Transcription Inhibitor Lurbinectedin and Oncolytic Peptide LTX-401 Trigger Immunogenic Cell Death and Synergize with Immune Checkpoint Blockade Wei Xie

Total Page:16

File Type:pdf, Size:1020Kb

Transcription Inhibitor Lurbinectedin and Oncolytic Peptide LTX-401 Trigger Immunogenic Cell Death and Synergize with Immune Checkpoint Blockade Wei Xie Transcription Inhibitor Lurbinectedin and Oncolytic Peptide LTX-401 trigger Immunogenic Cell Death and Synergize With Immune Checkpoint Blockade Wei Xie To cite this version: Wei Xie. Transcription Inhibitor Lurbinectedin and Oncolytic Peptide LTX-401 trigger Immunogenic Cell Death and Synergize With Immune Checkpoint Blockade. Cancer. Université Paris-Saclay, 2020. English. NNT : 2020UPASL072. tel-03186843 HAL Id: tel-03186843 https://tel.archives-ouvertes.fr/tel-03186843 Submitted on 31 Mar 2021 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. INDEX ABSTRACT ............................................................................................................... 1 RESUME.................................................................................................................... 2 ORIGINAL PAPERS ................................................................................................. 3 ABBREVIATION ...................................................................................................... 4 INTRODUCTION ..................................................................................................... 7 Cancer and anticancer immunity ............................................................................ 7 Cancer epidemiology .......................................................................................... 7 Cancer immunology ............................................................................................ 8 Immunogenic cell death in cancer therapy ...........................................................13 Major DAMPs of ICD .......................................................................................16 Methods to detect ICD ......................................................................................25 ICD inducers .....................................................................................................27 Induction of ICD sensitize cancers to immune checkpoint blockade ...............31 Transcription inhibitor Lurbinectedin ..................................................................33 Oncolytic peptide LTX-401 .................................................................................36 AIMS OF THE THESIS ..........................................................................................38 RESULTS ................................................................................................................39 Lurbinectedin synergizes with immune checkpoint blockade to generate anticancer immunity .............................................................................................39 Tumor lysis with LTX-401 creates anticancer immunity ....................................41 CONCLUDING REMARKS ...................................................................................42 ACKNOWLEDGEMENT .......................................................................................45 REFERENCES .........................................................................................................46 ANNEX 1: SCIENTIFIC PUBLICATIONS ...........................................................66 ANNEX 2: PAPERS NOT INCLUDED IN THIS THESIS (INSERTED IN PDF) ..................................................................................................................................69 ABSTRACT Cancer is the second leading cause of death worldwide, despite the existence of standard treatment, innovative therapeutic strategies and drugs are still in urgent demand. The combination of immunogenic cell death (ICD) inducing drugs and immune checkpoint blockade (ICB) seems to be a promising modality. In this thesis, we demonstrated Lurbinectedin, a transcription inhibitor newly approved for relapsed lung cancer treatment, triggers hallmarks of ICD in four different human and murine cell line in vitro. Vaccinated with Lurbinectedin-treated fibrosarcoma cell protects immunocompetent mice from rechallenge of syngeneic tumours. Lurbinectedin restrains transplanted fibrosarcoma growth in an immune dependent manner. Both transplanted MCA205 cancer and hormone/carcinogen induced breast cancer were sensitized by Lurbinectedin to PD-1 and CTLA- 4 double ICBs. Of note, long-term immunological memory was generated in cured mice. Further, we evaluated the anticancer capacity of LTX-401, an oncolytic peptide designed for local immunotherapy. Sequential intratumoral injections of LTX-401 dramatically retards subcutaneous MCA205 and TC-1 tumour growth in immunocompetent host, yet shows limited therapeutic effect on abscopal syngeneic tumours. Single LTX-401 administration boosts the efficacy of anti-CTLA-4 or anti-PD-1/anti-CTLA-4 ICBs. Moreover, sequential LTX-401 treatment with double ICBs exhibits systemic antitumor immunity to both treated and abscopal tumour. In conclusion, lurbinectedin and LTX-401 induce cancer cell immunogenic cell death and enhance the anticancer effects of immune checkpoint blockade. These results lay the experimental foundation of combination regiments and may facilitate the clinical trial designs. Key words: cancer, immunogenic cell death, immune checkpoint blockade, transcription, oncolytic peptide 1 RESUME Le cancer est la deuxième cause de mortalité dans le monde. Malgré l’existence des traitements standards, le développement et la recherche de stratégies thérapeutiques innovantes et de médicaments est toujours nécessaire. La combinaison des médicaments, induisant la mort cellulaire immunogène (ICD) et l’inhibition des points de contrôle immunitaire (ICB), semble être un protocole prometteur. Dans cette thèse, nous avons démontré que la Lurbinectédine, un inhibiteur de la transcription nouvellement approuvé pour le traitement du cancer du poumon récidivant, déclenche les caractéristiques de l’ICD dans quatre différentes lignées cellulaires humaines et murines in vitro. Vaccinée par des cellules de fibrosarcome traitées par la lurbinectine, les souris immunocompétentes sont protégées lors du rechallenge des tumeurs syngéniques. La lurbinectédine limite la croissance du fibrosarcome transplanté d'une manière immunodépendante. Dans les souris, le fibrosarcome murin (MCA205) transplanté et le cancer du sein, induit par des hormones en combinaison avec des cancérigènes, ont été sensibilisés par la lurbinectédine aux deux ICB : PD-1 et CTLA-4. Il convient de noter que la mémoire immunologique à long terme a été générée chez des souris guéries. En outre, nous avons évalué la capacité anticancéreuse de LTX-401, un peptide oncolytique conçu pour l'immunothérapie locale. Les injections intratumorales séquentielles de LTX-401 retardent considérablement la croissance des tumeurs sous-cutanées MCA205 et TC-1 chez un hôte immunocompétent, mais montrent un effet thérapeutique limité sur les tumeurs syngéniques abscopales. Une seule administration de LTX-401 augmente l'efficacité des ICB anti-CTLA-4 ou anti-PD-1 + anti-CTLA-4. De plus, le traitement séquentiel avec LTX-401 et les deux ICB présente une immunité antitumorale systémique à la fois contre la tumeur traitée et la tumeur abscopale. En conclusion, la lurbinectédine et le LTX-401 induisent la mort cellulaire immunogène des cellules cancéreuses et renforcent les effets anticancéreux des inhibiteurs de points de contrôle immunitaires. Ces résultats jettent les bases expérimentales de traitements combinés et peuvent faciliter les conceptions d'essais cliniques. Mots clés: cancer, mort cellulaire immunogène, inhibiteur de point de contrôle immunitaire, transcription, peptide oncolytique 2 ORIGINAL PAPERS This thesis is based on the following papers: Paper I. Lurbinectedin synergizes with immune checkpoint blockade to generate anticancer immunity. Xie W, Forveille S, Iribarren K, Sauvat A, Senovilla L, Wang Y, Humeau J, Perez-Lanzon M, Zhou H, Martínez-Leal JF, Kroemer G, Kepp O. Published in Oncoimmunology, 2019, VOL. 8, NO. 11, e1656502 (9 pages). Paper II. Tumor lysis with LTX-401 creates anticancer immunity. Xie W, Mondragón L, Mauseth B, Wang Y, Pol J, Lévesque S, Zhou H, Yamazaki T, Eksteen JJ, Zitvogel L, Sveinbjørnsson B, Rekdal Ø, Kepp O, Kroemer G. Published in Oncoimmunology, 2019, VOL. 8, NO. 7, e1594555 (8 pages). (In agreement with the doctoral school, Paper I&II are inserted to replace chapter ‘Materials and methods’ and ‘Results’.) 3 ABBREVIATION AIDS acquired immune deficiency syndrome ATF activating transcription factor AML acute myeloid leukaemia ATP adenosine triphosphate ANXA1 annexin A1 APCs antigen-presenting cells AMPs antimicrobial peptides LfcinB bovine lactoferricin BFA brefeldin A CALR calreticulin CRC colorectal cancer CXCL10 CXC-chemokine ligand 10 CTLA-4 cytotoxic T-lymphocyte-associated protein 4 DAMPs damage-associated molecular patterns DC dendritic cell DMBA dimethylbenzantracene DOXO doxorubicin ER endoplasmic reticulum ERAD ER-associated degradation eIF2α eukaryotic translation initiation factor 2 FDA federal drug administration FPR1 formyl peptide receptor 1 GCN2 general control nonderepressible 2 Hsp70 heat shock protein 70 HCC
Recommended publications
  • Specific Effects of Trabectedin and Lurbinectedin on Human Macrophage Function and Fate—Novel Insights
    cancers Article Specific Effects of Trabectedin and Lurbinectedin on Human Macrophage Function and Fate—Novel Insights 1, 1, 1 Adrián Povo-Retana y, Marina Mojena y, Adrian B. Stremtan , Victoria B. Fernández-García 1, Ana Gómez-Sáez 1, Cristina Nuevo-Tapioles 2,3 , José M. Molina-Guijarro 4 , José Avendaño-Ortiz 5, José M. Cuezva 2,3 , Eduardo López-Collazo 5, Juan F. Martínez-Leal 4 and Lisardo Boscá 1,5,6,* 1 Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain; [email protected] (A.P.-R.); [email protected] (M.M.); [email protected] (A.B.S.); [email protected] (V.B.F.-G.); [email protected] (A.G.-S.) 2 Centro de Biología Molecular (Centro Mixto CSIC-UAM), Nicolás Cabrera S/N, Ciudad Universitaria de Cantoblanco, 28049 Madrid, Spain; [email protected] (C.N.-T.); [email protected] (J.M.C.) 3 Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), 28029 Madrid, Spain 4 Pharma Mar SA, 28770 Colmenar Viejo, Spain; [email protected] (J.M.M.-G.); [email protected] (J.F.M.-L.) 5 Instituto de Investigación Sanitaria La Paz (IdiPaz), Hospital Universitario La Paz, 28046 Madrid, Spain; [email protected] (J.A.-O.); [email protected] (E.L.-C.) 6 Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain * Correspondence: [email protected]; Tel.: +34-9149-72747 These authors have equal contribution. y Received: 28 August 2020; Accepted: 16 October 2020; Published: 20 October 2020 Simple Summary: Trabectedin and lurbinectedin are two potent onco-therapeutic drugs for the treatment of advanced soft tissue sarcomas.
    [Show full text]
  • The Emerging Relevance of AIM2 in Liver Disease
    International Journal of Molecular Sciences Review The Emerging Relevance of AIM2 in Liver Disease Beatriz Lozano-Ruiz 1,2 and José M. González-Navajas 1,2,3,4,* 1 Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain; [email protected] 2 Department of Pharmacology, Paediatrics and Organic Chemistry, University Miguel Hernández (UMH), 03550 San Juan, Alicante, Spain 3 Networked Biomedical Research Center for Hepatic and Digestive Diseases (CIBERehd), Institute of Health Carlos III, 28029 Madrid, Spain 4 Institute of Research, Development and Innovation in Healthcare Biotechnology in Elche (IDiBE), University Miguel Hernández, 03202 Elche, Alicante, Spain * Correspondence: [email protected]; Tel.: +34-(965)-913-928 Received: 16 August 2020; Accepted: 4 September 2020; Published: 7 September 2020 Abstract: Absent in melanoma 2 (AIM2) is a cytosolic receptor that recognizes double-stranded DNA (dsDNA) and triggers the activation of the inflammasome cascade. Activation of the inflammasome results in the maturation of inflammatory cytokines, such as interleukin (IL)-1 β and IL-18, and a form of cell death known as pyroptosis. Owing to the conserved nature of its ligand, AIM2 is important during immune recognition of multiple pathogens. Additionally, AIM2 is also capable of recognizing host DNA during cellular damage or stress, thereby contributing to sterile inflammatory diseases. Inflammation, either in response to pathogens or due to sterile cellular damage, is at the center of the most prevalent and life-threatening liver diseases. Therefore, during the last 15 years, the study of inflammasome activation in the liver has emerged as a new research area in hepatology. Here, we discuss the known functions of AIM2 in the pathogenesis of different hepatic diseases, including non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH), hepatitis B, liver fibrosis, and hepatocellular carcinoma (HCC).
    [Show full text]
  • AIM2 Inflammasome Is Activated by Pharmacological Disruption Of
    AIM2 inflammasome is activated by pharmacological PNAS PLUS disruption of nuclear envelope integrity Antonia Di Miccoa,1, Gianluca Freraa,1,JérômeLugrina,1, Yvan Jamillouxa,b,Erh-TingHsuc,AubryTardivela, Aude De Gassarta, Léa Zaffalona, Bojan Bujisica, Stefanie Siegertd, Manfredo Quadronie, Petr Brozf, Thomas Henryb,ChristineA.Hrycynac,g, and Fabio Martinona,2 aDepartment of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland; bINSERM, U1111, Center for Infectiology Research, Lyon 69007, France; cDepartment of Chemistry, Purdue University, West Lafayette, IN 47907-2084; dFlow Cytometry Facility, Ludwig Center for Cancer Research, University of Lausanne, Epalinges 1066, Switzerland; eProtein Analysis Facility, Center for Integrative Genomics, University of Lausanne, Lausanne 1015, Switzerland; fFocal Area Infection Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland; and gPurdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907-2084 Edited by Zhijian J. Chen, University of Texas Southwestern Medical Center/Howard Hughes Medical Institute, Dallas, TX, and approved June 21, 2016 (received for review February 12, 2016) Inflammasomes are critical sensors that convey cellular stress and and in vitro (19, 20). Beyond their broad use as anti-HIV drugs, pathogen presence to the immune system by activating inflamma- these molecules display beneficial HIV-unrelated functions, anti- tory caspases and cytokines such as IL-1β. The nature of endogenous malaria, antituberculosis, and antitumor properties (20). At the stress signals that activate inflammasomes remains unclear. Here cellular level, the HIV-PIs trigger an atypical ER stress-like we show that an inhibitor of the HIV aspartyl protease, Nelfinavir, transcriptional response that relies mostly on the activation of the triggers inflammasome formation and elicits an IL-1R–dependent integrated stress response (19, 21).
    [Show full text]
  • A Phase I Dose-Finding, Pharmacokinetics and Genotyping
    www.nature.com/scientificreports OPEN A phase I dose‑fnding, pharmacokinetics and genotyping study of olaparib and lurbinectedin in patients with advanced solid tumors Andres Poveda1*, Ana Oaknin2, Ignacio Romero3, Angel Guerrero‑Zotano3, Lorena Fariñas‑Madrid2, Victor Rodriguez‑Freixinos4, Pedro Mallol5, Raquel Lopez‑Reig6,7 & Jose Antonio Lopez‑Guerrero6,7,8 The poly (ADP‑Ribose) polymerase (PARP) inhibitor olaparib has shown antitumor activity in patients with ovarian or breast cancer with or without BRCA1/2 mutations. Lurbinectedin is an ecteinascidin that generates DNA double‑strand breaks. We hypothesized that the combination of olaparib and lurbinectedin maximizes the DNA damage increasing the efcacy. A 3 + 3 dose‑escalation study examined olaparib tablets with lurbinectedin every 21 days. The purpose of this phase I study is to determine the dose‑limiting toxicities (DLTs) of the combination, to investigate the maximum tolerated dose (MTD), the recommended phase II dose (RP2D), efcacy, pharmacokinetics, in addition to genotyping and translational studies. In total, 20 patients with ovarian and endometrial cancers were included. The most common adverse events were asthenia, nausea, vomiting, constipation, abdominal pain, neutropenia, anemia. DLT grade 4 neutropenia was observed in two patients in dose level (DL) 5, DL4 was defned as the MTD, and the RP2D was lurbinectedin 1.5 mg/m2 + olaparib 250 mg twice a day (BID). Mutational analysis revealed a median of 2 mutations/case, 53% of patients with mutations in the homologous recombination (HR) pathway. None of the patients reached a complete or partial response; however, 60% of stable disease was achieved. In conclusion, olaparib in combination with lurbinectedin was well tolerated with a disease control rate of 60%.
    [Show full text]
  • Interleukin-18 in Health and Disease
    International Journal of Molecular Sciences Review Interleukin-18 in Health and Disease Koubun Yasuda 1 , Kenji Nakanishi 1,* and Hiroko Tsutsui 2 1 Department of Immunology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan; [email protected] 2 Department of Surgery, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan; [email protected] * Correspondence: [email protected]; Tel.: +81-798-45-6573 Received: 21 December 2018; Accepted: 29 January 2019; Published: 2 February 2019 Abstract: Interleukin (IL)-18 was originally discovered as a factor that enhanced IFN-γ production from anti-CD3-stimulated Th1 cells, especially in the presence of IL-12. Upon stimulation with Ag plus IL-12, naïve T cells develop into IL-18 receptor (IL-18R) expressing Th1 cells, which increase IFN-γ production in response to IL-18 stimulation. Therefore, IL-12 is a commitment factor that induces the development of Th1 cells. In contrast, IL-18 is a proinflammatory cytokine that facilitates type 1 responses. However, IL-18 without IL-12 but with IL-2, stimulates NK cells, CD4+ NKT cells, and established Th1 cells, to produce IL-3, IL-9, and IL-13. Furthermore, together with IL-3, IL-18 stimulates mast cells and basophils to produce IL-4, IL-13, and chemical mediators such as histamine. Therefore, IL-18 is a cytokine that stimulates various cell types and has pleiotropic functions. IL-18 is a member of the IL-1 family of cytokines. IL-18 demonstrates a unique function by binding to a specific receptor expressed on various types of cells.
    [Show full text]
  • The Essentials of Marine Biotechnology. Frontiers in Marine Science [Online], 8, Article 629629
    ROTTER, A., BARBIER, M., BERTONI, F. et al. 2021. The essentials of marine biotechnology. Frontiers in marine science [online], 8, article 629629. Available from: https://doi.org/10.3389/fmars.2021.629629 The essentials of marine biotechnology. ROTTER, A., BARBIER, M., BERTONI, F. et al. 2021 Copyright © 2021 Rotter, Barbier, Bertoni, Bones, Cancela, Carlsson, Carvalho, Cegłowska, Chirivella-Martorell, Conk Dalay, Cueto, Dailianis, Deniz, Díaz-Marrero, Drakulovic, Dubnika, Edwards, Einarsson, Erdoˇgan, Eroldoˇgan, Ezra, Fazi, FitzGerald, Gargan, Gaudêncio, Gligora Udoviˇc, Ivoševi´c DeNardis, Jónsdóttir, Kataržyt˙e, Klun, Kotta, Ktari, Ljubeši´c, Luki´c Bilela, Mandalakis, Massa-Gallucci, Matijošyt˙e, Mazur-Marzec, Mehiri, Nielsen, Novoveská, Overling˙e, Perale, Ramasamy, Rebours, Reinsch, Reyes, Rinkevich, Robbens, Röttinger, Rudovica, Sabotiˇc, Safarik, Talve, Tasdemir, Theodotou Schneider, Thomas, Toru´nska-Sitarz, Varese and Vasquez.. This article was first published in Frontiers in Marine Science on 16.03.2021. This document was downloaded from https://openair.rgu.ac.uk fmars-08-629629 March 10, 2021 Time: 14:8 # 1 REVIEW published: 16 March 2021 doi: 10.3389/fmars.2021.629629 The Essentials of Marine Biotechnology Ana Rotter1*, Michéle Barbier2, Francesco Bertoni3,4, Atle M. Bones5, M. Leonor Cancela6,7, Jens Carlsson8, Maria F. Carvalho9, Marta Cegłowska10, Jerónimo Chirivella-Martorell11, Meltem Conk Dalay12, Mercedes Cueto13, 14 15 16 17 Edited by: Thanos Dailianis , Irem Deniz , Ana R. Díaz-Marrero , Dragana Drakulovic , 18 19
    [Show full text]
  • Marine Vibrionaceae As a Reservoir for Bioprospecting and Ecology Studies
    Downloaded from orbit.dtu.dk on: Oct 09, 2021 Marine Vibrionaceae as a reservoir for bioprospecting and ecology studies Giubergia, Sonia Publication date: 2016 Document Version Publisher's PDF, also known as Version of record Link back to DTU Orbit Citation (APA): Giubergia, S. (2016). Marine Vibrionaceae as a reservoir for bioprospecting and ecology studies. Novo Nordisk Foundation Center for Biosustainability. General rights Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. Users may download and print one copy of any publication from the public portal for the purpose of private study or research. You may not further distribute the material or use it for any profit-making activity or commercial gain You may freely distribute the URL identifying the publication in the public portal If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Marine Vibrionaceae as a reservoir for bioprospecting and ecology studies. Sonia Giubergia PhD thesis April 2016 Copyright: Sonia Giubergia 2016 Novo Nordisk Foundation Center for Biosustainability Technical University of Denmark Kogle Allè 6 – 2970 Horshølm Denmark Cover: the Mediterranean Sea at Hyères, France (Sonia Giubergia) ii PREFACE The present PhD study has been conducted at the Department of Systems Biology and at the Novo Nordisk Foundation Centre for Biosustainability at the Technical University of Denmark from May 2013 to April 2016 under the supervision of Professor Lone Gram.
    [Show full text]
  • Complementary Regulation of Caspase-1 and IL-1Β Reveals Additional Mechanisms of Dampened Inflammation in Bats
    Complementary regulation of caspase-1 and IL-1β reveals additional mechanisms of dampened inflammation in bats Geraldine Goha,1, Matae Ahna,1, Feng Zhua, Lim Beng Leea, Dahai Luob,c, Aaron T. Irvinga,d,2, and Lin-Fa Wanga,e,2 aProgramme in Emerging Infectious Diseases, Duke–National University of Singapore Medical School, 169857, Singapore; bLee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore; cNTU Institute of Structural Biology, Nanyang Technological University, 636921, Singapore; dZhejiang University–University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University International Campus, Haining, 314400, China; and eSinghealth Duke–NUS Global Health Institute, 169857, Singapore Edited by Vishva M. Dixit, Genentech, San Francisco, CA, and approved September 14, 2020 (received for review February 21, 2020) Bats have emerged as unique mammalian vectors harboring a experimental confirmation is rare. We recently demonstrated diverse range of highly lethal zoonotic viruses with minimal that NLRP3 is dampened in bats as a result of loss-of-function clinical disease. Despite having sustained complete genomic loss bat-specific isoforms and impaired transcriptional priming (9). of AIM2, regulation of the downstream inflammasome response in The stimulator of IFN genes (STING), a key adaptor to the bats is unknown. AIM2 sensing of cytoplasmic DNA triggers ASC DNA-sensing cGAS protein, is also exclusively mutated at S358 aggregation and recruits caspase-1, the central inflammasome ef- in bats, resulting in a reduced IFN response to HSV1 (10). We fector enzyme, triggering cleavage of cytokines such as IL-1β and previously reported a complete absence of Absent in melanoma inducing GSDMD-mediated pyroptotic cell death.
    [Show full text]
  • Cancer Control Potential of Marine Natural Product Scaffolds Through
    Drug Discovery Today Volume 21, Number 11 November 2016 REVIEWS Teaser Greater emphasis on marine natural products research could selectively cure multiple human ailments with fewer hazardous effects. Cancer control potential of marine natural product scaffolds through FOUNDATION REVIEW inhibition of tumor cell migration and invasion Reviews 1 2 Mudit Mudit BS Mudit Mudit and Khalid A. El Sayed Pharmacy, PhD, is an assistant professor in the 1 Department of Pharmaceutical, Social and Administrative Sciences, D’Youville College School of Pharmacy, Department of Pharmaceutical, Social and Buffalo, NY 14201, USA 2 Administrative Sciences at Department of Basic Pharmaceutical Sciences, School of Pharmacy, The University of Louisiana at Monroe, the D’Youville College Monroe, LA 71201, USA School of Pharmacy. He is an active member of several professional organizations, including the The marine environment is a reliable source for the discovery of novel American Association of Colleges of Pharmacy, American Association of Pharmaceutical Scientists treatment options for numerous diseases. Past research efforts toward the (AAPS), American Chemical Society, and Rho Chi discovery of marine-derived anticancer agents have resulted in several Pharmacy Honor Society. He has given numerous research presentations both at regional and national commercially available marine-based drugs. The pharmaceutical value of meetings, and has been recognized by the AAPS for excellence in graduate education in the fields of Drug anticancer drugs from marine natural products (MNPs) ranges from Design and Discovery. He has also been a reviewer for US$563 billion to US$5.69 trillion. In this review, we highlight several many scientific manuscripts, research and educational grants, and annual professional meeting abstracts and marine-derived entities with the potential for cancer control and posters.
    [Show full text]
  • Lurbinectedin Specifically Triggers the Degradation of Phosphorylated RNA Polymerase II and the Formation of DNA Breaks in Cancer Cells
    Published OnlineFirst September 14, 2016; DOI: 10.1158/1535-7163.MCT-16-0172 Small Molecule Therapeutics Molecular Cancer Therapeutics Lurbinectedin Specifically Triggers the Degradation of Phosphorylated RNA Polymerase II and the Formation of DNA Breaks in Cancer Cells Gema Santamaría Nunez~ 1, Carlos Mario Genes Robles2, Christophe Giraudon2, Juan Fernando Martínez-Leal1, Emmanuel Compe2,Fred eric Coin2, Pablo Aviles1, Carlos María Galmarini1, and Jean-Marc Egly2 Abstract We have defined the mechanism of action of lurbinectedin, a ubiquitin/proteasome machinery; and (iii) the generation of marine-derived drug exhibiting a potent antitumor activity DNA breaks and subsequent apoptosis. The finding that inhi- across several cancer cell lines and tumor xenografts. This drug, bition of Pol II phosphorylation prevents its degradation and currently undergoing clinical evaluation in ovarian, breast, and the formation of DNA breaks after drug treatment underscores smallcelllungcancerpatients,inhibits the transcription pro- the connection between transcription elongation and DNA cess through (i) its binding to CG-rich sequences, mainly repair. Our results not only help to better understand the high located around promoters of protein-coding genes; (ii) the specificity of this drug in cancer therapy but also improve our irreversible stalling of elongating RNA polymerase II (Pol II) understanding of an important transcription regulation mech- on the DNA template and its specific degradation by the anism. Mol Cancer Ther; 15(10); 1–14. Ó2016 AACR. Introduction derivatives, anthracyclines, etc.; ref. 10). Currently, several laboratories are developing inhibitors of cyclin-dependent Cancer cells aberrantly deregulate specific gene expression kinases (CDK) that have a critical role in regulating transcrip- programs with critical functions in cell differentiation, prolifer- tion initiation, pause release, and elongation (e.g., CDK7, ation, and survival (1).
    [Show full text]
  • Mechanisms and Therapeutic Regulation of Pyroptosis in Inflammatory Diseases and Cancer
    International Journal of Molecular Sciences Review Mechanisms and Therapeutic Regulation of Pyroptosis in Inflammatory Diseases and Cancer Zhaodi Zheng and Guorong Li * Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan 250014, China; [email protected] * Correspondence: [email protected]; Tel.: +86-531-8618-2690 Received: 24 January 2020; Accepted: 17 February 2020; Published: 20 February 2020 Abstract: Programmed Cell Death (PCD) is considered to be a pathological form of cell death when mediated by an intracellular program and it balances cell death with survival of normal cells. Pyroptosis, a type of PCD, is induced by the inflammatory caspase cleavage of gasdermin D (GSDMD) and apoptotic caspase cleavage of gasdermin E (GSDME). This review aims to summarize the latest molecular mechanisms about pyroptosis mediated by pore-forming GSDMD and GSDME proteins that permeabilize plasma and mitochondrial membrane activating pyroptosis and apoptosis. We also discuss the potentiality of pyroptosis as a therapeutic target in human diseases. Blockade of pyroptosis by compounds can treat inflammatory disease and pyroptosis activation contributes to cancer therapy. Keywords: pyroptosis; GSDMD; GSDME; inflammatory disease; cancer therapy 1. Introduction Many disease states are cross-linked with cell death. The Nomenclature Committee on Cell Death make a series of recommendations to systematically classify cell death [1,2]. Programmed Cell Death (PCD) is mediated by specific cellular mechanisms and some signaling pathways are activated in these processes [3]. Apoptosis, autophagy and programmed necrosis are the three main types of PCD [4], and they may jointly determine the fate of malignant tumor cells.
    [Show full text]
  • Emerging Role of PYHIN Proteins As Antiviral Restriction Factors
    viruses Review Emerging Role of PYHIN Proteins as Antiviral Restriction Factors Matteo Bosso and Frank Kirchhoff * Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; [email protected] * Correspondence: frank.kirchhoff@uni-ulm.de; Tel.: +49-731-50065150 Academic Editor: Sébastien Nisole Received: 26 November 2020; Accepted: 16 December 2020; Published: 18 December 2020 Abstract: Innate immune sensors and restriction factors are cellular proteins that synergize to build an effective first line of defense against viral infections. Innate sensors are usually constitutively expressed and capable of detecting pathogen-associated molecular patterns (PAMPs) via specific pattern recognition receptors (PRRs) to stimulate the immune response. Restriction factors are frequently upregulated by interferons (IFNs) and may inhibit viral pathogens at essentially any stage of their replication cycle. Members of the Pyrin and hematopoietic interferon-inducible nuclear (HIN) domain (PYHIN) family have initially been recognized as important sensors of foreign nucleic acids and activators of the inflammasome and the IFN response. Accumulating evidence shows, however, that at least three of the four members of the human PYHIN family restrict viral pathogens independently of viral sensing and innate immune activation. In this review, we provide an overview on the role of human PYHIN proteins in the innate antiviral immune defense and on viral countermeasures. Keywords: PYHIN; DNA sensing; restriction factors; viral counteraction; immune evasion 1. Introduction Viruses strictly rely on their host cells for replication and spread. However, although viral pathogens are capable of exploiting numerous cellular factors and pathways, the cell does not provide a friendly environment. As a consequence of countless past encounters with viral pathogens, mammalian cells have evolved sensors of foreign invaders that alert and activate a large variety of antiviral effector proteins [1–5].
    [Show full text]