Chapter 2. The development of xenograft models for the in vivo study. of human T cells.

Similar documents
Animal Models of Normal and Leukemic Human Hematopoiesis

Animal Models to Understand Immunity

Transfer protocol of human HSC into NOG mice

Manipulation of T Cells in the Thnsplant Inoculum

Cover Page. The handle holds various files of this Leiden University dissertation.

Humanized Mouse Models for Vaccine Development Michael A. Brehm. Why Do We Need Humanized Mouse Models?

Immunology Lecture 4. Clinical Relevance of the Immune System

T cell manipulation of the graft: Yes

SEVENTH EDITION CHAPTER

Goals. Goals. From Bench to Cageside: Immunology of Humanizing Mice

From Bench to Cageside: Immunology of Humanizing Mice. Zachary T Freeman

Shiv Pillai Ragon Institute, Massachusetts General Hospital Harvard Medical School

Dr. Yi-chi M. Kong August 8, 2001 Benjamini. Ch. 19, Pgs Page 1 of 10 TRANSPLANTATION

Immune system. Aims. Immune system. Lymphatic organs. Inflammation. Natural immune system. Adaptive immune system

Chapter 5 quantitative assessment of human T lymphocytes in RAG2 -/- γc -/- mice: the impact of ex vivo

Transplantation. Immunology Unit College of Medicine King Saud University

T Cell Development. Xuefang Cao, MD, PhD. November 3, 2015

The Lymphatic System and Body Defenses

Medical Virology Immunology. Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University

The Immune System. A macrophage. ! Functions of the Immune System. ! Types of Immune Responses. ! Organization of the Immune System

One Day BMT Course by Thai Society of Hematology. Management of Graft Failure and Relapsed Diseases

CHAPTER 3 LABORATORY PROCEDURES

Tumor Immunology. Wirsma Arif Harahap Surgical Oncology Consultant

Immune Reconstitution Following Hematopoietic Cell Transplant

Mon, Wed, Fri 11:00 AM-12:00 PM. Owen, Judy, Jenni Punt, and Sharon Stranford Kuby-Immunology, 7th. Edition. W.H. Freeman and Co., New York.

Overview of the Lymphoid System

Immunology. Anas Abu-Humaidan M.D. Ph.D. Transplant immunology+ Secondary immune deficiency

T cell development October 28, Dan Stetson

COURSE: Medical Microbiology, PAMB 650/720 - Fall 2008 Lecture 16

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION

The Thymus as The Primary Site of T-cell Production

Achievement of Cellular Immunity and Discordant Xenogeneic Tolerance in Mice by Porcine Thymus Grafts

Immunity. Acquired immunity differs from innate immunity in specificity & memory from 1 st exposure

Immunopathology. 2-Patterned hemodynamic responses, cell surface associated and soluble mediator systems (e.g., complement and coagulation systems).

IN UTERO HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CANINES: THE GESTATIONAL WINDOW OF OPPORTUNITY TO MAXIMIZE ENGRAFTMENT

Chapter 3. A new xenograft model for graft-versus-host disease by. intravenous transfer of human peripheral blood mononuclear cells

Transplant Booklet D Page 1

Principles of Adaptive Immunity

Title: NATURAL KILLER CELL FUNCTIONS AND SURFACE RECEPTORS

HHS Public Access Author manuscript Bone Marrow Transplant. Author manuscript; available in PMC 2015 July 01.

Preface and Acknowledgments Preface and Acknowledgments to the Third Edition Preface to the Second Edition Preface to the First Edition

Micr-6005, Current Concepts of Immunology (Rutgers course number: 16:681:543) Spring 2009 Semester

ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS. Choompone Sakonwasun, MD (Hons), FRCPT

1. Overview of Adaptive Immunity

Haematopoietic stem cells

Development of B and T lymphocytes

Chapter 1. Chapter 1 Concepts. MCMP422 Immunology and Biologics Immunology is important personally and professionally!

The Immune System: Innate and Adaptive Body Defenses Outline PART 1: INNATE DEFENSES 21.1 Surface barriers act as the first line of defense to keep

FIT Board Review Corner March 2016

Mucosal Immune System

Overview B cell development T cell development

D Bonnet 1,2, M Bhatia 1,3, JCY Wang 1, U Kapp 1 and JE Dick 1. Summary:

Adaptive immune responses: T cell-mediated immunity

TRANSPLANT IMMUNOLOGY. Shiv Pillai Ragon Institute of MGH, MIT and Harvard

Natural Killer Cells: Development, Diversity, and Applications to Human Disease Dr. Michael A. Caligiuri

Leukine. Leukine (sargramostim) Description

The Immune System. Innate. Adaptive. - skin, mucosal barriers - complement - neutrophils, NK cells, mast cells, basophils, eosinophils

Modulating STAT Signaling to Promote Engraftment of Allogeneic Bone Marrow Transplant

Third line of Defense

TITLE: Assessing the Mechanisms of MDS and its Transformation to Leukemia in a Novel Humanized Mouse. REPORT DATE: September 2014

The development of T cells in the thymus

5/1/13. The proportion of thymus that produces T cells decreases with age. The cellular organization of the thymus

Corporate Medical Policy

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

Campbell's Biology: Concepts and Connections, 7e (Reece et al.) Chapter 24 The Immune System Multiple-Choice Questions

Nonspecific External Barriers skin, mucous membranes

Tolerance, autoimmunity and the pathogenesis of immunemediated inflammatory diseases. Abul K. Abbas UCSF

MHC class I MHC class II Structure of MHC antigens:

CD34+ Cells: A Comparison of Stem and Progenitor Cells in Cord Blood, Peripheral Blood, and the Bone Marrow

S. No Topic Class No Date

Scientific report: Delineating cellular stages and regulation of human NK cell development to improve NK cell-based therapy for cancer (Dnr )

immunity produced by an encounter with an antigen; provides immunologic memory. active immunity clumping of (foreign) cells; induced by crosslinking

Immune Regulation and Tolerance

Allergy and Immunology Review Corner: Chapter 19 of Immunology IV: Clinical Applications in Health and Disease, by Joseph A. Bellanti, MD.

Hematopoiesis. Hematopoiesis. Hematopoiesis

Understanding the role of ex vivo T cell depletion

General information. Cell mediated immunity. 455 LSA, Tuesday 11 to noon. Anytime after class.

T Lymphocyte Activation and Costimulation. FOCiS. Lecture outline

DEVELOPMENT OF CELLULAR IMMUNOLOGY

PIDPID GLOSSARYID GLOSSARY

TCR, MHC and coreceptors

Defensive mechanisms include :

BMTCN Review Course Basic Concepts and Indications for Transplantation How the Experts Treat Hematologic Malignancies Las Vegas, NV, March 10, 2016

Leukine. Leukine (sargramostim) Description

Dr.PSRK.Sastry MD, ECMO

Chapter 35 Active Reading Guide The Immune System

LYMPHOCYTES & IMMUNOGLOBULINS. Dr Mere Kende, Lecturer SMHS

Basic Immunology Final Exam

Third line of Defense. Topic 8 Specific Immunity (adaptive) (18) 3 rd Line = Prophylaxis via Immunization!

Immunology sheet. Done by: rasha rakan. Lecture: Introduction to medical immunology

Blood and Immune system Acquired Immunity

immunity defenses invertebrates vertebrates chapter 48 Animal defenses --

Immune-deficient mouse models for analysis of human stem cells

Chapter 22: The Lymphatic System and Immunity

What s a Transplant? What s not?

Chapter 24 The Immune System

CHAPTER-VII IMMUNOLOGY R.KAVITHA, M.PHARM, LECTURER, DEPARTMENT OF PHARMACEUTICS, SRM COLLEGE OF PHARMACY, SRM UNIVERSITY, KATTANKULATHUR.

CONTENTS. About the Authors, xv Contributors, xvi Preface and Acknowiedgments, xvii How to Use Your Textbook, xix About the Companion Website, xxiii

Tolerance 2. Regulatory T cells; why tolerance fails. Abul K. Abbas UCSF. FOCiS

Transcription:

Chapter 2 the development of xenograft models for the in vivo study of human T cells Chapter 2 The development of xenograft models for the in vivo study of human T cells. 33 Proefschrift Roos van Rijn.indb 33 19-10-2006 16:47:37

Preclinical evaluation of human T lymphocytes in RAG2 -/- γc -/- mice Rozemarijn S. van Rijn Human T cells can engraft in immunodeficient mice. The historical development of xenograft models for the study of human T cells is shown in Table 1 and amplified in the following paragraphs. The overview reflects the research groups that were successful in developing original models. In 1988, Mosier et al. transplanted human PBMCs for the first time into mice with severe combined immunodeficiency (SCID). 1 This resulted in the maintenance of mature T and B cells and high levels of immunoglobulins in the peripheral blood. The model was followed by several groups, but there were some setbacks. 2-8 Although intraperitoneal transfer was successful, intravenous transfer of up to 10 8 hupbmcs was ineffective. Other problems were a highly variable engraftment with very low maximal levels of human lymphoid cells in the peripheral blood (0.1-2%) and the development of human Epstein-Barr virus (EBV)-induced B-cell lymphomas over an extended period after human cell transfer when EBV-positive donor cells were used. Also, surprisingly little X-GVHD was observed in the hupbmc-scid chimeras, raising questions towards the alloreactivity of the human T cells in the murine environment. Of 800 hupbmc-scid mice reconstituted with 20-50x10 6 hupbmcs observed, only 8 mice injected with hupbmcs from the same donor displayed clinical symptoms consistent with a mild transient X-GVHD. 9 In vitro studies of chimera-derived long-term surviving human T cells suggested anergy of the T cells, as they were refractory to stimulation with anti-cd3 antibody, but proliferated in response to exogenous Il-2. 10 However, later on it was shown that failure to develop X-GVHD was a direct consequence of a low degree of T-cell chimerism (<10% human T cells) in most (80%) hupbmc-scid mice. 11 Although high numbers of engrafted human T cells can usually be found in the peritoneum or spleen of the mice, only the number of human T cells in the peripheral blood has been found to directly correlate with the development of X-GVHD. Low T-cell chimerism depends on the dose of hupbmcs and the conditioning regimen, but the main reason is a host-versus-graft reaction by the residual innate immune system of the SCID mice: natural killer (NK) cells, granulocytes and macrophages. 12-15 34 Proefschrift Roos van Rijn.indb 34 19-10-2006 16:47:37

Chapter 2 the development of xenograft models for the in vivo study of human T cells 35 Proefschrift Roos van Rijn.indb 35 19-10-2006 16:47:41

Preclinical evaluation of human T lymphocytes in RAG2 -/- γc -/- mice Rozemarijn S. van Rijn Several groups addressed this problem to obtain a better engraftment of human T cells. This was done in two general ways: 1. treatment of the SCID mice with antibodies that mediate cell kill of different parts of the innate immune system. 2. development of different strains of knockout mice with increased immunodeficiency, or a combination of 1 and 2. The models that were developed over the years are described in the following paragraphs. Unfortunately, not all groups have measured human T cell chimerism in the peripheral blood. For an optimal comparison, the site at which engraftment levels were measured, is specified as peripheral blood, spleen, bone marrow or peritoneum in Table 1. 1. Antibodies for attack of the innate immune system 1.1 Anti-NK-cell antibodies In 1983, Lotzova et al found in mice that NK cells formed a barrier for the engraftment of allogeneic bone marrow transplants and that rejection of the graft could be prevented by the administration of NK1.1 antiserum. 16 This was confirmed by other groups. 17,18 Anti-NK-cell antibodies were then also used to facilitate engraftment of hupbmcs. 19-21 Besides NK1.1 antiserum, other antibodies like TM-β1 or anti-asialo GM1 were used for in vivo NK cell depletion. 19,20,22-24 Similar results were obtained by diminishing NK-cell-derived interferon-γ with administration of antimurine interferon-γ monoclonal antibody. 14 1.2 Irradiation Another way to reduce host resistance was sublethal irradiation (100-400 cgy) 25 or a combination of irradiation with anti-asialo GM1. 26 Unfortunately, SCID mice are extremely radio-sensitive due to their inherent DNA repair defect and typically develop murine thymomas a few months after irradiation, which can be a problem for long-term observations. 27 1.3 Granulocyte or macrophage depletion Granulocyte depletion by treatment of the mice with anti-murine granulocyte monoclonal antibody and in vivo macrophage depletion by administration of clodronate both resulted in increased engraftment of human cells. 12,14,28 1.4 In vitro activation of hupbmcs Additional studies have demonstrated that activation of the freshly isolated hupbmcs before transfer facilitates engraftment and trafficking in the SCID host. 25,29 36 Proefschrift Roos van Rijn.indb 36 19-10-2006 16:47:41

Chapter 2 the development of xenograft models for the in vivo study of human T cells Activation of lymphocytes may induce adhesion molecules on the lymphocyte cell surface and permit trafficking of the cells to the murine lymphoid organs. However, little is known about the role of these homing receptors. Activation may also increase cytokine responsiveness of the transferred lymphocytes and promote their proliferation and engraftment. Activation regimens that have been tried include treatment with anti-cd3 antibody 25,29, chemokines 30, superantigens 31 and the administration of human growth hormone. 25,27 All these methods resulted in increased human cell engraftment, function or trafficking to the peripheral lymphoid organs of the SCID mice. Interestingly, activation with IL2 in vitro or in vivo has been found to inhibit human cell engraftment in these mice, which is suggested to be caused by the effect of IL2 on host effector cells resulting in an increased host resistance. 25,29 2. Development of knockout mice with increased immunodeficiency 2.1 Normal strains of mice One strategy is the engraftment of human cells in highly irradiated normal strains of mice. Lubin et al. successfully engrafted hupbmcs into Balb/c mice that received split total body irradiation of 4 Gy followed by 10 Gy 3 days later, before ip transfer of the human cells. The mice were radioprotected for the high-dose irradiation with iv infused SCID bone marrow. Sporadically X-GVHD was observed with maximal engraftment levels of up to 32% in peripheral blood. 32 2.2 Newborn mice Pflumio et al. transferred hupbmcs ip into newborn SCID mice, which still lack NK cell activity in the first weeks after birth, and reported more than 10% human cells after 4 weeks in 40% of mice, with a subsequent development of lethal X-GVHD as they mention in a significant number of mice. 33 2.3 BNX mice The bnx mouse strain was constructed by a combination of 3 recessive mutations, beige, nude and xid. 34 The spontaneous beige mutation is an analog of the human Chediak-Higashi syndrome in mice, and is associated with neutropenia and a lack of NK cytotoxic activity. The nude mutation affects the development of the thymic epithelium. Engraftment is comparable to SCID mice. 35 Bnx mice are severely immune deficient for T and B cells, but they still possess a certain amount of natural immunity, like intermediate levels of NK cells and macrophages. 37 Proefschrift Roos van Rijn.indb 37 19-10-2006 16:47:41

Preclinical evaluation of human T lymphocytes in RAG2 -/- γc -/- mice Rozemarijn S. van Rijn 2.4 NOD-SCID mice The non-obese diabetic (NOD) mice are characterized by deficiency of complement (2 base-pair deletion in a 5 exon of the murine C5 gene), molecularly undefined defects of macrophage function (defective regulation of colony-stimulating factor-1 and interferon-γ receptors, and reduced secretion of Il-1) and NK function. Introducing the scid mutation onto the NOD background by several groups has produced NOD/ LtSz-SCID 36 and NOD/Shi-SCID 37 mouse strains with diminished innate immunity. Knocking out the gene for β2-microglobulin in NOD-SCID mice results in a total absence of MHC class I and a total loss of NK activity. 38,39 Compared to SCID mice these mice have the advantage of a higher engraftment of human cells without the need for exogenous cytokines. Disadvantages are their comparable increased sensitivity to radiation and relatively short life span because of the common development of thymomas within a few months. They also exhibit a significant residual macrophage activity and can show leakiness, that is the development of functional B- or T-cells over time. 2.5 RAG1 deficient mice Another mouse strain is the recombinase-activating gene 1-knockout (RAG1-), which exhibits a total absence of mature T and B cells because of an arrest in their development. 40 The advantage of the RAG1-knockout compared to SCID mice would be the absence of leakiness. However, engraftment levels of human T cells and human immunoglobulins were reported to be significantly lower compared to the NOD-SCID strains: following ip transfer of 50x10 6 hupbmcs, human CD3 + cells were only detectable (<1%) in RAG1-knockout mice compared to levels up to 30% in peripheral blood within 20 days for NOD-SCID mice. 41 High levels of engraftment of 20x10 6 human T cells to 30-40% engraftment levels in the spleen, were reported also by ip transfer of hupbmcs into NOD/LtSz-RAG1 null mice, which are also claimed to be more radioresistant, have an increased mean life span due to later onset of T-cell lymphoma development and do not show leakiness. 42 Even higher engraftment levels to 50-60% were obtained by ip transfer of 20x10 6 hupbmcs into the more recently developed NOD/ LtSz-RAG1 null Pfp null mouse strain. A targeted mutation in the perforin (Pfp) structural gene, the major mediator of NK cell cytotoxicity, produced mice that lack NK cell cytotoxic function in addition to the NOD/LtSz-RAG1 already incorporated defects. 43 2.6 RAG2 -/- γc -/- mice In 1998, Goldman et al developed the RAG2 -/- γc -/- mouse strain, which was also used for the model described in this thesis. 44 Inactivation of the lymphoid-specific recombinase-activating RAG2 gene, like the RAG1 gene described above, in humans and mice, completely abrogates the production of thymus-derived T lymphocytes and peripheral B lymphocytes, although numbers of splenic macrophages and NK cells are increased. 38 Proefschrift Roos van Rijn.indb 38 19-10-2006 16:47:41

Chapter 2 the development of xenograft models for the in vivo study of human T cells The common cytokine receptor γ chain (γc) is a functional subunit of the receptors for IL-2, IL-4, IL-7, Il-9, IL-15 and IL-21, and plays an important role in lymphoid development. Also there is an absolute dependence on γc for development of NK cells and for export and survival of NK-T cells from the thymus. Inactivation of the gene is responsible for human X-linked SCID, and in homozygous mutant mice results in abnormal T-cell lymphopoiesis, dysfunction of residual peripheral T-lymphocyte cell populations and absence of NK cell function. Pilot studies with ip injection of 20-40x10 6 hupbmcs showed high but variable levels of engraftment from 0.1-77% in the spleens and high plasma levels of human immunoglobulins. Four to ten weeks following transfer of the hupbmcs, a mild X-GVHD was observed, characterized by lymphocyte infiltration in the liver and fibrosis of the spleen. 44 2.7 Stem cell engraftment for the study of human lymphoid cells Apart from the transfer of hupbmcs to study human T cells in vivo, a different approach used, is the engraftment of human CD34 + stem cells in immunodeficient mice resulting in development of human T cells within the humanized murine environment. In 1988, in the same month that Mosier published his results on transfer of hupbmcs, Mc Cune reported the development of mature human T and B cells following transplantation of human fetal liver hematopoietic cells, human fetal thymus, and human fetal lymph node under the kidney capsule of SCID mice. 45 Three months later Kamel- Reid & Dick transplanted human bone marrow into bnx mice, but although myeloid cells engrafted to 0.1-1.0% in the bone marrow and spleen, human lymphoid cells were not observed. 46 The levels of engraftment of human stem cells could be increased by the exogenous administration of a cocktail of human hematopoietic growth factors, which then did result in outgrowth of immature and mature lineages including detectable lymphoid progenitors. 35 Other groups improved this model of implanting fetal tissues together with stem cells into immunodeficient mice, generally resulting in a slow appearance of about 2% of functional human T cells in the peripheral circulation. 47-50 Recently, several groups reported relatively high engraftment levels of functional T cells following transplantation of CD34 + cord blood cells into variant strains of NOD- SCID mice. 51-55 Engraftment levels of up to 60% human T cells in the thymus were reported in an adapted NOD-SCID model, that included pretreatment of the mice with TM-β1. 54 Intrahepatic injection of CD34 + cord blood cells into RAG2 -/- γc -/- newborn mice regenerated adaptive immunity mediated by significant numbers of functional T and B cells. 55 Similar results were obtained in newborn NOD-SCID-IL2rγ null mice by Ishikawa et al. 52 Very recently, reconstitution of a functional human immune system was 39 Proefschrift Roos van Rijn.indb 39 19-10-2006 16:47:42

Preclinical evaluation of human T lymphocytes in RAG2 -/- γc -/- mice Rozemarijn S. van Rijn described in NOD-SCID mice through combined fetal thymus/liver transplantation under the kidney capsule and CD34 + cord blood cells iv with very high engraftment levels in peripheral blood (26-46%). 56 The advantage of these models over direct transfer of hupbmcs for the study of human T cells in vivo is that the stromal environment permits differentiation of human hematopoietic cells within the implanted tissue, leading to the possibility of a physiologic homeostatic proliferation of the lymphoid cells in a humanized model. However, the procedures generally require fetal material, are time consuming and technically complicated. In addition, high levels of T cell chimerism have not been obtained so far, so that for instance experiments with ex vivo manipulation of the human T cells have not yet been feasible. Therefore, the hupbmc model still remains the most accessible and simple means to examine human lymphocyte function and activity in vivo. 40 Proefschrift Roos van Rijn.indb 40 19-10-2006 16:47:42

Chapter 2 the development of xenograft models for the in vivo study of human T cells Reference List 1. Mosier DE, Gulizia RJ, Baird SM, Wilson DB. Transfer of a functional human immune system to mice with severe combined immunodeficiency. Nature. 1988;335:256-259. 2. Cannon MJ, Pisa P, Fox RI, Cooper NR. Epstein-Barr virus induces aggressive lymphoproliferative disorders of human B cell origin in SCID/hu chimeric mice. J Clin Invest. 1990;85:1333-1337. 3. Duchosal MA, McConahey PJ, Robinson CA, Dixon FJ. Transfer of human systemic lupus erythematosus in severe combined immunodeficient (SCID) mice. J Exp Med. 1990;172:985-988. 4. Krams SM, Dorshkind K, Gershwin ME. Generation of biliary lesions after transfer of human lymphocytes into severe combined immunodeficient (SCID) mice. J Exp Med. 1989;170:1919-1930. 5. Macht L, Fukuma N, Leader K et al. Severe combined immunodeficient (SCID) mice: a model for investigating human thyroid autoantibody synthesis. Clin Exp Immunol. 1991;84:34-42. 6. Purtilo DT, Falk K, Pirruccello SJ et al. SCID mouse model of Epstein-Barr virusinduced lymphomagenesis of immunodeficient humans. Int J Cancer. 1991;47:510-517. 7. Rowe M, Young LS, Crocker J et al. Epstein-Barr virus (EBV)-associated lymphoproliferative disease in the SCID mouse model: implications for the pathogenesis of EBV-positive lymphomas in man. J Exp Med. 1991;173:147-158. 8. Saxon A, Macy E, Denis K et al. Limited B cell repertoire in severe combined immunodeficient mice engrafted with peripheral blood mononuclear cells derived from immunodeficient or normal humans. J Clin Invest. 1991;87:658-665. 9. Mosier DE. Immunodeficient mice xenografted with human lymphoid cells: new models for in vivo studies of human immunobiology and infectious diseases. J Clin Immunol. 1990;10:185-191. 10. Tary-Lehmann M, Saxon A. Human mature T cells that are anergic in vivo prevail in SCID mice reconstituted with human peripheral blood. J Exp Med. 1992;175:503-516. 11. Hoffmannfezer G, Gall C, Zengerle U, Kranz B, Thierfelder S. Immunohistology and Immunocytology of Human T-Cell Chimerism and Graft-Versus-Host Disease in Scid Mice. Blood. 1993;81:3440-3448. 12. Fraser CC, Chen BP, Webb S, van RN, Kraal G. Circulation of human hematopoietic cells in severe combined immunodeficient mice after Cl2MDP-liposome-mediated macrophage depletion. Blood. 1995;86:183-192. 41 Proefschrift Roos van Rijn.indb 41 19-10-2006 16:47:42

Preclinical evaluation of human T lymphocytes in RAG2 -/- γc -/- mice Rozemarijn S. van Rijn 13. Sandhu JS, Gorczynski R, Shpitz B et al. A human model of xenogeneic graftversus-host disease in SCID mice engrafted with human peripheral blood lymphocytes. Transplantation. 1995;60:179-184. 14. Shibata S, Asano T, Noguchi A et al. Enhanced engraftment of human peripheral blood lymphocytes into anti-murine interferon-gamma monoclonal antibody-treated CB-17-scid mice. Cellular Immunology. 1998;183:60-69. 15. Xun CQ, Thompson JS, Jennings CD, Brown SA, Widmer MB. Effect of total body irradiation, busulfan-cyclophosphamide, or cyclophosphamide conditioning on inflammatory cytokine release and development of acute and chronic graft-versus-host disease in H-2-incompatible transplanted SCID mice. Blood. 1994;83:2360-2367. 16. Lotzova E, Savary CA, Pollack SB. Prevention of rejection of allogeneic bone marrow transplants by NK 1.1 antiserum. Transplantation. 1983;35:490-494. 17. Murphy WJ, Kumar V, Bennett M. Acute rejection of murine bone marrow allografts by natural killer cells and T cells. Differences in kinetics and target antigens recognized. J Exp Med. 1987;166:1499-1509. 18. Tiberghien P, Longo DL, Wine JW, Alvord WG, Reynolds CW. Anti-asialo GM1 antiserum treatment of lethally irradiated recipients before bone marrow transplantation: evidence that recipient natural killer depletion enhances survival, engraftment, and hematopoietic recovery. Blood. 1990;76:1419-1430. 19. Murphy WJ, Bennett M, Anver MR, Baseler M, Longo DL. Human-mouse lymphoid chimeras: host-vs.-graft and graft-vs.-host reactions. Eur J Immunol. 1992;22:1421-1427. 20. Sandhu J, Shpitz B, Gallinger S, Hozumi N. Human primary immune response in SCID mice engrafted with human peripheral blood lymphocytes. J Immunol. 1994;152:3806-3813. 21. Shpitz B, Chambers CA, Singhal AB et al. High level functional engraftment of severe combined immunodeficient mice with human peripheral blood lymphocytes following pretreatment with radiation and anti-asialo GM1. J Immunol Methods. 1994;169:1-15. 22. Christianson SW, Greiner DL, Schweitzer IB et al. Role of natural killer cells on engraftment of human lymphoid cells and on metastasis of human T-lymphoblastoid leukemia cells in C57BL/6J-scid mice and in C57BL/6J-scid bg mice. Cell Immunol. 1996;171:186-199. 23. Tournoy KG, Depraetere S, Pauwels RA, Leroux-Roels GG. Mouse strain and conditioning regimen determine survival and function of human leucocytes in immunodeficient mice. Clin Exp Immunol. 2000;119:231-239. 24. Tsuchida M, Brown SA, Tutt LM et al. A model of human anti-t-cell monoclonal antibody therapy in SCID mice engrafted with human peripheral blood lymphocytes. Clin Transplant. 1997;11:522-528. 42 Proefschrift Roos van Rijn.indb 42 19-10-2006 16:47:42

Chapter 2 the development of xenograft models for the in vivo study of human T cells 25. Cavacini LA, Kennel M, Lally EV, Posner MR, Quinn A. Human immunoglobulin production in immunodeficient mice: enhancement by immunosuppression of host and in vitro activation of human mononuclear cells. Clin Exp Immunol. 1992;90:135-140. 26. Barry TS, Jones DM, Richter CB, Haynes BF. Successful engraftment of human postnatal thymus in severe combined immune deficient (SCID) mice: differential engraftment of thymic components with irradiation versus anti-asialo GM-1 immunosuppressive regimens. J Exp Med. 1991;173:167-180. 27. Murphy WJ, Durum SK, Anver MR et al. Induction of T cell differentiation and lymphomagenesis in the thymus of mice with severe combined immune deficiency (SCID). J Immunol. 1994;153:1004-1014. 28. Santini SM, Spada M, Parlato S et al. Treatment of severe combined immunodeficiency mice with anti-murine granulocyte monoclonal antibody improves human leukocyte xenotransplantation. Transplantation. 1998;65:416-420. 29. Murphy WJ, Conlon KC, Sayers TJ et al. Engraftment and activity of anti-cd3- activated human peripheral blood lymphocytes transferred into mice with severe combined immune deficiency. J Immunol. 1993;150:3634-3642. 30. Murphy WJ, Tian ZG, Asai O et al. Chemokines and T lymphocyte activation: II. Facilitation of human T cell trafficking in severe combined immunodeficiency mice. J Immunol. 1996;156:2104-2111. 31. Martensson C, Ifversen P, Borrebaeck CA, Carlsson R. Enhancement of specific immunoglobulin production in SCID-hu-PBL mice after in vitro priming of human B cells with superantigen. Immunology. 1995;86:224-230. 32. Lubin I, Segall H, Marcus H et al. Engraftment of human peripheral blood lymphocytes in normal strains of mice. Blood. 1994;83:2368-2381. 33. Pflumio F, Lapidot T, Murdoch B, Patterson B, Dick JE. Engraftment of human lymphoid cells into newborn SCID mice leads to graft-versus-host disease. Int Immunol. 1993;5:1509-1522. 34. Andriole GL, Mule JJ, Hansen CT, Linehan WM, Rosenberg SA. Evidence that lymphokine-activated killer cells and natural killer cells are distinct based on an analysis of congenitally immunodeficient mice. J Immunol. 1985;135:2911-2913. 35. Dick JE, Lapidot T, Pflumio F. Transplantation of normal and leukemic human bone marrow into immune-deficient mice: development of animal models for human hematopoiesis. Immunol Rev. 1991;124:25-43. 36. Shultz LD, Schweitzer PA, Christianson SW et al. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J Immunol. 1995;154:180-191. 37. Koyanagi Y, Tanaka Y, Kira J et al. Primary human immunodeficiency virus type 1 viremia and central nervous system invasion in a novel hu-pbl-immunodeficient mouse strain. J Virol. 1997;71:2417-2424. 43 Proefschrift Roos van Rijn.indb 43 19-10-2006 16:47:42

Preclinical evaluation of human T lymphocytes in RAG2 -/- γc -/- mice Rozemarijn S. van Rijn 38. Christianson SW, Greiner DL, Hesselton RA et al. Enhanced human CD4+ T cell engraftment in beta2-microglobulin-deficient NOD-scid mice. J Immunol. 1997;158:3578-3586. 39. Wagar EJ, Cromwell MA, Shultz LD et al. Regulation of human cell engraftment and development of EBV-related lymphoproliferative disorders in Hu-PBL-scid mice. J Immunol. 2000;165:518-527. 40. Mombaerts P, Iacomini J, Johnson RS et al. RAG-1-deficient mice have no mature B and T lymphocytes. Cell. 1992;68:869-877. 41. Berney T, Molano RD, Pileggi A et al. Patterns of engraftment in different strains of immunodeficient mice reconstituted with human peripheral blood lymphocytes. Transplantation. 2001;72:133-140. 42. Shultz LD, Lang PA, Christianson SW et al. NOD/LtSz-Rag1null mice: an immunodeficient and radioresistant model for engraftment of human hematolymphoid cells, HIV infection, and adoptive transfer of NOD mouse diabetogenic T cells. J Immunol. 2000;164:2496-2507. 43. Shultz LD, Banuelos S, Lyons B et al. NOD/LtSz-Rag1nullPfpnull mice: a new model system with increased levels of human peripheral leukocyte and hematopoietic stem-cell engraftment. Transplantation. 2003;76:1036-1042. 44. Goldman JP, Blundell MP, Lopes L et al. Enhanced human cell engraftment in mice deficient in RAG2 and the common cytokine receptor gamma chain. Br J Haematol. 1998;103:335-342. 45. McCune JM, Namikawa R, Kaneshima H et al. The SCID-hu mouse: murine model for the analysis of human hematolymphoid differentiation and function. Science. 1988;241:1632-1639. 46. Kamel-Reid S, Dick JE. Engraftment of immune-deficient mice with human hematopoietic stem cells. Science. 1988;242:1706-1709. 47. Kollmann TR, Kim A, Zhuang X, Hachamovitch M, Goldstein H. Reconstitution of SCID mice with human lymphoid and myeloid cells after transplantation with human fetal bone marrow without the requirement for exogenous human cytokines. Proc Natl Acad Sci U S A. 1994;91:8032-8036. 48. Krowka JF, Sarin S, Namikawa R, McCune JM, Kaneshima H. Human T cells in the SCID-hu mouse are phenotypically normal and functionally competent. J Immunol. 1991;146:3751-3756. 49. Namikawa R, Weilbaecher KN, Kaneshima H, Yee EJ, McCune JM. Long-term human hematopoiesis in the SCID-hu mouse. J Exp Med. 1990;172:1055-1063. 50. Vandekerckhove BA, Krowka JF, McCune JM et al. Clonal analysis of the peripheral T cell compartment of the SCID-hu mouse. J Immunol. 1991;146:4173-4179. 51. Crisa L, Cirulli V, Smith KA et al. Human cord blood progenitors sustain thymic T-cell development and a novel form of angiogenesis. Blood. 1999;94:3928-3940. 44 Proefschrift Roos van Rijn.indb 44 19-10-2006 16:47:43

Chapter 2 the development of xenograft models for the in vivo study of human T cells 52. Ishikawa F, Yasukawa M, Lyons B et al. Development of functional human blood and immune systems in NOD/SCID/IL2 receptor {gamma} chain(null) mice. Blood. 2005;106:1565-1573. 53. Ito M, Hiramatsu H, Kobayashi K et al. NOD/SCID/gamma(c)(null) mouse: an excellent recipient mouse model for engraftment of human cells. Blood. 2002;100:3175-3182. 54. Kerre TC, De SG, de SM et al. Adapted NOD/SCID model supports development of phenotypically and functionally mature T cells from human umbilical cord blood CD34(+) cells. Blood. 2002;99:1620-1626. 55. Traggiai E, Chicha L, Mazzucchelli L et al. Development of a human adaptive immune system in cord blood cell-transplanted mice. Science. 2004;304:104-107. 56. Lan P, Tonomura N, Shimizu A, Wang S, Yang YG. Reconstitution of a functional human immune system in immunodeficient mice through combined human fetal thymus/liver and CD34+ cell transplantation. Blood. 2006. 45 Proefschrift Roos van Rijn.indb 45 19-10-2006 16:47:43

Preclinical evaluation of human T lymphocytes in RAG2 -/- γc -/- mice Rozemarijn S. van Rijn 46 Proefschrift Roos van Rijn.indb 46 19-10-2006 16:47:43