Structural Studies of a New Class of Immune Checkpoint

Total Page:16

File Type:pdf, Size:1020Kb

Structural Studies of a New Class of Immune Checkpoint STRUCTURAL STUDIES OF A NEW CLASS OF IMMUNE CHECKPOINT INHIBITOR FOR CANCER IMMUNOTHERAPY By NINA HARPELL A senior thesis submitted in partial fulfillment of the requirements for the degree of BACHELOR OF SCIENCE UNIVERSITY OF CALIFORNIA SANTA CRUZ Chemistry and Biochemistry APRIL 2020 ACKNOWLEDGMENT I would like to thank my advisor, Dr. Rebecca DuBois, for providing me with the opportunity to begin this project as well as encourage my pursuit of research beyond an undergraduate degree. I would also like to thank the additional members of the Dubois Lab: Natasha George, Ana Nunez Castrejon, Lena Meyer, John Dzimianski, Jordan Ford, and Kevin Delgado-Cunningham. Additional thanks go to Nicholas Lorig-Roach, for his mentorship and support in my research and education. ii Abstract Natural Killer (NK) cells are a key component of the innate immune system that play a role in eliminating both tumors and virus-infected cells. NK cells express a deactivating receptor, KIR2DL3, which inhibits their cytotoxic function when bound by its ligand, class I MHC. Lirilumab, an Anti-KIR antibody (mAb) in clinical trials, prevents deactivation of the NK cell by binding to the KIR2DL3 receptor. However, the structural basis of lirilumabâĂŹs disruption of KIR2DL3 activity is unknown. To that end, the first aim of this project is to use X-ray crystallography to determine the structure of the lirilumab-KIR2DL3 complex to reveal how the antibody blocks MHC binding to the KIR receptor. A series of expression constructs for the KIR2DL3 receptor with different tag configurations for affinity chromatography were cloned then expressed in HEK293 and CHO cells. Biolayer interferometry assays demonstrate binding of an expressed and purified KIR2DL3 receptor with a control mAb and will also be used to determine Lirilumab’s affinity. Identification of stable expression constructs, as well as structural epitopes of the KIR-mAb construct, will implement a foundation for the study of the Lirilumab mAb as a cancer therapeutic. TABLE OF CONTENTS Page ACKNOWLEDGMENT ................................ ii ABSTRACT ....................................... iii LIST OF FIGURES ................................... vi CHAPTER 1 Introduction .................................... 1 2 Background .................................... 3 2.1 Cancer and Immunotherapy ......................... 3 2.1.1 Immunotherapy for Cancer...................... 3 2.1.2 Immune Checkpoints ......................... 4 2.1.3 Immune Checkpoint Inhibitors.................... 4 2.2 Characterizing Different Antibodies and Their Function.......... 5 2.3 Natural Killer Cells.............................. 6 2.4 NK Cells KIR2DL3 Receptor and Function................. 7 2.5 Antibodies Bound to KIR2DL3 are Immune Checkpoint Inhibitors.... 7 2.6 Prior Work................................... 9 3 Work Completed ................................. 12 3.1 Cloning the KIR2DL3 Receptor ....................... 12 3.2 Protein Expression of KIR2DL3 in HEK293 Cells ............. 14 3.3 Protein Expression of KIR2DL3 in CHO Cells ............... 18 3.4 Cloning the TRL8605 mAb.......................... 19 3.5 Protein Expression of TRL8605 in HEK293 and CHO Cells . 20 3.6 Binding studies of TRL8605 mAb and KIR2DL3 Receptor . 21 3.7 Cloning the Lirilumab mAb ......................... 25 iv 3.8 KIR2DL3 Protein Expression in E.Coli Cells................ 25 4 Future Work ................................... 28 4.1 Lirilumab Protein Expression in CHO Cells................. 28 4.2 Binding Studies of Lirilumab mAb and KIR2DL3 receptor . 29 4.3 Crystallization of anti-KIR mAb bound to KIR2DL3 Receptor . 29 5 Conclusion ..................................... 30 REFERENCES ...................................... 34 APPENDIX A Materials ...................................... 36 B Detailed Methods ................................ 39 B.1 Polymerase Chain Reaction (PCR) ..................... 39 B.2 Restriction Digestion ............................. 39 B.3 Transformation into E. coli ......................... 40 B.4 SDS-PAGE, Western Blot .......................... 40 B.5 Small-scale Protein Purification (His-tag purification via affinity chromatog- raphy) ..................................... 40 B.6 Transfection of HEK293 Cells ........................ 41 B.7 Transfection of CHO cells .......................... 42 C Sequences ..................................... 44 C.1 Protein Sequence: TRL8605 Heavy Chain Variable Region . 44 C.2 Protein Sequence: TRL8605 Light Chain Variable Region . 44 C.3 Protein Sequence: Lirilumab SCFV..................... 44 C.4 Protein Sequence: KIR2DL3 Receptor.................... 45 v LIST OF FIGURES 2.1 IgG1, 2, 3, and 4 antibodies........................... 6 2.2 Antibody Bound to Target Cell and FC Receptor of NK Cell......... 7 2.3 NK Cell KIR Deactivating Receptor and Tumor Cell HLA Antigen (MHC). 8 2.4 NK Cell Activation through Anti-KIR mAb blockade............. 8 2.5 Tumor volume as a function of mAb administered................ 10 2.6 Binding affinity of TRL mAbs for KIR2DL3 receptor on NK cells measured on ELISA...................................... 11 3.1 KIR2DL3-cmyc-6His SDS-PAGE and Western Blot. ............. 17 3.2 SDS-Page of commercial His-tag KIR2DL3 Receptor.............. 17 3.3 The 10-hisKIR2DL3 receptor protein on SDS-Page gel............. 18 3.4 SDS-Page gel of KIR2DL3 his-tagged receptor protein. ............ 19 3.5 SDS-Page gel for TRL8605 mAb produced in HEK293 and CHO cells. 21 3.6 BLI assay scheme from left to right........................ 22 3.7 BLI assay of TRL8605 mAb and KIR2DL3 receptor produced in HEK293F cells. ....................................... 23 3.8 BLI assay of the TRL8605 (CHO) mAb and KIR2DL3 receptor (CHO). 24 3.9 BLI assay of KIR2DL3 receptor and TRL8605 mAb and control mAb. 24 vi 3.10 PCR products on agarose gel of Lirilumab light chain and heavy chain variable gene fragments................................... 26 3.11 Western Blot of KIR2DL3 receptor protein pellet and lysate prior to purification. 27 vii Chapter One Introduction Behind heart disease, cancer is the second most common cause of death in the United States. Treatments, such as chemotherapy and radiation therapy have been developed to combat cancer by targeting rapidly dividing cancer cells. However, healthy cells that divide at similar rates are often damaged or killed. Because these treatments do not target specific cancer cells, damaging healthy cells poses dangerous risks for the patient to develop toxic side effects or death. Even when these traditional methods appear to work and shrink tumor volumes, relapse and regrowth of tumors after treatment ends is a daunting problem. Cancer immunotherapy is a new and promising avenue for treatment that uses a patient’s own immune system to recognize and target cancer cells. Innovative therapies such as CAR T-Cell therapy have introduced new methodologies of engineering an individual’s T-cells for targeted tumor detection and cytotoxic function [3]. Other immunotherapies are utilizing engineered mAbs to serve as checkpoint inhibitors which retain activation of T-Cells and Natural Killer Cells in the presence of tumor cells. Ipilimumab and Avelumab are both FDA approved immune checkpoint inhibitors for the treatment of skin and lung cancer [17]. Lirilumab, a monoclonal antibody (mAb) in development by Innate Pharma and Brystol- Meyers Squibb, is currently in clinical trials for a variety of cancer indications, both alone and in combination with other therapeutics [18]. Lirilumab is an immune checkpoint inhibitor that binds to the KIR2DL3 receptor on Natural Killer (NK) cells, activating them and as- 1 sisting their killing of tumor cells. While the Lirilumab mAb is known to bind KIR2DL3 and block ligand interaction, there is no structural information on the Lirilumab-KIR complex, which could reveal how this antibody is able to specifically target inhibitory KIRs without affecting the homologous activating KIRs (or some sentence like this). Another company, Trellis Biosciences, has also developed a mAb, TRL8605, similar to lirilumab with higher binding affinity for the KIR receptor [19]. The structural basis for any similarities or differences between these therapeutic mAbs is unknown.Therefore, the first goal of this project is to compare binding affinity of the TRL8605 mAb with lirilumab to verify Trellis’ claim. The second goal is to use X-Ray crystallography to determine the structures of the Anti-KIR antibodies bound to the KIR receptor. Characterizing structural components will provide better understanding of which struc- tural epitopes are active in the complex binding site. Furthermore, understanding the mecha- nism these anti-KIR mAbs use to block interaction between the NK cell and MHC of tumor cells would provide insight into developing a mAb that can effectively target cancer cells without damaging healthy cells. 2 Chapter Two Background 2.1 Cancer and Immunotherapy Cancer is a group of diseases involving abnormal cell growth with the potential to invade or spread to other parts of the body [2]. In 2015, about 90.5 million people had cancer and about 14.1 million new cases occur a year [1]. Cancer accounts for 8.8 million (roughly 15.5%) deaths each year. The most common types of cancer such as breast cancer, lung cancer, and colon cancer which are initially treated with surgery [2]. Treatment options depend on both the cancer and the patient as well as available treatment options [2]. However, most available treatment options cannot specifically target individual cancer cells.
Recommended publications
  • Systemic Immunotherapy for Urothelial Cancer: Current Trends and Future Directions
    Review Systemic Immunotherapy for Urothelial Cancer: Current Trends and Future Directions Shilpa Gupta 1,†, David Gill 2,†, Austin Poole 2 and Neeraj Agarwal 2,* 1 Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; [email protected] 2 Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; [email protected] (D.G.); [email protected] (A.P.) * Correspondence: [email protected]; Tel.: +1-801-213-5658; Fax: +1-801-585-0124 † These authors contributed equally to this work. Academic Editor: Vita Golubovskaya Received: 14 December 2016; Accepted: 18 January 2017; Published: 27 January 2017 Abstract: Urothelial cancer of the bladder, renal pelvis, ureter, and other urinary organs is the fifth most common cancer in the United States, and systemic platinum-based chemotherapy remains the standard of care for first-line treatment of advanced/metastatic urothelial carcinoma (UC). Until recently, there were very limited options for patients who are refractory to chemotherapy, or do not tolerate chemotherapy due to toxicities and overall outcomes have remained very poor. While the role of immunotherapy was first established in non-muscle invasive bladder cancer in the 1970s, no systemic immunotherapy was approved for advanced disease until the recent approval of a programmed death ligand-1 (PD-L1) inhibitor, atezolizumab, in patients with advanced/metastatic UC who have progressed on platinum-containing regimens. This represents a significant milestone in this disease after a void of over 30 years. In addition to atezolizumab, a variety of checkpoint inhibitors have shown a significant activity in advanced/metastatic urothelial carcinoma and are expected to gain Food and Drug Administration (FDA) approval in the near future.
    [Show full text]
  • Classification Decisions Taken by the Harmonized System Committee from the 47Th to 60Th Sessions (2011
    CLASSIFICATION DECISIONS TAKEN BY THE HARMONIZED SYSTEM COMMITTEE FROM THE 47TH TO 60TH SESSIONS (2011 - 2018) WORLD CUSTOMS ORGANIZATION Rue du Marché 30 B-1210 Brussels Belgium November 2011 Copyright © 2011 World Customs Organization. All rights reserved. Requests and inquiries concerning translation, reproduction and adaptation rights should be addressed to [email protected]. D/2011/0448/25 The following list contains the classification decisions (other than those subject to a reservation) taken by the Harmonized System Committee ( 47th Session – March 2011) on specific products, together with their related Harmonized System code numbers and, in certain cases, the classification rationale. Advice Parties seeking to import or export merchandise covered by a decision are advised to verify the implementation of the decision by the importing or exporting country, as the case may be. HS codes Classification No Product description Classification considered rationale 1. Preparation, in the form of a powder, consisting of 92 % sugar, 6 % 2106.90 GRIs 1 and 6 black currant powder, anticaking agent, citric acid and black currant flavouring, put up for retail sale in 32-gram sachets, intended to be consumed as a beverage after mixing with hot water. 2. Vanutide cridificar (INN List 100). 3002.20 3. Certain INN products. Chapters 28, 29 (See “INN List 101” at the end of this publication.) and 30 4. Certain INN products. Chapters 13, 29 (See “INN List 102” at the end of this publication.) and 30 5. Certain INN products. Chapters 28, 29, (See “INN List 103” at the end of this publication.) 30, 35 and 39 6. Re-classification of INN products.
    [Show full text]
  • Citrullinated Protein Antibody Paratope Drives Epitope Spreading and Polyreactivity in Rheumatoid Arthritis
    Arthritis & Rheumatology Vol. 0, No. 0, Month 2019, pp 1–11 DOI 10.1002/art.40760 © 2019, American College of Rheumatology Affinity Maturation of the Anti–Citrullinated Protein Antibody Paratope Drives Epitope Spreading and Polyreactivity in Rheumatoid Arthritis Sarah Kongpachith, Nithya Lingampalli, Chia-Hsin Ju, Lisa K. Blum, Daniel R. Lu, Serra E. Elliott, Rong Mao and William H. Robinson Objective. Anti–citrullinated protein antibodies (ACPAs) are a hallmark of rheumatoid arthritis (RA). While epitope spreading of the serum ACPA response is believed to contribute to RA pathogenesis, little is understood regarding how this phenomenon occurs. This study was undertaken to analyze the antibody repertoires of individuals with RA to gain insight into the mechanisms leading to epitope spreading of the serum ACPA response in RA. Methods. Plasmablasts from the blood of 6 RA patients were stained with citrullinated peptide tetramers to identify ACPA- producing B cells by flow cytometry. Plasmablasts were single-cell sorted and sequenced to obtain antibody repertoires. Sixty-nine antibodies were recombinantly expressed, and their anticitrulline reactivities were characterized using a cyclic citrullinated peptide enzyme- linked immuosorbent assay and synovial antigen arrays. Thirty- six mutated antibodies designed either to represent ancestral antibodies or to test paratope residues critical for binding, as determined from molecular modeling studies, were also tested for anticitrulline reactivities. Results. Clonally related monoclonal ACPAs and their shared ancestral antibodies each exhibited differential re- activity against citrullinated antigens. Molecular modeling identified residues within the complementarity-determining region loops and framework regions predicted to be important for citrullinated antigen binding. Affinity maturation re- sulted in mutations of these key residues, which conferred binding to different citrullinated epitopes and/or increased polyreactivity to citrullinated epitopes.
    [Show full text]
  • Table S1. Medications Searched in Institutional Database Query Immune Checkpoint Inhibitors Additional Immune Modulators Atezol
    BMJ Publishing Group Limited (BMJ) disclaims all liability and responsibility arising from any reliance Supplemental material placed on this supplemental material which has been supplied by the author(s) J Immunother Cancer Table S1. Medications Searched in Institutional Database Query Immune Checkpoint Inhibitors Additional Immune Modulators Atezolizumab Abatacept Avelumab Adalimumab Cemiplimab Anakinra Durvalumab Belatacept Ipilimumab Certolizumab Lirilumab Cyclophosphamide Mogamulizumab Etanercept Nivolumab Golimumab Pembrolizumab Immune globulin IV Tremelimumab Infliximab Urelumab Mycophenolate mofetil Varlilumab Rituximab Tocilizumab Vedolizumab Beattie J, et al. J Immunother Cancer 2021; 9:e001884. doi: 10.1136/jitc-2020-001884 BMJ Publishing Group Limited (BMJ) disclaims all liability and responsibility arising from any reliance Supplemental material placed on this supplemental material which has been supplied by the author(s) J Immunother Cancer Table S2. Toxicity of additional immune modulators Treatment detail Toxicity Patient 1 day 18,32 Infliximab dosed day 59 P.aeruginosa, S.marcescens pneumonia; died Patient 2 day 9 Infliximab dosed day 44 Febrile neutropenia; P. aeruginosa SBP and day 26 Mycophenolate initiated sepsis; C. albicans fungemia; treated and discharged Patient 8 day 4,11 Infliximab dosed day 14 Disseminated HSV-1; died Patient 26 day 79-128 Infliximab dosed (x7) day 130 E. faecalis, P. aeruginosa bacteremia; died; day 81, 97 Cyclophosphamide dosed Fungal pneumonia on autopsy Beattie J, et al. J Immunother Cancer 2021; 9:e001884. doi: 10.1136/jitc-2020-001884 BMJ Publishing Group Limited (BMJ) disclaims all liability and responsibility arising from any reliance Supplemental material placed on this supplemental material which has been supplied by the author(s) J Immunother Cancer Table S3.
    [Show full text]
  • 2017 Immuno-Oncology Medicines in Development
    2017 Immuno-Oncology Medicines in Development Adoptive Cell Therapies Drug Name Organization Indication Development Phase ACTR087 + rituximab Unum Therapeutics B-cell lymphoma Phase I (antibody-coupled T-cell receptor Cambridge, MA www.unumrx.com immunotherapy + rituximab) AFP TCR Adaptimmune liver Phase I (T-cell receptor cell therapy) Philadelphia, PA www.adaptimmune.com anti-BCMA CAR-T cell therapy Juno Therapeutics multiple myeloma Phase I Seattle, WA www.junotherapeutics.com Memorial Sloan Kettering New York, NY anti-CD19 "armored" CAR-T Juno Therapeutics recurrent/relapsed chronic Phase I cell therapy Seattle, WA lymphocytic leukemia (CLL) www.junotherapeutics.com Memorial Sloan Kettering New York, NY anti-CD19 CAR-T cell therapy Intrexon B-cell malignancies Phase I Germantown, MD www.dna.com ZIOPHARM Oncology www.ziopharm.com Boston, MA anti-CD19 CAR-T cell therapy Kite Pharma hematological malignancies Phase I (second generation) Santa Monica, CA www.kitepharma.com National Cancer Institute Bethesda, MD Medicines in Development: Immuno-Oncology 1 Adoptive Cell Therapies Drug Name Organization Indication Development Phase anti-CEA CAR-T therapy Sorrento Therapeutics liver metastases Phase I San Diego, CA www.sorrentotherapeutics.com TNK Therapeutics San Diego, CA anti-PSMA CAR-T cell therapy TNK Therapeutics cancer Phase I San Diego, CA www.sorrentotherapeutics.com Sorrento Therapeutics San Diego, CA ATA520 Atara Biotherapeutics multiple myeloma, Phase I (WT1-specific T lymphocyte South San Francisco, CA plasma cell leukemia www.atarabio.com
    [Show full text]
  • Looking for Therapeutic Antibodies in Next Generation Sequencing Repositories
    bioRxiv preprint doi: https://doi.org/10.1101/572958; this version posted March 10, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Title: Looking for Therapeutic Antibodies in Next Generation Sequencing Repositories. Authors: Konrad Krawczyk1*, Matthew Raybould2, Aleksandr Kovaltsuk2, Charlotte M. Deane2 1 NaturalAntibody, Hamburg, Germany 2 Oxford University Department of Statistics, Oxford, UK *Correspondence to [email protected] Abstract: Recently it has become possible to query the great diversity of natural antibody repertoires using Next Generation Sequencing (NGS). These methods are capable of producing millions of sequences in a single experiment. Here we compare Clinical Stage Therapeutic antibodies to the ~1b sequences from 60 independent sequencing studies in the Observed Antibody Space Database. Of the 242 post Phase I antibodies, we find 16 with sequence identity matches of 95% or better for both heavy and light chains. There are also 54 perfect matches to therapeutic CDR-H3 regions in the NGS outputs, suggesting a nontrivial amount of convergence between naturally observed sequences and those developed artificially. This has potential implications for both the discovery of antibody therapeutics and the legal protection of commercial antibodies. Introduction Antibodies are proteins in jawed vertebrates that recognize noxious molecules (antigens) for elimination. An organism expresses millions of diverse antibodies to increase the chances that some of them will be able to bind the foreign antigen, initiating the adaptive immune response.
    [Show full text]
  • (12) United States Patent (10) Patent No.: US 8,796,427 B2 Spee Et Al
    USOO8796427B2 (12) United States Patent (10) Patent No.: US 8,796,427 B2 Spee et al. (45) Date of Patent: Aug. 5, 2014 (54) HUMANIZED ANTI-HUMAN NKG2A EP 1036327 A2 9, 2000 MONOCLONAL ANTIBODY JP O3112485 A 5, 1991 JP O3112486 A 5, 1991 (75) Inventors: Petrus Johannes Louis Spee, Allerød E. 2025. A 3.28. (DK); Jianhe Chen, Beijing (CN); JP O3112484 U. 8, 2005 Soren Berg Padkjaer, Vaerlose (DK); WO 99.28748 A2 6, 1999 Jing Su, Beijing (CN); Jinchao Zhang, W 94.9. A2 258 Beijing (CN); Jiujiu Yu, Zhejiang (CN) WO O3,OO8449 A1 1, 2003 WO O3,O95965 A2 11/2003 (73) Assignee: Novo Nordisk A/S, Bagsvaerd (DK) WO 2004.?003.019 A2 1/2004 WO WO-2004/056312 T 2004 (*) Notice: Subject to any disclaimer, the term of this WO WO 2006/070286 12, 2004 patent is extended or adjusted under 35 W. 3:39:23, A. i58. U.S.C. 154(b) by 153 days. WO WO 2006O70286 A2 * T 2006 WO 2007042573 A2 4/2007 (21) Appl. No.: 12/811,990 WO WO 2007042573 A2 * 4, 2007 WO WO 2008/OO9545 1, 2008 (22) PCT Filed: Jan. 23, 2009 WO 2009/092805 A1 T 2009 (86). PCT No.: PCT/EP2009/050795 OTHER PUBLICATIONS S371 (c)(1), Petrie, E. J., et al. (2008), J. Exp. Med. 205: 725-735.* (2), (4) Date: Nov. 19, 2010 Bagot et al., “Functional Inhibitory Receptors Expressed by a Cuta neous T-Cell Lymphoma-Specific Cytolytic Clonal T-Cell Popula (87) PCT Pub. No.: WO2009/0928.05 tion.” Journal ofInvestigative Dermatology, 2000, vol.
    [Show full text]
  • Neural Message Passing for Joint Paratope-Epitope Prediction
    Neural message passing for joint paratope-epitope prediction Alice Del Vecchio 1 Andreea Deac 2 3 4 Pietro Lio` 1 Petar Velickoviˇ c´ 4 Abstract Hence, both the antibody and the antigen may be viewed as sequences of amino acid residues. Their binding site Antibodies are proteins in the immune system consists of two regions: the paratope on the antibody, and which bind to antigens to detect and neutralise the epitope on its corresponding antigen. Predicting them them. The binding sites in an antibody-antigen can therefore be posed as a binary classification problem: interaction are known as the paratope and epitope, for each amino acid residue in the antibody and antigen, respectively, and the prediction of these regions respectively, do they participate in the binding? is key to vaccine and synthetic antibody develop- ment. Contrary to prior art, we argue that paratope However, proteins can also be considered as graphs with its and epitope predictors require asymmetric treat- residues as nodes, with two nodes sharing an edge if their ment, and propose distinct neural message passing residues are spatially close. Recently, such contact graphs architectures that are geared towards the specific have been directly leveraged for protein function prediction aspects of paratope and epitope prediction, re- by Gligorijevic et al.(2020). spectively. We obtain significant improvements The advantage of considering a sequence based approach on both tasks, setting the new state-of-the-art and over a graph-based approach is that structural information recovering favourable qualitative predictions on is much harder to obtain. However, recent advancements antigens of relevance to COVID-19.
    [Show full text]
  • Antigen Mimicry-Recognizing Paratope
    Structural Evaluation of a Mimicry-Recognizing Paratope: Plasticity in Antigen−Antibody Interactions Manifests in Molecular Mimicry This information is current as of September 28, 2021. Suman Tapryal, Vineet Gaur, Kanwal J. Kaur and Dinakar M. Salunke J Immunol published online 3 June 2013 http://www.jimmunol.org/content/early/2013/06/01/jimmun ol.1203260 Downloaded from Why The JI? Submit online. http://www.jimmunol.org/ • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average by guest on September 28, 2021 Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2013 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published June 3, 2013, doi:10.4049/jimmunol.1203260 The Journal of Immunology Structural Evaluation of a Mimicry-Recognizing Paratope: Plasticity in Antigen–Antibody Interactions Manifests in Molecular Mimicry Suman Tapryal,*,1 Vineet Gaur,*,1 Kanwal J. Kaur,* and Dinakar M. Salunke*,† Molecular mimicry manifests antagonistically with respect to the specificity of immune recognition. However, it often occurs because different Ags share surface topologies in terms of shape or chemical nature.
    [Show full text]
  • Intratumoral Immunization: a New Paradigm for Cancer Therapy
    Clinical Cancer Review Research Intratumoral Immunization: A New Paradigm for Cancer Therapy Aurelien Marabelle1, Holbrook Kohrt2, Christophe Caux1, and Ronald Levy2 Abstract Immune cell infiltration in the tumor microenvironment is of prognostic and therapeutic import. These immune cell subsets can be heterogeneous and are composed of mature antigen-presenting cells, helper and effector cytotoxic T cells, toleragenic dendritic cells, tumor-associated macrophages, and regulatory T cells, among other cell types. With the development of novel drugs that target the immune system rather than the cancer cells, the tumor immune microenvironment is not only prognostic for overall patient outcome, but also predictive for likelihood of response to these immune-targeted therapies. Such therapies aim to reverse the cancer immunotolerance and trigger an effective antitumor immune response. Two major families of immunostimulatory drugs are currently in clinical development: pattern recognition receptor agonists (PRRago) and immunostimulatory monoclonal antibodies (ISmAb). Despite their immune-targeted design, these agents have so far been developed clinically as if they were typical anticancer drugs. Here, we review the limitations of this conventional approach, specifically addressing the shortcomings of the usual schedules of intravenous infusions every 2 or 3 weeks. If the new modalities of immunotherapy target specific immune cells within the tumor microenvironment, it might be preferable to deliver them locally into the tumor rather than systemically. There is preclinical and clinical evidence that a therapeutic systemic antitumor immune response can be generated upon intratumoral immunomodulation. Moreover, pre- clinical results have shown that therapeutic synergy can be obtained by combining PRRagos and ISmAbs to the local tumor site.
    [Show full text]
  • The Two Tontti Tudiul Lui Hi Ha Unit
    THETWO TONTTI USTUDIUL 20170267753A1 LUI HI HA UNIT ( 19) United States (12 ) Patent Application Publication (10 ) Pub. No. : US 2017 /0267753 A1 Ehrenpreis (43 ) Pub . Date : Sep . 21 , 2017 ( 54 ) COMBINATION THERAPY FOR (52 ) U .S . CI. CO - ADMINISTRATION OF MONOCLONAL CPC .. .. CO7K 16 / 241 ( 2013 .01 ) ; A61K 39 / 3955 ANTIBODIES ( 2013 .01 ) ; A61K 31 /4706 ( 2013 .01 ) ; A61K 31 / 165 ( 2013 .01 ) ; CO7K 2317 /21 (2013 . 01 ) ; (71 ) Applicant: Eli D Ehrenpreis , Skokie , IL (US ) CO7K 2317/ 24 ( 2013. 01 ) ; A61K 2039/ 505 ( 2013 .01 ) (72 ) Inventor : Eli D Ehrenpreis, Skokie , IL (US ) (57 ) ABSTRACT Disclosed are methods for enhancing the efficacy of mono (21 ) Appl. No. : 15 /605 ,212 clonal antibody therapy , which entails co - administering a therapeutic monoclonal antibody , or a functional fragment (22 ) Filed : May 25 , 2017 thereof, and an effective amount of colchicine or hydroxy chloroquine , or a combination thereof, to a patient in need Related U . S . Application Data thereof . Also disclosed are methods of prolonging or increasing the time a monoclonal antibody remains in the (63 ) Continuation - in - part of application No . 14 / 947 , 193 , circulation of a patient, which entails co - administering a filed on Nov. 20 , 2015 . therapeutic monoclonal antibody , or a functional fragment ( 60 ) Provisional application No . 62/ 082, 682 , filed on Nov . of the monoclonal antibody , and an effective amount of 21 , 2014 . colchicine or hydroxychloroquine , or a combination thereof, to a patient in need thereof, wherein the time themonoclonal antibody remains in the circulation ( e . g . , blood serum ) of the Publication Classification patient is increased relative to the same regimen of admin (51 ) Int .
    [Show full text]
  • Natural Killer Cell-Based Immunotherapy for Acute Myeloid Leukemia
    Xu and Niu J Hematol Oncol (2020) 13:167 https://doi.org/10.1186/s13045-020-00996-x REVIEW Open Access Natural killer cell-based immunotherapy for acute myeloid leukemia Jing Xu and Ting Niu* Abstract Despite considerable progress has been achieved in the treatment of acute myeloid leukemia over the past decades, relapse remains a major problem. Novel therapeutic options aimed at attaining minimal residual disease-negative complete remission are expected to reduce the incidence of relapse and prolong survival. Natural killer cell-based immunotherapy is put forward as an option to tackle the unmet clinical needs. There have been an increasing num- ber of therapeutic dimensions ranging from adoptive NK cell transfer, chimeric antigen receptor-modifed NK cells, antibodies, cytokines to immunomodulatory drugs. In this review, we will summarize diferent forms of NK cell-based immunotherapy for AML based on preclinical investigations and clinical trials. Keywords: Acute myeloid leukemia, Natural killer cells, Immunotherapy, Adoptive NK cell transfer, Chimeric antigen receptor-modifed NK cells, Antibodies, Cytokines Background cells and substances in the immune system play pivotal Acute myeloid leukemia (AML) is a clinically and geneti- roles in detecting and destroying pathogen-infected or cally heterogeneous disease with unsatisfactory out- neoplastically transformed cells. But they become less comes. Over the last few years, considerable progress has potent in cancer elimination when malignant cells dis- been achieved in the treatment of AML with the devel- play the loss of antigenicity and/or immunogenicity and opment and implementation of new drugs [1, 2]. How- are surrounded by an immunosuppressive microenvi- ever, allogeneic hematopoietic cell transplantation (HCT) ronment [6].
    [Show full text]