6
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      IGF1 receptor inhibition amplifies the effects of cancer drugs by autophagy and immune-dependent mechanisms

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Pharmacological autophagy enhancement constitutes a preclinically validated strategy for preventing or treating most major age-associated diseases. Driven by this consideration, we performed a high-content/high-throughput screen on 65 000 distinct compounds on a robotized fluorescence microscopy platform to identify novel autophagy inducers.

          Results

          Here, we report the discovery of picropodophyllin (PPP) as a potent inducer of autophagic flux that acts on-target, as an inhibitor of the tyrosine kinase activity of the insulin-like growth factor-1 receptor (IGF1R). Thus, PPP lost its autophagy-stimulatory activity in cells engineered to lack IGF1R or to express a constitutively active AKT serine/threonine kinase 1 (AKT1) mutant. When administered to cancer-bearing mice, PPP improved the therapeutic efficacy of chemoimmunotherapy with a combination of immunogenic cytotoxicants and programmed cell death 1 (PDCD1, better known as PD-1) blockade. These PPP effects were lost when tumors were rendered PPP-insensitive or autophagy-incompetent. In combination with chemotherapy, PPP enhanced the infiltration of tumors by cytotoxic T lymphocytes, while reducing regulatory T cells. In human triple-negative breast cancer patients, the activating phosphorylation of IGF1R correlated with inhibited autophagy, an unfavorable local immune profile, and poor prognosis.

          Conclusion

          Altogether, these results suggest that IGF1R may constitute a novel and druggable therapeutic target for the treatment of cancer in conjunction with chemoimmunotherapies.

          Related collections

          Most cited references63

          • Record: found
          • Abstract: found
          • Article: not found

          Autophagy and the integrated stress response.

          Autophagy is a tightly regulated pathway involving the lysosomal degradation of cytoplasmic organelles or cytosolic components. This pathway can be stimulated by multiple forms of cellular stress, including nutrient or growth factor deprivation, hypoxia, reactive oxygen species, DNA damage, protein aggregates, damaged organelles, or intracellular pathogens. Both specific, stimulus-dependent and more general, stimulus-independent signaling pathways are activated to coordinate different phases of autophagy. Autophagy can be integrated with other cellular stress responses through parallel stimulation of autophagy and other stress responses by specific stress stimuli, through dual regulation of autophagy and other stress responses by multifunctional stress signaling molecules, and/or through mutual control of autophagy and other stress responses. Thus, autophagy is a cell biological process that is a central component of the integrated stress response. Copyright © 2010 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I

            Immune evasion is a major obstacle for cancer treatment. Common mechanisms include impaired antigen presentation through mutations or loss of heterozygosity (LOH) of the major histocompatibility complex class I (MHC-I), which has been implicated in resistance to immune checkpoint blockade (ICB) therapy 1–3 . However, in pancreatic ductal adenocarcinoma (PDAC), a malignancy refractory to most therapies including ICB 4 , mutations causing MHC-I loss are rarely found 5 despite the frequent downregulation of MHC-I expression 6–8 . Here we find that, in PDAC, MHC-I molecules are selectively targeted for lysosomal degradation through an autophagy-dependent mechanism that involves the autophagy cargo receptor NBR1. PDAC cells display reduced MHC-I cell surface expression and instead demonstrate predominant localization within autophagosomes and lysosomes. Notably, autophagy inhibition restores surface MHC-I levels, leading to improved antigen presentation, enhanced anti-tumour T cell response and reduced tumour growth in syngeneic hosts. Accordingly, anti-tumour effects of autophagy inhibition are reversed by depleting CD8+ T cells or reducing surface MHC-I expression. Autophagy inhibition, either genetically or pharmacologically with Chloroquine (CQ), synergizes with dual ICB (anti-PD1 and anti-CTLA4), and leads to an enhanced anti-tumour immune response. Our findings uncover a role for enhanced autophagy/lysosome function in immune evasion through selective targeting of MHC-I molecules for degradation, and provide a rationale for the combination of autophagy inhibition and dual ICB as a therapeutic strategy against PDAC.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Extending healthy life span--from yeast to humans.

              When the food intake of organisms such as yeast and rodents is reduced (dietary restriction), they live longer than organisms fed a normal diet. A similar effect is seen when the activity of nutrient-sensing pathways is reduced by mutations or chemical inhibitors. In rodents, both dietary restriction and decreased nutrient-sensing pathway activity can lower the incidence of age-related loss of function and disease, including tumors and neurodegeneration. Dietary restriction also increases life span and protects against diabetes, cancer, and cardiovascular disease in rhesus monkeys, and in humans it causes changes that protect against these age-related pathologies. Tumors and diabetes are also uncommon in humans with mutations in the growth hormone receptor, and natural genetic variants in nutrient-sensing pathways are associated with increased human life span. Dietary restriction and reduced activity of nutrient-sensing pathways may thus slow aging by similar mechanisms, which have been conserved during evolution. We discuss these findings and their potential application to prevention of age-related disease and promotion of healthy aging in humans, and the challenge of possible negative side effects.
                Bookmark

                Author and article information

                Journal
                J Immunother Cancer
                J Immunother Cancer
                jitc
                jitc
                Journal for Immunotherapy of Cancer
                BMJ Publishing Group (BMA House, Tavistock Square, London, WC1H 9JR )
                2051-1426
                2021
                14 June 2021
                : 9
                : 6
                : e002722
                Affiliations
                [1 ]departmentDepartment of Breast and Thyroid Surgery , Renmin Hospital of Wuhan University , Wuhan, Hubei, China
                [2 ]departmentCentre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer , Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France , Paris, France
                [3 ]departmentMetabolomics and Cell Biology Platforms , Gustave Roussy Cancer Center, Université Paris Saclay , Villejuif, France
                [4 ]departmentFaculty of Medicine , Université Paris Saclay , Kremlin Bicêtre, France
                [5 ]departmentDepartment of Pathology , Renmin Hospital of Wuhan University , Wuhan, Hubei, China
                [6 ]Samsara Therapeutics Ltd , Oxford, UK
                [7 ]departmentInstitute of Molecular Biosciences , NAWI Graz, University of Graz , Graz, Austria
                [8 ]BioTechMed-Graz , Graz, Austria
                [9 ]departmentField of Excellence BioHealth , University of Graz , Graz, Austria
                [10 ]departmentSuzhou Institute for Systems Medicine , Chinese Academy of Medical Sciences , Suzhou, Jiangsu, China
                [11 ]departmentPôle de Biologie , Hôpital Européen Georges Pompidou, AP-HP , Paris, France
                [12 ]departmentKarolinska Institutet, Department of Women's and Children's Health , Karolinska University Hospital , Stockholm, Sweden
                Author notes
                [Correspondence to ] Dr Oliver Kepp; captain.olsen@ 123456gmail.com ; Professor Yi Tu; ty701105@ 123456163.com ; Prof Guido Kroemer; kroemer@ 123456orange.fr

                QW, A-LT and BL are joint first authors.

                Author information
                http://orcid.org/0000-0002-6081-9558
                Article
                jitc-2021-002722
                10.1136/jitc-2021-002722
                8204183
                34127545
                ccdb9167-6110-41d0-b971-0bb7a7562eb5
                © Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

                This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See http://creativecommons.org/licenses/by-nc/4.0/.

                History
                : 04 May 2021
                Funding
                Funded by: Association pour la recherche sur le cancer (ARC);
                Funded by: BioTechMed;
                Funded by: University of Graz;
                Award ID: Fast4Health
                Funded by: Université Paris-Saclay, Plateforme Imagerie;
                Funded by: Île de France;
                Award ID: DIM Elicit
                Funded by: FundRef http://dx.doi.org/10.13039/501100004099, Ligue Contre le Cancer;
                Funded by: http://dx.doi.org/10.13039/501100003819, Natural Science Foundation of Hubei Province;
                Award ID: 2020CFA026
                Funded by: Agence National de la Recherche (ANR);
                Award ID: Projets blancs
                Funded by: European Union;
                Funded by: Fondation pour la Recherche Médicale (FRM);
                Funded by: High-end Foreign Expert Program in China;
                Award ID: GDW20171100085
                Funded by: Fondation Carrefour;
                Funded by: IdEx Université de Paris ANR;
                Award ID: 18-IDEX-0001
                Funded by: http://dx.doi.org/10.13039/501100004100, Labex;
                Award ID: ANR-18-IDEX-0001
                Funded by: Austrian Federal Ministry of Education, Science and Research;
                Award ID: BMWFW-80.109/0001-WF/V/3b/2015)
                Funded by: http://dx.doi.org/10.13039/501100002428, Austrian Science Fund;
                Award ID: W1226, P27893, P29203, P29262, P31727
                Award ID: SFB LIPOTOX F3007 & F3012
                Funded by: Chinese Scholarship Council;
                Funded by: AMMICa;
                Award ID: US23/CNRS UMS3655
                Funded by: Association Ruban Rose;
                Funded by: Canceropôle Île de France;
                Funded by: Elior;
                Funded by: Equipex Onco-Pheno-Screen;
                Funded by: European Joint Program on Rare Disease (EJPRD);
                Funded by: Gustave Roussy;
                Award ID: Odyssea
                Funded by: European Union Horizon 2020 Projects;
                Award ID: Oncobiome and Crimson
                Funded by: Fondation Carrefour;
                Funded by: http://dx.doi.org/10.13039/501100006364, Institut National Du Cancer;
                Funded by: INSERM (HTE);
                Funded by: http://dx.doi.org/10.13039/501100004795, Institut Universitaire de France;
                Funded by: RHU Torino Lumière;
                Funded by: Seerave Foundation;
                Funded by: SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE);
                Funded by: SIRIC Cancer Research and Personalized Medicine (CARPEM);
                Funded by: NAWI Granz;
                Award ID: BioHealth
                Funded by: BioTechMed-Graz;
                Award ID: EPIAge
                Categories
                Basic Tumor Immunology
                1506
                2434
                Original research
                Custom metadata
                unlocked

                immunotherapy,breast neoplasms,systems biology
                immunotherapy, breast neoplasms, systems biology

                Comments

                Comment on this article