Abstract
The immunogenicity of protein therapeutics has so far proven to be difficult to predict in patients, with many biologics inducing undesirable immune responses directed towards the therapeutic resulting in reduced efficacy, anaphylaxis and occasionally life threatening autoimmunity. The most common effect of administrating an immunogenic protein therapeutic is the development of a high affinity anti-therapeutic antibody response. Furthermore, it is clear from clinical studies that protein therapeutics derived from endogenous human proteins are capable of stimulating undesirable immune responses in patients, and as a consequence, the prediction and reduction of immunogenicity has been the focus of intense research. This review will outline the principle causes of the immunogenicity in protein therapeutics, and describe the development of pre-clinical models that can be used to aid in the prediction of the immunogenic potential of novel protein therapeutics prior to administration in man.
Key words: immunogenicity, deimmunization, protein therapeutic, T cells, neutralizing antibody
Introduction
The expectation that ‘self’ derived protein therapeutics (comprised of human germline sequence), such as recombinant human cytokines and ‘human’ antibodies, will avoid immunogenicity due to central tolerance is clearly flawed. There are now many examples of recombinant proteins (e.g., IFNβ1–3 IFNα4,5 GM-CSF6 and human anti-TNFα7,8 antibodies) which stimulate host immune responses that are directed against the therapeutic. Table 1 summarizes the frequency of anti-therapeutic antibodies (obtained from package inserts detailing clinical trial and post-approval data) observed against a number of FDA-approved biologics. Generation of anti-therapeutic antibodies involves stimulation of multiple components of the immune system, and therefore the immunogenicity of protein therapeutics cannot necessarily be attributed to a single factor. Indeed, stimulation of both adaptive (exemplified by the development of high affinity, highly specific antibodies and long lasting lymphocyte ‘memory’) and non-adaptive (often mediated by innate receptors which does not confer long-lasting protective immunity to the host) immune responses are normally involved in the development of a highly specific humoral response such as those directed against protein therapeutics. Such responses are normally polyclonal, and can have both a neutralizing and non-neutralizing effect on protein therapeutics. Anti-therapeutic antibodies that are detected in the serum of patients can comprise multiple isotypes (IgM, IgG and IgE) and sub-classes (IgG1-4) of heavy chain constant regions. In many instances such antibodies possess variable regions that bind with high affinity to the protein therapeutic, and will therefore have undergone somatic hypermutation of variable region genes. The ability to neutralize the protein therapeutic is a product of the B cell epitope(s) against which the humoral response is directed. For example, in the case of antibody therapeutics, human anti-mouse (HAMA) or human anti-human (HAHA) responses directed against the idiotype are typically neutralizing, and such responses have been observed for both humanized and fully-human antibodies.9,10 For protein therapeutics that are derived from endogenous proteins that serve a non-redundant function (e.g., recombinant human erythropoietin), a neutralizing antibody response can cross-react with the endogenous protein resulting in morbidity and mortality.11
Table 1.
Antibody name | Company | Type | Target | Indication(s) | Reported immunogenicity |
Muromanab (OKT3) | Ortho Biotech | Murine | CD3 | Allograft rejection | 25% (24) |
Abciximab (Reopro) | Centocor (Johnson & Johnson) | Chimeric Fab | GPIIb/IIIa | PTCA adjunct | 6%–44% (36) |
Rituximab (Rituxan) | Genentech (Roche)/Biogen Idec | Chimeric | CD20 | Non-Hodgkin lymphoma | 11% (2578) |
Daclizumab (Zenapax) | Hoffman LaRoche | Humanized | IL2R | Transplant rejection | 14–34% |
Trastuzumab (Herceptin) | Genentech (Roche) | Humanized | Her2/neu | Breast cancer | <1% |
Palivizumab (Synagis) | MedImmune (Astra Zeneca) | Humanized | RSVF | RSV prophylaxis | 0.7%–2% (1002–639) |
Basiliximab (Simulect) | Novartis | Chimeric | IL2R | Transplant rejection | 1–2% (138–339) |
Infliximab (Remicade) | Centocor (Johnson & Johnson) | Chimeric | TNFα | RA/Crohn | 10–15% |
Arcitumomab (CEA-scan) | Immunomedics | Murine | CEA | Colorectal cancer | <1% (3/400) |
Canakinumab (Ilaris) | Novartis | Human | IL-1β | Cryopirin-associated periodic syndrome | 0% (64) |
Fanolesomab (Neutrospec) | Palatin Tech. | Murine | CD15 | Imaging for appendicitis | 0–16.6% (30–54) |
Imciromab (Myoscint) | Centocor (Johnson & Johnson) | Murine | Myosin | Cardiac imaging for MI | <1% (914) |
Capromab (Prostascint) | Cytogen | Murine | PSMA | Prostate cancer diagnostic | 8%–19% (27–239) |
Nofetumomab (Verluma) | Boehringer Ingelheim | Murine | 40 KDa glycoprotein | Detection of SCLC | 6% (53) |
Gemtuzumab (Mylotarg) | Wyeth Pharma (Pfizer) | Humanized | CD33 | Acute myeloid leukemia | 0% (277) |
Alemtuzumab (Campath) | Ilex Pharma (Genzyme) | Humanized | CD52 | B cell chronic lymphocytic leukemia | 1.9–8.3% (133–211) |
Ibritumomab (Zevalin) | Idec Pharma (Biogen Idec) | Murine | CD20 | Non-Hodgkin lymphoma | 1.3% (446) |
Adalimumab (Humira) | Abbott | Human | TNFα | RA/Crohn/PsA/JIA/Ankylosing spondylitis/plaque psoriasis | 2.6%–26% |
Omalizumab (Xolair) | Genentech (Roche) | Humanized | IgE | Asthma | <0.1% (1723) |
Efalizumab (Raptiva) | Genentech (Roche) | Humanized | CD11a | Psoriasis | 6.3% (1063) |
Tositumomab (Bexxar) | GSK | Murine | CD20 | Non-Hodgkins lymphoma | 11% (230) |
Cetuximab (Erbitux) | Imclone (Eli Lilly) | Chimeric | EGFR | Colorectal cancer | 5% (1001) |
Bevacizumab (Avastin) | Genentech (Roche) | Humanized | VEGF | Colorectal, breast, renal and NSCL cancer | 0% (∼500) |
Panitumumab (Vectibix) | Amgen | Human | EGFR | Colorectal cancer | 4.6% (613) |
Ranibizumab (Lucentis) | Genentech (Roche) | Humanized | VEGF | Macular degeneration | 1–6% |
Eculizumab (Soliris) | Alexion Pharma | Humanized | C5 | Paroxysmal nocturnal hemoglobinuria | 2% (196) |
Natalizumab (Tysabri) | Biogen Idec | Humanized | α-4 integrin | MS & Crohn | 9% (627) |
Golimumab (Simponi) | Centocor (Johnson & Johnson) | Human | TNFα | RA/PsA/Ankylosing spondylitis | 4% (1425) |
Cetolizumab pegol (Cimzia) | UCB | Humanized | TNFα | RA/Crohn | 8% (1509) |
Ofatumumab (Arzerra) | GSK | Human | CD20 | CLL | 0% (79) |
Ustekinumab (Stelara) | Centocor (Johnson & Johnson) | Human | IL-12/IL-23 | Plaque psoriasis | 3–5% (743–1198) |
Tocilizumab (Actemra) | Genentech (Roche) | Humanized | IL-6R | Rheumatoid arthritis | 2% (2876) |
Denosumab (Prolia) | Amgen | Human | RANKL | Osteoporosis | <1% (8113) |
The frequency of anti-therapeutic antibody responses (both neutralizing and non-neutralizing antibodies) observed in patients is shown as % and the size of the patient group evaluated in immunogenicity studies given in brackets.
Development of Immune Responses Directed Against Protein Therapeutics
Initial events that trigger the development of immune responses against protein therapeutics may occur independently of CD4+ T-cell help. Such events can involve innate receptor activation (e.g., pattern recognition receptors, PRR) resulting in the stimulation of antigen presenting cells (APC), such as dendritic cells (DC) as well as B-cell subsets (reviewed in ref. 12–14). The involvement of innate receptors expressed on APC will greatly facilitate the development of a potent adaptive immune response.15–19 It is possible that the biophysical properties of the protein therapeutic, such as glycosylation, as well as excipients that may be present in the drug and/or formulation could provide the initial stimulation via PRR on DC, resulting in effective maturation and expression of lymphocyte co-stimulatory receptors.20,21 DC stimulated via PRR have an increased capacity to stimulate T cells and consequently support the generation of T-dependent (antigens that stimulate these responses are known as T-cell or thymus-dependent antigens) high affinity anti-therapeutic antibody response. Indeed, the biological activity of the protein therapeutic can itself have an adjuvant effect resulting in high levels of immunogenicity in patients.6 One possible explanation for immunogenicity associated with GM-CSF treatment (to help prevent infection during cancer therapy) is the fact that GM-CSF has an ‘adjuvant effect’ and efficiently activates DC, monocytes and lymphocytes. In contrast a G-CSF a similar size molecule also used in the treatment of cancer but with no immunomodulatory activity is non-immunogenic.
T-independent stimulation of B cells [antigens that stimulate these responses are known as T-cell or thymus-independent (T-independent) antigens] in which the protein therapeutic bypasses the need for T-cell help may occur if the protein forms a multimeric structure that can effectively cross-link the B-cell receptor (BCR) to an extent where co-stimulation from T cells is not required for an anti-therapeutic antibody response.22 It is envisaged that aggregated proteins could act in a manner similar to that described for T-independent type 2 antigens,23 via the polyclonal activation of splenic marginal zone B cells (MZ B cells). However, a number of factors regarding humoral responses against T-independent type 2 antigens do not support this hypothesis, including the fact that T-independent type 2 antigens are highly ordered protein structures (e.g., viral coat proteins) which enables the presentation of B-cell epitopes in a regular (5–10 nm) and repeating manner. Such regular repeating epitopes are critical for the activation of MZ B cells.24,25 Whether such organized protein structures are formed in aggregates that may be in formulations of protein therapeutics is at present unclear. The involvement of T cells also cannot be ruled out for T-independent type 2 antigens, since MZ B cells can efficiently present antigen to T cells.25 Stimulation of splenic MZ B cells in humans is characterized by production of IgM, and is not sufficient alone to induce class switch recombination to multiple IgG subclasses which are commonly observed in human immune responses against protein therapeutics.25
There is, however, a link between aggregated or multimeric proteins and enhanced immunogenicity. Studies have shown that aggregated protein therapeutics can stimulate enhanced humoral immune responses in a variety of models. Braun et al.26 showed that aggregated, but not monomeric, recombinant human IFNα was able to break tolerance and result in the generation of IFNα specific antibodies in human IFNα transgenic mice. A more detailed study of the immunogenicity of recombinant human IFNα aggregates with different physiochemical properties in human IFNα transgenic mice showed that maintenance of a ‘native’-like confirmation was associated with more immunogenic aggregates.27 The precise mechanism behind the influence of aggregates in the immunogenicity of protein therapeutics has not been defined, but earlier studies on antibody feedback regulation in the immune system provide an interesting view of how antibodies that form immune complexes with soluble or particulate antigen are able to influence the progression of an immune response. Administration of IgG to mice in combination with particulate antigen suppressed the humoral response,28 whereas combined administration of IgM and IgG3 enhanced antibody responses.29 Suppression by IgG is likely to be dependent on masking B-cell epitopes and suppression is dependent on antibody affinity,30,31 engagement of FcγRIIb and/or enhanced phagocytic clearance of immune complexes.32–35 By contrast enhancement of responses by IgM and IgG3 to antigens administered at sub-optimal doses in mice is dependent on fixing C1q of the classical complement cascade, since mice treated with cobra venom factor or lacking CR1/CR2 are impaired in their ability to mount an enhanced antibody response.36–38 A single IgM molecule can activate complement, and mouse IgG3 has the capacity to spontaneously multimerize to allow binding of C1q resulting in the subsequent activation of C3.39 Since it is possible to produce both IgM and IgG3 (in mice only) independently of T cells it is feasible that these isotypes are an important early trigger in initiating the first stages in the immune response cascade against preparations of protein therapeutics that may contain very low levels of aggregates. It should also be noted that certain antigens (e.g., derived from bacterial membranes) are effective in activating the alternative complement cascade in the absence of immune complexes and may also ultimately result in fixing C3d and effectively opsonising the protein for uptake by macrophages via C3d receptors (CR2).40
The generation of immune complexes against both particulate and soluble antigens as well as immune complex independent activation of the alternative complement cascade will lead to enhanced uptake by APC via FcR and complement receptors (CR1 and CR2). Getahun et al. have shown41 that transfer of D011.10 transgenic T cells into syngenic Balb/C mice that were immunized with IgG2a anti-TNP/TNP-Ova (without adjuvant) resulted in an enhanced T cell and Ova specific antibody response. These observations are consistent with an increase in presentation of T-cell epitopes to T-cells via enhanced uptake and processing through the APC via FcR-mediated uptake of immune complexes. Drug formulations are known to influence the conformation of protein therapeutics, such as multimerization and aggregation (reviewed in ref. 42). It is therefore possible that initial activation events involve the interaction of antibodies with the protein therapeutic to form immune complexes, and subsequent triggering of immune response suppression or enhancement is dependent on the conformation of the protein therapeutic and the binding of antibodies with specific isotypes. It should be noted that in naive patients, the amount of protein specific antibody is likely to be very low, although even small amounts of multimeric IgM binding to aggregated protein will be sufficient to activate complement. Pre-existing IgG antibodies in patients might be specific for a protein therapeutic that is derived from an endogenous counterpart, for example a recombinant human cytokine. Such antibodies may be produced as a result of incomplete central tolerance whereby low endogenous levels of protein expressed in specialized tissues will not facilitate deletion of auto-reactive B- and T-cell clones during lymphocyte development. Indeed transient and non-pathological autoimmune responses have been detected in healthy individuals and are probably suppressed by mechanisms involving peripheral tolerance such as regulatory T cells (Treg cells). For example, the development of anti-FVIII specific antibodies in healthy individuals has been frequently observed, and is thought to be produced as a result of tissue damage and activation of the clotting cascade.43,44 These responses have no obvious detrimental effect on healthy individuals presumably due to their transient appearance. Peripheral tolerance may be broken by treating patients with high doses of protein, particularly for chronic diseases, which will enable the production of antibodies specific for the protein therapeutic. Indeed the specificity of the humoral response may change during treatment as a result of B-cell epitope spreading, and this will be dependent on T-cell help which can be enhanced by the initial non-neutralizing antibody response allowing more efficient uptake of the protein by APC, and consequently the presentation of T-cell eptiopes.
Antigen Presentation and T-Dependent Responses
The sequence of events leading to the generation of a T-dependent humoral immune response against a protein therapeutic probably involves rapid induction of a T-independent response leading to expansion (1,000-fold) of epitope specific B cells that can increase the probability of a cognate interaction with CD4+ T cells.45 In parallel to this process, antigen will be phagocytosed by professional APC such as DC. If the therapeutic is injected sub-cutaneously, immature ‘steady state’ DC in the epidermis (e.g., Langerhans cells) will phagocytose and process the protein via the MHC class II processing pathway. For immature DC, activators or ‘danger signals’ that include proinflammatory factors, such as complement and chemokines as well as bacterial or viral products (LPS, CPG motifs and dsRNA), can enhance the activation of DC.46–48
One key function of immature DC is antigen capture and this can occur via a number of different receptors including members of the C-type lectin family (DEC-205, DCIR, Dectin-2 and CLEC-1), the immunoglobulin superfamily (DORA and ILT3), heat shock protein receptors, toll-like receptors (e.g., TLR-3 expressed on immature DC), and Fc receptors (FcγR and FcεR). As discussed above, one important factor in enhancing DC capture of therapeutic proteins may be the early formation of immune complexes with circulating IgM or IgG, which has been established to result in potent antibody responses in mice, and involves activation of complement and FcR binding. For example, in an experimental model the co-administration of IgG/KLH complexes results in a 1,000-fold enhancement (compared to KLH alone) in the IgG anti-KLH response.49 There is at least some circumstantial evidence for this in humans whereby small circulating immune complexes have been detected in patients receiving technetium-labelled infliximab.50 The presence of these immune complexes correlated with rapid uptake of infliximab in the liver and spleen, which is facilitated through interactions between antibody, FcR and complement-complement receptors.51 Thus, for protein therapeutics, initiation of anti-therapeutic antibody response may be dependent on the formation of immune complexes that can bind to APC through complement receptors or via the FcR.
Evidence that T-cell help is a central component of the pathway that results in antibody responses against protein therapeutics comes from a number of observations including the prominence of high affinity responses comprising multiple IgG isotypes, such as those observed against infliximab, adalimumab and FVIII.52,53 Indeed, T-cell subset polarization has been linked to the success of FVIII therapy in severe hemophiliacs where the predominance of Th2-driven IgG4 FVIII-specific antibodies correlates with the production of high inhibitor (neutralizing anti-FVIII antibodies) titres.54 By contrast, patients with FVIII inhibitors receiving immunosuppressive therapy have a predominance of Th1 driven IgG1 and IgG2 FVIII antibodies which are associated with a more successful therapeutic outcome.55 In some instances, T cells or T-cell help is targeted pharmacologically during treatment with protein therapeutics in order to treat the disease and/or reduce the immunogenicity of the protein therapeutic. The most frequent methods include the use of immunosuppressive therapies (such as methotrexate when administering infliximab for treating rheumatoid arthritis) or induction of neonatal tolerance, which is a primary strategy in reducing the incidence of inhibitors against FVIII (reviewed in ref. 56). It should be noted that other methods to reduce immunogenicity have also been adopted for therapeutic antibodies, such as infliximab, that induce peripheral tolerance where an inverse dose-immunogenicity relationship has been observed with high concentrations of antibody resulting in a reduction in the incidence of immunogenicity.57 One interesting method for inducing peripheral tolerance that may eventually be applied in a therapeutic context is the administration of proteins via a mucosal surface. Preliminary studies in mice with human IFNα and IFNβ have shown that anti-IFNα and IFNβ IgG responses are inhibited by oromucosal administration of the proteins.58 Induction of tolerance via this route is dependent on a unique class of ‘tolerizing’ DC and regulatory T cells (which suppress immune responses), which further highlights the importance of T cells in the development of anti-therapeutic antibodies.59
There has been considerable effort to establishing the role of MHC class II-restricted T-cell epitopes present in protein sequences and the development of humoral immune responses against protein therapeutics. Several studies have directly measured in vitro CD4+ T-cell responses against protein therapeutics (or peptide derivatives). Such studies have shown that many therapeutic proteins (including recombinant human proteins) contain potent T-cell epitopes.60–64 Stickler et al.65 used human in vitro T-cell assays to show the importance of T-cell epitopes in the immunogenicity of human IFNβ1b where the antibody response to deimmunized IFNβ1b (in which T-cell epitopes were mapped and removed by amino acid mutation) was compared in Balb/C mice. The immunogenicity of IFNβ1b was ameliorated by removing T-cell epitopes from the protein sequence. Moreover, removal of a single immunodominant but not a sub-dominant T-cell epitope, was sufficient to avoid an immune response.60 In a similar in vitro study the immunogenicity of human IFNβ (Rebif®) was attributed to the efficient uptake and processing of IFNβ1a by monocyte-derived DC in different clinical buffer formulations.66 Thus for recombinant human IFNβ1a, the formulation may influence protein conformation, leading to the possible development of protein multimers that can be efficiently engulfed by DC which may result in enhanced presentation of T-cell epitopes to CD4+ T cells. Analysis by in vitro T-cell assays revealed that the new Rebif® formulations were significantly less immunogenic than the existing formulation. These results were confirmed after a phase IIIb clinical trial with one new formulation that induced significantly fewer neutralizing anti-IFNβ antibodies in patients treated with the new formulation compared to standard Rebif®.67,68
Models for Predicting Immunogenicity
Traditional animal models used in safety and toxicological studies for small molecule drugs are of limited use in the assessment of the potential for immunogenicity of protein therapeutics in humans, especially for human proteins where the animal will not be tolerant to these proteins. Differences in antigen processing and MHC class II binding between humans and in-bred strains of rodents will also influence the repertoire of T-cell epitopes presented to T cells and consequently affect the immunogenicity of the protein therapeutic. In order to address some of these limitations, novel transgenic mice and xenograft transplantation models have been generated.
One of the more difficult issues with regard to studying the mechanisms behind immunogenicity using animal models is the fact that proteins administered in the absence of adjuvant are inherently non-immunogenic, indeed this observation was termed ‘immunologists’ dirty little secret' by Charles Janeway.69 Nevertheless, this has not hampered attempts to generate in vivo models that can be used to predict the immunogenicity of protein therapeutics. Mice expressing specific human HLA allotypes (and lacking endogenous mouse MHC class II) are frequently used for research to evaluate the involvement of human HLA alleles in diseases such as allergy and autoimmunity.70,71 Such models may be particularly useful where immune tolerance against a specific recombinant therapeutic or class of therapeutics is induced by either transgenic expression of the protein of interest or induction of tolerance during neonatal development.72,73 There are significant technical challenges associated with developing such multi-feature transgenic mice including: restricted diversity in HLA alleles (typically alleles would be selected to cover the main MHC class II supergroups),74 inbred stains of mice express restricted TCR repertoire, knocking out the murine homolog of the protein therapeutic whereby the endogenous murine gene and the human transgene may not always serve the same function in mice.
Human immune system xenografts offer a promising alternative to transgenic mouse models. Such models are based on immunodeficient (including lack of endogenous NK cells) NOD scid IL2Rγ-/- or Rag2-/-γc-/- mice in which human immune systems are transplanted by engraftment of human CD34+ stem cells.75,76 CD34+ stem cells typically derived from human peripheral blood mononuclear cells (PBMC) or from human umbilical vein are engrafted into neonatal mice. Differentiation of the human CD34+ stem cells results in the population of various lineages in different immune compartments including lymphocyte, monocyte/myeloid, granulocyte and dendritic cell. The fact that these mice express human MHC class II and are tolerant to human immunoglobulins (or do not recognize human immunoglobulins as “foreign”) may enable their use as a valuable in vivo model for the prediction of the immunogenicity of antibody therapeutics (e.g., humanized and/or fully human antibodies). There are however limitations in using these engraftment models including the fact that they are not tolerant against all human proteins, that there is no germline transfer of genes encoding human immune cells so that each mouse has to be engrafted on an individual basis, and that some studies have shown considerable variability in immune responses to antigens/factors that stimulate potent responses in humans.77
In addition to mouse models, non-human primates have commonly been used as a test system to evaluate the potential for immunogenicity of protein therapeutics. However, non-human primate MHC molecules are significantly different from the human equivalent making them less suited for use as models for immunogenicity. Indeed, studies using a marmoset model of experimental autoimmune encephalitis showed that, although the disease itself closely mimicked that found in humans, the peptide T-cell epitope driving the autoimmune response was different from that identified in humans.78 Moreover, the marmoset MHC class II molecule involved was of a lineage not found in humans. The fact that human protein therapeutics may still be significantly different from the endogenous primate homolog, coupled with the ethical issues involved with using non-human primates also significantly limit the utility of non-human primates as a model for immunogenicity in the clinic.
The generation of an anti-therapeutic antibody response in patients appears to correlate with the ability to stimulate epitopespecific human T-cell responses.79 Consequently tools to aide researchers in measuring CD4+ T-cell epitopes in both a quantitative and qualitative manner have been developed.
For high throughput screening of sequences in silico peptide-MHC class II algorithms have been developed that predict the ability of a peptide to bind to MHC class II. In addition to the advantage of rapidly screening large numbers of sequences these methods are considerably less expensive than traditional in vitro MHC class II binding assays which are time consuming and expensive. Current in silico tools are also reasonably accurate in terms of predicting MHC class II binding peptides against a large number of HLA alleles. The major limitation common to all methods of MHC class II binding analysis (in silico and in vitro) is the high level of false positive peptides that are identified as binding MHC class II but which fail to stimulate T-cell responses both in vitro and in vivo. Over-prediction is largely due to the inability of in silico tools to take into account other factors that influence the formation of epitopes, such as protein/peptide processing, recognition by the T-cell receptor (TCR), and T-cell tolerance to peptides.80
In vitro T cell culture systems offer the advantage of a direct measurement of antigen specific activation of T cells and are able to therefore overcome some of the limitations, in particular over-prediction, observed with in silico methods. PBMC are typically used as a source of CD4+ T cells for in vitro T-cell assays and these are stimulated with whole proteins or peptides derived from therapeutic proteins using a variety of in vitro culture formats. In vitro T-cell priming responses can be detected in healthy donors; furthermore there is evidence showing that the repertoire of T-cell epitopes recognized in non-primed healthy donors is represented in the primed patient population PBMC.61,62,81,83–87 This has enabled T-cell epitope maps to be generated using healthy donors that are relevant to the patient group.43,61–63 Data from in vitro T-cell assays, such as the number and potency (immunodominance) of T-cell epitopes, can be used to determine the relative risk of immunogenicity between multiple variant therapeutic proteins during pre-clinical development. In addition, strategies can be employed to reduce immunogenicity by removing T-cell epitopes through targeted amino acid substitutions (reviewed in ref. 88). Accurate detection of T-cell epitopes using in vitro T-cell assays requires selection and testing of peptides against large cohorts of individuals to ensure that a broad spectrum of MHC class II allotypes are present. The recent development of methods to store large well characterized banks of HLA-typed individuals has facilitated the judicious selection of cohorts of donors for studies which closely represent the frequency of the major MHC class II allotypes expressed in the world population. Qualitative and quantitative measurement of T-cell epitopes by in vitro T-cell assays has enabled strategies to remove T-cell epitopes (such as deimmunization) to be developed. Such strategies optimally rely on the use of in vitro methods for epitope identification combined with in silico tools for the design and selection of mutant peptides that can be incorporated into optimized T-cell epitope-depleted protein sequences which have reduced capacity for MHC binding.
Reducing Immunogenicity by Design
Depleting T-cell epitopes from protein therapeutics (by deimmunization) has proven to be successful and has lead to a number of deimmunized proteins progressing into clinical trials. The majority of these deimmunized protein therapeutics are antibodies, and none of these proteins has elicited any significant level of undesirable immunogenicity in patients to date. One of the more clinically advanced deimmunized antibodies targets prostate specific membrane antigen, and several clinical trials have been performed using this antibody (conjugated to radiolabels or the toxin DM1) with no anti-therapeutic antibody responses observed in any patient.89–92 Overall, this clinical data provides support for the importance of T-cell epitopes in the generation of anti-therapeutic antibody responses and indicate that deimmunized protein therapeutics may provide a safer less immunogenic class of new biologic.
The way forward for dealing with issues associated with immunogenicity in protein therapeutics is likely to involve a combined approach whereby, new molecules can be generated through rational sequence design (via methods such as deimmunization) and where the lead proteins are also tested in the appropriate animal model and/or in vitro assay during pre-clinical development from which the least immunogenic leads can be selected. Testing of other product-specific triggers of immune responses should be considered including: appropriate physicochemical analysis of the therapeutic; aggregation, post-translational modifications, and effects of excipients that may be present in the final product formulation. Such a combined approach will undoubtedly facilitate the generation of protein therapeutics with reduced potential for immunogenicity.
Table 1.
Biologic name | Company | Type | Target | Indication(s) | Reported immunogenicity |
Prolastin | Talecris biotherapeutics | PD | α1-proteinase inhibitor | α1-antitrypsin deficiency | None reported |
Aralast | Baxter Healthcare | PD | α1-proteinase inhibitor | α1-antitrypsin deficiency | None reported |
Zemaira | Aventis Behring (CSL Behring) | PD | α1-proteinase inhibitor | α1-antitrypsin deficiency | None reported |
Kogenate FS | Bayer (Bayer Schering Pharma) | rHu | Factor VIII | Hemophilia A | 15% |
ReFacto | Genetics Institute (Wyeth) | rHu | Factor VIII | Hemophilia A | 30% |
Zyntha | Wyeth (Pfizer) | rHu | Factor VIII | Hemophilia A | 2.2% (89) |
NovoSeven | NovoNordisk | rHu | Factor FVII | Hemophilia | <1% |
Benefix | Wyeth (Pfizer) | rHu | Factor IX | Hemophilia B | 3% |
ATryn | GTC Biotherapeutics | rHu | Anti-thrombin | Thromboembolism | None reported |
BabyBIG | California Department of Health Services | PD | Botulism Immune Globulin Intravenous Human | Infant botulism | |
Berinert | CSL Behring | rHu | C1 Esterase Inhibitor | Angioedema | |
Cinryze | Lev Pharmaceuticals | rHu | C1 Esterase Inhibitor | Angioedema | |
Rhophylac | CSL Behring | PD | Rho(D) Immune Globulin | ITP | 0% (447) |
Evithrom | OMRI X Biopharmaceuticals | rHu | Thrombin, Topical | Coagulation | 3.3% |
Recothrom | ZymoGenetics | rHu | Thrombin, Topical | Coagulation | 1.2–1.5% |
Wilate | Octapharma | PD | von Willebrand Factor | Coagulation | 1.5–3% |
Cerezyme | Genzyme | rHu | ß-glucocerebrosidase; | Gacher Disease | 15% |
Exenatide or Byetta | Amylin Pharmaceuticals/Eli Lilly | R | Glucagon Like Peptide-1 | Type II diabetes | 6% |
IntronA | Schering Corp (Bayer Schering Pharma) | rHu | IFNα | Leukemia, Kaposi sarcoma, hepatitis B/C | <3–13% |
Betaseron | Bayer Schering Pharma | rHu | IFNβ | Multiple sclerosis | 16.5–25.2% |
NovoLog | NovoNordisk | rHu | Insulin analog | Type II diabetes | Transient antibodies |
Leukine | Genzyme | rHu | GM-CSF | Preventing infection in cancer | 2.3% |
NEUPOGEN (Filgrastim) | Amgen | rHu | G-CSF. | Preventing infection in cancer | 3% |
Retavase | PDL Biopharma | rHu | TPA | Myocardial infarction, pulmonary embolism | 0% (2400) |
Humatrope | Eli Lilly | rHu | Growth hormone | Dwarfism | 1.6% |
Adagen | Enzon Pharmaceuticals | Bovine | ADA Adenosine deaminase | Inherited immunodeficiency | Not reported (SCID) |
Pulmozyme | Genentech (Roche) | rHu | DNase I | Cystic fibrosis | 2–4% |
Procrit | Amgen | rHu | EPO | Anemia in chronic renal disease | |
Proleukin | Novartis | rHu | IL-2 | Oncology | <1% |
Recombinant human (rHu), plasma derived (PD) and recombinant (r) products are indicated. Companies that acquired a majority stake holding in any of the companies that performed the initial drug development of the biologics listed are shown in brackets
Acknowledgements
The authors would like to acknowledge the kind assistance of Drs. Robin Thorpe, Laura Perry, Frank Carr and Tim Jones in proof reading this manuscript.
Footnotes
Previously published online: www.landesbioscience.com/journals/selfnonself/article/13904
References
- 1.Bertolotto A, Malucchi S, Sala A, Orefice G, Carrieri PB, Capobianco M, et al. Differential effects of three interfeorn betas on neutralising antibodies in patients with multiple sclerosis: a follow up study in an independent laboratory. J Neurol Neurosurg Psychiatry. 2002;73:148–153. doi: 10.1136/jnnp.73.2.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bertolotto A, Sala A, Malucchi S, Marnetto F, Caldano M, Di Sapio A, et al. Biological activity of interferon betas in patients with multiple sclerosis is affected by treatment regimen and neutralising antibodies. J Neurol Neurosurg Psychiatry. 2004;75:1294–1299. doi: 10.1136/jnnp.2004.037259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Cook SD, Quinless JR, Jotkowitz A, Beaton P. Serum IFN neutralizing antibodies and neopterin levels in a cross-section of MS patients. Neurology. 2001;57:1080–1084. doi: 10.1212/wnl.57.6.1080. [DOI] [PubMed] [Google Scholar]
- 4.Ryff JC. Clinical investigation of the immunogenicity of interferon-alpha2a. J Interferon Cytokine Res. 1997;17:29–33. [PubMed] [Google Scholar]
- 5.Von Wussow P, Hehlmann R, Hochhaus T, Jakschies D, Nolte KU, Prümmer O, et al. Roferon (rIFNalpha2a) is more immunogenic than intron A (rIFNalpha2b) in patients with chronic myelogenous leukemia. J Interferon Res. 1994;14:217–219. doi: 10.1089/jir.1994.14.217. [DOI] [PubMed] [Google Scholar]
- 6.Wadhwa M, Meager A, Dilger P, Bird C, Dolman C, Das RG, et al. Neutralizing antibodies to granulocyte-macrophage colony-stimulating factor, interleukin-1alpha and interferon-alpha but not other cytokines in human immunoglobulin preparations. Immunology. 2000;99:113–123. doi: 10.1046/j.1365-2567.2000.00949.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Baert F, Noman M, Vermeire S, Van Assche G, D' Haens G, Carbonez A, et al. Influence of immunogenicity on the long-term efficacy of infliximab in Crohn's disease. N Engl J Med. 2003;348:601–608. doi: 10.1056/NEJMoa020888. [DOI] [PubMed] [Google Scholar]
- 8.Radstake TR, Svenson M, Eijsbouts AM, van den Hoogen FH, Enevold C, van Riel PL, et al. Formation of antibodies against infliximab and adalimumab strongly correlates with functional drug levels and clinical responses in rheumatoid arthritis. Ann Rheum Dis. 2009;68:1739–1745. doi: 10.1136/ard.2008.092833. [DOI] [PubMed] [Google Scholar]
- 9.Ritter G, Cohen LS, Williams C, Jr, Richards EC, Old LJ, Welt S. Serological analysis of human anti-human antibody responses in colon cancer patients treated with repeated doses of humanized monoclonal antibody A33. Cancer Res. 2001;61:6851–6859. [PubMed] [Google Scholar]
- 10.Cassinotti A, Ardizzone S, Porro GB. Adalimumab for the treatment of Crohn's disease. Biologics. 2008;2:763–777. doi: 10.2147/btt.s3292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Casadevall N, Nataf J, Viron B, Kolta A, Kiladjian JJ, Martin-Dupont P, et al. Pure red-cell aplasia and anti-erythropoietin antibodies in patients treated with recombinant erythropoietin. N Engl J Med. 2002;346:469–475. doi: 10.1056/NEJMoa011931. [DOI] [PubMed] [Google Scholar]
- 12.Vos Q, Lees A, Wu ZQ, Snapper CM, Mond JJ. B-cell activation by T-cell-independent type 2 antigens as an integral part of the humoral immune response to pathogenic microorganisms. Immunol Rev. 2000;176:154–170. doi: 10.1034/j.1600-065x.2000.00607.x. [DOI] [PubMed] [Google Scholar]
- 13.Geijtenbeek TB, van Vliet SJ, Engering A, 't Hart BA, van Kooyk Y. Self- and nonself-recognition by C-type lectins on dendritic cells. Annu Rev Immunol. 2004;22:33–54. doi: 10.1146/annurev.immunol.22.012703.104558. [DOI] [PubMed] [Google Scholar]
- 14.Diebold SS. Activation of dendritic cells by toll-like receptors and C-type lectins. Handb Exp Pharmacol. 2009;188:3–30. doi: 10.1007/978-3-540-71029-5_1. [DOI] [PubMed] [Google Scholar]
- 15.Bonifaz LC, Bonnyay DP, Charalambous A, Darguste DI, Fujii S, Soares H, et al. In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T cell vaccination. J Exp Med. 2004;199:815–824. doi: 10.1084/jem.20032220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Tacken PJ, de Vries IJ, Gijzen K, Joosten B, Wu D, Rother RP, et al. Effective induction of naive and recall T-cell responses by targeting antigen to human dendritic cells via a humanized anti-DC-SIGN antibody. Blood. 2005;106:1278–1285. doi: 10.1182/blood-2005-01-0318. [DOI] [PubMed] [Google Scholar]
- 17.Merlo A, Calcaterra C, Mínard S, Balsari A. Cross-talk between toll-like receptors 5 and 9 on activation of human immune responses. J Leukoc Biol. 2007;82:509–518. doi: 10.1189/jlb.0207100. [DOI] [PubMed] [Google Scholar]
- 18.Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C, Yamazaki S, et al. Differential antigen processing by dendritic cell subsets in vivo. Science. 2007;315:107–111. doi: 10.1126/science.1136080. [DOI] [PubMed] [Google Scholar]
- 19.Adams EW, Ratner DM, Seeberger PH, Hacohen N. Carbohydratemediated targeting of antigen to dendritic cells leads to enhanced presentation of antigen to T cells. Chembiochem. 2008;9:294–303. doi: 10.1002/cbic.200700310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hou B, Reizis B, DeFranco AL. Toll-like receptors activate innate and adaptive immunity by using dendritic cell-intrinsic and -extrinsic mechanisms. Immunity. 2008;29:272–282. doi: 10.1016/j.immuni.2008.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Honda K, Ohba Y, Yanai H, Negishi H, Mizutani T, Takaoka A, et al. Spatiotemporal regulation of MyD88-IRF-7 signalling for robust type-I interferon induction. Nature. 2005;434:1035–1040. doi: 10.1038/nature03547. [DOI] [PubMed] [Google Scholar]
- 22.Martin F, Oliver A, Kearney J. Marginal zone and B1 B cells unite in the early response against T-independent blood-borne particulate antigens. Immunity. 2001;14:617–629. doi: 10.1016/s1074-7613(01)00129-7. [DOI] [PubMed] [Google Scholar]
- 23.Fehr T, Bachmann MF, Bucher E, Kalinke U, Di Padova FE, Lang AB, et al. Role of repetitive antigen patterns for induction of antibodies against antibodies. J Exp Med. 1997;185:1785–1792. doi: 10.1084/jem.185.10.1785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Fehr T, Naim HY, Bachmann MF, Ochsenbein AF, Spielhofer P, Bucher E, et al. T-cell independent IgM and enduring protective IgG antibodies induced by chimeric measles viruses. Nat Med. 1998;4:945–948. doi: 10.1038/nm0898-945. [DOI] [PubMed] [Google Scholar]
- 25.Bachmann MF, Zinkernagel RM. Neutralizing antiviral B cell responses. Annu Rev Immunol. 1997;15:235–270. doi: 10.1146/annurev.immunol.15.1.235. [DOI] [PubMed] [Google Scholar]
- 26.Braun A, Kwee L, Labow MA, Alsenz J. Protein aggregates seem to play a key role among the parameters influencing the antigenicity of interferon alpha (IFNalpha) in normal and transgenic mice. Pharm Res. 1997;14:1472–1478. doi: 10.1023/a:1012193326789. [DOI] [PubMed] [Google Scholar]
- 27.Hermeling S, Schellekens H, Maas C, Gebbink MF, Crommelin DJ, Jiskoot W. Antibody response to aggregated human interferon alpha2b in wild-type and transgenic immune tolerant mice depends on type and level of aggregation. J Pharm Sci. 2006;95:1084–1096. doi: 10.1002/jps.20599. [DOI] [PubMed] [Google Scholar]
- 28.Cerottini JC, McConahey PJ, Dixon FJ. The immunosuppressive effect of passively administered antibody IgG fragments. J Immunol. 1969;102:1008–1015. [PubMed] [Google Scholar]
- 29.Klaus GGB. Generation of memory cells. III. Antibody class requirements for the generation of B-memory cells by antigen-antibody complexes. Immunology. 1979;37:345–351. [PMC free article] [PubMed] [Google Scholar]
- 30.Karlsson MCI, Getahun A, Heyman B. FccRIIB in IgG-mediated suppression of antibody responses: different impact in vivo and in vitro. J Immunol. 2001;67:5558–5564. doi: 10.4049/jimmunol.167.10.5558. [DOI] [PubMed] [Google Scholar]
- 31.Heyman B, Wiersma E, Nose M. Complement activation is not required for IgG-mediated suppression of the antibody response. Eur J Immunol. 1988;18:1739–1743. doi: 10.1002/eji.1830181113. [DOI] [PubMed] [Google Scholar]
- 32.Daëron M. Fc receptor biology. Annu Rev Immunol. 1997;15:203–234. doi: 10.1146/annurev.immunol.15.1.203. [DOI] [PubMed] [Google Scholar]
- 33.Amigorena S, Bonnerot C, Drake JR, Choquet D, Hunziker W, Guillet JG, et al. Cytoplasmic domain heterogeneity and functions of IgG Fc receptors in B lymphocytes. Science. 1992;256:1808–1812. doi: 10.1126/science.1535455. [DOI] [PubMed] [Google Scholar]
- 34.Muta T, Kurosaki T, Misulovin Z, Sanchez M, Nussenzweig MC, Ravetch JV. A 13-amino-acid motif in the cytoplasmic domain of FccRIIB modulates B-cell receptor signalling. Nature. 1994;368:70–73. doi: 10.1038/368070a0. [DOI] [PubMed] [Google Scholar]
- 35.Getahun A, Heyman B. Studies on the mechanism by which antigen-specific IgG suppresses primary antibody responses: evidence for epitope masking and decreased localization of antigen in the spleen. Scand J Immunol. 2009;70:277–287. doi: 10.1111/j.1365-3083.2009.02298.x. [DOI] [PubMed] [Google Scholar]
- 36.Heyman B, Pilstrom L, Shulman MJ. Complement activation is required for IgM-mediated enhancement of the antibody response. J Exp Med. 1988;167:1999–2004. doi: 10.1084/jem.167.6.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Applequist SE, Dahlstrom J, Jiang N, Molina H, Heyman B. Antibody production in mice deficient for complement receptors 1 and 2 can be induced by IgG/Ag and IgE/Ag, but not IgM/Ag complexes. J Immunol. 2000;165:2398–2403. doi: 10.4049/jimmunol.165.5.2398. [DOI] [PubMed] [Google Scholar]
- 38.Diaz de Stahl T, Dahlstrom J, Carroll MC, Heyman B. A role for complement in feedback-enhancement of antibody responses by IgG3. J Exp Med. 2003;197:1183–1190. doi: 10.1084/jem.20022232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Greenspan NS, Cooper LJN. Intermolecular cooperativity: a clue to why mice have IgG3? Immunol Today. 1992;13:164–168. doi: 10.1016/0167-5699(92)90120-V. [DOI] [PubMed] [Google Scholar]
- 40.Dempsey PW, Allison ME, Akkaraju S, Goodnow CC, Fearon DT. C3d of complement as a molecular adjuvant: bridging innate and acquired immunity. Science. 1996;271:348–350. doi: 10.1126/science.271.5247.348. [DOI] [PubMed] [Google Scholar]
- 41.Getahun A, Dahlstrom J, Wernersson S, Heyman B. IgG2a-mediated enhancement of Ab- and T-cell responses and its relation to inhibitory and activating FccRs. J Immunol. 2004;172:5269–5276. doi: 10.4049/jimmunol.172.9.5269. [DOI] [PubMed] [Google Scholar]
- 42.Hermeling S, Crommelin DJ, Schellekens H, Jiskoot W. Structure-immunogenicity relationships of therapeutic proteins. Pharm Res. 2004;21:897–903. doi: 10.1023/b:pham.0000029275.41323.a6. [DOI] [PubMed] [Google Scholar]
- 43.Reding MT, Wu H, Krampf M, Okita DK, Diethelm-Okita BM, Key NS, et al. CD4+ T cell response to Factor VIII in haemophilia A, acquired haemophilia and healthy subjects. Thromb Haemost. 1999;82:509–515. [PubMed] [Google Scholar]
- 44.Reding MT, Wu H, Krampf M, Okita DK, Diethelm-Okita BM, Christie BA, et al. Sensitization of CD4+ T cells to coagulation Factor VIII: Response in congenital and acquired haemophilia patients and in healthy subjects. Thromb Haemost. 2000;84:643–652. [PubMed] [Google Scholar]
- 45.Bachmann MF, Kundig TM, Kalberer CP, Hengartner H, Zinkernagel RM. How many specific B cells are needed to protect against a virus? J Immunol. 1994;152:4235–4241. [PubMed] [Google Scholar]
- 46.Sallusto F, Schaerli P, Loetscher P, Schaniel C, Lenig D, Mackay CR, et al. Rapid and coordinated switch in chemokine receptor expression during dendritic cell maturation. Eur J Immunol. 1998;28:2760–2769. doi: 10.1002/(SICI)1521-4141(199809)28:09<2760::AID-IMMU2760>3.0.CO;2-N. [DOI] [PubMed] [Google Scholar]
- 47.Sparwasser T, Koch ES, Vabulas RM, Heeg K, Lipford GB, Ellwart JW, et al. Bacterial DNA and immunostimulatory CpG oligonucleotides trigger maturation and activation of murine dendritic cells. Eur J Immunol. 1998;28:2045–2054. doi: 10.1002/(SICI)1521-4141(199806)28:06<2045::AID-IMMU2045>3.0.CO;2-8. [DOI] [PubMed] [Google Scholar]
- 48.De Smedt T, Pajak B, Muraille E, Lespagnard L, Heinen E, DeBaetselier P, et al. Regulation of dendritic cell numbers and maturation by lipopolysaccharide in vivo. J Exp Med. 1996;184:1413–1424. doi: 10.1084/jem.184.4.1413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Coulie P, Van Snick J. Enhancement of IgG anti-carrier responses by IgG2-anti-hapten antibodies in mice. Eur J Immunol. 1985;15:793–799. doi: 10.1002/eji.1830150810. [DOI] [PubMed] [Google Scholar]
- 50.van der Laken CJ, Voskuyl AE, Roos JC, Stigter van WM, de Groot ER, Wolbink G, et al. Imaging and serum analysis of immune complex formation of radio-labeled infliximab and anti-infliximab in responders and non-responders to therapy for rheumatoid arthritis. Ann Rheum Dis. 2007;66:253–256. doi: 10.1136/ard.2006.057406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Bajtay Z, Csomor E, Sándor N, Erdei A. Expression and role of Fc- and complement-receptors on human dendritic cells. Immunol Lett. 2006;104:46–52. doi: 10.1016/j.imlet.2005.11.023. [DOI] [PubMed] [Google Scholar]
- 52.Bartelds GM, Wijbrandts CA, Nurmohamed MT, Stapel S, Lems WF, Aarden L, et al. Anti-infliximab and anti-adalimumab antibodies in relation to response to adalimumab in infliximab switchers and anti-tumour necrosis factor naive patients: a cohort study. Ann Rheum Dis. 2010;69:817–821. doi: 10.1136/ard.2009.112847. [DOI] [PubMed] [Google Scholar]
- 53.Ehrenforth S, Kreuz W, Scharrer I, Linde R, Funk M, Güngör T, et al. Incidence of development of factor VIII and factor IX inhibitors in haemophiliacs. Lancet. 1992;339:594–598. doi: 10.1016/0140-6736(92)90874-3. [DOI] [PubMed] [Google Scholar]
- 54.Reding MT, Lei S, Lei H, Green D, Gill J, Conti-Fine BM. Distribution of Th1- and Th2-induced anti-factor VIII IgG subclasses in congenital and acquired hemophilia patients. Thromb Haemost. 2002;88:568–575. [PubMed] [Google Scholar]
- 55.Reding MT. Immunological aspects of inhibitor development. Haemophilia. 2006;12:30–35. doi: 10.1111/j.1365-2516.2006.01363.x. [DOI] [PubMed] [Google Scholar]
- 56.DiMichele DM, Hoots WK, Pipe SW, Rivard GE, Santagostino E. International workshop on immune tolerance induction: consensus recommendations. Haemophilia. 2007;13:1–22. doi: 10.1111/j.1365-2516.2007.01497.x. [DOI] [PubMed] [Google Scholar]
- 57.Wagner CL, Schantz A, Barnathan E, Olson A, Mascelli MA, Ford J, et al. Consequences of immunogenicity to the therapeutic monoclonal antibodies ReoPro and Remicade. Dev Biol (Basel) 2003;112:37–53. [PubMed] [Google Scholar]
- 58.Meritet JF, Maury C, Tovey MG. Induction of tolerance to recombinant therapeutic proteins. J Interferon Cytokine Res. 2001;21:1031–1038. doi: 10.1089/107999001317205150. [DOI] [PubMed] [Google Scholar]
- 59.Nagler-Anderson C, Terhoust C, Bhan AK, Podolsky DK. Mucosal antigen presentation and the control of tolerance and immunity. Trends Immunol. 2001;22:120–122. doi: 10.1016/s1471-4906(00)01830-5. [DOI] [PubMed] [Google Scholar]
- 60.Stickler M, Valdes AM, Gebel W, Razo OJ, Faravashi N, Chin R, et al. The HLA-DR2 haplotype is associated with an increased proliferative response to the immunodominant CD4(+) T-cell epitope in human interferon-beta. Genes Immun. 2004;5:1–7. doi: 10.1038/sj.gene.6364027. [DOI] [PubMed] [Google Scholar]
- 61.Jones TD, Phillips WJ, Smith BJ, Bamford CA, Nayee PD, Baglin TP, et al. Identification and removal of a promiscuous CD4+ T cell epitope from the C1 domian of factor VIII. J Thromb Haemost. 2005;3:991–1000. doi: 10.1111/j.1538-7836.2005.01309.x. [DOI] [PubMed] [Google Scholar]
- 62.Tangri S, Mothe BF, Eisenbraun J, Sidney J, Southwood S, Briggs K, et al. Rationally engineered therapeutic proteins with reduced immunogenicity. J Immunol. 2005;174:3187–3196. doi: 10.4049/jimmunol.174.6.3187. [DOI] [PubMed] [Google Scholar]
- 63.Barbosa MD, Vielmetter J, Chu S, Smith DD, Jacinto J. Clinical link between MHC class II haplotype and interferon-beta (IFNbeta) immunogenicity. Clin Immunol. 2006;118:42–50. doi: 10.1016/j.clim.2005.08.017. [DOI] [PubMed] [Google Scholar]
- 64.Harding FA, Stickler MM, Razo J, Dubridge RB. The immunogenicity of humanized and fully human antibodies: Residual immunogenicity resides in the CDR regions. MAbs. 2010;1:2. doi: 10.4161/mabs.2.3.11641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Yeung VP, Chang J, Miller J, Barnett C, Stickler M, Harding FA. Elimination of an immunodominant CD4+ T cell epitope in human IFNbeta does not result in an in vivo response directed at the subdominant epitope. J Immunol. 2004;172:6658–6665. doi: 10.4049/jimmunol.172.11.6658. [DOI] [PubMed] [Google Scholar]
- 66.Jaber A, Baker M. Assessment of the immunogenicity of different interferon beta-1a formulations using ex vivo T-cell assays. J Pharm Biomed Anal. 2007;43:1256–1261. doi: 10.1016/j.jpba.2006.10.023. [DOI] [PubMed] [Google Scholar]
- 67.Jaber A, Driebergen R, Giovannoni G, Schellekens H, Simsarian J, Antonelli M. The Rebif new formulation story: it's not trials and error. Drugs R D. 2007;8:335–348. doi: 10.2165/00126839-200708060-00002. [DOI] [PubMed] [Google Scholar]
- 68.Giovannoni G, Barbarash O, Casset-Semanaz F, King J, Metz L, Pardo G, et al. On behalf of the Rebif® New Formulation Study Group. Safety and immunogenicity of a new formulation of interferon {beta}-1a (Rebif(R) New Formulation) in a Phase IIIb study in patients with relapsing multiple sclerosis: 96-week results. Mult Scler. 2009;15:219–228. doi: 10.1177/1352458508097299. [DOI] [PubMed] [Google Scholar]
- 69.Janeway CA. “Approaching the asymptote? Evolution and revolution in immunology,”. Cold Spring Harb Symp Quant Biol. 1989;54:1–13. doi: 10.1101/sqb.1989.054.01.003. [DOI] [PubMed] [Google Scholar]
- 70.Rosloniec EF, Brand DD, Myers LK, Whittington KB, Gumanovskaya M, Zaller DM, et al. An HLA-DR1 transgene confers susceptibility to collagen-induced arthritis elicited with human type II collagen. J Exp Med. 1997;185:1113–1122. doi: 10.1084/jem.185.6.1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Black KE, Murray JA, David CS. HLA-DQ determines the response to exogenous wheat proteins: a model of gluten sensitivity in transgenic knockout mice. J Immunol. 2002;169:5595–5600. doi: 10.4049/jimmunol.169.10.5595. [DOI] [PubMed] [Google Scholar]
- 72.Madoiwa S, Yamauchi T, Hakamata Y, Kobayashi E, Arai M, Sugo T, et al. Induction of immune tolerance by neonatal intravenous injection of human factor VIII in murine hemophilia A. J Thromb Haemost. 2004;2:754–762. doi: 10.1111/j.1538-7933.2004.00671.x. [DOI] [PubMed] [Google Scholar]
- 73.Madoiwa S, Yamauchi T, Kobayashi E, Hakamata Y, Dokai M, Makino N, et al. Induction of factor VIII-specific unresponsiveness by intrathymic factor VIII injection in murine hemophilia A. J Thromb Haemost. 2009;2:754–762. doi: 10.1111/j.1538-7836.2009.03314.x. Erratum in: J Thromb Haemost 2004; 2:1504. [DOI] [PubMed] [Google Scholar]
- 74.Southwood S, Sidney J, Kondo A, del Guercio MF, Appella E, Hoffman S, et al. Several common HLADR types share largely overlapping peptide binding repertoires. J Immunol. 1998;160:3363–3373. [PubMed] [Google Scholar]
- 75.Traggiai E, Chicha L, Mazzucchelli L, Bronz L, Piffaretti JC, Lanzavecchia A, et al. Development of a human adaptive immune system in cord blood cell-transplanted mice. Science. 2004;304:104–107. doi: 10.1126/science.1093933. [DOI] [PubMed] [Google Scholar]
- 76.Ishikawa F, Yasukawa M, Lyons B, Yoshida S, Miyamoto T, Yoshimoto G, et al. Development of functional human blood and immune systems in NOD/SCID/IL2 receptor {gamma} chain(null) mice. Blood. 2005;106:1565–1573. doi: 10.1182/blood-2005-02-0516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Watanabe Y, Takahashi T, Okajima A, Shiokawa M, Ishii N, Katano I, et al. The analysis of the functions of human B and T cells in humanized NOD/shi-scid/gammac(null) (NOG) mice (hu-HSC NOG mice) Int Immunol. 2009;21:843–858. doi: 10.1093/intimm/dxp050. [DOI] [PubMed] [Google Scholar]
- 78.Bontrop RE, Otting N, de Groot NG, Doxiadis GG. Major histocompatibility complex class II polymorphisms in primates. Immunol Rev. 1999;167:339–350. doi: 10.1111/j.1600-065x.1999.tb01403.x. [DOI] [PubMed] [Google Scholar]
- 79.Baker MP, Jones TD. Identification and removal of immunogenicity in therapeutic proteins. Curr Opin Drug Discov Devel. 200;10:219–227. [PubMed] [Google Scholar]
- 80.Perry LC, Jones TD, Baker MP. New approaches to prediction of immune responses to therapeutic proteins during preclinical development. Drugs RD. 2008;9:385–396. doi: 10.2165/0126839-200809060-00004. [DOI] [PubMed] [Google Scholar]
- 81.Harding FA, Stickler MM, Razo J, Dubridge RB. The immunogenicity of humanized and fully human antibodies: Residual immunogenicity resides in the CDR regions. MAbs. 2010:2. doi: 10.4161/mabs.2.3.11641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Marshall SA, Lazar GA, Chirino AJ, Desjarlais JR. Rational design and engineering of therapeutic proteins. Drug Discov Today. 2003;8:212–221. doi: 10.1016/s1359-6446(03)02610-2. [DOI] [PubMed] [Google Scholar]
- 83.Reding MT, Okita DK, Diethelm-Okita BM, Anderson TA, Conti-Fine BM. Epitope repertoire of human CD4(+) T cells on the A3 domain of coagulation factor VIII. J Thromb Haemost. 2004;2:1385–1394. doi: 10.1111/j.1538-7836.2004.00850.x. [DOI] [PubMed] [Google Scholar]
- 84.Reding MT, Okita DK, Diethelm-Okita BM, Anderson TA, Conti-Fine BM. Human CD4+ T-cell epitope repertoire on the C2 domain of coagulation factor VIII. J Thromb Haemost. 2003;1:1777–1784. doi: 10.1046/j.1538-7836.2003.00251.x. [DOI] [PubMed] [Google Scholar]
- 85.Jones TD, Hanlon M, Smith BJ, Heise CT, Nayee PD, Sanders DA, et al. The development of a modified human IFNalpha2b linked to the Fc portion of human IgG1 as a novel potential therapeutic for the treatment of hepatitis C virus infection. J Interferon Cytokine Res. 2004;24:560–572. doi: 10.1089/jir.2004.24.560. [DOI] [PubMed] [Google Scholar]
- 86.Stickler M, Rochanayon N, Razo OJ, Mucha J, Gebel W, Faravashi N, et al. An in vitro human cell-based assay to rank the relative immunogenicity of proteins. Toxicol Sci. 2004;77:280–289. doi: 10.1093/toxsci/kfh021. [DOI] [PubMed] [Google Scholar]
- 87.Stickler M, Chin R, Faravashi N, Gebel W, Razo OJ, Rochanayon N, et al. Human population-based identification of CD4(+) T-cell peptide epitope determinants. J Immunol Methods. 2003;281:95–108. doi: 10.1016/s0022-1759(03)00279-5. [DOI] [PubMed] [Google Scholar]
- 88.Jones TD, Crompton LJ, Carr FJ, Baker MP. Deimmunization of monoclonal antibodies. Methods Mol Biol. 2009;525:405–423. doi: 10.1007/978-1-59745-554-1_21. [DOI] [PubMed] [Google Scholar]
- 89.Morris MJ, Divgi CR, Pandit-Taskar N, Batraki M, Warren N, Nacca A, et al. Pilot trial of unlabeled and indium-111-labeled anti-prostate-specific membrane antigen antibody J591 for castrate metastatic prostate cancer. Clin Cancer Res. 2005;11:7454–7461. doi: 10.1158/1078-0432.CCR-05-0826. [DOI] [PubMed] [Google Scholar]
- 90.Vallabhajosula S, Goldsmith SJ, Kostakoglu L, Milowsky MI, Nanus DM, Bander NH. Radioimmunotherapy of prostate cancer using 90Y- and 177Lu-labeled J591 monoclonal antibodies: effect of multiple treatments on myelotoxicity. Clin Cancer Res. 2005;11:7195–7200. doi: 10.1158/1078-0432.CCR-1004-0023. [DOI] [PubMed] [Google Scholar]
- 91.Pandit-Taskar N, O'Donoghue JA, Morris MJ, Wills EA, Schwartz LH, Gonen M, et al. Antibody mass escalation study in patients with castration-resistant prostate cancer using 111In-J591: lesion detectability and dosimetric projections for 90Y radioimmunotherapy. J Nucl Med. 2008;49:1066–1074. doi: 10.2967/jnumed.107.049502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Galsky MD, Eisenberger M, Moore-Cooper S, Kelly WK, Slovin SF, DeLaCruz A, et al. Phase I trial of the prostate-specific membrane antigen-directed immunoconjugate MLN2704 in patients with progressive metastatic castration-resistant prostate cancer. J Clin Oncol. 2008;26:2147–2154. doi: 10.1200/JCO.2007.15.0532. [DOI] [PubMed] [Google Scholar]