Superparamagnetic Iron Oxide Nanoparticles Reprogram the Tumor Microenvironment and Reduce Lung Cancer Regrowth after Crizotinib TreatmentClick to copy article linkArticle link copied!
- Natalie K. HorvatNatalie K. HorvatDepartment of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, GermanyMolecular Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, GermanyRuprecht Karl University of Heidelberg, 69120, Heidelberg, GermanyMore by Natalie K. Horvat
- Sara ChocarroSara ChocarroDivision of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, GermanyRuprecht Karl University of Heidelberg, 69120, Heidelberg, GermanyMore by Sara Chocarro
- Oriana MarquesOriana MarquesDepartment of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, GermanyMolecular Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, GermanyMore by Oriana Marques
- Tobias A. BauerTobias A. BauerLeiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The NetherlandsMore by Tobias A. Bauer
- Ruiyue QiuRuiyue QiuDepartment of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, GermanyMore by Ruiyue Qiu
- Alberto Diaz-JimenezAlberto Diaz-JimenezDivision of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, GermanyRuprecht Karl University of Heidelberg, 69120, Heidelberg, GermanyMore by Alberto Diaz-Jimenez
- Barbara HelmBarbara HelmDivision of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, GermanyGerman Center for Lung Research (DZL) and Translational Lung Research Center Heidelberg (TRLC), 69120, Heidelberg, GermanyMore by Barbara Helm
- Yuanyuan ChenYuanyuan ChenDivision of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, GermanyMore by Yuanyuan Chen
- Stefan SawallStefan SawallX-ray Imaging and CT, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, GermanyMore by Stefan Sawall
- Richard SparlaRichard SparlaDepartment of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, GermanyMore by Richard Sparla
- Lu SuLu SuLeiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The NetherlandsMore by Lu Su
- Ursula KlingmüllerUrsula KlingmüllerDivision of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, GermanyGerman Center for Lung Research (DZL) and Translational Lung Research Center Heidelberg (TRLC), 69120, Heidelberg, GermanyGerman Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, GermanyMore by Ursula Klingmüller
- Matthias BarzMatthias BarzLeiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The NetherlandsDepartment of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, GermanyMore by Matthias Barz
- Matthias W. Hentze*Matthias W. Hentze*E-mail: [email protected]Molecular Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, GermanyEuropean Molecular Biology Laboratory (EMBL), Meyerhofstr.1, 69117, Heidelberg, GermanyMore by Matthias W. Hentze
- Rocío Sotillo*Rocío Sotillo*E-mail: [email protected]Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, GermanyGerman Center for Lung Research (DZL) and Translational Lung Research Center Heidelberg (TRLC), 69120, Heidelberg, GermanyGerman Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, GermanyMore by Rocío Sotillo
- Martina U. Muckenthaler*Martina U. Muckenthaler*E-mail: [email protected]Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, GermanyMolecular Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, GermanyGerman Center for Lung Research (DZL) and Translational Lung Research Center Heidelberg (TRLC), 69120, Heidelberg, GermanyGerman Centre for Cardiovascular Research (DZHK), Partner Site, 69120, Heidelberg/Mannheim, GermanyMore by Martina U. Muckenthaler
Abstract
ALK-positive NSCLC patients demonstrate initial responses to ALK tyrosine kinase inhibitor (TKI) treatments, but eventually develop resistance, causing rapid tumor relapse and poor survival rates. Growing evidence suggests that the combination of drug and immune therapies greatly improves patient survival; however, due to the low immunogenicity of the tumors, ALK-positive patients do not respond to currently available immunotherapies. Tumor-associated macrophages (TAMs) play a crucial role in facilitating lung cancer growth by suppressing tumoricidal immune activation and absorbing chemotherapeutics. However, they can also be programmed toward a pro-inflammatory tumor suppressive phenotype, which represents a highly active area of therapy development. Iron loading of TAMs can achieve such reprogramming correlating with an improved prognosis in lung cancer patients. We previously showed that superparamagnetic iron oxide nanoparticles containing core-cross-linked polymer micelles (SPION-CCPMs) target macrophages and stimulate pro-inflammatory activation. Here, we show that SPION-CCPMs stimulate TAMs to secrete reactive nitrogen species and cytokines that exert tumoricidal activity. We further show that SPION-CCPMs reshape the immunosuppressive Eml4-Alk lung tumor microenvironment (TME) toward a cytotoxic profile hallmarked by the recruitment of CD8+ T cells, suggesting a multifactorial benefit of SPION-CCPM application. When intratracheally instilled into lung cancer-bearing mice, SPION-CCPMs delay tumor growth and, after first line therapy with a TKI, halt the regrowth of relapsing tumors. These findings identify SPIONs-CCPMs as an adjuvant therapy, which remodels the TME, resulting in a delay in the appearance of resistant tumors.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Results
Synthesis and Characterization of SPION-CCPMs
SPION-CCPM-Loaded Macrophages Reduce Cancer Cell Viability and Proliferation in Cocultures
SPION-CCPM Uptake in Cocultured Macrophages Induces an Acute Inflammatory Response and Increased Phagocytic Activity
Secreted Factors from SPION-CCPM-Loaded Macrophages Reduce Cancer Viability
SPION-CCPMs are Taken up by Innate Immune Cells of the Lung and Stimulate Pro-Inflammatory Responses
Pulmonary SPION-CCPM Instillation Diminishes Lung Tumor Growth
SPION-CCPMs Reprogram the Tumor-Associated Immune Landscape by Inducing Pro-Inflammatory and Cytotoxic Immune Cells
SPION-CCPM Adjuvant Therapy Effectively Reduces Cancer Regrowth
Discussion
Conclusions
Study Strengths
Study Limitations
Methods/Experimental
SPION CCPM Synthesis
Dynamic Light Scattering
Transmission Electron Microscopy
Cell Lines
Bone Marrow-Derived Macrophage Isolation, Differentiation, and Coculture
Flow Cytometry
Protein Extraction and Western Blotting
RNA Extraction, Reverse Transcription, and Quantitative Real-Time PCR Analysis
Sample Preparation and Protein Digestion
LC-MS/MS Analysis
Database Search and Proteomic Data Analysis
GSH Assay
Mice and Adenoviral Infection
Tissue Non-Heme Iron Measurement
Serum Iron Measurement
Immunostainings
Perls’ Prussian Blue Staining and DAB-enhanced Perls’ staining
μCT Imaging
Data Availability
Statistics and Reproducibility
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsnano.3c08335.
Figure S1, SPION-CCPM uptake in cocultures, EML4-Alk proteomics, gene analysis, γ-H2AX microscopy, Nqo1/Gclc RT-PCR, and GSH assays; Figure S2, cell viability, lipid peroxidation, Gpx4 RT-PCR, LDH, drug assays, antibody neutralization assay, and coculture proteomics; Figure S3, SPION-CCPM intratracheal instillation data in mice; Figure S4, supplementary results for Figures 4 and 5; Table ST1, RT-PCR primer pairs; Table ST2, flow cytometry antibodies (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We would like to thank the DKFZ Core Facilities of Light Microscopy and the Small Animal Imaging Center for the technical assistance and the Central Animal Laboratory for animal care and husbandry. Authors would also like to acknowledge the EMBL Flow Cytometry Core Facility, especially Dr. Malte Paulsen and Dr. Diana Ordonez, for training, discussions, and technical assistance. We thank Dr. Sandro Altamura, Dr. Silvia Colucci, and Dr. Francesca Vinchi for insightful discussions, inspiration, and support spanning the course of this study. Authors would like to thank Dr. Jessica Konen for scientific consulting and Prof. Annette Kopp-Schneider for help with statistical analyses. Schemes were generated using BioRender.com.
References
This article references 77 other publications.
- 1Sung, H.; Ferlay, J.; Siegel, R. L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. Ca Cancer J. Clin 2021, 71 (3), 209– 249, DOI: 10.3322/caac.21660Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3srpsVKnug%253D%253D&md5=f3e54fabe5ac5f2397292bf9a97ce5bfGlobal Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 CountriesSung Hyuna; Siegel Rebecca L; Jemal Ahmedin; Ferlay Jacques; Laversanne Mathieu; Soerjomataram Isabelle; Bray FreddieCA: a cancer journal for clinicians (2021), 71 (3), 209-249 ISSN:.This article provides an update on the global cancer burden using the GLOBOCAN 2020 estimates of cancer incidence and mortality produced by the International Agency for Research on Cancer. Worldwide, an estimated 19.3 million new cancer cases (18.1 million excluding nonmelanoma skin cancer) and almost 10.0 million cancer deaths (9.9 million excluding nonmelanoma skin cancer) occurred in 2020. Female breast cancer has surpassed lung cancer as the most commonly diagnosed cancer, with an estimated 2.3 million new cases (11.7%), followed by lung (11.4%), colorectal (10.0 %), prostate (7.3%), and stomach (5.6%) cancers. Lung cancer remained the leading cause of cancer death, with an estimated 1.8 million deaths (18%), followed by colorectal (9.4%), liver (8.3%), stomach (7.7%), and female breast (6.9%) cancers. Overall incidence was from 2-fold to 3-fold higher in transitioned versus transitioning countries for both sexes, whereas mortality varied <2-fold for men and little for women. Death rates for female breast and cervical cancers, however, were considerably higher in transitioning versus transitioned countries (15.0 vs 12.8 per 100,000 and 12.4 vs 5.2 per 100,000, respectively). The global cancer burden is expected to be 28.4 million cases in 2040, a 47% rise from 2020, with a larger increase in transitioning (64% to 95%) versus transitioned (32% to 56%) countries due to demographic changes, although this may be further exacerbated by increasing risk factors associated with globalization and a growing economy. Efforts to build a sustainable infrastructure for the dissemination of cancer prevention measures and provision of cancer care in transitioning countries is critical for global cancer control.
- 2Sica, A.; Mantovani, A. Macrophage Plasticity and Polarization: In Vivo Veritas. J. Clin Invest 2012, 122 (3), 787– 795, DOI: 10.1172/JCI59643Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XjsV2ms7g%253D&md5=2789f6879df4dea15e294b335f086598Macrophage plasticity and polarization: in vivo veritasSica, Antonio; Mantovani, AlbertoJournal of Clinical Investigation (2012), 122 (3), 787-795CODEN: JCINAO; ISSN:0021-9738. (American Society for Clinical Investigation)A review. Diversity and plasticity are hallmarks of cells of the monocyte-macrophage lineage. In response to IFNs, Toll-like receptor engagement, or IL-4/IL-13 signaling, macrophages undergo M1 (classical) or M2 (alternative) activation, which represent extremes of a continuum in a universe of activation states. Progress has now been made in defining the signaling pathways, transcriptional networks, and epigenetic mechanisms underlying M1-M2 or M2-like polarized activation. Functional skewing of mononuclear phagocytes occurs in vivo under physiol. conditions (e.g., ontogenesis and pregnancy) and in pathol. (allergic and chronic inflammation, tissue repair, infection, and cancer). However, in selected preclin. and clin. conditions, coexistence of cells in different activation states and unique or mixed phenotypes have been obsd., a reflection of dynamic changes and complex tissue-derived signals. The identification of mechanisms and mols. assocd. with macrophage plasticity and polarized activation provides a basis for macrophage-centered diagnostic and therapeutic strategies.
- 3Franklin, R. A.; Li, M. O. Ontogeny of Tumor-Associated Macrophages and Its Implication in Cancer Regulation. Trends Cancer 2016, 2 (1), 20– 34, DOI: 10.1016/j.trecan.2015.11.004Google ScholarThere is no corresponding record for this reference.
- 4Sedighzadeh, S. S.; Khoshbin, A. P.; Razi, S.; Keshavarz-Fathi, M.; Rezaei, N. A Narrative Review of Tumor-Associated Macrophages in Lung Cancer: Regulation of Macrophage Polarization and Therapeutic Implications. Transl Lung Cancer Res. 2021, 10 (4), 1889– 1916, DOI: 10.21037/tlcr-20-1241Google ScholarThere is no corresponding record for this reference.
- 5Condeelis, J.; Pollard, J. W. Macrophages: Obligate Partners for Tumor Cell Migration, Invasion, and Metastasis. Cell 2006, 124 (2), 263– 266, DOI: 10.1016/j.cell.2006.01.007Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xht1KktLo%253D&md5=ddd258f9109498d4f1c252bfc5bc0a63Macrophages: obligate partners for tumor cell migration, invasion, and metastasisCondeelis, John; Pollard, Jeffrey W.Cell (Cambridge, MA, United States) (2006), 124 (2), 263-266CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Macrophages within the tumor microenvironment facilitate angiogenesis and extracellular-matrix breakdown and remodeling and promote tumor cell motility. Recent studies reveal that direct communication between macrophages and tumor cells leads to invasion and egress of tumor cells into the blood vessels (intravasation). Thus, macrophages are at the center of the invasion microenvironment and are an important drug target for cancer therapy.
- 6Shi, L.; Wang, L.; Hou, J.; Zhu, B.; Min, Z.; Zhang, M.; Song, D.; Cheng, Y.; Wang, X. Targeting Roles of Inflammatory Microenvironment in Lung Cancer and Metastasis. Cancer Metast Rev. 2015, 34 (2), 319– 331, DOI: 10.1007/s10555-015-9570-4Google ScholarThere is no corresponding record for this reference.
- 7Calles, A.; Riess, J. W.; Brahmer, J. R. Checkpoint Blockade in Lung Cancer With Driver Mutation: Choose the Road Wisely. Am. Soc. Clin Oncol Educ Book 2020, 40 (40), 372– 384, DOI: 10.1200/EDBK_280795Google ScholarThere is no corresponding record for this reference.
- 8Cooper, A. J.; Sequist, L. V.; Lin, J. J. Third-Generation EGFR and ALK Inhibitors: Mechanisms of Resistance and Management. Nat. Rev. Clin Oncol 2022, 19 (8), 499– 514, DOI: 10.1038/s41571-022-00639-9Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsFyjtr%252FN&md5=9d2ce0593c9b0d6a05cc92fe023103ceThird-generation EGFR and ALK inhibitors: mechanisms of resistance and managementCooper, Alissa J.; Sequist, Lecia V.; Lin, Jessica J.Nature Reviews Clinical Oncology (2022), 19 (8), 499-514CODEN: NRCOAA; ISSN:1759-4774. (Nature Portfolio)A review. Abstr.: The discoveries of EGFR mutations and ALK rearrangements as actionable oncogenic drivers in non-small-cell lung cancer (NSCLC) has propelled a biomarker-directed treatment paradigm for patients with advanced-stage disease. Numerous EGFR and ALK tyrosine kinase inhibitors (TKIs) with demonstrated efficacy in patients with EGFR-mutant and ALK-rearranged NSCLCs have been developed, culminating in the availability of the highly effective third-generation TKIs osimertinib and lorlatinib, resp. Despite their marked efficacy, resistance to these agents remains an unsolved fundamental challenge. Both 'on-target' mechanisms (largely mediated by acquired resistance mutations in the kinase domains of EGFR or ALK) and 'off-target' mechanisms of resistance (mediated by non-target kinase alterations such as bypass signaling activation or phenotypic transformation) have been identified in patients with disease progression on osimertinib or lorlatinib. A growing understanding of the biol. and spectrum of these mechanisms of resistance has already begun to inform the development of more effective therapeutic strategies. In this Review, we discuss the development of third-generation EGFR and ALK inhibitors, predominant mechanisms of resistance, and approaches to tackling resistance in the clinic, ranging from novel fourth-generation TKIs to combination regimens and other investigational therapies.
- 9Schenk, E. L. Narrative Review: Immunotherapy in Anaplastic Lymphoma Kinase (ALK)+ Lung Cancer─Current Status and Future Directions. Transl Lung Cancer Res. 2023, 12 (2), 322– 336, DOI: 10.21037/tlcr-22-883Google ScholarThere is no corresponding record for this reference.
- 10Poltavets, A. S.; Vishnyakova, P. A.; Elchaninov, A. V.; Sukhikh, G. T.; Fatkhudinov, T. Kh. Macrophage Modification Strategies for Efficient Cell Therapy. Cells 2020, 9 (6), 1535, DOI: 10.3390/cells9061535Google ScholarThere is no corresponding record for this reference.
- 11Hirsch, F. R.; Scagliotti, G. V.; Mulshine, J. L.; Kwon, R.; Curran, W. J.; Wu, Y.-L.; Paz-Ares, L. Lung Cancer: Current Therapies and New Targeted Treatments. Lancet 2017, 389 (10066), 299– 311, DOI: 10.1016/S0140-6736(16)30958-8Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVamsbzP&md5=36ace1a455a803dc6e7af83dbffb2e34Lung cancer: current therapies and new targeted treatmentsHirsch, Fred R.; Scagliotti, Giorgio V.; Mulshine, James L.; Kwon, Regina; Curran, Walter J., Jr.; Wu, Yi-Long; Paz-Ares, LuisLancet (2017), 389 (10066), 299-311CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)Lung cancer is the most frequent cause of cancer-related deaths worldwide. Every year, 1·8 million people are diagnosed with lung cancer, and 1·6 million people die as a result of the disease. 5-yr survival rates vary from 4-17% depending on stage and regional differences. In this Seminar, we discuss existing treatment for patients with lung cancer and the promise of precision medicine, with special emphasis on new targeted therapies. Some subgroups, eg-patients with poor performance status and elderly patients-are not specifically addressed, because these groups require special treatment considerations and no frameworks have been established in terms of new targeted therapies. We discuss prevention and early detection of lung cancer with an emphasis on lung cancer screening. Although we acknowledge the importance of smoking prevention and cessation, this is a large topic beyond the scope of this Seminar.
- 12Bai, R.; Li, L.; Chen, X.; Chen, N.; Song, W.; Cui, J. Neoadjuvant and Adjuvant Immunotherapy: Opening New Horizons for Patients With Early-Stage Non-Small Cell Lung Cancer. Frontiers Oncol 2020, 10, 575472 DOI: 10.3389/fonc.2020.575472Google ScholarThere is no corresponding record for this reference.
- 13Mills, C. D.; Ley, K. M1 and M2Macrophages: The Chicken and the Egg of Immunity. J. Innate Immun 2014, 6 (6), 716– 726, DOI: 10.1159/000364945Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhslyrsbrK&md5=face8a56588de3398ff368a2697538d9M1 and M2 Macrophages: The Chicken and the Egg of ImmunityMills, Charles D.; Ley, KlausJournal of Innate Immunity (2014), 6 (6), 716-726CODEN: JIIOB2; ISSN:1662-811X. (S. Karger AG)The purpose of this perspective is to describe a crit. advance in understanding how immune responses work. Macrophages are required for all animal life: 'Inhibit' type macrophages in all animals (called M1) can rapidly kill pathogens, and are thus the primary host defense, and 'Heal' type macrophages (M2) routinely repair and maintain tissue integrity. Macrophages perform these activities in all animals without T cells, and also in T cell-deficient vertebrates. Although adaptive immunity can amplify macrophage polarization, the long-held notion that macrophages need to be 'activated' or 'alternatively activated' by T cells is incorrect; indeed, immunol. has had it backward. M1/M2-type macrophages necessarily direct T cells toward Th1- or Th2-like activities, resp. That such macrophage-innate activities are the central directing element in immune responses is a dramatic change in understanding how immune systems operate. Most important, this revelation is opening up whole new approaches to immunotherapy. For example, many modern diseases, such as cancer and atherosclerosis, may not display 'foreign' antigens. However, there are clear imbalances in M1/M2-type responses. Correcting such innate imbalances can result in better health. Macrophages are the chicken and the egg of immunity.
- 14Donovan, A.; Lima, C. A.; Pinkus, J. L.; Pinkus, G. S.; Zon, L. I.; Robine, S.; Andrews, N. C. The Iron Exporter Ferroportin/Slc40a1 Is Essential for Iron Homeostasis. Cell Metab 2005, 1 (3), 191– 200, DOI: 10.1016/j.cmet.2005.01.003Google ScholarThere is no corresponding record for this reference.
- 15Vinchi, F.; da Silva, M. C.; Ingoglia, G.; Petrillo, S.; Brinkman, N.; Zuercher, A.; Cerwenka, A.; Tolosano, E.; Muckenthaler, M. U. Hemopexin Therapy Reverts Heme-Induced Proinflammatory Phenotypic Switching of Macrophages in a Mouse Model of Sickle Cell Disease. Blood 2016, 127 (4), 473– 486, DOI: 10.1182/blood-2015-08-663245Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xht1Cis7zM&md5=49e09b37adb6ee5ad1e84b32cbe6c294Hemopexin therapy reverts heme-induced proinflammatory phenotypic switching of macrophages in a mouse model of sickle cell diseaseVinchi, Francesca; Costa da Silva, Milene; Ingoglia, Giada; Petrillo, Sara; Brinkman, Nathan; Zuercher, Adrian; Cerwenka, Adelheid; Tolosano, Emanuela; Muckenthaler, Martina U.Blood (2016), 127 (4), 473-486CODEN: BLOOAW; ISSN:1528-0020. (American Society of Hematology)Hemolytic diseases, such as sickle cell anemia and thalassemia, are characterized by enhanced release of Hb and heme into the circulation, heme-iron loading of reticulo-endothelial system macrophages, and chronic inflammation. Here we show that in addn. to activating the vascular endothelium, Hb and heme excess alters the macrophage phenotype in sickle cell disease. We demonstrate that exposure of cultured macrophages to hemolytic aged red blood cells, heme, or iron causes their functional phenotypic change toward a proinflammatory state. In addn., hemolysis and macrophage heme/iron accumulation in a mouse model of sickle disease trigger similar proinflammatory phenotypic alterations in hepatic macrophages. On the mechanistic level, this critically depends on reactive oxygen species prodn. and activation of the Toll-like receptor 4 signaling pathway. We further demonstrate that the heme scavenger hemopexin protects reticulo-endothelial macrophages from heme overload in heme-loaded Hx-null mice and reduces prodn. of cytokines and reactive oxygen species. Importantly, in sickle mice, the administration of human exogenous hemopexin attenuates the inflammatory phenotype of macrophages. Taken together, our data suggest that therapeutic administration of hemopexin is beneficial to counteract heme-driven macrophage-mediated inflammation and its pathophysiol. consequences in sickle cell disease.
- 16Recalcati, S.; Locati, M.; Marini, A.; Santambrogio, P.; Zaninotto, F.; De Pizzol, M.; Zammataro, L.; Girelli, D.; Cairo, G. Differential Regulation of Iron Homeostasis during Human Macrophage Polarized Activation. Eur. J. Immunol. 2010, 40 (3), 824– 835, DOI: 10.1002/eji.200939889Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjtVWktLg%253D&md5=762786d1111efa80786c318ae77b1318Differential regulation of iron homeostasis during human macrophage polarized activationRecalcati, Stefania; Locati, Massimo; Marini, Agnese; Santambrogio, Paolo; Zaninotto, Federica; De Pizzol, Maria; Zammataro, Luca; Girelli, Domenico; Cairo, GaetanoEuropean Journal of Immunology (2010), 40 (3), 824-835CODEN: EJIMAF; ISSN:0014-2980. (Wiley-VCH Verlag GmbH & Co. KGaA)Iron metab. in inflammation has been mostly characterized in macrophages exposed to pathogens or inflammatory conditions, mimicked by the combined action of LPS and IFN-γ (M1 polarization). However, macrophages can undergo an alternative type of activation stimulated by Th2 cytokines, and acquire a role in cell growth and tissue repair control (M2 polarization). We characterized the expression of genes related to iron homeostasis in fully differentiated unpolarized (M0), M1 and M2 human macrophages. The mol. signature of the M1 macrophages showed changes in gene expression (ferroportin repression and H ferritin induction) that favor iron sequestration in the reticuloendothelial system, a hallmark of inflammatory disorders, whereas the M2 macrophages had an expression profile (ferroportin upregulation and the downregulation of H ferritin and heme oxygenase) that enhanced iron release. The conditioned media from M2 macrophages promoted cell proliferation more efficiently than those of M1 cells and the effect was blunted by iron chelation. The role of ferroportin-mediated iron release was demonstrated by the absence of differences from the media of macrophages of a patient with loss of function ferroportin mutation. The distinct regulation of iron homeostasis in M2 macrophages provides insights into their role under pathophysiol. conditions.
- 17Recalcati, S.; Gammella, E.; Cairo, G. Ironing out Macrophage Immunometabolism. Pharm. 2019, 12 (2), 94, DOI: 10.3390/ph12020094Google ScholarThere is no corresponding record for this reference.
- 18Pawate, S.; Shen, Q.; Fan, F.; Bhat, N. R. Redox Regulation of Glial Inflammatory Response to Lipopolysaccharide and Interferonγ. J. Neurosci. Res. 2004, 77 (4), 540– 551, DOI: 10.1002/jnr.20180Google ScholarThere is no corresponding record for this reference.
- 19Mertens, C.; Marques, O.; Horvat, N. K.; Simonetti, M.; Muckenthaler, M. U.; Jung, M. The Macrophage Iron Signature in Health and Disease. Int. J. Mol. Sci. 2021, 22 (16), 8457, DOI: 10.3390/ijms22168457Google ScholarThere is no corresponding record for this reference.
- 20Fu, H.; Zhang, Z.; Li, D.; Lv, Q.; Chen, S.; Zhang, Z.; Wu, M. LncRNA PELATON, a Ferroptosis Suppressor and Prognositic Signature for GBM. Frontiers Oncol 2022, 12, 817737 DOI: 10.3389/fonc.2022.817737Google ScholarThere is no corresponding record for this reference.
- 21Yano, T.; Obata, Y.; Ishikawa, G.; Ichikawa, T. Enhancing Effect of High Dietary Iron on Lung Tumorigenesis in Mice. Cancer Lett. 1994, 76 (1), 57– 62, DOI: 10.1016/0304-3835(94)90134-1Google ScholarThere is no corresponding record for this reference.
- 22Zhao, H.; Tanaka, T.; Mitlitski, V.; Heeter, J.; Balazs, E. A.; Darzynkiewicz, Z. Protective Effect of Hyaluronate on Oxidative DNA Damage in WI-38 and A549 Cells. Int. J. Oncol. 2008, 32 (6), 1159– 1167, DOI: 10.3892/ijo_32_6_1159Google ScholarThere is no corresponding record for this reference.
- 23Shen, Y.; Li, X.; Zhao, B.; Xue, Y.; Wang, S.; Chen, X.; Yang, J.; Lv, H.; Shang, P. Iron Metabolism Gene Expression and Prognostic Features of Hepatocellular Carcinoma. J. Cell Biochem 2018, 119 (11), 9178– 9204, DOI: 10.1002/jcb.27184Google ScholarThere is no corresponding record for this reference.
- 24Torti, S. V.; Torti, F. M. Iron and Cancer: More Ore to Be Mined. Nat. Rev. Cancer 2013, 13 (5), 342– 355, DOI: 10.1038/nrc3495Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXlvF2isL8%253D&md5=9d950f3dab0010974d1ef25f8c9c1733Iron and cancer: more ore to be minedTorti, Suzy V.; Torti, Frank M.Nature Reviews Cancer (2013), 13 (5), 342-355CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Iron is an essential nutrient that facilitates cell proliferation and growth. However, iron also has the capacity to engage in redox cycling and free radical formation. Therefore, iron can contribute to both tumor initiation and tumor growth; recent work has also shown that iron has a role in the tumor microenvironment and in metastasis. Pathways of iron acquisition, efflux, storage and regulation are all perturbed in cancer, suggesting that reprogramming of iron metab. is a central aspect of tumor cell survival. Signalling through hypoxia-inducible factor (HIF) and WNT pathways may contribute to altered iron metab. in cancer. Targeting iron metabolic pathways may provide new tools for cancer prognosis and therapy.
- 25Land, W. G. Transfusion-Related Acute Lung Injury: The Work of DAMPs*. Transfus Med. Hemoth 2013, 40 (1), 3– 13, DOI: 10.1159/000345688Google ScholarThere is no corresponding record for this reference.
- 26Nairz, M.; Theurl, I.; Swirski, F. K.; Weiss, G. Pumping Iron”─How Macrophages Handle Iron at the Systemic, Microenvironmental, and Cellular Levels. Pflugers Arch. 2017, 469, 397– 418, DOI: 10.1007/s00424-017-1944-8Google ScholarThere is no corresponding record for this reference.
- 27da Silva, M. C.; Breckwoldt, M. O.; Vinchi, F.; Correia, M. P.; Stojanovic, A.; Thielmann, C. M.; Meister, M.; Muley, T.; Warth, A.; Platten, M.; Hentze, M. W.; Cerwenka, A.; Muckenthaler, M. U. Iron Induces Anti-Tumor Activity in Tumor-Associated Macrophages. Front Immunol 2017, 8, 1479, DOI: 10.3389/fimmu.2017.01479Google ScholarThere is no corresponding record for this reference.
- 28Thielmann, C. M.; da Silva, M. C.; Muley, T.; Meister, M.; Herpel, E.; Muckenthaler, M. U. Iron Accumulation in Tumor-Associated Macrophages Marks an Improved Overall Survival in Patients with Lung Adenocarcinoma. Sci. Rep-uk 2019, 9 (1), 11326 DOI: 10.1038/s41598-019-47833-xGoogle ScholarThere is no corresponding record for this reference.
- 29Bauer, T. A.; Horvat, N. K.; Marques, O.; Chocarro, S.; Mertens, C.; Colucci, S.; Schmitt, S.; Carrella, L. M.; Morsbach, S.; Koynov, K.; Fenaroli, F.; Blümler, P.; Jung, M.; Sotillo, R.; Hentze, M. W.; Muckenthaler, M. U.; Barz, M. Core Cross-Linked Polymeric Micelles for Specific Iron Delivery: Inducing Sterile Inflammation in Macrophages. Adv. Healthc Mater. 2021, 10, 2100385, DOI: 10.1002/adhm.202100385Google ScholarThere is no corresponding record for this reference.
- 30Schäfer, O.; Huesmann, D.; Barz, M. Poly(S-ethylsulfonyl-l-cysteines) for Chemoselective Disulfide Formation. Macromolecules 2016, 49 (21), 8146– 8153, DOI: 10.1021/acs.macromol.6b02064Google ScholarThere is no corresponding record for this reference.
- 31Klinker, K.; Schäfer, O.; Huesmann, D.; Bauer, T.; Capelôa, L.; Braun, L.; Stergiou, N.; Schinnerer, M.; Dirisala, A.; Miyata, K.; Osada, K.; Cabral, H.; Kataoka, K.; Barz, M. Secondary-Structure-Driven Self-Assembly of Reactive Polypept(o)Ides: Controlling Size, Shape, and Function of Core Cross-Linked Nanostructures. Angew. Chem., Int. Ed. 2017, 56 (32), 9608– 9613, DOI: 10.1002/anie.201702624Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFensrzE&md5=3476193910ff8e0647b88ce30bb96d74Secondary-Structure-Driven Self-Assembly of Reactive Polypept(o)ides: Controlling Size, Shape, and Function of Core Cross-Linked NanostructuresKlinker, Kristina; Schaefer, Olga; Huesmann, David; Bauer, Tobias; Capeloa, Leon; Braun, Lydia; Stergiou, Natascha; Schinnerer, Meike; Dirisala, Anjaneyulu; Miyata, Kanjiro; Osada, Kensuke; Cabral, Horacio; Kataoka, Kazunori; Barz, MatthiasAngewandte Chemie, International Edition (2017), 56 (32), 9608-9613CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Achieving precise control over the morphol. and function of polymeric nanostructures during self-assembly remains a challenge in materials as well as biomedical science, esp. when independent control over particle properties is desired. Herein, we report on nanostructures derived from amphiphilic block copolypept(o)ides by secondary-structure-directed self-assembly, presenting a strategy to adjust core polarity and function sep. from particle prepn. in a bioreversible manner. The peptide-inherent process of secondary-structure formation allows for the synthesis of spherical and worm-like core-crosslinked architectures from the same block copolymer, introducing a simple yet powerful approach to versatile peptide-based core-shell nanostructures.
- 32Bauer, T. A.; Schramm, J.; Fenaroli, F.; Siemer, S.; Seidl, C. I.; Rosenauer, C.; Bleul, R.; Stauber, R. H.; Koynov, K.; Maskos, M.; Barz, M. Complex Structures Made Simple – Continuous Flow Production of Core Cross-Linked Polymeric Micelles for Paclitaxel Pro-Drug-Delivery. Adv. Mater. 2023, 35 (21), e2210704 DOI: 10.1002/adma.202210704Google ScholarThere is no corresponding record for this reference.
- 33Bauer, T. A.; Imschweiler, J.; Muhl, C.; Weber, B.; Barz, M. Secondary Structure-Driven Self-Assembly of Thiol-Reactive Polypept(o)Ides. Biomacromolecules 2021, 22 (5), 2171– 2180, DOI: 10.1021/acs.biomac.1c00253Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXotFygsr0%253D&md5=57b9257d9a451bf5555d7ba3c8befca1Secondary Structure-Driven Self-Assembly of Thiol-Reactive Polypept(o)idesBauer, Tobias A.; Imschweiler, Jan; Muhl, Christian; Weber, Benjamin; Barz, MatthiasBiomacromolecules (2021), 22 (5), 2171-2180CODEN: BOMAF6; ISSN:1525-7797. (American Chemical Society)Secondary structure formation differentiates polypeptides from most of the other synthetic polymers, and the transitions from random coils to rod-like α-helixes or β-sheets represent an addnl. parameter to direct self-assembly and the morphol. of nanostructures. We investigated the influence of distinct secondary structures on the self-assembly of reactive amphiphilic polypept(o)ides. The individual morphologies can be preserved by core crosslinking via chemoselective disulfide bond formation. A series of thiol-responsive copolymers of racemic polysarcosine-block-poly(S-ethylsulfonyl-DL-cysteine) (pSar-b-p(DL)Cys), enantiopure polysarcosine-block-poly(S-ethylsulfonyl-L-cysteine) (pSar-b-p(L)Cys), and polysarcosine-block-poly(S-ethylsulfonyl-L-homocysteine) (pSar-b-p(L)Hcy) was prepd. by N-carboxyanhydride polymn. The secondary structure of the peptide segment varies from α-helixes (pSar-b-p(L)Hcy) to antiparallel β-sheets (pSar-b-p(L)Cys) and disrupted β-sheets (pSar-b-p(DL)Cys). When subjected to nanopptn., copolymers with antiparallel β-sheets display the strongest tendency to self-assemble, whereas disrupted β-sheets hardly induce aggregation. This translates to worm-like micelles, solely spherical micelles, or ellipsoidal structures, as analyzed by at. force microscopy and cryogenic transmission electron microscopy, which underlines the potential of secondary structure-driven self-assembly of synthetic polypeptides.
- 34Schäfer, O.; Klinker, K.; Braun, L.; Huesmann, D.; Schultze, J.; Koynov, K.; Barz, M. Combining Orthogonal Reactive Groups in Block Copolymers for Functional Nanoparticle Synthesis in a Single Step. ACS Macro Lett. 2017, 6 (10), 1140– 1145, DOI: 10.1021/acsmacrolett.7b00678Google ScholarThere is no corresponding record for this reference.
- 35Schäfer, O.; Huesmann, D.; Muhl, C.; Barz, M. Rethinking Cysteine Protective Groups: S-Alkylsulfonyl-l-cysteines for Chemoselective Disulfide Formation. Chem.─Eur. J. 2016, 22 (50), 18085– 18091, DOI: 10.1002/chem.201604391Google ScholarThere is no corresponding record for this reference.
- 36Schlüter, C.; Duchrow, M.; Wohlenberg, C.; Becker, M. H.; Key, G.; Flad, H. D.; Gerdes, J. The Cell Proliferation-Associated Antigen of Antibody Ki-67: A Very Large, Ubiquitous Nuclear Protein with Numerous Repeated Elements, Representing a New Kind of Cell Cycle-Maintaining Proteins. J. Cell Biol. 1993, 123 (3), 513– 522, DOI: 10.1083/jcb.123.3.513Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXltV2rtQ%253D%253D&md5=8eaf81089de4298736ef038380d67ea7The cell proliferation-associated antigen of antibody Ki-67: A very large, ubiquitous nuclear protein with numerous repeated elements, representing a new kind of cell cycle-maintaining proteinsSchlueter, Carsten; Duchrow, Michael; Wohlenberg, Claudia; Becker, Michael H. G.; Key, Goeran; Flad, Hans D.; Gerdes, JohannesJournal of Cell Biology (1993), 123 (3), 513-22CODEN: JCLBA3; ISSN:0021-9525.The antigen defined by monoclonal antibody (mAb) Ki-67 is a human nuclear protein, the expression of which is strictly assocd. with cell proliferation and which is widely used in routine pathol. as a proliferation marker to measure the growth fraction of cells in human tumors. Ki-67 detects a double band with apparent mol. wts. of 395 and 345 kDa in immunoblots of proteins from proliferating cells. The authors cloned and sequenced the full length cDNA, identified 2 differentially spliced isoforms of mRNA with open reading frames of 9768 and 8688 bp encoding for this cell proliferation-assocd. protein with calcd. mol. wts. of 358,761 Da and 319,508 Da, resp. New mAbs against a bacterially expressed part and a synthetic polypeptide deduced from the isolated cDNA react with the native Ki-67 antigen, thus providing a circle of evidence that the authors have cloned the authentic Ki-67 antigen cDNA. The central part of the Ki-67 antigen cDNA contains a large 6845-bp exon with 16 tandemly repeated 366-bp elements, the Ki-67 repeats, each including a highly conserved new motif of 66 bp, the Ki-67 motif, which encodes for the epitope detected by Ki-67. Computer anal. of the nucleic acid and the deduced amino acid sequence of the Ki-67 antigen confirmed that the cDNA encodes for a nuclear and short-lived protein without any significant homol. to known sequences. Ki-67 antigen-specific antisense oligonucleotides inhibit the proliferation of IM-9 cells, indicating that the Ki-67 antigen may be an abs. requirement for maintaining cell proliferation. Evidently, the Ki-67 antigen defines a new category of cell cycle-assocd. nuclear nonhistone proteins.
- 37Rogakou, E. P.; Pilch, D. R.; Orr, A. H.; Ivanova, V. S.; Bonner, W. M. DNA Double-Stranded Breaks Induce Histone H2AX Phosphorylation on Serine 139*. J. Biol. Chem. 1998, 273 (10), 5858– 5868, DOI: 10.1074/jbc.273.10.5858Google ScholarThere is no corresponding record for this reference.
- 38Butler, L. M.; Zhou, X.; Xu, W.-S.; Scher, H. I.; Rifkind, R. A.; Marks, P. A.; Richon, V. M. The Histone Deacetylase Inhibitor SAHA Arrests Cancer Cell Growth, up-Regulates Thioredoxin-Binding Protein-2, and down-Regulates Thioredoxin. Proc. National Acad. Sci. 2002, 99 (18), 11700– 11705, DOI: 10.1073/pnas.182372299Google ScholarThere is no corresponding record for this reference.
- 39Cai, B.; Kasikara, C.; Doran, A. C.; Ramakrishnan, R.; Birge, R. B.; Tabas, I. MerTK Signaling in Macrophages Promotes the Synthesis of Inflammation Resolution Mediators by Suppressing CaMKII Activity. Sci. Signal 2018, 11 (549), na, DOI: 10.1126/scisignal.aar3721Google ScholarThere is no corresponding record for this reference.
- 40MULERO, V.; WEI, X.; LIEW, F. Y.; BROCK, J. H. Regulation of Phagosomal Iron Release from Murine Macrophages by Nitric Oxide. Biochem. J. 2002, 365 (1), 127– 132, DOI: 10.1042/bj20011875Google ScholarThere is no corresponding record for this reference.
- 41Aldrovandi, M.; Conrad, M. Ferroptosis: The Good, the Bad and the Ugly. Cell Res. 2020, 30 (12), 1061– 1062, DOI: 10.1038/s41422-020-00434-0Google ScholarThere is no corresponding record for this reference.
- 42Lemaire, G.; Alvarez-Pachon, F.-J.; Beuneu, C.; Lepoivre, M.; Petit, J.-F. Differential Cytostatic Effects of NO Donors and NO Producing Cells. Free Radical Bio Med. 1999, 26 (9–10), 1274– 1283, DOI: 10.1016/S0891-5849(98)00331-1Google ScholarThere is no corresponding record for this reference.
- 43Van den Bossche, J.; Baardman, J.; Otto, N. A.; van der Velden, S.; Neele, A. E.; van den Berg, S. M.; Luque-Martin, R.; Chen, H.-J.; Boshuizen, M.; Ahmed, M.; Hoeksema, M. A.; de Vos, A. F.; de Winther, M. Mitochondrial Dysfunction Prevents Repolarization of Inflammatory Macrophages. Cell Reports 2016, 17 (3), 684– 696, DOI: 10.1016/j.celrep.2016.09.008Google ScholarThere is no corresponding record for this reference.
- 44Maddalo, D.; Manchado, E.; Concepcion, C. P.; Bonetti, C.; Vidigal, J. A.; Han, Y.-C.; Ogrodowski, P.; Crippa, A.; Rekhtman, N.; de Stanchina, E.; Lowe, S. W.; Ventura, A. In Vivo Engineering of Oncogenic Chromosomal Rearrangements with the CRISPR/Cas9 System. Nature 2014, 516, 423– 427, DOI: 10.1038/nature13902Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVemtrvL&md5=ded8f269a1342b56ecf0ec48f61af33aIn vivo engineering of oncogenic chromosomal rearrangements with the CRISPR/Cas9 systemMaddalo, Danilo; Manchado, Eusebio; Concepcion, Carla P.; Bonetti, Ciro; Vidigal, Joana A.; Han, Yoon-Chi; Ogrodowski, Paul; Crippa, Alessandra; Rekhtman, Natasha; de Stanchina, Elisa; Lowe, Scott W.; Ventura, AndreaNature (London, United Kingdom) (2014), 516 (7531), 423-427CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Chromosomal rearrangements have a central role in the pathogenesis of human cancers and often result in the expression of therapeutically actionable gene fusions. A recently discovered example is a fusion between the genes echinoderm microtubule-assocd. protein like 4 (EML4) and anaplastic lymphoma kinase (ALK), generated by an inversion on the short arm of chromosome 2: inv(2)(p21p23). The EML4-ALK oncogene is detected in a subset of human non-small cell lung cancers (NSCLC) and is clin. relevant because it confers sensitivity to ALK inhibitors. Despite their importance, modeling such genetic events in mice has proven challenging and requires complex manipulation of the germ line. Here we describe an efficient method to induce specific chromosomal rearrangements in vivo using viral-mediated delivery of the CRISPR/Cas9 system to somatic cells of adult animals. We apply it to generate a mouse model of Eml4-Alk-driven lung cancer. The resulting tumors invariably harbor the Eml4-Alk inversion, express the Eml4-Alk fusion gene, display histopathol. and mol. features typical of ALK+ human NSCLCs, and respond to treatment with ALK inhibitors. The general strategy described here substantially expands our ability to model human cancers in mice and potentially in other organisms.
- 45da Silva, M. C.; Breckwoldt, M. O.; Vinchi, F.; Correia, M. P.; Stojanovic, A.; Thielmann, C. M.; Meister, M.; Muley, T.; Warth, A.; Platten, M.; Hentze, M. W.; Cerwenka, A.; Muckenthaler, M. U. Iron Induces Anti-Tumor Activity in Tumor-Associated Macrophages. Front Immunol 2017, 8, 1479, DOI: 10.3389/fimmu.2017.01479Google ScholarThere is no corresponding record for this reference.
- 46Scagliotti, G.; Stahel, R. A.; Rosell, R.; Thatcher, N.; Soria, J.-C. ALK Translocation and Crizotinib in Non-Small Cell Lung Cancer: An Evolving Paradigm in Oncology Drug Development. Eur. J. Cancer 2012, 48 (7), 961– 973, DOI: 10.1016/j.ejca.2012.02.001Google ScholarThere is no corresponding record for this reference.
- 47Shrestha, N.; Nimick, M.; Dass, P.; Rosengren, R. J.; Ashton, J. C. Mechanisms of Suppression of Cell Growth by Dual Inhibition of ALK and MEK in ALK-Positive Non-Small Cell Lung Cancer. Sci. Rep-uk 2019, 9 (1), 18842 DOI: 10.1038/s41598-019-55376-4Google ScholarThere is no corresponding record for this reference.
- 48Guo, Q.; Liu, L.; Chen, Z.; Fan, Y.; Zhou, Y.; Yuan, Z.; Zhang, W. Current Treatments for Non-Small Cell Lung Cancer. Frontiers Oncol 2022, 12, 945102 DOI: 10.3389/fonc.2022.945102Google ScholarThere is no corresponding record for this reference.
- 49Zanganeh, S.; Hutter, G.; Spitler, R.; Lenkov, O.; Mahmoudi, M.; Shaw, A.; Pajarinen, J. S.; Nejadnik, H.; Goodman, S.; Moseley, M.; Coussens, L. M.; Daldrup-Link, H. E. Iron Oxide Nanoparticles Inhibit Tumour Growth by Inducing Pro-Inflammatory Macrophage Polarization in Tumour Tissues. Nat. Nanotechnol 2016, 11 (11), 986– 994, DOI: 10.1038/nnano.2016.168Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFyms7fO&md5=ddc5109f0cce0aa4b79adf816808fb4bIron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissuesZanganeh, Saeid; Hutter, Gregor; Spitler, Ryan; Lenkov, Olga; Mahmoudi, Morteza; Shaw, Aubie; Pajarinen, Jukka Sakari; Nejadnik, Hossein; Goodman, Stuart; Moseley, Michael; Coussens, Lisa Marie; Daldrup-Link, Heike ElisabethNature Nanotechnology (2016), 11 (11), 986-994CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)Until now, the Food and Drug Administration (FDA)-approved iron supplement ferumoxytol and other iron oxide nanoparticles have been used for treating iron deficiency, as contrast agents for magnetic resonance imaging and as drug carriers. Here, the authors show an intrinsic therapeutic effect of ferumoxytol on the growth of early mammary cancers, and lung cancer metastases in liver and lungs. In vitro, adenocarcinoma cells coincubated with ferumoxytol and macrophages showed increased caspase-3 activity. Macrophages exposed to ferumoxytol displayed increased mRNA assocd. with pro-inflammatory Th1-type responses. In vivo, ferumoxytol significantly inhibited growth of s.c. adenocarcinomas in mice. In addn., i.v. ferumoxytol treatment before i.v. tumor cell challenge prevented development of liver metastasis. Fluorescence-activated cell sorting (FACS) and histopathol. studies showed that the obsd. tumor growth inhibition was accompanied by increased presence of pro-inflammatory M1 macrophages in the tumor tissues. The results suggest that ferumoxytol could be applied 'off label' to protect the liver from metastatic seeds and potentiate macrophage-modulating cancer immunotherapies.
- 50Cameron, D. J.; Churchill, W. H. Cytotoxicity of Human Macrophages for Tumor Cells. Enhancement by Human Lymphocyte Mediators. J. Clin Invest 1979, 63 (5), 977– 984, DOI: 10.1172/JCI109398Google ScholarThere is no corresponding record for this reference.
- 51Hudson, S. V.; Miller, H. A.; Mahlbacher, G. E.; Saforo, D.; Beverly, L. J.; Arteel, G. E.; Frieboes, H. B. Computational/Experimental Evaluation of Liver Metastasis Post Hepatic Injury: Interactions with Macrophages and Transitional ECM. Sci. Rep-uk 2019, 9 (1), 15077 DOI: 10.1038/s41598-019-51249-yGoogle ScholarThere is no corresponding record for this reference.
- 52Bartha, L.; Eftimie, R. Mathematical Investigation into the Role of Macrophage Heterogeneity on the Temporal and Spatio-Temporal Dynamics of Non-Small Cell Lung Cancers. J. Theor. Biol. 2022, 549, 111207 DOI: 10.1016/j.jtbi.2022.111207Google ScholarThere is no corresponding record for this reference.
- 53Li, X.; Jolly, M. K.; George, J. T.; Pienta, K. J.; Levine, H. Computational Modeling of the Crosstalk Between Macrophage Polarization and Tumor Cell Plasticity in the Tumor Microenvironment. Frontiers Oncol 2019, 9, 10, DOI: 10.3389/fonc.2019.00010Google ScholarThere is no corresponding record for this reference.
- 54Hegedűs, C.; Kovács, K.; Polgár, Z.; Regdon, Z.; Szabó, É.; Robaszkiewicz, A.; Forman, H. J.; Martner, A.; Virág, L. Redox Control of Cancer Cell Destruction. Redox Biol. 2018, 16, 59– 74, DOI: 10.1016/j.redox.2018.01.015Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjtFOit7w%253D&md5=a2829fe76b47302092d8e0feb1e6de67Redox control of cancer cell destructionHegedus, Csaba; Kovacs, Katalin; Polgar, Zsuzsanna; Regdon, Zsolt; Szabo, Eva; Robaszkiewicz, Agnieszka; Forman, Henry Jay; Martner, Anna; Virag, LaszloRedox Biology (2018), 16 (), 59-74CODEN: RBEIB3; ISSN:2213-2317. (Elsevier B.V.)A review. Redox regulation has been proposed to control various aspects of carcinogenesis, cancer cell growth, metab., migration, invasion, metastasis and cancer vascularization. As cancer has many faces, the role of redox control in different cancers and in the numerous cancer-related processes often point in different directions. In this review, we focus on the redox control mechanisms of tumor cell destruction. The review covers the tumor-intrinsic role of oxidants derived from the redn. of oxygen and nitrogen in the control of tumor cell proliferation as well as the roles of oxidants and antioxidant systems in cancer cell death caused by traditional anticancer weapons (chemotherapeutic agents, radiotherapy, photodynamic therapy). Emphasis is also put on the role of oxidants and redox status in the outcome following interactions between cancer cells, cytotoxic lymphocytes and tumor infiltrating macrophages.
- 55Kim, S. J.; Kim, H. S.; Seo, Y. R. Understanding of ROS-Inducing Strategy in Anticancer Therapy. Oxid Med. Cell Longev 2019, 2019, 5381692 DOI: 10.1155/2019/5381692Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MbptlektA%253D%253D&md5=365ea194a3a48a0107703bce40d04de4Understanding of ROS-Inducing Strategy in Anticancer TherapyKim Su Ji; Kim Hyun Soo; Seo Young Rok; Kim Su Ji; Kim Hyun Soo; Seo Young RokOxidative medicine and cellular longevity (2019), 2019 (), 5381692 ISSN:.Redox homeostasis is essential for the maintenance of diverse cellular processes. Cancer cells have higher levels of reactive oxygen species (ROS) than normal cells as a result of hypermetabolism, but the redox balance is maintained in cancer cells due to their marked antioxidant capacity. Recently, anticancer therapies that induce oxidative stress by increasing ROS and/or inhibiting antioxidant processes have received significant attention. The acceleration of accumulative ROS disrupts redox homeostasis and causes severe damage in cancer cells. In this review, we describe ROS-inducing cancer therapy and the anticancer mechanism employed by prooxidative agents. To understand the comprehensive biological response to certain prooxidative anticancer drugs such as 2-methoxyestradiol, buthionine sulfoximine, cisplatin, doxorubicin, imexon, and motexafin gadolinium, we propose and visualize the drug-gene, drug-cell process, and drug-disease interactions involved in oxidative stress induction and antioxidant process inhibition as well as specific side effects of these drugs using pathway analysis with a big data-based text-mining approach. Our review will be helpful to improve the therapeutic effects of anticancer drugs by providing information about biological changes that occur in response to prooxidants. For future directions, there is still a need for pharmacogenomic studies on prooxidative agents as well as the molecular mechanisms underlying the effects of the prooxidants and/or antioxidant-inhibitor agents for effective anticancer therapy through selective killing of cancer cells.
- 56Wang, K.; Jiang, J.; Lei, Y.; Zhou, S.; Wei, Y.; Huang, C. Targeting Metabolic–Redox Circuits for Cancer Therapy. Trends Biochem. Sci. 2019, 44 (5), 401– 414, DOI: 10.1016/j.tibs.2019.01.001Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXpsVOgtA%253D%253D&md5=5b51ed067632d8be3067545e53792010Targeting Metabolic-Redox Circuits for Cancer TherapyWang, Kui; Jiang, Jingwen; Lei, Yunlong; Zhou, Shengtao; Wei, Yuquan; Huang, CanhuaTrends in Biochemical Sciences (2019), 44 (5), 401-414CODEN: TBSCDB; ISSN:0968-0004. (Elsevier Ltd.)A review. Metabolic alterations and elevated levels of reactive oxygen species (ROS) are two characteristics of cancer. The metabolic patterns of cancer cells are elaborately reprogrammed to fulfill the high biomass demands of rapid propagation. ROS, the byproducts of metabolic processes, are accumulated in cancer cells partially due to metabolic abnormalities or oncogenic mutations. To prevent oxidative damage, cancer cells can orchestrate metabolic adaptation to maintain redn.-oxidn. (redox) balance by producing reducing equiv. ROS, acting as second messengers, can in turn manipulate metabolic pathways by directly or indirectly affecting the function of metabolic enzymes. In this review we discuss how cancer cell metab. and redox signaling are intertwined, with an emphasis on the perspective of targeting metabolic-redox circuits for cancer therapy.
- 57Teppo, H.-R.; Soini, Y.; Karihtala, P. Reactive Oxygen Species-Mediated Mechanisms of Action of Targeted Cancer Therapy. Oxid Med. Cell Longev 2017, 2017, 1485283 DOI: 10.1155/2017/1485283Google ScholarThere is no corresponding record for this reference.
- 58Samarin, J.; Fabrowski, P.; Kurilov, R.; Nuskova, H.; Hummel-Eisenbeiss, J.; Pink, H.; Li, N.; Weru, V.; Alborzinia, H.; Yildiz, U.; Grob, L.; Taubert, M.; Czech, M.; Morgen, M.; Brandstädter, C.; Becker, K.; Mao, L.; Jayavelu, A. K.; Goncalves, A.; Uhrig, U.; Seiler, J.; Lyu, Y.; Diederichs, S.; Klingmüller, U.; Muckenthaler, M.; Kopp-Schneider, A.; Teleman, A.; Miller, A. K.; Gunkel, N. Low Level of Antioxidant Capacity Biomarkers but Not Target Overexpression Predicts Vulnerability to ROS-Inducing Drugs. Redox Biol. 2023, 62, 102639 DOI: 10.1016/j.redox.2023.102639Google ScholarThere is no corresponding record for this reference.
- 59Wang, Y.; Zhang, X.; Yang, L.; Xue, J.; Hu, G. Blockade of CCL2 Enhances Immunotherapeutic Effect of Anti-PD1 in Lung Cancer. J. Bone Oncol 2018, 11, 27– 32, DOI: 10.1016/j.jbo.2018.01.002Google ScholarThere is no corresponding record for this reference.
- 60Binnewies, M.; Roberts, E. W.; Kersten, K.; Chan, V.; Fearon, D. F.; Merad, M.; Coussens, L. M.; Gabrilovich, D. I.; Ostrand-Rosenberg, S.; Hedrick, C. C.; Vonderheide, R. H.; Pittet, M. J.; Jain, R. K.; Zou, W.; Howcroft, T. K.; Woodhouse, E. C.; Weinberg, R. A.; Krummel, M. F. Understanding the Tumor Immune Microenvironment (TIME) for Effective Therapy. Nat. Med. 2018, 24 (5), 541– 550, DOI: 10.1038/s41591-018-0014-xGoogle Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXosVCnsLk%253D&md5=c8a5823d49b18653b960f920c1fdbedfUnderstanding the tumor immune microenvironment (TIME) for effective therapyBinnewies, Mikhail; Roberts, Edward W.; Kersten, Kelly; Chan, Vincent; Fearon, Douglas F.; Merad, Miriam; Coussens, Lisa M.; Gabrilovich, Dmitry I.; Ostrand-Rosenberg, Suzanne; Hedrick, Catherine C.; Vonderheide, Robert H.; Pittet, Mikael J.; Jain, Rakesh K.; Zou, Weiping; Howcroft, T. Kevin; Woodhouse, Elisa C.; Weinberg, Robert A.; Krummel, Matthew F.Nature Medicine (New York, NY, United States) (2018), 24 (5), 541-550CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)A review. The clin. successes in immunotherapy have been both astounding and at the same time unsatisfactory. Countless patients with varied tumor types have seen pronounced clin. response with immunotherapeutic intervention; however, many more patients have experienced minimal or no clin. benefit when provided the same treatment. As technol. has advanced, so has the understanding of the complexity and diversity of the immune context of the tumor microenvironment and its influence on response to therapy. It has been possible to identify different subclasses of immune environment that have an influence on tumor initiation and response and therapy; by parsing the unique classes and subclasses of tumor immune microenvironment (TIME) that exist within a patient's tumor, the ability to predict and guide immunotherapeutic responsiveness will improve, and new therapeutic targets will be revealed.
- 61Zeisberger, S. M.; Odermatt, B.; Marty, C.; Zehnder-Fjällman, A. H. M.; Ballmer-Hofer, K.; Schwendener, R. A. Clodronate-Liposome-Mediated Depletion of Tumour-Associated Macrophages: A New and Highly Effective Antiangiogenic Therapy Approach. Br. J. Cancer 2006, 95 (3), 272– 281, DOI: 10.1038/sj.bjc.6603240Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xnsl2iu7o%253D&md5=10ef4032a292c1709a91b672f323a22eClodronate-liposome-mediated depletion of tumour-associated macrophages: a new and highly effective antiangiogenic therapy approachZeisberger, S. M.; Odermatt, B.; Marty, C.; Zehnder-Fjaellman, A. H. M.; Ballmer-Hofer, K.; Schwendener, R. A.British Journal of Cancer (2006), 95 (3), 272-281CODEN: BJCAAI; ISSN:0007-0920. (Nature Publishing Group)Tumor-assocd. macrophages, TAMs, play a pivotal role in tumor growth and metastasis by promoting tumor angiogenesis. Treatment with clodronate encapsulated in liposomes (clodrolip) efficiently depleted these phagocytic cells in the murine F9 teratocarcinoma and human A673 rhabdomyosarcoma mouse tumor models resulting in significant inhibition of tumor growth ranging from 75 to >92%, depending on therapy and schedule. Tumor inhibition was accompanied by a drastic redn. in blood vessel d. in the tumor tissue. Vascular endothelial growth factor (VEGF) is one of the major inducers of tumor angiogenesis and is also required for macrophage recruitment. The strongest effects were obsd. with the combination therapy of clodrolip and a VEGF-neutralizing antibody, whereas free clodronate was not significantly active. Immunohistol. evaluation of the tumors showed significant depletion of F4/80+ and MOMA-1+ and a less pronounced depletion of CD11b+ TAMs. Blood vessel staining (CD31) and quantification of the vessels as well as TAMs and tumor-assocd. dendritic cells (TADCs) in the A673 model showed redn. rates of 85 to >94%, even 9 days after the end of therapy. In addn., CD11c+ TADCs, which have been shown to potentially differentiate into endothelial-like cells upon stimulation by tumor released growth and differentiation factors, were similarly reduced by clodrolip or antibody treatment. These results validate clodrolip therapy in combination with angiogenesis inhibitors as a promising novel strategy for an indirect cancer therapy aimed at the hematopoietic precursor cells that stimulate tumor growth and dissemination and as a tool to study the role of macrophages and dendritic cells in tumorigenesis.
- 62Gubin, M. M.; Esaulova, E.; Ward, J. P.; Malkova, O. N.; Runci, D.; Wong, P.; Noguchi, T.; Arthur, C. D.; Meng, W.; Alspach, E.; Medrano, R. F. V.; Fronick, C.; Fehlings, M.; Newell, E. W.; Fulton, R. S.; Sheehan, K. C. F.; Oh, S. T.; Schreiber, R. D.; Artyomov, M. N. High-Dimensional Analysis Delineates Myeloid and Lymphoid Compartment Remodeling during Successful Immune-Checkpoint Cancer Therapy. Cell 2018, 175 (4), 1014– 1030, DOI: 10.1016/j.cell.2018.09.030Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFKjtbbM&md5=873a47c26202c2a47e7750d719cfb530High-Dimensional Analysis Delineates Myeloid and Lymphoid Compartment Remodeling during Successful Immune-Checkpoint Cancer TherapyGubin, Matthew M.; Esaulova, Ekaterina; Ward, Jeffrey P.; Malkova, Olga N.; Runci, Daniele; Wong, Pamela; Noguchi, Takuro; Arthur, Cora D.; Meng, Wei; Alspach, Elise; Medrano, Ruan F. V.; Fronick, Catrina; Fehlings, Michael; Newell, Evan W.; Fulton, Robert S.; Sheehan, Kathleen C. F.; Oh, Stephen T.; Schreiber, Robert D.; Artyomov, Maxim N.Cell (Cambridge, MA, United States) (2018), 175 (4), 1014-1030.e19CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Although current immune-checkpoint therapy (ICT) mainly targets lymphoid cells, it is assocd. with a broader remodeling of the tumor micro-environment. Here, using complementary forms of high-dimensional profiling, the authors define differences across all hematopoietic cells from syngeneic mouse tumors during unrestrained tumor growth or effective ICT. Unbiased assessment of gene expression of tumor-infiltrating cells by single-cell RNA sequencing (scRNAseq) and longitudinal assessment of cellular protein expression by mass cytometry (CyTOF) revealed significant remodeling of both the lymphoid and myeloid intratumoral compartments. Surprisingly, the authors obsd. multiple subpopulations of monocytes/macrophages, distinguishable by the markers CD206, CX3CR1, CD1d, and iNOS, that change over time during ICT in a manner partially dependent on IFNγ. The authors' data support the hypothesis that this macrophage polarization/activation results from effects on circulatory monocytes and early macrophages entering tumors, rather than on pre-polarized mature intratumoral macrophages.
- 63Giordano, S.; Petrelli, A. From Single- to Multi-Target Drugs in Cancer Therapy: When Aspecificity Becomes an Advantage. Curr. Med. Chem. 2008, 15 (5), 422– 432, DOI: 10.2174/092986708783503212Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXnsVyisL0%253D&md5=d90df6a670921b6fad58caa46db01fa5From single- to multi-target drugs in cancer therapy: when a specificity becomes an advantagePetrelli, A.; Giordano, S.Current Medicinal Chemistry (2008), 15 (5), 422-432CODEN: CMCHE7; ISSN:0929-8673. (Bentham Science Publishers Ltd.)A review. Targeted therapies by means of compds. that inhibit a specific target mol. represent a new perspective in the treatment of cancer. In contrast to conventional chemotherapy which acts on all dividing cells generating toxic effects and damage of normal tissues, targeted drugs allow to hit, in a more specific manner, subpopulations of cells directly involved in tumor progression. Mols. controlling cell proliferation and death, such as Tyrosine Kinase Receptors (RTKs) for growth factors, are among the best targets for this type of therapeutic approach. Two classes of compds. targeting RTKs are currently used in clin. practice: monoclonal antibodies and tyrosine kinase inhibitors. The era of targeted therapy began with the approval of Trastuzumab, a monoclonal antibody against HER2, for treatment of metastatic breast cancer, and Imatinib, a small tyrosine kinase inhibitor targeting BCR-Abl, in Chronic Myeloid Leukemia. Despite the initial enthusiasm for the efficacy of these treatments, clinicians had to face soon the problem of relapse, as almost invariably cancer patients developed drug resistance, often due to the activation of alternative RTKs pathways. In this view, the rationale at the basis of targeting drugs is radically shifting. In the past, the main effort was aimed at developing highly specific inhibitors acting on single RTKs. Now, there is a general agreement that mols. interfering simultaneously with multiple RTKs might be more effective than single target agents. With the recent approval by FDA of Sorafenib and Sunitinib - targeting VEGFR, PDGFR, FLT-3 and c-Kit - a different scenario has been emerging, where a new generation of anti-cancer drugs, able to inhibit more than one pathway, would probably play a major role.
- 64Bedard, P. L.; Hyman, D. M.; Davids, M. S.; Siu, L. L. Small Molecules, Big Impact: 20 Years of Targeted Therapy in Oncology. Lancet 2020, 395 (10229), 1078– 1088, DOI: 10.1016/S0140-6736(20)30164-1Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlvFGrt7c%253D&md5=1538cc035cda58fae77a81025d485027Small molecules, big impact: 20 years of targeted therapy in oncologyBedard, Philippe L.; Hyman, David M.; Davids, Matthew S.; Siu, Lillian L.Lancet (2020), 395 (10229), 1078-1088CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)A review. The identification of mol. targets and the growing knowledge of their cellular functions have led to the development of small mol. inhibitors as a major therapeutic class for cancer treatment. Both multitargeted and highly selective kinase inhibitors are used for the treatment of advanced treatment-resistant cancers, and many have also achieved regulatory approval for early clin. settings as adjuvant therapies or as first-line options for recurrent or metastatic disease. Lessons learned from the development of these agents can accelerate the development of next-generation inhibitors to optimize the therapeutic index, overcome drug resistance, and establish combination therapies. The future of small mol. inhibitors is promising as there is the potential to investigate novel difficult-to-drug targets, to apply predictive non-clin. models to select promising drug candidates for human evaluation, and to use dynamic clin. trial interventions with liq. biopsies to deliver precision medicine.
- 65Greten, F. R.; Grivennikov, S. I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51 (1), 27– 41, DOI: 10.1016/j.immuni.2019.06.025Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlyls7jP&md5=0d39409f4a26c18100163b7b49367d46Inflammation and Cancer: Triggers, Mechanisms, and ConsequencesGreten, Florian R.; Grivennikov, Sergei I.Immunity (2019), 51 (1), 27-41CODEN: IUNIEH; ISSN:1074-7613. (Elsevier Inc.)A review. Inflammation predisposes to the development of cancer and promotes all stages of tumorigenesis. Cancer cells, as well as surrounding stromal and inflammatory cells, engage in well-orchestrated reciprocal interactions to form an inflammatory tumor microenvironment (TME). Cells within the TME are highly plastic, continuously changing their phenotypic and functional characteristics. Here, we review the origins of inflammation in tumors, and the mechanisms whereby inflammation drives tumor initiation, growth, progression, and metastasis. We discuss how tumor-promoting inflammation closely resembles inflammatory processes typically found during development, immunity, maintenance of tissue homeostasis, or tissue repair and illuminate the distinctions between tissue-protective and pro-tumorigenic inflammation, including spatiotemporal considerations. Defining the cornerstone rules of engagement governing mol. and cellular mechanisms of tumor-promoting inflammation will be essential for further development of anti-cancer therapies.
- 66Ries, C. H.; Cannarile, M. A.; Hoves, S.; Benz, J.; Wartha, K.; Runza, V.; Rey-Giraud, F.; Pradel, L. P.; Feuerhake, F.; Klaman, I.; Jones, T.; Jucknischke, U.; Scheiblich, S.; Kaluza, K.; Gorr, I. H.; Walz, A.; Abiraj, K.; Cassier, P. A.; Sica, A.; Gomez-Roca, C.; de Visser, K. E.; Italiano, A.; Le Tourneau, C.; Delord, J.-P.; Levitsky, H.; Blay, J.-Y.; Rüttinger, D. Targeting Tumor-Associated Macrophages with Anti-CSF-1R Antibody Reveals a Strategy for Cancer Therapy. Cancer Cell 2014, 25 (6), 846– 859, DOI: 10.1016/j.ccr.2014.05.016Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXpslCntrw%253D&md5=023e0585a753021f10b204c6aeb72d4bTargeting Tumor-Associated Macrophages with Anti-CSF-1R Antibody Reveals a Strategy for Cancer TherapyRies, Carola H.; Cannarile, Michael A.; Hoves, Sabine; Benz, Joerg; Wartha, Katharina; Runza, Valeria; Rey-Giraud, Flora; Pradel, Leon P.; Feuerhake, Friedrich; Klaman, Irina; Jones, Tobin; Jucknischke, Ute; Scheiblich, Stefan; Kaluza, Klaus; Gorr, Ingo H.; Walz, Antje; Abiraj, Keelara; Cassier, Philippe A.; Sica, Antonio; Gomez-Roca, Carlos; de Visser, Karin E.; Italiano, Antoine; Le Tourneau, Christophe; Delord, Jean-Pierre; Levitsky, Hyam; Blay, Jean-Yves; Ruettinger, DominikCancer Cell (2014), 25 (6), 846-859CODEN: CCAECI; ISSN:1535-6108. (Elsevier Inc.)Macrophage infiltration has been identified as an independent poor prognostic factor in several cancer types. The major survival factor for these macrophages is macrophage colony-stimulating factor 1 (CSF-1). We generated a monoclonal antibody (RG7155) that inhibits CSF-1 receptor (CSF-1R) activation. In vitro RG7155 treatment results in cell death of CSF-1-differentiated macrophages. In animal models, CSF-1R inhibition strongly reduces F4/80+ tumor-assocd. macrophages accompanied by an increase of the CD8+/CD4+ T cell ratio. Administration of RG7155 to patients led to striking redns. of CSF-1R+CD163+ macrophages in tumor tissues, which translated into clin. objective responses in diffuse-type giant cell tumor (Dt-GCT) patients.
- 67Muzumdar, M. D.; Tasic, B.; Miyamichi, K.; Li, L.; Luo, L. A Global Double-fluorescent Cre Reporter Mouse. Genesis 2007, 45 (9), 593– 605, DOI: 10.1002/dvg.20335Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtlWjs7nK&md5=0de0e4d9a2768c46b17023656cada120A global double-fluorescent Cre reporter mouseMuzumdar, Mandar Deepak; Tasic, Bosiljka; Miyamichi, Kazunari; Li, Ling; Luo, LiqunGenesis (Hoboken, NJ, United States) (2007), 45 (9), 593-605CODEN: GNESFY; ISSN:1526-954X. (Wiley-Liss, Inc.)The Cre/loxP system has been used extensively for conditional mutagenesis in mice. Reporters of Cre activity are important for defining the spatial and temporal extent of Cre-mediated recombination. Here we describe mT/mG, a double-fluorescent Cre reporter mouse that expresses membrane-targeted tandem dimer Tomato (mT) prior to Cre-mediated excision and membrane-targeted green fluorescent protein (mG) after excision. We show that reporter expression is nearly ubiquitous, allowing visualization of fluorescent markers in live and fixed samples of all tissues examd. We further demonstrate that mG labeling is Cre-dependent, complementary to mT at single cell resoln., and distinguishable by fluorescence-activated cell sorting. Both membrane-targeted markers outline cell morphol., highlight membrane structures, and permit visualization of fine cellular processes. In addn. to serving as a global Cre reporter, the mT/mG mouse may also be used as a tool for lineage tracing, transplantation studies, and anal. of cell morphol. in vivo.
- 68Oikonomou, N.; Mouratis, M.-A.; Tzouvelekis, A.; Kaffe, E.; Valavanis, C.; Vilaras, G.; Karameris, A.; Prestwich, G. D.; Bouros, D.; Aidinis, V. Pulmonary Autotaxin Expression Contributes to the Pathogenesis of Pulmonary Fibrosis. Am. J. Resp Cell Mol. 2012, 47 (5), 566– 574, DOI: 10.1165/rcmb.2012-0004OCGoogle ScholarThere is no corresponding record for this reference.
- 69Guida, C.; Altamura, S.; Klein, F. A.; Galy, B.; Boutros, M.; Ulmer, A. J.; Hentze, M. W.; Muckenthaler, M. U. A Novel Inflammatory Pathway Mediating Rapid Hepcidin-Independent Hypoferremia. Blood 2015, 125 (14), 2265– 2275, DOI: 10.1182/blood-2014-08-595256Google ScholarThere is no corresponding record for this reference.
- 70Hodgkin, P. D.; Lee, J. H.; Lyons, A. B. B Cell Differentiation and Isotype Switching Is Related to Division Cycle Number. J. Exp Medicine 1996, 184 (1), 277– 281, DOI: 10.1084/jem.184.1.277Google ScholarThere is no corresponding record for this reference.
- 71Müller, T.; Kalxdorf, M.; Longuespée, R.; Kazdal, D. N.; Stenzinger, A.; Krijgsveld, J. Automated Sample Preparation with SP3 for Low-input Clinical Proteomics. Mol. Syst. Biol. 2020, 16 (1), e9111 DOI: 10.15252/msb.20199111Google Scholar71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFOis7o%253D&md5=5bf4201cc5b52478697c68c60cae6ee0Automated sample preparation with SP3 for low-input clinical proteomicsMueller, Torsten; Kalxdorf, Mathias; Longuespee, Remi; Kazdal, Daniel N.; Stenzinger, Albrecht; Krijgsveld, JeroenMolecular Systems Biology (2020), 16 (1), e9111CODEN: MSBOC3; ISSN:1744-4292. (Wiley-VCH Verlag GmbH & Co. KGaA)High-throughput and streamlined workflows are essential in clin. proteomics for standardized processing of samples from a variety of sources, including fresh-frozen tissue, FFPE tissue, or blood. To reach this goal, we have implemented single-pot solid-phase-enhanced sample prepn. (SP3) on a liq. handling robot for automated processing (autoSP3) of tissue lysates in a 96-well format. AutoSP3 performs unbiased protein purifn. and digestion, and delivers peptides that can be directly analyzed by LCMS, thereby significantly reducing hands-on time, reducing variability in protein quantification, and improving longitudinal reproducibility. We demonstrate the distinguishing ability of autoSP3 to process low-input samples, reproducibly quantifying 500-1,000 proteins from 100 to 1,000 cells. Furthermore, we applied this approach to a cohort of clin. FFPE pulmonary adenocarcinoma (ADC) samples and recapitulated their sepn. into known histol. growth patterns. Finally, we integrated autoSP3 with AFA ultrasonication for the automated end-to-end sample prepn. and LCMS anal. of 96 intact tissue samples. Collectively, this constitutes a generic, scalable, and cost-effective workflow with minimal manual intervention, enabling reproducible tissue proteomics in a broad range of clin. and non-clin. applications.
- 72Heming, S.; Hansen, P.; Vlasov, A.; Schwörer, F.; Schaumann, S.; Frolovaitė, P.; Lehmann, W.-D.; Timmer, J.; Schilling, M.; Helm, B.; Klingmüller, U. MSPypeline: A Python Package for Streamlined Data Analysis of Mass Spectrometry-Based Proteomics. Bioinform Adv. 2022, 2 (1), na, DOI: 10.1093/bioadv/vbac004Google ScholarThere is no corresponding record for this reference.
- 73Ritchie, M. E.; Phipson, B.; Wu, D.; Hu, Y.; Law, C. W.; Shi, W.; Smyth, G. K. Limma Powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic Acids Res. 2015, 43 (7), e47– e47, DOI: 10.1093/nar/gkv007Google Scholar73https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFaiu7%252FN&md5=e8a5f44160c650ad89adc48a9912bd96limma powers differential expression analyses for RNA-sequencing and microarray studiesRitchie, Matthew E.; Phipson, Belinda; Wu, Di; Hu, Yifang; Law, Charity W.; Shi, Wei; Smyth, Gordon K.Nucleic Acids Research (2015), 43 (7), e47/1-e47/13CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Limma is an R/Bioconductor software package that provides an integrated soln. for analyzing data from gene expression expts. It contains rich features for handling complex exptl. designs and for information borrowing to overcome the problem of small sample sizes. Over the past decade, limma has been a popular choice for gene discovery through differential expression analyses of microarray and high-throughput PCR data. The package contains particularly strong facilities for reading, normalizing and exploring such data. Recently, the capabilities of limma have been significantly expanded in two important directions. First, the package can now perform both differential expression and differential splicing analyses of RNA sequencing (RNA-seq) data. All the downstream anal. tools previously restricted to microarray data are now available for RNA-seq as well. These capabilities allow users to analyze both RNA-seq and microarray data with very similar pipelines. Second, the package is now able to go past the traditional gene-wise expression analyses in a variety of ways, analyzing expression profiles in terms of co-regulated sets of genes or in terms of higher-order expression signatures. This provides enhanced possibilities for biol. interpretation of gene expression differences. This article reviews the philosophy and design of the limma package, summarizing both new and historical features, with an emphasis on recent enhancements and features that have not been previously described.
- 74Zhou, Y.; Zhou, B.; Pache, L.; Chang, M.; Khodabakhshi, A. H.; Tanaseichuk, O.; Benner, C.; Chanda, S. K. Metascape Provides a Biologist-Oriented Resource for the Analysis of Systems-Level Datasets. Nat. Commun. 2019, 10 (1), 1523, DOI: 10.1038/s41467-019-09234-6Google Scholar74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M%252Fhtl2rtg%253D%253D&md5=cb130c08668f9c8221f464a84039164dMetascape provides a biologist-oriented resource for the analysis of systems-level datasetsZhou Yingyao; Zhou Bin; Khodabakhshi Alireza Hadj; Tanaseichuk Olga; Pache Lars; Chanda Sumit K; Chang Max; Benner ChristopherNature communications (2019), 10 (1), 1523 ISSN:.A critical component in the interpretation of systems-level studies is the inference of enriched biological pathways and protein complexes contained within OMICs datasets. Successful analysis requires the integration of a broad set of current biological databases and the application of a robust analytical pipeline to produce readily interpretable results. Metascape is a web-based portal designed to provide a comprehensive gene list annotation and analysis resource for experimental biologists. In terms of design features, Metascape combines functional enrichment, interactome analysis, gene annotation, and membership search to leverage over 40 independent knowledgebases within one integrated portal. Additionally, it facilitates comparative analyses of datasets across multiple independent and orthogonal experiments. Metascape provides a significantly simplified user experience through a one-click Express Analysis interface to generate interpretable outputs. Taken together, Metascape is an effective and efficient tool for experimental biologists to comprehensively analyze and interpret OMICs-based studies in the big data era.
- 75Torrance, J. D.; Bothwell, T. H. A Simple Technique for Measuring Storage Iron Concentrations in Formalinised Liver Samples. South Afr J. Medical Sci. 1968, 33 (1), 9– 11Google ScholarThere is no corresponding record for this reference.
- 76Stirling, D. R.; Swain-Bowden, M. J.; Lucas, A. M.; Carpenter, A. E.; Cimini, B. A.; Goodman, A. CellProfiler 4: Improvements in Speed, Utility and Usability. Bmc Bioinformatics 2021, 22 (1), 433, DOI: 10.1186/s12859-021-04344-9Google Scholar76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2crmtVyksA%253D%253D&md5=048dceafeec6cbb75e0b3c7886b2722aCellProfiler 4: improvements in speed, utility and usabilityStirling David R; Lucas Alice M; Carpenter Anne E; Cimini Beth A; Goodman Allen; Swain-Bowden Madison JBMC bioinformatics (2021), 22 (1), 433 ISSN:.BACKGROUND: Imaging data contains a substantial amount of information which can be difficult to evaluate by eye. With the expansion of high throughput microscopy methodologies producing increasingly large datasets, automated and objective analysis of the resulting images is essential to effectively extract biological information from this data. CellProfiler is a free, open source image analysis program which enables researchers to generate modular pipelines with which to process microscopy images into interpretable measurements. RESULTS: Herein we describe CellProfiler 4, a new version of this software with expanded functionality. Based on user feedback, we have made several user interface refinements to improve the usability of the software. We introduced new modules to expand the capabilities of the software. We also evaluated performance and made targeted optimizations to reduce the time and cost associated with running common large-scale analysis pipelines. CONCLUSIONS: CellProfiler 4 provides significantly improved performance in complex workflows compared to previous versions. This release will ensure that researchers will have continued access to CellProfiler's powerful computational tools in the coming years.
- 77Vizcaíno, J. A.; Côté, R. G.; Csordas, A.; Dianes, J. A.; Fabregat, A.; Foster, J. M.; Griss, J.; Alpi, E.; Birim, M.; Contell, J.; O’Kelly, G.; Schoenegger, A.; Ovelleiro, D.; Pérez-Riverol, Y.; Reisinger, F.; Ríos, D.; Wang, R.; Hermjakob, H. The Proteomics Identifications (PRIDE) Database and Associated Tools: Status in 2013. Nucleic Acids Res. 2012, 41 (D1), D1063– D1069, DOI: 10.1093/nar/gks1262Google ScholarThere is no corresponding record for this reference.
Cited By
This article is cited by 1 publications.
- Raghav Soni, Atsushi Mahara. Modulation of Macrophage Polarization via Fluorescein Conjugation with Iron Oxide Nanoparticles for MR Imaging. ACS Applied Nano Materials 2024, 7
(19)
, 22417-22422. https://doi.org/10.1021/acsanm.4c02947
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 77 other publications.
- 1Sung, H.; Ferlay, J.; Siegel, R. L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. Ca Cancer J. Clin 2021, 71 (3), 209– 249, DOI: 10.3322/caac.216601https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3srpsVKnug%253D%253D&md5=f3e54fabe5ac5f2397292bf9a97ce5bfGlobal Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 CountriesSung Hyuna; Siegel Rebecca L; Jemal Ahmedin; Ferlay Jacques; Laversanne Mathieu; Soerjomataram Isabelle; Bray FreddieCA: a cancer journal for clinicians (2021), 71 (3), 209-249 ISSN:.This article provides an update on the global cancer burden using the GLOBOCAN 2020 estimates of cancer incidence and mortality produced by the International Agency for Research on Cancer. Worldwide, an estimated 19.3 million new cancer cases (18.1 million excluding nonmelanoma skin cancer) and almost 10.0 million cancer deaths (9.9 million excluding nonmelanoma skin cancer) occurred in 2020. Female breast cancer has surpassed lung cancer as the most commonly diagnosed cancer, with an estimated 2.3 million new cases (11.7%), followed by lung (11.4%), colorectal (10.0 %), prostate (7.3%), and stomach (5.6%) cancers. Lung cancer remained the leading cause of cancer death, with an estimated 1.8 million deaths (18%), followed by colorectal (9.4%), liver (8.3%), stomach (7.7%), and female breast (6.9%) cancers. Overall incidence was from 2-fold to 3-fold higher in transitioned versus transitioning countries for both sexes, whereas mortality varied <2-fold for men and little for women. Death rates for female breast and cervical cancers, however, were considerably higher in transitioning versus transitioned countries (15.0 vs 12.8 per 100,000 and 12.4 vs 5.2 per 100,000, respectively). The global cancer burden is expected to be 28.4 million cases in 2040, a 47% rise from 2020, with a larger increase in transitioning (64% to 95%) versus transitioned (32% to 56%) countries due to demographic changes, although this may be further exacerbated by increasing risk factors associated with globalization and a growing economy. Efforts to build a sustainable infrastructure for the dissemination of cancer prevention measures and provision of cancer care in transitioning countries is critical for global cancer control.
- 2Sica, A.; Mantovani, A. Macrophage Plasticity and Polarization: In Vivo Veritas. J. Clin Invest 2012, 122 (3), 787– 795, DOI: 10.1172/JCI596432https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XjsV2ms7g%253D&md5=2789f6879df4dea15e294b335f086598Macrophage plasticity and polarization: in vivo veritasSica, Antonio; Mantovani, AlbertoJournal of Clinical Investigation (2012), 122 (3), 787-795CODEN: JCINAO; ISSN:0021-9738. (American Society for Clinical Investigation)A review. Diversity and plasticity are hallmarks of cells of the monocyte-macrophage lineage. In response to IFNs, Toll-like receptor engagement, or IL-4/IL-13 signaling, macrophages undergo M1 (classical) or M2 (alternative) activation, which represent extremes of a continuum in a universe of activation states. Progress has now been made in defining the signaling pathways, transcriptional networks, and epigenetic mechanisms underlying M1-M2 or M2-like polarized activation. Functional skewing of mononuclear phagocytes occurs in vivo under physiol. conditions (e.g., ontogenesis and pregnancy) and in pathol. (allergic and chronic inflammation, tissue repair, infection, and cancer). However, in selected preclin. and clin. conditions, coexistence of cells in different activation states and unique or mixed phenotypes have been obsd., a reflection of dynamic changes and complex tissue-derived signals. The identification of mechanisms and mols. assocd. with macrophage plasticity and polarized activation provides a basis for macrophage-centered diagnostic and therapeutic strategies.
- 3Franklin, R. A.; Li, M. O. Ontogeny of Tumor-Associated Macrophages and Its Implication in Cancer Regulation. Trends Cancer 2016, 2 (1), 20– 34, DOI: 10.1016/j.trecan.2015.11.004There is no corresponding record for this reference.
- 4Sedighzadeh, S. S.; Khoshbin, A. P.; Razi, S.; Keshavarz-Fathi, M.; Rezaei, N. A Narrative Review of Tumor-Associated Macrophages in Lung Cancer: Regulation of Macrophage Polarization and Therapeutic Implications. Transl Lung Cancer Res. 2021, 10 (4), 1889– 1916, DOI: 10.21037/tlcr-20-1241There is no corresponding record for this reference.
- 5Condeelis, J.; Pollard, J. W. Macrophages: Obligate Partners for Tumor Cell Migration, Invasion, and Metastasis. Cell 2006, 124 (2), 263– 266, DOI: 10.1016/j.cell.2006.01.0075https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xht1KktLo%253D&md5=ddd258f9109498d4f1c252bfc5bc0a63Macrophages: obligate partners for tumor cell migration, invasion, and metastasisCondeelis, John; Pollard, Jeffrey W.Cell (Cambridge, MA, United States) (2006), 124 (2), 263-266CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Macrophages within the tumor microenvironment facilitate angiogenesis and extracellular-matrix breakdown and remodeling and promote tumor cell motility. Recent studies reveal that direct communication between macrophages and tumor cells leads to invasion and egress of tumor cells into the blood vessels (intravasation). Thus, macrophages are at the center of the invasion microenvironment and are an important drug target for cancer therapy.
- 6Shi, L.; Wang, L.; Hou, J.; Zhu, B.; Min, Z.; Zhang, M.; Song, D.; Cheng, Y.; Wang, X. Targeting Roles of Inflammatory Microenvironment in Lung Cancer and Metastasis. Cancer Metast Rev. 2015, 34 (2), 319– 331, DOI: 10.1007/s10555-015-9570-4There is no corresponding record for this reference.
- 7Calles, A.; Riess, J. W.; Brahmer, J. R. Checkpoint Blockade in Lung Cancer With Driver Mutation: Choose the Road Wisely. Am. Soc. Clin Oncol Educ Book 2020, 40 (40), 372– 384, DOI: 10.1200/EDBK_280795There is no corresponding record for this reference.
- 8Cooper, A. J.; Sequist, L. V.; Lin, J. J. Third-Generation EGFR and ALK Inhibitors: Mechanisms of Resistance and Management. Nat. Rev. Clin Oncol 2022, 19 (8), 499– 514, DOI: 10.1038/s41571-022-00639-98https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsFyjtr%252FN&md5=9d2ce0593c9b0d6a05cc92fe023103ceThird-generation EGFR and ALK inhibitors: mechanisms of resistance and managementCooper, Alissa J.; Sequist, Lecia V.; Lin, Jessica J.Nature Reviews Clinical Oncology (2022), 19 (8), 499-514CODEN: NRCOAA; ISSN:1759-4774. (Nature Portfolio)A review. Abstr.: The discoveries of EGFR mutations and ALK rearrangements as actionable oncogenic drivers in non-small-cell lung cancer (NSCLC) has propelled a biomarker-directed treatment paradigm for patients with advanced-stage disease. Numerous EGFR and ALK tyrosine kinase inhibitors (TKIs) with demonstrated efficacy in patients with EGFR-mutant and ALK-rearranged NSCLCs have been developed, culminating in the availability of the highly effective third-generation TKIs osimertinib and lorlatinib, resp. Despite their marked efficacy, resistance to these agents remains an unsolved fundamental challenge. Both 'on-target' mechanisms (largely mediated by acquired resistance mutations in the kinase domains of EGFR or ALK) and 'off-target' mechanisms of resistance (mediated by non-target kinase alterations such as bypass signaling activation or phenotypic transformation) have been identified in patients with disease progression on osimertinib or lorlatinib. A growing understanding of the biol. and spectrum of these mechanisms of resistance has already begun to inform the development of more effective therapeutic strategies. In this Review, we discuss the development of third-generation EGFR and ALK inhibitors, predominant mechanisms of resistance, and approaches to tackling resistance in the clinic, ranging from novel fourth-generation TKIs to combination regimens and other investigational therapies.
- 9Schenk, E. L. Narrative Review: Immunotherapy in Anaplastic Lymphoma Kinase (ALK)+ Lung Cancer─Current Status and Future Directions. Transl Lung Cancer Res. 2023, 12 (2), 322– 336, DOI: 10.21037/tlcr-22-883There is no corresponding record for this reference.
- 10Poltavets, A. S.; Vishnyakova, P. A.; Elchaninov, A. V.; Sukhikh, G. T.; Fatkhudinov, T. Kh. Macrophage Modification Strategies for Efficient Cell Therapy. Cells 2020, 9 (6), 1535, DOI: 10.3390/cells9061535There is no corresponding record for this reference.
- 11Hirsch, F. R.; Scagliotti, G. V.; Mulshine, J. L.; Kwon, R.; Curran, W. J.; Wu, Y.-L.; Paz-Ares, L. Lung Cancer: Current Therapies and New Targeted Treatments. Lancet 2017, 389 (10066), 299– 311, DOI: 10.1016/S0140-6736(16)30958-811https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVamsbzP&md5=36ace1a455a803dc6e7af83dbffb2e34Lung cancer: current therapies and new targeted treatmentsHirsch, Fred R.; Scagliotti, Giorgio V.; Mulshine, James L.; Kwon, Regina; Curran, Walter J., Jr.; Wu, Yi-Long; Paz-Ares, LuisLancet (2017), 389 (10066), 299-311CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)Lung cancer is the most frequent cause of cancer-related deaths worldwide. Every year, 1·8 million people are diagnosed with lung cancer, and 1·6 million people die as a result of the disease. 5-yr survival rates vary from 4-17% depending on stage and regional differences. In this Seminar, we discuss existing treatment for patients with lung cancer and the promise of precision medicine, with special emphasis on new targeted therapies. Some subgroups, eg-patients with poor performance status and elderly patients-are not specifically addressed, because these groups require special treatment considerations and no frameworks have been established in terms of new targeted therapies. We discuss prevention and early detection of lung cancer with an emphasis on lung cancer screening. Although we acknowledge the importance of smoking prevention and cessation, this is a large topic beyond the scope of this Seminar.
- 12Bai, R.; Li, L.; Chen, X.; Chen, N.; Song, W.; Cui, J. Neoadjuvant and Adjuvant Immunotherapy: Opening New Horizons for Patients With Early-Stage Non-Small Cell Lung Cancer. Frontiers Oncol 2020, 10, 575472 DOI: 10.3389/fonc.2020.575472There is no corresponding record for this reference.
- 13Mills, C. D.; Ley, K. M1 and M2Macrophages: The Chicken and the Egg of Immunity. J. Innate Immun 2014, 6 (6), 716– 726, DOI: 10.1159/00036494513https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhslyrsbrK&md5=face8a56588de3398ff368a2697538d9M1 and M2 Macrophages: The Chicken and the Egg of ImmunityMills, Charles D.; Ley, KlausJournal of Innate Immunity (2014), 6 (6), 716-726CODEN: JIIOB2; ISSN:1662-811X. (S. Karger AG)The purpose of this perspective is to describe a crit. advance in understanding how immune responses work. Macrophages are required for all animal life: 'Inhibit' type macrophages in all animals (called M1) can rapidly kill pathogens, and are thus the primary host defense, and 'Heal' type macrophages (M2) routinely repair and maintain tissue integrity. Macrophages perform these activities in all animals without T cells, and also in T cell-deficient vertebrates. Although adaptive immunity can amplify macrophage polarization, the long-held notion that macrophages need to be 'activated' or 'alternatively activated' by T cells is incorrect; indeed, immunol. has had it backward. M1/M2-type macrophages necessarily direct T cells toward Th1- or Th2-like activities, resp. That such macrophage-innate activities are the central directing element in immune responses is a dramatic change in understanding how immune systems operate. Most important, this revelation is opening up whole new approaches to immunotherapy. For example, many modern diseases, such as cancer and atherosclerosis, may not display 'foreign' antigens. However, there are clear imbalances in M1/M2-type responses. Correcting such innate imbalances can result in better health. Macrophages are the chicken and the egg of immunity.
- 14Donovan, A.; Lima, C. A.; Pinkus, J. L.; Pinkus, G. S.; Zon, L. I.; Robine, S.; Andrews, N. C. The Iron Exporter Ferroportin/Slc40a1 Is Essential for Iron Homeostasis. Cell Metab 2005, 1 (3), 191– 200, DOI: 10.1016/j.cmet.2005.01.003There is no corresponding record for this reference.
- 15Vinchi, F.; da Silva, M. C.; Ingoglia, G.; Petrillo, S.; Brinkman, N.; Zuercher, A.; Cerwenka, A.; Tolosano, E.; Muckenthaler, M. U. Hemopexin Therapy Reverts Heme-Induced Proinflammatory Phenotypic Switching of Macrophages in a Mouse Model of Sickle Cell Disease. Blood 2016, 127 (4), 473– 486, DOI: 10.1182/blood-2015-08-66324515https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xht1Cis7zM&md5=49e09b37adb6ee5ad1e84b32cbe6c294Hemopexin therapy reverts heme-induced proinflammatory phenotypic switching of macrophages in a mouse model of sickle cell diseaseVinchi, Francesca; Costa da Silva, Milene; Ingoglia, Giada; Petrillo, Sara; Brinkman, Nathan; Zuercher, Adrian; Cerwenka, Adelheid; Tolosano, Emanuela; Muckenthaler, Martina U.Blood (2016), 127 (4), 473-486CODEN: BLOOAW; ISSN:1528-0020. (American Society of Hematology)Hemolytic diseases, such as sickle cell anemia and thalassemia, are characterized by enhanced release of Hb and heme into the circulation, heme-iron loading of reticulo-endothelial system macrophages, and chronic inflammation. Here we show that in addn. to activating the vascular endothelium, Hb and heme excess alters the macrophage phenotype in sickle cell disease. We demonstrate that exposure of cultured macrophages to hemolytic aged red blood cells, heme, or iron causes their functional phenotypic change toward a proinflammatory state. In addn., hemolysis and macrophage heme/iron accumulation in a mouse model of sickle disease trigger similar proinflammatory phenotypic alterations in hepatic macrophages. On the mechanistic level, this critically depends on reactive oxygen species prodn. and activation of the Toll-like receptor 4 signaling pathway. We further demonstrate that the heme scavenger hemopexin protects reticulo-endothelial macrophages from heme overload in heme-loaded Hx-null mice and reduces prodn. of cytokines and reactive oxygen species. Importantly, in sickle mice, the administration of human exogenous hemopexin attenuates the inflammatory phenotype of macrophages. Taken together, our data suggest that therapeutic administration of hemopexin is beneficial to counteract heme-driven macrophage-mediated inflammation and its pathophysiol. consequences in sickle cell disease.
- 16Recalcati, S.; Locati, M.; Marini, A.; Santambrogio, P.; Zaninotto, F.; De Pizzol, M.; Zammataro, L.; Girelli, D.; Cairo, G. Differential Regulation of Iron Homeostasis during Human Macrophage Polarized Activation. Eur. J. Immunol. 2010, 40 (3), 824– 835, DOI: 10.1002/eji.20093988916https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjtVWktLg%253D&md5=762786d1111efa80786c318ae77b1318Differential regulation of iron homeostasis during human macrophage polarized activationRecalcati, Stefania; Locati, Massimo; Marini, Agnese; Santambrogio, Paolo; Zaninotto, Federica; De Pizzol, Maria; Zammataro, Luca; Girelli, Domenico; Cairo, GaetanoEuropean Journal of Immunology (2010), 40 (3), 824-835CODEN: EJIMAF; ISSN:0014-2980. (Wiley-VCH Verlag GmbH & Co. KGaA)Iron metab. in inflammation has been mostly characterized in macrophages exposed to pathogens or inflammatory conditions, mimicked by the combined action of LPS and IFN-γ (M1 polarization). However, macrophages can undergo an alternative type of activation stimulated by Th2 cytokines, and acquire a role in cell growth and tissue repair control (M2 polarization). We characterized the expression of genes related to iron homeostasis in fully differentiated unpolarized (M0), M1 and M2 human macrophages. The mol. signature of the M1 macrophages showed changes in gene expression (ferroportin repression and H ferritin induction) that favor iron sequestration in the reticuloendothelial system, a hallmark of inflammatory disorders, whereas the M2 macrophages had an expression profile (ferroportin upregulation and the downregulation of H ferritin and heme oxygenase) that enhanced iron release. The conditioned media from M2 macrophages promoted cell proliferation more efficiently than those of M1 cells and the effect was blunted by iron chelation. The role of ferroportin-mediated iron release was demonstrated by the absence of differences from the media of macrophages of a patient with loss of function ferroportin mutation. The distinct regulation of iron homeostasis in M2 macrophages provides insights into their role under pathophysiol. conditions.
- 17Recalcati, S.; Gammella, E.; Cairo, G. Ironing out Macrophage Immunometabolism. Pharm. 2019, 12 (2), 94, DOI: 10.3390/ph12020094There is no corresponding record for this reference.
- 18Pawate, S.; Shen, Q.; Fan, F.; Bhat, N. R. Redox Regulation of Glial Inflammatory Response to Lipopolysaccharide and Interferonγ. J. Neurosci. Res. 2004, 77 (4), 540– 551, DOI: 10.1002/jnr.20180There is no corresponding record for this reference.
- 19Mertens, C.; Marques, O.; Horvat, N. K.; Simonetti, M.; Muckenthaler, M. U.; Jung, M. The Macrophage Iron Signature in Health and Disease. Int. J. Mol. Sci. 2021, 22 (16), 8457, DOI: 10.3390/ijms22168457There is no corresponding record for this reference.
- 20Fu, H.; Zhang, Z.; Li, D.; Lv, Q.; Chen, S.; Zhang, Z.; Wu, M. LncRNA PELATON, a Ferroptosis Suppressor and Prognositic Signature for GBM. Frontiers Oncol 2022, 12, 817737 DOI: 10.3389/fonc.2022.817737There is no corresponding record for this reference.
- 21Yano, T.; Obata, Y.; Ishikawa, G.; Ichikawa, T. Enhancing Effect of High Dietary Iron on Lung Tumorigenesis in Mice. Cancer Lett. 1994, 76 (1), 57– 62, DOI: 10.1016/0304-3835(94)90134-1There is no corresponding record for this reference.
- 22Zhao, H.; Tanaka, T.; Mitlitski, V.; Heeter, J.; Balazs, E. A.; Darzynkiewicz, Z. Protective Effect of Hyaluronate on Oxidative DNA Damage in WI-38 and A549 Cells. Int. J. Oncol. 2008, 32 (6), 1159– 1167, DOI: 10.3892/ijo_32_6_1159There is no corresponding record for this reference.
- 23Shen, Y.; Li, X.; Zhao, B.; Xue, Y.; Wang, S.; Chen, X.; Yang, J.; Lv, H.; Shang, P. Iron Metabolism Gene Expression and Prognostic Features of Hepatocellular Carcinoma. J. Cell Biochem 2018, 119 (11), 9178– 9204, DOI: 10.1002/jcb.27184There is no corresponding record for this reference.
- 24Torti, S. V.; Torti, F. M. Iron and Cancer: More Ore to Be Mined. Nat. Rev. Cancer 2013, 13 (5), 342– 355, DOI: 10.1038/nrc349524https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXlvF2isL8%253D&md5=9d950f3dab0010974d1ef25f8c9c1733Iron and cancer: more ore to be minedTorti, Suzy V.; Torti, Frank M.Nature Reviews Cancer (2013), 13 (5), 342-355CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Iron is an essential nutrient that facilitates cell proliferation and growth. However, iron also has the capacity to engage in redox cycling and free radical formation. Therefore, iron can contribute to both tumor initiation and tumor growth; recent work has also shown that iron has a role in the tumor microenvironment and in metastasis. Pathways of iron acquisition, efflux, storage and regulation are all perturbed in cancer, suggesting that reprogramming of iron metab. is a central aspect of tumor cell survival. Signalling through hypoxia-inducible factor (HIF) and WNT pathways may contribute to altered iron metab. in cancer. Targeting iron metabolic pathways may provide new tools for cancer prognosis and therapy.
- 25Land, W. G. Transfusion-Related Acute Lung Injury: The Work of DAMPs*. Transfus Med. Hemoth 2013, 40 (1), 3– 13, DOI: 10.1159/000345688There is no corresponding record for this reference.
- 26Nairz, M.; Theurl, I.; Swirski, F. K.; Weiss, G. Pumping Iron”─How Macrophages Handle Iron at the Systemic, Microenvironmental, and Cellular Levels. Pflugers Arch. 2017, 469, 397– 418, DOI: 10.1007/s00424-017-1944-8There is no corresponding record for this reference.
- 27da Silva, M. C.; Breckwoldt, M. O.; Vinchi, F.; Correia, M. P.; Stojanovic, A.; Thielmann, C. M.; Meister, M.; Muley, T.; Warth, A.; Platten, M.; Hentze, M. W.; Cerwenka, A.; Muckenthaler, M. U. Iron Induces Anti-Tumor Activity in Tumor-Associated Macrophages. Front Immunol 2017, 8, 1479, DOI: 10.3389/fimmu.2017.01479There is no corresponding record for this reference.
- 28Thielmann, C. M.; da Silva, M. C.; Muley, T.; Meister, M.; Herpel, E.; Muckenthaler, M. U. Iron Accumulation in Tumor-Associated Macrophages Marks an Improved Overall Survival in Patients with Lung Adenocarcinoma. Sci. Rep-uk 2019, 9 (1), 11326 DOI: 10.1038/s41598-019-47833-xThere is no corresponding record for this reference.
- 29Bauer, T. A.; Horvat, N. K.; Marques, O.; Chocarro, S.; Mertens, C.; Colucci, S.; Schmitt, S.; Carrella, L. M.; Morsbach, S.; Koynov, K.; Fenaroli, F.; Blümler, P.; Jung, M.; Sotillo, R.; Hentze, M. W.; Muckenthaler, M. U.; Barz, M. Core Cross-Linked Polymeric Micelles for Specific Iron Delivery: Inducing Sterile Inflammation in Macrophages. Adv. Healthc Mater. 2021, 10, 2100385, DOI: 10.1002/adhm.202100385There is no corresponding record for this reference.
- 30Schäfer, O.; Huesmann, D.; Barz, M. Poly(S-ethylsulfonyl-l-cysteines) for Chemoselective Disulfide Formation. Macromolecules 2016, 49 (21), 8146– 8153, DOI: 10.1021/acs.macromol.6b02064There is no corresponding record for this reference.
- 31Klinker, K.; Schäfer, O.; Huesmann, D.; Bauer, T.; Capelôa, L.; Braun, L.; Stergiou, N.; Schinnerer, M.; Dirisala, A.; Miyata, K.; Osada, K.; Cabral, H.; Kataoka, K.; Barz, M. Secondary-Structure-Driven Self-Assembly of Reactive Polypept(o)Ides: Controlling Size, Shape, and Function of Core Cross-Linked Nanostructures. Angew. Chem., Int. Ed. 2017, 56 (32), 9608– 9613, DOI: 10.1002/anie.20170262431https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFensrzE&md5=3476193910ff8e0647b88ce30bb96d74Secondary-Structure-Driven Self-Assembly of Reactive Polypept(o)ides: Controlling Size, Shape, and Function of Core Cross-Linked NanostructuresKlinker, Kristina; Schaefer, Olga; Huesmann, David; Bauer, Tobias; Capeloa, Leon; Braun, Lydia; Stergiou, Natascha; Schinnerer, Meike; Dirisala, Anjaneyulu; Miyata, Kanjiro; Osada, Kensuke; Cabral, Horacio; Kataoka, Kazunori; Barz, MatthiasAngewandte Chemie, International Edition (2017), 56 (32), 9608-9613CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Achieving precise control over the morphol. and function of polymeric nanostructures during self-assembly remains a challenge in materials as well as biomedical science, esp. when independent control over particle properties is desired. Herein, we report on nanostructures derived from amphiphilic block copolypept(o)ides by secondary-structure-directed self-assembly, presenting a strategy to adjust core polarity and function sep. from particle prepn. in a bioreversible manner. The peptide-inherent process of secondary-structure formation allows for the synthesis of spherical and worm-like core-crosslinked architectures from the same block copolymer, introducing a simple yet powerful approach to versatile peptide-based core-shell nanostructures.
- 32Bauer, T. A.; Schramm, J.; Fenaroli, F.; Siemer, S.; Seidl, C. I.; Rosenauer, C.; Bleul, R.; Stauber, R. H.; Koynov, K.; Maskos, M.; Barz, M. Complex Structures Made Simple – Continuous Flow Production of Core Cross-Linked Polymeric Micelles for Paclitaxel Pro-Drug-Delivery. Adv. Mater. 2023, 35 (21), e2210704 DOI: 10.1002/adma.202210704There is no corresponding record for this reference.
- 33Bauer, T. A.; Imschweiler, J.; Muhl, C.; Weber, B.; Barz, M. Secondary Structure-Driven Self-Assembly of Thiol-Reactive Polypept(o)Ides. Biomacromolecules 2021, 22 (5), 2171– 2180, DOI: 10.1021/acs.biomac.1c0025333https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXotFygsr0%253D&md5=57b9257d9a451bf5555d7ba3c8befca1Secondary Structure-Driven Self-Assembly of Thiol-Reactive Polypept(o)idesBauer, Tobias A.; Imschweiler, Jan; Muhl, Christian; Weber, Benjamin; Barz, MatthiasBiomacromolecules (2021), 22 (5), 2171-2180CODEN: BOMAF6; ISSN:1525-7797. (American Chemical Society)Secondary structure formation differentiates polypeptides from most of the other synthetic polymers, and the transitions from random coils to rod-like α-helixes or β-sheets represent an addnl. parameter to direct self-assembly and the morphol. of nanostructures. We investigated the influence of distinct secondary structures on the self-assembly of reactive amphiphilic polypept(o)ides. The individual morphologies can be preserved by core crosslinking via chemoselective disulfide bond formation. A series of thiol-responsive copolymers of racemic polysarcosine-block-poly(S-ethylsulfonyl-DL-cysteine) (pSar-b-p(DL)Cys), enantiopure polysarcosine-block-poly(S-ethylsulfonyl-L-cysteine) (pSar-b-p(L)Cys), and polysarcosine-block-poly(S-ethylsulfonyl-L-homocysteine) (pSar-b-p(L)Hcy) was prepd. by N-carboxyanhydride polymn. The secondary structure of the peptide segment varies from α-helixes (pSar-b-p(L)Hcy) to antiparallel β-sheets (pSar-b-p(L)Cys) and disrupted β-sheets (pSar-b-p(DL)Cys). When subjected to nanopptn., copolymers with antiparallel β-sheets display the strongest tendency to self-assemble, whereas disrupted β-sheets hardly induce aggregation. This translates to worm-like micelles, solely spherical micelles, or ellipsoidal structures, as analyzed by at. force microscopy and cryogenic transmission electron microscopy, which underlines the potential of secondary structure-driven self-assembly of synthetic polypeptides.
- 34Schäfer, O.; Klinker, K.; Braun, L.; Huesmann, D.; Schultze, J.; Koynov, K.; Barz, M. Combining Orthogonal Reactive Groups in Block Copolymers for Functional Nanoparticle Synthesis in a Single Step. ACS Macro Lett. 2017, 6 (10), 1140– 1145, DOI: 10.1021/acsmacrolett.7b00678There is no corresponding record for this reference.
- 35Schäfer, O.; Huesmann, D.; Muhl, C.; Barz, M. Rethinking Cysteine Protective Groups: S-Alkylsulfonyl-l-cysteines for Chemoselective Disulfide Formation. Chem.─Eur. J. 2016, 22 (50), 18085– 18091, DOI: 10.1002/chem.201604391There is no corresponding record for this reference.
- 36Schlüter, C.; Duchrow, M.; Wohlenberg, C.; Becker, M. H.; Key, G.; Flad, H. D.; Gerdes, J. The Cell Proliferation-Associated Antigen of Antibody Ki-67: A Very Large, Ubiquitous Nuclear Protein with Numerous Repeated Elements, Representing a New Kind of Cell Cycle-Maintaining Proteins. J. Cell Biol. 1993, 123 (3), 513– 522, DOI: 10.1083/jcb.123.3.51336https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXltV2rtQ%253D%253D&md5=8eaf81089de4298736ef038380d67ea7The cell proliferation-associated antigen of antibody Ki-67: A very large, ubiquitous nuclear protein with numerous repeated elements, representing a new kind of cell cycle-maintaining proteinsSchlueter, Carsten; Duchrow, Michael; Wohlenberg, Claudia; Becker, Michael H. G.; Key, Goeran; Flad, Hans D.; Gerdes, JohannesJournal of Cell Biology (1993), 123 (3), 513-22CODEN: JCLBA3; ISSN:0021-9525.The antigen defined by monoclonal antibody (mAb) Ki-67 is a human nuclear protein, the expression of which is strictly assocd. with cell proliferation and which is widely used in routine pathol. as a proliferation marker to measure the growth fraction of cells in human tumors. Ki-67 detects a double band with apparent mol. wts. of 395 and 345 kDa in immunoblots of proteins from proliferating cells. The authors cloned and sequenced the full length cDNA, identified 2 differentially spliced isoforms of mRNA with open reading frames of 9768 and 8688 bp encoding for this cell proliferation-assocd. protein with calcd. mol. wts. of 358,761 Da and 319,508 Da, resp. New mAbs against a bacterially expressed part and a synthetic polypeptide deduced from the isolated cDNA react with the native Ki-67 antigen, thus providing a circle of evidence that the authors have cloned the authentic Ki-67 antigen cDNA. The central part of the Ki-67 antigen cDNA contains a large 6845-bp exon with 16 tandemly repeated 366-bp elements, the Ki-67 repeats, each including a highly conserved new motif of 66 bp, the Ki-67 motif, which encodes for the epitope detected by Ki-67. Computer anal. of the nucleic acid and the deduced amino acid sequence of the Ki-67 antigen confirmed that the cDNA encodes for a nuclear and short-lived protein without any significant homol. to known sequences. Ki-67 antigen-specific antisense oligonucleotides inhibit the proliferation of IM-9 cells, indicating that the Ki-67 antigen may be an abs. requirement for maintaining cell proliferation. Evidently, the Ki-67 antigen defines a new category of cell cycle-assocd. nuclear nonhistone proteins.
- 37Rogakou, E. P.; Pilch, D. R.; Orr, A. H.; Ivanova, V. S.; Bonner, W. M. DNA Double-Stranded Breaks Induce Histone H2AX Phosphorylation on Serine 139*. J. Biol. Chem. 1998, 273 (10), 5858– 5868, DOI: 10.1074/jbc.273.10.5858There is no corresponding record for this reference.
- 38Butler, L. M.; Zhou, X.; Xu, W.-S.; Scher, H. I.; Rifkind, R. A.; Marks, P. A.; Richon, V. M. The Histone Deacetylase Inhibitor SAHA Arrests Cancer Cell Growth, up-Regulates Thioredoxin-Binding Protein-2, and down-Regulates Thioredoxin. Proc. National Acad. Sci. 2002, 99 (18), 11700– 11705, DOI: 10.1073/pnas.182372299There is no corresponding record for this reference.
- 39Cai, B.; Kasikara, C.; Doran, A. C.; Ramakrishnan, R.; Birge, R. B.; Tabas, I. MerTK Signaling in Macrophages Promotes the Synthesis of Inflammation Resolution Mediators by Suppressing CaMKII Activity. Sci. Signal 2018, 11 (549), na, DOI: 10.1126/scisignal.aar3721There is no corresponding record for this reference.
- 40MULERO, V.; WEI, X.; LIEW, F. Y.; BROCK, J. H. Regulation of Phagosomal Iron Release from Murine Macrophages by Nitric Oxide. Biochem. J. 2002, 365 (1), 127– 132, DOI: 10.1042/bj20011875There is no corresponding record for this reference.
- 41Aldrovandi, M.; Conrad, M. Ferroptosis: The Good, the Bad and the Ugly. Cell Res. 2020, 30 (12), 1061– 1062, DOI: 10.1038/s41422-020-00434-0There is no corresponding record for this reference.
- 42Lemaire, G.; Alvarez-Pachon, F.-J.; Beuneu, C.; Lepoivre, M.; Petit, J.-F. Differential Cytostatic Effects of NO Donors and NO Producing Cells. Free Radical Bio Med. 1999, 26 (9–10), 1274– 1283, DOI: 10.1016/S0891-5849(98)00331-1There is no corresponding record for this reference.
- 43Van den Bossche, J.; Baardman, J.; Otto, N. A.; van der Velden, S.; Neele, A. E.; van den Berg, S. M.; Luque-Martin, R.; Chen, H.-J.; Boshuizen, M.; Ahmed, M.; Hoeksema, M. A.; de Vos, A. F.; de Winther, M. Mitochondrial Dysfunction Prevents Repolarization of Inflammatory Macrophages. Cell Reports 2016, 17 (3), 684– 696, DOI: 10.1016/j.celrep.2016.09.008There is no corresponding record for this reference.
- 44Maddalo, D.; Manchado, E.; Concepcion, C. P.; Bonetti, C.; Vidigal, J. A.; Han, Y.-C.; Ogrodowski, P.; Crippa, A.; Rekhtman, N.; de Stanchina, E.; Lowe, S. W.; Ventura, A. In Vivo Engineering of Oncogenic Chromosomal Rearrangements with the CRISPR/Cas9 System. Nature 2014, 516, 423– 427, DOI: 10.1038/nature1390244https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVemtrvL&md5=ded8f269a1342b56ecf0ec48f61af33aIn vivo engineering of oncogenic chromosomal rearrangements with the CRISPR/Cas9 systemMaddalo, Danilo; Manchado, Eusebio; Concepcion, Carla P.; Bonetti, Ciro; Vidigal, Joana A.; Han, Yoon-Chi; Ogrodowski, Paul; Crippa, Alessandra; Rekhtman, Natasha; de Stanchina, Elisa; Lowe, Scott W.; Ventura, AndreaNature (London, United Kingdom) (2014), 516 (7531), 423-427CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Chromosomal rearrangements have a central role in the pathogenesis of human cancers and often result in the expression of therapeutically actionable gene fusions. A recently discovered example is a fusion between the genes echinoderm microtubule-assocd. protein like 4 (EML4) and anaplastic lymphoma kinase (ALK), generated by an inversion on the short arm of chromosome 2: inv(2)(p21p23). The EML4-ALK oncogene is detected in a subset of human non-small cell lung cancers (NSCLC) and is clin. relevant because it confers sensitivity to ALK inhibitors. Despite their importance, modeling such genetic events in mice has proven challenging and requires complex manipulation of the germ line. Here we describe an efficient method to induce specific chromosomal rearrangements in vivo using viral-mediated delivery of the CRISPR/Cas9 system to somatic cells of adult animals. We apply it to generate a mouse model of Eml4-Alk-driven lung cancer. The resulting tumors invariably harbor the Eml4-Alk inversion, express the Eml4-Alk fusion gene, display histopathol. and mol. features typical of ALK+ human NSCLCs, and respond to treatment with ALK inhibitors. The general strategy described here substantially expands our ability to model human cancers in mice and potentially in other organisms.
- 45da Silva, M. C.; Breckwoldt, M. O.; Vinchi, F.; Correia, M. P.; Stojanovic, A.; Thielmann, C. M.; Meister, M.; Muley, T.; Warth, A.; Platten, M.; Hentze, M. W.; Cerwenka, A.; Muckenthaler, M. U. Iron Induces Anti-Tumor Activity in Tumor-Associated Macrophages. Front Immunol 2017, 8, 1479, DOI: 10.3389/fimmu.2017.01479There is no corresponding record for this reference.
- 46Scagliotti, G.; Stahel, R. A.; Rosell, R.; Thatcher, N.; Soria, J.-C. ALK Translocation and Crizotinib in Non-Small Cell Lung Cancer: An Evolving Paradigm in Oncology Drug Development. Eur. J. Cancer 2012, 48 (7), 961– 973, DOI: 10.1016/j.ejca.2012.02.001There is no corresponding record for this reference.
- 47Shrestha, N.; Nimick, M.; Dass, P.; Rosengren, R. J.; Ashton, J. C. Mechanisms of Suppression of Cell Growth by Dual Inhibition of ALK and MEK in ALK-Positive Non-Small Cell Lung Cancer. Sci. Rep-uk 2019, 9 (1), 18842 DOI: 10.1038/s41598-019-55376-4There is no corresponding record for this reference.
- 48Guo, Q.; Liu, L.; Chen, Z.; Fan, Y.; Zhou, Y.; Yuan, Z.; Zhang, W. Current Treatments for Non-Small Cell Lung Cancer. Frontiers Oncol 2022, 12, 945102 DOI: 10.3389/fonc.2022.945102There is no corresponding record for this reference.
- 49Zanganeh, S.; Hutter, G.; Spitler, R.; Lenkov, O.; Mahmoudi, M.; Shaw, A.; Pajarinen, J. S.; Nejadnik, H.; Goodman, S.; Moseley, M.; Coussens, L. M.; Daldrup-Link, H. E. Iron Oxide Nanoparticles Inhibit Tumour Growth by Inducing Pro-Inflammatory Macrophage Polarization in Tumour Tissues. Nat. Nanotechnol 2016, 11 (11), 986– 994, DOI: 10.1038/nnano.2016.16849https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFyms7fO&md5=ddc5109f0cce0aa4b79adf816808fb4bIron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissuesZanganeh, Saeid; Hutter, Gregor; Spitler, Ryan; Lenkov, Olga; Mahmoudi, Morteza; Shaw, Aubie; Pajarinen, Jukka Sakari; Nejadnik, Hossein; Goodman, Stuart; Moseley, Michael; Coussens, Lisa Marie; Daldrup-Link, Heike ElisabethNature Nanotechnology (2016), 11 (11), 986-994CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)Until now, the Food and Drug Administration (FDA)-approved iron supplement ferumoxytol and other iron oxide nanoparticles have been used for treating iron deficiency, as contrast agents for magnetic resonance imaging and as drug carriers. Here, the authors show an intrinsic therapeutic effect of ferumoxytol on the growth of early mammary cancers, and lung cancer metastases in liver and lungs. In vitro, adenocarcinoma cells coincubated with ferumoxytol and macrophages showed increased caspase-3 activity. Macrophages exposed to ferumoxytol displayed increased mRNA assocd. with pro-inflammatory Th1-type responses. In vivo, ferumoxytol significantly inhibited growth of s.c. adenocarcinomas in mice. In addn., i.v. ferumoxytol treatment before i.v. tumor cell challenge prevented development of liver metastasis. Fluorescence-activated cell sorting (FACS) and histopathol. studies showed that the obsd. tumor growth inhibition was accompanied by increased presence of pro-inflammatory M1 macrophages in the tumor tissues. The results suggest that ferumoxytol could be applied 'off label' to protect the liver from metastatic seeds and potentiate macrophage-modulating cancer immunotherapies.
- 50Cameron, D. J.; Churchill, W. H. Cytotoxicity of Human Macrophages for Tumor Cells. Enhancement by Human Lymphocyte Mediators. J. Clin Invest 1979, 63 (5), 977– 984, DOI: 10.1172/JCI109398There is no corresponding record for this reference.
- 51Hudson, S. V.; Miller, H. A.; Mahlbacher, G. E.; Saforo, D.; Beverly, L. J.; Arteel, G. E.; Frieboes, H. B. Computational/Experimental Evaluation of Liver Metastasis Post Hepatic Injury: Interactions with Macrophages and Transitional ECM. Sci. Rep-uk 2019, 9 (1), 15077 DOI: 10.1038/s41598-019-51249-yThere is no corresponding record for this reference.
- 52Bartha, L.; Eftimie, R. Mathematical Investigation into the Role of Macrophage Heterogeneity on the Temporal and Spatio-Temporal Dynamics of Non-Small Cell Lung Cancers. J. Theor. Biol. 2022, 549, 111207 DOI: 10.1016/j.jtbi.2022.111207There is no corresponding record for this reference.
- 53Li, X.; Jolly, M. K.; George, J. T.; Pienta, K. J.; Levine, H. Computational Modeling of the Crosstalk Between Macrophage Polarization and Tumor Cell Plasticity in the Tumor Microenvironment. Frontiers Oncol 2019, 9, 10, DOI: 10.3389/fonc.2019.00010There is no corresponding record for this reference.
- 54Hegedűs, C.; Kovács, K.; Polgár, Z.; Regdon, Z.; Szabó, É.; Robaszkiewicz, A.; Forman, H. J.; Martner, A.; Virág, L. Redox Control of Cancer Cell Destruction. Redox Biol. 2018, 16, 59– 74, DOI: 10.1016/j.redox.2018.01.01554https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjtFOit7w%253D&md5=a2829fe76b47302092d8e0feb1e6de67Redox control of cancer cell destructionHegedus, Csaba; Kovacs, Katalin; Polgar, Zsuzsanna; Regdon, Zsolt; Szabo, Eva; Robaszkiewicz, Agnieszka; Forman, Henry Jay; Martner, Anna; Virag, LaszloRedox Biology (2018), 16 (), 59-74CODEN: RBEIB3; ISSN:2213-2317. (Elsevier B.V.)A review. Redox regulation has been proposed to control various aspects of carcinogenesis, cancer cell growth, metab., migration, invasion, metastasis and cancer vascularization. As cancer has many faces, the role of redox control in different cancers and in the numerous cancer-related processes often point in different directions. In this review, we focus on the redox control mechanisms of tumor cell destruction. The review covers the tumor-intrinsic role of oxidants derived from the redn. of oxygen and nitrogen in the control of tumor cell proliferation as well as the roles of oxidants and antioxidant systems in cancer cell death caused by traditional anticancer weapons (chemotherapeutic agents, radiotherapy, photodynamic therapy). Emphasis is also put on the role of oxidants and redox status in the outcome following interactions between cancer cells, cytotoxic lymphocytes and tumor infiltrating macrophages.
- 55Kim, S. J.; Kim, H. S.; Seo, Y. R. Understanding of ROS-Inducing Strategy in Anticancer Therapy. Oxid Med. Cell Longev 2019, 2019, 5381692 DOI: 10.1155/2019/538169255https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MbptlektA%253D%253D&md5=365ea194a3a48a0107703bce40d04de4Understanding of ROS-Inducing Strategy in Anticancer TherapyKim Su Ji; Kim Hyun Soo; Seo Young Rok; Kim Su Ji; Kim Hyun Soo; Seo Young RokOxidative medicine and cellular longevity (2019), 2019 (), 5381692 ISSN:.Redox homeostasis is essential for the maintenance of diverse cellular processes. Cancer cells have higher levels of reactive oxygen species (ROS) than normal cells as a result of hypermetabolism, but the redox balance is maintained in cancer cells due to their marked antioxidant capacity. Recently, anticancer therapies that induce oxidative stress by increasing ROS and/or inhibiting antioxidant processes have received significant attention. The acceleration of accumulative ROS disrupts redox homeostasis and causes severe damage in cancer cells. In this review, we describe ROS-inducing cancer therapy and the anticancer mechanism employed by prooxidative agents. To understand the comprehensive biological response to certain prooxidative anticancer drugs such as 2-methoxyestradiol, buthionine sulfoximine, cisplatin, doxorubicin, imexon, and motexafin gadolinium, we propose and visualize the drug-gene, drug-cell process, and drug-disease interactions involved in oxidative stress induction and antioxidant process inhibition as well as specific side effects of these drugs using pathway analysis with a big data-based text-mining approach. Our review will be helpful to improve the therapeutic effects of anticancer drugs by providing information about biological changes that occur in response to prooxidants. For future directions, there is still a need for pharmacogenomic studies on prooxidative agents as well as the molecular mechanisms underlying the effects of the prooxidants and/or antioxidant-inhibitor agents for effective anticancer therapy through selective killing of cancer cells.
- 56Wang, K.; Jiang, J.; Lei, Y.; Zhou, S.; Wei, Y.; Huang, C. Targeting Metabolic–Redox Circuits for Cancer Therapy. Trends Biochem. Sci. 2019, 44 (5), 401– 414, DOI: 10.1016/j.tibs.2019.01.00156https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXpsVOgtA%253D%253D&md5=5b51ed067632d8be3067545e53792010Targeting Metabolic-Redox Circuits for Cancer TherapyWang, Kui; Jiang, Jingwen; Lei, Yunlong; Zhou, Shengtao; Wei, Yuquan; Huang, CanhuaTrends in Biochemical Sciences (2019), 44 (5), 401-414CODEN: TBSCDB; ISSN:0968-0004. (Elsevier Ltd.)A review. Metabolic alterations and elevated levels of reactive oxygen species (ROS) are two characteristics of cancer. The metabolic patterns of cancer cells are elaborately reprogrammed to fulfill the high biomass demands of rapid propagation. ROS, the byproducts of metabolic processes, are accumulated in cancer cells partially due to metabolic abnormalities or oncogenic mutations. To prevent oxidative damage, cancer cells can orchestrate metabolic adaptation to maintain redn.-oxidn. (redox) balance by producing reducing equiv. ROS, acting as second messengers, can in turn manipulate metabolic pathways by directly or indirectly affecting the function of metabolic enzymes. In this review we discuss how cancer cell metab. and redox signaling are intertwined, with an emphasis on the perspective of targeting metabolic-redox circuits for cancer therapy.
- 57Teppo, H.-R.; Soini, Y.; Karihtala, P. Reactive Oxygen Species-Mediated Mechanisms of Action of Targeted Cancer Therapy. Oxid Med. Cell Longev 2017, 2017, 1485283 DOI: 10.1155/2017/1485283There is no corresponding record for this reference.
- 58Samarin, J.; Fabrowski, P.; Kurilov, R.; Nuskova, H.; Hummel-Eisenbeiss, J.; Pink, H.; Li, N.; Weru, V.; Alborzinia, H.; Yildiz, U.; Grob, L.; Taubert, M.; Czech, M.; Morgen, M.; Brandstädter, C.; Becker, K.; Mao, L.; Jayavelu, A. K.; Goncalves, A.; Uhrig, U.; Seiler, J.; Lyu, Y.; Diederichs, S.; Klingmüller, U.; Muckenthaler, M.; Kopp-Schneider, A.; Teleman, A.; Miller, A. K.; Gunkel, N. Low Level of Antioxidant Capacity Biomarkers but Not Target Overexpression Predicts Vulnerability to ROS-Inducing Drugs. Redox Biol. 2023, 62, 102639 DOI: 10.1016/j.redox.2023.102639There is no corresponding record for this reference.
- 59Wang, Y.; Zhang, X.; Yang, L.; Xue, J.; Hu, G. Blockade of CCL2 Enhances Immunotherapeutic Effect of Anti-PD1 in Lung Cancer. J. Bone Oncol 2018, 11, 27– 32, DOI: 10.1016/j.jbo.2018.01.002There is no corresponding record for this reference.
- 60Binnewies, M.; Roberts, E. W.; Kersten, K.; Chan, V.; Fearon, D. F.; Merad, M.; Coussens, L. M.; Gabrilovich, D. I.; Ostrand-Rosenberg, S.; Hedrick, C. C.; Vonderheide, R. H.; Pittet, M. J.; Jain, R. K.; Zou, W.; Howcroft, T. K.; Woodhouse, E. C.; Weinberg, R. A.; Krummel, M. F. Understanding the Tumor Immune Microenvironment (TIME) for Effective Therapy. Nat. Med. 2018, 24 (5), 541– 550, DOI: 10.1038/s41591-018-0014-x60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXosVCnsLk%253D&md5=c8a5823d49b18653b960f920c1fdbedfUnderstanding the tumor immune microenvironment (TIME) for effective therapyBinnewies, Mikhail; Roberts, Edward W.; Kersten, Kelly; Chan, Vincent; Fearon, Douglas F.; Merad, Miriam; Coussens, Lisa M.; Gabrilovich, Dmitry I.; Ostrand-Rosenberg, Suzanne; Hedrick, Catherine C.; Vonderheide, Robert H.; Pittet, Mikael J.; Jain, Rakesh K.; Zou, Weiping; Howcroft, T. Kevin; Woodhouse, Elisa C.; Weinberg, Robert A.; Krummel, Matthew F.Nature Medicine (New York, NY, United States) (2018), 24 (5), 541-550CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)A review. The clin. successes in immunotherapy have been both astounding and at the same time unsatisfactory. Countless patients with varied tumor types have seen pronounced clin. response with immunotherapeutic intervention; however, many more patients have experienced minimal or no clin. benefit when provided the same treatment. As technol. has advanced, so has the understanding of the complexity and diversity of the immune context of the tumor microenvironment and its influence on response to therapy. It has been possible to identify different subclasses of immune environment that have an influence on tumor initiation and response and therapy; by parsing the unique classes and subclasses of tumor immune microenvironment (TIME) that exist within a patient's tumor, the ability to predict and guide immunotherapeutic responsiveness will improve, and new therapeutic targets will be revealed.
- 61Zeisberger, S. M.; Odermatt, B.; Marty, C.; Zehnder-Fjällman, A. H. M.; Ballmer-Hofer, K.; Schwendener, R. A. Clodronate-Liposome-Mediated Depletion of Tumour-Associated Macrophages: A New and Highly Effective Antiangiogenic Therapy Approach. Br. J. Cancer 2006, 95 (3), 272– 281, DOI: 10.1038/sj.bjc.660324061https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xnsl2iu7o%253D&md5=10ef4032a292c1709a91b672f323a22eClodronate-liposome-mediated depletion of tumour-associated macrophages: a new and highly effective antiangiogenic therapy approachZeisberger, S. M.; Odermatt, B.; Marty, C.; Zehnder-Fjaellman, A. H. M.; Ballmer-Hofer, K.; Schwendener, R. A.British Journal of Cancer (2006), 95 (3), 272-281CODEN: BJCAAI; ISSN:0007-0920. (Nature Publishing Group)Tumor-assocd. macrophages, TAMs, play a pivotal role in tumor growth and metastasis by promoting tumor angiogenesis. Treatment with clodronate encapsulated in liposomes (clodrolip) efficiently depleted these phagocytic cells in the murine F9 teratocarcinoma and human A673 rhabdomyosarcoma mouse tumor models resulting in significant inhibition of tumor growth ranging from 75 to >92%, depending on therapy and schedule. Tumor inhibition was accompanied by a drastic redn. in blood vessel d. in the tumor tissue. Vascular endothelial growth factor (VEGF) is one of the major inducers of tumor angiogenesis and is also required for macrophage recruitment. The strongest effects were obsd. with the combination therapy of clodrolip and a VEGF-neutralizing antibody, whereas free clodronate was not significantly active. Immunohistol. evaluation of the tumors showed significant depletion of F4/80+ and MOMA-1+ and a less pronounced depletion of CD11b+ TAMs. Blood vessel staining (CD31) and quantification of the vessels as well as TAMs and tumor-assocd. dendritic cells (TADCs) in the A673 model showed redn. rates of 85 to >94%, even 9 days after the end of therapy. In addn., CD11c+ TADCs, which have been shown to potentially differentiate into endothelial-like cells upon stimulation by tumor released growth and differentiation factors, were similarly reduced by clodrolip or antibody treatment. These results validate clodrolip therapy in combination with angiogenesis inhibitors as a promising novel strategy for an indirect cancer therapy aimed at the hematopoietic precursor cells that stimulate tumor growth and dissemination and as a tool to study the role of macrophages and dendritic cells in tumorigenesis.
- 62Gubin, M. M.; Esaulova, E.; Ward, J. P.; Malkova, O. N.; Runci, D.; Wong, P.; Noguchi, T.; Arthur, C. D.; Meng, W.; Alspach, E.; Medrano, R. F. V.; Fronick, C.; Fehlings, M.; Newell, E. W.; Fulton, R. S.; Sheehan, K. C. F.; Oh, S. T.; Schreiber, R. D.; Artyomov, M. N. High-Dimensional Analysis Delineates Myeloid and Lymphoid Compartment Remodeling during Successful Immune-Checkpoint Cancer Therapy. Cell 2018, 175 (4), 1014– 1030, DOI: 10.1016/j.cell.2018.09.03062https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvFKjtbbM&md5=873a47c26202c2a47e7750d719cfb530High-Dimensional Analysis Delineates Myeloid and Lymphoid Compartment Remodeling during Successful Immune-Checkpoint Cancer TherapyGubin, Matthew M.; Esaulova, Ekaterina; Ward, Jeffrey P.; Malkova, Olga N.; Runci, Daniele; Wong, Pamela; Noguchi, Takuro; Arthur, Cora D.; Meng, Wei; Alspach, Elise; Medrano, Ruan F. V.; Fronick, Catrina; Fehlings, Michael; Newell, Evan W.; Fulton, Robert S.; Sheehan, Kathleen C. F.; Oh, Stephen T.; Schreiber, Robert D.; Artyomov, Maxim N.Cell (Cambridge, MA, United States) (2018), 175 (4), 1014-1030.e19CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Although current immune-checkpoint therapy (ICT) mainly targets lymphoid cells, it is assocd. with a broader remodeling of the tumor micro-environment. Here, using complementary forms of high-dimensional profiling, the authors define differences across all hematopoietic cells from syngeneic mouse tumors during unrestrained tumor growth or effective ICT. Unbiased assessment of gene expression of tumor-infiltrating cells by single-cell RNA sequencing (scRNAseq) and longitudinal assessment of cellular protein expression by mass cytometry (CyTOF) revealed significant remodeling of both the lymphoid and myeloid intratumoral compartments. Surprisingly, the authors obsd. multiple subpopulations of monocytes/macrophages, distinguishable by the markers CD206, CX3CR1, CD1d, and iNOS, that change over time during ICT in a manner partially dependent on IFNγ. The authors' data support the hypothesis that this macrophage polarization/activation results from effects on circulatory monocytes and early macrophages entering tumors, rather than on pre-polarized mature intratumoral macrophages.
- 63Giordano, S.; Petrelli, A. From Single- to Multi-Target Drugs in Cancer Therapy: When Aspecificity Becomes an Advantage. Curr. Med. Chem. 2008, 15 (5), 422– 432, DOI: 10.2174/09298670878350321263https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXnsVyisL0%253D&md5=d90df6a670921b6fad58caa46db01fa5From single- to multi-target drugs in cancer therapy: when a specificity becomes an advantagePetrelli, A.; Giordano, S.Current Medicinal Chemistry (2008), 15 (5), 422-432CODEN: CMCHE7; ISSN:0929-8673. (Bentham Science Publishers Ltd.)A review. Targeted therapies by means of compds. that inhibit a specific target mol. represent a new perspective in the treatment of cancer. In contrast to conventional chemotherapy which acts on all dividing cells generating toxic effects and damage of normal tissues, targeted drugs allow to hit, in a more specific manner, subpopulations of cells directly involved in tumor progression. Mols. controlling cell proliferation and death, such as Tyrosine Kinase Receptors (RTKs) for growth factors, are among the best targets for this type of therapeutic approach. Two classes of compds. targeting RTKs are currently used in clin. practice: monoclonal antibodies and tyrosine kinase inhibitors. The era of targeted therapy began with the approval of Trastuzumab, a monoclonal antibody against HER2, for treatment of metastatic breast cancer, and Imatinib, a small tyrosine kinase inhibitor targeting BCR-Abl, in Chronic Myeloid Leukemia. Despite the initial enthusiasm for the efficacy of these treatments, clinicians had to face soon the problem of relapse, as almost invariably cancer patients developed drug resistance, often due to the activation of alternative RTKs pathways. In this view, the rationale at the basis of targeting drugs is radically shifting. In the past, the main effort was aimed at developing highly specific inhibitors acting on single RTKs. Now, there is a general agreement that mols. interfering simultaneously with multiple RTKs might be more effective than single target agents. With the recent approval by FDA of Sorafenib and Sunitinib - targeting VEGFR, PDGFR, FLT-3 and c-Kit - a different scenario has been emerging, where a new generation of anti-cancer drugs, able to inhibit more than one pathway, would probably play a major role.
- 64Bedard, P. L.; Hyman, D. M.; Davids, M. S.; Siu, L. L. Small Molecules, Big Impact: 20 Years of Targeted Therapy in Oncology. Lancet 2020, 395 (10229), 1078– 1088, DOI: 10.1016/S0140-6736(20)30164-164https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlvFGrt7c%253D&md5=1538cc035cda58fae77a81025d485027Small molecules, big impact: 20 years of targeted therapy in oncologyBedard, Philippe L.; Hyman, David M.; Davids, Matthew S.; Siu, Lillian L.Lancet (2020), 395 (10229), 1078-1088CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)A review. The identification of mol. targets and the growing knowledge of their cellular functions have led to the development of small mol. inhibitors as a major therapeutic class for cancer treatment. Both multitargeted and highly selective kinase inhibitors are used for the treatment of advanced treatment-resistant cancers, and many have also achieved regulatory approval for early clin. settings as adjuvant therapies or as first-line options for recurrent or metastatic disease. Lessons learned from the development of these agents can accelerate the development of next-generation inhibitors to optimize the therapeutic index, overcome drug resistance, and establish combination therapies. The future of small mol. inhibitors is promising as there is the potential to investigate novel difficult-to-drug targets, to apply predictive non-clin. models to select promising drug candidates for human evaluation, and to use dynamic clin. trial interventions with liq. biopsies to deliver precision medicine.
- 65Greten, F. R.; Grivennikov, S. I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51 (1), 27– 41, DOI: 10.1016/j.immuni.2019.06.02565https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlyls7jP&md5=0d39409f4a26c18100163b7b49367d46Inflammation and Cancer: Triggers, Mechanisms, and ConsequencesGreten, Florian R.; Grivennikov, Sergei I.Immunity (2019), 51 (1), 27-41CODEN: IUNIEH; ISSN:1074-7613. (Elsevier Inc.)A review. Inflammation predisposes to the development of cancer and promotes all stages of tumorigenesis. Cancer cells, as well as surrounding stromal and inflammatory cells, engage in well-orchestrated reciprocal interactions to form an inflammatory tumor microenvironment (TME). Cells within the TME are highly plastic, continuously changing their phenotypic and functional characteristics. Here, we review the origins of inflammation in tumors, and the mechanisms whereby inflammation drives tumor initiation, growth, progression, and metastasis. We discuss how tumor-promoting inflammation closely resembles inflammatory processes typically found during development, immunity, maintenance of tissue homeostasis, or tissue repair and illuminate the distinctions between tissue-protective and pro-tumorigenic inflammation, including spatiotemporal considerations. Defining the cornerstone rules of engagement governing mol. and cellular mechanisms of tumor-promoting inflammation will be essential for further development of anti-cancer therapies.
- 66Ries, C. H.; Cannarile, M. A.; Hoves, S.; Benz, J.; Wartha, K.; Runza, V.; Rey-Giraud, F.; Pradel, L. P.; Feuerhake, F.; Klaman, I.; Jones, T.; Jucknischke, U.; Scheiblich, S.; Kaluza, K.; Gorr, I. H.; Walz, A.; Abiraj, K.; Cassier, P. A.; Sica, A.; Gomez-Roca, C.; de Visser, K. E.; Italiano, A.; Le Tourneau, C.; Delord, J.-P.; Levitsky, H.; Blay, J.-Y.; Rüttinger, D. Targeting Tumor-Associated Macrophages with Anti-CSF-1R Antibody Reveals a Strategy for Cancer Therapy. Cancer Cell 2014, 25 (6), 846– 859, DOI: 10.1016/j.ccr.2014.05.01666https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXpslCntrw%253D&md5=023e0585a753021f10b204c6aeb72d4bTargeting Tumor-Associated Macrophages with Anti-CSF-1R Antibody Reveals a Strategy for Cancer TherapyRies, Carola H.; Cannarile, Michael A.; Hoves, Sabine; Benz, Joerg; Wartha, Katharina; Runza, Valeria; Rey-Giraud, Flora; Pradel, Leon P.; Feuerhake, Friedrich; Klaman, Irina; Jones, Tobin; Jucknischke, Ute; Scheiblich, Stefan; Kaluza, Klaus; Gorr, Ingo H.; Walz, Antje; Abiraj, Keelara; Cassier, Philippe A.; Sica, Antonio; Gomez-Roca, Carlos; de Visser, Karin E.; Italiano, Antoine; Le Tourneau, Christophe; Delord, Jean-Pierre; Levitsky, Hyam; Blay, Jean-Yves; Ruettinger, DominikCancer Cell (2014), 25 (6), 846-859CODEN: CCAECI; ISSN:1535-6108. (Elsevier Inc.)Macrophage infiltration has been identified as an independent poor prognostic factor in several cancer types. The major survival factor for these macrophages is macrophage colony-stimulating factor 1 (CSF-1). We generated a monoclonal antibody (RG7155) that inhibits CSF-1 receptor (CSF-1R) activation. In vitro RG7155 treatment results in cell death of CSF-1-differentiated macrophages. In animal models, CSF-1R inhibition strongly reduces F4/80+ tumor-assocd. macrophages accompanied by an increase of the CD8+/CD4+ T cell ratio. Administration of RG7155 to patients led to striking redns. of CSF-1R+CD163+ macrophages in tumor tissues, which translated into clin. objective responses in diffuse-type giant cell tumor (Dt-GCT) patients.
- 67Muzumdar, M. D.; Tasic, B.; Miyamichi, K.; Li, L.; Luo, L. A Global Double-fluorescent Cre Reporter Mouse. Genesis 2007, 45 (9), 593– 605, DOI: 10.1002/dvg.2033567https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtlWjs7nK&md5=0de0e4d9a2768c46b17023656cada120A global double-fluorescent Cre reporter mouseMuzumdar, Mandar Deepak; Tasic, Bosiljka; Miyamichi, Kazunari; Li, Ling; Luo, LiqunGenesis (Hoboken, NJ, United States) (2007), 45 (9), 593-605CODEN: GNESFY; ISSN:1526-954X. (Wiley-Liss, Inc.)The Cre/loxP system has been used extensively for conditional mutagenesis in mice. Reporters of Cre activity are important for defining the spatial and temporal extent of Cre-mediated recombination. Here we describe mT/mG, a double-fluorescent Cre reporter mouse that expresses membrane-targeted tandem dimer Tomato (mT) prior to Cre-mediated excision and membrane-targeted green fluorescent protein (mG) after excision. We show that reporter expression is nearly ubiquitous, allowing visualization of fluorescent markers in live and fixed samples of all tissues examd. We further demonstrate that mG labeling is Cre-dependent, complementary to mT at single cell resoln., and distinguishable by fluorescence-activated cell sorting. Both membrane-targeted markers outline cell morphol., highlight membrane structures, and permit visualization of fine cellular processes. In addn. to serving as a global Cre reporter, the mT/mG mouse may also be used as a tool for lineage tracing, transplantation studies, and anal. of cell morphol. in vivo.
- 68Oikonomou, N.; Mouratis, M.-A.; Tzouvelekis, A.; Kaffe, E.; Valavanis, C.; Vilaras, G.; Karameris, A.; Prestwich, G. D.; Bouros, D.; Aidinis, V. Pulmonary Autotaxin Expression Contributes to the Pathogenesis of Pulmonary Fibrosis. Am. J. Resp Cell Mol. 2012, 47 (5), 566– 574, DOI: 10.1165/rcmb.2012-0004OCThere is no corresponding record for this reference.
- 69Guida, C.; Altamura, S.; Klein, F. A.; Galy, B.; Boutros, M.; Ulmer, A. J.; Hentze, M. W.; Muckenthaler, M. U. A Novel Inflammatory Pathway Mediating Rapid Hepcidin-Independent Hypoferremia. Blood 2015, 125 (14), 2265– 2275, DOI: 10.1182/blood-2014-08-595256There is no corresponding record for this reference.
- 70Hodgkin, P. D.; Lee, J. H.; Lyons, A. B. B Cell Differentiation and Isotype Switching Is Related to Division Cycle Number. J. Exp Medicine 1996, 184 (1), 277– 281, DOI: 10.1084/jem.184.1.277There is no corresponding record for this reference.
- 71Müller, T.; Kalxdorf, M.; Longuespée, R.; Kazdal, D. N.; Stenzinger, A.; Krijgsveld, J. Automated Sample Preparation with SP3 for Low-input Clinical Proteomics. Mol. Syst. Biol. 2020, 16 (1), e9111 DOI: 10.15252/msb.2019911171https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFOis7o%253D&md5=5bf4201cc5b52478697c68c60cae6ee0Automated sample preparation with SP3 for low-input clinical proteomicsMueller, Torsten; Kalxdorf, Mathias; Longuespee, Remi; Kazdal, Daniel N.; Stenzinger, Albrecht; Krijgsveld, JeroenMolecular Systems Biology (2020), 16 (1), e9111CODEN: MSBOC3; ISSN:1744-4292. (Wiley-VCH Verlag GmbH & Co. KGaA)High-throughput and streamlined workflows are essential in clin. proteomics for standardized processing of samples from a variety of sources, including fresh-frozen tissue, FFPE tissue, or blood. To reach this goal, we have implemented single-pot solid-phase-enhanced sample prepn. (SP3) on a liq. handling robot for automated processing (autoSP3) of tissue lysates in a 96-well format. AutoSP3 performs unbiased protein purifn. and digestion, and delivers peptides that can be directly analyzed by LCMS, thereby significantly reducing hands-on time, reducing variability in protein quantification, and improving longitudinal reproducibility. We demonstrate the distinguishing ability of autoSP3 to process low-input samples, reproducibly quantifying 500-1,000 proteins from 100 to 1,000 cells. Furthermore, we applied this approach to a cohort of clin. FFPE pulmonary adenocarcinoma (ADC) samples and recapitulated their sepn. into known histol. growth patterns. Finally, we integrated autoSP3 with AFA ultrasonication for the automated end-to-end sample prepn. and LCMS anal. of 96 intact tissue samples. Collectively, this constitutes a generic, scalable, and cost-effective workflow with minimal manual intervention, enabling reproducible tissue proteomics in a broad range of clin. and non-clin. applications.
- 72Heming, S.; Hansen, P.; Vlasov, A.; Schwörer, F.; Schaumann, S.; Frolovaitė, P.; Lehmann, W.-D.; Timmer, J.; Schilling, M.; Helm, B.; Klingmüller, U. MSPypeline: A Python Package for Streamlined Data Analysis of Mass Spectrometry-Based Proteomics. Bioinform Adv. 2022, 2 (1), na, DOI: 10.1093/bioadv/vbac004There is no corresponding record for this reference.
- 73Ritchie, M. E.; Phipson, B.; Wu, D.; Hu, Y.; Law, C. W.; Shi, W.; Smyth, G. K. Limma Powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic Acids Res. 2015, 43 (7), e47– e47, DOI: 10.1093/nar/gkv00773https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFaiu7%252FN&md5=e8a5f44160c650ad89adc48a9912bd96limma powers differential expression analyses for RNA-sequencing and microarray studiesRitchie, Matthew E.; Phipson, Belinda; Wu, Di; Hu, Yifang; Law, Charity W.; Shi, Wei; Smyth, Gordon K.Nucleic Acids Research (2015), 43 (7), e47/1-e47/13CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Limma is an R/Bioconductor software package that provides an integrated soln. for analyzing data from gene expression expts. It contains rich features for handling complex exptl. designs and for information borrowing to overcome the problem of small sample sizes. Over the past decade, limma has been a popular choice for gene discovery through differential expression analyses of microarray and high-throughput PCR data. The package contains particularly strong facilities for reading, normalizing and exploring such data. Recently, the capabilities of limma have been significantly expanded in two important directions. First, the package can now perform both differential expression and differential splicing analyses of RNA sequencing (RNA-seq) data. All the downstream anal. tools previously restricted to microarray data are now available for RNA-seq as well. These capabilities allow users to analyze both RNA-seq and microarray data with very similar pipelines. Second, the package is now able to go past the traditional gene-wise expression analyses in a variety of ways, analyzing expression profiles in terms of co-regulated sets of genes or in terms of higher-order expression signatures. This provides enhanced possibilities for biol. interpretation of gene expression differences. This article reviews the philosophy and design of the limma package, summarizing both new and historical features, with an emphasis on recent enhancements and features that have not been previously described.
- 74Zhou, Y.; Zhou, B.; Pache, L.; Chang, M.; Khodabakhshi, A. H.; Tanaseichuk, O.; Benner, C.; Chanda, S. K. Metascape Provides a Biologist-Oriented Resource for the Analysis of Systems-Level Datasets. Nat. Commun. 2019, 10 (1), 1523, DOI: 10.1038/s41467-019-09234-674https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M%252Fhtl2rtg%253D%253D&md5=cb130c08668f9c8221f464a84039164dMetascape provides a biologist-oriented resource for the analysis of systems-level datasetsZhou Yingyao; Zhou Bin; Khodabakhshi Alireza Hadj; Tanaseichuk Olga; Pache Lars; Chanda Sumit K; Chang Max; Benner ChristopherNature communications (2019), 10 (1), 1523 ISSN:.A critical component in the interpretation of systems-level studies is the inference of enriched biological pathways and protein complexes contained within OMICs datasets. Successful analysis requires the integration of a broad set of current biological databases and the application of a robust analytical pipeline to produce readily interpretable results. Metascape is a web-based portal designed to provide a comprehensive gene list annotation and analysis resource for experimental biologists. In terms of design features, Metascape combines functional enrichment, interactome analysis, gene annotation, and membership search to leverage over 40 independent knowledgebases within one integrated portal. Additionally, it facilitates comparative analyses of datasets across multiple independent and orthogonal experiments. Metascape provides a significantly simplified user experience through a one-click Express Analysis interface to generate interpretable outputs. Taken together, Metascape is an effective and efficient tool for experimental biologists to comprehensively analyze and interpret OMICs-based studies in the big data era.
- 75Torrance, J. D.; Bothwell, T. H. A Simple Technique for Measuring Storage Iron Concentrations in Formalinised Liver Samples. South Afr J. Medical Sci. 1968, 33 (1), 9– 11There is no corresponding record for this reference.
- 76Stirling, D. R.; Swain-Bowden, M. J.; Lucas, A. M.; Carpenter, A. E.; Cimini, B. A.; Goodman, A. CellProfiler 4: Improvements in Speed, Utility and Usability. Bmc Bioinformatics 2021, 22 (1), 433, DOI: 10.1186/s12859-021-04344-976https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2crmtVyksA%253D%253D&md5=048dceafeec6cbb75e0b3c7886b2722aCellProfiler 4: improvements in speed, utility and usabilityStirling David R; Lucas Alice M; Carpenter Anne E; Cimini Beth A; Goodman Allen; Swain-Bowden Madison JBMC bioinformatics (2021), 22 (1), 433 ISSN:.BACKGROUND: Imaging data contains a substantial amount of information which can be difficult to evaluate by eye. With the expansion of high throughput microscopy methodologies producing increasingly large datasets, automated and objective analysis of the resulting images is essential to effectively extract biological information from this data. CellProfiler is a free, open source image analysis program which enables researchers to generate modular pipelines with which to process microscopy images into interpretable measurements. RESULTS: Herein we describe CellProfiler 4, a new version of this software with expanded functionality. Based on user feedback, we have made several user interface refinements to improve the usability of the software. We introduced new modules to expand the capabilities of the software. We also evaluated performance and made targeted optimizations to reduce the time and cost associated with running common large-scale analysis pipelines. CONCLUSIONS: CellProfiler 4 provides significantly improved performance in complex workflows compared to previous versions. This release will ensure that researchers will have continued access to CellProfiler's powerful computational tools in the coming years.
- 77Vizcaíno, J. A.; Côté, R. G.; Csordas, A.; Dianes, J. A.; Fabregat, A.; Foster, J. M.; Griss, J.; Alpi, E.; Birim, M.; Contell, J.; O’Kelly, G.; Schoenegger, A.; Ovelleiro, D.; Pérez-Riverol, Y.; Reisinger, F.; Ríos, D.; Wang, R.; Hermjakob, H. The Proteomics Identifications (PRIDE) Database and Associated Tools: Status in 2013. Nucleic Acids Res. 2012, 41 (D1), D1063– D1069, DOI: 10.1093/nar/gks1262There is no corresponding record for this reference.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsnano.3c08335.
Figure S1, SPION-CCPM uptake in cocultures, EML4-Alk proteomics, gene analysis, γ-H2AX microscopy, Nqo1/Gclc RT-PCR, and GSH assays; Figure S2, cell viability, lipid peroxidation, Gpx4 RT-PCR, LDH, drug assays, antibody neutralization assay, and coculture proteomics; Figure S3, SPION-CCPM intratracheal instillation data in mice; Figure S4, supplementary results for Figures 4 and 5; Table ST1, RT-PCR primer pairs; Table ST2, flow cytometry antibodies (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.