Immune Escape Mechanisms As a Guide for Cancer Immunotherapy Gregory L

Total Page:16

File Type:pdf, Size:1020Kb

Immune Escape Mechanisms As a Guide for Cancer Immunotherapy Gregory L Published OnlineFirst December 12, 2014; DOI: 10.1158/1078-0432.CCR-14-1860 Perspectives Clinical Cancer Research Immune Escape Mechanisms as a Guide for Cancer Immunotherapy Gregory L. Beatty and Whitney L. Gladney Abstract Immunotherapy has demonstrated impressive outcomes for exploited by cancer and present strategies for applying this some patients with cancer. However, selecting patients who are knowledge to improving the efficacy of cancer immunotherapy. most likely to respond to immunotherapy remains a clinical Clin Cancer Res; 21(4); 1–6. Ó2014 AACR. challenge. Here, we discuss immune escape mechanisms Introduction fore, promote tumor outgrowth. In this process termed "cancer immunoediting," cancer clones evolve to avoid immune-medi- The immune system is a critical regulator of tumor biology with ated elimination by leukocytes that have antitumor properties the capacity to support or inhibit tumor development, growth, (6). However, some tumors may also escape elimination by invasion, and metastasis. Strategies designed to harness the recruiting immunosuppressive leukocytes, which orchestrate a immune system are the focus of several recent promising thera- microenvironment that spoils the productivity of an antitumor peutic approaches for patients with cancer. For example, adoptive immune response (7). Thus, although the immune system can be T-cell therapy has produced impressive remissions in patients harnessed, in some cases, for its antitumor potential, clinically with advanced malignancies (1). In addition, therapeutic mono- relevant tumors appear to be marked by an immune system that clonal antibodies designed to disrupt inhibitory signals received actively selects for poorly immunogenic tumor clones and/or by T cells through the cytotoxic T-lymphocyte–associated antigen establishes a microenvironment that suppresses productive anti- 4 (CTLA-4; also known as CD152) and programmed cell death-1 tumor immunity (Fig. 1). (PD-1; also known as CD279) molecules are demonstrating long- Immune signatures for cancer immunotherapy are actively term survival benefits for some patients with metastatic melano- being explored across many clinical studies (8). However, unlike ma (2, 3). However, not all tumors appear to respond to these small-molecule inhibitors, for which the presence or absence of a immunomodulatory maneuvers. This observation emphasizes target mutation may predict response, the efficacy of immuno- the heterogeneity of cancer and suggests the existence of addi- therapy relies on the functional competence of multiple immu- tional immunoregulatory mechanisms in many patients. A major nologic elements. For example, strategies designed to harness the challenge for cancer immunotherapy, therefore, lies in under- antitumor potential of endogenous T cells rely on the proper standing these resistance mechanisms for selecting patients who execution of a series of steps that have been described in the cancer are most likely to benefit. immunity cycle (9). In this cycle, tumor cells must release immu- A central tenet of cancer immunotherapy is that the immune nogenic tumor antigens for the priming and activation of tumor- system actively surveys for malignant transformation and can be specific T cells. Tumor-reactive T cells must then infiltrate tumor induced to recognize and eliminate malignant cells. This premise tissue and recognize cancer cells in the context of a peptide–MHC was initially proposed by Thomas and Burnet in 1957 in the complex to induce cancer cell death. To evade immune-mediated immunosurveillance hypothesis, which postulated a role for the elimination, tumors must then develop strategies that disrupt this immune system in controlling the development and outgrowth of cycle. Therefore, predicting the clinical benefit of T-cell immuno- nascent transformed cells (4, 5). This hypothesis has since been therapy is likely to require an understanding of each of these steps refined based on knowledge that the immune system cannot only as it relates to a patient's individual tumor. Here, we discuss tumor protect against tumor development but can also select for tumors antigenicity, tumor immunogenicity and the tumor microenvi- with decreased antigenicity and/or immunogenicity and there- ronment as key elements of this cycle that may be used to predict clinical benefit with T-cell immunotherapy and guide the devel- opment of rational combinations. Abramson Cancer Center, Department of Medicine, Division of Hema- tology-Oncology, University of Pennsylvania Perelman School of Med- icine, Philadelphia, Pennsylvania. Antigenicity Corresponding Author: Gregory L. Beatty, University of Pennsylvania Perelman The ability of the immune system to distinguish between School of Medicine, Department of Medicine, Division of Hematology/Oncology normal and malignant cells is fundamental to cancer immuno- Smilow Center for Translational Research, Room 8-112, 3400 Civic Center therapy and relies, in part, on malignant cells retaining sufficient Boulevard Building, 421 Philadelphia, PA 19105-5156. Phone: 215-746-7764; Fax: antigenicity. Tumors can express a variety of nonmutated and 215-573-8590; E-mail: [email protected] mutated antigens that have the potential to elicit tumor-specific doi: 10.1158/1078-0432.CCR-14-1860 immune responses (10). However, to avoid immune-mediated Ó2014 American Association for Cancer Research. elimination, cancer cells may lose their antigenicity. Loss of www.aacrjournals.org OF1 Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2014 American Association for Cancer Research. Published OnlineFirst December 12, 2014; DOI: 10.1158/1078-0432.CCR-14-1860 Beatty and Gladney cells may inform the potential susceptibility of a cancer to Translational Relevance immune elimination by endogenous T cells. Immunotherapy is a promising treatment modality for Tumor antigens can be derived from viral proteins, proteins cancer. However, predicting which patients will benefit encoded by cancer-germline genes, differentiation antigens, and remains a challenge. Fundamental to the outgrowth of malig- proteins arising from somatic mutations or gene rearrangements nant cells is their ability to evade immune elimination, and (10). However, because malignant cells across patients and even therefore understanding the mechanisms exploited by a cancer within the same patient can be quite different, it remains unclear may help guide the application of immunotherapy. Here, we how to effectively quantify the antigenicity of a cancer. One propose that the tailoring of immunotherapy to patients may approach might be to determine the frequency of somatic muta- require knowledge of tumor antigenicity and immunogenicity tions within a cancer as a predictor of neoantigens. Until recently as well as the composition of the tumor microenvironment. In though, it was unclear whether mutated proteins that produce our discussion, we describe challenges and future directions in "neoantigens" could even serve as effective targets for cancer applying this knowledge to enhancing the efficacy of immu- immunotherapy. Recent studies have now demonstrated that T notherapy by improving patient selection and by informing cells recognizing neoantigens infiltrate tumor tissue and can the development of rational combination therapies. mediate strong tumor regressions after ex vivo expansion and adoptive transfer (11–13). These findings raise the possibility that tumor mutational frequency might correlate with respon- siveness to immunotherapy (14). antigenicity can arise due to the immune selection of cancer cells Mutational analysis of solid tumor malignancies has recently fi that lack or mutate immunogenic tumor antigens as well as revealed signi cant variability with some immunogenic cancers, through the acquisition of defects or deficiencies in antigen such as melanoma, displaying a high mutation rate and other presentation [e.g., loss of major histocompatibility (MHC) nonimmunogenic cancers, such as PDAC, showing a low muta- fi expression or dysregulation of antigen processing machinery; tion rate (15, 16). Although this nding has sometimes been used ref. 6]. As a result, knowledge of the antigenicity of malignant to explain the poor immunogenicity of cancers such as pancreatic Tumor (i) Loss of antigenicity (ii) Loss of immunogenicity (iii) Immunosuppressive microenvironment Immunosuppressive leukocytes (e.g., macrophages) Angenic tumor cell Antumor leukocytes (e.g., effector T cells) Poorly angenic tumor cell Immunosuppressive molecule (e.g., PD-L1) Figure 1. Immune escape mechanisms in cancer. Clinically apparent tumors must evolve mechanisms to evade immune elimination. During this process, nascent transformed cells may be selected for based on (i) a loss of antigenicity and/or (ii) a loss of immunogenicity. Loss of antigenicity may be achieved through the acquisition of defects in antigen processing and presentation or through the loss of immunogenic tumor antigens leading to a lack of immunogenic peptides presented in the context of a peptide–MHC complex. Although a loss of antigenicity is also associated with a loss of immunogenicity, malignant cells can gain additional immunosuppressive properties, such as expression of PD-L1 or secretion of suppressive cytokines (e.g., IL10 and TGFb), which further reduces their immunogenicity. (iii) Tumors may also escape immune elimination by orchestrating an immunosuppressive microenvironment. Malignant transformation induced by alterations in oncogenes and tumor-suppressor genes can
Recommended publications
  • Effects of Chemotherapy Agents on Circulating Leukocyte Populations: Potential Implications for the Success of CAR-T Cell Therapies
    cancers Review Effects of Chemotherapy Agents on Circulating Leukocyte Populations: Potential Implications for the Success of CAR-T Cell Therapies Nga T. H. Truong 1, Tessa Gargett 1,2,3, Michael P. Brown 1,2,3,† and Lisa M. Ebert 1,2,3,*,† 1 Translational Oncology Laboratory, Centre for Cancer Biology, University of South Australia and SA Pathology, North Terrace, Adelaide, SA 5000, Australia; [email protected] (N.T.H.T.); [email protected] (T.G.); [email protected] (M.P.B.) 2 Cancer Clinical Trials Unit, Royal Adelaide Hospital, Port Rd, Adelaide, SA 5000, Australia 3 Adelaide Medical School, University of Adelaide, North Terrace, Adelaide, SA 5000, Australia * Correspondence: [email protected] † These authors contributed equally. Simple Summary: CAR-T cell therapy is a new approach to cancer treatment that is based on manipulating a patient’s own T cells such that they become able to seek and destroy cancer cells in a highly specific manner. This approach is showing remarkable efficacy in treating some types of blood cancers but so far has been much less effective against solid cancers. Here, we review the diverse effects of chemotherapy agents on circulating leukocyte populations and find that, despite some negative effects over the short term, chemotherapy can favourably modulate the immune systems of cancer patients over the longer term. Since blood is the starting material for CAR-T cell Citation: Truong, N.T.H.; Gargett, T.; production, we propose that these effects could significantly influence the success of manufacturing, Brown, M.P.; Ebert, L.M.
    [Show full text]
  • Guideline on Immunogenicity Assessment of Biotechnology-Derived Therapeutic Proteins
    European Medicines Agency London, 13 December 2007 Doc. Ref. EMEA/CHMP/BMWP/14327/2006 COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) GUIDELINE ON IMMUNOGENICITY ASSESSMENT OF BIOTECHNOLOGY-DERIVED THERAPEUTIC PROTEINS DRAFT AGREED BY BMWP July 2006 ADOPTION BY CHMP FOR RELEASE FOR CONSULTATION January 2007 END OF CONSULTATION (DEADLINE FOR COMMENTS) July 2007 AGREED BY BMWP October 2007 ADOPTION BY CHMP December 2007 DATE FOR COMING INTO EFFECT April 2008 KEYWORDS Immunogenicity, unwanted immune response, biotechnology derived proteins, immunogenicity risk factors, assays, clinical efficacy and safety, risk management 7 Westferry Circus, Canary Wharf, London, E14 4HB, UK Tel. (44-20) 74 18 84 00 Fax (44-20) 74 18 86 13 E-mail: [email protected] http://www.emea.europa.eu ©EMEA 2007 Reproduction and/or distribution of this document is authorised for non commercial purposes only provided the EMEA is acknowledged GUIDELINE ON IMMUNOGENICITY ASSESSMENT OF BIOTECHNOLOGY-DERIVED THERAPEUTIC PROTEINS TABLE OF CONTENTS EXECUTIVE SUMMARY................................................................................................................... 3 1. INTRODUCTION......................................................................................................................... 3 2. SCOPE............................................................................................................................................ 4 3. LEGAL BASIS .............................................................................................................................
    [Show full text]
  • Risk Assessment and Mitigation Strategies for Immune Responses to Therapeutic Proteins: the FDA Perspective
    Risk Assessment and Mitigation Strategies for Immune Responses to Therapeutic Proteins: the FDA Perspective Amy S. Rosenberg, M.D. Supervisory Medical Officer, Office of Biotechnology Products CDER, FDA Risk is a Specific Knowledge Set Encompassing Both Consequences and Probabilities (modified from Stirling and Gee 2002) Knowledge about Consequences Knowledge about likelihoods Consequences Consequences well-defined poorly-defined Some basis Ambiguity Risk (I) for (III) probabilities Incertitude No basis for Uncertainty Ignorance probabilities (II) (IV) “Guidance for Industry Immunogenicity Assessment for Therapeutic Protein Products” U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluation and Research (CBER) August 2014 Clinical/Medical 3 Immunogenicity Risk Assessment: Consequences for Safety • Fatality/Severe Morbidity – Anaphylaxis: clinical definition, does not imply mechanism • Proteins of non-human origin, eg, aprotinin, asparaginase • Replacement human proteins in knock out phenotype: eg, Factor IX in hemophilia B – Cross reactive neutralization of endogenous factor or receptor homolog with unique function resulting in deficiency syndrome or cytokine release syndrome – Immune complex disease and delayed hypersensitivity • Serum sickness; nephropathy • Most often seen when high doses of therapeutic proteins are administered in setting of a sustained high titered antibody response Immunogenicity Risk Assessment Consequences for Efficacy
    [Show full text]
  • Measuring the Immunogenicity of Allogeneic Adult Mesenchymal Stem Cells Alix K
    Berglund et al. Stem Cell Research & Therapy (2017) 8:288 DOI 10.1186/s13287-017-0742-8 REVIEW Open Access Immunoprivileged no more: measuring the immunogenicity of allogeneic adult mesenchymal stem cells Alix K. Berglund1*, Lisa A. Fortier2, Douglas F. Antczak3 and Lauren V. Schnabel1* Abstract Background: Autologous and allogeneic adult mesenchymal stem/stromal cells (MSCs) are increasingly being investigated for treating a wide range of clinical diseases. Allogeneic MSCs are especially attractive due to their potential to provide immediate care at the time of tissue injury or disease diagnosis. The prevailing dogma has been that allogeneic MSCs are immune privileged, but there have been very few studies that control for matched or mismatched major histocompatibility complex (MHC) molecule expression and that examine immunogenicity in vivo. Studies that control for MHC expression have reported both cell-mediated and humoral immune responses to MHC-mismatched MSCs. The clinical implications of immune responses to MHC-mismatched MSCs are still unknown. Pre-clinical and clinical studies that document the MHC haplotype of donors and recipients and measure immune responses following MSC treatment are necessary to answer this critical question. Conclusions: This review details what is currently known about the immunogenicity of allogeneic MSCs and suggests contemporary assays that could be utilized in future studies to appropriately identify and measure immune responses to MHC-mismatched MSCs. Keywords: Mesenchymal stem cell, Allogeneic, Immunogenicity, Major histocompatibility complex, Mixed leukocyte reaction, Cytotoxicity, ELISPOT, Microcytotoxicity Background in vivo [9], questioning the relevance of differentiation Mesenchymal stem cells (MSCs) are currently defined as to the therapeutic properties of MSCs when injected in a plastic-adherent cells with a fibroblast-like morphology naive state.
    [Show full text]
  • Size, Affinity, and Immunogenicity Peptide-Binding Repertoires Of
    HLA Class I Alleles Are Associated with Peptide-Binding Repertoires of Different Size, Affinity, and Immunogenicity This information is current as Sinu Paul, Daniela Weiskopf, Michael A. Angelo, John of September 29, 2021. Sidney, Bjoern Peters and Alessandro Sette J Immunol 2013; 191:5831-5839; Prepublished online 4 November 2013; doi: 10.4049/jimmunol.1302101 http://www.jimmunol.org/content/191/12/5831 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2013/11/05/jimmunol.130210 Material 1.DC1 http://www.jimmunol.org/ References This article cites 51 articles, 12 of which you can access for free at: http://www.jimmunol.org/content/191/12/5831.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision by guest on September 29, 2021 • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average 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. The Journal of Immunology HLA Class I Alleles Are Associated with Peptide-Binding Repertoires of Different Size, Affinity, and Immunogenicity Sinu Paul,1 Daniela Weiskopf,1 Michael A.
    [Show full text]
  • Monoclonal Antibody Therapeutics and Apoptosis
    Oncogene (2003) 22, 9097–9106 & 2003 Nature Publishing Group All rights reserved 0950-9232/03 $25.00 www.nature.com/onc Monoclonal antibody therapeutics and apoptosis Dale L Ludwig*,1, Daniel S Pereira1, Zhenping Zhu1, Daniel J Hicklin1 and Peter Bohlen1 1ImClone Systems Incorporated, 180 Varick Street, New York, NY 10014, USA The potential for disease-specific targeting and low including the generation of human antibody phage toxicity profiles have made monoclonal antibodies attrac- display libraries, human immunoglobulin-producing tive therapeutic drug candidates. Antibody-mediated transgenic mice, and directed affinity maturation meth- target cell killing is frequently associated with immune odologies, have further improved on the efficiency, effector mechanisms such as antibody-directed cellular specificity, and reactivity of monoclonal antibodies for cytotoxicity, but they can also be induced by apoptotic their target antigens (Schier et al., 1996; Mendez et al., processes. Antibody-directed mechanisms, including anti- 1997; de Haard et al., 1999; Hoogenboom and Chames, gen crosslinking, activation of death receptors, and 2000; Knappik et al., 2000). As a result, the isolation of blockade of ligand-receptor growth or survival pathways, high-affinity fully human monoclonal antibodies is now can elicit the induction of apoptosis in targeted cells. commonplace. Owing to their inherent specificity for a Depending on their mechanism of action, monoclonal particular target antigen, monoclonal antibodies pro- antibodies can induce targeted cell-specific killing alone or mise precise selectivity for target cells, avoiding non- can enhance target cell susceptibility to chemo- or reacting normal cells. radiotherapeutics by effecting the modulation of anti- To be effective as therapeutics for cancer, antibodies apoptotic pathways.
    [Show full text]
  • Immunogenicity Assessment of Therapeutic Proteins
    European Medicines Agency London, 24 January 2007 Doc. Ref. EMEA/CHMP/BMWP/14327/2006 COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) DRAFT GUIDELINE ON IMMUNOGENICITY ASSESSMENT OF BIOTECHNOLOGY-DERIVED THERAPEUTIC PROTEINS DRAFT AGREED BY BMWP July 2006 ADOPTION BY CHMP FOR RELEASE FOR CONSULTATION 24 January 2007 END OF CONSULTATION (DEADLINE FOR COMMENTS) 31 July 2007 Comments should be provided electronically in word format using this template to: [email protected] KEYWORDS Immunogenicity, biotechnology derived proteins, antigenicity risk factors, assays, clinical efficacy 7 Westferry Circus, Canary Wharf, London, E14 4HB, UK Tel. (44-20) 74 18 84 00 Fax (44-20) 74 18 86 13 E-mail: [email protected] http://www.emea.europa.eu ©EMEA 2007 Reproduction and/or distribution of this document is authorised for non commercial purposes only provided the EMEA is acknowledged GUIDELINE ON IMMUNOGENICITY ASSESSMENT OF BIOTECHNOLOGY-DERIVED THERAPEUTIC PROTEINS TABLE OF CONTENTS EXECUTIVE SUMMARY................................................................................................................... 3 1. INTRODUCTION......................................................................................................................... 3 2. SCOPE............................................................................................................................................ 3 3. LEGAL BASIS .............................................................................................................................
    [Show full text]
  • SEREX Analysis for Tumor Antigen Identification in a Mouse Model Of
    © 2000 Nature America, Inc. 0929-1903/00/$15.00/ϩ0 www.nature.com/cgt SEREX analysis for tumor antigen identification in a mouse model of adenocarcinoma Tracy A. Hampton,1–3 Robert M. Conry,2 M. B. Khazaeli,2 Denise R. Shaw,2 David T. Curiel,1,2 Albert F. LoBuglio,2 and Theresa V. Strong1–3 1Gene Therapy Center, 2Division of Hematology/Oncology, Department of Medicine, and 3Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama 35294. Evaluation of immunotherapy strategies in mouse models of carcinoma is hampered by the limited number of known murine tumor antigens (Ags). Although tumor Ags can be identified based on cytotoxic T-cell activation, this approach is not readily accomplished for many tumor types. We applied an alternative strategy based on a humoral immune response, SEREX, to the identification of tumor Ags in the murine colon adenocarcinoma cell line MC38. Immunization of syngeneic C57BL/6 mice with MC38 cells by three different methods induced a protective immune response with concomitant production of anti-MC38 antibodies. Immunoscreening of an MC38-derived expression library resulted in the identification of the endogenous ecotropic leukemia virus envelope (env) protein and the murine ATRX protein as candidate tumor Ags. Northern blot analysis demonstrated high levels of expression of the env transcript in MC38 cells and in several other murine tumor cell lines, whereas expression in normal colonic epithelium was absent. ATRX was found to be variably expressed in tumor cell lines and in normal tissue. Further analysis of the expressed env sequence indicated that it represents a nonmutated tumor Ag.
    [Show full text]
  • Tumor Immunology 101 for the Non-Immunologist
    Tumor Immunology 101 For the Non-Immunologist Louis M. Weiner, MD Director, Lombardi Comprehensive Cancer Center Francis L. and Charlotte G. Gragnani Chair Professor and Chairman, Department of Oncology Georgetown University Medical Center Disclosure Consulting Fees Ownership Interest Abbvie Pharmaceuticals Celldex Novartis Jounce Merck Merrimack Genetech Symphogen Cytomax Immunovative Therapies, Ltd. Contracted Research Symphogen We Have Been at War Against Cancer Throughout Human History Tumor President Nixon declares a “War on Cancer” in 1971 Medieval Saxon man with a large tumor of the left femur Which Target? Hanahan, Weinberg, Cell 2000 The “War on Cancer” is fought one person at a time… • Primary Combatants: – Malignant cell population – Host immune system • The host immune system is the dominant active enemy faced by a developing cancer • All “successful” cancers must solve the challenges of overcoming defenses erected by host immune systems Which System? Hanahan, Weinberg, Cell 2011 The Case for Cancer Immunotherapy • Surprisingly few new truly curative anti- cancer cytotoxic drugs or targeted therapies in 20+ years – Tumor heterogeneity – Too many escape routes? • The immune response is designed to identify and disable “escape routes ” that cancers employ • Immunotherapy can cure cancers Immunotherapy • Treatment of disease by inducing, enhancing, or suppressing an immune response • “Treating the immune system so it can treat the cancer” (J. Wolchok) Some Examples of Successful Cancer Immunotherapy • Type 1 interferons – bladder
    [Show full text]
  • Vaccine Immunology Claire-Anne Siegrist
    2 Vaccine Immunology Claire-Anne Siegrist To generate vaccine-mediated protection is a complex chal- non–antigen-specifc responses possibly leading to allergy, lenge. Currently available vaccines have largely been devel- autoimmunity, or even premature death—are being raised. oped empirically, with little or no understanding of how they Certain “off-targets effects” of vaccines have also been recog- activate the immune system. Their early protective effcacy is nized and call for studies to quantify their impact and identify primarily conferred by the induction of antigen-specifc anti- the mechanisms at play. The objective of this chapter is to bodies (Box 2.1). However, there is more to antibody- extract from the complex and rapidly evolving feld of immu- mediated protection than the peak of vaccine-induced nology the main concepts that are useful to better address antibody titers. The quality of such antibodies (e.g., their these important questions. avidity, specifcity, or neutralizing capacity) has been identi- fed as a determining factor in effcacy. Long-term protection HOW DO VACCINES MEDIATE PROTECTION? requires the persistence of vaccine antibodies above protective thresholds and/or the maintenance of immune memory cells Vaccines protect by inducing effector mechanisms (cells or capable of rapid and effective reactivation with subsequent molecules) capable of rapidly controlling replicating patho- microbial exposure. The determinants of immune memory gens or inactivating their toxic components. Vaccine-induced induction, as well as the relative contribution of persisting immune effectors (Table 2.1) are essentially antibodies— antibodies and of immune memory to protection against spe- produced by B lymphocytes—capable of binding specifcally cifc diseases, are essential parameters of long-term vaccine to a toxin or a pathogen.2 Other potential effectors are cyto- effcacy.
    [Show full text]
  • Monoclonal Antibodies in Cancer Therapy
    Clin Oncol Cancer Res (2011) 8: 215–219 215 DOI 10.1007/s11805-011-0583-7 Monoclonal Antibodies in Cancer Therapy Yu-Ting GUO1,2 ABSTRACT Monoclonal antibodies (MAbs) are a relatively new Qin-Yu HOU1,2 innovation in cancer treatment. At present, some monoclonal antibodies have increased the efficacy of the treatment of certain 1,2 Nan WANG tumors with acceptable safety profiles. When monoclonal antibodies enter the body and attach to cancer cells, they function in several different ways: first, they can trigger the immune system to attack and kill that cancer cell; second, they can block the growth signals; third, they can prevent the formation of new blood vessels. Some naked 1 Key Laboratory of Industrial Fermentation MAbs such as rituximab can be directed to attach to the surface of Microbiology, Ministry of Education, Tianjin, cancer cells and make them easier for the immune system to find and 300457, China. destroy. The ability to produce antibodies with limited immunogeni- 2 Tianjin Key Laboratory of Industrial Micro- city has led to the production and testing of a host of agents, several biology, College of Biotechnology, Tianjin of which have demonstrated clinically important antitumor activity University of Science and Technology, Tianjin, 300457, China. and have received U.S. Food & Drug Administration (FDA) approval as cancer treatments. To reduce the immunogenicity of murine anti- bodies, murine molecules are engineered to remove the immuno- genic content and to increase their immunologic efficiency. Radiolabeled antibodies composed of antibodies conjugated to radionuclides show efficacy in non-Hodgkin’s lymphoma. Anti- vascular endothelial growth factor (VEGF) antibodies such as bevacizumab intercept the VEGF signal of tumors, thereby stopping them from connecting with their targets and blocking tumor growth.
    [Show full text]
  • The Evaluation of Immunogenicity of Therapeutic Antibody Drug Aggregates Using 2D and 3D in Vitro Models
    Dissertation zur Erlangung des Doktorgrades der Fakultät Chemie und Pharmazie der Ludwig-Maximilians-Universität München The evaluation of immunogenicity of therapeutic antibody drug aggregates using 2D and 3D in vitro models Teresa Franziska Kraus aus Weingarten 2019 Erklärung Diese Dissertation wurde im Sinne von § 7 der Promotionsordnung vom 28. November 2011 von Frau PD Dr. habil. Julia Engert betreut. Eidesstattliche Versicherung Diese Dissertation wurde eigenständig und ohne unerlaubte Hilfe erarbeitet. München, den 01.05.2019 ____________________________ Teresa Kraus Dissertation eingereicht am: 16.05.2019 1. Gutachter: PD Dr. habil. Julia Engert 2. Gutachter: Prof. Dr. Gerhard Winter Mündliche Prüfung am: 02.07.2019 Für Opa SALUS AEGROTI SUPREMA LEX Hippokrates Acknowledgements This thesis was prepared between March 2015 and December 2018 at the Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics at the Ludwig-Maximilians-Universität München under the supervision of PD Dr. habil. Julia Engert. First and foremost, I would like to express my deepest gratitude to PD Dr. habil. Julia Engert for her excellent supervision and scientific guidance during my PhD time. I’m deeply thankful for her support and help in all research issues, the lively scientific discussions and for always having an open ear. In addition, I highly appreciated her help in preparing publications. I’m deeply grateful to Prof. Dr. Gerhard Winter for giving me the chance to work on an innovative and challenging project. I highly appreciated his outstanding scientific advice and support on a professional but also personal level. Besides the professional aims, he also attached importance to a good team spirit and always supported group activities which created an excellent working atmosphere.
    [Show full text]