diagnosed at the asymptomatic stage. The introduction of immune checkpoint inhibitors and targeted therapies has revolutionized treatment in several frequently BM-causing entities like metastatic lung cancer, melanoma and breast cancer. However, registered trials of new targeted and immunotherapy mostly excluded patients with BM resulting in limited knowledge of the intracranial efficacy of new systemic agents.Areas covered: The present review highlights recent advances in systemic therapies for the treatment and prophylaxis of the three leading BM causing tumors: NSCLC, melanoma and breast cancer.Expert opinion: High intracranial efficacy was observed for several next-generation tyrosine kinase inhibitors as well as immune checkpoint inhibitors, especially in patients with asymptomatic disease. Ongoing discussions addressed the need for local therapies in patients with asymptomatic BM and the availability of systemic therapy with high intracranial efficacy. Further BM-specific studies as well as BM-specific endpoints in registered trials are needed to define the role of systemic monotherapies in patients with BM.
Collapse
Affiliation(s)
- Ariane Steindl
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
39
|
Fattore L, Mancini R, Ciliberto G. Cancer Stem Cells and the Slow Cycling Phenotype: How to Cut the Gordian Knot Driving Resistance to Therapy in Melanoma. Cancers (Basel) 2020; 12:cancers12113368. [PMID: 33202944 PMCID: PMC7696527 DOI: 10.3390/cancers12113368] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Cancer stem cells play a central role in the development of cancer and are poorly sensitive to standard chemotherapy and radiotherapy. Furthermore, they are also responsible for the onset of drug resistance. This also occurs in malignant melanoma, the deadliest form of skin cancer. Hence, cancer stem cells eradication is one of the main challenges for medical oncology. Here, we conducted a bioinformatics approach aimed to identify the main circuits and proteins underpinning cancer stem cell fitness in melanoma. Several lessons emerged from our work and may help to conceptualize future therapeutic approaches to prolong the efficacy of current therapies. Abstract Cancer stem cells (CSCs) have historically been defined as slow cycling elements that are able to differentiate into mature cells but without dedifferentiation in the opposite direction. Thanks to advances in genomic and non-genomic technologies, the CSC theory has more recently been reconsidered in a dynamic manner according to a “phenotype switching” plastic model. Transcriptional reprogramming rewires this plasticity and enables heterogeneous tumors to influence cancer progression and to adapt themselves to drug exposure by selecting a subpopulation of slow cycling cells, similar in nature to the originally defined CSCs. This model has been conceptualized for malignant melanoma tailored to explain resistance to target therapies. Here, we conducted a bioinformatics analysis of available data directed to the identification of the molecular pathways sustaining slow cycling melanoma stem cells. Using this approach, we identified a signature of 25 genes that were assigned to four major clusters, namely (1) kinases and metabolic changes, (2) melanoma-associated proteins, (3) Hippo pathway and (4) slow cycling/CSCs factors. Furthermore, we show how a protein−protein interaction network may be the main driver of these melanoma cell subpopulations. Finally, mining The Cancer Genome Atlas (TCGA) data we evaluated the expression levels of this signature in the four melanoma mutational subtypes. The concomitant alteration of these genes correlates with the worst overall survival (OS) for melanoma patients harboring BRAF-mutations. All together these results underscore the potentiality to target this signature to selectively kill CSCs and to achieve disease control in melanoma.
Collapse
Affiliation(s)
- Luigi Fattore
- Department of Research, Advanced Diagnostics and Technological Innovation, SAFU Laboratory, Translational Research Area, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS, “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Sant’ Andrea Hospital, Sapienza University of Rome, 00161 Rome, Italy;
| | - Gennaro Ciliberto
- Scientific Directorate, IRCSS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Correspondence:
| |
Collapse
|
40
|
Xu Y, Xue D, Bankhead A, Neamati N. Why All the Fuss about Oxidative Phosphorylation (OXPHOS)? J Med Chem 2020; 63:14276-14307. [PMID: 33103432 DOI: 10.1021/acs.jmedchem.0c01013] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Certain subtypes of cancer cells require oxidative phosphorylation (OXPHOS) to survive. Increased OXPHOS dependency is frequently a hallmark of cancer stem cells and cells resistant to chemotherapy and targeted therapies. Suppressing the OXPHOS function might also influence the tumor microenvironment by alleviating hypoxia and improving the antitumor immune response. Thus, targeting OXPHOS is a promising strategy to treat various cancers. A growing arsenal of therapeutic agents is under development to inhibit this biological process. This Perspective provides an overview of the structure and function of OXPHOS complexes, their biological functions in cancer, relevant research tools and models, as well as the limitations of OXPHOS as drug targets. We also focus on the current development status of OXPHOS inhibitors and potential therapeutic strategies to strengthen their clinical applications.
Collapse
Affiliation(s)
- Yibin Xu
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ding Xue
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Armand Bankhead
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States.,Department of Biostatistics, University of Michigan, School of Public Health, Ann Arbor, Michigan 48109, United States
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
41
|
Klein K, He K, Younes AI, Barsoumian HB, Chen D, Ozgen T, Mosaffa S, Patel RR, Gu M, Novaes J, Narayanan A, Cortez MA, Welsh JW. Role of Mitochondria in Cancer Immune Evasion and Potential Therapeutic Approaches. Front Immunol 2020; 11:573326. [PMID: 33178201 PMCID: PMC7596324 DOI: 10.3389/fimmu.2020.573326] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/18/2020] [Indexed: 11/13/2022] Open
Abstract
The role of mitochondria in cancer formation and progression has been studied extensively, but much remains to be understood about this complex relationship. Mitochondria regulate many processes that are known to be altered in cancer cells, from metabolism to oxidative stress to apoptosis. Here, we review the evolving understanding of the role of mitochondria in cancer cells, and highlight key evidence supporting the role of mitochondria in cancer immune evasion and the effects of mitochondria-targeted antitumor therapy. Also considered is how knowledge of the role of mitochondria in cancer can be used to design and improve cancer therapies, particularly immunotherapy and radiation therapy. We further offer critical insights into the mechanisms by which mitochondria influence tumor immune responses, not only in cancer cells but also in immune cells. Given the central role of mitochondria in the complex interactions between cancer and the immune system, high priority should be placed on developing rational strategies to address mitochondria as potential targets in future preclinical and clinical studies. We believe that targeting mitochondria may provide additional opportunities in the development of novel antitumor therapeutics.
Collapse
Affiliation(s)
- Katherine Klein
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,McGovern Medical School at UTHealth, Houston, TX, United States
| | - Kewen He
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China
| | - Ahmed I Younes
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dawei Chen
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China
| | - Tugce Ozgen
- Ankara University Faculty of Medicine, Ankara, Turkey
| | - Sara Mosaffa
- Department of Molecular Biosciences, The University of Texas at Austin, Houston, TX, United States
| | - Roshal R Patel
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Meidi Gu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jose Novaes
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, The Bronx, NY, United States
| | - Aarthi Narayanan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Angelica Cortez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
42
|
Aminzadeh-Gohari S, Weber DD, Catalano L, Feichtinger RG, Kofler B, Lang R. Targeting Mitochondria in Melanoma. Biomolecules 2020; 10:biom10101395. [PMID: 33007949 PMCID: PMC7599575 DOI: 10.3390/biom10101395] [Citation(s) in RCA: 17] [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: 08/31/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
Drastically elevated glycolytic activity is a prominent metabolic feature of cancer cells. Until recently it was thought that tumor cells shift their entire energy production from oxidative phosphorylation (OXPHOS) to glycolysis. However, new evidence indicates that many cancer cells still have functional OXPHOS, despite their increased reliance on glycolysis. Growing pre-clinical and clinical evidence suggests that targeting mitochondrial metabolism has anti-cancer effects. Here, we analyzed mitochondrial respiration and the amount and activity of OXPHOS complexes in four melanoma cell lines and normal human dermal fibroblasts (HDFs) by Seahorse real-time cell metabolic analysis, immunoblotting, and spectrophotometry. We also tested three clinically approved antibiotics, one anti-parasitic drug (pyrvinium pamoate), and a novel anti-cancer agent (ONC212) for effects on mitochondrial respiration and proliferation of melanoma cells and HDFs. We found that three of the four melanoma cell lines have elevated glycolysis as well as OXPHOS, but contain dysfunctional mitochondria. The antibiotics produced different effects on the melanoma cells and HDFs. The anti-parasitic drug strongly inhibited respiration and proliferation of both the melanoma cells and HDFs. ONC212 reduced respiration in melanoma cells and HDFs, and inhibited the proliferation of melanoma cells. Our findings highlight ONC212 as a promising drug for targeting mitochondrial respiration in cancer.
Collapse
Affiliation(s)
- Sepideh Aminzadeh-Gohari
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (S.A.-G.); (D.D.W.); (L.C.); (R.G.F.)
| | - Daniela D. Weber
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (S.A.-G.); (D.D.W.); (L.C.); (R.G.F.)
| | - Luca Catalano
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (S.A.-G.); (D.D.W.); (L.C.); (R.G.F.)
| | - René G. Feichtinger
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (S.A.-G.); (D.D.W.); (L.C.); (R.G.F.)
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (S.A.-G.); (D.D.W.); (L.C.); (R.G.F.)
- Correspondence: (B.K.); (R.L.); Tel.: +43-57255-26274 (B.K.); +43-57255-58200 (R.L.)
| | - Roland Lang
- Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
- Correspondence: (B.K.); (R.L.); Tel.: +43-57255-26274 (B.K.); +43-57255-58200 (R.L.)
| |
Collapse
|
43
|
Sun X, Zhang N, Yin C, Zhu B, Li X. Ultraviolet Radiation and Melanomagenesis: From Mechanism to Immunotherapy. Front Oncol 2020; 10:951. [PMID: 32714859 PMCID: PMC7343965 DOI: 10.3389/fonc.2020.00951] [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: 03/01/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022] Open
Abstract
Melanoma is the deadliest form of skin cancer, and nearly 90% of melanomas are believed to be caused by ultraviolet radiation (UVR), mainly from sunlight. UVR induces DNA damage, forming products such as cyclobutane pyrimidine dimers (CPD) and 6-4-pyrimidone photoproducts (6-4PP) in a wavelength-dependent manner and causes oxidative DNA damage. These DNA lesions lead to DNA mutations and contribute to the formation of melanoma. In this review, we discuss the protective role of melanocytes against UV-induced DNA damage and how genetic variations, including those in p53 and melanocortin-1 receptor (MC1R), or epigenetic histone modifications in melanocytes result in a tendency toward melanoma. We also provide a summary of prevention and treatment strategies against melanoma, including the most recent immunotherapies. Collectively, this work contributes to the understanding of the molecular pathogenesis of UV-induced melanoma.
Collapse
Affiliation(s)
- Xiaoying Sun
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Na Zhang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chengqian Yin
- Department of Dermatology, Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Bo Zhu
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Xin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
44
|
Rytelewski M, Harutyunyan K, Baran N, Mallampati S, Zal MA, Cavazos A, Butler JM, Konoplev S, El Khatib M, Plunkett S, Marszalek JR, Andreeff M, Zal T, Konopleva M. Inhibition of Oxidative Phosphorylation Reverses Bone Marrow Hypoxia Visualized in Imageable Syngeneic B-ALL Mouse Model. Front Oncol 2020; 10:991. [PMID: 32695673 PMCID: PMC7339962 DOI: 10.3389/fonc.2020.00991] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Abnormally low level of interstitial oxygen, or hypoxia, is a hallmark of tumor microenvironment and a known promoter of cancer chemoresistance. Inside a solid tumor mass, the hypoxia stems largely from inadequate supply of oxygenated blood through sparse or misshapen tumor vasculature whilst oxygen utilization rates are low in typical tumor's glycolytic metabolism. In acute leukemias, however, markers of intracellular hypoxia such as increased pimonidazole adduct staining and HIF-1α stabilization are observed in advanced leukemic bone marrows (BM) despite an increase in BM vasculogenesis. We utilized intravital fast scanning two-photon phosphorescence lifetime imaging microscopy (FaST-PLIM) in a BCR-ABL B-ALL mouse model to image the extracellular oxygen concentrations (pO2) in leukemic BM, and we related the extracellular oxygen levels to intracellular hypoxia, vascular markers and local leukemia burden. We observed a transient increase in BM pO2 in initial disease stages with intermediate leukemia BM burden, which correlated with an expansion of blood-carrying vascular network in the BM. Yet, we also observed increased formation of intracellular pimonidazole adducts in leukemic BM at the same time. This intermediate stage was followed by a significant decrease of extracellular pO2 and further increase of intracellular hypoxia as leukemia cellularity overwhelmed BM in disease end-stage. Remarkably, treatment of leukemic mice with IACS-010759, a pharmacological inhibitor of mitochondrial Complex I, substantially increased pO2 in the BM with advanced B-ALL, and it alleviated intracellular hypoxia reported by pimonidazole staining. High rates of oxygen consumption by B-ALL cells were confirmed by Seahorse assay including in ex vivo cells. Our results suggest that B-ALL expansion in BM is associated with intense oxidative phosphorylation (OxPhos) leading to the onset of metabolic BM hypoxia despite increased BM vascularization. Targeting mitochondrial respiration may be a novel approach to counteract BM hypoxia in B-ALL and, possibly, tumor hypoxia in other OxPhos-reliant malignancies.
Collapse
Affiliation(s)
- Mateusz Rytelewski
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Karine Harutyunyan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Saradhi Mallampati
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - M Anna Zal
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Antonio Cavazos
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jason M Butler
- Weill Cornell Medicine, Medical School of Biological Sciences, Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ, United States
| | - Sergej Konoplev
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mirna El Khatib
- Department of Biochemistry and Biophysics, The University of Pennsylvania, Philadelphia, PA, United States
| | - Shane Plunkett
- Department of Biochemistry and Biophysics, The University of Pennsylvania, Philadelphia, PA, United States
| | - Joseph R Marszalek
- TRACTION, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Tomasz Zal
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
45
|
Lee SH, Griffiths JR. How and Why Are Cancers Acidic? Carbonic Anhydrase IX and the Homeostatic Control of Tumour Extracellular pH. Cancers (Basel) 2020; 12:cancers12061616. [PMID: 32570870 PMCID: PMC7352839 DOI: 10.3390/cancers12061616] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
The acidic tumour microenvironment is now recognized as a tumour phenotype that drives cancer somatic evolution and disease progression, causing cancer cells to become more invasive and to metastasise. This property of solid tumours reflects a complex interplay between cellular carbon metabolism and acid removal that is mediated by cell membrane carbonic anhydrases and various transport proteins, interstitial fluid buffering, and abnormal tumour-associated vessels. In the past two decades, a convergence of advances in the experimental and mathematical modelling of human cancers, as well as non-invasive pH-imaging techniques, has yielded new insights into the physiological mechanisms that govern tumour extracellular pH (pHe). In this review, we examine the mechanisms by which solid tumours maintain a low pHe, with a focus on carbonic anhydrase IX (CAIX), a cancer-associated cell surface enzyme. We also review the accumulating evidence that suggest a role for CAIX as a biological pH-stat by which solid tumours stabilize their pHe. Finally, we highlight the prospects for the clinical translation of CAIX-targeted therapies in oncology.
Collapse
Affiliation(s)
- Shen-Han Lee
- Department of Otorhinolaryngology, Hospital Sultanah Bahiyah, Jalan Langgar, Alor Setar 05460, Kedah, Malaysia
- Correspondence:
| | - John R. Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK;
| |
Collapse
|
46
|
Chen D, Barsoumian HB, Fischer G, Yang L, Verma V, Younes AI, Hu Y, Masropour F, Klein K, Vellano C, Marszalek J, Davies M, Cortez MA, Welsh J. Combination treatment with radiotherapy and a novel oxidative phosphorylation inhibitor overcomes PD-1 resistance and enhances antitumor immunity. J Immunother Cancer 2020; 8:e000289. [PMID: 32581056 PMCID: PMC7319777 DOI: 10.1136/jitc-2019-000289] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2020] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Despite outstanding responses to anti-PD-1 agents in a subset of non-small cell lung cancer (NSCLC) patients, approximately 80% of patients fail to have prolonged favorable response. Recent studies show that tumor cell oxidative metabolism is a barrier to PD-1 immunotherapy and radiotherapy could overcome PD-1 resistance, so it is urgent to determine if combination treatment with radiotherapy and a novel oxidative phosphorylation (OXPHOS) inhibitor (IACS-010759) is an effective strategy against PD-1 resistance in NSCLC. METHODS The antitumor effect of this combinational treatment was evaluated in vitro and in vivo. For in vivo experiments, we treated 129Sv/Ev mice with anti-PD1-sensitive and anti-PD1-resistant 344SQ NSCLC adenocarcinoma xenografts with oral IACS-010759 combined with radiotherapy (XRT). In vitro experiments included PCR, seahorse bioenergetic profiling, flow cytometry phenotyping, and clonogenic survival assay. RESULTS In the current study, we found that our PD-1-resistant model utilized OXPHOS to a significantly greater extent than the PD-1-sensitive model and XRT increased OXPHOS in vitro and in vivo. Thus, we explored the effect of the novel OXPHOS inhibitor IACS-010759 on PD-1-resistant NSCLC in an effort to overcome XRT-induced immunosuppression and maximize response to PD-1. Additionally, combined XRT and IACS-010759 promoted antitumor effects in the PD-1-resistant model, but not in the sensitive model. After elucidation of the most optimal dose/fractionation scheme of XRT with IACS-010759, the combinatorial therapy with this regimen did not increase the abscopal antitumor effect, although IACS-010549 did not decrease CD45+, CD4+, and CD8+ immune cells. Finally, triple therapy with IACS-010759, XRT, and anti-PD-1 promoted abscopal responses and prolonged survival time. CONCLUSION OXPHOS inhibition as part of a combinatorial regimen with XRT is a promising strategy to address PD-1-resistant NSCLC, and this combination is being tested clinically.
Collapse
Affiliation(s)
- Dawei Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Grant Fischer
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, United States
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Liangpeng Yang
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vivek Verma
- Department of Radiation oncology, Allegheny General Hospital, Pittsburgh, United States
| | - Ahmed I Younes
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yun Hu
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fatemeh Masropour
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Katherine Klein
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Christopher Vellano
- Translational Research to Advance Therapeutics and Innovation in Oncology Platform, The University of Texas MD Anderson Cancer Center, Houston, United States
- Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Joseph Marszalek
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, United States
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Houston, TX, United States
| | - Maria Angelica Cortez
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - James Welsh
- Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
47
|
Teh JLF, Purwin TJ, Han A, Chua V, Patel P, Baqai U, Liao C, Bechtel N, Sato T, Davies MA, Aguirre-Ghiso J, Aplin AE. Metabolic Adaptations to MEK and CDK4/6 Cotargeting in Uveal Melanoma. Mol Cancer Ther 2020; 19:1719-1726. [PMID: 32430489 DOI: 10.1158/1535-7163.mct-19-1016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/19/2020] [Accepted: 05/15/2020] [Indexed: 12/25/2022]
Abstract
Frequent GNAQ and GNA11 mutations in uveal melanoma hyperactivate the MEK-ERK signaling pathway, leading to aberrant regulation of cyclin-dependent kinases (CDK) and cell-cycle progression. MEK inhibitors (MEKi) alone show poor efficacy in uveal melanoma, raising the question of whether downstream targets can be vertically inhibited to provide long-term benefit. CDK4/6 selective inhibitors are FDA-approved in patients with estrogen receptor (ER)-positive breast cancer in combination with ER antagonists/aromatase inhibitors. We determined the effects of MEKi plus CDK4/6 inhibitors (CDK4/6i) in uveal melanoma. In vitro, palbociclib, a CDK4/6i, enhanced the effects of MEKi via downregulation of cell-cycle proteins. In contrast, in vivo CDK4/6 inhibition alone led to cytostasis and was as effective as MEKi plus CDK4/6i treatment at delaying tumor growth. RNA sequencing revealed upregulation of the oxidative phosphorylation (OxPhos) pathway in both MEKi-resistant tumors and CDK4/6i-tolerant tumors. Furthermore, oxygen consumption rate was increased following MEKi + CDK4/6i treatment. IACS-010759, an OxPhos inhibitor, decreased uveal melanoma cell survival in combination with MEKi + CDK4/6i. These data highlight adaptive upregulation of OxPhos in response to MEKi + CDK4/6i treatment in uveal melanoma and suggest that suppression of this metabolic state may improve the efficacy of MEKi plus CDK4/6i combinations.
Collapse
Affiliation(s)
| | | | - Anna Han
- Department of Cancer Biology, Philadelphia, Pennsylvania
| | - Vivian Chua
- Department of Cancer Biology, Philadelphia, Pennsylvania
| | - Prem Patel
- Department of Cancer Biology, Philadelphia, Pennsylvania
| | - Usman Baqai
- Department of Cancer Biology, Philadelphia, Pennsylvania
| | - Connie Liao
- Department of Cancer Biology, Philadelphia, Pennsylvania
| | - Nelisa Bechtel
- Department of Cancer Biology, Philadelphia, Pennsylvania
| | - Takami Sato
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Julio Aguirre-Ghiso
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Oncological Sciences and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Andrew E Aplin
- Department of Cancer Biology, Philadelphia, Pennsylvania. .,Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|