24.08.2015 Views

Check GAD antibody positivity with the Diamyd anti-GAD RIA plate*

GAD in Metabolic - Diamyd Medical AB

GAD in Metabolic - Diamyd Medical AB

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

forewordResearch Scientists throughout <strong>the</strong> world are currently faced<strong>with</strong> a time-old challenge – to define and understand <strong>the</strong>mechanisms leading to development of autoimmune diseases,and <strong>the</strong>n to determine and develop efficient means oftreating or preventing <strong>the</strong>m. While <strong>the</strong>se might sound like two distinctchallenges, <strong>the</strong> definition of <strong>the</strong> molecules targeted in an autoimmune diseaseprocess also provides <strong>the</strong> candidates for <strong>the</strong>rapeutic targeting.To date, <strong>the</strong>re is no vaccine for any of <strong>the</strong> many autoimmune diseases thataffect millions of people throughout <strong>the</strong> world. A number of candidate moleculeshave been identified and targeted, such as insulin in Type 1 diabetesand myelin basic protein in Multiple Sclerosis, but clinical vaccination trialsusing <strong>the</strong>se molecules have not yet been successful. Given <strong>the</strong> recent advancesin gene technology and knowledge of <strong>the</strong> genetic codes comprising bothMan and experimental animals, <strong>the</strong> potential for discovery of new candidatemolecules is great. However, one candidate molecule identified manyyears ago still stands <strong>the</strong> test of time.Glutamic acid decarboxylase 65 (<strong>GAD</strong>65) is a candidate auto<strong>anti</strong>genimplicated in development of <strong>the</strong> autoimmune disease Type 1 diabetes. Inautumn 1994, ‘The Story of <strong>GAD</strong>’ by Robert Dinsmoor appeared in <strong>the</strong>Countdown magazine, a publication of <strong>the</strong> Juvenile Diabetes ResearchFoundation International. This article described <strong>the</strong> series of research discoveriesencompassing <strong>the</strong> period 1982-1993 that led to definition of<strong>GAD</strong>65 as a prime candidate auto<strong>anti</strong>gen in Type 1 diabetes. The articleended <strong>with</strong> two pertinent statements – that access to recombinant <strong>GAD</strong>in industrial-sized qu<strong>anti</strong>ties would be required for testing of <strong>GAD</strong> <strong>the</strong>rapiesin humans, and that <strong>GAD</strong>-based <strong>the</strong>rapies should provide a meansof preventing diabetes.During <strong>the</strong> last 10 years, intensive efforts have led to development ofhigh quality recombinant <strong>GAD</strong>65 proteins, which have been tested inPhase I and Phase II clinical trials.Additionally, recent research efforts havealso identified <strong>GAD</strong> as an important elementin Parkinson’s and o<strong>the</strong>r neurologicaldiseases, indicating that <strong>the</strong> candidate moleculehas not only remained a potential keyto prevention of diabetes, but also import<strong>anti</strong>n treatment of o<strong>the</strong>r diseases as well. Thereis obviously still more to learn about thisfascinating protein.This issue is dedicated to <strong>GAD</strong>65 <strong>with</strong> anumber of contributions made by several prominent <strong>GAD</strong> researchers. Aswell as personal reflections of <strong>the</strong>ir own past experiences of <strong>GAD</strong>, <strong>the</strong>current status and proposals for future applications of <strong>GAD</strong> are presented.We hope you find <strong>the</strong>se articles interesting.Assoc. Prof. Robert A. HarrisintroductionIntroduction byLars KlareskogIn 1999 diamyd medical initiated a unique cooperation<strong>with</strong> <strong>the</strong> Karolinska Hospital whensetting up its Competence Center forAutoimmune Diabetes (DMCCAD) at <strong>the</strong>Center for Molecular Medicine (CMM) atKarolinska. Since its inception, <strong>with</strong> Robert Harris at<strong>the</strong> helm, DMCCAD has become an interesting pointfor cross-fertilization of ideas related to various autoimmunediseases including Type 1 diabetes, MS andRheumatoid Arthritis.It is generally agreed that Glutamic AcidDecarboxylase 65 (<strong>GAD</strong>) is an important <strong>anti</strong>gen inType 1 diabetes. As in o<strong>the</strong>r autoimmune diseases,recent research is focused on induction of tolerance todisease specific <strong>anti</strong>gens. This may protect self structuresunder attack <strong>with</strong>out interfering <strong>with</strong> <strong>the</strong> immunesystem’s capabilities in o<strong>the</strong>r areas, such as combattingviral infections or cancer.While it is already generally accepted that <strong>GAD</strong><strong>anti</strong>bodies (<strong>GAD</strong>A) is a major diagnostic marker forType 1 diabetes and predicting <strong>the</strong> course of developmentof Type 2 diabetes (LADA-patients), toleranceinduction to <strong>GAD</strong> may become a way to prevent <strong>the</strong>development of insulin dependency in LADA patients.I have had <strong>the</strong> pleasure of being able to follow <strong>the</strong>development of <strong>Diamyd</strong> Medical’s <strong>GAD</strong>-based vaccinefor LADA patients. The outcome of <strong>the</strong> Phase IIstudy in LADA patients is important, not only becauseit may lead to a new drug for diabetes <strong>the</strong>rapy, butits results may also be of importance when designingfuture clinical trials in o<strong>the</strong>r diseases such as MS.page 4 dmccad june 2003


Lars Klareskog, MD, Ph.D., is Professor of Rheumatology and Head of<strong>the</strong> Rheumatology Research group at <strong>the</strong> Karolinska Hospital inStockholm and formerly Professor of Medical Immunology and Headof Clinical Immunology at <strong>the</strong> Uppsala Teaching Hospital. Klareskog'sresearch is specifically aimed at <strong>the</strong> causes and treatment of autoimmunedisorders. Klareskog is a member of <strong>the</strong> Nobel Foundation.Comments on <strong>Diamyd</strong> Diabetes Vaccineby Hans WigzellThat <strong>the</strong> human immune system can react against selfstructures is well known. Most such reactions do notcause disease – but some do.In <strong>the</strong> efforts to fight autoimmune disease such as Type1 diabetes, one <strong>the</strong>rapeutic approach is to modify <strong>the</strong> immune reactionand induce functional tolerance to potentially relevant moleculessuch as <strong>GAD</strong> by administration of <strong>the</strong> autologous <strong>anti</strong>gen itself.Similar approaches have been successful before. For exampleadministration of auto<strong>anti</strong>gens have alleviated autoimmune diseasein animal models. Hyposensitization (where increasing doses of allergenare used to treat allergies) is ano<strong>the</strong>r example.How should <strong>GAD</strong> be used to induce functional tolerance?Using our knowledge from conventional as well as from autologousvaccines it seems logical to use doses from a few micrograms to 500Hans Wigzell, MD, D.Sc, is Professor of Immunology, Dean of <strong>the</strong>Karolinska Institute and Chief Scientific Advisor to <strong>the</strong> SwedishGovernment. Wigzell is one of Sweden´s most prominent and internationallyrenowned scientists in <strong>the</strong> field of Immunology. Wigzellwas Director General of <strong>the</strong> National Bacteriological Laboratory1988-1993; Director General of <strong>the</strong> Swedish Institute for InfectiousDiseases 1993-1994; Wigzell is since 1990 Chairman of <strong>the</strong> ECConcerted Research Programme: European Vaccine against AIDS(EVA). Wigzell was Chairman of <strong>the</strong> Nobel Committee 1990-1992and is Chairman of <strong>the</strong> Nobel Foundation.micrograms.To use alum as adjuvant seems highly recommendable. It is conventionaland it is biased to <strong>the</strong> humoral ra<strong>the</strong>r than <strong>the</strong> cellularimmune response which is logical when <strong>the</strong> cellular response is to bereduced.Subcutaneous administration is recommendable and conventional.Intravenous administration is much more problematic and intramuscularis less efficient in terms of immunogenicity.The <strong>GAD</strong>-vaccine differs from a conventional vaccine in that <strong>the</strong>administered <strong>anti</strong>gen is already present naturally in <strong>the</strong> body. Thisprobably means that <strong>the</strong> vaccine needs to be injected a couple oftimes. Therefore a prime and boost strategy seems logical.In summary, <strong>the</strong> diamyd approach to induce functional tolerance to<strong>GAD</strong> seems logical.dmccad june 2003page 5


The Story of<strong>GAD</strong>Robert Dinsmoor1975Researchersdiscover isletcell surface<strong>anti</strong>bodies inchildren <strong>with</strong>diabetes1982Scientistsfind 64K<strong>anti</strong>bodiesin diabeticBB rats1986Researchshows that64K <strong>anti</strong>bodiesdevelopyears beforediabetes1988Scientistsfind <strong>anti</strong>bodiesto <strong>GAD</strong> inpatients <strong>with</strong>Stiff ManSyndromeResearchersdiscover Islet CellAntibodies (ICAs)in people<strong>with</strong> newlydiagnoseddiabetes1978Scientistsfind <strong>anti</strong>bodiesto 64Kprotein indiabeticchildren1984Molecularbiologistssequencegene formaking <strong>GAD</strong>1987Researchersfind 64K<strong>anti</strong>bodies indiabeticNOD miceThis article’s original form appeared in Countdown magazine, a publication of <strong>the</strong> Juvenile Diabetes Research Foundation International, Autumn 1994.At one time, scientists thought that diabetes wascaused by a virus or toxic environmentalagent that destroyed <strong>the</strong>insulin-producing beta cells of <strong>the</strong>pancreas. But decades ago, scientistsdiscovered that people <strong>with</strong> newlydiagnosed diabetes had inflammationof <strong>the</strong> islet cells of <strong>the</strong> pancreas, or insulitis.The islets were filled <strong>with</strong> white blood cells, which<strong>the</strong> body’s immune system uses to fightinfection.This led researchers to speculatethat diabetes might be an autoimmunedisease, caused by a misdirectedattack by <strong>the</strong> body’s own immunesystem.page 6 dmccad june 2003


More support for <strong>the</strong> autoimmune<strong>the</strong>ory came in <strong>the</strong> early1970’s when Dr. Bottazzo andDr. Doniach and colleagues at<strong>the</strong> Middlesex Hospital inLondon first detected <strong>anti</strong>bodiesto pancreatic islet cells in people <strong>with</strong> newly diagnoseddiabetes. The <strong>anti</strong>bodies, which react to <strong>the</strong> cytoplasminside all islet cells, came to be called cytoplasmicislet cell <strong>anti</strong>bodies or ICAs.Antibodies are a part of <strong>the</strong> immune system responsecalled <strong>the</strong> humoral response. Made by white blood cellscalled B lymphocytes, specific <strong>anti</strong>bodies are designed toattack specific foreign proteins or <strong>anti</strong>gens. It was unclear,however, whe<strong>the</strong>r <strong>the</strong> beta cells were actually destroyedby <strong>the</strong>se <strong>anti</strong>bodies or by components of <strong>the</strong> cellularimmune response, such as <strong>the</strong> killer T lymphocytes or“T cells”.In <strong>the</strong> mid-1970’s, Dr. Lernmark, who had developed atechnique for isolating beta cells from isolated islets of1990In a majorbreakthrough,scientistsidentify <strong>the</strong>64K proteinas <strong>GAD</strong>1991Scientistsdiscover that 64Kprotein is<strong>GAD</strong>65, <strong>the</strong>smaller formof <strong>GAD</strong>Study showsthat T cellstarget<strong>GAD</strong>651993Research teamsprevent diabetesin NOD mice byinactivating <strong>GAD</strong>-Reactive T cellsResearcherssuggest 64K<strong>anti</strong>bodies maybe earlypredictor ofdiabetesMolecularbiologistssequence twoforms of <strong>GAD</strong>– <strong>GAD</strong>65 and<strong>GAD</strong>67 – fromislet cellsStudies showthat someICAs only reactto <strong>GAD</strong>65Two studiesestablish <strong>GAD</strong>as first knowntarget of T cellsin diabeticNOD mice<strong>the</strong> Langerhans, was researching proteins on <strong>the</strong> surface ofbeta cells at <strong>the</strong> University of Chicago. While involved inthis research, he attended a lecture in ICAs in Chicago byDr. Doniach. During <strong>the</strong> lecture, he realized that it mustbe <strong>the</strong> proteins on <strong>the</strong> surface of <strong>the</strong> beta cells that provoked<strong>anti</strong>bodies in diabetes, and he turned his research inthat direction. He and o<strong>the</strong>r researchers at <strong>the</strong> Universityof Chicago treated pancreatic islets cells from rats <strong>with</strong>blood serum from children who had diabetes for less thanfive years. They discovered that some of <strong>the</strong>se childrenhad <strong>anti</strong>bodies to proteins on <strong>the</strong> surface of <strong>the</strong> islet cells,called islet cell surface <strong>anti</strong>bodies or ICAs. However, <strong>the</strong>ydid not know which specific proteins had attracted <strong>the</strong><strong>anti</strong>bodies.The 64,000 Mystery ProteinIn <strong>the</strong> late 1970’s, Dr. Lernmark became Director ofResearch at <strong>the</strong> Hagedorn Research Laboratory inDenmark, where he continued to work <strong>with</strong> ICAs. Heenlisted <strong>the</strong> aid of Dr. Baekkeskov, who had developeddmccad june 2003page 7


Åke Lernmark, MD, Ph.D., and his colleagues atHagedorn Research Laboratory in Denmark, firstreported 64K proteins in humans in 1982.Steinunn Baekkeskov, Ph.D., played a primary role indetecting <strong>the</strong> 64K protein in humans and later worked incollaboration <strong>with</strong> Yale researchers to identify <strong>the</strong> proteinas <strong>GAD</strong>.Mark Atkinsson, Ph.D., and colleagues at <strong>the</strong> Universityof Florida in Gainesville, concluded in 1990 that 64Kauto<strong>anti</strong>bodies might be <strong>the</strong> earliest and best marker fordiabetes yet identified.techniques for detecting membrane proteins while studying<strong>the</strong> role of <strong>anti</strong>gens in African sleeping sickness. Usingnewly developed and very sensitive tests such as <strong>the</strong>“immunoprecipitation technique”, Drs. Baekkeskov,Lernmark and colleagues at <strong>the</strong> Hagedorn found 80-90percent of blood serum samples from children <strong>with</strong>newly diagnosed diabetes had <strong>anti</strong>bodies to an unidentifiedICSA <strong>with</strong> molecular weight of approximately 64,000daltons-or 64 kilodaltons. They dubbed <strong>the</strong> mystery protein64K, and reported <strong>the</strong>ir findings in 1982 in Nature.In 1984, <strong>the</strong>y reported that 64K <strong>anti</strong>bodies were oftenfound in diabetic BB rats (a commonly used animalmodel for human diabetes). One intriguing aspect of thisfinding was that <strong>the</strong> <strong>anti</strong>bodies appeared 40 to 70 daysbefore <strong>the</strong> onset of diabetes. This suggested that <strong>the</strong> 64Kprotein was an early and major target of <strong>the</strong> immunesystem. But did this same predictive effect hold true forhumans as well?They next looked at blood serum samples of 14 peoplewho later developed diabetes. Eleven of <strong>the</strong>m had <strong>anti</strong>bodiesto <strong>the</strong> 64K protein - for up to seven years before <strong>the</strong>ybegan to show symptoms of diabetes. This suggested that64K <strong>anti</strong>bodies might be an early warning sign for <strong>the</strong>eventual development of diabetes.In <strong>the</strong> summer of 1986 Dr. Atkinson who had beenstudying environmental influences on diabetes in rats at<strong>the</strong> University of Florida in Gainesville, traveled toHagedorn as a visiting scientist. There Baekkeskov taughthim <strong>the</strong> immunoprecipitation technique for detecting64K <strong>anti</strong>bodies. Armed <strong>with</strong> this new skill, he returned to<strong>the</strong> University of Florida, where he and Dr. NoelMaclaren, detected <strong>the</strong> 64K <strong>anti</strong>bodies in nonobese diabetic(NOD) mice, a newly bred type of animal model fordiabetes. Finding <strong>the</strong> 64K <strong>anti</strong>bodies in animals helpedconfirm that <strong>the</strong> 64K <strong>anti</strong>gen was an important feature indiabetes.Drs. Atkinson, Maclaren, and colleagues also studiedfirst-degree relatives of people <strong>with</strong> diabetes, who were athigher risk for developing diabetes <strong>the</strong>mselves. In 1990, in<strong>the</strong> British Journal The Lancet, <strong>the</strong>y reported finding 64Kauto<strong>anti</strong>bodies in 23 of 28 people who developed diabetesup to seven years later. Based on <strong>the</strong>se findings and thoseof <strong>the</strong> Hagedorn researchers <strong>the</strong>y concluded that 64Kmight be <strong>the</strong> earliest and best marker for diabetes yetidentified and might be especially useful for predictingdiabetes.As diabetes researchers became increasingly aware ofhow important this protein was, <strong>the</strong>y set out to analyzeits chemical characteristics to compare <strong>with</strong> known proteins.In 1988, Drs. Solimena, Camilli, and <strong>the</strong>ir colleagues atYale University reported in The New England Journal ofMedicine that <strong>the</strong>y had found <strong>the</strong> target <strong>anti</strong>gen in patients<strong>with</strong> an autoimmune disorder called Stiff-ManSyndrome (“SMS”). SMS is a rare disorder of <strong>the</strong> centralnervous system in which a person’s muscles become moreand more rigid, and painful spasms occur. Patients <strong>with</strong><strong>the</strong> disorder were found to have <strong>anti</strong>bodies to an enzymecalled Glutamic acid Decarboxylase, or <strong>GAD</strong>.<strong>GAD</strong> is responsible for converting <strong>the</strong> amino acid glutamateinto a protein called GABA, which <strong>the</strong> brain cells useto communicate. It turned out that many patients <strong>with</strong>page 8 dmccad june 2003


<strong>GAD</strong> <strong>anti</strong>bodies had diabetes – even <strong>the</strong> ones who neverdeveloped SMS. Moreover, <strong>the</strong>y found that <strong>anti</strong>bodies from<strong>the</strong> serum of SMS patients targeted <strong>the</strong> beta cells, and <strong>the</strong>sewere not any of <strong>the</strong> previously known <strong>anti</strong>bodies.This led to an historic collaboration between Dr.Baekkeskov, now at <strong>the</strong> University of California, SanFrancisco, and <strong>the</strong> Yale research team. Dr. Camilli sentBaekkeskov blood serum samples from SMS patients (containing<strong>GAD</strong> <strong>anti</strong>bodies), and she sent him blood serumsamples from diabetes patients (containing 64K <strong>anti</strong>bodies).Then <strong>the</strong>y used various methods to demonstrate that<strong>the</strong> proteins were completely identical. The end result ofthis meeting of <strong>the</strong> minds was <strong>the</strong>ir landmark paper inNature identifying <strong>the</strong> 64K protein as <strong>the</strong> enzyme <strong>GAD</strong>.The discovery set off a flurry of interest in <strong>GAD</strong> by diabetesresearchers.Send in <strong>the</strong> ClonesDr. Kaufman, who had been studying <strong>the</strong> role of <strong>GAD</strong> inneurological diseases such as epilepsy, became interested indiabetes one day in 1990 when his car broke down. Whilewaiting for it to be fixed, he went to <strong>the</strong> library, wherehe stumbled on <strong>the</strong> article by Dr. Atkinson and colleaguesin The Lancet describing <strong>the</strong> 64K <strong>anti</strong>bodies as predictorsof diabetes.According to Kaufman, <strong>the</strong> landmark paper identifying64K as <strong>GAD</strong> had yet to appear in Nature, but he was ableto put <strong>the</strong> two toge<strong>the</strong>r for himself: He knew about <strong>the</strong> roleof <strong>GAD</strong> in SMS and <strong>the</strong> fact that it was found in beta cells,and he also knew that that molecular weight of <strong>GAD</strong> wasapproximately 64kD. He joined forces <strong>with</strong> <strong>the</strong> researchteam at <strong>the</strong> University of Florida, as well as <strong>with</strong> his formermentor at <strong>the</strong> UCLA Dr. Tobin, who was researching <strong>the</strong>role of <strong>GAD</strong> in Huntington’s disease and epilepsy. Therewere known to be slightly different molecules of <strong>GAD</strong>found in <strong>the</strong> brain – one weighing 67kD (<strong>GAD</strong>67) and asmaller one weighing 65kD (<strong>GAD</strong>65). As a graduate studentworking <strong>with</strong> Tobin, Kaufman had worked out <strong>the</strong> geneticsequence for <strong>GAD</strong>67, and ano<strong>the</strong>r graduate student Dr.Erlander, had done <strong>the</strong> same for <strong>GAD</strong>65. Thanks in part tothis work, Dr. Tobin’s laboratory was now able to makeboth forms of <strong>GAD</strong> in qu<strong>anti</strong>ty using genetically alteredbacteria.Spurred by <strong>the</strong> news that 64K was <strong>GAD</strong>, o<strong>the</strong>r researchgroups cloned <strong>GAD</strong>. In 1988, Dr. Lernmark had moved to<strong>the</strong> University of Washington in Seattle <strong>with</strong> some membersof his former Hagedorn research team. One of <strong>the</strong>seDr. Allan Karlsen, cloned <strong>the</strong> gene for human islet<strong>GAD</strong>65. One of Lernmark’s former graduate student’s, Dr.Birgitte Michelsen, cloned <strong>GAD</strong>67. According to Dr.Lernmark, cloning <strong>GAD</strong> enabled researchers to makerecombinant <strong>GAD</strong> in qu<strong>anti</strong>ties <strong>the</strong>y had never dreamedAllan Tobin, Ph.D., and Daniel L. Kaufman, Ph.D., at <strong>the</strong> University of California at Los Angeles published one of two1993 papers in “Nature” confirming that <strong>GAD</strong> is <strong>the</strong> first known target of <strong>the</strong> T cell attack in diabetes.of. Cloning <strong>GAD</strong>65 also enabled researchers to determinewhich molecule was <strong>the</strong> alter ego of 64K.The researchers in Tobin’s lab sought help from aUCLA diabetes expert, Dr. Michael Clare-Salzler, tostudy <strong>GAD</strong>’s role in diabetes. Using blood serum samplesfrom diabetic patients <strong>with</strong> newly diagnosed diabetes providedby Dr. Clare-Salzler, <strong>the</strong> researchers showed that96% of patients <strong>with</strong> newly diagnosed diabetes had <strong>anti</strong>bodiesto one or both forms of <strong>GAD</strong>. Fur<strong>the</strong>rmore, itappeared that 64K was <strong>GAD</strong>65, <strong>the</strong> smaller of <strong>the</strong> twomolecules that had been identified.Dr. Erlander also used a computer search that uncoveredstructural similarities between both forms of <strong>GAD</strong>and proteins in <strong>the</strong> Coxsackie virus, a virus known toinfect many people who eventually developed diabetes.“That moment was probably <strong>the</strong> most exciting moment inmy whole scientific life!” Dr. Tobin recalled. This findingsupported a <strong>the</strong>ory known as “molecular mimcry”, whichsuggested that infection by <strong>the</strong> Coxsackie virus might setoff <strong>the</strong> immune reaction in diabetes. According to <strong>the</strong>“true identity”pageAs diabetes researchers became increasinglyaware of how important thismystery protein was, <strong>the</strong>y were eager to discover itsdmccad june 20039


<strong>the</strong>ory, <strong>the</strong> immune system mistakenlyattacks <strong>GAD</strong> in <strong>the</strong> beta cells aswell.<strong>GAD</strong> Provokes an Immune AttackIn 1992, a number of studies showedthat some of <strong>the</strong> ICAs, <strong>the</strong> <strong>anti</strong>bodiesdescribed back in 1974 that werevery predictive for diabetes, targeted<strong>GAD</strong>. One study from Bottazzo’sresearch group in London found thatICAs from <strong>the</strong> blood serum samplesof pre-diabetic individuals seemed toattack <strong>GAD</strong>. Likewise Dr. GeorgeEisenbarth and Dr. Robert Gianiniand colleagues at Joslin DiabetesCenter in Boston studied ICAs fromHugh McDevitt, at Stanford University suggested in 1993that diabetes could be prevented in its earliest stages by relatives of people <strong>with</strong> diabetes.preventing <strong>the</strong> T cell attack against <strong>GAD</strong>.They found that <strong>GAD</strong> was one targetof ICAs, but <strong>the</strong>re might be o<strong>the</strong>r<strong>anti</strong>gens as well.These reports showed an attack on <strong>GAD</strong> by <strong>the</strong> humoralimmune system – that is <strong>anti</strong>bodies. But <strong>the</strong>se <strong>anti</strong>bodiesare probably not what destroys <strong>the</strong> beta cells. Morelikely, <strong>the</strong>y are destroyed by <strong>the</strong> cellular immune responsein <strong>the</strong> form of T cells, white blood cells that infiltrate <strong>the</strong>pancreatic islets.The same year, Drs. Atkinson and Maclaren, working inconjunction <strong>with</strong> <strong>the</strong> UCLA research team, took T cellsfrom people <strong>with</strong> newly diagnosed diabetes, relatives ofpeople <strong>with</strong> diabetes, and nondiabetic individuals, andexposed <strong>the</strong>m all to <strong>GAD</strong>65. The T cells that reacted andmultiplied in <strong>the</strong> presence of <strong>GAD</strong>65 tended to be thosefrom people <strong>with</strong> diabetes and <strong>the</strong>ir relatives who hadICAs and would later develop diabetes. This showed that<strong>GAD</strong> provokes a T cell attack, which may be whatdestroys <strong>the</strong> beta cell in diabetes.In 1993, two studies published in Nature helped confirm<strong>GAD</strong> as <strong>the</strong> first known target of <strong>the</strong> T cells andsuggested that diabetes could be prevented in its earlieststages by preventing <strong>the</strong> T cell attack against <strong>GAD</strong>. Onestudy was from Kaufman and colleagues at UCLA in conjunction<strong>with</strong> Dr. Atkinson, and <strong>the</strong> o<strong>the</strong>r was from Dr.Hugh McDevitt, Dr. Roland Tisch and <strong>the</strong>ir colleagues atStanford University. In each case, <strong>the</strong> researchers testedNOD mice for reactivity to some known <strong>anti</strong>gens in diabetes,including <strong>the</strong> two forms of <strong>GAD</strong>. They found that<strong>the</strong> attack on <strong>GAD</strong> by T cells coincided <strong>with</strong> <strong>the</strong> developmentof insulitis – infiltration of <strong>the</strong> beta cells <strong>with</strong> whiteblood cells. Only later did <strong>the</strong> T cells attack o<strong>the</strong>r known<strong>anti</strong>gens in <strong>the</strong> beta cells.This suggested that <strong>GAD</strong> is <strong>the</strong> first <strong>anti</strong>gen to beattacked by T cells, and that this attack <strong>the</strong>n diversifies toinclude o<strong>the</strong>r <strong>anti</strong>gens, and that <strong>the</strong>se were recognizedand attacked by <strong>the</strong> immune system. Was this actually <strong>the</strong>cause, or was <strong>the</strong>re some o<strong>the</strong>r <strong>anti</strong>gen, as yet unknown,that provoked <strong>the</strong> first attack?To help answer <strong>the</strong>se questions, both groups of researcherstried deactivating <strong>the</strong> T cell response in <strong>the</strong>ir NODmice. The UCLA group injected NOD mice <strong>with</strong> <strong>GAD</strong> atthree weeks of age, a treatment already shown to inactivateT lymphocytes against <strong>GAD</strong>. Most of <strong>the</strong> mice treated<strong>with</strong> <strong>GAD</strong> showed no T cell response against <strong>GAD</strong>, indicatingcomplete immune system tolerance to <strong>GAD</strong>. The<strong>GAD</strong> tolerant mice showed no reaction to <strong>the</strong> o<strong>the</strong>r betacell <strong>anti</strong>gens, and never developed any degree of insulitisor diabetes. This suggested that <strong>GAD</strong> – and not any of<strong>the</strong> o<strong>the</strong>r known <strong>anti</strong>gens – provoked diabetes. On <strong>the</strong>o<strong>the</strong>r hand, mice treated <strong>with</strong> o<strong>the</strong>r <strong>anti</strong>gens did developa T cell attack to <strong>the</strong> o<strong>the</strong>r beta cell to <strong>anti</strong>gens, did developinsulitis, and went on to develop diabetes.The Stanford researchers likewise tried inactivating<strong>GAD</strong>-reactive T cells – but in this case by injecting NODmice <strong>with</strong> <strong>GAD</strong>65 into <strong>the</strong>ir thymus (a major site ofimmune system programming). The 70 percent of NODmice who actually became tolerant of <strong>GAD</strong> had markedlyreduced T lymphocyte responses to <strong>the</strong> o<strong>the</strong>r beta cell<strong>anti</strong>gens, reduced insulitis, and no development of diabetes.“I think this is an important step toward understanding<strong>the</strong> disease process,” Dr. Tisch explained. “It appears that<strong>the</strong> disease occurs in specific stages, and now we may beable to categorize <strong>the</strong> <strong>anti</strong>gens <strong>with</strong> regard to reactivity<strong>with</strong>in those stages.”The Prediction and Prevention Pay-OffNow that <strong>GAD</strong> has been identified, cloned, and shown tobe important in <strong>the</strong> development of diabetes, it can playan important role in diabetes treatment, to predict whowill get diabetes – probably years before <strong>the</strong>y develop anysymptoms of <strong>the</strong> disease. Now we have a very specificassay to show whe<strong>the</strong>r a healthy person has <strong>GAD</strong> <strong><strong>anti</strong>body</strong>.But <strong>the</strong> ultimate implications of <strong>GAD</strong> go far beyondpredicting who will get diabetes. The recent studies inNOD mice show that <strong>GAD</strong>, too, might play a role in preventingdiabetes.Unfortunately, testing oral <strong>GAD</strong> in humans won’t befeasible until researchers have access to recombinant<strong>GAD</strong> in industrial-sized qu<strong>anti</strong>ties and at an affordablecost. “We’ve actually discovered a direction for potentiallypreventing diabetes in human beings. Using treatmentssuch as <strong>with</strong> <strong>GAD</strong> we should be able to prevent diabetesin all individuals who are at risk of developing it,” concludedDr. Clare-Salzler.page 10 dmccad june 2003


<strong>Diamyd</strong>, Inc., is pleased to provide a comprehensive portfolio ofimmunoassays for In Vitro Diagnostic UseRECOMBINANT PROTEINS10-65702-13-01 T cell <strong>GAD</strong> 1mg/vial10-65702-01 rh<strong>GAD</strong>65 1mg/vial10-IA2-01Human IA-2 0.5 mg/vialAUTOANTIBODY DETECTION KITS10-1121-01 IAA ELISA10-1123-01 <strong>Diamyd</strong> <strong>anti</strong> <strong>GAD</strong>65 <strong>RIA</strong>DIABETES HUMAN IMMUNOASSAYS10-1113-01 Insulin ELISA10-1132-01 Ultra Sensitive Insulin ELISA10-1128-01 Iso-Insulin ELISA10-1136-01 C-Peptide Specific ELISA10-1141-01 Ultra Sensitive C-Peptide ELISA10-1118-01 Proinsulin ELISADIABETES MAMMALIAN IMMUNOASSAYS10-1124-01 Rat Insulin ELISA10-1137-01 Ultra Sensitive Rat Insulin ELISA10-1145-01 High Range Rat Insulin ELISA10-1149-01 Mouse Insulin ELISA10-1150-01 Ultra Sensitive Mouse Insulin ELISA10-1129-01 Porcine Insulin ELISA10-1130-01 Sheep Insulin ELISA10-1131-01 Bovine Insulin ELISACONTROL SERUM10-1134-01 Diabetes <strong>anti</strong>gen control, human L and H 2X 0.5 ml10-1135-01 Insulin control, Mammalian L and H 2X 0.5 mlCARDIOVASCULAR DISEASE10-1103-01 Apo(a) ELISA10-1106-01 Apo(a) <strong>RIA</strong>10-1143-01 Oxidized LDL ELISAT cell AND B-CELL SEPARATION KITS20-7200-01 Human T cell Kit20-7100-01 Mouse T cell Kit20-6900-01 Rat T cell Kit20-6900-01 B-Cell Accessory Kit (Human, Mouse, Rat)NEUROLOGICAL DISEASE10-1198-01 <strong>Diamyd</strong> S-100β Concentration Measurement Kit10-1199-01 <strong>Diamyd</strong> S-100-β Antibody ELISAwww.diamyd.comproducts@diamyd.com


<strong>GAD</strong> Back to <strong>the</strong> Future…Åke LernmarkÅke Lernmark, MD, Ph.D., is R.H.Williams Professor of Medicine at<strong>the</strong> University of Washington inSeattle. Lernmark focused hisattention on diabetes and <strong>the</strong><strong>anti</strong>gen that later proved to be<strong>GAD</strong> at an early stage. Lernmarkwas first to demonstrate <strong>the</strong> presenceof <strong>anti</strong>bodies against <strong>GAD</strong>in patients <strong>with</strong> insulin-dependentdiabetes, and to use molecularmethods to define HLA genes thatare necessary, but not sufficient todevelop <strong>the</strong> disorder.The beta cell specific loss associated <strong>with</strong><strong>the</strong> clinical onset of Type 1 diabetesmellitus (T1DM) is a remarkable phenomenon.The ablation of <strong>the</strong> insulinproducingcells does not seem to affectneighboring cells producing glucagon,somatostatin or pancreas polypeptide. The eradication of <strong>the</strong>beta cells is a testimony to <strong>the</strong> laser knife-like precision of<strong>the</strong> immune system to recognize and attack <strong>anti</strong>gen. Theimmune mediated loss of beta cells is well documented inpancreas specimens from T1DM patients of short diseaseduration. The degree and character of <strong>the</strong> inflammatory cellinfiltration vary from patient to patient and would not predictclinical onset. The loss of beta cells appears more predictiveand <strong>the</strong> major question that we and o<strong>the</strong>rs try to answeris whe<strong>the</strong>r <strong>the</strong> immune attack on <strong>the</strong> beta cells can bereversed by controlling <strong>the</strong> autoimmune reaction againstglutamic acid decarboxylase (<strong>GAD</strong>65) ei<strong>the</strong>r before or afteronset. While auto<strong>anti</strong>bodies to insulin and IA-2 are predictiveof T1DM primarily in <strong>the</strong> young, <strong>the</strong> predictive value of<strong>GAD</strong>65 auto<strong>anti</strong>bodies appears less age-dependent. It is <strong>the</strong>reforeoften argued that <strong>GAD</strong>65 is <strong>the</strong> major auto<strong>anti</strong>gen inT1DM, which is consistent <strong>with</strong> <strong>GAD</strong>65 auto<strong>anti</strong>bodiesbeing most prevalent (more than 80% of new onset patients)at onset. Although recent follow up studies suggestthat <strong>GAD</strong>65 auto<strong>anti</strong>bodies are not necessarily <strong>the</strong> firstauto<strong><strong>anti</strong>body</strong> to appear before <strong>the</strong> clinical diagnosis is made,appearance of all three auto<strong>anti</strong>bodies to <strong>GAD</strong>65, IA-2 andinsulin is <strong>the</strong> best predictor of disease. Recognition of<strong>GAD</strong>65 by helper CD4 and killer CD8 positive cells are likelyto precede <strong>the</strong> auto<strong><strong>anti</strong>body</strong> appearance. At present,<strong>the</strong>re is a lack of reliable and standardized tests that detectT cell reactivity against islet auto<strong>anti</strong>gens (1). This is in contrastto <strong>GAD</strong>65 and IA-2 auto<strong>anti</strong>bodies, which can be reliablymeasured against a WHO standard (2). At least <strong>the</strong>rewill be one reliable way to monitor whe<strong>the</strong>r SpecificImmune Therapy (SIT) <strong>with</strong> <strong>GAD</strong>65 might alter <strong>the</strong> courseof T1DM disease associated autoimmunity.Following <strong>the</strong> molecular cloning of <strong>the</strong> human (3) androdent <strong>GAD</strong> proteins (4) it was possible to carry out experiments<strong>with</strong> recombinant <strong>GAD</strong>65 to demonstrate that spontaneousdiabetes in <strong>the</strong> NOD mouse (5, 6) was preventable.Of equal importance was <strong>the</strong> observation that diabetes wasnot inducible by <strong>GAD</strong>65 in rats (7) and mice (8). T1D <strong>the</strong>reforedid not immediately seem to mimic o<strong>the</strong>r autoimmunediseases, which have animal models that are based on <strong>the</strong>actual immunization of an auto<strong>anti</strong>gen. About 10 yearsafter <strong>the</strong> demonstration that NOD mouse diabetes was preventable,Phase I (toxicity) and Phase II (safety) clinical trialsof <strong>GAD</strong>65 have been completed. In <strong>the</strong> absence of adverseevents among <strong>the</strong> <strong>GAD</strong>65 auto<strong><strong>anti</strong>body</strong> positive Type 2 diabetespatients (so-called LADA patients or LatentAutoimmune Diabetes in <strong>the</strong> Adult) it may now be possibleto move into Phase III clinical trials to test whe<strong>the</strong>r<strong>GAD</strong>65 SIT is efficacious.The future approach to determine whe<strong>the</strong>r <strong>GAD</strong>65 SIT1994Researchers showthat transplantedislets survivelonger if <strong>the</strong>y arepretreated <strong>with</strong><strong>GAD</strong>1998Researcherstransfer diabetesbetween animals<strong>with</strong> <strong>GAD</strong>-reactiveT cellsStart ofPhase Iclinical trial<strong>Diamyd</strong> Phase Iclinical trial in 24healthyindividualscompleted<strong>Diamyd</strong> Medicalinitiatesdevelopment of a<strong>GAD</strong> baseddiabetes vaccine1996Researchers showthat progressionto T1DM isassociated <strong>with</strong> achange in <strong>GAD</strong>Aisotypes1999Researchers showthat ß-cellexpression of <strong>GAD</strong>is required fordevelopment ofT1DM in <strong>the</strong> NODmouse2000Start of Phase IIclinical trial


is truly inducing immune tolerance, deviation, or both, is tocarefully move from adult patients who already have diabetessuch as LADA patients to new onset adult T1DM patientsand <strong>the</strong>n possibly to triple auto<strong><strong>anti</strong>body</strong> positive firstdegree relatives since our ability to predict disease in suchsubjects is now well documented thanks to <strong>the</strong> DPT-1study <strong>with</strong> parenteral insulin administration (9). Theattempts to approach prevention in <strong>the</strong> fashion modeled inanimals will by necessity require a carefully staked outpathway. Novel observations are particularly tantalizing asto <strong>the</strong> possibility to use <strong>GAD</strong>65 SIT and cause no harm inhealthy subjects.The beta cell expression of <strong>GAD</strong>65 in human beta cells iswell established. There is a lack of qualitative and qu<strong>anti</strong>tativestudies on <strong>GAD</strong>65 in <strong>the</strong> non-insulin producing endocrineislet cells. Also, we still do not fully understand <strong>the</strong> roleof GABA for normal beta cell function. Recent observationssuggest that increased body mass index (BMI) is associated<strong>with</strong> <strong>GAD</strong>65 auto<strong>anti</strong>bodies in subjects not developing diabetes(10, 11). It will be critical to find out if subclinical<strong>GAD</strong>65 autoimmunity is related to obesity phenotypes arisingbecause of altered beta cell function, insulin resistance,or both. Future studies will also require detailed studies of<strong>GAD</strong>65 gene expression in CNS areas controlling food intakesince <strong>GAD</strong>65 gene polymorphisms affecting GABA productionmay be related to morbid obesity (P. Boutin and P.Frougel, personal communication). The role of <strong>GAD</strong>65 genepolymorphism in food intake regulation, obesity developmentand beta cell function may <strong>the</strong>refore need to be fur<strong>the</strong>rstudied before we can embark on large scale <strong>GAD</strong>65SIT.The relationship between Stiff Man Syndrome and <strong>GAD</strong>autoimmunity was critical to <strong>the</strong> demonstration that T1DMsera immunoprecipitated 64K protein <strong>with</strong> <strong>GAD</strong> enzymeactivity (12). We have now learned that <strong>the</strong> immune responseto <strong>GAD</strong>65 in SMS is qualitatively and qu<strong>anti</strong>tatively diffferentfrom T1DM and <strong>the</strong> approach to prevent ei<strong>the</strong>r SMS,T1DM, or both may require different<strong>GAD</strong>65 SIT. One approach may be tobase <strong>the</strong> SIT on <strong>the</strong> HLA association<strong>with</strong> <strong>the</strong> disease. In <strong>the</strong> InsulinAutoimmune Syndrome <strong>the</strong> auto<strong><strong>anti</strong>body</strong>formation is strongly associated<strong>with</strong> HLA DRB1*0401. This haplotypeis insufficient for <strong>the</strong> association <strong>with</strong>insulin auto<strong>anti</strong>bodies in T1DM, whichis ra<strong>the</strong>r associated <strong>with</strong> HLADQA1*0501-B1*0201. Hence insulin aswell as <strong>GAD</strong>65 is <strong>the</strong> auto<strong>anti</strong>gen ofmore than one disorder of autoimmunecharacter. Future studies will needto take <strong>the</strong>se similarities and differencesinto account to design interventiontrials that may lead to <strong>the</strong> identificationof novel approaches to prevent orreverse auto<strong>anti</strong>gen-specific autoimmunity.Don’t sit around, <strong>GAD</strong> back to<strong>the</strong> future…References1. Peakman M, et al,Characterization of preparations of <strong>GAD</strong>65, proinsulin, and <strong>the</strong> islet tyrosinephosphatase IA-2 for use in detection of autoreactive T cells in Type 1 diabetes:report of phase II of <strong>the</strong> Second International Immunology of Diabetes SocietyWorkshop for Standardization of T cell assays in Type 1 diabetes.Diabetes 50:1749-1754, 2001.2. Mire-Sluis AR, et al,The World Health Organization International Collaborative Study for Islet CellAntibodies.Diabetologia 43:1282-1292., 2003. Karlsen AE, et al,Cloning and primary structure of a human islet isoform of glutamic acid decarboxylasefrom chromosome 10.Proc Natl Acad Sci US 88:8337-8341, 1991.4. Kaufman DJ, et al,Autoimmunity to two forms of glutamate decarboxylase in Insulin-dependentdiabetes mellitus.J. Clin. Invest. 89:283-292, 19925. Kaufman DL, et al,Spontaneous loss of T cell tolerance to glutamic acid decarboxylase in murineinsulin-dependent diabetes.Nature 366:69-72, 1993.6. Tisch R, et al,Immune response to glutamic acid decarboxylase correlates <strong>with</strong> insulitis in nonobesediabetic mice.Nature 366:72-75, 1993.7. Bieg S, et al,<strong>GAD</strong>65 and insulin B chain peptide (9-23) are not primary auto<strong>anti</strong>gens in <strong>the</strong>Type 1 diabetes syndrome of <strong>the</strong> BB rat.Autoimmunity 31:15-24, 1999.8. Plesner A, et al,Immunization of diabetes-prone or non-diabetes-prone mice <strong>with</strong> <strong>GAD</strong>65 doesnot induce diabetes or islet cell pathology.J Autoimmun 11:335-341, 1998.9. DPT-1: Effects of insulin in relatives of patients <strong>with</strong> Type 1 diabetes mellitus. NEngl J Med 346:1685-1691, 2002.10. Rolandsson O, et al,Levels of glutamate decarboxylase (<strong>GAD</strong>65) and tyrosine phosphatase-like protein(IA-2) auto<strong>anti</strong>bodies in <strong>the</strong> general population are related to glucose intoleranceand body mass index.Diabetologia 42:555-559, 1999.11. Weets I, et al,Male-to-female excess in diabetes diagnosed in early adulthood is not specific for<strong>the</strong> immune-mediated form nor is it HLA-DQ restricted: possible relation toincreased body mass index.Diabetologia 44:40-47, 2001.12. Baekkeskov S, et al,Identification of <strong>the</strong> 64K auto<strong>anti</strong>gen in insulin-dependent diabetes as <strong>the</strong> GABAsyn<strong>the</strong>sizingenzyme glutamic acid decarboxylase.Nature 347:151-156, 1990.Researchers preventT1DM in <strong>the</strong>NOD mouse byvaccination <strong>with</strong><strong>GAD</strong>-plasmid DNAScientists prevent T1DM in<strong>the</strong> NOD mouse <strong>with</strong> <strong>GAD</strong>genemodified Vaccinia virusResearchers report that<strong>GAD</strong> gene polymorphismmay be associated <strong>with</strong>obesity200120022003ADA says in its Clinical PracticeRecommendations for 2002 that“auto<strong><strong>anti</strong>body</strong> tests may be of value toidentify which newly diagnosedpatients have immune-mediated Type 1-diabetes in circumstances where it is notobvious".Resarchers successfullyuse <strong>GAD</strong> gene <strong>the</strong>rapy fortreatment of Parkinson’sdisease in an animalmodel<strong>Diamyd</strong> Phase IIclinical trial in47 LADApatientscompleted


100<strong>GAD</strong> in Graphs<strong>GAD</strong>65 DNA vaccination preventsdiabetes in NOD micediabetic mice (in percent)90ppInsulin (n=25)80 ICA69+<strong>GAD</strong>65+ppIns (n=14)control (untreated) (n=14)70 control (vector) (n=12)<strong>GAD</strong>65 (n=14)6050403020DNA vaccination1000 10 20 30age (in weeks)Karges et al, Diabetes 51:3237; 2002Proliferation (cpm)• 4-6 week old female NOD• Different DNA vaccinations• Insulin construct does not protectfrom diabetes• <strong>GAD</strong>65 construct preventsdiabetes• Combining constructs abolishes<strong>GAD</strong>65-induced protectionBaculovirus-encoded recombinant<strong>GAD</strong> outperforms bacterial andyeast <strong>GAD</strong> preparations in T cellproliferation assays100000100001000baculovirusyeast100bacterial1 10Peakman et al (2002) Characterization of preparationsof <strong>GAD</strong>65, proinsulin and IA-2 for use indetection of autoreactive T-celsl in Type 1 diabetes.Diabetes 50:1749-1754• T cell recall immune responseswere studied by proliferation assaysusing defined T cell lines and clonesand different preparations of <strong>GAD</strong>65• Two independently produced<strong>GAD</strong>65 preparations expressed bybaculovirus gave higher proliferationindices than ei<strong>the</strong>r yeast- orbacterial-produced <strong>GAD</strong>65Untangling <strong>the</strong> <strong>GAD</strong>sAllan TobinMy interest in GABA and <strong>GAD</strong> came from my interest in Huntington’s disease – adegenerative neurological disorder that had killed <strong>the</strong> mo<strong>the</strong>r of two of my friends.I decided in <strong>the</strong> early 1980s that <strong>GAD</strong> was a reasonable “candidate gene” forHuntington’s disease, and I set out to isolate it.The time was ripe for gene isolationsince new molecular techniques were emerging every month – both new ways ofmaking collections of genes and new methods for screening <strong>the</strong>m for genes of interest.Just at that time, Dan Kaufman, whohad worked in my laboratory as aUCLA undergraduate and <strong>the</strong>nmoved to Berkeley to do graduatework, decided to move back to LosAngeles, to rejoin my laboratory, andto work on <strong>the</strong> brain.Never suspecting that I would everend up contributing to that field, I counseled Danto work on a more tractable problem – <strong>the</strong> isolationof <strong>the</strong> <strong>GAD</strong> gene. At <strong>the</strong> time, few neuroscientistswere using molecular techniques, and findingthat gene would undoubtedly make a significantcontribution to <strong>the</strong> field. Even ifHuntington’s disease is not caused by a mutationin <strong>GAD</strong>, I reasoned, having <strong>the</strong> gene in handwould accelerate progress in understanding <strong>the</strong>inhibitory circuits that are important in normalbrain function and that fail in Huntington’s disease.Dan quickly succeeded in finding <strong>the</strong> <strong>GAD</strong> gene(which we now call <strong>GAD</strong>67), using a combinationof immunological, molecular, and biochemical techniques.Pure luck – <strong>the</strong> ability of bacteria to makea functional <strong>GAD</strong> – allowed us to short circuitmost of <strong>the</strong> slogging that we thought would benecessary to prove we had <strong>the</strong> right gene. But <strong>the</strong>rewere some anomalies that later caused ano<strong>the</strong>r graduatestudent, Mark Erlander, to challenge our initialview that “<strong>the</strong>re is only one <strong>GAD</strong>”. Using a newset of techniques, in <strong>the</strong> early days of PCR (<strong>the</strong>polymerase chain reaction), Mark found ano<strong>the</strong>r<strong>GAD</strong> gene, which we now call <strong>GAD</strong>65.In 1990, Dan Kaufman, <strong>the</strong>n a postdoctoral felllowat <strong>the</strong> Salk Institute, again returned to <strong>the</strong>lab, again interested in autoimmune disease. On<strong>the</strong> basis of a paper by Pietro Di Camilli on StiffMan Syndrome – a rare complication of diabetesand of o<strong>the</strong>r conditions, Dan suspected that<strong>GAD</strong>65 or <strong>GAD</strong>67 might be identical to <strong>the</strong>unnamed 64 kD <strong>anti</strong>gen first identified by ÅkeLernmark in 1982 as <strong>the</strong> first target of Type 1(T1D) <strong>anti</strong>bodies. About <strong>the</strong> same time, SteinunnBaekkeskov, Pietro Di Camilli, and <strong>the</strong>ir colleaguescame to a similar conclusion though <strong>the</strong>y didn’tknow about <strong>GAD</strong>65, which we had onlyrecently discovered. In collaboration <strong>with</strong> NoelMaclaren and Mark Atkinson at <strong>the</strong> Universityof Florida, we examined <strong>GAD</strong> autoimmunity inT1D patients, using <strong>GAD</strong>65 and <strong>GAD</strong>67 that wecould produce from recombinant DNA.Auto<strong>anti</strong>bodies to <strong>GAD</strong> indeed provided anpage 14dmccad june 2003


“our real excitement came from a thoroughly unexpected(and still unexplained) finding, that part of <strong>GAD</strong>65 showed astriking similarity to a piece of Coxsackievirus, a virus”that isepidemiologically associated <strong>with</strong> Type 1 diabeteseffective diagnostic or predictive test for T1D.While auto<strong>anti</strong>bodies are hallmarks of <strong>the</strong> autoimmuneprocess and are valuable prediagnosticmarkers, autoreactive T cells, ra<strong>the</strong>r than auto<strong>anti</strong>bodies,are likely to be <strong>the</strong> underlying cause ofT1D. In fur<strong>the</strong>r collaboration <strong>with</strong> Maclaren andAtkinson, we showed that newly diagnosed T1Dpatients, in contrast to healthy controls, have significantT cell responses to purified recombinanthuman <strong>GAD</strong>65.We knew that <strong>the</strong>se discoveries would be usefuland important, but our real excitement came froma thoroughly unexpected finding – that part of<strong>GAD</strong>65 showed a striking similarity to a piece ofCoxsackie virus a virus that is epidemiologicallyassociated <strong>with</strong> T1D. This sequence match sugggesteda possible mechanism for <strong>the</strong> initiation ofan immune response. We thought that T1D mightinvolve “molecular mimicry,” in which <strong>the</strong>Coxsackie viral <strong>anti</strong>gen triggered an immuneresponse that cross-reacted <strong>with</strong> <strong>GAD</strong>. With thishighly speculative hypo<strong>the</strong>sis of molecular mimicryin mind, we set about to look for evidencethat <strong>GAD</strong> autoimmunity might actually be pathogenicra<strong>the</strong>r than merely diagnostic in T1D.To test <strong>the</strong> hypo<strong>the</strong>sis that an autoimmuneresponse to<strong>GAD</strong>65 initiatesautoimmunity,DanKaufman andour o<strong>the</strong>rUCLA colleaguesturned to<strong>the</strong> non-obesediabetic mouse(NOD mice).Allan Tobin, Ph.D., is aProfessor and Directorof <strong>the</strong> Brain ResearchInstitute at UCLA, LosAngeles. Tobin is alsoScientific Director of <strong>the</strong>Hereditary DiseaseFoundation, which participatedin <strong>the</strong> identification of <strong>the</strong> gene thatcauses Huntington´s disease. Tobin has specializedin <strong>the</strong> use of molecular methods for syn<strong>the</strong>sis,function and breaking down of GABA.GABA provides a significant inhibitory signal in<strong>the</strong> brain and <strong>the</strong> pancreas.Early treatment <strong>with</strong> <strong>GAD</strong>65 induced tolerancenot only to <strong>GAD</strong>65 itself, as expected, but alsoprevented <strong>the</strong> development of autoimmunity too<strong>the</strong>r ß cell <strong>anti</strong>gens as well.Still more amazingly, mice made tolerant to<strong>GAD</strong>65 never developed insulitis or diabetes. Thiswas our first strong indication that <strong>GAD</strong>-specificautoimmunity may be important in <strong>the</strong> pathogenesisof T1D. This finding, toge<strong>the</strong>r <strong>with</strong> subsequentwork by Dan Kaufman and o<strong>the</strong>rs, was <strong>the</strong>basis for <strong>the</strong> clinical trial now being conductedby diamyd medical.dmccad june 2003page 15


In Nature, Anything that CanHappen Does HappenDaniel Kaufman, Ph.D.,is a Professor, active<strong>with</strong>in <strong>the</strong> departmentof Molecular andMedical Pharmacologyat <strong>the</strong> UCLA School ofMedicine in Los Angeles.In a paper in November1993, Kaufman demonstrated that <strong>the</strong> administrationof <strong>GAD</strong> to mice that would o<strong>the</strong>rwisedevelop insulin-dependent diabetes prevented<strong>the</strong> outbreak of this disorder.Kaufman was also a member of <strong>the</strong> groupassociated <strong>with</strong> Allan Tobin, which was <strong>the</strong>first to submit a patent application for <strong>the</strong>full cDNA code for <strong>GAD</strong>, <strong>the</strong> application that<strong>Diamyd</strong> Medical licenses. Kaufman’s sphereof interest is focused on <strong>GAD</strong> and its relationto diabetes.IDan Kaufmandid not want to clone <strong>GAD</strong>. It was <strong>the</strong>mid-1980’s, and I was first year graduatestudent of Allan Tobin at UCLA.New cloning technologies had just beendeveloped that could allow neurobiologiststo clone rare brain mRNAs. Allanand I thought that some of <strong>the</strong> most importantgenes in brain development and disease would beneurotransmitter receptors and neurotransmittersyn<strong>the</strong>sizing proteins. I wanted to use <strong>anti</strong>bodiesto screen brain cDNA expression libraries forGABA receptors, or use serum from multiple sclerosispatients to identify <strong>the</strong> protein targets of<strong>the</strong>ir autoimmune response. Allan, however, knewthat <strong>the</strong>re was a good <strong><strong>anti</strong>body</strong> available against<strong>GAD</strong> (developed by Oertel) and instructed me togo after <strong>GAD</strong>.I constructed and screened a brain cDNAexpression library and quickly isolated one immunoreactiveclone. In those days, <strong>the</strong>re was greatcontention about <strong>the</strong> molecular weight of <strong>GAD</strong>,so I tested <strong>the</strong> putative <strong>GAD</strong> clone for its <strong>GAD</strong>enzymatic activity. Luckily, <strong>the</strong> one immunoreactiveclone I had isolated encoded an enzymaticallyactive protein when expressed in E. coli. Ourreport describing <strong>the</strong> cloning of <strong>GAD</strong> (Science,1986) showed that expression libraries could bescreened for functional activity, led to a greater ofunderstanding <strong>GAD</strong> biochemistry, and providedneuroscientists <strong>with</strong> a cDNA probe for studying<strong>GAD</strong> and GABA in brain development and disease.My discussions <strong>with</strong> a new graduate student in<strong>the</strong> Tobin lab, Mark Erlander, led us to suspect thatano<strong>the</strong>r <strong>GAD</strong> gene existed, despite our failure todetect ano<strong>the</strong>r gene by low stringency screening.Using degenerate PCR techniques, Mark was <strong>the</strong>first to isolate <strong>the</strong> second <strong>GAD</strong> gene, which encodeda 65 kD protein (<strong>GAD</strong>65). The clone that I had isolatedwas renamed <strong>GAD</strong>67, based on its molecularweight.Using recombinantly produced <strong>GAD</strong>67 protein,I next generated a <strong>GAD</strong>67-specific <strong><strong>anti</strong>body</strong>. This<strong><strong>anti</strong>body</strong> has been widely used by neuroscientiststo study GABAergic neurons. Using this <strong><strong>anti</strong>body</strong>toge<strong>the</strong>r <strong>with</strong> a <strong>GAD</strong>65-specific <strong><strong>anti</strong>body</strong>, I showedfor <strong>the</strong> first time that while <strong>GAD</strong>67 is distributedthroughout <strong>the</strong> neuron, <strong>GAD</strong>65 is localizedin <strong>the</strong> nerve axon terminals. While most of<strong>GAD</strong>67 is saturated <strong>with</strong> its cofactor, less thanhalf of <strong>GAD</strong>65 exists as active holoenzyme. Wesuggested that <strong>the</strong> modulation of <strong>GAD</strong>65apo/holoenzyme levels in nerve terminals couldcouple GABA production to neuronal activity (J.Neurochem, 1991).My change in course to study Type I diabetes(T1D) resulted from my car breaking down. I was<strong>the</strong>n a postdoc at <strong>the</strong> Salk Institute in San Diego.After working in <strong>the</strong> lab and finding that my carwouldn’t start, I decided to pass time in <strong>the</strong> library.I picked up <strong>the</strong> latest issue of Lancet, and it fellopen to a report entitled “64,000 Mr auto<strong>anti</strong>bodiesas predictors of insulin-dependent diabetes” byMark Atkinson and colleagues. A <strong>the</strong>n littleknown factlet came to mind that besides <strong>the</strong> brain,<strong>GAD</strong> was also expressed in <strong>the</strong> cells that madeinsulin. From my work in <strong>the</strong> Tobin lab, I knewthat <strong>the</strong>re were two forms of <strong>GAD</strong>, both of whichcould be candidates for this 64K auto<strong>anti</strong>gen.I learned that <strong>the</strong> 64K auto<strong>anti</strong>gen was a longsought after ß-cell <strong>anti</strong>gen described by SteinunnBaekkeskov and Åke Lernmark. In addition,Michele Solimena and Pietro De Camilli had justreported an association between <strong>GAD</strong> autoimmunityand Stiffman syndrome. However, <strong>the</strong>y alsoreported that <strong>the</strong>y did not detect <strong>GAD</strong> autoimmmunityin T1D patients, which in retrospect, wasmissed only because <strong>the</strong> <strong>GAD</strong> <strong>anti</strong>bodies in T1Dpatients could not recognize <strong>the</strong> denatured <strong>GAD</strong>epitopes in <strong>the</strong>ir assay system. Despite this reportedlack of association, I decided that it wasworth a try to test whe<strong>the</strong>r T1D sera could recognizerecombinant <strong>GAD</strong>67 or <strong>GAD</strong>65. This wouldbe an easy experiment using <strong>the</strong> recombinant<strong>GAD</strong>s available in <strong>the</strong> Tobin lab. I was again fortunate,since at this time, <strong>the</strong> existence of two<strong>GAD</strong>s was only known, and <strong>the</strong>ir cDNAs onlyavailable, in <strong>the</strong> Tobin lab.I got permission from Allan to do an immunoprecipitationexperiment in his lab over <strong>the</strong> nextweekend. I got T1D sera from Michael Clare-Salzler, and later, from Mark Atkinson and NoelMaclaren. Using <strong>the</strong> recombinant <strong>GAD</strong>s, I quicklypage 16 dmccad june 2003


found that sera from T1D patients contained <strong>GAD</strong>auto<strong>anti</strong>bodies that were primarily directed against<strong>GAD</strong>65. Using recombinantly produced fragmentsof <strong>GAD</strong>65, I was able to map some of <strong>the</strong> auto<strong><strong>anti</strong>body</strong>epitope recognition patterns. These were <strong>the</strong>first T1D diagnostics based on using recombinant<strong>GAD</strong>s. I had also read about an epidemiologicalassociation between Coxsackie virus and T1D.When we compared Mark Erlander’s <strong>GAD</strong>65 DNAsequence <strong>with</strong> o<strong>the</strong>r sequences in <strong>the</strong> DNA sequencedata banks, we found a large sequence similarity<strong>with</strong> a Coxsackie virus. This was a very provocativefinding, which could explain how ß-cell autoimmunitywas started, or augmented.We submitted a manuscript <strong>with</strong> our findingsto Science essentially at <strong>the</strong> same time asSteinunn Baekkeskov and colleagues submitted<strong>the</strong>ir findings to Nature. Unfortunately, a reviewerinsisted that we provide fur<strong>the</strong>r proof of<strong>GAD</strong>-Coxsackie virus molecular mimicry beforeit could be published. Consequently, we were notable to publish our work until after that ofBaekkeskov and colleagues. While very disappointingat <strong>the</strong> time, this may have been in a way fortuitous– I had just become a new AssistantProfessor at UCLA, and it prompted me to goafter <strong>the</strong> next major question – what was <strong>the</strong> roleof <strong>GAD</strong> in T1D?As T1D is thought to be T cell, and not auto<strong><strong>anti</strong>body</strong>mediated, it was crucial to understand Tcell responses to ß-cell <strong>anti</strong>gens. I was again extremelyfortunate to have Jide Tian join my newlab, who was highly knowledgeable and skilled incellular immunology. Using <strong>the</strong> NOD mousemodel of T1D, Jide found that a Th1-type responsefirst arose to <strong>GAD</strong>65, and <strong>the</strong>n spread intraandinter-molecularly to o<strong>the</strong>r ß-cell <strong>anti</strong>gens. Jidetreated very young NOD mice <strong>with</strong> <strong>GAD</strong>65 oro<strong>the</strong>r ß-cell <strong>anti</strong>gens in a way that inactivatedreactive T cells. The mice that were tolerized to<strong>GAD</strong>65 had no autoimmune responses or insulitis,while those tolerized to o<strong>the</strong>r <strong>anti</strong>gens did displayß-cell autoreactivity (Nature, 1993). Thus,<strong>the</strong> early inactivation of <strong>GAD</strong>65 reactive T cellscould circumvent <strong>the</strong> development of autoimmunity,qualifying <strong>GAD</strong>65 as a key <strong>anti</strong>gen in <strong>the</strong>induction of murine T1D.We next examined what could be done to inhibitan autoimmune process after it had alreadystarted. Jide showed that injecting <strong>GAD</strong>65 in anadjuvant that induced Th2 responses to <strong>GAD</strong>65could greatly inhibit <strong>the</strong> progression of <strong>the</strong> autoimmuneprocess in NOD mice that already had“These and subsequent experiments suggested that <strong>the</strong> greaterprotective effect of <strong>GAD</strong>65 was due to its greater abilityto induce Th2 responses. It is <strong>the</strong>se experiments that”led to <strong>the</strong> <strong>GAD</strong>65vaccine that <strong>Diamyd</strong> is testing in clinical trialsan established autoimmune process. Moreover,<strong>GAD</strong>65 vaccination could also protect transplantedislets in diabetic mice, significantly betterthan <strong>the</strong> o<strong>the</strong>r ß-cell auto<strong>anti</strong>gens (Nature-Medicine, 1996). These and subsequent experimentssuggested that <strong>the</strong> greater protective effectof <strong>GAD</strong>65 was due to its greater ability to induceTh2 responses. It is <strong>the</strong>se experiments that led to<strong>the</strong> <strong>GAD</strong>65 vaccine that diamyd is testing in clinicaltrails.We also began to examine <strong>the</strong> possibility of Tcell cross-reactivity between <strong>GAD</strong> and Coxsackievirus. Jide showed that T cell cross-reactivity didoccur, both at <strong>the</strong> peptide level and at <strong>the</strong> wholeprotein level between <strong>GAD</strong>65 and <strong>the</strong> Coxsackievirus protein – but only in mice <strong>with</strong> <strong>the</strong> diabetessusceptibly MHC II allele, and not in mice <strong>with</strong>o<strong>the</strong>r MHC II alleles (J. Exp. Med, 1994). Thejury is still out as to whe<strong>the</strong>r <strong>the</strong> epidemiologicalassociation of Coxsackie virus <strong>with</strong> T1D is due to<strong>the</strong> ability of Coxsackie virus to infect islet cells,or due to T cell cross-reactivity <strong>with</strong> <strong>GAD</strong>. As itis generally found that in nature, anything thatcan happen does happen, it would not be surprisingto find that both mechanisms can occur, andin some cases, initiate or augment ß-cell autoimmmunity.It has been most gratifying to see our work at<strong>the</strong> bench contribute to new diagnostics andpotential treatments for diabetes, as well as neurologicaldiseases. Our work has been highly dependenton <strong>the</strong> support of Allan Tobin, as well ascontributions by Jide Tian, Michael Clare-Salzler,Mark Atkinson and Paul Lehmann – who havealso had <strong>the</strong> patience of saints in teaching meimmunology.dmccad june 2003page 17


References1. Baekkeskov, S., et al,Auto<strong>anti</strong>bodies in newly diagnosed diabetic childrenimmunoprecipitate specific human pancreatic islet cellproteins.Nature 298, 167-169 (1982).2. Baekkeskov, S., et al,Antibodies to a Mr 64,000 human islet cell <strong>anti</strong>gen precede<strong>the</strong> clinical onset of insulin-dependent diabetes. J.Clin. Invest. 79, 926-934 (1987).3. Baekkeskov, S., et al,Revelation of specificity of 64k auto<strong>anti</strong>bodies in IDDMserums by high-resolution 2D-gel electrophoresis.Unambiguous identification of 64k target <strong>anti</strong>gen.Diabetes 38, 1133-1141 (1989).4. Christgau, S., et al,Pancreatic ß-cells express two auto<strong>anti</strong>genic forms of glutamicacid decarboxylase, a 65kDa hydrophilic form anda 64kDa amphiphilic form which can be both membrane-boundand soluble.J. Biol. Chem. 266, 21257-21264 (1991).5. Baekkeskov, S., et al,Identification of <strong>the</strong> 64k auto<strong>anti</strong>gen in insulin-dependentdiabetes as <strong>the</strong> GABA-syn<strong>the</strong>sizing enzyme glutamicacid decarboxylase.Nature 347, 151-156 (1990).6. Kim, J., et al,Higher auto<strong><strong>anti</strong>body</strong> levels and recognition of a linearN-terminal epitope in <strong>the</strong> auto<strong>anti</strong>gen <strong>GAD</strong>65 distinguishstiff-man syndrome from insulin dependent diabetes.J. Exp. Med. 180, 595-606 (1994).7. Kanaani, J., et al,A combination of three distinct trafficking signals mediatesaxonal targeting and presynaptic clustering of <strong>GAD</strong>65.J. Cell Biol. 158, 1229-1238. (2002).8. 8. Kash, S. F., et al,Increased anxiety and altered responses to anxiolytics inmice deficient in <strong>the</strong> 65 kDa isoform of glutamic aciddecarboxylase.Proc. Natl. Acad. Sci. 96, 1698-1703 (1999).9. Shi, Y., et al,Increased expression of glutamic acid decarboxylase andGABA in pancreatic b-cells impairs glucose-stimulatedinsulin secretion at a step proximal to membrane depolarization.Am. J. Physiol. Endocrinol. Metab. 279, 684-694, (2000).10. Schwartz, H., et al,High resolution epitope mapping and structural modelingof human glutamic acid decarboxylase 65.J. Mol. Biol. 287, 983-999 (1999).11. Jaume, J. C., et al,Suppressive effect of <strong>GAD</strong>65-specific autoimmune Blymphocytes on processing of T cell determinants located<strong>with</strong>in <strong>the</strong> <strong><strong>anti</strong>body</strong> epitope.J. Immunology 169, 665-672, (2002).Contributions to<strong>the</strong> <strong>GAD</strong>65 FieldISteinunn Baekkeskovn <strong>the</strong> 1960’’s and 1970’s several investigatorsdiscovered evidence to show thatinsulin dependent diabetes mellitus(Type 1 diabetes or T1D) is an autoimmmunedisease characterized by circulatingauto<strong>anti</strong>bodies to <strong>the</strong> β cell in humanpancreatic islets and infiltration of lymphocytesinto <strong>the</strong> islets. However, <strong>the</strong> target <strong>anti</strong>gen of thisautoimmunity was completely unknown, when Ijoined <strong>the</strong> Hagedorn Laboratory in Copenhagen inlate 1979 to work <strong>with</strong> Åke Lernmark. Usingexpertise I had gained in studying target <strong>anti</strong>gensin trypanosomes, I developed a method that alloweda very sensitive detection of human β cellproteins that bound to circulating auto<strong>anti</strong>bodiesin <strong>the</strong> blood of patients. To our surprise, we discoveredthat auto<strong>anti</strong>bodies in <strong>the</strong> blood from 8 outof 10 diabetic children bound to <strong>the</strong> same protein,a 64kD <strong>anti</strong>gen from human islets while none of10 healthy children recognized this protein. Thiswas <strong>the</strong> very first indication that <strong>the</strong>re was a specifictarget <strong>anti</strong>gen in <strong>the</strong> autoimmune response inT1D (1). We <strong>the</strong>n showed that <strong>the</strong> auto<strong>anti</strong>bodiesto <strong>the</strong> 64kD protein can be present many yearsbefore clinical onset of T1D (2), suggesting that<strong>the</strong>se auto<strong>anti</strong>bodies can be used as a sensitivemarker to identify individuals at risk of developingT1D. Also, in all autoimmune diseases <strong>the</strong> target<strong>anti</strong>gen is of critical importance because of itspotential to be used to prevent <strong>the</strong> autoimmunedisease. Having established my own laboratoryfirst at <strong>the</strong> Hagedorn Research Laboratory andlater at <strong>the</strong> University of California, San Francisco,we developed amethod for partiallypurifying<strong>the</strong> 64kD protein(2-3). Wecharacterizedimportantbiochemicaland biophysicalparameters of<strong>the</strong> <strong>anti</strong>gen (2-4) in a painstakingeffort thatculminated inits identificationas <strong>the</strong>Steinunn Baekkeskov,Ph.D., is a Professor at<strong>the</strong> University ofCalifornia. Baekkeskov´scurrent research projectsfocus on four mainareas: i) structure,function, and cell biologyof <strong>GAD</strong>65, IA-2 and glima 38; ii) characterizationof disease specific B-cell epitopesin <strong>GAD</strong>65, and IA2, and <strong>the</strong> temporalpattern of <strong>the</strong>ir recognition during early andlate phases of §-cell destruction; iii) characterizationof autoimmune T cell epitopes in<strong>GAD</strong>65 and IA2, and how <strong>the</strong>y can be used toinduce apoptosis in autoimmune T cells during<strong>the</strong> preclinical phase to prevent disease; iv)and mechanisms of transplantation tolerance,and development of methods to preventallo- as well as autoimmune destruction ofislet cell transplants.smaller isoform of <strong>the</strong> GABA-syn<strong>the</strong>sizing enzymeglutamic acid decarboxylase or <strong>GAD</strong>65 (5). Theidentification of a component of <strong>the</strong> 64kD auto<strong>anti</strong>genas <strong>GAD</strong>65 transformed <strong>the</strong> field ofImmunology of Diabetes, because it became possibleto use recombinant protein for developmentof auto<strong><strong>anti</strong>body</strong> assays and for studies of autoimmunemechanisms in <strong>the</strong> NOD mouse. A fewyears later, a second component of <strong>the</strong> 64kD auto<strong>anti</strong>gen,left behind by our purification method,was identified as <strong>the</strong> protein IA-2 by Michaelpage 18 dmccad june 2003


<strong>GAD</strong>65 moleculeChristie, my first postdoctoral fellow, in his ownlab. Toge<strong>the</strong>r, <strong>GAD</strong>65 and IA-2, which are expresssedin both ß cells and neurons, are recognized by80-90% of T1D patients. In neurons, <strong>GAD</strong>65 isalso a target of autoimmunity in a rare neurologicaldisorder Stiff-man syndrome (6).Following <strong>the</strong> discovery of <strong>GAD</strong>65 as a major targetauto<strong>anti</strong>gen in T1D (5), my lab has focused onunderstanding <strong>the</strong> structure, cell biology, and functionof this molecule, and how it is recognized by<strong>the</strong> immune system. The subcellular trafficking of<strong>GAD</strong>65 is <strong>the</strong> parameter that most clearly distinguishesit from <strong>the</strong> highly homologous <strong>GAD</strong> isoform,<strong>GAD</strong>67, and is dependent on unique trafficking signalsin <strong>the</strong> N-terminal region (7 and refs <strong>the</strong>rein).By knocking-out <strong>GAD</strong>65 in <strong>the</strong> mouse, we showedthat GABA generated by <strong>GAD</strong>65 is involved infine tuning of inhibitory neurotransmission inresponse to a variety of environmental stimuli (8and refs <strong>the</strong>rein), while overexpression of <strong>the</strong> proteinin β cells revealed a role of GABA in negativeregulation of first phase insulin secretion (9). Finemapping of <strong>the</strong> autoimmune epitopes recognizedby <strong>GAD</strong>65 auto<strong>anti</strong>bodies in human patients revealedthat <strong>the</strong>y target almost <strong>the</strong> entire surface of <strong>the</strong>molecule (10 and refs <strong>the</strong>rein) and led to <strong>the</strong> firstThe figure is printed <strong>with</strong> permission from Academic Press (Journal of Molecular Biology, Vol.287,No 5, April 16, 1999, pp. 983-999, Baekkeskov S, Schwartz HL).3D model of <strong>the</strong> <strong>GAD</strong>65 dimer (10). This structuralinformation enabled us to show how <strong>the</strong> epitopespecificity of autoimmune B cells, a critical playerin <strong>GAD</strong>65 presentation to T cells and developmentof T1D, influences <strong>the</strong> autoimmune T cellepitope repertoire in <strong>the</strong> protein (11).dmccad june 2003 page 19


<strong>GAD</strong> in GraphsIncidence of diabetes (%)10080604020In Type 1 diabetes patients, <strong>GAD</strong>65-reactive T cells are primarily memoryT cellsAnti–B7-1[ 3 H] Thymidine604020(%) Inhibition0IFN-γ6040200IL-136040200<strong>GAD</strong>65 (µg/ml)Viglietta et al (2002) <strong>GAD</strong>65-reactive T cells areactivated in patients <strong>with</strong> autoimmune Type 1adiabetes. J Clin Invest 109:895-903.• T cell recall immune responseswere studied by proliferation andcytokine assays using <strong>GAD</strong>65 andinhibition of <strong>the</strong> costimulatorymolecule B7.1• T cells from Type 1 diabetics werenot inhibited by <strong>anti</strong>-B7.1, whilethose from controls were. This indicatesthat <strong>the</strong> Type 1 diabetic T cellsare of a memory T cell phenotype.Treatment of young NOD mice <strong>with</strong>rrVV-<strong>GAD</strong>65 prevents diabetesdevelopmentrVV-<strong>GAD</strong>65rVV-MJ601rVV-NS1Uninjected010 20 30age (in weeks)Sun et al. (2002). Prevention of autoimmunediabetes by immunogene <strong>the</strong>rapy using recombinantvaccinia virus expressing glutamic aciddecarboxylase.Diabetologia 45:668-676• 3 week old female NOD mice wereinjected <strong>with</strong> vaccina constructscoding for <strong>GAD</strong> or control <strong>anti</strong>gens• Mice receiving <strong>the</strong> <strong>GAD</strong>65construct were protected fromdiabetes40The <strong>GAD</strong>65 Stanford PerspectiveHugh McDevittInitial work <strong>with</strong> <strong>GAD</strong>65 identified it as one of <strong>the</strong> first to elicit a spontaneousimmune response in 3-4 week old NOD mice. Next, we used <strong>GAD</strong>65 to suppress ordelay <strong>the</strong> diabetic process. Treatment <strong>with</strong> 4 doses of <strong>GAD</strong>65 intravenously at 12weeks of age resulted in a large decrease in diabetes incidence at 30 weeks. O<strong>the</strong>rislet cell proteins, or foreign proteins had no effect.Tcell hybridomas were used toidentify immunodominantpeptide epitopes of <strong>GAD</strong> 65in NOD mice: <strong>GAD</strong>65 aminoacids 206-220, 221-235, and286-300.Transgenic mice <strong>with</strong> T Cell Receptors (TCRs)for <strong>the</strong> 206 and 286 peptides, and a hybrid TCRtransgenic <strong>with</strong> <strong>the</strong> 221α chain and <strong>the</strong> 286βchain, revealed that in all 3 TCR transgenic NODlines nei<strong>the</strong>r insulitis nor Type 1 diabetes developed.T cells in all 3 lines are reactive to cognatepeptide and produce IL-2, IFNγ, IL-10, and someTNFα. The T cells are positively selected in <strong>the</strong>thymus, but only escape negative selection via <strong>the</strong>expression of a second α chain. The 206 and 286TCR transgenic lines on <strong>the</strong> Cα -/- backgroundhad near complete deletion of T cells. MHC tetrameranalysis of 286 TCR affinity of <strong>the</strong> 286 TCRshowed that it was of extremely low affinity.T cells were partially negatively selected in <strong>the</strong>thymus, and underwent fur<strong>the</strong>r negative selectionin <strong>the</strong> periphery. Only 10-20% of peripheral CD4 +T cells are 286 tetramer positive, while 30-40% ofCD8 + T cells are tetramer positive. Cell transferexperiments showed that <strong>the</strong>se mice have T cellscapable of partially suppressing, and definitelydelaying <strong>the</strong> onset of Type 1 diabetes in a standardtransfer system.To date, none of <strong>the</strong>se mice have developed diabetesor insulitis. Crosses <strong>with</strong> a RIP-Hu<strong>GAD</strong>65high expressing transgenic NOD line has not resultedin diabetes or insulitis in ei<strong>the</strong>r <strong>the</strong> 206 or 286lines. O<strong>the</strong>r studies are currently underway.However, it appears that in all 3 TCR transgenicmouse lines,<strong>GAD</strong>65 specificT cells areprotective.A review of<strong>the</strong> literatureshows thatmost <strong>GAD</strong>65-specific T cellclones ei<strong>the</strong>rdo not causediabetes orHugh McDevitt, MD, isProfessor of <strong>the</strong>Department ofMicrobiology andImmunology atStanford UniversitySchool of Medicine,<strong>with</strong> a joint appointmentin <strong>the</strong> Department of Medicine.Following his discovery of linkage of <strong>the</strong> majorhistocompatibility complex <strong>with</strong> specificimmune responses, McDevitt has pursued <strong>the</strong>mechanisms of this association over <strong>the</strong> pastseveral decades, and most recently has beenstudying <strong>the</strong> immune response of NOD miceto <strong>GAD</strong>65.insulitis, or in some cases are protective. (Oneclone, reported by Robert Sherwin, did cause Type1 diabetes.) Fur<strong>the</strong>r, von Boehmer (J. Exp. Med., inpress) has shown that induction of recessive toleranceto <strong>GAD</strong>65 <strong>with</strong> a transgene insuring highlevel <strong>GAD</strong>65 expression in endosomal compartmentsdoes not change Type 1 diabetes incidence.These results suggest that <strong>GAD</strong>65 is primarilya protective <strong>anti</strong>gen in Type 1 diabetes in NODmice. Whe<strong>the</strong>r it is also a protective <strong>anti</strong>gen inpatients is an open question. None<strong>the</strong>less, experimentsfrom our and o<strong>the</strong>r laboratories suggestthat administration of <strong>GAD</strong>65 in non-inflammatoryvehicles result in a delay in onset, or adecrease in incidence of NOD diabetes. This raises<strong>the</strong> question of whe<strong>the</strong>r <strong>GAD</strong>65 is a protectiveislet cell <strong>anti</strong>gen because of its unusual locationin ß cells (in vesicles similar to neuronalvesicles), <strong>with</strong> this presentation resulting in apreferential elicitation of a TH2 response to thisislet cell protein. This increase in TH2 T cells in<strong>the</strong> islets may <strong>the</strong>n be a factor in delaying onsetof diabetes and/or decreasing its incidence.page 20 dmccad june 2003


Islet Cell Antibody Dependent Progression to Insulin Treatment in LADA Patientsnon-insulin requiring diabetes (%)age4510090807060504030201000 1 2 3 4(years)5 6The Search for Islet AntigensTDavid Leslie and Marco Londeihe search for islet <strong>anti</strong>gens associated<strong>with</strong> <strong>the</strong> immuneresponse which precedes andpredicts <strong>the</strong> clinical onset ofType 1 diabetes received a subst<strong>anti</strong>alboost <strong>with</strong> <strong>the</strong> identificationof <strong>GAD</strong> as one <strong>the</strong> major <strong>anti</strong>gens involvedin <strong>the</strong> aberrant immune response. We used a largecohort of identical twins which had been followedprospectively for several years to show for <strong>the</strong> firsttime that in <strong>the</strong>se twins <strong>GAD</strong> <strong>anti</strong>bodies were highlypredictive of progression to clinical diabetes.Fur<strong>the</strong>r studies found that <strong>the</strong> isotypes of <strong>the</strong>se<strong>GAD</strong> <strong>anti</strong>bodies were largely restricted to a singleisotype IgG1. In contrast, patients <strong>with</strong> a rare nervedisease Stiff Man Syndrome, who also have <strong>GAD</strong><strong>anti</strong>bodies, and in whom about half <strong>the</strong> cases alsohave diabetes, were found to have a broad isotypeprofile involving <strong><strong>anti</strong>body</strong> isotypes o<strong>the</strong>r than IgG1.These observations suggested that <strong>the</strong> <strong><strong>anti</strong>body</strong>immune response to <strong>GAD</strong> in Type 1 diabetes showedmaturation at an early stage and was distinctfrom that found in Stiff Man Syndrome. We showedthat <strong>the</strong> T cell responses to <strong>GAD</strong> also differedin Type 1 diabetes as compared <strong>with</strong> normal subjectsand patients <strong>with</strong> Stiff man syndrome.Specifically, <strong>the</strong> dominant T cell response in diabeteswas to peptide epitopes (fragments) at <strong>the</strong> carboxy-“These studies, takentoge<strong>the</strong>r, suggest adistinct mature immune response to<strong>GAD</strong> involving cellular and humoralimmunity in Type 1 diabetes”terminal end of <strong>the</strong> <strong>GAD</strong> molecule. These studies,taken toge<strong>the</strong>r, suggest a distinct mature immuneresponse to <strong>GAD</strong> involving cellular and humoralimmunity in Type 1 diabetes and raised <strong>the</strong> possibilitythat modulation of that response might modify<strong>the</strong> destructive immune process associated <strong>with</strong> progressionto insulin dependence.The potential to modify that <strong>GAD</strong> immuneresponse and <strong>the</strong>reby modify <strong>the</strong> disease coursewas illustrated by o<strong>the</strong>rs in animal models and isnow being tested in patients <strong>with</strong> autoimmunediabetes. Should <strong>the</strong>se studies prove successful <strong>the</strong>next stage would be to consider intervention in atrisksubjects since <strong>GAD</strong> <strong>anti</strong>bodies, allied to o<strong>the</strong>rdisease markers, are highly predictive of diseaseprogression.David Leslie, MD,FRCP., is Professor ofDiabetes and Autoimmunityat <strong>the</strong> RoyalLondon and St.Bartholomew´s Schoolof Medicine,University of London.Leslie has been involved in diabetes researchand clinical studies since 1975. Lesliehas been Director of <strong>the</strong> British DiabeticTwin Study since 1982, <strong>the</strong> world’s largesttwin study of its type. By studying twinsProfessor Leslie has been able to show <strong>the</strong>possibilities for predicting and preventingdiabetes.Marco Londei, MD, isa Professor at <strong>the</strong>Imperial CollegeSchool of Medicine inLondon, is one ofGreat Britain's mostinternationally renownedscientists in <strong>the</strong>field of autoimmunity. Londei´s research hasbeen concentrated on T cells in autoimmunity.dmccad june 2003page 21


References1. Quinn A, et al,MHC class I-restricted determinants on <strong>the</strong> glutamicacid decarboxylase 65 molecule induce spontaneousCTL activity.J Immunol. 2001;167:1748-57.2. Bowie L, et al,Generation and maintenance of auto<strong>anti</strong>gen-specificCD8+ T cell clones isolated from NOD mice.J. Immunol. Methods. 1999;228:87-95.3. Gauvrit A, et al,Identification of a relevant <strong>GAD</strong>65 H-2Kd-restrictedepitope recognized by CD8+ T cells in NOD mice.2003. Unpublished data.4. Bach J-M, et al,High affinity presentation of an auto<strong>anti</strong>genic peptidein Type I diabetes by an HLA class II protein encodedin a haplotype protecting from disease.J. Autoimmun. 19975. Bach JM, et al,Identification of mimicry peptides based on structuralmotifs of epitopes derived from 65-kDa glutamic aciddecarboxylase.Eur. J. Immunol. 1998;28:1902-1910.6. Endl J, et al,Identification of naturally processed T cell epitopesfrom glutamic acid decarboxylase presented in <strong>the</strong> contextof HLA-Dr. alleles by T lymphocytes of recentonset IDDM patients.J. Clin. Invest. 1997;99:2405-2415.J.-M. Bach, Ph.D., D.V.M., is AssistantProfessor in Endocrinology and Director of<strong>the</strong> ImmunoendocrinologyUnit of Nantes, France. Doctor Bach hasspecialized in <strong>the</strong> study of <strong>the</strong> cellularimmune response to <strong>GAD</strong> in Type 1 diabetes(mouse, dog and human).<strong>GAD</strong> Peptide Vaccines forT1DM:Not Just a Blueprint?TJ.-M. Bachype 1 diabetes is an autoimmunedisease resulting in <strong>the</strong>destruction of pancreatic islet ß-cells, which secrete insulin, byautoreactive T lymphocytes.One of <strong>the</strong> major challenges of<strong>the</strong> new century will be <strong>the</strong> treatment of organspecificautoimmune disease. For Type 1 diabetes, itmeans disease prevention before <strong>the</strong> completedestruction of <strong>the</strong> ß-cell mass, or replacement ofinsulin-secretive cells after <strong>the</strong>ir disappearance.Pancreas and isletcell transplantation are effectiveß-cell replacement <strong>the</strong>rapy for diabetes, but <strong>the</strong>shortage of human donor pancreata has led tosearch for potential alternative sources of isleT cells(pig islets, in vitro generation of ß-cells from pancreaticduct cells or from stem cells). Beside replacementapproaches, diabetes prevention implicates<strong>the</strong> development of immuno<strong>the</strong>rapies, especiallyspecific immunomodulations of auto-<strong>anti</strong>gensinvolved in Type 1 diabetes pathogenesis.Immuno<strong>the</strong>rapies are also necessary for extensive ß-cell replacement approaches, to block <strong>the</strong> autoimmmuneprocess and to avoid strong immunosuppressortreatments. To be attractive, immuno<strong>the</strong>rapiesof Type 1 diabetes have to produce a long-lastingprotective response specific for self-<strong>anti</strong>gens.On one hand, specific modulations and tolerancecould be obtained in rodent models <strong>with</strong>whole self-proteins or DNA encoding auto-<strong>anti</strong>gens.On a second hand, syn<strong>the</strong>tic peptide-basedvaccines could be a very attractive approach forspecific immunomodulations, permitting to targetclonal elimination or unresponsiveness of aggressiveT cells. Peptides can be produced in largequ<strong>anti</strong>ties <strong>with</strong> high purity at low cost, and aresafer than recombinant proteins. To allow <strong>the</strong>design of <strong>anti</strong>gen-specific immune <strong>the</strong>rapies targetingpathogenic autoreactive T cells, we need tocharacterize peptide specificities of autoreactive Tcells, which should also provide fabulous markersof isleT cell damage, useful for <strong>the</strong>rapeutic trialsand diagnosis of individuals <strong>with</strong> increased riskfor disease, and could help to unravel <strong>the</strong> pathophysiologyof autoimmune diabetes.Several self-<strong>anti</strong>gens have been identified in Type1 diabetes. Among <strong>the</strong>m, <strong>GAD</strong> (Glutamic AcidDecarboxylase) is a crucial early target involved indiabetes in human and non-obese diabetic (NOD)mouse. Its role is considered presently as decisive inautoimmunity initiation as well as in progression toovert disease. A majority of studies of diabetes autoimmunityprocess and prevention are focused onthis auto<strong>anti</strong>gen. In rodent models, <strong>the</strong> role of apeculiar set of T lymphocytes, expressing <strong>the</strong> CD8marker, in ß-cell aggression is not clearly understood.CD8 + T cells could be implicated not only in diabetesinduction, but also in progression to destructiveinsulitis and overt diabetes. Teams of E. Sercarz (1)and A. Cooke (2) have identified two peptides derivedfrom <strong>GAD</strong> and recognized by CD8 + T lymphocytesin NOD mouse (peptide 546 and 515,respectively). CD8 + T cells specific for <strong>the</strong>se peptidesare detected early in young NOD mouse. Oursquad have very recently specific ano<strong>the</strong>r <strong>GAD</strong>derivedpeptide (peptide 90), which may be crucialin diabetes progression in mice [3]. For <strong>the</strong> firsttime, we showed that CD8 + T cells specific for a<strong>GAD</strong>-derived peptide are implicated in diabetesaggravation. Specific cytotoxic CD8 + T cells wereactivated and could play an important role in progressionof insulitis to overt disease. We presentlytest immunoprevention of spontaneous diabetes inNOD mice using CD8 + T cell-inducing peptides.And in humans? In humans, while auto<strong><strong>anti</strong>body</strong>responses have been extensively characterized,very little is known of <strong>the</strong> natural history of Tcell autoreactivities, nei<strong>the</strong>r for CD8 + T cells (probably<strong>the</strong> first T lymphocytes to colonize pancreaticislets), nor for T lymphocytes expressing CD4<strong>anti</strong>gen. We and o<strong>the</strong>rs have already identified<strong>GAD</strong>-derived peptides presented to CD4 + T lymphocytesof recent-onset diabetic patients[4-6]. Inhumans, we need also to identify relevant CD8 + Tcell-inducing peptides of <strong>GAD</strong>. In <strong>the</strong> future, discoveryof <strong>GAD</strong>-peptides recognized by self-reactiveaggressive CD8 + T lymphocytes and elucidationof key immunological events leading to diabetesinitiation or aggravation in humans may permit todefine efficient peptide-vaccine strategies.page 22 dmccad june 2003


No <strong>GAD</strong> No Type 1 Diabetes?TJi-Won Yoon, Hee-Sook Jun and Julia McFarlanehe prevention of Type 1 diabeteshas long been a primaryarea of research for our laboratory.Several ß cell auto<strong>anti</strong>genshave been implicated in <strong>the</strong>triggering of ß cell-specificautoimmunity, and <strong>GAD</strong>65 is a strong candidatein both humans and <strong>the</strong> diabetes-prone nonobesediabetic (NOD) mouse, which is one of <strong>the</strong> bestanimal models for human autoimmune diabetes.In <strong>the</strong> NOD mouse, <strong>GAD</strong>, as compared <strong>with</strong>o<strong>the</strong>r ß cell auto<strong>anti</strong>gens examined, provokes <strong>the</strong>earliest T cell proliferative response.We hypo<strong>the</strong>sized that <strong>GAD</strong> expression in <strong>the</strong> ßcells may be important for disease initiation. Toaddress this, we took a transgenic approach and selectivelysuppressed <strong>GAD</strong> expression in <strong>the</strong> ß cells ofNOD mice. We found that complete suppression of<strong>GAD</strong> expression in <strong>the</strong> ß cells of <strong>anti</strong>-sense <strong>GAD</strong>transgenic mice ([SJL x C57BL/6] F2 mice) backcrosssed<strong>with</strong> NOD mice resulted in <strong>the</strong> prevention ofautoimmune Type 1 diabetes. These results support<strong>the</strong> hypo<strong>the</strong>sis that <strong>GAD</strong> may play an important rolein <strong>the</strong> development of T cell-mediated autoimmunediabetes. However, we cannot exclude <strong>the</strong> possibilitythat diabetes-resistance genes from <strong>the</strong> founder micemay have been transmitted to <strong>the</strong> <strong>anti</strong>-sense <strong>GAD</strong>transgenic mice. To address this issue, we are presentlyexamining <strong>the</strong> development of insulitis and diabetesin ß cell-specific <strong>GAD</strong> knock-out NOD mice to confirm<strong>the</strong> role of <strong>GAD</strong> in <strong>the</strong> initiation of Type 1autoimmune diabetes.Several different approaches for immune <strong>the</strong>rapyusing <strong>GAD</strong> have been tried for <strong>the</strong> prevention ofdiabetes in animal models. We used a recombinantvaccinia virus (rVV) expressing <strong>GAD</strong> as a vaccine,as rVVs can induce humoral and cell-mediatedimmune responses to target proteins and <strong>the</strong> inducedimmune responses can be long lived. We wereable to show that administration of <strong>GAD</strong>-expresssingrVV effectively prevented autoimmune diabetesin an age- and dose-dependent manner throughactive suppression of effector T cells in NOD mice.Although <strong>the</strong> <strong>the</strong>rapeutic effect of <strong>GAD</strong>-basedimmuno<strong>the</strong>rapy is different depending on <strong>the</strong>route of administration, experimental conditions,and quality of <strong>anti</strong>gen, treatment using <strong>GAD</strong> maybe of importance <strong>with</strong> regard to future strategiesfor <strong>the</strong> prevention of Type 1 diabetes in humans.What is <strong>the</strong>Genetic Basis of T1DM?Michael Clare-SalzlerDr. Ji-Won Yoon holds aCanada Research Chairin Diabetes and is <strong>the</strong>director of <strong>the</strong> Laboratoryof Viral andImmunopathogenesis ofDiabetes in <strong>the</strong> Facultyof Medicine, Universityof Calgary. Previously, Yoon spent 10 years as asenior investigator at <strong>the</strong> National Institutesof Health. Over <strong>the</strong> past 5 years, Yoon hasbeen involved in studies on <strong>the</strong> role of glutamicacid decarboxylase (<strong>GAD</strong>) in <strong>the</strong> pathogenesisof diabetes and <strong>the</strong> development of methodsto prevent autoimmune diabetes byimmunogene <strong>the</strong>rapy using recombinant vaccciniavirus vectors expressing <strong>GAD</strong>.The goal of my research is to establish<strong>the</strong> cellular, molecular, andgenetic basis for <strong>the</strong> immunopathogenesisof Type 1 diabetes(T1D) and to develop methodsto prevent this disease. I haveconcentrated on identifying auto<strong>anti</strong>gens and determining<strong>the</strong> role of <strong>anti</strong>gen presenting cells, e.g. dendriticcells (DC), in T1D. Our publications wereamong <strong>the</strong> first defining glutamate decarboxylase(<strong>GAD</strong>) as an auto<strong>anti</strong>gen. Our subsequent studiesdemonstrated <strong>GAD</strong> administration prevented diseasein diabetes-prone NOD mice. Recently, <strong>the</strong> NIHbaseddiabetes prevention study, TrialNet, developedplans for a trial to test whe<strong>the</strong>r <strong>GAD</strong> administrationprevents T1D in humans.Michael Clare-Salzler, MD. Professor ofPathology, Immunology and LaboratoryMedicine, University of Florida, Director ofResearch and Academic affairs, research; part of<strong>the</strong> UCLA team that characterized <strong>GAD</strong> as anauto<strong>anti</strong>gen in NOD mice, demonstrated <strong>GAD</strong>responses in NOD mice, study of <strong>the</strong> role <strong>anti</strong>genpresenting cells in Type 1 diabetes, first todemonstrate a tolerogenic role for dendritic cellsin <strong>the</strong> autoimmunity of <strong>the</strong> NOD mouse, first labto demonstrate abnormal regulation Cox-2regulation and prostaglandin metabolism in Type1 diabetes.10090807060Percent Diabetic5040<strong>GAD</strong> in GraphsInduction of <strong>GAD</strong>65-specific regulatoryT cells modulates diabetes inNOD mice30alone20 + OVA T cells+ <strong>GAD</strong> T cells10010 20 30 40 50 60Days Post TransferTisch et al. (1998) Induction of <strong>GAD</strong>65-specificregulatory T cells inhibits ongoing autoimmunediabetes in nonobese diabetic miceDiabetes 47:894-899Insulitis severity• Irradiated NOD mice developdiabetes when diabetogenicsplenic cells are transferred alone orcotransfered <strong>with</strong> OVA-specific Tcells.• Cotransfer of <strong>GAD</strong>-specificregulatory T cells prevents diabetesdevelopmentImmunization <strong>with</strong> <strong>GAD</strong>65 does notinduce murine diabetesA2.01.51.00.50.0C2.01.51.00.5BALB/cB2.01.51.00.50.0D2.01.51.00.5C57B10.00.0NMRINODPlesner et al. (1998) Immunization of diabetesproneor non-diabetes-prone mice <strong>with</strong> <strong>GAD</strong>65does not induce diabetes or islet cell pathologyJ Autoimmunity 11:335-341• 11 wk old NOD mice were injectedtwice sc <strong>with</strong> 75mg <strong>GAD</strong>65 or BSA.• Non-diabetes prone mice (Balb/c,C57/Bl and NMRI) did not developdiabetes.• NOD mice injected <strong>with</strong> <strong>GAD</strong>65developed less insulitisdmccad june 2003page 23


References1. Tisch, R., et al,Induction of <strong>GAD</strong>65-specific regulatory T cells inhibitsongoing autoimmune diabetes in nonobese diabeticmice.Diabetes, 47:894, 1998.2. Tisch, R., et al,Induction of <strong>GAD</strong>65-specific Th2 cells and preventionof autoimmune diabetes at late stages of disease developmentis epitope dependent.J. Immunol. 163:1178, 1999.3. Tisch, R., et al,Antigen-specific mediated suppression of ß cell autoimmunityby plasmid DNA vaccination.J. Immunol. 166:2122, 2001.4. Seifarth, C., et al,. More stringent conditions of plasmidDNA vaccination are required to protect graftedversus endogenous islets in nonobese diabetic mice.J. Immunol. (in press). 2003.Roland Tisch, Professor in Microbiology andImmunology at <strong>the</strong> University of NorthCarolina. Currently, <strong>the</strong> laboratory is studyingType I diabetes, an autoimmune diseasecharacterized by <strong>the</strong> T cell mediateddestruction of insulin produ-cing beta cells.The non-obese diabetic (NOD) mouse, aspontaneous animal model for Type I diabetesis being utilized to eluci-date <strong>the</strong> keymolecular and cellular events involved in <strong>the</strong>diabetogenic response.<strong>GAD</strong>65 and <strong>the</strong> Immunoregulationof Autoimmune DiabetesORoland Tischur group has used <strong>GAD</strong>65as a model ß cell auto<strong>anti</strong>genfor <strong>the</strong> purpose ofdeveloping strategies of<strong>anti</strong>gen-specific basedimmuno<strong>the</strong>rapy, and definingevents involved in <strong>the</strong> breakdown of selftolerance<strong>with</strong>in <strong>the</strong> T cell compartment in nonobesediabetic (NOD) mice. As in Man, <strong>GAD</strong>65-specific reactivity can be detected in NOD miceearly in <strong>the</strong> diabetogenic response, suggesting akey role for this ß cell auto<strong>anti</strong>gen in both murineand human Type 1 diabetes (T1D).The goal of <strong>anti</strong>gen-specific based immuno<strong>the</strong>rapyis to selectively prevent and/or suppress pathologicalautoimmunity <strong>with</strong>out hindering <strong>the</strong>“normal” function of <strong>the</strong> immune system. In anumber of models of autoimmunity includingT1D, prevention and/or treatment of disease istypically accomplished through induction of socalledimmunoregulatory T cells, of which <strong>the</strong>reappear to be a number of “types” exhibitingvarious physical and functional properties. Weand o<strong>the</strong>rs have found that administration of<strong>GAD</strong>65 protein or peptides via different routes toyoung NOD mice can prevent initiation of <strong>the</strong>diabetogenic response. More importantly, administrationof <strong>GAD</strong>65 can prevent <strong>the</strong> onset ofovert diabetes even at late stages of preclinicalT1D in NOD mice (1-4). This latter scenario isreflective of a clinical setting in which prediabeticindividuals exhibiting ongoing ß cell autoimmunitywould be considered for some form of intervention.Fur<strong>the</strong>rmore, we have recently foundthat <strong>GAD</strong>65 vaccination can also be effectivelyapplied to induce islet graft tolerance. Specifically,islets transplanted into diabetic NOD mice areprotected long-term via administration of geneticvaccines encoding a portion of <strong>GAD</strong>65 and cytokinesknown to promote T cells <strong>with</strong> immunoregulatoryfunction. One key characteristic of <strong>the</strong>protection mediated by <strong>GAD</strong>65 administration isthat infiltration of T cells and o<strong>the</strong>r immune cellsinto <strong>the</strong> islets is efficiently blocked regardless of<strong>the</strong> stage of disease progression. This propertyappears to be unique to <strong>GAD</strong>65 since protectionmediated by administration of o<strong>the</strong>r ß cell auto<strong>anti</strong>gensis usually marked by continued infiltrationof <strong>the</strong> islets, especially when <strong>the</strong> diabetogenicresponse is ongoing at <strong>the</strong> time of treatment.Currently, <strong>the</strong>re is a need to fur<strong>the</strong>r define <strong>the</strong>properties of <strong>GAD</strong>65-specific immunoregulatoryT cells. For example, different “types” of T cellsexhibiting distinct as well as overlapping immunoregulatoryfunctions appear to be induced byadministration of <strong>GAD</strong>65 under <strong>the</strong> appropriateconditions. Therefore, <strong>the</strong> general immuno<strong>the</strong>rapeuticefficacy of <strong>GAD</strong>65 vaccination may reflect<strong>the</strong> relative number and “variety” of immunoregulatoryT cells elicited by treatment.Indeed, <strong>GAD</strong>65-specific T cells may “normally”regulate <strong>the</strong> progression of T1D. We have establishedNOD mice in which a significant frequencyof developing T cells express a “transgenic” T cellreceptor (TCR) specific for a peptide derivedfrom <strong>GAD</strong>65. T cells expressing <strong>the</strong> transgenicTCR are efficiently deleted through mechanismsthat prevent <strong>the</strong> development of autoreactive Tcells and maintain tolerance to self-proteins suchas <strong>GAD</strong>65. Interestingly, T1D is exacerbated in<strong>the</strong>se transgenic NOD mice. This finding suggeststhat deletion of T cells expressing <strong>the</strong> transgenicTCR results in <strong>the</strong> loss of <strong>GAD</strong>65-specific T cellswhich regulate <strong>the</strong> diabetogenic response. In <strong>the</strong>context of immuno<strong>the</strong>rapy, <strong>GAD</strong>65 vaccinationmay in part lead to <strong>the</strong> expansion of this pool ofimmunoregulatory T cells. The task at hand is todetermine whe<strong>the</strong>r findings regarding <strong>the</strong> role of<strong>GAD</strong>65-specific immunoregulatory T cells inNOD mice can be extrapolated to diabetic individualsand/or those at high risk and exploited for<strong>the</strong> purpose of immuno<strong>the</strong>rapy.page 24 dmccad june 2003


Using DNA Vaccines Expressing IsletAntigens to Prevent Type 1 DiabetesOMatthias von Herrathur laboratory is interested indevising novel strategies toprevent Type 1 diabetes byutilizing beta cell derivedauto<strong>anti</strong>gens for immunization.The underlying rationaleis that self-<strong>anti</strong>gens might frequently induceimmune responses that are regulatory in nature.Indeed, during <strong>the</strong> past 8 years, we and o<strong>the</strong>rgroups have identified autoreactive regulatory lymphocytesthat can be induced by administering betacell <strong>anti</strong>gens, such as <strong>GAD</strong>. For example, oral administrationof <strong>the</strong>se <strong>anti</strong>gens can induce immunedeviation in <strong>the</strong> sense that islet-specific responsesare skewed away from TH1 more towards a TH2or TH3-like effector phenotype. In some cases <strong>the</strong>secells are dependent on TGF-beta, in our hands <strong>the</strong>yfunctionally rely on secretion of interleukin-4 (IL-4).In our experimental systems, insulin-B induced IL-4producing CD4 + lymphocytes can act as bystandersuppressors by reducing numbers of autoaggressiveCD8 T cells in <strong>the</strong> pancreatic draining lymph nodeand <strong>the</strong> islets. At least in mouse models oral auto<strong>anti</strong>gensrequire quite large dosages to be effective.Therefore, we have developed DNA vaccines thatexpress <strong>the</strong> beta cell <strong>anti</strong>gens insB and <strong>GAD</strong>. Bothvaccines can prevent diabetes in <strong>the</strong> RIP-LCMV orNOD mouse models. However, in order to avoid augmentationof autoaggression (which one might fearas <strong>the</strong> most undesirable outcome when immunizing<strong>with</strong> auto<strong>anti</strong>gens), response modifiers in <strong>the</strong> form ofcytokines and possibly <strong>anti</strong>-CD3 should be given.This strategy does not only increase <strong>the</strong> safety marginbut also enhances efficacy.We would like to propose that immunization<strong>with</strong> auto<strong>anti</strong>gens such as <strong>GAD</strong> can be an effectivemeans to prevent Type 1 diabetes, if augmentationof autoaggression is prevented by suitable responsemodifiers. It is noteworthy that in certain experimentalsituations, diabetes was prevented <strong>with</strong>outresponse modifiers. This allows for <strong>the</strong> speculationthat some islet <strong>anti</strong>gen specific immune responsesmight be more prone to a regulatory ra<strong>the</strong>r thanaggressive phenotype.Peptide derivedfrom self-<strong>anti</strong>genBystander suppression by Ag-specific TregsMHC class I or IImoleculeT cell receptorAPCSolublemediatorsCell-cellcontactAPCCytokine/chemokineAPCTregEffectorT cell of differentAg specificitythan TregNaive or <strong>anti</strong>gen-experienced autoaggressors in<strong>the</strong> affected organ or draining lymphoid nodesare, as a consequence, not activated or amplifiedGeneration of moreTregsAfter interaction <strong>with</strong>Tregs, APC expressesless co-stimulatormolecules and IL-12,and more <strong>anti</strong>inflammatoryIL-10Matthias von Herrath,MD, Associate Member<strong>with</strong> Tenure, La JollaInstitute for Allergyand Immunology. Thefocus of von Herrath’sresearch is in devisingnovel strategies toprevent Type 1 diabetes by inducing autoreactiveregulatory T cells. Their modes ofaction via modulation of <strong>anti</strong>gen presentingcells and combinatorial application <strong>with</strong>o<strong>the</strong>r immune interventions (aCD3, aCD40L)are of particular interest. Positive and negativeassociations between viral infections andautoimmune disease are being investigated.dmccad june 2003page 25


Vaccination <strong>with</strong> <strong>GAD</strong> PlasmidSuppresses Diabetes in <strong>the</strong> NOD MouseEven After Development of InsulitisNora Sarvetnick is Professor of Immunologyat The Scripps Research Institute (TSRI) in LaJolla. Sarvetnick is focusing on new <strong>the</strong>rapiesfor <strong>the</strong> prevention of autoimmune diabetes.TNora SarvetnickThe NOD mouse developsspontaneous T cell dependentautoimmune diabetes and isused as an experimental modelto explore many features shared<strong>with</strong> Type 1 diabetes patients.Much research has focused on pancreaticauto<strong>anti</strong>gens and <strong>GAD</strong>65 is one of <strong>the</strong> bestknown targets of autoreactive T cells during <strong>the</strong>early phase of disease development. <strong>GAD</strong> expresssionin <strong>the</strong> pancreatic islets increases <strong>with</strong> age in<strong>the</strong> NOD mouse and when its expression is suppressed,diabetogenic T cells are not generated,resulting in protection from diabetes. The importanceof <strong>GAD</strong> in Type 1 diabetes has also beenconfirmed by adoptive transfer experiments, thatis, a <strong>GAD</strong>-specific CD4 T cell clone from a NODmice that normally develops diabetes transferreddiabetes into NOD-scid/scid mice that normallydo not develops diabetes.Autoimmune diabetes has been successfullysuppressed at <strong>the</strong> prediabetic stage via Th2 immunedeviation. For example, young NOD mice injected<strong>with</strong> <strong>GAD</strong> protein (intravenously or intraperitoneally)or its peptides (intranasally) responded<strong>with</strong> a reduction of T cell proliferation to <strong>GAD</strong>65and <strong>the</strong>ir insulitis and diabetes decreased. Likewise,induction of a <strong>GAD</strong>-reactive Th2 response prolonged<strong>the</strong> survival of syngeneic islets grafted indiabetic NOD mice and inhibited disease progresssionin animals <strong>with</strong> early signs of Type 1 diabetes.Additionally, <strong>the</strong> transgenic expression of IL-4 in<strong>the</strong> pancreas of NOD mice completely protected<strong>the</strong>m from <strong>the</strong> disease due to <strong>the</strong> inhibition ofdiabetogenic Th1 lymphocytes by islet-reactiveTh2 cells.Recently it was shown that vaccination of mice<strong>with</strong> plasmids encoding foreign (viral) <strong>anti</strong>gensinduced effective T cell responses and circumvented<strong>the</strong> requirement for conventional immunization<strong>with</strong> proteins in complete Freunds adjuvant.We <strong>the</strong>refore hypo<strong>the</strong>zised that genetic vaccination<strong>with</strong> plasmids encoding <strong>GAD</strong> may interfere<strong>with</strong> development of autoimmune diabetes.Indeed, <strong>GAD</strong>-plasmid vaccination effectively preventeddiabetes in NOD female mice when <strong>the</strong>ywere treated at 4 to 5 weeks of age (before insulitis)and even at 10 to 11 weeks of age (after insulitis).This protection, however, did not stem frominduction of <strong>the</strong> classical Th2 shift as reported inseveral o<strong>the</strong>r <strong>GAD</strong>-vaccination protocols. On <strong>the</strong>contrary, <strong>the</strong> mechanism of <strong>GAD</strong>-plasmid mediateddiabetes protection was due to insufficientcostimulation of <strong>GAD</strong>-specific T cells at <strong>the</strong> siteof <strong>anti</strong>gen presentation. In fact when costimulationwas provided in vivo at <strong>the</strong> time of <strong>GAD</strong>-plasmidvaccination, <strong>the</strong> protective effect was abrogated.With <strong>the</strong>se results our lab was first to report(Clinical Immunology, May, 2001) that <strong>GAD</strong>65-plasmid vaccination is effective in suppressing diabetesin <strong>the</strong> NOD mouse even after developmentof insulitis and that that this protection is notdependent on a systemic or pancreatic Th2 immuneresponse.page 26 dmccad june 2003


<strong>GAD</strong> in GraphsImmune deviation of T cell responsesto <strong>GAD</strong>65 inhibits disease progressionand protects islet transplantsin NOD mice100<strong>GAD</strong> Peptides and T1DM-Associated HLA MoleculesMay Hold <strong>the</strong> Key to DiseasePrediction and PreventionC.B. TSanjeevi1DM is an autoimmune diseasewhich occurs predominantlyin children.Auto<strong>anti</strong>bodies to T1DMauto<strong>anti</strong>gens (<strong>GAD</strong>65 orGlutamic acid decarboxylaseisoform 65, IA-2 or Tyrosine phosphatase andinsulin) are used to identify ongoing autoimmuneprocess both in newly diagnosed T1DM childrenand prediabetic individuals. The autoreactive Tcells are implicated in <strong>the</strong> destruction of <strong>the</strong> insulinproducing beta cells leading to <strong>the</strong> disease and<strong>the</strong> auto<strong>anti</strong>bodies are considered only markersfor <strong>the</strong> disease.We have studied HLA-DQ that is associated<strong>with</strong> T1DM in my laboratory. These studies havehelped us to identify naturally processed and presentedpeptides from susceptible and protectiveHLA-DQ, sequence <strong>the</strong>se peptides and show <strong>the</strong>binding motifs (from peptide sequencing andmolecular modelling).We have screened <strong>the</strong> diabetes auto<strong>anti</strong>gen<strong>GAD</strong>65 and identified several 15 amino acid longpeptides that carry <strong>the</strong> right binding motifs forHLA-DQ associated <strong>with</strong> T1DM.We believe that T cells reacting to <strong>the</strong> DQrestricted peptides from T1DM auto<strong>anti</strong>gen(<strong>GAD</strong>65) are in <strong>the</strong> peripheral blood of newlyCarani B. Sanjeevi isan Associate Professorin <strong>the</strong> division ofDiabetes andEndocrinology at <strong>the</strong>Department ofMolecular Medicine at<strong>the</strong> KarolinskaInstitute in Stockholm. Sanjeevi is also headat <strong>the</strong> ‘Molecular Immunogenetics’ researchgroup at Karolinska Hospital. Sanjeevi isassociated <strong>with</strong> his contribution of MHC inautoimmune Diabetes and is currently workingon <strong>the</strong> role of peptides from diabetesauto<strong>anti</strong>gens (<strong>GAD</strong>65) and viruses that carrymotifs for diabetes associated HLA-DQ, indisease prediction.diagnosedT1DM childrenand prediabeticchildren and<strong>the</strong>ir detectionis predictive of<strong>the</strong> disease.An assay toidentify <strong>the</strong> Tcells reacting to<strong>the</strong> DQ restrictedpeptides hasbeen standardizedand is being tested in newly diagnosed Type 1diabetes children and <strong>the</strong>ir first degree relatives. Wehope that this assay will help us to both diagnose<strong>the</strong> disease and also to predict <strong>the</strong> disease in firstdegree relatives.Our subsequent aim would be to use <strong>the</strong>se peptidesin some form of peptide-based vaccine approach.Future potential for <strong>GAD</strong>65:<strong>GAD</strong>65 has a very good potential in <strong>the</strong> diagnostic,predictive as well as preventive approach. The keyis to find <strong>the</strong> form in which <strong>GAD</strong>65 could beused to achieve <strong>the</strong> goals. It could be whole<strong>GAD</strong>65 or modified <strong>GAD</strong>65 or peptides from<strong>GAD</strong>65.Diabetes Incidence (%)% Diabetes Recurrence1008060402080604020§-GAL.Insulin B-chainHSP277<strong>GAD</strong>6500 15 30 45 60 75 90 105Post-transplantation Time (days)Tian et al. (1996) Modulating autoimmuneresponses to <strong>GAD</strong> inhibits disease progressionand prolongs islet graft survival in diabetespronemiceNature Medicine 2:1348-1353• Pancreatic islets transplantedinto diabetic NOD mice that hadbeen pretreated <strong>with</strong> <strong>GAD</strong>65survived longer than in those thathad been pretreated <strong>with</strong> ei<strong>the</strong>rß-galactosidase, insulin B chainor hsp65• <strong>GAD</strong>65 treatment increases islettransplant efficacyNasal administration of <strong>GAD</strong>65 preventsdiabetes in NOD mice<strong>GAD</strong>65 peptidesControl peptide010 15 20 25 30 35 40 45 50 55 60Time (weeks)Tian et al. (1996) Nasal administration of glutamatedecarboxylase (<strong>GAD</strong>65) peptides inducesTh2 responses and prevents murine insulindependentdiabetesJ Exp Med 183:1561-1567• Intranasal administration of<strong>GAD</strong>65 peptide into 2-3wk old NODmice induced IgG1 <strong>anti</strong>-<strong>GAD</strong> <strong>anti</strong>bodies,reduced IFN-γ and increasedIL-5responses to <strong>GAD</strong>65 (i.e. diversion of<strong>the</strong> <strong>GAD</strong> response from Th1-Th2)• Nasal administration of<strong>GAD</strong>65 prevents diabetesdmccad june 2003page 27


<strong>GAD</strong><strong>GAD</strong> in Graphs<strong>GAD</strong> ELISPOT outperforms proliferationassays for detection of <strong>GAD</strong>reactiveT cellsStimulation IndexIL-5 (pg/ml)121086420300250200150100500Control Type 2 Type 1*(n=23) (n=12) (n=31)Kotani et al (2002) Detection of <strong>GAD</strong>65 reactiveT cells in Type 1 diabetes by immunoglobulinfreeELISPOT assays. Diabetes Care 25:1390-1397• Cellular immune responses toß-cell auto<strong>anti</strong>gens were studiedby proliferation and ELISPOT inPMBCs of Type 1, Type 2 diabetespatients and healthy controls• <strong>GAD</strong>65-specific proliferation wasdetected in 32% Type 1 patients,while IFN-γ ELISPOT <strong>GAD</strong>65-specificactivity was detected in 66% patientsImmunization <strong>with</strong> a <strong>GAD</strong>65/IL-4plasmid DNA vaccine prevents diabetesin NOD miceCMedium*untreatedHEL<strong>GAD</strong><strong>GAD</strong>/IL-4<strong>GAD</strong>65 HSP60 CPHTisch et al. (2001) Antigen-specific mediatedsuppression of cell autoimmunity by plasmidDNA vaccination.J Immunol 166:2122-2132p=.003• Enhanced secretion of IL-5 anddecreased secretion of IFN-γ by Tcells from NOD mice immunized at12 wks old <strong>with</strong> pDNA encoding<strong>GAD</strong>65-IgFc and IL-4 and restimulated<strong>with</strong> <strong>GAD</strong>65.• Vaccination caused a shift in cytokineresponse to <strong>GAD</strong>65 stimulation*Human T Cells Recognizing<strong>GAD</strong>65 in Type 1a DiabetesODavid Hafler, Jack Sadie, David Breakstone and Sally Kentur laboratory has beeninterested in <strong>the</strong> functionof autoreactive T cells inhumans <strong>with</strong> autoimmunedisease and in particular,Multiple Sclerosis (MS) formany years. The T cell response is regulated in partthrough two signaling events. The first signal isthrough <strong>the</strong> T cell receptor by peptide processedfrom an <strong>anti</strong>gen in <strong>the</strong> context MajorHistocompatibility Complex (MHC) proteins onan <strong>anti</strong>gen-presenting cell (APC) and <strong>the</strong> second isthrough costimulation proteins, CD28 and CTLA-4by B7-1 and B7-2 proteins on APCs. We and o<strong>the</strong>rshad previously found that myelin basic protein(MBP) reactive T cells in patients <strong>with</strong> MS werecostimulation-independent (second signal) as comparedto T cells from normal individuals. This indicatesthat <strong>the</strong> autoreactive T cells behaved morelike T cells in a memory response and that <strong>the</strong> patienthas had T cells reactive to MBP for some time.This also suggests a method for differentiating patientT cell responses from those of controls andways of intervening in <strong>the</strong> autodestructive immuneresponse by <strong>the</strong> T cells.These data prompted us to examine this issuein Type 1 diabetes. Insulin-dependent Type 1a diabetesis an autoimmune disease mediated by Tlymphocytes recognizing pancreatic islet cell <strong>anti</strong>gens.Glutamic acid decarboxylase 65 (<strong>GAD</strong>65)appears to be an important auto<strong>anti</strong>gen in <strong>the</strong> disease.We found that in patients <strong>with</strong> new-onsetType 1a diabetes, <strong>GAD</strong>65-reactive T cells werestrikingly less dependent on CD28 and B7-1 costimulationto enter into cell cycle and proliferatethan were equivalent cells derived from healthycontrols. B7-2 appears to be <strong>the</strong> primary costimulatorymolecule engaging CD28 in T cell activationof <strong>GAD</strong>65-reactive T cells, and its engagement<strong>with</strong> CTLA-4 appears to deliver a negativesignal. We hypo<strong>the</strong>size that <strong>the</strong>se autoreactive Tcells have been activated in vivo and have differentiatedinto memory cells, suggesting a pathogenicrole in Type 1 diabetes. These findings stronglyindicate that <strong>the</strong> activation state of <strong>anti</strong>gen-specificcells plays a role in <strong>the</strong> autoimmune processand selected costimulatory molecules may represent<strong>the</strong> target of future <strong>the</strong>rapies.We are currently utilizing <strong>GAD</strong>65 for o<strong>the</strong>r studiesin Type 1a diabetes. These include studies examining<strong>the</strong> qu<strong>anti</strong>tation and phenotype of <strong>GAD</strong>65reactive T cells from controls and Type 1 diabetics<strong>with</strong> a <strong>GAD</strong> peptide-loaded tetramer (HLADR*0401 loaded <strong>with</strong> <strong>GAD</strong> p555-567) and monitoringpotentially destructive <strong>GAD</strong>65 T cell autoreactiveresponses in long-term Type 1a diabetics receivingislet transplants. We are enthusiastic to continueto utilize <strong>GAD</strong>65 as a means of examining autoreactiveT cellresponses inhuman Type 1adiabetes.David Hafler, MD. is Professor of Neurology(Neuroscience) at Brigham and Women’sHospital and Harvard Medical School inBoston, MA., and <strong>the</strong> head of <strong>the</strong> MolecularImmunology Laboratory. Hafler’s main clinicaland research interests are in human autoimmunediseases: Multiple Sclerosis, Type 1diabetes and rheumatoid arthritis. His goalsare to understand <strong>the</strong> nature of self-recognitionby T cells, to understand how that immuneresponse leads to autoimmune disease andhow one can alter this response to developnovel <strong>the</strong>rapiesSally Kent, Ph.D., is an Instructor in Neurologyand Associate Immunologist at Brigham andWomen’s Hospital and Harvard MedicalSchool in Boston, MA. Kent has specialized inNKT cell function in Type 1a diabetics by examiningperipheral blood and pancreatic draininglymph nodes. In Type 1a diabetes, Kenthas focused on <strong>GAD</strong>65 T cell reactivity as ameasure of autoreactivity and memory T cellresponses in controls and patients and inpatients undergoing islet cell transplantation.page 28 dmccad june 2003


<strong>GAD</strong> in Graphs<strong>GAD</strong>65 Specific RegulatoryT Cells May ProvideProtection from DiabetesNAnthony Quinnumerous studies haveshown that T cells of <strong>the</strong>CD8 + and CD4 + subsetsare both involved in <strong>the</strong>immunopathogenesis ofType 1 diabetes (T1D) inhumans. Likewise, both <strong>the</strong>se cell types are alsonecessary for T1D in NOD mice, <strong>the</strong> murine modelfor spontaneous autoimmune diabetes. We havebeen studying <strong>the</strong> cellular immune responses toself-<strong>anti</strong>gens in NOD mice to determine significancein <strong>the</strong> initiation of islet-specific damage. Thefocus has been on two very relevant issues regarding<strong>the</strong> initiation of autoimmune disease: <strong>the</strong> identificationof host-derived factors that influence susceptibilityto autoimmune disease and <strong>the</strong> role ofinfectious agents in <strong>the</strong> induction and persistenceof autoimmunity. Although both issues are broadgeneralized topics – particularly <strong>the</strong> study of susceptibilityfactors – <strong>the</strong>re are very specific issuesthat can be readily addressed and that provide valuableinformation for our understanding of <strong>the</strong> pathogenicprocesses in autoimmune disease.First, does <strong>the</strong> quality of <strong>the</strong> autoimmuneresponse to self-<strong>anti</strong>gens influence <strong>the</strong> susceptibilityto autoimmune disease? Recently, we have beenseeking to determine if CD8 + T cell responses toglutamic acid decarboxylase (<strong>GAD</strong>65) are clinicalllyrelevant in <strong>the</strong> NOD mouse model of T1D.<strong>GAD</strong>65 is one of <strong>the</strong> first beta cell <strong>anti</strong>gens toprime autoimmune responses that are detectablein <strong>the</strong> spleens of naive prediabetic NOD mice.Our preliminary data demonstrates that <strong>GAD</strong>65-reactive CD8 + T cells can be found in prediabeticNOD mice, while o<strong>the</strong>rs have shown that suchcells are present in <strong>the</strong> peripheral blood of humanpatients recently diagnosed <strong>with</strong> diabetes.Currently, we are investigating <strong>the</strong> nature of<strong>GAD</strong>65-specific regulatory T cells which may provideprotection from diabetes in individuals whoare considered to be at-risk but remain diabetesfree.Such mechanisms may provide protection tosiblings/relatives of diabetic individuals and maybe represented in male NOD mice and F1 mice,both of which display a reduced susceptibility todiabetes compared to female NOD mice.Secondly, can islet proteins, or fragments from<strong>the</strong>m, be used to prevent diabetes in young NODmice or humans? Autoimmune diseases such as diabetesare likely <strong>the</strong> result of immunological dysregulationsuch that <strong>the</strong> balance between <strong>the</strong> antagonisticforces of regulation and pathogenesis tilts in favor of<strong>the</strong> pathogenic mechanisms. One of our objectives isto initiate <strong>GAD</strong>65-specific mechanisms that are capableof restoring <strong>the</strong> immunological balance in diabetes-pronemice. Hopefully, <strong>the</strong> clues gained in <strong>the</strong>murine models can also provide insights into applicationsto humandisease.Tony Quinn, Ph.D., isAssociate Professor inBiological Sciences at<strong>the</strong> University ofToledo, and AdjunctProfessor inMicrobiology andImmunology at <strong>the</strong>Medical College of Ohio, Toledo, Ohio. Quinnis an immunologist <strong>with</strong> a long-standinginterest in <strong>the</strong> regulation of immunity, as itrelates to <strong>the</strong> control of autoimmune disease,and <strong>the</strong> enhancement of targeted immuneresponses. Quinn is currently studying <strong>the</strong>cellular immune responses to self-<strong>anti</strong>gens inautoimmune diabetes and <strong>the</strong>ir roles in <strong>the</strong>initiation of pancreas-specific damage.Relative amount of <strong>GAD</strong>65immunoprecipitated (% of PAS)(%) Diabetes10080604020Clinical onset of IDDM is associated<strong>with</strong> a decreased Th2 <strong>GAD</strong> response2520151050IgM<strong>GAD</strong>65-GMCSFPre. IDDM -IDDM -IDDM(High)Petersen et al (1999). Progression to Type 1 diabetesis associated <strong>with</strong> a change in <strong>the</strong> immunoglobulinisotype profile of auto<strong>anti</strong>bodies toglutamic acid decarboxylase (<strong>GAD</strong>65). ChildhoodDiabetes in Finland Study Group. Clin Immunol90:2 276-81• Isotype-specific IgE and IgM<strong>GAD</strong>65 auto<strong>anti</strong>bodies in firstdegree relatives before clinical onset(Pre), IDDM at onset (IDDM), lowrisk IDDM relatives (-IDDM) andhigh risk relatives (-IDDM High).• Individuals <strong>with</strong> a low risk forType 1 diabetes have a higher Th2response to <strong>GAD</strong>65 than do individualsat high risk or already <strong>with</strong>diseaseAdoptive transfer of <strong>GAD</strong>-reactiveCD4 + Th1 cells induces diabetes inNOD/SCID mice5A injeced miceOVA3E3 injected miceSALINE injected mice00 2 4 6 8 10 12Time (wk)Zekzer et al. (1998) <strong>GAD</strong>-reactive CD4 + Th1 cellsinduce diabetes in NOD/SCID miceJ Clin Invest 101:68-73• Adoptive transfer of <strong>the</strong> <strong>GAD</strong>specificCD4 + T cell line 5A inducesdiabetes In NOD/SCID mice, buttransfer of an OVA-specific controlT cell line (OVA3E3) does not• <strong>GAD</strong>-specific T cells cause diabetes**dmccad june 2003page 29


<strong>GAD</strong> in GraphsA1004 wks of age% Incidence of diabetes% Incidence of diabetes80604020B10001480604018 22 2610 wks of age(n=12)Plasmids<strong>GAD</strong>65<strong>GAD</strong>65-GMCSFcontrol(n=8)(n=13)Vaccination <strong>with</strong> <strong>GAD</strong> plasmid DNAprevents diabetes in NOD mice(n=11)Plasmids<strong>GAD</strong>65<strong>GAD</strong>65-GMCSFcontrol20(n=8)(n=7)016 14 20 24 28 32 36Balasa et al, 2001. Vaccination <strong>with</strong> <strong>GAD</strong> plasmidDNA protects mice from spontaneousautoimmune diabetes and B7/CD28 costimulationcircumvents that protectionClin Immunol 99:241-252• Mice vaccinated at both 4-5 (A)and 10-11 weeks of age (B) wereprotected from developing diabetesIs <strong>GAD</strong> All There Is?TBart Roephe discovery of <strong>GAD</strong>65 as oneof <strong>the</strong> first islet auto<strong>anti</strong>gens in<strong>the</strong> pathogenesis of type 1 diabeteswas a breakthrough thatenabled study of specific autoimmuneresponses. I worked asa postdoc in <strong>the</strong> laboratory of Steinunn Baekkeskovin San Francisco at <strong>the</strong> time that <strong>the</strong> identity of<strong>the</strong> 64,000 kDa target of auto<strong>anti</strong>bodies was unraveledas <strong>GAD</strong>65. My aim as a T cell immunologistwas to determine T cell autoreactivity to this proteinin humans, and to study whe<strong>the</strong>r immunizationof genetically predisposed mice <strong>with</strong> <strong>GAD</strong>65led to <strong>the</strong> induction of insulitis or diabetes. Theresults in <strong>the</strong>se pioneer studies to check for T cellautoimmunity were unexpected since I foundvery few differences between patients and controlsubjects, although in both groups significantresponses were detectable. With few exceptions,this finding turned out to be reproducible bymany colleagues all over <strong>the</strong> world during <strong>the</strong> folllowingdecade. My second aim to induce diabetesby immunization <strong>with</strong> <strong>GAD</strong>65 was in vain:although very strong immune responses could beinduced in various strains of mice, none of <strong>the</strong>sedeveloped insulitis or diabetes. The latter observationproved useful for <strong>the</strong> application of using<strong>GAD</strong>65 as an immune modulator ra<strong>the</strong>r than asan inducer of pathogenesis. Currently <strong>the</strong>re is consensusthat, immunization <strong>with</strong> <strong>GAD</strong>65 delays orprevents diabetes in mice, ra<strong>the</strong>r than inducing oraccelerating <strong>the</strong> disease.For <strong>the</strong> next step as T cell immunologists, weintroduced <strong>GAD</strong>65 as a candidate target auto<strong>anti</strong>genin our immune monitoring of Type 1 diabetespatients receiving islet allografts. Indeed, chronicrecurrent loss of islet allograft function wasaccompanied <strong>with</strong> increment in T cell autoreactivityto <strong>GAD</strong>65, even in <strong>the</strong> absence of increases in<strong><strong>anti</strong>body</strong> titers, while this reactivity was neverobserved in patients successfully transplanted <strong>with</strong>islets. This clinical trial proved that longitudinalstudies on cellular immune responses to <strong>GAD</strong>65were informative to determine <strong>the</strong> clinical fate ofbeta-cells in Type 1 diabetes patients. It proved oftremendous importance to test <strong>GAD</strong>65 of highpurity to avoid reactivity to contaminants in <strong>the</strong>preparation of recombinant <strong>GAD</strong>65. This was <strong>the</strong>prime result from <strong>the</strong> first international workshopon T cell reactivity to islet auto<strong>anti</strong>gens. Eversince, I have used <strong>GAD</strong>65 produced and purifiedby diamyd as golden standard, since this materialwas of reproducibly high quality and void of toxicor mitogenic contaminants. A second satisfactoryencounter <strong>with</strong> <strong>GAD</strong>65 came from studies in arare autoimmune disorder called Stiff-ManSyndrome (SMS) that shares <strong>GAD</strong>65 <strong>with</strong> Type 1diabetes as major target auto<strong>anti</strong>gen. One-third ofSMS patients develop Type 1 diabetes. We studieda case of SMS <strong>with</strong>out Type 1 diabetes to understandwhy SMS patients often do not developType 1 diabetes despite phenomenal autoimmunityto this neuroendocrine auto<strong>anti</strong>gen <strong>GAD</strong>65, inorder to develop immuno<strong>the</strong>rapy for Type 1 diabetesin patients that lost tolerance to this protein.Indeed, <strong>the</strong> <strong>GAD</strong>65 reactivity measured in thispage 30 dmccad june 2003


“Indeed, chronic recurrent loss of islet allograftfunction was accompanied <strong>with</strong> incrementin T cell autoreactivity to <strong>GAD</strong>65, even”in <strong>the</strong>absence of increases in <strong><strong>anti</strong>body</strong> titersnon-diabetic SMS patient was characterized as nonoreven <strong>anti</strong>-inflammatory by its IL-10 producingnature. Unexpectedly, this SMS patient did not elicitprimary proliferative responses to <strong>GAD</strong>65.However, when she developed Type 1 diabetesfour years later, strong primary responses weredetectable, while <strong>the</strong> only cytokine produced by<strong>the</strong>se T cells was <strong>the</strong> proinflammatory cytokineinterferon-γ. At that time, all symptoms of SMShad resolved. This observation demonstrated that<strong>the</strong> nature of cellular autoimmunity to <strong>GAD</strong>65 isan important parameter to distinguish healthy subjectsfrom Type 1 diabetics from SMS patients.Moreover, this was a clinical demonstration thatan <strong>anti</strong>-inflammatory cytokine profile was favorablefor diabetes-prone subjects against developmentof Type 1 diabetes.Thirteen years after <strong>the</strong> discovery of <strong>GAD</strong>65 astarget auto<strong>anti</strong>gen in Type 1 diabetes mellitusthrough its recognition by auto<strong>anti</strong>bodies, we arestill left <strong>with</strong> many open issues. The disease specificityof immune responses to <strong>GAD</strong>65 is unclear.Auto<strong>anti</strong>bodies against <strong>GAD</strong>65 are rarely found innon-diabetic subjects, but <strong>the</strong> majority of subjectswho do have such <strong>anti</strong>bodies will remain healthy,while o<strong>the</strong>rs suffer from different autoimmunediseases. With regard to T cell responses to<strong>GAD</strong>65, <strong>the</strong> consensus is that <strong>the</strong>re are only minimaldifferences in T cell proliferation to <strong>GAD</strong>65in Type 1 diabetes patients versus controls. Thedifference most likely lies in <strong>the</strong> quality of <strong>the</strong>immune response, and how <strong>GAD</strong>65 autoimmunityis regulated. The big question now, however, iswhe<strong>the</strong>r islet auto<strong>anti</strong>gens such as <strong>GAD</strong>65 can beused as an immuno<strong>the</strong>rapeutic to divert autoimmmunityfrom destruction to regulation.It should also be considered to assess whe<strong>the</strong>rcombinations of immuno<strong>the</strong>rapies including<strong>GAD</strong>65 as an immune modifying agent are effectiveto suppress disease activity. In preliminary invitro experiments we have observed that stimulationof <strong>GAD</strong>65 specific autoreactive T cell clonesisolated from a prediabetic subject is affected by<strong>the</strong> new generation of humanized monoclonal<strong>anti</strong>bodies against CD3, and only upon stimulation<strong>with</strong> <strong>GAD</strong>65.Bart Roep is AssociateProfessor in Medicine,in particular <strong>the</strong>immunology andimmunogenetics ofType 1 diabetes, at <strong>the</strong>Leiden UniversityMedical Center, Leiden,The Ne<strong>the</strong>rlands. Roep pioneered studies on<strong>the</strong> role of T cells in <strong>the</strong> pathogenesis ofType 1 diabetes, demonstrating that autoreactiveT cells play a key role in beta celldestruction in humans. Roep is currentlyinvolved in <strong>the</strong> design and evaluation of newimmuno<strong>the</strong>rapies to prevent beta-cell autoimmunity.dmccad june 2003page 31


Mark Atkinson, Ph.D.,is an AmericanDiabetes AssociationProfessor for DiabetesResearch, and <strong>the</strong>Director of <strong>the</strong> Centerfor Immunology andTransplantation at <strong>the</strong>University of Florida. Atkinson was amongst<strong>the</strong> first groups of researchers to identify<strong>the</strong> value of measuring immune responsesagainst <strong>GAD</strong>, and to describe <strong>the</strong> whiteblood cell response against this protein inpersons <strong>with</strong> <strong>the</strong> disease. Atkinson holdspositions on a number of scientific advisoryboards/research panels including <strong>the</strong>Juvenile Diabetes Foundation Inter-national(JDFI), <strong>the</strong> American Diabetes Associationand <strong>the</strong> National Institutes of Health (NIH).While Atkinson’s current research extends tounderstanding <strong>the</strong> molecular immunologicaland genetic mechanisms underlying <strong>the</strong> formationof diabetes, his primary researchgoal lies in <strong>the</strong> development of an effectivemethod for preventing insulin-dependentdiabetes. Professors Atkinson and NoelMaclaren were first to file a US patent for<strong>GAD</strong> and diabetes <strong>the</strong>rapy which is exclusivelylicensed by <strong>Diamyd</strong> Medical.More Effective Methods forpreventative Interventionβ cell MassLower Accuracy inDisease PredictionHigher Accuracy inDisease PredictionLess Effective Methodsfor PreventativeInterventionOnset of OvertType 1 DiabetesAbility to Predict Type 1DiabetesAbility to Predict Type 1DiabetesType 1 Diabetes: a Dilemmafor Clinical TreatmentTMark Atkinsonhroughout much of <strong>the</strong> lastdecade, guarded hope existedthat an agent capable of preventingor reversing Type 1diabetes would be uncovered.As of today, such an agentdoes not unequivocally exist. As a result, manyhave addressed <strong>the</strong> question ‘why?’ The answers tothis question are many; some of which are readilyaddressable, o<strong>the</strong>rs are by <strong>the</strong>ir nature more inherentlydifficult. Among <strong>the</strong> latter obstacles facing<strong>the</strong> diabetes prevention field is a situation that hasbeen referred to as <strong>the</strong> ‘treatment dilemma’. Awide body of evidence, both in animal models ofType 1 diabetes as well as in persons <strong>with</strong> or – atincreased risk for – <strong>the</strong> disease, supports <strong>the</strong>notion that <strong>the</strong> most effective interventions willbe those that are begun early in <strong>the</strong> autoimmunedisease process. In contrast, <strong>the</strong> process of diseaseprediction [that is, using immunologic (e.g. <strong>GAD</strong>auto<strong>anti</strong>bodies), genetic (e.g. HLA types), andmetabolic (e.g. glucose tolerance tests) markers of<strong>the</strong> disease to identify risk for eventual diseasedevelopment] is most accurate in <strong>the</strong> period closeto <strong>the</strong> onset of overt diabetes. As a result, a conflict(both ethical and clinical) exists wherein <strong>the</strong>most effective forms of <strong>the</strong>rapy may involve <strong>the</strong>early treatment of subjects in a period in whichdisease prediction is less accurate; a situation thathas positioned <strong>the</strong> need for a safe and benignform of <strong>the</strong>rapy against treating persons who maynever develop Type 1 diabetes. The identificationof such an idealized agent has thus far provenextremely difficult to uncover.Yet ano<strong>the</strong>r challenge relates to properlyaddressing, in combination, <strong>the</strong> questions of “whodo we treat” and “what agent will we use”? In <strong>the</strong>ory,attempts to prevent Type 1 diabetes will mostlikely address two distinct populations. The firstwould involve <strong>the</strong>rapy of high-risk individuals (e.g.<strong>GAD</strong>/islet auto<strong><strong>anti</strong>body</strong> positive relatives of aproband <strong>with</strong> Type 1 diabetes) or those who alreadyhave one form of <strong>the</strong> disease (e.g. LADA subjects).The second would be that of a generalpopulation approach such as is common practicefor vaccinations against infectious disease. Bothmodels have inherent strengths and weaknesses interms of <strong>the</strong>rapeutic intervention. In <strong>the</strong> lattermodel, a safe and benign <strong>the</strong>rapy capable of interrruptingadverse immune events/environmentalagents (e.g. a vaccine) or alterations in lifestyle providingavoidance of disease risk factors (e.g. diabetogenicdietary components) would ideally beimplemented while <strong>the</strong> costs associated <strong>with</strong> screeninggeneral populations forms a barrier. Indeed,one could speculate that in designing preventativemeasures <strong>with</strong>in <strong>the</strong> general population, <strong>the</strong> diseasefrequency and unpredictable time of onsetform major obstacles that screening would be eliminatedand vaccination would become universal.Performing clinical trials in increased-risk populationsor those already diagnosed <strong>with</strong> <strong>the</strong> diseasemay prove more cost effective (in terms of a trial)and efficient, yet in terms of humanitarian benefit,it could be argued that <strong>the</strong> general populationapproach may ultimately be more importantas approximately 85% of newly-diagnosedpatients have no family history of <strong>the</strong> disease.A final barrier for this discussion is <strong>the</strong> lack ofobvious candidates for <strong>the</strong> next round of largeprevention trials. As a result, current interest isdirected at studies involving recent-onset Type 1diabetes and LADA patients for <strong>the</strong> purpose ofidentifying new and perhaps more promisingagents such as diamyd . If diamyd is shown to beclinically effective, <strong>the</strong>n prospective, randomizedcontrolled studies <strong>with</strong> appropriate statisticalpower and objective endpoints can be designedand prevention strategies in different populationgroups at different stages of <strong>the</strong> disease processcan be undertaken. Not only will <strong>the</strong>se studiesascertain potential efficacy and safety, but shouldalso lead to greater insight into disease pathogenesis.page 32 dmccad june 2003


<strong>Diamyd</strong> S-100ß ConcentrationMeasurement ELISADetermination of S-100β protein concentrations in serum is used in neurologyto assess <strong>the</strong> extent of brain damage in stroke, in head injuries, during extracorporealcirculation and during circular arrest.It is also used for follow-up and prognosis of malignant melanoma.<strong>Diamyd</strong>, Inc has developed prototype ELISA kits for detecting <strong>the</strong> presence ofS-100ß protein in human serum/plasma.<strong>Diamyd</strong> S-100ß-<strong><strong>anti</strong>body</strong> ELISAS-100β auto<strong>anti</strong>bodies have recently been shown to be a possiblemarker for autoimmune diabetes. An article (Nature 2003*) shows evidencethat islet cell death is related to autoreactive T- and β-cellresponses to neighbouring peri-islet Schwann cells, which express S-100β protein. <strong>Diamyd</strong>, Inc has developed prototype ELISA kits fordetecting <strong>the</strong> presence of auto<strong>anti</strong>bodies to S-100ß protein in humanserum/plasma. The ELISA uses visual detection (requires a visibleplate reader that measures absorbance at 492 nm).page 32www.diamyd.comproducts@diamyd.compage 21dmccad june 2003


<strong>GAD</strong> in GraphsIL-4 (pg/ml)250100755025Immunization <strong>with</strong> a <strong>GAD</strong>65/IL-4plasmid DNA vaccine preventsdiabetes in NOD miceB *p=.002untreatedHEL<strong>GAD</strong><strong>GAD</strong>/IL-40Medium <strong>GAD</strong>65 HSP60 CPHTisch et al. (2001) Antigen-specific mediatedsuppression of cell autoimmunity by plasmidDNA vaccination.J Immunol 166:2122-2132• Enhanced secretion of IL-4 anddecreased secretion of IFN-γ by Tcells from NOD mice immunized at12 wks old <strong>with</strong> pDNA encoding<strong>GAD</strong>65-IgFc and IL-4 and restimulated<strong>with</strong> <strong>GAD</strong>65.• Vaccination caused a shift in cytokineresponse to <strong>GAD</strong>65 stimulationOur Story of <strong>GAD</strong>– Serendipity in SciencePaul Zimmet and Ian MackayOur involvement <strong>with</strong> <strong>the</strong> <strong>GAD</strong> story is one of serendipity. One of us (PZ) was attendinga major international pharmaceutical company advisory board meeting inRome in 1991 and heard <strong>the</strong> presentation of one of <strong>the</strong>ir lead scientists, Dr. BillKnowles. He presented work that he was involved in on islet cell <strong>anti</strong>bodies.This wasjust around <strong>the</strong> time that <strong>GAD</strong> had been identified by Baekkeskov as <strong>the</strong> 64kD <strong>anti</strong>genthat she had discovered in <strong>the</strong> 1980’s – a putative key auto<strong>anti</strong>gen in Type 1 diabetes.I told Bill that I was very keen to have access to a method to measure <strong>anti</strong>-<strong>GAD</strong>. Over a quiet glass of Chi<strong>anti</strong>, Bill told me that he had purified <strong>GAD</strong> from pigbrain and was attempting to develop an assay for <strong>anti</strong>bodies but his company was% Islets100β cell-specific expression of <strong>GAD</strong> isrequired for development of autoimmunediabetes806040200H-AS-<strong>GAD</strong>Tg(-)Islets Status4 retracted3 >50%2 25-50%1 >25%0 NormalYoon et al. (1999) Control of autoimmunediabetes in NOD mice by <strong>GAD</strong> expression orsuppression in β cells Science 284:1183-1190• Islet grafts from transgenic miceexpressing high levels of <strong>anti</strong>sense<strong>GAD</strong>65/67 (H-AS-<strong>GAD</strong>) survivewhen grafted into acutely diabeticNOD mice, while grafts expressing<strong>GAD</strong> (Tg-) are infiltrated anddestroyed• <strong>GAD</strong> is <strong>the</strong> target of autoimmuneattack in diabetesnot all that interested as <strong>the</strong>y were more interested in Type 2 diabetes. I asked himwhe<strong>the</strong>r I could have some <strong>GAD</strong> and he agreed. Knowles became a valued advisorSand collaborator in <strong>the</strong> next phase of our work.o <strong>the</strong> <strong>GAD</strong> came to Melbourneand <strong>with</strong>in a few weeks, <strong>the</strong> nimblefingers of Dr. Merrill Rowleyin our laboratory resulted in <strong>the</strong>development of <strong>the</strong> first radioimmmunoassay(<strong>RIA</strong>) for <strong>anti</strong>-<strong>GAD</strong>,based on radioiodine labelling of <strong>the</strong> Knowles’<strong>GAD</strong> preparation. I was involved <strong>with</strong> <strong>the</strong> developmentof a Type 1 Diabetes Register in ourisland state of Tasmania so I was able to quicklyfind samples to test and we demonstrated highlevels in newly diagnosed and long-standing casesof Type 1 diabetes. The results were publishedin Diabetes as <strong>the</strong> first <strong>RIA</strong> for <strong>anti</strong>-<strong>GAD</strong>.Then again, serendipity came into play. Wewere very interested in <strong>the</strong> fact that a numberof adults were identified <strong>with</strong> diabetes that presentedclinically as Type 2 yet <strong>the</strong>ir naturalhistory over a period of a year or more was indicativeof insulin dependency. We were awarethat a close friend and colleague, Leif Groop,<strong>the</strong>n in Finland, had undertaken a study someyears previously on a group of <strong>the</strong>se patients andhad demonstrated a positive test for islet cell<strong>anti</strong>bodies. Fortuitously, one of his outstandingyoung researchers, Dr. Tiinamaija Tuomi, hadcome to our laboratory as a visiting researcher.She obtained <strong>the</strong> serum samples from Groop’sstudy and we were quickly able to measure <strong>anti</strong>-<strong>GAD</strong> in <strong>the</strong>se subjects: <strong>the</strong> frequency of <strong>anti</strong>-<strong>GAD</strong> in <strong>the</strong>se adult subjects was very high andappeared to be a better predictor of insulindependency than ICA. Indeed, when vigorouslychallenged by a leading authority on ICA at anAmerican Diabetes Association presentation asto our ability to perform <strong>the</strong> measurement ofpage 34 dmccad june 2003


ICA properly, Dr. Tuomi was able to point outthat <strong>the</strong> ICA assays had been performed in hislaboratory!We <strong>the</strong>n had a vigorous debate on what weshould call this slow-onset Type 1 diabetes. LeifGroop favoured <strong>the</strong> term AIDA (autoimmunediabetes in adults) and PZ suggested SODA(slow onset diabetes of adults), but <strong>the</strong> wisdomand status of IM prevailed and we adopted <strong>the</strong>term LADA (latent immune diabetes in adults),not to be confused <strong>with</strong> <strong>the</strong> Mexican telephonecompany or <strong>the</strong> Czech motor car! Tuomi quicklywrote up <strong>the</strong> results and submitted <strong>the</strong> paperto Diabetes – it was accepted <strong>with</strong>in 5 days.With a number of o<strong>the</strong>r studies <strong>with</strong> internationalcollaborators we were able to confirm ourfindings in several different countries and ethnicgroups. The biggest challenge still lay ahead. Wewere battling against <strong>the</strong> ICA “mafia” who stillbelieved that this test was <strong>the</strong> gold standard.Would it be feasible to use <strong>the</strong> <strong>anti</strong>-<strong>GAD</strong> test topredict future diabetes?We <strong>the</strong>n came again through serendipity to astudy we called “Back to <strong>the</strong> Future”.Fortuitously, my colleague in Finland, ProfessorJaakko Tuomilehto, brought to our attentionthat <strong>the</strong> Finnish Women’s Register had takenblood samples from every woman during pregnancysince 1984 and <strong>the</strong>se samples were storedaway at <strong>the</strong> National Finnish Public HealthInstitute. By linking <strong>the</strong>se samples to <strong>the</strong> FinnishType 1 Diabetes Register, and testing <strong>the</strong>m allfor <strong>anti</strong>-<strong>GAD</strong>, we were able to show that up to10 years prior to a woman developing Type 1diabetes, <strong>anti</strong>bodies to <strong>GAD</strong> were present. Thiswas a critical study that confirmed <strong>the</strong> utility of<strong>the</strong> <strong>anti</strong>-<strong>GAD</strong> test as a powerful weapon for <strong>the</strong>prediction of Type 1 diabetes, and establishedthat <strong>the</strong> “latent period” for an autoimmune diseasecould be very long indeed.Later, in a landmark collaboration <strong>with</strong> <strong>the</strong>late Professor Robert Turner on <strong>the</strong> UKPDScohort, we were able to demonstrate that 10% of<strong>the</strong> “pedigree” Type 2 diabetics of this cohortactually had LADA. It is now well demonstratedthat this 10% figure applies in many countriesaround <strong>the</strong> world in terms of <strong>the</strong> number ofsubjects “misclassified” as having Type 2 diabetes.Of course, early prediction of Type 1 diabetesin its long preclinical phase is not that much of“It is now welldemonstrated that this 10%figure applies in many countriesaround <strong>the</strong> world interms of <strong>the</strong> number of subjects“misclassified” as”having Type 2 diabetesa blessing in <strong>the</strong> absence of an effective preventiveregimen. At <strong>the</strong> time of our studies onLADA referred to above, <strong>the</strong>re was a peaking ofinterest in administration of auto<strong>anti</strong>genic preparationsmucosally (oral tolerance) to abrogateautoimmune disorders diabetes included.Administration of <strong>GAD</strong> to NOD mice to retardonset of diabetes, in our studies and those ofo<strong>the</strong>rs, have given at best “promising” results.While earlier enthusiasm for oral tolerance maybe diminishing, we certainly look forward toseeing results of human trials of <strong>the</strong> preventativeuse of <strong>GAD</strong> in LADA.Paul Zimmet isDirector of <strong>the</strong>InternationalDiabetes Instituteand Professor ofDiabetes, MonashUniversity inMelbourne, Australia.His current research includes Type 1 diabetesetiology and <strong>the</strong> molecular mechanismsof Type 2 diabetes, insulin resistance andobesity and <strong>the</strong> effects of life-style changeleading to diabetes, obesity, coronary heartdisease and hypertension in developingcountries in <strong>the</strong> Asia-Pacific region.Intraperitoneal injection of <strong>GAD</strong>65prevents diabetes in NOD mice100 NaClMBP<strong>GAD</strong>-MBP8060402000 10 20 30<strong>GAD</strong>treatment Age (weeks)Pleau et al. (1995). Prevention of autoimmunediabetes in nonobese diabetic female mice bytreatment <strong>with</strong> recombinant glutamic aciddecarboxylase (<strong>GAD</strong>65). Clin Immunol Pathol76:90-95Diabetic animals %Stimulation index log100101<strong>GAD</strong> in Graphs• 4 wk old NOD mice which hadbeen injected ip 3 times <strong>with</strong> <strong>GAD</strong>65were protected from diabetes developmentcompared to mice injected<strong>with</strong> fusion protein (MBP) or saline(NaCl)• ip administration of <strong>GAD</strong>65prevents diabetes<strong>GAD</strong>65 is a key target <strong>anti</strong>gen in <strong>the</strong>induction of murine IDDM<strong>GAD</strong>hsp65cbphinsulin3 4 6 8 12 15Age of mice (weeks)Kaufman et al. (1993) Spontaneous loss of T celltolerance to glutamic acid in murine insulindependentdiabetes. Nature 366:69-72• Spontaneous T cell proliferativeresponses in NOD micefirst develop to <strong>GAD</strong>65, <strong>the</strong>n tohsp65, carboxypeptidase H andto insulin in a defined order.• <strong>GAD</strong>65 is a key diabetogenicauto<strong>anti</strong>gen in murine IDDMdmccad june 2003page 35


References1. Kobayashi T, et al,IsleT cell <strong>anti</strong>bodies in Insulin-dependent and non-insulindependent diabetics in Japan: <strong>the</strong>ir prevalence andclinical significance. In clinico-genetic genesis of diabetesmellitus. Mimura G, Baba S, Goto Y, Kobberling J,Eds. Amsterdam,Excerpta Med 150-160 (ICS No. 597), 1982.2. Kobayashi T, et al,Time course of islet cell <strong>anti</strong>bodies and beta cell functionin non-insulin-dependent stage of Type I diabetes.Diabetes 36: 510-7, 1987.3. Kobayashi T, et al,Maleness as risk factor for slowly progressive IDDM.Diabetes Care 12: 7-11, 1989.4. Kobayashi TSubtype of insulin-dependent diabetes mellitus (IDDM)in Japan: slowly progressive IDDM-<strong>the</strong> clinical characteristicsand pathogenesis of <strong>the</strong> syndrome.Diabetes Res Clin Pract 24 Suppl: S95-9. Review, 1994.5. Kobayashi T, et al,<strong>GAD</strong> <strong>anti</strong>bodies seldom disappear in slowly progressiveIDDM.Diabetes Care 19: 1031, 1996.6. Kobayashi T, et al,Immunogenetic and clinical characterization of slowlyprogressive IDDM.Diabetes Care 16: 780-8, 1993.7. Nakanishi K, et al,Predictive value of insulin auto<strong>anti</strong>bodies for fur<strong>the</strong>rprogression of beta cell dysfunction in non-insulindependentdiabetics.Diabetes Res 9: 105-109, 1988.8. Nakanishi K, et al,Exocrine pancreatic ductograms in insulin-dependentdiabetes mellitus.Am J Gastroenterol 89: 762-6,1994.9. Nakanishi K, et al,Relationships among residual beta cells, exocrine pancreas,and islet cell <strong>anti</strong>bodies in insulin-dependent diabetesmellitus.Metabolism. 142: 196-203, 1993.10. Groop LC, et al,Islet cell <strong>anti</strong>bodies identify latent Type I diabetes inpatients aged 35-75 years at diagnosis.Diabetes 35: 237-41, 1986.11. Zimmet PZ, et al,Latent autoimmune diabetes mellitus in adults(LADA): <strong>the</strong> role of <strong>anti</strong>bodies to glutamic acid decarboxylasein diagnosis and prediction of insulin dependency.Diabet Med 11: 299-303, 1994.12. Thai AC, et al,Anti-<strong>GAD</strong> <strong>anti</strong>bodies in Chinese patients <strong>with</strong> youthand adult-onset IDDM and NIDDM. Diabetologia 40:1425-1430, 1997.13. Sutanegara D, et al,The epidemiology and management of diabetes mellitusin Indonesia.Diabetes Res Clin Pract 50 Suppl 2: S9-S16, 2000.14. Brooks-Worrell BM, et al,Cellular immune responses to human islet proteins in<strong><strong>anti</strong>body</strong>-positive Type 2 diabetic patients. Diabetes48: 983-8, 1999.15. Kobayashi T, et al,Small doses of subcutaneous insulin as a strategy forpreventing slowly progressive beta cell failure in isletcell <strong><strong>anti</strong>body</strong>-positive patients <strong>with</strong> clinical features ofNIDDM.Diabetes 45: 622-6, 1996.16. Nakanishi K, et al,Residual beta cell function and HLA-A24 in IDDM.Markers of glycemic control and subsequent developmentof diabetic retinopathy.Diabetes 44: 1334-9, 1995.17. Kobayashi T, et al,Insulin Intervention to Preserve ß-cells in SlowlyProgressive Insulin-Dependent (Type 1) DiabetesMellitus.Ann NY Acad Sci 958: 117-30, 2002.Tetsuro Kobayashi, MD, Ph.D., KanazawaUniversity School of Medicine, Kanazawa,Professor and Chairman, Third Departmentof Internal Medicine, School of Medicine,University of Yamanashi, Tamaho City,Yamanashi, 409-3898 Japan 1968-1974.Chief, Department of Endocnology andMetabolism, Toranomon Hospital andOkinaka Memorial Institute of MedicalResearch, Tokyo. Main Works: Discovery of<strong>the</strong> presence of SPIDDM, Insulin interventionof SPIDDM.Intervention to Preserve ß-Cells in SPIDDMTetsuro Kobayashi , Shoichiro Tanakaa, Kaoru Aidaa and Taro MaruyamabSince <strong>the</strong> late 1970’s our laboratoryhas made rigorous efforts to examine<strong>the</strong> clinical significance of <strong>the</strong> presenceof islet cell <strong>anti</strong>bodies (ICA) inpatients <strong>with</strong> non-insulin-dependentdiabetes mellitus (NIDDM) (1-6). Wehave found that <strong>the</strong> clinical features of ICA-positiveNIDDM patients are largely different fromICA-negative NIDDM patients because ß-cell dysfunctionis progressive and most of <strong>the</strong>m lapse intoan insulin dependent state indistinguishable fromthat of insulin-dependent diabetes mellitus(IDDM) (2-6). In 1982, we described ICA-positiveNIDDM as slowly progressive insulin-dependentdiabetes mellitus (SPIDDM) based on <strong>the</strong> characteristicclinical courses (1). The clinical characteristicsof SPIDDM include; 1) Late age onset <strong>with</strong> an initialclinical phenotype of NIDDM <strong>with</strong> progressiveß-cell failure and subsequent features of IDDM (1-4); 2) Persistent pancreatic humoral autoimmunemarkers including glutamic acid decarboxylaseauto<strong><strong>anti</strong>body</strong> (<strong>GAD</strong>Ab), ICA, ICA512/IA-2 auto<strong>anti</strong>bodies(IA-2Ab) and insulin auto<strong>anti</strong>bodies (IAA)(2, 5, 7); 3) Male predominance (3, 6); 4)Involvement of exocrine as well as endocrine pancreas(8); 5) Less marked insulitis <strong>with</strong> preserved ß-cell mass (9); 6) Association <strong>with</strong> specific HLA-DQA1*0302-DQB1*0401 haplotype (6). After developmentof a convenient <strong>GAD</strong>Ab assay, an increasingnumber of studies have shown that NIDDM patientswho have <strong>GAD</strong>Ab as well as ICA are confirmedto be distinct from <strong>GAD</strong>Ab-negative NIDDMpatients and are called latent Type 1 diabetes (10)or latent autoimmune diabetes in adults (LADA)(11).The clinical importance for intervention to maintainß-cell function or to prevent ß-cell failure inSPIDDM is based on <strong>the</strong> following points: 1)SPIDDM is more prevalent than classical IDDMand <strong>the</strong> prevalence is as high as 10% amongNIDDM patients in some ethnic groups includingCaucasian (10, 11), Japanese (5), Chinese (12),Indonesian (13), and Thai (4) populations. 2) <strong>the</strong> Tcell response to islet <strong>anti</strong>gens in SPIDDM is weakerthan that in classical IDDM (14). 3) A pilot studydemonstrated that small doses of insulin prevent ß-cell failure in SPIDDM patients (15). 4) Insulinsecreted from preserved ß-cells in SPIDDM contributesto stable glycemic control and subsequentlyprevents late diabetic complications (16). In 1996,we organized a multicenter randomized clinical trial(The Tokyo Study) to examine <strong>the</strong> effect of earlytreatment <strong>with</strong> insulin in SPIDDM. At seven hospitalsin <strong>the</strong> Tokyo area, about 4000 NIDDM patientswere screened for auto<strong>anti</strong>bodies against <strong>GAD</strong>.Patients were randomly assigned to one of twogroups. One group received subcutaneous insulininjection (Insulin group) and <strong>the</strong> o<strong>the</strong>r received oralsulfonylurea (SU group). The primary outcomemeasures for <strong>the</strong> study were serum C-peptideresponse and level of blood glucose during 75gOGTT.During <strong>the</strong> trial C-peptide responses to OGTT(Sigma C-peptide) decreased progressively in <strong>the</strong> SUgroup and became significant at 24 and 36 months(17). Seven patients lapsed into an insulin-dependentstage when <strong>the</strong>ir sigma C-peptide reached lessthan 4 ng/ml. In contrast, <strong>the</strong> sigma C-peptidevalue remained unchanged in <strong>the</strong> patients in <strong>the</strong>Insulin group and <strong>the</strong> value was significantly differentfrom that of <strong>the</strong> SU group at 36 months (17).Our multicenter randomized study demonstratedthat insulin intervention is effective and safe for gradualß-cell failure in SPIDDM, specifically in <strong>the</strong> patients<strong>with</strong> preserved ß-cell function and high titer of<strong>GAD</strong>Ab at <strong>the</strong> initiation of insulin.In a recent study, we have found <strong>GAD</strong>Abs to aunique epitope in <strong>the</strong> N-terminal region of <strong>the</strong><strong>GAD</strong>65 molecule. This region includes anchoringdomains of <strong>GAD</strong>65 molecules, which potentiallycan be accessed by <strong>GAD</strong>Ab during <strong>the</strong> exocytosisof GABA from <strong>the</strong> ß-cell (unpublished data). Theseresults open <strong>the</strong> door to <strong>the</strong> prevention of ß-cell failureby vaccination of <strong>GAD</strong> in SPIDDM patients,because <strong>GAD</strong>Ab may have a causative role in ß-celldysfunction and vaccination <strong>with</strong> <strong>GAD</strong> may modify<strong>the</strong> pathological process of ß-cell failure in thissyndrome.page 36 dmccad june 2003


<strong>GAD</strong> Antibodies and LatentAutoimmune Diabetes of <strong>the</strong>Adult (LADA)TA.G. Unnikrishnan and S. K. Singhhe prevalence of Type 2 diabetesis rapidly increasing in <strong>the</strong>Indian subcontinent.While <strong>the</strong>majority of subjects <strong>with</strong> Type2 diabetes in developed countriesare obese, those fromIndia are mostly non-obese, and many of <strong>the</strong>m arelean (1). The proposed hypo<strong>the</strong>ses to explain thisinclude coexistent malnutrition, autoimmunityand metabolic abnormalities.Recent research from North India shows thatone-fourth of <strong>the</strong> recently diagnosed Type 2 diabeticswho are lean (i.e <strong>with</strong> a body mass index ofless than 18.5 kg/m 2 ) have <strong>positivity</strong> to <strong>GAD</strong><strong>anti</strong>bodies. These adult subjects <strong>with</strong> autoimmunitywere significantly younger, had a lower waisthip ratio and beta cell function (HOMA) as comparedto lean Type 2 diabetics <strong>with</strong>out <strong><strong>anti</strong>body</strong><strong>positivity</strong>. Hence <strong>the</strong>y had a clinical profile consistent<strong>with</strong> latent autoimmune diabetes of <strong>the</strong>adult (LADA) . They also had lower insulinresistance (HOMA), showing that reduced betacell function is <strong>the</strong> predominant metabolic abnormalityin <strong>the</strong>se subjects (2).The above study included only adult-onsetType 2 diabetes, which is seen commonly in India.There is also evidence that 23% of emaciated, veryyoung subjects <strong>with</strong> a ketosis-resistant form ofdiabetes, a phenotype which is rare yet peculiar toIndia, are positive for <strong>GAD</strong> <strong>anti</strong>bodies (3). Thisform of diabetes has also been termed malnutritionmodulated diabetes mellitus (MMDM).Exciting work is in progress to unravel <strong>the</strong> geneticbasis of Indian diabetics <strong>with</strong> <strong>GAD</strong>65 <strong><strong>anti</strong>body</strong><strong>positivity</strong>. It has been reported that MHC-relatedgenes can discriminate between acute-onset andslow-onset Type 1 diabetes in India, and can alsodistinguish <strong>the</strong> MMDM phenotype (4).Clearly, it is important to avoid <strong>the</strong> misclassificationof <strong>the</strong>se lean diabetic subjects, as some of<strong>the</strong>m could have LADA. The detection of <strong>GAD</strong><strong>anti</strong>bodies could predict <strong>the</strong> early onset of insulindependency, prompting more aggressive glucoselowering<strong>the</strong>rapy. This could prevent unneccessaryexposure to hyperglycemia. Fur<strong>the</strong>r Indian studiesare needed to assess <strong>the</strong> prevalence of eventualbeta cell failure in <strong>the</strong>se subjects, and <strong>the</strong> speed ofprogression.References1. Mohan V, et al,Clinical Profile of lean NIDDM in South India.Diabetes Res Clin Pract 38(2): 101-8, 1997.2. Unnikrishnan AG, et al,Clinical and immunological profile of underweightType 2 diabetes in north India.( Abstract number: A-03-255-EASD). Accepted for presentationat <strong>the</strong> 18th International Diabetes FederationCongress, 24 - 29 August 2003, Paris, France.3. Singh AK, et al,Role of islet autoimmunity in <strong>the</strong> aetiology of differentclinical subtypes of diabetes mellitus in young northIndians.Diabet Med Apr;17(4):275-80, 2000.4. Sanjeevi CB, et al,MHC class I chain-related gene a alleles distinguishmalnutrition-modulated diabetes, insulin-dependent diabetes,and non-insulin- dependent diabetes mellitus patientsfrom eastern India.Ann N Y Acad Sci Apr 958:341-4, 2002.A.G. Unnikrishnan 1 , S. K. Singh 21 Asst. Professor, Department of Diabetesand Endocrinology, Amrita Institute ofMedical Sciences, Kochi, India, 2 Reader,Department of Endocrinology, Institute ofMedical Sciences, Banaras Hindu University,Varanasi, India.dmccad june 2003page 37


References1. A.Falorni, et al,Radioimmunoassays for glutamic acid decarboxylase(<strong>GAD</strong>65) and <strong>GAD</strong>65 auto<strong>anti</strong>bodies using 35S or 3Hrecombinant human ligands.J.Immunol.Methods 186:89-99,19952. C.L.Vandewalle, et al,Belgian Diabetes Registry: High diagnostic sensitivityof glutamate decarboxylase auto<strong>anti</strong>bodies in IDDM<strong>with</strong> clinical onset between age 20 and 40 years.J.Clin.Endocrinol. & Metab. 80:846-851,19953. A.Falorni, et al,Diagnostic sensitivity of immunodominant epitopes ofglutamic acid decarboxylase (<strong>GAD</strong>65) auto<strong>anti</strong>bodiesin childhood IDDM.Diabetologia 39:1091-98,19964. A.Falorni, et al,Auto<strong><strong>anti</strong>body</strong> recognition of COOH-terminal epitopesof <strong>GAD</strong>65 marks <strong>the</strong> risk for insulin requirement inadult-onset diabetes mellitus. J.Clin.Endocrinol.Metab85:309-316,2000.5. L.Avesani, et al,Improved in planta expression of <strong>the</strong> human isletauto<strong>anti</strong>gen glutamic acid decarboxylase (<strong>GAD</strong>65).Transgenic Research 12:203-212,20036. Marcovina et al,Evaluation of a novel radioimmunoassay using 125 I-labelled human recombinant <strong>GAD</strong>65 for <strong>the</strong> determinationof glutamic acid decarboxylase (<strong>GAD</strong>65) auto<strong>anti</strong>bodiesInt J Lab Res (2000) 30: 21-26Alberto Falorni, MD, PhD is AssociateProfessor in Internal Medicine at <strong>the</strong>University of Perugia, Italy. Falorni has specializedin <strong>the</strong> techniques for <strong>the</strong> study ofhumoral autoimmunity and genetics of Type1 diabetes mellitus and o<strong>the</strong>r endocrine autoimmunediseases. Falorni’s clinical activity isfocused on <strong>the</strong> diagnosis and treatment ofendocrine diseases. Falorni’s main scientificinterests are pathogenesis and prevention ofType 1 diabetes mellitus and of autoimmuneprimary adrenal insufficiency.LADA Diagnostics and <strong>GAD</strong>Transgenic PlantsMAlberto Falorniy main research interest ispathogenesis and preventionof Type 1 diabetesmellitus and o<strong>the</strong>r endocrineautoimmune diseases. Ihave initially focused myattention on <strong>the</strong> role of glutamic acid decarboxylase(<strong>GAD</strong>65) as a target molecule of auto<strong>anti</strong>bodies in Type1 diabetes. The development of a semi-automated procedurefor <strong>the</strong> radioimmunological determination of<strong>GAD</strong>65 auto<strong>anti</strong>bodies in human serum (1) has made itpossible to test <strong>the</strong> diagnostic sensitivity (frequency ofType 1 diabetic patients positive) and specificity (frequencyof non-diabetic subjects negative) of this immunemarker for <strong>the</strong> disease. It was shown that <strong>GAD</strong>65 auto<strong>anti</strong>bodiescan be detected in over 80% of recently diagnosedType 1 diabetic patients and occur more frequentlyamong diabetic females than males. Interestingly,<strong>GAD</strong>65Ab have emerged as <strong>the</strong> immune marker athighest diagnostic sensitivity for adult-onset Type 1 diabetes(2) which has paved <strong>the</strong> way to <strong>the</strong> diagnosticuse of this marker in routine clinical practice to discriminateautoimmune from non-autoimmune cases.In <strong>the</strong> attempt of both identifying novel markersat highest diagnostic accuracy for Type 1 diabetesand elucidating <strong>the</strong> molecular mechanisms of auto<strong><strong>anti</strong>body</strong>formation, I have constructed chimeric molecules,generated by substitution of regions of human<strong>GAD</strong>65 <strong>with</strong> homologous regions of <strong>GAD</strong>67 (aLeanType II diabetesLow risk forinsulin requirement-Clinical diagnosis of adult-onset diabetes mellitus28ObeseType II diabetesScreening for o<strong>the</strong>rautoimmune diseases<strong>GAD</strong> isoenzyme which is not a major diabetes-relatedauto<strong>anti</strong>gen), to define <strong>the</strong> epitope regions of <strong>the</strong>auto<strong>anti</strong>gen recognized by human auto<strong>anti</strong>bodies (3).It was shown that human <strong>GAD</strong>65 auto<strong>anti</strong>bodies areprimarily directed against epitopes located in <strong>the</strong>middle and COOH-terminal regions of <strong>the</strong> enzymeand that levels of <strong>GAD</strong>65 <strong>anti</strong>bodies specific for <strong>the</strong>COOH-terminal end of <strong>the</strong> auto<strong>anti</strong>gen discriminateType 1 diabetic children from <strong><strong>anti</strong>body</strong>-positive childrenwho do not progress towards clinical diabetes.In addition, I have constructed a mutant form ofhuman <strong>GAD</strong>65, generated by site-directed mutagenesisof <strong>the</strong> active site of <strong>the</strong> enzyme, which has provenenzymatically inactive but immunologically indistinguishablefrom “wild-type” human <strong>GAD</strong>65.More recently, I have focused my interest on <strong>the</strong>diagnosis and clinical characteristics of <strong>the</strong> so-calledlatent autoimmune diabetes in adults (LADA). Ihave demonstrated that <strong>GAD</strong>65 <strong>anti</strong>bodies can bedetected in approximately 10% of a hospital-basedpopulation of patients diagnosed <strong>with</strong> Type 2 diabeteson clinical grounds in Italy (4). Among<strong>GAD</strong>65Ab-positive individuals, high <strong><strong>anti</strong>body</strong> levelsand presence of <strong>anti</strong>bodies directed to <strong>the</strong> COOHterminalend of <strong>the</strong> auto<strong>anti</strong>gen predicted an extremelyhigh risk of progression towards insulin-dependencyand of associated organ-specific autoimmunediseases, such as thyroid autoimmune diseases orautoimmune Addison’s disease. In contrast, <strong>the</strong> presenceof low levels of <strong>GAD</strong>65 <strong>anti</strong>bodies directedonly against <strong>the</strong> middle region of <strong>the</strong> enzyme discriminateda sub-population of LADA patients <strong>with</strong>clinical characteristics very similar to that of <strong><strong>anti</strong>body</strong>-negativeType 2 patients.At present, I am testing <strong>the</strong> hypo<strong>the</strong>sis that <strong>the</strong>islet autoimmune process may be modulated and <strong>the</strong>appearance of clinical signs of <strong>the</strong> disease delayed orprevented by <strong>the</strong> oral administration of recombinanthuman <strong>GAD</strong>65. To make this strategy potentiallyapplicable to clinical studies of primary prevention,we have constructed transgenic plants expressinghuman <strong>GAD</strong>65 (5), that can potentially allow us toadminister <strong>the</strong> human auto<strong>anti</strong>gen <strong>with</strong>out <strong>the</strong> needfor costly and time-consuming procedures of proteinpurification. We have demonstrated that, in transgenicplants, <strong>GAD</strong>65 accumulates in chloroplast tylacoidsand mitochondria and that <strong>the</strong> targeting ofhuman <strong>GAD</strong>65 to <strong>the</strong> plant cell cytosol (by substitutionof <strong>the</strong> NH 2 -terminal end of <strong>the</strong> protein <strong>with</strong> ahomologous region of <strong>GAD</strong>67) is associated <strong>with</strong> a 5-fold increase of expression levels. Ongoing studies aretesting <strong>the</strong> effect of <strong>the</strong> oral administration of transgenicplant material containing human <strong>GAD</strong>65 inanimal models of spontaneous autoimmune diabetes.page 38 dmccad june 2003


ReferencesT1DM and LADA Differ in<strong>GAD</strong>A Epitope SpecificityDChristiane Hampe, Rattan Juneja, Åke Lernmark, Jerry Palmeriabetes Mellitus is classifiedinto two major forms, Type1 and Type 2 diabetes. Type1 diabetes is characterizedby an autoimmune-mediateddestruction of betacells,leading to insulin deficiency. The autoimmunereaction involves both T cells and <strong>anti</strong>bodies directedagainst islet cell auto<strong>anti</strong>gens that can be detectedin <strong>the</strong> majority of Type 1 diabetes patients (1, 2).The main auto<strong>anti</strong>gens identified are insulin (3), <strong>the</strong>Mr 65,000 isoform of glutamic acid decarboxylase(<strong>GAD</strong>65) (4), and <strong>the</strong> tyrosine phosphatase-like IA-2<strong>anti</strong>gen (5). These auto<strong>anti</strong>bodies are often detectedlong before <strong>the</strong> clinical onset of Type 1 diabetes andare useful to predict disease risk (5, 6). <strong>GAD</strong>65 andIA-2 auto<strong>anti</strong>bodies (Ab) are readily detected bynow standardized (7), precise and reproducible radioimmunoassays(4, 7, 8) suitable for large scale analysisand population screening (6, 9). In contrast, classicalType 2 diabetes patients do not show evidence ofautoimmune beta cell destruction. Patients <strong>with</strong>Type 1 diabetes usually require insulin treatment at<strong>the</strong> time of diagnosis whereas Type 2 patients can besuccessfully treated by diet and oral agents for manyyears. These patients do not show evidence of autoimmunebeta cell destruction. A third group of patientsis referred to as latent autoimmune diabetes inadults (LADA) (10), Type 1.5 diabetes (11), or slowlyprogressive insulin dependent diabetes mellitus(SPIDDM) (12). These patients lose beta cell function,fail oral agents early and require insulin treatment(13, 14). Evidence for an underlying autoimmmunepathogenesis is provided by <strong>the</strong> observationthat many of <strong>the</strong>se patients have islet cell <strong>anti</strong>bodies(ICA), auto<strong>anti</strong>bodies to <strong>GAD</strong>65 (<strong>GAD</strong>65Ab) (10,15, 16), or both. The presence of <strong>GAD</strong>65Ab alone isa sufficient marker for future insulin requirementin younger patients (44 years or younger) while inolder patients <strong>positivity</strong> for both ICA and<strong>GAD</strong>65Ab is a stronger predictor of insulin requirement(17). The question has been raised whe<strong>the</strong>rType 1.5 diabetes represents a separate clinical diseaseor is a slowly progressive form of Type 1 diabetes(18, 19). Epitope mapping of <strong>GAD</strong>65Ab can assist in<strong>the</strong> classification of <strong>the</strong> underlying autoimmunity.Using both <strong>GAD</strong>65/67 fusion proteins (20, 21) and<strong>GAD</strong>65-specific recombinant Fab we and o<strong>the</strong>rswere able to identify phenotype-specific <strong>GAD</strong>65Abepitopes. <strong>GAD</strong>65Ab in newly diagnosed young Type1 diabetes patients recognize restricted epitopes primarilylocated at <strong>the</strong> combined middle-carboxyterminalconformational epitope of <strong>GAD</strong>65, while bindingto <strong>GAD</strong>67 or <strong>the</strong> N-terminus of <strong>GAD</strong>65 isdetected only at a low level. In contrast, <strong>GAD</strong>65Abpositive Type 1.5 diabetes patients exhibit a<strong>GAD</strong>65Ab epitope pattern that is characterized bybinding to both <strong>the</strong> N-terminus of <strong>GAD</strong>65 and to atentative conformational epitope formed of <strong>the</strong>middle and carboxyterminal part of <strong>GAD</strong>65 (22).This <strong>GAD</strong>65Ab epitope profile clearly differs fromthat found in Type 1 diabetes patients and moreresembles <strong>the</strong> broader <strong>GAD</strong>65Ab epitope specificityfound in <strong>GAD</strong>65Ab-positive healthy individuals andfirst-degree relatives (20). This difference in <strong>the</strong> bindingpattern of <strong>GAD</strong>65Ab of Type 1.5 diabetes patientscompared to that of Type 1 diabetes patientssupports <strong>the</strong> notion that <strong>the</strong> disease process maydiffer between <strong>the</strong>se two types of patients. We <strong>the</strong>reforesuggest that Type 1.5 diabetes might be a subtypeof Type 1 diabetes characterized by separateimmunologic features. <strong>GAD</strong>65Ab epitope patternsmay be useful to identify Type 1.5.Christiane Hampe, Ph.D., has a position asjunior faculty at <strong>the</strong> University of Washingtonin Seattle. Her research interests are <strong>the</strong> disssectionof <strong>the</strong> role of <strong>GAD</strong>65 and its auto<strong>anti</strong>bodiesin <strong>the</strong> pathogenesis of Type 1 diabetes.While <strong>the</strong>se auto<strong>anti</strong>bodies are widely acceptedas markers for <strong>the</strong> disease, preliminarydata indicate that disease-specific <strong>GAD</strong>65Abmodulate T cell responses. Hampe’s currentresearch goals are to understand <strong>the</strong> effect of<strong>GAD</strong>65Ab on processing and presentation of<strong>GAD</strong>65.1. Bonifacio E, et al,Islet auto<strong><strong>anti</strong>body</strong> markers in IDDM: risk assessmentstrategies yielding high sensitivity.Diabetologia 38:816-822, 19952. Landin-Olsson M, et al,Islet cell and o<strong>the</strong>r organ-specific auto<strong>anti</strong>bodies in allchildren developing Type 1 (insulin-independent) diabetesmellitus in Sweden during one year and in matchedcontrols.Diabetologia 32:387-395, 19893. Palmer JP, et al,Insulin <strong>anti</strong>bodies in insulin-dependent diabetics beforeinsulin treatment. Science 222:1337-1339, 19834. Grubin CE, et al,A novel radioligand binding assay to determine diagnosticaccuracy of isoform-specific glutamic acid decarboxylase<strong>anti</strong>bodies in childhood IDDM.Diabetologia 37:344-350, 19945. Verge CF, et al,Prediction of Type I diabetes in first-degree relativesusing a combination6. Bingley PJ, et al,Prediction of IDDM in <strong>the</strong> general population:Strategies based on combinations of auto<strong><strong>anti</strong>body</strong>markers.Diabetes 46:1701-1710, 19977. Mire-Sluis AR, et al,The development of a World Health Organisationinternational standard for islet cell <strong>anti</strong>bodies: <strong>the</strong> aimsand design of an international collaborative study.Diabetes Metab Res Rev 15:72-77, 19998. Verge CF, et al,Combined use of auto<strong>anti</strong>bodies (IA-2) auto<strong><strong>anti</strong>body</strong>,<strong>GAD</strong> auto<strong><strong>anti</strong>body</strong>, insulin auto<strong><strong>anti</strong>body</strong>, cytoplasmicislet cell <strong>anti</strong>bodies) in Type 1 diabetes: CombinatorialIslet Auto<strong><strong>anti</strong>body</strong> Workshop.Diabetes 47:1857-1866, 19989. Rolandsson O, et al,Glutamate decarboxylase (<strong>GAD</strong>65) and tyrosine phosphatase-likeprotein (IA-2) auto<strong>anti</strong>bodies index in aregional population is related to glucose intoleranceand body mass index.Diabetologia 42:555-559, 199910. Tuomi T, et al,Antibodies to glutamic acid decarboxylase reveallatent autoimmune diabetes mellitus in adults <strong>with</strong> anon-insulin-dependent onset of disease.Diabetes 42:359-362, 199311. Harris MI, et al,Classification of diabetes mellitus and o<strong>the</strong>r catagoriesof glucose intolerance. In: Keen H, DeFronzo R,Alberti K, Zimmet P, ed.The international textbook of diabetes mellitus.London: Wiley, 1992, 3-18.12. Ludvigsson J, et al,HLA-DR3 is associated <strong>with</strong> amore slowly progressiveform of Type 1 (insulin-dependent) diabetes.Diabetologia 29:207-210, 198613. Temple RC, et al,Insulin deficiency in non-insulin-dependent diabetes.Lancet 1:293-295, 198914. Gjessing HJ, et al,Fasting plasma c-peptide, glucagon stimulated plasmac-peptide, and urinary c-peptide in relation to clinicaltype of diabetes.Diabetologia 32:305-311, 198915. Groop LC, et al,Islet cell <strong>anti</strong>bodies identify latent Type 1 diabetes inpatients aged 35-75 years at diagnosis.Diabetes 35:237-241, 198616. Rowley MJ, et al,Antibodies to glutamic acid decarboxylase discriminatemajor types of diabetes mellitus.Diabetes 41:548-551, 199217. Turner R, et al,UKPDS 25: auto<strong>anti</strong>bodies to isle T cell cytoplasm andglutamic acid decarboxylase for prediction of insulinrequirement in Type 2 diabetes.UK Prospective Diabetes Study Group [published erratumappears in Lancet 1998 Jan 31;351 (9099): 376].Lancet 350:1288-1293, 199718. Juneja R, et al,Autoimmunity 29:65-83, 199919. Tuomi T, et al,Clinical and genetic characteristics of Type 2 diabetes<strong>with</strong> and <strong>with</strong>out <strong>GAD</strong> <strong>anti</strong>bodies.Diabetes 48:150-157, 199920. Hampe CS, et al,Recognition of Glutamic Acid Decarboxylase (<strong>GAD</strong>)by Auto<strong>anti</strong>bodies from Different <strong>GAD</strong> Antibody-Positive Phenotypes.J Clin Endocrinol Metab 85:4671-4679, 200021. Falorni A, et al,Diagnostic sensitivity of immunodominant epitopes ofglutamic acid decarboxylase (<strong>GAD</strong>65) auto<strong>anti</strong>bodiesepitopes in childhood IDDM.Diabetologia 39:1091-1098, 199622. Hampe CS, et al,<strong>GAD</strong>65 <strong><strong>anti</strong>body</strong> epitope patterns of Type 1.5 diabeticpatients are consistent <strong>with</strong> slow-onset autoimmunediabetes.Diabetes Care 25:1481-1482., 2002dmccad june 2003page 39


References1. Chattopadhyay, S., et al,An auto<strong><strong>anti</strong>body</strong> inhibitory to glutamic acid decarboxylasein <strong>the</strong> neurodegeneartive disorder Batten disease.Hum. Mol. Genet. 11, 1421-1431, 2002.2. International Batten Disease Consortium. Isolation ofa novel gene underlying Batten disease.Cell 82, 949-957, 1995.3. Mitchison, H. M., et al,Targeted disruption of <strong>the</strong> Cln3 gene provides a mousemodel for Batten disease.Neurobiol. Dis. 6, 321-334, 1999.4. Chattopadhyay, S., et al,An auto<strong><strong>anti</strong>body</strong> to <strong>GAD</strong>65 in sera of patients <strong>with</strong>juvenile neuronal ceroid lipofuscinoses.Neurology. 59, 1816-1817, 2002.David Pearce is Assistant Professor ofBiochemistry and Biophysics in <strong>the</strong> Centerfor Aging and Developmental Biology at <strong>the</strong>University of Rochester School of Medicineand Dentistry, Rochester, NY. Pearce is researching<strong>the</strong> molecular basis of juvenileneuronal ceroid lipofuscinosis (JNCL), orBatten disease. Pearce’s group discovered <strong>the</strong>presence of auto<strong>anti</strong>bodies to <strong>GAD</strong>65 inindividuals <strong>with</strong> Batten disease as well as ina mouse model for <strong>the</strong> human disease.<strong>GAD</strong> and Batten’s DiseaseTDavid Pearcehe discovery that individuals<strong>with</strong> juvenile neuronal ceroidlipofuscinosis, or Batten diseasehave circulating auto<strong>anti</strong>bodiesto <strong>GAD</strong>65 is perhaps<strong>the</strong> most recent chapter on<strong>GAD</strong>65 auto<strong>anti</strong>bodies and disease, and certainlyone that requires fur<strong>the</strong>r exploration. We firstpublished that individuals <strong>with</strong> Batten disease, anda mouse model for <strong>the</strong> disease had circulatingauto<strong>anti</strong>bodies to <strong>GAD</strong>65 in Human MolecularGenetics in 2002 (1). The circumstances that ledto this report do not necessarily follow conventionalscientific reasoning, as much as to intuition.Batten disease is a pediatric onset devastatingfatal neurodegenerative disease. Individuals <strong>with</strong>Batten disease have inherited mutations in bothcopies of a still to be functionally characterizedgene product, designated CLN3 (2). We were studyinga mouse model that lacks a CLN3-gene productconstructed by o<strong>the</strong>rs (3). Gene expressionstudies indicated that in <strong>the</strong> brain of cln3-knockoutmice, a shift in <strong>the</strong> expression of enzymesassociated to <strong>the</strong> syn<strong>the</strong>sis and utilization of <strong>the</strong>neurotransmitter glutamate was altered. We nextconfirmed that endogenous levels ofglutamate/glutamic acid were elevated in <strong>the</strong>brains of cln3-knockout mice. There was no reasonto suspect, and still isn’t, that <strong>the</strong> CLN3-gene productis directly involved in glutamate metabolism.I convinced Subrata Chattopadhyay <strong>the</strong> postdoctoralfellow working <strong>with</strong> me on this to use serumdrawn from <strong>the</strong> cln3-knockout mice as <strong>the</strong> primary<strong><strong>anti</strong>body</strong> in a western blot against a brain proteinextract. The basis for this was that a block in<strong>GAD</strong> activity, which converts glutamate to GABAcould be mediated by <strong>the</strong> presence of an auto<strong><strong>anti</strong>body</strong>to <strong>GAD</strong>, thus causing <strong>the</strong> elevation in glutamatethat we observed. It turned out that <strong>the</strong>cln3-knockout mice sera had circulating auto<strong>anti</strong>bodiesreactive to a number of brain proteins.Most notably, <strong>the</strong>re was a predominant protein ofaround 65kD. We knew that this would be<strong>GAD</strong>65, we just had to prove it. Thanks to <strong>the</strong>vast amount of research on <strong>GAD</strong>65 in diabetesand stiff persons syndrome we were able to benefitfrom reagents available and a plethora ofpublished techniques. Within <strong>the</strong> space of a fewmonths Subrata confirmed that <strong>the</strong> cln3-knockoutmice in addition to having elevated glutamate in<strong>the</strong> brain, also had a decrease in enzymatic activityof <strong>GAD</strong>, most likely due to a demonstrated associationof auto<strong><strong>anti</strong>body</strong> in <strong>the</strong> brain. His in vitrostudies showed that serum drawn from <strong>the</strong> cln3-knockout mice also contained an element thatcould inhibit <strong>the</strong> activity of <strong>GAD</strong>, and that wecould block <strong>the</strong> reactivity of <strong>the</strong> auto<strong><strong>anti</strong>body</strong> to<strong>GAD</strong>65 by pre-incubation <strong>with</strong> recombinant<strong>GAD</strong>. At this point I felt we had biochemicallymet <strong>the</strong> criteria of showing that <strong>the</strong> auto<strong><strong>anti</strong>body</strong>could block an enzyme, <strong>GAD</strong>, resulting in accumulationof it’s substrate, glutamate, and that associationto <strong>the</strong> brain resulted in a decrease in <strong>the</strong>activity of <strong>the</strong> enzyme. The final piece of this initialstudy was to show a possible link between <strong>the</strong>auto<strong><strong>anti</strong>body</strong> and <strong>the</strong> disease. I might add that thisin many ways is an ongoing task.Never<strong>the</strong>less, this is where Jim Powers, a neuropathologistcame to <strong>the</strong> rescue. With <strong>the</strong> aid of hispostdoctoral fellow, Masumi Ito, he confirmed adecrease in <strong>GAD</strong>65 positive neurons in Batten diseasepost-mortem brain. I might add that Jim did ahuge amount of o<strong>the</strong>r work that aided our understandingof this phenomenon, and most importantly,he believed what initially was a strange idea,namely that <strong>GAD</strong>65 auto<strong>anti</strong>bodies may be presentin <strong>the</strong> disease. Of course we have gone on toshow that individuals <strong>with</strong> Batten disease haveauto<strong>anti</strong>bodies to <strong>GAD</strong>65, in fact to date everychild we have tested has come up positive.Fur<strong>the</strong>rmore, we have shown that o<strong>the</strong>r pediatricneurodegenerative diseases that fall in a similarcategory to Batten disease do not have circulatingauto<strong>anti</strong>bodies to <strong>GAD</strong>65 (4). Our focus right nowis to deduce whe<strong>the</strong>r <strong>GAD</strong>65 auto<strong>anti</strong>bodies arereally pathological, or simply epi-phenomenal inBatten disease. This of course is a mammoth biologicaltask, however, thanks again to <strong>the</strong> resourcesalready put forth in researching <strong>GAD</strong>65 auto<strong>anti</strong>bodiesin o<strong>the</strong>r diseases, and some excellent collaborations<strong>with</strong> researchers in this field, I believe wewill again benefit in expediting answering thisquestion. Importantly, establishing <strong>the</strong> potentialcontribution of <strong>the</strong> auto<strong><strong>anti</strong>body</strong> to Batten disease,will reveal whe<strong>the</strong>r or not this is an element of <strong>the</strong>disease process that should be targeted.page 40 dmccad june 2003


<strong>GAD</strong> and Parkinson’s DiseaseOHelen Fitzsimons and Mat<strong>the</strong>w Duringur primary interest in<strong>GAD</strong> has been in <strong>the</strong> developmentof gene <strong>the</strong>rapybased treatments for neurologicaldisorders that involveabnormalities in neuronalactivity. As neurotransmission in <strong>the</strong> brain canbe modulated by manipulation of GABA levels,we are using adeno-associated viral vector (AAV)-mediated gene transfer of <strong>GAD</strong> into targetedbrain areas to augment inhibition. The expressionof <strong>GAD</strong> in neurons, which contain high intracelllularlevels of glutamate and co-factors, leads tobiosyn<strong>the</strong>sis and release of GABA, which actingon GABAA receptors facilitates transport of chlorideions into <strong>the</strong> cell, hyperpolarization and relevantsilencing of cell activity. AAV is our preferrredgene delivery vector as it provides a high levelof stable gene expression <strong>with</strong> minimal inflammatoryor immune response (Xu et al, 2001;Mastakov et al, 2002) and in addition, direct injectionof AAV allows transduction of specificneuronal pathways <strong>with</strong>out affecting <strong>the</strong> rest of<strong>the</strong> brain (Xu et al, 2001; During et al, 2003).Our main focus to date has been on <strong>the</strong> developmentof an AAV<strong>GAD</strong> gene <strong>the</strong>rapy based treatmentfor Parkinson’s disease (PD). The motorabnormalities of PD are caused by alterations inbasal ganglia network activity. Briefly, <strong>the</strong> deathof dopaminergic neurons in <strong>the</strong> subst<strong>anti</strong>a nigrapars compacta and <strong>the</strong> associated depletion ofdopamine in <strong>the</strong> striatum causes disinhibition of<strong>the</strong> subthalamic nucleus (STN). This in turn causesoveractivation of <strong>the</strong> output nuclei of <strong>the</strong>basal ganglia, <strong>the</strong> subst<strong>anti</strong>a nigra pars reticulata(SNr) and <strong>the</strong> internal segment of <strong>the</strong> globus palllidus,leading to impaired motor function.We rationalized that an increase in GABAergictransmission from <strong>the</strong> STN to <strong>the</strong> SNr wouldlead to suppression of firing activity of SNrneurons. AAV<strong>GAD</strong>65 or AAV<strong>GAD</strong>67 gene transferto <strong>the</strong> rat STN via stereotactic surgery resultedin robust expression of recombinant <strong>GAD</strong>protein that was restricted to STN neurons, and afour-fold increase in GABA release from <strong>the</strong> SNrfollowing stimulation of <strong>the</strong> STN. Single unitrecording from <strong>the</strong> SNr following stimulation of<strong>the</strong> STN showed rare (6%) inhibitory responses incontrol rats, which was increased to 78% in<strong>GAD</strong>65-treated rats (P


<strong>Diamyd</strong>’s Commercial Development of a <strong>GAD</strong> VaccineJohn Robertson<strong>Diamyd</strong> Medical has been pioneering both <strong>the</strong> evaluation of clinical safetyand clinical efficacy of <strong>GAD</strong> – <strong>with</strong> a view to its potential use as a vaccine toprevent autoimmune diabetes. Both <strong>the</strong>se achievements were made possibleby <strong>Diamyd</strong>’s early definition of a manufacturing process – capable ofproviding <strong>the</strong> qu<strong>anti</strong>ty and quality of <strong>GAD</strong> required for different stages ofcommercial drug development.Our manufacturing process relies on <strong>the</strong> expression ofrecombinant human <strong>GAD</strong>65 in an insect cell line -grown under special conditions – after infection <strong>with</strong>an insect specific baculovirus containing <strong>the</strong> <strong>GAD</strong>cDNA (our “baculo<strong>GAD</strong>” recombinant clone). This is referred to as<strong>the</strong> baculovirus/insect cell expression system (or BVES). Both <strong>the</strong>qu<strong>anti</strong>ty and quality of <strong>GAD</strong> manufactured by our BVES processhave proven appropriate up to <strong>the</strong> current stage of development – andseem likely to meet our future requirements up to market introduction.While currently at <strong>the</strong> 50 litre scale (<strong>with</strong> each 50L batch providingsufficient <strong>GAD</strong> for thousands of vaccinations) we have alreadyfound that our process can readily be scaled-up to 500 litres (providingtens of thousands of vaccinations per batch). So, pending <strong>the</strong>successful outcome of our Phase II, our manufacturing process seemscapable of producing sufficient <strong>GAD</strong> for fur<strong>the</strong>r clinical developmentand market introduction.Apart from its suitability for manufacturing active <strong>GAD</strong>, <strong>the</strong>BVES also has inherent safety advantages – as a non-mammalianexpression system modified to avoid contact <strong>with</strong> any mammaliancomponents. This implies <strong>the</strong> low risk of contamination of our vaccineby harmful viruses. Moreover, because <strong>the</strong> vast majority of humanviruses are not able to grow in <strong>the</strong> insect cells used, <strong>the</strong> likelihood of<strong>the</strong>se inadvertently being propagated during manufacture and contaminatingour vaccine is considerably reduced. Similarly, because <strong>the</strong>“baculo<strong>GAD</strong>” recombinant clone is an insect-specific virus (that cannot infect mammalian cells) <strong>the</strong> risk imposed by possible residual tracesof residual virus in our vaccine is greatly reduced.A final attribute of our <strong>GAD</strong> <strong>the</strong>rapeutic strategy has recentlySf9Insectcells<strong>Diamyd</strong> - cGMP manufacturerh<strong>GAD</strong>65 Bulk Drug50LfermenterExtract/PurifyrhGA D65Bulk Drug<strong>Diamyd</strong><strong>GAD</strong>recombinantbaculovirusFormulation<strong>Diamyd</strong>Formulation/FillAlum<strong>Diamyd</strong> Vaccinebecome apparent. Despite only one (or a few) small regions (ca. 12amino acids) of <strong>GAD</strong> being thought as likely to be active, we made<strong>the</strong> decision at <strong>the</strong> outset (now 9 years ago) that we would not riskpage 42 dmccad june 2003


No Impact on Experimental Autoimmune Encephalomyelitis DevelopmentClinical Score3.532.521.510.50-70 7 8 9 10 11 12 13 14 15 16 17 18DayPBSALUM-<strong>GAD</strong> 2 µgALUM-<strong>GAD</strong> 20 µgALUM-<strong>GAD</strong> 200 µgCTRL-ALUMMean cpm immunoprecipitated<strong>with</strong> serum dilution 1:1000Immunogeneticity of <strong>GAD</strong> is Increased <strong>with</strong> Use of Adjuvant100009000800070006000500040003000200010000Mean <strong>GAD</strong> bufferMean ALUM-<strong>GAD</strong>Mean IFA-<strong>GAD</strong>Assoc. Prof. Robert Harris CMM, Karolinska Institute, Sweden, 2000• Clinical Course of MBP 63-88 –EAE in Lewis ratsProf. Åke Lenmark, University of Washington, Seattle, USA, 2000• Total rat serum lgG <strong>anti</strong>-<strong>GAD</strong> radioimmunoassaychoosing <strong>the</strong> “wrong” portion - and our vaccine would contain <strong>the</strong>full-length <strong>GAD</strong> protein (consisting of 585 amino acids). So, despite<strong>the</strong> obvious difficulties imposed in manufacturing a recombinant proteinof this size (which would be avoided by syn<strong>the</strong>sising a peptideinstead) we decided that our vaccine would contain full-length, natural<strong>GAD</strong> protein - leaving <strong>anti</strong>gen processing and presentation of <strong>the</strong>appropriate region (“determinant”) to that individual patients immunesystem. This decision not to pursue <strong>GAD</strong> peptide <strong>the</strong>rapy seems tohave paid off in view of recent scientific reports that repeat injectionsof protein fragments (“peptides”) can confuse <strong>the</strong> immune system andresult in immune over-reaction (anaphylaxis) – that can be life threateningin experimental animals. This response is not shown by <strong>the</strong>respective (intact) proteins. Our use of <strong>the</strong> full-length <strong>GAD</strong>65 proteinin our vaccine avoids this.Our initial manufacturing was conducted to <strong>the</strong> principles of GLP(“Good Laboratory Practice”) and was used successfully for all preclinicalsafety studies and clinical Phase I (in healthy volunteers).These ca. 25 different pre-clinical safety studies used various qu<strong>anti</strong>tiesof ei<strong>the</strong>r <strong>GAD</strong> alone, or <strong>the</strong> <strong>GAD</strong> vaccine (alum-<strong>GAD</strong>), in severaldifferent species, and administered by different routes. All <strong>the</strong>sestudies followed international regulatory requirements to establish <strong>the</strong>pre-clinical safety after <strong>GAD</strong> administration, and <strong>the</strong>reby provided<strong>the</strong> basis for Phase II development. In contrast to <strong>the</strong> pre-clinical andPhase I stages, however, <strong>the</strong> <strong>GAD</strong> vaccineused for Phase II is manufactured to <strong>the</strong> highestquality standard available for manufactureof clinical <strong>the</strong>rapeutics. This is “cGMP”(or “current Good Manufacturing Practice”)– that will also be required for all future clinicaldevelopment.<strong>Diamyd</strong>’s development of <strong>the</strong> <strong>GAD</strong> vaccinehas now culminated <strong>with</strong> completion of ourPhase II clinical trial in 47 “Type 2” diabetespatients <strong>with</strong> <strong>GAD</strong>-<strong>anti</strong>bodies (LADA). Thisstudy is truly pioneering – in that this is <strong>the</strong>first time patients have received <strong>the</strong> <strong>GAD</strong>vaccine.Prior to un-blinding, I am highly optimisticregarding <strong>the</strong> outcome of this PhaseII. I think this study will be a resoundingsuccess if <strong>the</strong> study outcome includes <strong>the</strong>following:1. <strong>the</strong>re are no safety concerns of alum-<strong>GAD</strong> vaccination2. <strong>the</strong>re is evidence for responses that areconsistent <strong>with</strong> a positive <strong>the</strong>rapeutic effect.John Robertson, Ph.D., has beenDirector of Research Development in<strong>Diamyd</strong> Medical since 1994.Robertson has experience in biotechnologyand toxicology, both fromindustry (Inveresk ResearchInternational, U.K. and ScheringAgrochemicals, U.K.) and academicinstitutes (Karolinska Institute,Stockholm; The Pasteur Institute,Paris; and <strong>the</strong> NIH in Washington).dmccad june 2003page 43


T cell <strong>GAD</strong>65For use of <strong>GAD</strong> in immunological assaysbulk rh<strong>GAD</strong>65For use of <strong>GAD</strong> in enzymatic assays<strong>Diamyd</strong>, Inc.,Research Triangle Park6213-D Angus DriveRaleigh, NC. 27617USAwww.diamyd.comproducts@diamyd.com<strong>Diamyd</strong>, Medical AB (publ)Djurgårdsbrunnsvägen 54SE-115 25 StockholmSweden

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!