232 results on '"Gabrilovich DI"'
Search Results
2. Safety, pharmacokinetics, and pharmacodynamics of oral omaveloxolone (RTA 408), a synthetic triterpenoid, in a first-in-human trial of patients with advanced solid tumors
- Author
-
Creelan BC, Gabrilovich DI, Gray JE, Williams CC, Tanvetyanon T, Haura EB, Weber JS, Gibney GT, Markowitz J, Proksch JW, Reisman SA, McKee MD, Chin MP, Meyer CJ, and Antonia SJ
- Subjects
Bardoxolone methyl ,Nitrotyrosine ,Non-small cell lung cancer ,Myeloid-derived suppressor cells ,Antioxidant inflammation modulator ,Nitric oxide synthase ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Melanoma ,lcsh:RC254-282 ,Immuno-Oncology - Abstract
Ben C Creelan,1 Dmitry I Gabrilovich,2 Jhanelle E Gray,1 Charles C Williams,1 Tawee Tanvetyanon,1 Eric B Haura,1 Jeffrey S Weber,3 Geoffrey T Gibney,4 Joseph Markowitz,5 Joel W Proksch,6 Scott A Reisman,6 Mark D McKee,7 Melanie P Chin,6 Colin J Meyer,6 Scott J Antonia11Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Magnolia Drive, Tampa, FL, USA; 2The Wistar Institute, Philadelphia, PA, USA; 3Laura and Isaac Perlmutter Cancer Center, New York, NY, USA; 4Department of Medicine, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC, USA; 5Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Magnolia Drive, Tampa, FL, USA; 6Reata Pharmaceuticals, Inc., Irving, TX, USA; 7AbbVie, Inc., North Chicago, IL, USABackground: Omaveloxolone is a semisynthetic oleanane triterpenoid that potently activates Nrf2 with subsequent antioxidant function. Weconducted a first-in-human Phase I clinical trial (NCT02029729) with the primary objectives to determine the appropriate dose for Phase II studies, characterize pharmacokinetic and pharmacodynamic parameters, and assess antitumor activity.Methods: Omaveloxolone was administered orally once daily continuously in a 28-day cycle for patients with stage 4 relapsed/refractory melanoma or non-small cell lung cancer. An accelerated titration design was employed until a grade 2-related adverse event (AE) occurred. A standard 3+3 dose escalation was employed. Single-dose and steady-state plasma pharmacokinetics of the drug were characterized. Downstream Nrf2 activation was assessed in peripheral blood mononuclear cells by quantification of target gene mRNA expression.Results: Omaveloxolone was tested at four dose levels up to 15 mg given orally once daily. No dose-limiting toxicities were detected, and the maximum tolerated dose was not determined. All drug-related AEs were either grade 1 or 2 in severity, and none required clinical action. The most common drug-related AEs were elevated alkaline phosphatase (18%) and anemia (18%). No drug interruptions or reductions were required. Omaveloxolone was rapidly absorbed and exhibited proportional increases in exposure across dose levels. With some exceptions, an overall trend toward time-dependent and dose-dependent activation of Nrf2 antioxidant genes was observed. No confirmed radiologic responses were seen, although one lung cancer subject did have stable disease exceeding 1 year.Conclusions: Omaveloxolone has favorable tolerability at biologically active doses, although this trial had a small sample size which limits definitive conclusions. These findings support further investigation of omaveloxolone in cancer.Keywords: antioxidant inflammation modulator, nitrotyrosine, nitric oxide synthase, melanoma, non-small cell lung cancer, immuno-oncology, myeloid-derived suppressor cells, bardoxolone methyl
- Published
- 2017
3. Abstract BS1-2: Myeloid-derived suppressor cells: many facets of regulation of tumor progression
- Author
-
Gabrilovich, DI, primary
- Published
- 2018
- Full Text
- View/download PDF
4. Lectin-type oxidized LDL receptor 1 defines a population of polymorphonuclear myeloid-derived suppressor cells in cancer patients
- Author
-
Condamine, T, Dominguez, GA, Youn, JI, Kossenkov, A, Mony, S, Alicea-Torres, K, Tsyganov, E, Hashimoto, A, Nefedova, Y, Lin, C, Partlova, S, Garfall, A, Vogl, DT, Xu, X, Knight, SC, Malietzsis, G, Lee, GH, Eruslanov, E, Albelda, SM, Wang, X, Mehta, JL, Bewtra, M, Rustgi, A, Hockstein, N, Witt, R, Masters, G, Nam, B, Smirnov, D, Sepulveda, MA, Gabrilovich, DI, Biotechnology and Biological Sciences Research Cou, and Biotechnology and Biological Sciences Research Council
- Abstract
Polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) are important regulators of immune responses in cancer and have been directly implicated in the promotion of tumor progression. However, the heterogeneity of these cells and the lack of distinct markers hamper the progress in understanding the biology and clinical importance of these cells. Using partial enrichment of PMN-MDSC with gradient centrifugation, we determined that low-density PMN-MDSC and high-density neutrophils from the same cancer patients had a distinct gene profile. The most prominent changes were observed in the expression of genes associated with endoplasmic reticulum (ER) stress. Unexpectedly, low-density lipoprotein (LDL) was one of the most increased regulators, and its receptor oxidized LDL receptor 1 (OLR1) was one of the most overexpressed genes in PMN-MDSC. Lectin-type oxidized LDL receptor-1 (LOX-1) encoded by OLR1 was practically undetectable in neutrophils in peripheral blood of healthy donors, whereas 5 to 15% of total neutrophils in cancer patients and 15 to 50% of neutrophils in tumor tissues were LOX-1+. In contrast to their LOX-1− counterparts, LOX-1+ neutrophils had gene signature, potent immunosuppressive activity, up-regulation of ER stress, and other biochemical characteristics of PMN-MDSCs. Moreover, induction of ER stress in neutrophils from healthy donors up-regulated LOX-1 expression and converted these cells to suppressive PMN-MDSCs. Thus, we identified a specific marker of human PMN-MDSC associated with ER stress and lipid metabolism, which provides new insights into the biology and potential therapeutic targeting of these cells.
- Published
- 2016
5. [Untitled]
- Author
-
Kolesiak K, Young Mr, Wright Ma, and Gabrilovich Di
- Subjects
Cancer Research ,Matrigel ,Pathology ,medicine.medical_specialty ,CD34 ,Lewis lung carcinoma ,General Medicine ,Biology ,Endothelial stem cell ,medicine.anatomical_structure ,Oncology ,Cancer research ,Myeloid-derived Suppressor Cell ,medicine ,Bone marrow ,Stem cell ,Progenitor cell - Abstract
Patients and animals with GM-CSF-producing tumors have an increased number of mobilized CD34+ progenitor cells within their peripheral blood and tumor tissue. These CD34+ cells are inhibitory to the activity of intratumoral T-cells. The present study used the murine Lewis lung carcinoma (LLC) model to assess mechanisms that could lead to the accumulation of CD34+ cells within the tumor tissue. In vitro analyses showed that LLC tumor explants released chemoattractants for normal femoral CD34+ cells. The LLC tumor cells contributed to the production of this activity since CD34+ cell chemoattractants were also released by cultured LLC cells. Antibody neutralization studies showed that most, although not all, of the chemotactic activity that was produced by LLC cells could be attributed to VEGF. In vivo studies with fluorescent-tagged CD34+ cells showed their accumulation within the tumor tissue, but not within the lungs, spleen or bone marrow, suggesting a selective accumulation within the tumor. Whether or not VEGF could chemoattract CD34+ cells in vivo was measured with a VEGF-containing Matrigel plug assay. Infusion of fluorescent-tagged CD34+ cells into mice after the plugs became vascularized revealed the accumulation of fluorescent-tagged cells within the plugs. However, these CD34+ cells failed to accumulate within the VEGF-containing Matrigel plugs when they were infused together with neutralizing anti-VEGF antibody. Through a combination of in vitro and in vivo analyses, the LLC cells were shown to be capable of chemoattracting CD34+ cells, with most of the tumor-derived chemotactic activity being due to tumor release of VEGF.
- Published
- 1999
6. Myeloid-derived suppressor cells in human cancer.
- Author
-
Nagaraj S and Gabrilovich DI
- Abstract
Myeloid-derived suppressor cells are one of the major factors responsible for immune suppression in cancer. They also contribute to limited efficacy of current vaccination strategies. Here, we give an overview of the myeloid-derived suppressor cells field focusing primarily on the studies in cancer patients and current and future therapeutic options targeting these cells. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
7. Combination of chemotherapy and immunotherapy for cancer: a paradigm revisited.
- Author
-
Gabrilovich DI
- Published
- 2007
- Full Text
- View/download PDF
8. A guideline on the molecular ecosystem regulating ferroptosis.
- Author
-
Dai E, Chen X, Linkermann A, Jiang X, Kang R, Kagan VE, Bayir H, Yang WS, Garcia-Saez AJ, Ioannou MS, Janowitz T, Ran Q, Gu W, Gan B, Krysko DV, Zhu X, Wang J, Krautwald S, Toyokuni S, Xie Y, Greten FR, Yi Q, Schick J, Liu J, Gabrilovich DI, Liu J, Zeh HJ, Zhang DD, Yang M, Iovanna J, Kopf M, Adolph TE, Chi JT, Li C, Ichijo H, Karin M, Sankaran VG, Zou W, Galluzzi L, Bush AI, Li B, Melino G, Baehrecke EH, Lotze MT, Klionsky DJ, Stockwell BR, Kroemer G, and Tang D
- Subjects
- Humans, Animals, Signal Transduction, Reactive Oxygen Species metabolism, Iron metabolism, Ferroptosis genetics, Lipid Peroxidation
- Abstract
Ferroptosis, an intricately regulated form of cell death characterized by uncontrolled lipid peroxidation, has garnered substantial interest since this term was first coined in 2012. Recent years have witnessed remarkable progress in elucidating the detailed molecular mechanisms that govern ferroptosis induction and defence, with particular emphasis on the roles of heterogeneity and plasticity. In this Review, we discuss the molecular ecosystem of ferroptosis, with implications that may inform and enable safe and effective therapeutic strategies across a broad spectrum of diseases., (© 2024. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
9. Defining myeloid-derived suppressor cells.
- Author
-
Akkari L, Amit I, Bronte V, Fridlender ZG, Gabrilovich DI, Ginhoux F, Hedrick CC, and Ostrand-Rosenberg S
- Published
- 2024
- Full Text
- View/download PDF
10. International consensus guidelines for the definition, detection, and interpretation of autophagy-dependent ferroptosis.
- Author
-
Chen X, Tsvetkov AS, Shen HM, Isidoro C, Ktistakis NT, Linkermann A, Koopman WJH, Simon HU, Galluzzi L, Luo S, Xu D, Gu W, Peulen O, Cai Q, Rubinsztein DC, Chi JT, Zhang DD, Li C, Toyokuni S, Liu J, Roh JL, Dai E, Juhasz G, Liu W, Zhang J, Yang M, Liu J, Zhu LQ, Zou W, Piacentini M, Ding WX, Yue Z, Xie Y, Petersen M, Gewirtz DA, Mandell MA, Chu CT, Sinha D, Eftekharpour E, Zhivotovsky B, Besteiro S, Gabrilovich DI, Kim DH, Kagan VE, Bayir H, Chen GC, Ayton S, Lünemann JD, Komatsu M, Krautwald S, Loos B, Baehrecke EH, Wang J, Lane JD, Sadoshima J, Yang WS, Gao M, Münz C, Thumm M, Kampmann M, Yu D, Lipinski MM, Jones JW, Jiang X, Zeh HJ, Kang R, Klionsky DJ, Kroemer G, and Tang D
- Subjects
- Humans, Animals, Consensus, Ferroptosis physiology, Autophagy physiology
- Abstract
Macroautophagy/autophagy is a complex degradation process with a dual role in cell death that is influenced by the cell types that are involved and the stressors they are exposed to. Ferroptosis is an iron-dependent oxidative form of cell death characterized by unrestricted lipid peroxidation in the context of heterogeneous and plastic mechanisms. Recent studies have shed light on the involvement of specific types of autophagy (e.g. ferritinophagy, lipophagy, and clockophagy) in initiating or executing ferroptotic cell death through the selective degradation of anti-injury proteins or organelles. Conversely, other forms of selective autophagy (e.g. reticulophagy and lysophagy) enhance the cellular defense against ferroptotic damage. Dysregulated autophagy-dependent ferroptosis has implications for a diverse range of pathological conditions. This review aims to present an updated definition of autophagy-dependent ferroptosis, discuss influential substrates and receptors, outline experimental methods, and propose guidelines for interpreting the results. Abbreviation : 3-MA:3-methyladenine; 4HNE: 4-hydroxynonenal; ACD: accidentalcell death; ADF: autophagy-dependentferroptosis; ARE: antioxidant response element; BH2:dihydrobiopterin; BH4: tetrahydrobiopterin; BMDMs: bonemarrow-derived macrophages; CMA: chaperone-mediated autophagy; CQ:chloroquine; DAMPs: danger/damage-associated molecular patterns; EMT,epithelial-mesenchymal transition; EPR: electronparamagnetic resonance; ER, endoplasmic reticulum; FRET: Försterresonance energy transfer; GFP: green fluorescent protein;GSH: glutathione;IF: immunofluorescence; IHC: immunohistochemistry; IOP, intraocularpressure; IRI: ischemia-reperfusion injury; LAA: linoleamide alkyne;MDA: malondialdehyde; PGSK: Phen Green™ SK;RCD: regulatedcell death; PUFAs: polyunsaturated fatty acids; RFP: red fluorescentprotein;ROS: reactive oxygen species; TBA: thiobarbituricacid; TBARS: thiobarbituric acid reactive substances; TEM:transmission electron microscopy.
- Published
- 2024
- Full Text
- View/download PDF
11. The ATR inhibitor ceralasertib potentiates cancer checkpoint immunotherapy by regulating the tumor microenvironment.
- Author
-
Hardaker EL, Sanseviero E, Karmokar A, Taylor D, Milo M, Michaloglou C, Hughes A, Mai M, King M, Solanki A, Magiera L, Miragaia R, Kar G, Standifer N, Surace M, Gill S, Peter A, Talbot S, Tohumeken S, Fryer H, Mostafa A, Mulgrew K, Lam C, Hoffmann S, Sutton D, Carnevalli L, Calero-Nieto FJ, Jones GN, Pierce AJ, Wilson Z, Campbell D, Nyoni L, Martins CP, Baker T, Serrano de Almeida G, Ramlaoui Z, Bidar A, Phillips B, Boland J, Iyer S, Barrett JC, Loembé AB, Fuchs SY, Duvvuri U, Lou PJ, Nance MA, Gomez Roca CA, Cadogan E, Critichlow SE, Fawell S, Cobbold M, Dean E, Valge-Archer V, Lau A, Gabrilovich DI, and Barry ST
- Subjects
- Humans, Animals, Mice, B7-H1 Antigen, Tumor Microenvironment, Cell Line, Tumor, Immunotherapy, Disease Models, Animal, Ataxia Telangiectasia Mutated Proteins, CD8-Positive T-Lymphocytes, Neoplasms, Indoles, Morpholines, Pyrimidines, Sulfonamides
- Abstract
The Ataxia telangiectasia and Rad3-related (ATR) inhibitor ceralasertib in combination with the PD-L1 antibody durvalumab demonstrated encouraging clinical benefit in melanoma and lung cancer patients who progressed on immunotherapy. Here we show that modelling of intermittent ceralasertib treatment in mouse tumor models reveals CD8
+ T-cell dependent antitumor activity, which is separate from the effects on tumor cells. Ceralasertib suppresses proliferating CD8+ T-cells on treatment which is rapidly reversed off-treatment. Ceralasertib causes up-regulation of type I interferon (IFNI) pathway in cancer patients and in tumor-bearing mice. IFNI is experimentally found to be a major mediator of antitumor activity of ceralasertib in combination with PD-L1 antibody. Improvement of T-cell function after ceralasertib treatment is linked to changes in myeloid cells in the tumor microenvironment. IFNI also promotes anti-proliferative effects of ceralasertib on tumor cells. Here, we report that broad immunomodulatory changes following intermittent ATR inhibition underpins the clinical therapeutic benefit and indicates its wider impact on antitumor immunity., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
12. S100A8/A9-alarmin promotes local myeloid-derived suppressor cell activation restricting severe autoimmune arthritis.
- Author
-
von Wulffen M, Luehrmann V, Robeck S, Russo A, Fischer-Riepe L, van den Bosch M, van Lent P, Loser K, Gabrilovich DI, Hermann S, Roth J, and Vogl T
- Subjects
- Animals, Mice, T-Lymphocytes cytology, T-Lymphocytes immunology, Disease Models, Animal, Cell Differentiation, Nitric Oxide metabolism, Signal Transduction, Toll-Like Receptor 4 metabolism, Arthritis immunology, Arthritis metabolism, Arthritis pathology, Myeloid-Derived Suppressor Cells cytology, Myeloid-Derived Suppressor Cells immunology, Calgranulin A metabolism, Calgranulin B metabolism
- Abstract
Immune-suppressive effects of myeloid-derived suppressor cells (MDSCs) are well characterized during anti-tumor immunity. The complex mechanisms promoting MDSC development and their regulatory effects during autoimmune diseases are less understood. We demonstrate that the endogenous alarmin S100A8/A9 reprograms myeloid cells to a T cell suppressing phenotype during autoimmune arthritis. Treatment of myeloid precursors with S100-alarmins during differentiation induces MDSCs in a Toll-like receptor 4-dependent manner. Consequently, knockout of S100A8/A9 aggravates disease activity in collagen-induced arthritis due to a deficit of MDSCs in local lymph nodes, which could be corrected by adoptive transfer of S100-induced MDSCs. Blockade of MDSC function in vivo aggravates disease severity in arthritis. Therapeutic application of S100A8 induces MDSCs in vivo and suppresses the inflammatory phenotype of S100A9ko mice. Accordingly, the interplay of T cell-mediated autoimmunity with a defective innate immune regulation is crucial for autoimmune arthritis, which should be considered for future innovative therapeutic options., Competing Interests: Declaration of interests Dmitry I. Gabrilovich is an employee and shareholder of AstraZeneca., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
13. Ferroptosis of immune cells in the tumor microenvironment.
- Author
-
Kim R, Taylor D, Vonderheide RH, and Gabrilovich DI
- Subjects
- Humans, Tumor Microenvironment, Cell Death, Lipid Metabolism, Ferroptosis, Neoplasms drug therapy
- Abstract
Ferroptosis is a distinct form of cell death driven by the accumulation of peroxidized lipids. Characterized by alterations in redox lipid metabolism, ferroptosis has been implicated in a variety of cellular processes, including cancer. Induction of ferroptosis is considered a novel way to kill tumor cells, especially cells resistant to radiation and chemotherapy. However, in recent years, a new paradigm has emerged. In addition to promoting tumor cell death, ferroptosis causes potent immune suppression in the tumor microenvironment (TME) by affecting both innate and adaptive immune responses. In this review, we discuss the dual role of ferroptosis in the antitumor and protumorigenic functions of immune cells in cancer. We suggest strategies for targeting ferroptosis, taking into account its ambiguous role in cancer., Competing Interests: Declaration of interests D.T. and D.I.G. are employees of and stakeholders in AstraZeneca., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
14. Cell-Autonomous Cxcl1 Sustains Tolerogenic Circuitries and Stromal Inflammation via Neutrophil-Derived TNF in Pancreatic Cancer.
- Author
-
Bianchi A, De Castro Silva I, Deshpande NU, Singh S, Mehra S, Garrido VT, Guo X, Nivelo LA, Kolonias DS, Saigh SJ, Wieder E, Rafie CI, Dosch AR, Zhou Z, Umland O, Amirian H, Ogobuiro IC, Zhang J, Ban Y, Shiau C, Nagathihalli NS, Montgomery EA, Hwang WL, Brambilla R, Komanduri K, Villarino AV, Toska E, Stanger BZ, Gabrilovich DI, Merchant NB, and Datta J
- Subjects
- Humans, Neutrophils, Receptors, Tumor Necrosis Factor, Type II therapeutic use, Proto-Oncogene Proteins p21(ras) genetics, Inflammation genetics, Tumor Microenvironment physiology, Chemokine CXCL1 genetics, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology
- Abstract
We have shown that KRAS-TP53 genomic coalteration is associated with immune-excluded microenvironments, chemoresistance, and poor survival in pancreatic ductal adenocarcinoma (PDAC) patients. By treating KRAS-TP53 cooperativity as a model for high-risk biology, we now identify cell-autonomous Cxcl1 as a key mediator of spatial T-cell restriction via interactions with CXCR2+ neutrophilic myeloid-derived suppressor cells in human PDAC using imaging mass cytometry. Silencing of cell-intrinsic Cxcl1 in LSL-KrasG12D/+;Trp53R172H/+;Pdx-1Cre/+(KPC) cells reprograms the trafficking and functional dynamics of neutrophils to overcome T-cell exclusion and controls tumor growth in a T cell-dependent manner. Mechanistically, neutrophil-derived TNF is a central regulator of this immunologic rewiring, instigating feed-forward Cxcl1 overproduction from tumor cells and cancer-associated fibroblasts (CAF), T-cell dysfunction, and inflammatory CAF polarization via transmembrane TNF-TNFR2 interactions. TNFR2 inhibition disrupts this circuitry and improves sensitivity to chemotherapy in vivo. Our results uncover cancer cell-neutrophil cross-talk in which context-dependent TNF signaling amplifies stromal inflammation and immune tolerance to promote therapeutic resistance in PDAC., Significance: By decoding connections between high-risk tumor genotypes, cell-autonomous inflammatory programs, and myeloid-enriched/T cell-excluded contexts, we identify a novel role for neutrophil-derived TNF in sustaining immunosuppression and stromal inflammation in pancreatic tumor microenvironments. This work offers a conceptual framework by which targeting context-dependent TNF signaling may overcome hallmarks of chemoresistance in pancreatic cancer. This article is highlighted in the In This Issue feature, p. 1275., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
15. Therapeutic targeting of tumour myeloid cells.
- Author
-
Barry ST, Gabrilovich DI, Sansom OJ, Campbell AD, and Morton JP
- Subjects
- Humans, Myeloid Cells, Immunotherapy, Neutrophils, Tumor Microenvironment, Neoplasms drug therapy, Myeloid-Derived Suppressor Cells
- Abstract
Myeloid cells are pivotal within the immunosuppressive tumour microenvironment. The accumulation of tumour-modified myeloid cells derived from monocytes or neutrophils - termed 'myeloid-derived suppressor cells' - and tumour-associated macrophages is associated with poor outcome and resistance to treatments such as chemotherapy and immune checkpoint inhibitors. Unfortunately, there has been little success in large-scale clinical trials of myeloid cell modulators, and only a few distinct strategies have been used to target suppressive myeloid cells clinically so far. Preclinical and translational studies have now elucidated specific functions for different myeloid cell subpopulations within the tumour microenvironment, revealing context-specific roles of different myeloid cell populations in disease progression and influencing response to therapy. To improve the success of myeloid cell-targeted therapies, it will be important to target tumour types and patient subsets in which myeloid cells represent the dominant driver of therapy resistance, as well as to determine the most efficacious treatment regimens and combination partners. This Review discusses what we can learn from work with the first generation of myeloid modulators and highlights recent developments in modelling context-specific roles for different myeloid cell subtypes, which can ultimately inform how to drive more successful clinical trials., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
16. S100A8/S100A9 Promote Progression of Multiple Myeloma via Expansion of Megakaryocytes.
- Author
-
Lin C, Garcia-Gerique L, Bonner EE, Mastio J, Rosenwasser M, Cruz Z, Lawler M, Bernabei L, Muthumani K, Liu Q, Poncz M, Vogl T, Törngren M, Eriksson H, Vogl DT, Gabrilovich DI, and Nefedova Y
- Subjects
- Humans, Megakaryocytes metabolism, Lenalidomide, Proteasome Inhibitors, Calgranulin B metabolism, Calgranulin A metabolism, Tumor Microenvironment, Multiple Myeloma drug therapy
- Abstract
Multiple myeloma is characterized by clonal proliferation of plasma cells that accumulate preferentially in the bone marrow (BM). The tumor microenvironment is one of the leading factors that promote tumor progression. Neutrophils and monocytes are a major part of the BM tumor microenvironment, but the mechanism of their contribution to multiple myeloma progression remains unclear. Here, we describe a novel mechanism by which S100A8/S100A9 proteins produced by BM neutrophils and monocytes promote the expansion of megakaryocytes supporting multiple myeloma progression. S100A8/S100A9 alone was not sufficient to drive megakaryopoiesis but markedly enhanced the effect of thrombopoietin, an effect that was mediated by Toll-like receptor 4 and activation of the STAT5 transcription factor. Targeting S100A9 with tasquinimod as a single agent and in combination with lenalidomide and with proteasome inhibitors has potent antimyeloma effect that is at least partly independent of the adaptive immune system. This newly identified axis of signaling involving myeloid cells and megakaryocytes may provide a new avenue for therapeutic targeting in multiple myeloma., Significance: We identified a novel mechanism by which myeloid cells promote myeloma progression independently of the adaptive immune system. Specifically, we discovered a novel role of S100A8/S100A9, the most abundant proteins produced by neutrophils and monocytes, in regulation of myeloma progression via promotion of the megakaryocyte expansion and angiogenesis. Tasquinimod, an inhibitor of S100A9, has potent antimyeloma effects as a single agent and in combination with lenalidomide and with proteasome inhibitors., (© 2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
17. Functional states of myeloid cells in cancer.
- Author
-
van Vlerken-Ysla L, Tyurina YY, Kagan VE, and Gabrilovich DI
- Subjects
- Humans, Myeloid Cells, Macrophages pathology, Monocytes pathology, Tumor Microenvironment, Neoplasms therapy, Myeloid-Derived Suppressor Cells pathology
- Abstract
Myeloid cells, comprised of macrophages, dendritic cells, monocytes, and granulocytes, represent a major component of the tumor microenvironment (TME) and are critically involved in regulation of tumor progression and metastasis. In recent years, single-cell omics technologies have identified multiple phenotypically distinct subpopulations. In this review, we discuss recent data and concepts suggesting that the biology of myeloid cells is largely defined by a very limited number of functional states that transcend the narrowly defined cell populations. These functional states are primarily centered around classical and pathological states of activation, with the latter state commonly defined as myeloid-derived suppressor cells. We discuss the concept that lipid peroxidation of myeloid cells represents a major mechanism that governs their pathological state of activation in the TME. Lipid peroxidation is associated with ferroptosis mediating suppressive activity of these cells and thus could be considered an attractive target for therapeutic intervention., Competing Interests: Declaration of interests L.v.V.-Y. and D.G. are employee and shareholders of AstraZeneca., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. Syntaphilin Regulates Neutrophil Migration in Cancer.
- Author
-
Fu S, Deng H, Bertolini I, Perego M, Chen ES, Sanseviero E, Mostafa A, Alicea-Torres K, Garcia-Gerique L, Stone EL, Kossenkov AV, Schug ZT, Nam B, Mulligan C, Altieri DC, Nefedova Y, and Gabrilovich DI
- Subjects
- Animals, Mice, Cell Movement, Neutrophils metabolism, Membrane Proteins metabolism, Myeloid-Derived Suppressor Cells metabolism, Neoplasms pathology, Nerve Tissue Proteins
- Abstract
Pathologically activated neutrophils (PMN) with immunosuppressive activity, which are termed myeloid-derived suppressor cells (PMN-MDSC), play a critical role in regulating tumor progression. These cells have been implicated in promoting tumor metastases by contributing to premetastatic niche formation. This effect was facilitated by enhanced spontaneous migration of PMN from bone marrow to the premetastatic niches during the early-stage of cancer development. The molecular mechanisms underpinning this phenomenon remained unclear. In this study, we found that syntaphilin (SNPH), a cytoskeletal protein previously known for anchoring mitochondria to the microtubule in neurons and tumor cells, could regulate migration of PMN. Expression of SNPH was decreased in PMN from tumor-bearing mice and patients with cancer as compared with PMN from tumor-free mice and healthy donors, respectively. In Snph-knockout (SNPH-KO) mice, spontaneous migration of PMN was increased and the mice showed increased metastasis. Mechanistically, in SNPH-KO mice, the speed and distance travelled by mitochondria in PMN was increased, rates of oxidative phosphorylation and glycolysis were elevated, and generation of adenosine was increased. Thus, our study reveals a molecular mechanism regulating increased migratory activity of PMN during cancer progression and suggests a novel therapeutic targeting opportunity., (©2022 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
19. Protection of Regulatory T Cells from Fragility and Inactivation in the Tumor Microenvironment.
- Author
-
Zhang H, Tomar VS, Li J, Basavaraja R, Yan F, Gui J, McBrearty N, Costich TL, Beiting DP, Blanco MA, Conejo-Garcia JR, Saggu G, Berger A, Nefedova Y, Gabrilovich DI, and Fuchs SY
- Subjects
- Humans, Tumor Microenvironment, Neuropilin-1, Immunotherapy, T-Lymphocytes, Regulatory, Neoplasms
- Abstract
Fragility of regulatory T (Treg) cells manifested by the loss of neuropilin-1 (NRP1) and expression of IFNγ undermines the immune suppressive functions of Treg cells and contributes to the success of immune therapies against cancers. Intratumoral Treg cells somehow avoid fragility; however, the mechanisms by which Treg cells are protected from fragility in the tumor microenvironment are not well understood. Here, we demonstrate that the IFNAR1 chain of the type I IFN (IFN1) receptor was downregulated on intratumoral Treg cells. Downregulation of IFNAR1 mediated by p38α kinase protected Treg cells from fragility and maintained NRP1 levels, which were decreased in response to IFN1. Genetic or pharmacologic inactivation of p38α and stabilization of IFNAR1 in Treg cells induced fragility and inhibited their immune suppressive and protumorigenic activities. The inhibitor of sumoylation TAK981 (Subasumstat) upregulated IFNAR1, eliciting Treg fragility and inhibiting tumor growth in an IFNAR1-dependent manner. These findings describe a mechanism by which intratumoral Treg cells retain immunosuppressive activities and suggest therapeutic approaches for inducing Treg fragility and increasing the efficacy of immunotherapies., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
20. Ferroptosis of tumour neutrophils causes immune suppression in cancer.
- Author
-
Kim R, Hashimoto A, Markosyan N, Tyurin VA, Tyurina YY, Kar G, Fu S, Sehgal M, Garcia-Gerique L, Kossenkov A, Gebregziabher BA, Tobias JW, Hicks K, Halpin RA, Cvetesic N, Deng H, Donthireddy L, Greenberg A, Nam B, Vonderheide RH, Nefedova Y, Kagan VE, and Gabrilovich DI
- Subjects
- Humans, Mice, Animals, Tumor Microenvironment, Neoplasms
- Abstract
Ferroptosis is a non-apoptotic form of regulated cell death that is triggered by the discoordination of regulatory redox mechanisms culminating in massive peroxidation of polyunsaturated phospholipids. Ferroptosis inducers have shown considerable effectiveness in killing tumour cells in vitro, yet there has been no obvious success in experimental animal models, with the notable exception of immunodeficient mice
1,2 . This suggests that the effect of ferroptosis on immune cells remains poorly understood. Pathologically activated neutrophils (PMNs), termed myeloid-derived suppressor cells (PMN-MDSCs), are major negative regulators of anti-tumour immunity3-5 . Here we found that PMN-MDSCs in the tumour microenvironment spontaneously die by ferroptosis. Although decreasing the presence of PMN-MDSCs, ferroptosis induces the release of oxygenated lipids and limits the activity of human and mouse T cells. In immunocompetent mice, genetic and pharmacological inhibition of ferroptosis abrogates suppressive activity of PMN-MDSCs, reduces tumour progression and synergizes with immune checkpoint blockade to suppress the tumour growth. By contrast, induction of ferroptosis in immunocompetent mice promotes tumour growth. Thus, ferroptosis is a unique and targetable immunosuppressive mechanism of PMN-MDSCs in the tumour microenvironment that can be pharmacologically modulated to limit tumour progression., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
21. Mechanisms regulating transitory suppressive activity of neutrophils in newborns: PMNs-MDSCs in newborns.
- Author
-
Perego M, Fu S, Cao Y, Kossenkov A, Yao M, Bonner E, Alicea-Torres K, Liu W, Jiang Z, Chen Z, Fuchs SY, Zhou J, and Gabrilovich DI
- Subjects
- Mice, Animals, Neutrophils, Lactoferrin metabolism, NF-kappa B metabolism, Cytokines metabolism, Lipoproteins, LDL metabolism, Myeloid-Derived Suppressor Cells
- Abstract
Transitory appearance of immune suppressive polymorphonuclear neutrophils (PMNs) defined as myeloid-derived suppressor cells (PMNs-MDSCs) in newborns is important for their protection from inflammation associated with newly established gut microbiota. Here, we report that inhibition of the type I IFN (IFN1) pathway played a major role in regulation of PMNs-MDSCs-suppressive activity during first weeks of life. Expression of the IFN1 receptor IFNAR1 was markedly lower in PMNs-MDSCs. However, in newborn mice, down-regulation of IFNAR1 was not sufficient to render PMNs immune suppressive. That also required the presence of a positive signal from lactoferrin via its receptor low-density lipoprotein receptor-related protein 2. The latter effect was mediated via NF-κB activation, which was tempered by IFN1 in a manner that involved suppressor of cytokine signaling 3. Thus, we discovered a mechanism of tight regulation of immune suppressive PMNs-MDSCs in newborns, which may be used in the development of therapies of neonatal pathologies., (©2022 Society for Leukocyte Biology.)
- Published
- 2022
- Full Text
- View/download PDF
22. PPT1 inhibition enhances the antitumor activity of anti-PD-1 antibody in melanoma.
- Author
-
Sharma G, Ojha R, Noguera-Ortega E, Rebecca VW, Attanasio J, Liu S, Piao S, Lee JJ, Nicastri MC, Harper SL, Ronghe A, Jain V, Winkler JD, Speicher DW, Mastio J, Gimotty PA, Xu X, Wherry EJ, Gabrilovich DI, and Amaravadi RK
- Published
- 2022
- Full Text
- View/download PDF
23. Peroxynitrite in the tumor microenvironment changes the profile of antigens allowing escape from cancer immunotherapy.
- Author
-
Tcyganov EN, Sanseviero E, Marvel D, Beer T, Tang HY, Hembach P, Speicher DW, Zhang Q, Donthireddy LR, Mostafa A, Tsyganova S, Pisarev V, Laufer T, Ignatov D, Ferrone S, Meyer C, Maby-El Hajjami H, Speiser DE, Altiok S, Antonia S, Xu X, Xu W, Zheng C, Schuchter LM, Amaravadi RK, Mitchell TC, Karakousis GC, Yuan Z, Montaner LJ, Celis E, and Gabrilovich DI
- Subjects
- Animals, Antigens, Neoplasm metabolism, Epitopes, Histocompatibility Antigens Class I metabolism, Immunotherapy, Mice, Oxidants metabolism, Peptides, Peroxynitrous Acid metabolism, T-Lymphocytes, Cytotoxic, Melanoma metabolism, Tumor Microenvironment
- Abstract
Cancer immunotherapy often depends on recognition of peptide epitopes by cytotoxic T lymphocytes (CTLs). The tumor microenvironment (TME) is enriched for peroxynitrite (PNT), a potent oxidant produced by infiltrating myeloid cells and some tumor cells. We demonstrate that PNT alters the profile of MHC class I bound peptides presented on tumor cells. Only CTLs specific for PNT-resistant peptides have a strong antitumor effect in vivo, whereas CTLs specific for PNT-sensitive peptides are not effective. Therapeutic targeting of PNT in mice reduces resistance of tumor cells to CTLs. Melanoma patients with low PNT activity in their tumors demonstrate a better clinical response to immunotherapy than patients with high PNT activity. Our data suggest that intratumoral PNT activity should be considered for the design of neoantigen-based therapy and also may be an important immunotherapeutic target., Competing Interests: Declaration of interests E.C. is a paid consultant for Oncovir, Inc. E.M., Q.Z., A.M., and D.I.G. are employees and shareholders of AstraZeneca., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. A Novel Selective Inhibitor JBI-589 Targets PAD4-Mediated Neutrophil Migration to Suppress Tumor Progression.
- Author
-
Deng H, Lin C, Garcia-Gerique L, Fu S, Cruz Z, Bonner EE, Rosenwasser M, Rajagopal S, Sadhu MN, Gajendran C, Zainuddin M, Gosu R, Sivanandhan D, Shelef MA, Nam B, Vogl DT, Gabrilovich DI, and Nefedova Y
- Subjects
- Animals, Disease Models, Animal, Immune Checkpoint Inhibitors, Mice, Neutrophils, Protein-Arginine Deiminase Type 4, Receptors, Chemokine metabolism, Extracellular Traps metabolism, Lung Neoplasms pathology
- Abstract
Neutrophils are closely involved in the regulation of tumor progression and formation of premetastatic niches. However, the mechanisms of their involvement and therapeutic regulation of these processes remain elusive. Here, we report a critical role of neutrophil peptidylarginine deiminase 4 (PAD4) in neutrophil migration in cancer. In several transplantable and genetically engineered mouse models, tumor growth was accompanied by significantly elevated enzymatic activity of neutrophil PAD4. Targeted deletion of PAD4 in neutrophils markedly decreased the intratumoral abundance of neutrophils and led to delayed growth of primary tumors and dramatically reduced lung metastases. PAD4-mediated neutrophil accumulation by regulating the expression of the major chemokine receptor CXCR2. PAD4 expression and activity as well as CXCR2 expression were significantly upregulated in neutrophils from patients with lung and colon cancers compared with healthy donors, and PAD4 and CXCR2 expression were positively correlated in neutrophils from patients with cancer. In tumor-bearing mice, pharmacologic inhibition of PAD4 with the novel PAD4 isoform-selective small molecule inhibitor JBI-589 resulted in reduced CXCR2 expression and blocked neutrophil chemotaxis. In mouse tumor models, targeted deletion of PAD4 in neutrophils or pharmacologic inhibition of PAD4 with JBI-589 reduced both primary tumor growth and lung metastases and substantially enhanced the effect of immune checkpoint inhibitors. Taken together, these results suggest a therapeutic potential of targeting PAD4 in cancer., Significance: PAD4 regulates tumor progression by promoting neutrophil migration and can be targeted with a small molecule inhibitor to suppress tumor growth and metastasis and increase efficacy of immune checkpoint blockade therapy., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
25. Myeloid-Derived Suppressor Cells and Radiotherapy.
- Author
-
Jiménez-Cortegana C, Galassi C, Klapp V, Gabrilovich DI, and Galluzzi L
- Subjects
- Humans, Tumor Microenvironment, Myeloid-Derived Suppressor Cells pathology, Neoplasms pathology
- Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of pathologically activated, mostly immature, myeloid cells that exert robust immunosuppressive functions. MDSCs expand during oncogenesis and have been linked to accelerated disease progression and resistance to treatment in both preclinical tumor models and patients with cancer. Thus, MDSCs stand out as promising targets for the development of novel immunotherapeutic regimens with superior efficacy. Here, we summarize accumulating preclinical and clinical evidence indicating that MDSCs also hamper the efficacy of radiotherapy (RT), as we critically discuss the potential of MDSC-targeting strategies as tools to achieve superior immunotherapeutic tumor control by RT in the clinic., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
26. ONP-302 Nanoparticles Inhibit Tumor Growth By Altering Tumor-Associated Macrophages And Cancer-Associated Fibroblasts.
- Author
-
Donthireddy L, Vonteddu P, Murthy T, Kwak T, Eraslan RN, Podojil JR, Elhofy A, Boyne MT 2nd, Puisis JJ, Veglia F, Singh SS, Dotiwala F, Montaner LJ, and Gabrilovich DI
- Abstract
In this study, we evaluated the ability of negatively charged bio-degradable nanoparticles, ONP- 302, to inhibit tumor growth. Therapeutic treatment with ONP-302 in vivo resulted in a marked delay in tumor growth in three different syngeneic tumor models in immunocompetent mice. ONP- 302 efficacy persisted with depletion of CD8+ T cells in immunocompetent mice and also was effective in immune deficient mice. Examination of ONP-302 effects on components of the tumor microenvironment (TME) were explored. ONP-302 treatment caused a gene expression shift in TAMs toward the pro-inflammatory M1 type and substantially inhibited the expression of genes associated with the pro-tumorigenic function of CAFs. ONP-302 also induced apoptosis in CAFs in the TME. Together, these data support further development of ONP-302 as a novel first-in- class anti-cancer therapeutic that can be used as a single-agent as well as in combination therapies for the treatment of solid tumors due to its ability to modulate the TME., Competing Interests: Competing Interests: TM, JRP, AE, MTB, JJP are employees of Cour Pharmaceuticals, RE-is employee of invivotek, DIG - is employee of AstraZeneca., (© The author(s).)
- Published
- 2022
- Full Text
- View/download PDF
27. Myeloid Cell-Derived Oxidized Lipids and Regulation of the Tumor Microenvironment.
- Author
-
Hicks KC, Tyurina YY, Kagan VE, and Gabrilovich DI
- Subjects
- Animals, Humans, Neoplasms pathology, Oxidation-Reduction, Immune Tolerance, Lipid Metabolism immunology, Myeloid Cells immunology, Myeloid Cells metabolism, Neoplasms immunology, Neoplasms metabolism, Tumor Microenvironment immunology
- Abstract
Immunosuppressive myeloid cells play a major role in cancer by negatively regulating immune responses, promoting tumor progression, and limiting the efficacy of cancer immunotherapy. Immunosuppression is mediated by various mechanisms dependent upon the type of myeloid cell involved. In recent years, a more universal mechanism of immunosuppressive activity of myeloid cells has emerged: Generation of oxidized lipids. Oxidized lipids accumulate in all types of myeloid cells and are often transferred between cells. In this review, we discuss mechanisms involved in the generation and biological role of myeloid cell-derived oxidized lipids in cancer., (©2021 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
28. Myeloid-Derived Suppressor Cells: A Propitious Road to Clinic.
- Author
-
Grover A, Sanseviero E, Timosenko E, and Gabrilovich DI
- Subjects
- Humans, Monocytes, Neutrophils, Myeloid-Derived Suppressor Cells, Neoplasms
- Abstract
Myeloid-derived suppressor cells (MDSC) are important regulators of immune responses in cancer. They represent a relatively stable form of pathologic activation of neutrophils and monocytes and are characterized by distinct transcriptional, biochemical, functional, and phenotypical features. The close association of MDSCs with clinical outcomes in cancer suggests that these cells can be an attractive target for therapeutic intervention. However, the complex nature of MDSC biology represents a substantial challenge for the development of selective therapies. Here, we discuss the mechanisms regulating MDSC development and fate and recent research advances that have demonstrated opportunities for therapeutic regulation of these cells. SIGNIFICANCE: MDSCs are attractive therapeutic targets because of their close association with negative clinical outcomes in cancer and established biology as potent immunosuppressive cells. However, the complex nature of MDSC biology presents a substantial challenge for therapeutic targeting. In this review, we discuss those challenges and possible solutions., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
29. Upregulation of C/EBPα Inhibits Suppressive Activity of Myeloid Cells and Potentiates Antitumor Response in Mice and Patients with Cancer.
- Author
-
Hashimoto A, Sarker D, Reebye V, Jarvis S, Sodergren MH, Kossenkov A, Sanseviero E, Raulf N, Vasara J, Andrikakou P, Meyer T, Huang KW, Plummer R, Chee CE, Spalding D, Pai M, Khan S, Pinato DJ, Sharma R, Basu B, Palmer D, Ma YT, Evans J, Habib R, Martirosyan A, Elasri N, Reynaud A, Rossi JJ, Cobbold M, Habib NA, and Gabrilovich DI
- Subjects
- Animals, Humans, Mice, Treatment Outcome, Tumor Cells, Cultured, Antineoplastic Agents therapeutic use, CCAAT-Enhancer-Binding Protein-alpha physiology, Carcinoma, Hepatocellular drug therapy, Liver Neoplasms drug therapy, Myeloid Cells physiology, Sorafenib therapeutic use, Up-Regulation
- Abstract
Purpose: To evaluate the mechanisms of how therapeutic upregulation of the transcription factor, CCAAT/enhancer-binding protein alpha (C/EBPα), prevents tumor progression in patients with advanced hepatocellular carcinoma (HCC) and in different mouse tumor models., Experimental Design: We conducted a phase I trial in 36 patients with HCC (NCT02716012) who received sorafenib as part of their standard care, and were given therapeutic C/EBPα small activating RNA (saRNA; MTL-CEBPA) as either neoadjuvant or adjuvant treatment. In the preclinical setting, the effects of MTL-CEBPA were assessed in several mouse models, including BNL-1ME liver cancer, Lewis lung carcinoma (LLC), and colon adenocarcinoma (MC38)., Results: MTL-CEBPA treatment caused radiologic regression of tumors in 26.7% of HCC patients with an underlying viral etiology with 3 complete responders. MTL-CEBPA treatment in those patients caused a marked decrease in peripheral blood monocytic myeloid-derived suppressor cell (M-MDSC) numbers and an overall reduction in the numbers of protumoral M2 tumor-associated macrophages (TAM). Gene and protein analysis of patient leukocytes following treatment showed CEBPA activation affected regulation of factors involved in immune-suppressive activity. To corroborate this observation, treatment of all the mouse tumor models with MTL-CEBPA led to a reversal in the suppressive activity of M-MDSCs and TAMs, but not polymorphonuclear MDSCs (PMN-MDSC). The antitumor effects of MTL-CEBPA in these tumor models showed dependency on T cells. This was accentuated when MTL-CEBPA was combined with checkpoint inhibitors or with PMN-MDSC-targeted immunotherapy., Conclusions: This report demonstrates that therapeutic upregulation of the transcription factor C/EBPα causes inactivation of immune-suppressive myeloid cells with potent antitumor responses across different tumor models and in cancer patients. MTL-CEBPA is currently being investigated in combination with pembrolizumab in a phase I/Ib multicenter clinical study (NCT04105335)., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
30. Pathway signatures derived from on-treatment tumor specimens predict response to anti-PD1 blockade in metastatic melanoma.
- Author
-
Du K, Wei S, Wei Z, Frederick DT, Miao B, Moll T, Tian T, Sugarman E, Gabrilovich DI, Sullivan RJ, Liu L, Flaherty KT, Boland GM, Herlyn M, and Zhang G
- Subjects
- Antibodies, Monoclonal therapeutic use, Biomarkers, Tumor, Humans, Immunotherapy, Antibodies, Monoclonal immunology, Melanoma genetics, Melanoma metabolism, Programmed Cell Death 1 Receptor immunology, Transcriptome
- Abstract
Both genomic and transcriptomic signatures have been developed to predict responses of metastatic melanoma to immune checkpoint blockade (ICB) therapies; however, most of these signatures are derived from pre-treatment biopsy samples. Here, we build pathway-based super signatures in pre-treatment (PASS-PRE) and on-treatment (PASS-ON) tumor specimens based on transcriptomic data and clinical information from a large dataset of metastatic melanoma treated with anti-PD1-based therapies as the training set. Both PASS-PRE and PASS-ON signatures are validated in three independent datasets of metastatic melanoma as the validation set, achieving area under the curve (AUC) values of 0.45-0.69 and 0.85-0.89, respectively. We also combine all test samples and obtain AUCs of 0.65 and 0.88 for PASS-PRE and PASS-ON signatures, respectively. When compared with existing signatures, the PASS-ON signature demonstrates more robust and superior predictive performance across all four datasets. Overall, we provide a framework for building pathway-based signatures that is highly and accurately predictive of response to anti-PD1 therapies based on on-treatment tumor specimens. This work would provide a rationale for applying pathway-based signatures derived from on-treatment tumor samples to predict patients' therapeutic response to ICB therapies., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
31. Distinct mechanisms govern populations of myeloid-derived suppressor cells in chronic viral infection and cancer.
- Author
-
Tcyganov EN, Hanabuchi S, Hashimoto A, Campbell D, Kar G, Slidel TW, Cayatte C, Landry A, Pilataxi F, Hayes S, Dougherty B, Hicks KC, Mulgrew K, Tang CA, Hu CA, Guo W, Grivennikov S, Ali MA, Beltra JC, Wherry EJ, Nefedova Y, and Gabrilovich DI
- Subjects
- Animals, Cell Line, Tumor, Chronic Disease, Endoplasmic Reticulum Stress genetics, Endoplasmic Reticulum Stress immunology, Female, Humans, Immune Tolerance genetics, Interferon-gamma immunology, Lymphocytic Choriomeningitis genetics, Lymphocytic Choriomeningitis immunology, Lymphocytic Choriomeningitis virology, Lymphocytic choriomeningitis virus classification, Lymphocytic choriomeningitis virus immunology, Lymphocytic choriomeningitis virus pathogenicity, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Myeloid-Derived Suppressor Cells classification, Myeloid-Derived Suppressor Cells metabolism, Neoplasms genetics, Neoplasms metabolism, Neoplasms, Experimental genetics, Neoplasms, Experimental immunology, Neoplasms, Experimental metabolism, Transcriptome, Virus Diseases genetics, Virus Diseases metabolism, Myeloid-Derived Suppressor Cells immunology, Neoplasms immunology, Virus Diseases immunology
- Abstract
Myeloid-derived suppressor cells (MDSCs) are major negative regulators of immune responses in cancer and chronic infections. It remains unclear if regulation of MDSC activity in different conditions is controlled by similar mechanisms. We compared MDSCs in mice with cancer and lymphocytic choriomeningitis virus (LCMV) infection. Chronic LCMV infection caused the development of monocytic MDSCs (M-MDSCs) but did not induce polymorphonuclear MDSCs (PMN-MDSCs). In contrast, both MDSC populations were present in cancer models. An acquisition of immune-suppressive activity by PMN-MDSCs in cancer was controlled by IRE1α and ATF6 pathways of the endoplasmic reticulum (ER) stress response. Abrogation of PMN-MDSC activity by blockade of the ER stress response resulted in an increase in tumor-specific immune response and reduced tumor progression. In contrast, the ER stress response was dispensable for suppressive activity of M-MDSCs in cancer and LCMV infection. Acquisition of immune-suppressive activity by M-MDSCs in spleens was mediated by IFN-γ signaling. However, it was dispensable for suppressive activity of M-MDSCs in tumor tissues. Suppressive activity of M-MDSCs in tumors was retained due to the effect of IL-6 present at high concentrations in the tumor site. These results demonstrate disease- and population-specific mechanisms of MDSC accumulation and the need for targeting different pathways to achieve inactivation of these cells.
- Published
- 2021
- Full Text
- View/download PDF
32. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity.
- Author
-
Veglia F, Sanseviero E, and Gabrilovich DI
- Subjects
- Autoimmune Diseases immunology, Autoimmune Diseases metabolism, COVID-19 immunology, COVID-19 prevention & control, COVID-19 virology, Cytokines metabolism, Humans, Monocytes metabolism, Myeloid Cells metabolism, Myeloid-Derived Suppressor Cells metabolism, Neoplasms immunology, Neoplasms metabolism, Neutrophils metabolism, SARS-CoV-2 immunology, SARS-CoV-2 physiology, Cytokines immunology, Monocytes immunology, Myeloid Cells immunology, Myeloid-Derived Suppressor Cells immunology, Neutrophils immunology
- Abstract
Myeloid-derived suppressor cells (MDSCs) are pathologically activated neutrophils and monocytes with potent immunosuppressive activity. They are implicated in the regulation of immune responses in many pathological conditions and are closely associated with poor clinical outcomes in cancer. Recent studies have indicated key distinctions between MDSCs and classical neutrophils and monocytes, and, in this Review, we discuss new data on the major genomic and metabolic characteristics of MDSCs. We explain how these characteristics shape MDSC function and could facilitate therapeutic targeting of these cells, particularly in cancer and in autoimmune diseases. Additionally, we briefly discuss emerging data on MDSC involvement in pregnancy, neonatal biology and COVID-19., (© 2021. Springer Nature Limited.)
- Published
- 2021
- Full Text
- View/download PDF
33. The Dawn of Myeloid-Derived Suppressor Cells: Identification of Arginase I as the Mechanism of Immune Suppression.
- Author
-
Gabrilovich DI
- Subjects
- Immunosuppression Therapy, Myeloid Cells immunology, T-Lymphocytes immunology, Arginase, Myeloid-Derived Suppressor Cells immunology
- Abstract
A study published in Cancer Research in 2004 by Rodriguez and colleagues identified the existence of arginase-producing myeloid cells in tumors distinct from macrophages. They demonstrated the role of arginase in negative regulation of T-cell function in vivo This was one of the first reports implicating cells, which later were named myeloid-derived suppressor cells (MDSC), in T-cell suppression in vivo and linking this effect with arginase activity and expression. This work was important in advancing the field of MDSC research and helped to bring these cells to the forefront of cancer immunology. See related article by Rodriguez et al., Cancer Res 2004;64:5839-49 ., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
34. Analysis of classical neutrophils and polymorphonuclear myeloid-derived suppressor cells in cancer patients and tumor-bearing mice.
- Author
-
Veglia F, Hashimoto A, Dweep H, Sanseviero E, De Leo A, Tcyganov E, Kossenkov A, Mulligan C, Nam B, Masters G, Patel J, Bhargava V, Wilkinson P, Smirnov D, Sepulveda MA, Singhal S, Eruslanov EB, Cristescu R, Loboda A, Nefedova Y, and Gabrilovich DI
- Subjects
- Animals, Carcinoma, Lewis Lung pathology, Carcinoma, Non-Small-Cell Lung blood, Case-Control Studies, Cell Line, Tumor, Disease Models, Animal, Female, Humans, Lung Neoplasms blood, Lymphoma pathology, Mice, Mice, Inbred C57BL, RNA-Seq, Single-Cell Analysis, Transcriptome, Carcinoma, Lewis Lung immunology, Carcinoma, Non-Small-Cell Lung immunology, Lung Neoplasms immunology, Lymphoma immunology, Neutrophils classification, Neutrophils immunology
- Abstract
In this study, using single-cell RNA-seq, cell mass spectrometry, flow cytometry, and functional analysis, we characterized the heterogeneity of polymorphonuclear neutrophils (PMNs) in cancer. We describe three populations of PMNs in tumor-bearing mice: classical PMNs, polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), and activated PMN-MDSCs with potent immune suppressive activity. In spleens of mice, PMN-MDSCs gradually replaced PMNs during tumor progression. Activated PMN-MDSCs were found only in tumors, where they were present at the very early stages of the disease. These populations of PMNs in mice could be separated based on the expression of CD14. In peripheral blood of cancer patients, we identified two distinct populations of PMNs with characteristics of classical PMNs and PMN-MDSCs. The gene signature of tumor PMN-MDSCs was similar to that in mouse activated PMN-MDSCs and was closely associated with negative clinical outcome in cancer patients. Thus, we provide evidence that PMN-MDSCs are a distinct population of PMNs with unique features and potential for selective targeting opportunities., Competing Interests: Disclosures: V. Bhargava is a full-time employee of Janssen Research and Development LLC. J. Patel and P. Wilkinson are employees of Janssen Research and Development LLC. D. Smirnov reported other from Janssen outside the submitted work and is an employee and shareholder of Janssen. M.A. Sepulveda is a full-time employee of Johnson & Johnson pharmaceuticals in Discovery Oncology. They collaborated with Dr. Gabrilovich and his team in the work included in this publication. R. Cristescu reported other from Merck during the conduct of the study and other from Merck outside the submitted work. A. Loboda is a full-time employee of Merck & Co. D.I. Gabrilovich reported other from AstraZeneca outside the submitted work and is a current employee of AstraZeneca. No other disclosures were reported., (© 2021 Veglia et al.)
- Published
- 2021
- Full Text
- View/download PDF
35. Immune suppressive activity of myeloid-derived suppressor cells in cancer requires inactivation of the type I interferon pathway.
- Author
-
Alicea-Torres K, Sanseviero E, Gui J, Chen J, Veglia F, Yu Q, Donthireddy L, Kossenkov A, Lin C, Fu S, Mulligan C, Nam B, Masters G, Denstman F, Bennett J, Hockstein N, Rynda-Apple A, Nefedova Y, Fuchs SY, and Gabrilovich DI
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Antineoplastic Agents pharmacology, Bone Marrow, Cell Line, Tumor, Disease Models, Animal, Female, Gene Expression Regulation, Neoplastic, Humans, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Middle Aged, Monocytes immunology, Neutrophils immunology, Receptor, Interferon alpha-beta genetics, p38 Mitogen-Activated Protein Kinases drug effects, p38 Mitogen-Activated Protein Kinases metabolism, Interferon Type I metabolism, Myeloid-Derived Suppressor Cells immunology, Neoplasms metabolism, Receptor, Interferon alpha-beta metabolism
- Abstract
Myeloid-derived suppressor cells (MDSC) are pathologically activated neutrophils and monocytes with potent immune suppressive activity. These cells play an important role in accelerating tumor progression and undermining the efficacy of anti-cancer therapies. The natural mechanisms limiting MDSC activity are not well understood. Here, we present evidence that type I interferons (IFN1) receptor signaling serves as a universal mechanism that restricts acquisition of suppressive activity by these cells. Downregulation of the IFNAR1 chain of this receptor is found in MDSC from cancer patients and mouse tumor models. The decrease in IFNAR1 depends on the activation of the p38 protein kinase and is required for activation of the immune suppressive phenotype. Whereas deletion of IFNAR1 is not sufficient to convert neutrophils and monocytes to MDSC, genetic stabilization of IFNAR1 in tumor bearing mice undermines suppressive activity of MDSC and has potent antitumor effect. Stabilizing IFNAR1 using inhibitor of p38 combined with the interferon induction therapy elicits a robust anti-tumor effect. Thus, negative regulatory mechanisms of MDSC function can be exploited therapeutically.
- Published
- 2021
- Full Text
- View/download PDF
36. Entinostat plus Pembrolizumab in Patients with Metastatic NSCLC Previously Treated with Anti-PD-(L)1 Therapy.
- Author
-
Hellmann MD, Jänne PA, Opyrchal M, Hafez N, Raez LE, Gabrilovich DI, Wang F, Trepel JB, Lee MJ, Yuno A, Lee S, Brouwer S, Sankoh S, Wang L, Tamang D, Schmidt EV, Meyers ML, Ramalingam SS, Shum E, and Ordentlich P
- Subjects
- Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized adverse effects, Antibodies, Monoclonal, Humanized pharmacology, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacology, B7-H1 Antigen antagonists & inhibitors, Benzamides adverse effects, Benzamides pharmacology, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung mortality, Carcinoma, Non-Small-Cell Lung pathology, Drug Resistance, Neoplasm immunology, Female, Follow-Up Studies, Humans, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Lung Neoplasms immunology, Lung Neoplasms mortality, Lung Neoplasms pathology, Male, Middle Aged, Neoplasm Recurrence, Local immunology, Neoplasm Recurrence, Local mortality, Neoplasm Recurrence, Local pathology, Progression-Free Survival, Pyridines adverse effects, Pyridines pharmacology, Antibodies, Monoclonal, Humanized administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Benzamides administration & dosage, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Neoplasm Recurrence, Local drug therapy, Pyridines administration & dosage
- Abstract
Purpose: New therapies are needed to treat immune checkpoint inhibitor-resistant non-small cell lung cancer (NSCLC) and identify biomarkers to personalize treatment. Epigenetic therapies, including histone deacetylase inhibitors, may synergize with programmed cell death-1 (PD-1) blockade to overcome resistance. We report outcomes in patients with anti-programmed cell death ligand-1 [PD-(L)1]-resistant/refractory NSCLC treated with pembrolizumab plus entinostat in ENCORE 601., Patients and Methods: The expansion cohort of ENCORE 601 included patients with NSCLC who previously experienced disease progression with immune checkpoint inhibitors. The primary endpoint for the phase II expansion cohort is overall response rate (ORR); safety, tolerability, and exploratory endpoints are described., Results: Of 76 treated patients, 71 were evaluable for efficacy. immune-regulated RECIST-assessed ORR was 9.2% [95% confidence interval (CI): 3.8-18.1], which did not meet the prespecified threshold for positivity. Median duration of response was 10.1 months (95% CI: 3.9-not estimable), progression-free survival (PFS) at 6 months was 22%, median PFS was 2.8 months (95% CI: 1.5-4.1), and median overall survival was 11.7 months (95% CI: 7.6-13.4). Benefit was enriched among patients with high levels of circulating classical monocytes at baseline. Baseline tumor PD-L1 expression and IFNγ gene expression were not associated with benefit. Treatment-related grade ≥3 adverse events occurred in 41% of patients., Conclusions: In anti-PD-(L)1-experienced patients with NSCLC, entinostat plus pembrolizumab did not achieve the primary response rate endpoint but provided a clinically meaningful benefit, with objective response in 9% of patients. No new toxicities, including immune-related adverse events, were seen for either drug. Future studies will continue to evaluate the association of monocyte levels and response., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
37. Myeloid-Derived Suppressor Cells Are a Major Source of Wnt5A in the Melanoma Microenvironment and Depend on Wnt5A for Full Suppressive Activity.
- Author
-
Douglass SM, Fane ME, Sanseviero E, Ecker BL, Kugel CH 3rd, Behera R, Kumar V, Tcyganov EN, Yin X, Liu Q, Chhabra Y, Alicea GM, Kuruvilla R, Gabrilovich DI, and Weeraratna AT
- Subjects
- Animals, Antigens, CD metabolism, Arginase metabolism, Cell Line, Tumor, Female, Lung Neoplasms secondary, Lymphocytes, Tumor-Infiltrating metabolism, Male, Melanoma secondary, Mice, Mice, Inbred C57BL, Mice, Transgenic, Myeloid-Derived Suppressor Cells immunology, Neoplasm Invasiveness, Programmed Cell Death 1 Receptor metabolism, T-Lymphocytes, Regulatory metabolism, Transforming Growth Factor beta1 metabolism, Lymphocyte Activation Gene 3 Protein, Melanoma metabolism, Myeloid-Derived Suppressor Cells metabolism, Tumor Microenvironment, Wnt-5a Protein metabolism
- Abstract
Metastatic dissemination remains a significant barrier to successful therapy for melanoma. Wnt5A is a potent driver of invasion in melanoma and is believed to be secreted from the tumor microenvironment (TME). Our data suggest that myeloid-derived suppressor cells (MDSC) in the TME are a major source of Wnt5A and are reliant upon Wnt5A for multiple actions. Knockdown of Wnt5A specifically in the myeloid cells demonstrated a clear decrease in Wnt5A expression within the TME in vivo as well as a decrease in intratumoral MDSC and regulatory T cell (Treg). Wnt5A knockdown also decreased the immunosuppressive nature of MDSC and decreased expression of TGFβ1 and arginase 1. In the presence of Wnt5A-depleted MDSC, tumor-infiltrating lymphocytes expressed decreased PD-1 and LAG3, suggesting a less exhausted phenotype. Myeloid-specific Wnt5A knockdown also led to decreased lung metastasis. Tumor-infiltrating MDSC from control animals showed a strong positive correlation with Treg, which was completely ablated in animals with Wnt5A-negative MDSC. Overall, our data suggest that while MDSC contribute to an immunosuppressive and less immunogenic environment, they exhibit an additional function as the major source of Wnt5A in the TME. SIGNIFICANCE: These findings demonstrate that myeloid cells provide a major source of Wnt5A to facilitate metastatic potential in melanoma cells and rely on Wnt5A for their immunosuppressive function., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
38. Isolation and Phenotyping of Splenic Myeloid-Derived Suppressor Cells in Murine Cancer Models.
- Author
-
Sanseviero E, Kim R, and Gabrilovich DI
- Subjects
- Animals, Disease Models, Animal, Mice, Monocytes pathology, Neutrophils pathology, Phenotype, Cell Separation methods, Immunophenotyping methods, Myeloid-Derived Suppressor Cells pathology, Neoplasms pathology, Spleen pathology
- Abstract
Myeloid-derived suppressor cells (MDSC) are immunosuppressive myeloid cells that accumulate in tumor sites and peripheral lymphoid organs such as the spleen. In murine cancer models, the spleen is a major reservoir for MDSC, representing an easily accessible tissue from which to isolate high numbers of these cell population for downstream applications. Here we describe an efficient method to phenotype as well as to isolate and assess the functionality of murine splenic MDSC.
- Published
- 2021
- Full Text
- View/download PDF
39. Distinct Populations of Immune-Suppressive Macrophages Differentiate from Monocytic Myeloid-Derived Suppressor Cells in Cancer.
- Author
-
Kwak T, Wang F, Deng H, Condamine T, Kumar V, Perego M, Kossenkov A, Montaner LJ, Xu X, Xu W, Zheng C, Schuchter LM, Amaravadi RK, Mitchell TC, Karakousis GC, Mulligan C, Nam B, Masters G, Hockstein N, Bennett J, Nefedova Y, and Gabrilovich DI
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, CCAAT-Enhancer-Binding Proteins genetics, Cell Line, Tumor, Female, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Microarray Analysis, Middle Aged, Myeloid-Derived Suppressor Cells immunology, Tumor Microenvironment, CCAAT-Enhancer-Binding Proteins metabolism, Calgranulin A physiology, Calgranulin B physiology, Immunosuppression Therapy, Macrophages metabolism, Monocytes metabolism, Myeloid-Derived Suppressor Cells metabolism
- Abstract
Here, we report that functional heterogeneity of macrophages in cancer could be determined by the nature of their precursors: monocytes (Mons) and monocytic myeloid-derived suppressor cells (M-MDSCs). Macrophages that are differentiated from M-MDSCs, but not from Mons, are immune suppressive, with a genomic profile matching that of M-MDSCs. Immune-suppressive activity of M-MDSC-derived macrophages is dependent on the persistent expression of S100A9 protein in these cells. S100A9 also promotes M2 polarization of macrophages. Tissue-resident- and Mon-derived macrophages lack expression of this protein. S100A9-dependent immune-suppressive activity of macrophages involves transcription factor C/EBPβ. The presence of S100A9-positive macrophages in tumor tissues is associated with shorter survival in patients with head and neck cancer and poor response to PD-1 antibody treatment in patients with metastatic melanoma. Thus, this study reveals the pathway of the development of immune-suppressive macrophages and suggests an approach to their selective targeting., Competing Interests: Declaration of Interests D.I.G. is a current employee of AstraZeneca., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
40. Reactivation of dormant tumor cells by modified lipids derived from stress-activated neutrophils.
- Author
-
Perego M, Tyurin VA, Tyurina YY, Yellets J, Nacarelli T, Lin C, Nefedova Y, Kossenkov A, Liu Q, Sreedhar S, Pass H, Roth J, Vogl T, Feldser D, Zhang R, Kagan VE, and Gabrilovich DI
- Subjects
- Calgranulin A, Humans, Lipids, Neoplasm Recurrence, Local, Calgranulin B, Neutrophils
- Abstract
Tumor recurrence years after seemingly successful treatment of primary tumors is one of the major causes of mortality in patients with cancer. Reactivation of dormant tumor cells is largely responsible for this phenomenon. Using dormancy models of lung and ovarian cancer, we found a specific mechanism, mediated by stress and neutrophils, that may govern this process. Stress hormones cause rapid release of proinflammatory S100A8/A9 proteins by neutrophils. S100A8/A9 induce activation of myeloperoxidase, resulting in accumulation of oxidized lipids in these cells. Upon release from neutrophils, these lipids up-regulate the fibroblast growth factor pathway in tumor cells, causing tumor cell exit from the dormancy and formation of new tumor lesions. Higher serum concentrations of S100A8/A9 were associated with shorter time to recurrence in patients with lung cancer after complete tumor resection. Targeting of S100A8/A9 or β2-adrenergic receptors abrogated stress-induced reactivation of dormant tumor cells. These observations demonstrate a mechanism linking stress and specific neutrophil activation with early recurrence in cancer., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
41. A Novel Inhibitor of HSP70 Induces Mitochondrial Toxicity and Immune Cell Recruitment in Tumors.
- Author
-
Barnoud T, Leung JC, Leu JI, Basu S, Poli ANR, Parris JLD, Indeglia A, Martynyuk T, Good M, Gnanapradeepan K, Sanseviero E, Moeller R, Tang HY, Cassel J, Kossenkov AV, Liu Q, Speicher DW, Gabrilovich DI, Salvino JM, George DL, and Murphy ME
- Subjects
- Adenosine Triphosphate metabolism, Alarmins metabolism, Animals, Cell-Free System, Colorectal Neoplasms immunology, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, HMGB1 Protein metabolism, HT29 Cells, High-Throughput Screening Assays, Humans, Male, Mice, Inbred C57BL, Mice, Inbred NOD, Mitochondria metabolism, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Colorectal Neoplasms drug therapy, HSP70 Heat-Shock Proteins antagonists & inhibitors, Mitochondria drug effects
- Abstract
The protein chaperone HSP70 is overexpressed in many cancers including colorectal cancer, where overexpression is associated with poor survival. We report here the creation of a uniquely acting HSP70 inhibitor (HSP70i) that targets multiple compartments in the cancer cell, including mitochondria. This inhibitor was mitochondria toxic and cytotoxic to colorectal cancer cells, but not to normal colon epithelial cells. Inhibition of HSP70 was efficacious as a single agent in primary and metastatic models of colorectal cancer and enabled identification of novel mitochondrial client proteins for HSP70. In a syngeneic colorectal cancer model, the inhibitor increased immune cell recruitment into tumors. Cells treated with the inhibitor secreted danger-associated molecular patterns (DAMP), including ATP and HMGB1, and functioned effectively as a tumor vaccine. Interestingly, the unique properties of this HSP70i in the disruption of mitochondrial function and the inhibition of proteostasis both contributed to DAMP release. This HSP70i constitutes a promising therapeutic opportunity in colorectal cancer and may exhibit antitumor activity against other tumor types. SIGNIFICANCE: These findings describe a novel HSP70i that disrupts mitochondrial proteostasis, demonstrating single-agent efficacy that induces immunogenic cell death in treated tumors., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
42. PPT1 inhibition enhances the antitumor activity of anti-PD-1 antibody in melanoma.
- Author
-
Sharma G, Ojha R, Noguera-Ortega E, Rebecca VW, Attanasio J, Liu S, Piao S, Lee JJ, Nicastri MC, Harper SL, Ronghe A, Jain V, Winkler JD, Speicher DW, Mastio J, Gimotty PA, Xu X, Wherry EJ, Gabrilovich DI, and Amaravadi RK
- Subjects
- Animals, Antibodies immunology, Antineoplastic Combined Chemotherapy Protocols, Enzyme Inhibitors administration & dosage, Enzyme Inhibitors therapeutic use, Hydroxychloroquine administration & dosage, Hydroxychloroquine therapeutic use, Immune Checkpoint Inhibitors administration & dosage, Immune Checkpoint Inhibitors pharmacology, Interferon-beta metabolism, Macrophages drug effects, Macrophages immunology, Melanoma immunology, Mice, Mice, Inbred C57BL, Nucleotidyltransferases metabolism, Programmed Cell Death 1 Receptor immunology, RAW 264.7 Cells, T-Lymphocytes immunology, Thiolester Hydrolases genetics, Thiolester Hydrolases metabolism, Tumor Cells, Cultured, Enzyme Inhibitors pharmacology, Hydroxychloroquine pharmacology, Immune Checkpoint Inhibitors therapeutic use, Melanoma drug therapy, Programmed Cell Death 1 Receptor antagonists & inhibitors, Thiolester Hydrolases antagonists & inhibitors
- Abstract
New strategies are needed to enhance the efficacy of anti-programmed cell death protein antibody (anti-PD-1 Ab) in cancer. Here, we report that inhibiting palmitoyl-protein thioesterase 1 (PPT1), a target of chloroquine derivatives like hydroxychloroquine (HCQ), enhances the antitumor efficacy of anti-PD-1 Ab in melanoma. The combination resulted in tumor growth impairment and improved survival in mouse models. Genetic suppression of core autophagy genes, but not Ppt1, in cancer cells reduced priming and cytotoxic capacity of primed T cells. Exposure of antigen-primed T cells to macrophage-conditioned medium derived from macrophages treated with PPT1 inhibitors enhanced melanoma-specific killing. Genetic or chemical Ppt1 inhibition resulted in M2 to M1 phenotype switching in macrophages. The combination was associated with a reduction in myeloid-derived suppressor cells in the tumor. Ppt1 inhibition by HCQ, or DC661, induced cyclic GMP-AMP synthase/stimulator of interferon genes/TANK binding kinase 1 pathway activation and the secretion of interferon-β in macrophages, the latter being a key component for augmented T cell-mediated cytotoxicity. Genetic Ppt1 inhibition produced similar findings. These data provide the rationale for this combination in melanoma clinical trials and further investigation in other cancers.
- Published
- 2020
- Full Text
- View/download PDF
43. Changes in Aged Fibroblast Lipid Metabolism Induce Age-Dependent Melanoma Cell Resistance to Targeted Therapy via the Fatty Acid Transporter FATP2.
- Author
-
Alicea GM, Rebecca VW, Goldman AR, Fane ME, Douglass SM, Behera R, Webster MR, Kugel CH 3rd, Ecker BL, Caino MC, Kossenkov AV, Tang HY, Frederick DT, Flaherty KT, Xu X, Liu Q, Gabrilovich DI, Herlyn M, Blair IA, Schug ZT, Speicher DW, and Weeraratna AT
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cell Line, Tumor, Cellular Senescence, Coculture Techniques, Coenzyme A Ligases antagonists & inhibitors, Dermis cytology, Dermis pathology, Drug Resistance, Neoplasm drug effects, Humans, Keratinocytes metabolism, Lipid Metabolism, Melanoma pathology, Molecular Targeted Therapy methods, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Skin Neoplasms pathology, Tumor Microenvironment, Antineoplastic Combined Chemotherapy Protocols pharmacology, Coenzyme A Ligases metabolism, Fibroblasts metabolism, Melanoma drug therapy, Skin Neoplasms drug therapy
- Abstract
Older patients with melanoma (>50 years old) have poorer prognoses and response rates to targeted therapy compared with young patients (<50 years old), which can be driven, in part, by the aged microenvironment. Here, we show that aged dermal fibroblasts increase the secretion of neutral lipids, especially ceramides. When melanoma cells are exposed to the aged fibroblast lipid secretome, or cocultured with aged fibroblasts, they increase the uptake of lipids via the fatty acid transporter FATP2, which is upregulated in melanoma cells in the aged microenvironment and known to play roles in lipid synthesis and accumulation. We show that blocking FATP2 in melanoma cells in an aged microenvironment inhibits their accumulation of lipids and disrupts their mitochondrial metabolism. Inhibiting FATP2 overcomes age-related resistance to BRAF/MEK inhibition in animal models, ablates tumor relapse, and significantly extends survival time in older animals. SIGNIFICANCE: These data show that melanoma cells take up lipids from aged fibroblasts, via FATP2, and use them to resist targeted therapy. The response to targeted therapy is altered in aged individuals because of the influences of the aged microenvironment, and these data suggest FATP2 as a target to overcome resistance. See related commentary by Montal and White, p. 1255 . This article is highlighted in the In This Issue feature, p. 1241 ., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
44. BTN3A1 governs antitumor responses by coordinating αβ and γδ T cells.
- Author
-
Payne KK, Mine JA, Biswas S, Chaurio RA, Perales-Puchalt A, Anadon CM, Costich TL, Harro CM, Walrath J, Ming Q, Tcyganov E, Buras AL, Rigolizzo KE, Mandal G, Lajoie J, Ophir M, Tchou J, Marchion D, Luca VC, Bobrowicz P, McLaughlin B, Eskiocak U, Schmidt M, Cubillos-Ruiz JR, Rodriguez PC, Gabrilovich DI, and Conejo-Garcia JR
- Subjects
- Animals, Antibodies, Monoclonal therapeutic use, Antigens, CD genetics, Butyrophilins genetics, Female, Humans, Immunotherapy methods, Mice, Mice, Transgenic, Receptors, Antigen, T-Cell, alpha-beta immunology, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antigens, CD immunology, Butyrophilins antagonists & inhibitors, Butyrophilins immunology, Intraepithelial Lymphocytes immunology, Ovarian Neoplasms immunology, Ovarian Neoplasms therapy
- Abstract
Gamma delta (γδ) T cells infiltrate most human tumors, but current immunotherapies fail to exploit their in situ major histocompatibility complex-independent tumoricidal potential. Activation of γδ T cells can be elicited by butyrophilin and butyrophilin-like molecules that are structurally similar to the immunosuppressive B7 family members, yet how they regulate and coordinate αβ and γδ T cell responses remains unknown. Here, we report that the butyrophilin BTN3A1 inhibits tumor-reactive αβ T cell receptor activation by preventing segregation of N-glycosylated CD45 from the immune synapse. Notably, CD277-specific antibodies elicit coordinated restoration of αβ T cell effector activity and BTN2A1-dependent γδ lymphocyte cytotoxicity against BTN3A1
+ cancer cells, abrogating malignant progression. Targeting BTN3A1 therefore orchestrates cooperative killing of established tumors by αβ and γδ T cells and may present a treatment strategy for tumors resistant to existing immunotherapies., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2020
- Full Text
- View/download PDF
45. Polymorphonuclear myeloid-derived suppressor cells limit antigen cross-presentation by dendritic cells in cancer.
- Author
-
Ugolini A, Tyurin VA, Tyurina YY, Tcyganov EN, Donthireddy L, Kagan VE, Gabrilovich DI, and Veglia F
- Subjects
- Animals, Cross-Priming immunology, Female, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Neoplasms pathology, Tumor Cells, Cultured, Tumor Microenvironment immunology, Xenograft Model Antitumor Assays, Antigen Presentation immunology, Antigens, Neoplasm immunology, Dendritic Cells immunology, Myeloid-Derived Suppressor Cells immunology, Neoplasms immunology, Neutrophils immunology, Peroxidase physiology
- Abstract
DCs are a critical component of immune responses in cancer primarily due to their ability to cross-present tumor-associated antigens. Cross-presentation by DCs in cancer is impaired, which may represent one of the obstacles for the success of cancer immunotherapies. Here, we report that polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) blocked cross-presentation by DCs without affecting direct presentation of antigens by these cells. This effect did not require direct cell-cell contact and was associated with transfer of lipids. Neutrophils (PMN) and PMN-MDSC transferred lipid to DCs equally well; however, PMN did not affect DC cross-presentation. PMN-MDSC generate oxidatively truncated lipids previously shown to be involved in impaired cross-presentation by DCs. Accumulation of oxidized lipids in PMN-MDSC was dependent on myeloperoxidase (MPO). MPO-deficient PMN-MDSC did not affect cross-presentation by DCs. Cross-presentation of tumor-associated antigens in vivo by DCs was improved in MDSC-depleted or tumor-bearing MPO-KO mice. Pharmacological inhibition of MPO in combination with checkpoint blockade reduced tumor progression in different tumor models. These data suggest MPO-driven lipid peroxidation in PMN-MDSC as a possible non-cell autonomous mechanism of inhibition of antigen cross-presentation by DCs and propose MPO as potential therapeutic target to enhance the efficacy of current immunotherapies for patients with cancer.
- Published
- 2020
- Full Text
- View/download PDF
46. All Myeloid-Derived Suppressor Cells Are Not Created Equal: How Gender Inequality Influences These Cells and Affects Cancer Therapy.
- Author
-
Gabrilovich DI
- Subjects
- Female, Humans, Male, Glioblastoma genetics, Glioblastoma therapy, Myeloid-Derived Suppressor Cells
- Abstract
In this issue of Cancer Discovery , Bayik and colleagues demonstrated sexual dimorphism in accumulation of different populations of myeloid-derived suppressor cells in glioblastoma and showed that they could be targeted by different agents. See related article by Bayik et al., p. 1210 ., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
47. Activation of p38α stress-activated protein kinase drives the formation of the pre-metastatic niche in the lungs.
- Author
-
Gui J, Zahedi F, Ortiz A, Cho C, Katlinski KV, Alicea-Torres K, Li J, Todd L, Zhang H, Beiting DP, Sander C, Kirkwood JM, Snow BE, Wakeham AC, Mak TW, Diehl JA, Koumenis C, Ryeom SW, Stanger BZ, Puré E, Gabrilovich DI, and Fuchs SY
- Subjects
- Fibroblasts pathology, Humans, Lung pathology, Protein Kinases, Lung Neoplasms pathology, Signal Transduction
- Abstract
Primary tumor-derived factors (TDFs) act upon normal cells to generate a pre-metastatic niche, which promotes colonization of target organs by disseminated malignant cells. Here we report that TDFs-induced activation of the p38α kinase in lung fibroblasts plays a critical role in the formation of a pre-metastatic niche in the lungs and subsequent pulmonary metastases. Activation of p38α led to inactivation of type I interferon signaling and stimulation of expression of fibroblast activation protein (FAP). FAP played a key role in remodeling of the extracellular matrix as well as inducing the expression of chemokines that enable lung infiltration by neutrophils. Increased activity of p38 in normal cells was associated with metastatic disease and poor prognosis in human melanoma patients whereas inactivation of p38 suppressed lung metastases. We discuss the p38α-driven mechanisms stimulating the metastatic processes and potential use of p38 inhibitors in adjuvant therapy of metastatic cancers., Competing Interests: Competing Interests Statement The authors have declared that no conflict of interest exists
- Published
- 2020
- Full Text
- View/download PDF
48. Therapies for tuberculosis and AIDS: myeloid-derived suppressor cells in focus.
- Author
-
Dorhoi A, Kotzé LA, Berzofsky JA, Sui Y, Gabrilovich DI, Garg A, Hafner R, Khader SA, Schaible UE, Kaufmann SH, Walzl G, Lutz MB, Mahon RN, Ostrand-Rosenberg S, Bishai W, and du Plessis N
- Subjects
- Humans, Myeloid-Derived Suppressor Cells pathology, Acquired Immunodeficiency Syndrome immunology, Acquired Immunodeficiency Syndrome pathology, Acquired Immunodeficiency Syndrome therapy, HIV-1 immunology, Mycobacterium tuberculosis immunology, Myeloid-Derived Suppressor Cells immunology, Tuberculosis immunology, Tuberculosis pathology, Tuberculosis therapy
- Abstract
The critical role of suppressive myeloid cells in immune regulation has come to the forefront in cancer research, with myeloid-derived suppressor cells (MDSCs) as a main oncology immunotherapeutic target. Recent improvement and standardization of criteria classifying tumor-induced MDSCs have led to unified descriptions and also promoted MDSC research in tuberculosis (TB) and AIDS. Despite convincing evidence on the induction of MDSCs by pathogen-derived molecules and inflammatory mediators in TB and AIDS, very little attention has been given to their therapeutic modulation or roles in vaccination in these diseases. Clinical manifestations in TB are consequences of complex host-pathogen interactions and are substantially affected by HIV infection. Here we summarize the current understanding and knowledge gaps regarding the role of MDSCs in HIV and Mycobacterium tuberculosis (co)infections. We discuss key scientific priorities to enable application of this knowledge to the development of novel strategies to improve vaccine efficacy and/or implementation of enhanced treatment approaches. Building on recent findings and potential for cross-fertilization between oncology and infection biology, we highlight current challenges and untapped opportunities for translating new advances in MDSC research into clinical applications for TB and AIDS.
- Published
- 2020
- Full Text
- View/download PDF
49. Detecting Prostate Cancer Using Pattern Recognition Neural Networks With Flow Cytometry-Based Immunophenotyping in At-Risk Men.
- Author
-
Dominguez GA, Polo AT, Roop J, Campisi AJ, Somer RA, Perzin AD, Gabrilovich DI, and Kumar A
- Abstract
Current screening methods for prostate cancer (PCa) result in a large number of false positives making it difficult for clinicians to assess disease status, thus warranting advancements in screening and early detection methods. The goal of this study was to design a liquid biopsy test that uses flow cytometry-based immunophenotyping and artificial neural network (ANN) analysis to detect PCa. Numerous myeloid and lymphoid cell populations, including myeloid-derived suppressor cells, were measured from 156 patients with PCa, 123 with benign prostatic hyperplasia (BPH), and 99 male healthy donor (HD) controls. Using pattern recognition neural network (PRNN) analysis, a type of ANN, PCa detection compared against HD resulted in 96.6% sensitivity, 87.5% specificity, and an area under the curve (AUC) value of 0.97. Detecting patients with higher risk disease (⩾Gleason 7) against lower risk disease (BPH/Gleason 6) resulted in 92.0% sensitivity, 42.7% specificity, and an AUC of 0.72. This study suggests that analyzing flow cytometry immunophenotyping data with PRNNs may prove to be a useful tool to improve PCa detection and reduce the number of unnecessary prostate biopsies performed each year., Competing Interests: Declaration of Conflicting Interests:The author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: G.A.D., A.T.P*, J.R., A.J.C., and A.K. are employees (or former employees*) of Anixa Diagnostics Corp. and have ownership (including patents) interests., (© The Author(s) 2020.)
- Published
- 2020
- Full Text
- View/download PDF
50. Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death.
- Author
-
Kapralov AA, Yang Q, Dar HH, Tyurina YY, Anthonymuthu TS, Kim R, St Croix CM, Mikulska-Ruminska K, Liu B, Shrivastava IH, Tyurin VA, Ting HC, Wu YL, Gao Y, Shurin GV, Artyukhova MA, Ponomareva LA, Timashev PS, Domingues RM, Stoyanovsky DA, Greenberger JS, Mallampalli RK, Bahar I, Gabrilovich DI, Bayır H, and Kagan VE
- Subjects
- Animals, Arachidonate 15-Lipoxygenase metabolism, Arachidonate 15-Lipoxygenase physiology, Cell Death, Female, Iron metabolism, Iron physiology, Leukotrienes metabolism, Lipid Peroxidation physiology, Lipid Peroxides metabolism, Male, Mice, Mice, Inbred C57BL, Microglia metabolism, Nitric Oxide Synthase Type II physiology, Oxidation-Reduction, Reactive Oxygen Species metabolism, Ferroptosis physiology, Macrophages metabolism, Nitric Oxide Synthase Type II metabolism
- Abstract
Ferroptotic death is the penalty for losing control over three processes-iron metabolism, lipid peroxidation and thiol regulation-that are common in the pro-inflammatory environment where professional phagocytes fulfill their functions and yet survive. We hypothesized that redox reprogramming of 15-lipoxygenase (15-LOX) during the generation of pro-ferroptotic signal 15-hydroperoxy-eicosa-tetra-enoyl-phosphatidylethanolamine (15-HpETE-PE) modulates ferroptotic endurance. Here, we have discovered that inducible nitric oxide synthase (iNOS)/NO
• -enrichment of activated M1 (but not alternatively activated M2) macrophages/microglia modulates susceptibility to ferroptosis. Genetic or pharmacologic depletion/inactivation of iNOS confers sensitivity on M1 cells, whereas NO• donors empower resistance of M2 cells to ferroptosis. In vivo, M1 phagocytes, in comparison to M2 phagocytes, exert higher resistance to pharmacologically induced ferroptosis. This resistance is diminished in iNOS-deficient cells in the pro-inflammatory conditions of brain trauma or the tumour microenvironment. The nitroxygenation of eicosatetraenoyl (ETE)-PE intermediates and oxidatively truncated species by NO• donors and/or suppression of NO• production by iNOS inhibitors represent a novel redox mechanism of regulation of ferroptosis in pro-inflammatory conditions.- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.