Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2014 Sep 25.
Published in final edited form as: J Carcinog Mutagen. 2014 May 31;5(3):1000176. doi: 10.4172/2157-2518.1000176

IgE-based Immunotherapy of Cancer -A Comparative Oncology Approach

Josef Singer 1, Erika Jensen-Jarolim 1,2,*
PMCID: PMC4174352  EMSID: EMS60228  PMID: 25264496

Abstract

Antibody-based immunotherapies are important therapy options in human oncology. Although human humoral specific immunity is constituted of five different immunoglobulin classes, currently only IgG-based immunotherapies have proceeded to clinical application.

This review, however, discusses the benefits and difficulties of IgE-based immunotherapy of cancer, with special emphasis on how to translate promising preclinical results into clinical studies. Pursuing the “Comparative Oncology” approach, novel drug candidates are investigated in clinical trials with veterinary cancer patients, most often dogs. By this strategy drug development could be speeded up, animal experiments could be reduced and novel therapy options could be introduced benefitting humans as well as man’s best friend.

Keywords: IgE, AllergoOncology, Comparative oncology, Comparative medicine, Immunotherapy

Targeted Therapies of Cancer

Cancer is a huge burden of our societies with an overall worldwide incidence of 182,3 cases per 100.000 inhabitants and an overall mortality of 102,4/100.000 according to the International Agency for Research on Cancer of the World Health Organization (estimated age-standardized incidence and mortality rates (ASR) for both sexes). Highest incidence rates are reported for breast, colorectal and cervical cancer in women and lung and prostate cancer in men [1].

Current treatment options comprise of surgery, chemotherapy or radiation plus more recently introduced targeted therapies. Targeted therapies aim to specifically address malignantly transformed cells while sparing healthy tissues [2]. Thus, receptors, which are important during embryonic development and readopted by cancer cells, belong to the most promising targets. One of the most prominent molecules of that kind is the human epidermal growth factor receptor-2 (HER-2). HER-2 is a receptor tyrosine kinase, mediating signals for cell proliferation, cell mobility and survival [3]. In the absence of a known ligand [4,5], activation is achieved by homo- or heterodimerization [6]. HER-2 is very important during embryonic development, e.g. it plays a role in ductal morphogenesis of the mammary gland [7], but it is almost not expressed on adult tissue, except the heart [8]. On the contrary, HER-2 is overexpressed in breast, ovarian, gastric, colorectal, pancreatic, and endometrial cancers [9].

Another closely related receptor tyrosine-kinase is the epidermal growth factor receptor (EGFR). Its overexpression is associated with head and neck squamous cell carcinoma (HNSCC), non-small-cell lung cancer (NSCLC), colorectal cancer (CRC), breast and pancreatic cancer, but also with certain types of brain cancer [10]. In contrast to HER-2, EGFR senses the epidermal growth factor (EGF) and other important growth signals, such as transforming growth factor-α (TGF-α) or amphiregulin [11-14]. EGFR is physiologically required for promoting cell proliferation and DNA repair [15], but can also lead to tumor growth, progression, and evasion of apoptosis via the activation of PLC-γ-PKC, Ras-Raf-MEK, PI-3K-Akt-mTOR and JAK2-STAT3 pathways [12,16]. Overall, EGFR and HER-2 together with HER-3 and HER-4 belong to the ErbB-family [17], which derives its name from the homology to the erythroblastic leukemia viral oncogene protein (v-erb-B, [18,19]).

Currently two forms of targeted therapies against EGFR and HER-2 are in clinical use: i) blocking the intracellular receptor tyrosine kinase with small molecules and ii) attacking the extracellular domains of the receptor with monoclonal antibodies.

Small molecules targeting EGFR comprise erlotinib (Tarceva®, Roche) and gefitinib (Iressa®, AstraZeneca) plus the dual kinase inhibitors lapatinib (Tykerb®, GlaxoSmithKline) and afatinib (Gilotrif®, Böhringer Ingelheim), the latter inhibiting HER-2 as well ([20], see Table 1). Especially the reversible inhibitors gefitinib, being FDA-approved in May 2003 [21] and erlotinib, with FDA-approval in November 2004 [22], are successfully applied in non-small-cell lung cancer [23]. Although gefitinib was recalled from that indication in the US, it is still widely used in Japan, where patients display a higher rate of EGFR-mutations in NSCLC [24], and also received marketing authorization in the European Union in 2009 [25]. Moreover, erlotinib is approved for the treatment of advanced pancreatic cancer [26] and several next generation irreversible EGFR-tyrosine kinase inhibitors, like canertinib, are under investigation for their efficacy in breast [27], colorectal, lung, pancreatic, renal, head and neck, gynecologic and prostate cancer [28].

Table 1.

Overview of indications for approved targeted therapies against EGFR and HER-2.

Cancer Type EGFR expression HER-2 expression Approved Targeted Therapy
Breast Cancer Yes Yes Lapatinib, trastuzumab, pertuzumab, T-DM1
Colorectal Cancer Yes Yes Cetuximab, panitumumab
Gastric Cancer (GEJ) No Yes Trastuzumab
HNSCC Yes No Cetuximab
NSCLC Yes No Afatinib, Erlotinib, Gefitinib
Pancreatic Cancer Yes Yes Erlotinib

The most prominent tyrosine kinase inhibitor (TKI) for HER-2 is lapatinib (Tykerb®, GlaxoSmithKline), the above mentioned reversible dual inhibitor of HER-2 and EGFR [29], which was FDA-approved in March 2007 for the treatment of advanced breast cancer [30]. Also in this case, irreversible inhibitors, like neratinib or again canertinib are widely investigated [31].

In contrast to small molecules that intracellularly interfere signaling via blocking the kinase activity, monoclonal antibodies directed against EGFR and HER-2 aim to extracellularly inhibit ligand binding or dimerization of these receptors, respectively [32].

For targeting EGFR, two monoclonal antibodies are currently in clinical use, cetuximab (Erbitux®, Merck KGaA), which was FDA-approved in February 2004 [33] and panitumumab (Vectibix®, Amgen), which received FDA-approval in September 2006 ([34], Table 1). In particular cetuximab, a human-murine chimeric IgG1 antibody has become an indispensable cornerstone in the treatment of advanced-stage metastatic CRC and advanced HNSCC [35,36]. Cetuximab finds its epitope within the ligand-binding site of EGFR (extracellular domain III) and can thus block binding of growth signals [37,38]. Panitumumab works mechanistically similar; it can also prevent EGF-binding via sterical hindrance [39], but on a different epitope on domain III, though partially overlapping [40]. Panitumumab is successfully applied in the treatment of metastatic colorectal cancer [41,42].

For all above mentioned therapeutics, wild type (wt) Kirsten rat sarcoma viral oncogene homolog (KRAS)-status of the patient is of uttermost importance, as it could be demonstrated, that acquired KRAS mutations lead to resistance against EGFR targeting [43]. As KRAS is a downstream effector-protein in the EGFR-signaling pathway, mutations that lead to constitutive activation of KRAS counteract growth signal inhibition of all EGFR targeting drugs [44]. Therefore KRAS-status is meanwhile routinely determined for every human patient before any EGFR-specific treatment is initiated [45].

Also in case of HER-2 targeting, two monoclonal antibody therapies are FDA-approved: trastuzumab (Herceptin®, Roche) and pertuzumab (Perjeta®, Genentech). Especially trastuzumab, being FDA approved in September 1998 [46], proved to be highly successful for the treatment of metastatic breast cancer [3] and has later received importance also for treatment of metastatic gastric cancer [47] and tumors of the gastroesophageal junction (GEJ, [48]). Trastuzumab has been so successful in breast cancer therapy, that very recently, in February 2013, also a drug-conjugated trastuzumab derivate, trastuzumabemtansine (T-DM1, Kadcyla®, Roche) was approved by the FDA for treatment of advanced breast cancer [49,50], fuelling the emerging field of antibody-drug conjugates [51].

The other HER-2 targeting antibody, pertuzumab received FDA-approval in June 2012 [52] and is also applied for treatment of metastatic breast cancer [53], as specified in Table 1.

Trastuzumab and Pertuzumab target different epitopes on HER-2: trastuzumab binds to subdomain IV on the extracellular domain (ECD) of HER-2 [54], whereas pertuzumab targets subdomain II [55]. As HER-2 has no endogenous ligand [5], the mechanisms of action of trastuzumab and pertuzumab differ from those of the mentioned EGFR-targeting immunoglobulins. Trastuzumab sterically prevents the formation of HER-2 homodimers or highly active heterodimers with other ErbB-family members [3], mainly HER-3 [56,57] and EGFR ([58,59]. Moreover, upon trastuzumab binding, HER-2 gets endocytosed and shedding of the receptor is inhibited, which otherwise would lead to an actively-signaling p95-remnant on the cancer cell surface [3]. Pertuzumab, on the other hand, binds more distant from the cell membrane, is more efficient in preventing heterodimer formation [60] and in contrast to trastuzumab, which inhibits ligand-independent dimerization, pertuzumab especially inhibits ligand-induced HER-2 heterodimers [57].

This different mode of action prompted researchers to investigate a combination therapy of both antibodies in preclinical models, resulting in more complete HER-2 blockade, and efficacy in cases where cancer had progressed after trastuzumab monotherapy [61]. Also in clinical settings, this combination therapy proved to be highly effective and peaked in June 2012 in the first FDA approval of a dual HER-2 targeting regimen for metastatic breast cancer [62].

Immunological Effects of Antibodies

The mechanism of action of monoclonal antibodies, however, cannot be confined to their growth signal inhibitory capacity only; as all mentioned monoclonal antibodies feature fully functional human constant regions, thus they are also able to attract immune effector cells to the site of the tumor and trigger immune cell mediated cancer cell death. Among the attracted immune cells are monocytes and macrophages dominant [63], which are known for their tumoricidic potential in mediating antibody-dependent cell-mediated cytotoxicity (ADCC) via insertion of granzyme B and caspase enzymes [64]. Moreover, our group could demonstrate that monocytes are also able to mediate high levels of antibody-dependent cell-mediated phagocytosis (ADCP) upon trastuzumab treatment [65]. Other attracted cell types expressing Fc-receptors for antibody binding, are NK-cells and neutrophilic granulocytes, which have been shown to bear high tumoricidic potential with regard to ADCC [66,67]. Moreover, professional antigen-presenting cells, that also express Fcreceptors such as dendritic [68] or Langerhans Cells [69] can be attracted and can induce activation of tumor-reactive T-cells upon facilitated antigen uptake and presentation. This mechanism has been demonstrated to lead to tumor regression in a xenograft model of cetuximab treatment in concert with reconstituted immune cells [70].

Upon a closer look, all mentioned EGFR or HER-2 targeting antibodies belong to the IgG class of immunoglobulins, with cetuximab, trastuzumab and pertuzumab being IgG1 antibodies [71,72] and panitumumab, being an IgG2 [73]. In fact, all currently FDA-approved monoclonal antibodies for therapy of cancer are gamma-Immunoglobulins [71]. This is indeed astonishing as the human Immunoglobulin repertoire consists of 5 different classes: IgA, IgD, IgE, IgG and IgM [74].

Based on their different constant domains, all of them bear distinct physiological properties with respect to distribution, tissue penetration and function, such as complement-activation or ADCC and ADCP mediation [75]:

IgM, the primary antibody response against pathogens [76] is highly active in opsonization, which leads to pathogen clearance by phagocytic cells [77].

IgA protects body surfaces [78], such as the respiratory, gastrointestinal or genitourinary tract [79] and is abundantly found in secrets like tear fluid [80] or saliva [78], where it exerts neutralizing functions [81]. The function of IgA in mother’s milk [82] is of special interest as it protects the infant against pathogens in the mother’s environment, which are also of high risk to the child [83].

IgG is the most abundant antibody isotype in the bloodstream, as it constitutes 70% of all serum immunoglobulins [79]. IgG antibodies, which can be further subdivided in IgG1-4 in humans, exert systemic immune-protection by binding to bacteria, viruses and fungi with high affinity. In general, IgG1 and IgG3 antibodies are mainly produced in response to protein antigens, whereas IgG2 and IgG4 antibodies react against pathogens with polysaccharide capsules, like Streptococcus pneumoniae [75,79].

The biological role of IgD, however, remains still enigmatic; discovered late (in 1965) in the serum of a myeloma patient [84,85], it was long neglected, because it is only cleaved in minor amounts. Membrane-bound IgD on the surface of B-cells though, was found to regulate B-cell activation. Despite thorough investigation in recent years, the physiological and pathophysiological role of IgD is still unclear; however, an interesting aspect is, that IgD was found to bind to certain bacterial proteins with relatively high affinity, but not via its antigen binding-site, as rather through sugar residues on its constant region [86]. Moreover, it has recently been shown, that circulating IgD can activate antimicrobial, proinflammatory and B cell-stimulating effects in basophilic granulocytes [87].

IgE, finally, is the most recently discovered immunoglobulin subclass, as it was only first described in 1966 by Teruko and Kimishige Ishizaka as a novel immunoglobulin in the serum of an atopic individual [88]. At the same time, Bennich and Johannsson could purify a paraprotein from serum of a myeloma patient, which they termed “IgND” [89]. They could soon link this novel “IgND” to asthma [90] and it turned out to be the same protein as the group from Japan had found. On a meeting in Lausanne in February 1968, finally, under the guidance of the WHO Immunoglobuline Reference Laboratory, it was decided to designate this novel immunoglobuline “IgE” [91,92].

IgE plays an important role in defense against parasites [93] and is a key molecule in the pathophysiology of allergic diseases such as atopic dermatitis, asthma, food allergy or anaphylaxis [94]. Upon crosslinking of IgE-antibodies, bound to FcεRI-receptors on the surface of mast cells or basophils, histamine and leukotrienes are released leading to the manifestation of allergic symptoms [95].

IgE and Cancer

IgE is mostly known for its detrimental role in allergy, but several studies have for long pointed towards a natural tumor surveillance function of this antibody isotype [96,97]. Interestingly, large epidemiologic studies could reveal an inverse association between the history of atopic diseases and cancer [98]. In 2005, Turner et al. published a study enrolling 1.1 million US-american adults with self-reported, physician-diagnosed asthma or hay fever, who had no cancer at baseline and were followed up for 18 years. In this population, a relative-risk reduction for all cancer mortality could be observed [relative risk (RR) = 0.88; 95% confidence interval (CI) 0.83-0.93]. However, in a separate analysis of never-smokers, this effect still persisted, but was not significant anymore [99].

In a following literature analysis of studies from the MEDLINE® database from 1966 to August 2005, the same group described “strong inverse associations for pancreatic cancer and glioma, whereas lung cancer was positively associated with asthma”. However, methodical issues to these historical studies with regard to exposure assessment, confounding and bias were addressed by the authors [100].

The most recent study investigating a possible association between IgE and cancer was published in 2010: Van Hemelrijck et al. reviewed 27 studies from PubMed and EMBASE™ and surveyed a Swedish cohort of 24.820 people, who underwent IgE measurements. Here, the authors could show a weak inverse association in their cohort, and a pattern by cancer type in the meta-analysis of the historical studies [101].

Another interesting observation was made when the anti-IgE antibody omalizumab (Xolair®, Novartis) underwent clinical trials and pooled phase I to III data was evaluated. Omalizumab, which removes IgE from the circulation, is currently approved for therapy of severe persistent asthma [102]. In those mentioned phase I to III trials with allergics undergoing omalizumab treatment, a slightly higher number of malignant neoplasms was observed in the anti-IgE-treated group (20 of 4127=0.5% compared to 5 of 2236=0.2% in the control group). Malignancies that occurred in the treatment group comprised of breast, non-melanoma skin, prostate, melanoma and parotid cancer. Subsequently, Busse et al. analyzed 67 phase I to IV trials of omalizumab and could not confirm any possible association between omalizumab treatment and cancer in this extended study [103].

Summarizing all epidemiologic observations one can only state, that a possible association between IgE and cancer remains still unclear due to the lack of big prospective studies.

A more mechanistic approach, however, was pursued, when Fu et al. purified immunoglobulins from tissue surrounding pancreatic cancers. They could isolate IgE antibodies which were not only specific for a 50 kDa pancreatic cancer antigen but were indeed able to mediate ADCC of pancreatic cancer cells in vitro [104], pointing towards a benefitial role of IgE-antibodies in defense against cancer.

IgE-based Immunotherapies of Cancer - Pioneer Studies

Pioneer studies with IgG and IgE antibodies of the same epitope specificity tested head-to-head revealed a higher potential of the IgE in terms of cytotoxicity. The very first studies were performed with Mov18IgG and IgE [105], antibodies that target the folate receptor (FR)-α. FR-α (also known as folate-binding protein, LK26 trophoblastic antigen or GP38) is a glycosylphosphatidylinositol (GPI)-anchored membrane protein that binds folic acid and is regarded as a tumor-associated-antigen (TAA) in gynecologic malignancies [106], due to its overexpression in more than 90% of epithelial ovarian cancers and in a subpopulation of uterine carcinomas. As folate is a necessary micronutrient of replicating cells, overexpression of FR-α facilitates enhanced growth of cancer cells [107].

For targeting of this receptor, Gould et al. could demonstrate in an ovarian cancer model, that Mov18IgE was able to mediate ADCC of FR-α expressing tumor cells in vitro and in vivo. In a mouse model using xenografted human FR-α overexpressing cells, mice that received Mov18IgE treatment developed in the presence of human peripheral blood mononuclear cells (PBMCs) significantly smaller tumors than those treated with Mov18IgG [105]. In a follow-up study, it could further be demonstrated, that also cytotoxic killing by monocytes can be efficiently triggered with IgE. In a subsequent nude mouse study, where again FR-α overexpressing tumors were grafted and PBMCs were reconstituted, the IgE-treated group had shown monocytic infiltration of the tumor xenografts, which was still persistent after 3 weeks and led to significantly longer survival. Moreover, upon a closer look in an in vitro flow cytometric model, specific ADCC of tumor cells, executed by monocytes upon Mov18IgE stimulation could be displayed. Finally phagocytosis of FR-α positive tumor cells by monocytes armed with FR-α specific IgE could be displayed by fluorescence microscopy [108].

Subsequently, we could demonstrate similar results for the HER-2 system in close collaboration with Prof. Gould and Dr. Karagiannis: upon generation of a recombinant trastuzumab-like IgE, constituted of the same variable regions as original trastuzumab (being an IgG1), it was shown in a flow cytometric assay, that the IgE antibody is highly effective in mediating ADCC of monocytes against HER-2 overexpressing cells. Interestingly, in this model, the IgE antibody mediated high levels of ADCC but only background ADCP, whereas the picture was completely opposite for the IgG, which mediated killing of tumor cells almost exclusively via ADCP [65]. This could be a first hint towards distinct mechanisms of tumor cell killing mediated by different immunoglobulin classes; however, this still has to be confirmed in more extensive experiments and has to be investigated also for other cancer types.

Possible Advantages and Pitfalls of IgE-based Cancer Immunotherapies

Apart from a possibly higher potential for mediating ADCC, IgE-based immunotherapies of cancer could have other beneficial effects: first, IgE antibodies have a uniquely high affinity to their receptors on immune cells (Ka~ 1010/M for FcεRI and Ka~ 108-109/M for the CD23 trimer complex), which significantly exceeds the affinities of IgG1-4 to their high-affinity receptor FcRI [94,109]. Thus, due to its rapid binding to Fcε-receptors on cells, IgE is quickly removed from the circulation, which is advantageous in terms of side-effects because of the short duration of the compound in the bloodstream. Moreover, potential IgE-immunotherapies would be effectively distributed to tumor tissues, as IgE antibodies bound to Fcε-receptors on e.g. mast cells can use those cells as shuttle systems to penetrate malignancies and as mast cells are tissue-resident immune cells [110], this transport would be highly efficient.

Consistently, we would like to quote the review “Problems of Delivery of Monoclonal Antibodies” by Reilly et al., who wrote that “the clinical success of monoclonal antibody-based cancer diagnosis and therapy depends, however, on solving a number of pharmacokinetic delivery problems. These include: (i) slow elimination of monoclonal antibodies from the blood and poor vascular permeability; (ii) low and heterogeneous tumour uptake” [111], and state that those two substantial challenges of anti-tumor immunotherapy could be simply addressed by using IgE.

Other possible advantages include the high sensitivity of IgE-effector cells to activation by antigens and the speed and amplitude of the response, which can most impressively be seen during allergic and anaphylactic reactions, typically beginning within minutes upon allergen exposure.

That is at the same time also the biggest concern of using IgE-based immunotherapies against cancer: recombinant IgE, applied intravenously, always bears the risk of anaphylactic reactions; therefore, careful selection of the target epitope is of uttermost importance in this regard.

During an anaphylactic reaction, preformed IgE, that is bound to Fcε-receptors on the surface of mast cells or basophilic granulocytes is cross-linked by allergens, which induces release of stored granules, containing vasoactive amines (e.g.: histamine) or lipid mediators (e.g. Prostaglandin D2, Platelet-activating factor, or leukotrienes) [112]. This rapid release can lead within minutes to fatal symptoms like asphyxiation from laryngeal swelling, circulatory collapse from hypotensive shock, cardiac arrest, or respiratory failure because of bronchoconstriction [113].

In order to prevent such effects, the target structure for designing passive immunotherapies with IgE-antibodies should not be expected to be cross-linking, which means, that the epitope should be

  • monovalent and

  • it should not circulate in the blood, or if,

  • it should only circulate in a monomeric form [114].

These requirements are fulfilled for the mentioned anti-FR-α antibody “Mov18IgE”, but also for the anti-HER-2 antibody “trastuzumab-like IgE”. For both antibodies it could be demonstrated, that monomeric target molecules do not trigger mediator release of mast cells, which were preloaded with their specific IgE-antibodies, respectively [65,115]. Furthermore, Rudman et al. could demonstrate, that although serum levels of FR-α were increased in ovarian cancer patients (up to 40 ng/ml) compared to healthy controls (mean=1,73; SD=3,45), basophilic granulocytes loaded with Mov18IgE were not significantly activated upon incubation with FR-α even at a concentration of 300 ng/ml [115].

On the other hand, both antibodies were shown to mediate mast cell degranulation upon incubation with tumor cells, displaying high numbers of target molecules in a repetitive manner on their surface [65,115]. Here, cross-linking of Fcε-receptors is highly efficient, and therefore local anaphylaxis at the tumor site could be expected, which would again be beneficial, as it results in initiation of a strong immune response. As mast cells also store tumorinhibiting agents in their granules, e.g. tumor necrosis factor (TNF)-α [116], this degranulation could also result in direct tumor cell killing. Moreover, also other cells involved in anaphylactic reactions, such as eosinophils, have been shown to execute tumoricidic functions, e.g. via secretion of granzyme A [117] or eosinophilic peroxidase [118].

Another big challenge of current immunotherapies with IgG antibodies is that not all human Fcγ-receptors are immune-activating, but one among them, FcγRIIb is -inhibiting [119]. Therefore, the tumoricidic effects of IgG-based immunotherapies also depend on the net ratio of binding to activating and inhibiting receptors. As it has recently been shown for IgG4, a subclass that shows relatively high binding affinity to FcγRIIb [120], this antibody is not able to trigger immune cell-mediated tumor cell killing in vitro, despite being TAA-specific. Moreover it was demonstrated, that IgG4 antibodies significantly impaired the killing potential of IgG1 antibodies of the same specificity in vitro and in vivo [121]. Strategies to overcome this limitation include modification of the posttranslational glycosylation of the IgG-constant regions’ heavy chains, as these sugar residues have been identified to be of high relevance for distinct binding affinities to different Fc-receptors [75]. For IgE on the other hand, there are no inhibitory receptors [114], so again this isotype could contribute to overcome a current challenge of immunotherapies of cancer.

However the Fcε-receptor-biology differs considerably between humans and mice, as the high affinity IgE receptor FcεRI is only expressed on mouse basophils and mast cells [122], whereas it has been described in humans on mast cells, basophils, eosinophils, monocytes, Langerhans and dendritic cells [123,124]. This is a huge limitation of current mouse models in displaying all mentioned in vivo benefits and risks of IgE-based immunotherapy of cancer and the great benefit of using a “Comparative Oncology” approach.

Comparative Medicine

Although human and veterinary medicine share the same goals and aims, namely to treat patients and promote health, currently both of them are distinct sciences with distinct studies taught on separate universities. Research is presently going on in one or the other, but there is little crosstalk between the two specialties.

The concept of Comparative Medicine, however, aims to study naturally occurring diseases across species to improve both human and veterinary medicine [125,126]. There is in fact no explicit reason for a strict separation of studies for humans or other mammals, as many pathophysiological processes have been shown to be similar and highly comparable, or as the german pathologist Rudolf Virchow stated: “Between animal and human medicine there are no dividing lines - nor should there be. The object is different but the experience obtained constitutes the basis of all medicine” [127].

This view, which is also in line with the wider concepts of “One Medicine”, or “One Health”, bringing together aspects of health of humans, animals and also their environment [128] is not really novel. Browsing through the history of medicine, this approach appears at many points, starting with Hippocrates and Plato in ancient Greece [129]. Even further back in time, physicians in Ancient India were trained to treat humans and animals, especially cattle, elephants and horses [130] and in Ancient China, medical treatment for horses was highly elaborated as well [131].

Also in Europe and North America human and veterinary medicine were closely interconnected for a long period of time, peaking in the 18th, 19th and early 20th century. Many important findings were made by comparative observation and experimentation during that time, for instance Edward Jenner (1749-1823) noted that milkers who had been in contact with cowpox-infected cows did not develop smallpox (variola minor) but only the milder cowpox. He also observed that this protection could be mediated by inoculation of a small amount of pus from cowpox blisters, a method he called “vaccination” because of the Latin word for cow, vacca [132]. Similarly, Louis Pasteur (1822-1895) worked on cholera in humans and chicken [133], Robert Koch (1843-1910) researched on human and bovine tuberculosis [134], and the mentioned Rudolf Virchow (1821-1902) worked on trichinella infections in humans and pigs [135].

Besides Virchow, the most important proponent of Comparative Medicine was Sir William Osler, a Canadian physician (1849-1919), who studied, worked and taught in London, Berlin, Vienna, Toronto and Montreal, and was one of the four founding professors of Johns Hopkins Hospital in Baltimore. Osler lectured at McGill University for medical students as well as for students from the Veterinary Medical College with emphasis on comparative topics. His research in this field concentrated on infectious diseases in dogs, pigs and cattle and his deep interest is documented in many editorials of the “Journal of Comparative Medicine and Surgery” [136].

Recent developments are no longer dependent on distinguished individuals, but comprise the establishment of special departments for Comparative Medicine within universities, such as at Stanford, Yale or at the University of California, Davis. Whereas Comparative Medicine focused mainly on infectious diseases in previous centuries, modern approaches tend to tackle another big burden of our societies, cancer, via the principle of “Comparative Oncology”.

Comparative Oncology

The Comparative Oncology approach aims to speed up drug development simultaneously for human and animal cancer patients via clinical trials in pet patients, primarily cats and dogs [137]. Although murine models have been proven to be highly effective with regards for understanding basic principles of malignant transformation, cancer signal transduction pathways or drug resistance formation, these models often poorly mimic human cancer for drug testing [138]. For many compounds, the translation of a safe and efficacious agent in mice into an actual drug fails [139], due to the poor presentation of key features of human cancer in murine tumor models, such as genomic instability, long latency periods or the lack of intra-tumor heterogeneity [140]. Moreover, the concept of toxicity studies, which are conducted in healthy animals, followed immediately by Phase I and Phase II trials in humans, often leaves many important questions unanswered, before treating a relatively high number of human patients [140,141].

On the other hand, also more and more extensive studies in animals cannot be the solution, as it would increase the ethical dilemma that potential benefits for humans stand against the costs sustained by animals. Thus, legislators and regulatory bodies state in their directives on drug development that the “3Rs” should be applied on animal experiments [142], which are: Replacement, Reduction, and Refinement [143,144 ].

In line with these concepts, clinical studies in animal patients, which suffer from spontaneously developed tumors, would allow the investigation of drug effects on malignancies that developed naturally within intact immune systems, in the context of their original tumor microenvironment in pet animals that share similar environmental factors as their owners, for instance pollution. Such trials in veterinary patients could replace many preclinical experiments, refine our models and ultimately reduce animal experiments. The information obtained from these studies would be highly relevant and valuable, while the treated veterinary patients would be provided with cutting edge research simultaneously [140].

Studies in this field of Comparative Oncology with treated dog patients, led to the advancement of surgical techniques, like limb sparing for sarcoma patients, elucidating hyperthermia or evaluating novel delivery strategies, like inhalation of cytokines or chemotherapies [137].

This development peaked recently in the formation of the “Comparative Oncology Trials Consortium” (COTC), a multicenter initiative of twenty Comparative Oncology centers throughout the USA. Founded and centrally managed by the National Cancer Institute of the National Institutes of Health (NIH), 11 clinical trials have been conducted so far, ranging from all fields of cancer therapy attempts like “Evaluation of RGD Targeted Delivery of Phage Expressing TNF-alpha to Tumor Bearing Dogs” (COTC001, closed trial) via “Preclinical Comparison of Three Indenoisoquinolines Candidates in Tumor Bearing Dogs” (COTC007b, open trial) to “Evaluation of the mTOR inhibitor Rapamycin in Dogs with Metastatic Osteosarcoma” (COTC008, closed trial) [145].

One of the most successful example of a recent clinical Comparative Oncology trial was published in 2009, in which the tyrosine kinase inhibitor toceranib phosphate (Palladia®, SU11654, Pfizer), targeting kit, vascular endothelial growth factor receptor 2 (VEGFR2) and platelet derived growth factor receptor-beta (PDGFRβ) [146] was tested in dog cancer patients with recurrent mast cell tumors. This large clinical phase III trial could demonstrate significant effects of toceranib phosphate with regard to overall response rates, median duration of objective responses and time to tumor progression [147], which has finally led to the approval of toceranib phosphate for mast cell tumors in dogs [148] and to the approval of sunitinib (Sutent®, SU11248, Pfizer), a similar compound, for therapy of human renal cell cancer and gastrointestinal stromal tumors (GIST, [149,150]).

Also for evaluation of IgE-based immunotherapies of cancer, trials in dog cancer patients would be highly valuable, due to the fact that dogs, in contrast to mice, share important principles of the IgE-biology with humans [151,152], underlined by the clinical observation that dogs also suffer from IgE-mediated diseases, such as atopic dermatitis [153] or food allergies [154]. Thus, the introduction of passive immunotherapy in veterinary clinical oncology would be highly valuable to elucidate the full potential of IgE-based Immunotherapy of Cancer. We contributed to this strategy by our recent molecular characterization of canine EGFR and the generation of a recombinant canine IgE of the exact cetuximab specificity [155, 156].

Acknowledgement

The study was supported by grants P23398-B11 and JS by W1205-B09 of the Austrian Science Fund (FWF). This text is the introductory chapter of the PhD-thesis of Dr. Josef Singer, submitted in January 2014, entitled “IgE-based Immunotherapies of Cancer - A Comparative Oncology Approach”.

References

  • 1.International Agency for Research on Cancer (IARC) of the World Health Organization (WHO) Section of Cancer Information Globocan 2012. 2012
  • 2.Cheng YD, Yang H, Chen GQ, Zhang ZC. Molecularly targeted drugs for metastatic colorectal cancer. Drug Des Devel Ther. 2013;7:1315–1322. doi: 10.2147/DDDT.S52485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hudis CA. Trastuzumab--mechanism of action and use in clinical practice. N Engl J Med. 2007;357:39–51. doi: 10.1056/NEJMra043186. [DOI] [PubMed] [Google Scholar]
  • 4.Avraham R, Yarden Y. Feedback regulation of EGFR signalling: decision making by early and delayed loops. Nat Rev Mol Cell Biol. 2011;12:104–117. doi: 10.1038/nrm3048. [DOI] [PubMed] [Google Scholar]
  • 5.Normanno N, De Luca A, Bianco C, Strizzi L, Mancino M, et al. Epidermal growth factor receptor (EGFR) signaling in cancer. Gene. 2006;366:2–16. doi: 10.1016/j.gene.2005.10.018. [DOI] [PubMed] [Google Scholar]
  • 6.Spector NL, Blackwell KL. Understanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol. 2009;27:5838–5847. doi: 10.1200/JCO.2009.22.1507. [DOI] [PubMed] [Google Scholar]
  • 7.Sternlicht MD. Key stages in mammary gland development: the cues that regulate ductal branching morphogenesis. Breast Cancer Res. 2006;8:201. doi: 10.1186/bcr1368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wadugu B, Kühn B. The role of neuregulin/ErbB2/ErbB4 signaling in the heart with special focus on effects on cardiomyocyte proliferation. Am J Physiol Heart Circ Physiol. 2012;302:H2139–2147. doi: 10.1152/ajpheart.00063.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.English DP, Roque DM, Santin AD. HER2 expression beyond breast cancer: therapeutic implications for gynecologic malignancies. Mol Diagn Ther. 2013;17:85–99. doi: 10.1007/s40291-013-0024-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Brand TM, Iida M, Li C, Wheeler DL. The nuclear epidermal growth factor receptor signaling network and its role in cancer. Discov Med. 2011;12:419–432. [PMC free article] [PubMed] [Google Scholar]
  • 11.Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2:127–137. doi: 10.1038/35052073. [DOI] [PubMed] [Google Scholar]
  • 12.Yarden Y, Shilo BZ. SnapShot: EGFR signaling pathway. Cell. 2007;131:1018. doi: 10.1016/j.cell.2007.11.013. [DOI] [PubMed] [Google Scholar]
  • 13.Busser B1, Sancey L, Brambilla E, Coll JL, Hurbin A. The multiple roles of amphiregulin in human cancer. Biochim Biophys Acta. 2011;1816:119–131. doi: 10.1016/j.bbcan.2011.05.003. [DOI] [PubMed] [Google Scholar]
  • 14.Citri A, Yarden Y. EGF-ERBB signalling: towards the systems level. Nat Rev Mol Cell Biol. 2006;7:505–516. doi: 10.1038/nrm1962. [DOI] [PubMed] [Google Scholar]
  • 15.Lo HW. Nuclear mode of the EGFR signaling network: biology, prognostic value, and therapeutic implications. Discov Med. 2010;10:44–51. [PMC free article] [PubMed] [Google Scholar]
  • 16.Han W, Lo HW. Landscape of EGFR signaling network in human cancers: biology and therapeutic response in relation to receptor subcellular locations. Cancer Lett. 2012;318:124–134. doi: 10.1016/j.canlet.2012.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Desai MD, Saroya BS, Lockhart AC. Investigational therapies targeting the ErbB (EGFR, HER2, HER3, HER4) family in GI cancers. Expert Opin Investig Drugs. 2013;22:341–356. doi: 10.1517/13543784.2013.761972. [DOI] [PubMed] [Google Scholar]
  • 18.Muleris M, Almeida A, Malfoy B, Dutrillaux B. Assignment of v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2 (ERBB2) to human chromosome band 17q21.1 by in situ hybridization. Cytogenet Cell Genet. 1997;76:34–35. doi: 10.1159/000134507. [DOI] [PubMed] [Google Scholar]
  • 19.Downward J, Yarden Y, Mayes E, Scrace G, Totty N, et al. Close similarity of epidermal growth factor receptor and v-erb-B oncogene protein sequences. Nature. 1984;307:521–527. doi: 10.1038/307521a0. [DOI] [PubMed] [Google Scholar]
  • 20.Cohen RB. Current challenges and clinical investigations of epidermal growth factor receptor (EGFR)- and ErbB family-targeted agents in the treatment of head and neck squamous cell carcinoma (HNSCC) Cancer Treat Rev. 2014;40:567–577. doi: 10.1016/j.ctrv.2013.10.002. [DOI] [PubMed] [Google Scholar]
  • 21.Cohen MH, Williams GA, Sridhara R, Chen G, Pazdur R. FDA drug approval summary: gefitinib (ZD1839) (Iressa) tablets. Oncologist. 2003;8:303–306. doi: 10.1634/theoncologist.8-4-303. [DOI] [PubMed] [Google Scholar]
  • 22.Cohen MH, Johnson JR, Chen YF, Sridhara R, Pazdur R. FDA drug approval summary: erlotinib (Tarceva) tablets. Oncologist. 2005;10:461–466. doi: 10.1634/theoncologist.10-7-461. [DOI] [PubMed] [Google Scholar]
  • 23.Reck M, Heigener DF, Mok T, Soria JC, Rabe KF. Management of non-small-cell lung cancer: recent developments. Lancet. 2013;382:709–719. doi: 10.1016/S0140-6736(13)61502-0. [DOI] [PubMed] [Google Scholar]
  • 24.Jiang H. Overview of gefitinib in non-small cell lung cancer: an Asian perspective. Jpn J Clin Oncol. 2009;39:137–150. doi: 10.1093/jjco/hyn139. [DOI] [PubMed] [Google Scholar]
  • 25.Vivanco I, Mellinghoff IK. Epidermal growth factor receptor inhibitors in oncology. Curr Opin Oncol. 2010;22:573–578. doi: 10.1097/CCO.0b013e32833edbdf. [DOI] [PubMed] [Google Scholar]
  • 26.Bareschino MA, Schettino C, Troiani T, Martinelli E, Morgillo F, et al. Erlotinib in cancer treatment. Ann Oncol. 2007;18(Suppl 6):vi35–41. doi: 10.1093/annonc/mdm222. [DOI] [PubMed] [Google Scholar]
  • 27.Ocaña A, Amir E. Irreversible pan-ErbB tyrosine kinase inhibitors and breast cancer: current status and future directions. Cancer Treat Rev. 2009;35:685–691. doi: 10.1016/j.ctrv.2009.08.001. [DOI] [PubMed] [Google Scholar]
  • 28.Rocha-Lima CM, Soares HP, Raez LE, Singal R. EGFR targeting of solid tumors. Cancer Control. 2007;14:295–304. doi: 10.1177/107327480701400313. [DOI] [PubMed] [Google Scholar]
  • 29.Hurvitz SA, Hu Y, O’Brien N, Finn RS. Current approaches and future directions in the treatment of HER2-positive breast cancer. Cancer Treat Rev. 2013;39:219–229. doi: 10.1016/j.ctrv.2012.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ryan Q, Ibrahim A, Cohen MH, Johnson J, Ko CW, et al. FDA drug approval summary: lapatinib in combination with capecitabine for previously treated metastatic breast cancer that overexpresses HER-2. Oncologist. 2008;13:1114–1119. doi: 10.1634/theoncologist.2008-0816. [DOI] [PubMed] [Google Scholar]
  • 31.Roy V, Perez EA. Beyond trastuzumab: small molecule tyrosine kinase inhibitors in HER-2-positive breast cancer. Oncologist. 2009;14:1061–1069. doi: 10.1634/theoncologist.2009-0142. [DOI] [PubMed] [Google Scholar]
  • 32.Roskoski R., Jr The ErbB/HER family of protein-tyrosine kinases and cancer. Pharmacol Res. 2014;79:34–74. doi: 10.1016/j.phrs.2013.11.002. [DOI] [PubMed] [Google Scholar]
  • 33.Goldberg RM. Cetuximab. Nat Rev Drug Discov Suppl. 2005:S10–11. doi: 10.1038/nrd1728. [DOI] [PubMed] [Google Scholar]
  • 34.Giusti RM, Shastri KA, Cohen MH, Keegan P, Pazdur R. FDA drug approval summary: panitumumab (Vectibix) Oncologist. 2007;12:577–583. doi: 10.1634/theoncologist.12-5-577. [DOI] [PubMed] [Google Scholar]
  • 35.Burotto M, Hartley ML, Marshall JL, Pishvaian MJ. Future of targeted agents in metastatic colorectal cancer. Colorectal Cancer. 2012:1. doi: 10.2217/crc.12.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Specenier P, Vermorken JB. Cetuximab: its unique place in head and neck cancer treatment. Biologics. 2013;7:77–90. doi: 10.2147/BTT.S43628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Masui H, Kawamoto T, Sato JD, Wolf B, Sato G, et al. Growth inhibition of human tumor cells in athymic mice by anti-epidermal growth factor receptor monoclonal antibodies. Cancer Res. 1984;44:1002–1007. [PubMed] [Google Scholar]
  • 38.Li S, Schmitz KR, Jeffrey PD, Wiltzius JJ, Kussie P, et al. Structural basis for inhibition of the epidermal growth factor receptor by cetuximab. Cancer Cell. 2005;7:301–311. doi: 10.1016/j.ccr.2005.03.003. [DOI] [PubMed] [Google Scholar]
  • 39.Dubois EA, Cohen AF. Panitumumab. Br J Clin Pharmacol. 2009;68:482–483. doi: 10.1111/j.1365-2125.2009.03492.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Voigt M1, Braig F, Göthel M, Schulte A, Lamszus K, et al. Functional dissection of the epidermal growth factor receptor epitopes targeted by panitumumab and cetuximab. Neoplasia. 2012;14:1023–1031. doi: 10.1593/neo.121242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.de Mello RA, Marques AM, Araújo A. Epidermal growth factor receptor and metastatic colorectal cancer: insights into target therapies. World J Gastroenterol. 2013;19:6315–6318. doi: 10.3748/wjg.v19.i38.6315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Prenen H, Vecchione L, Van Cutsem E. Role of targeted agents in metastatic colorectal cancer. Target Oncol. 2013;8:83–96. doi: 10.1007/s11523-013-0281-x. [DOI] [PubMed] [Google Scholar]
  • 43.de Mello RA, Madureira P, Carvalho LS, Araújo A, O’Brien M, et al. EGFR and KRAS mutations, and ALK fusions: current developments and personalized therapies for patients with advanced non-small-cell lung cancer. Pharmacogenomics. 2013;14:1765–1777. doi: 10.2217/pgs.13.177. [DOI] [PubMed] [Google Scholar]
  • 44.Grady WM, Pritchard CC. Molecular alterations and biomarkers in colorectal cancer. Toxicol Pathol. 2014;42:124–139. doi: 10.1177/0192623313505155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Parsons BL, Myers MB. Personalized cancer treatment and the myth of KRAS wild-type colon tumors. Discov Med. 2013;15:259–267. [PubMed] [Google Scholar]
  • 46.Miller JL. Progress in breast cancer treatment: prevention, new therapies come to forefront. Am J Health Syst Pharm. 1998;55:2326, 2328, 2330. doi: 10.1093/ajhp/55.22.2326. [DOI] [PubMed] [Google Scholar]
  • 47.Schinzari G, Cassano A, Orlandi A, Basso M, Barone C1. Targeted therapy in advanced gastric carcinoma: the future is beginning. Curr Med Chem. 2014;21:1026–1038. doi: 10.2174/0929867321666131129124054. [DOI] [PubMed] [Google Scholar]
  • 48.Almhanna K, Meredith KL, Hoffe SE, Shridhar R, Coppola D. Targeting the human epidermal growth factor receptor 2 in esophageal cancer. Cancer Control. 2013;20:111–116. doi: 10.1177/107327481302000204. [DOI] [PubMed] [Google Scholar]
  • 49.Verma S, Miles D, Gianni L, Krop IE, Welslau M, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367:1783–1791. doi: 10.1056/NEJMoa1209124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Jelovac D, Emens LA. HER2-directed therapy for metastatic breast cancer. Oncology (Williston Park) 2013;27:166–175. [PubMed] [Google Scholar]
  • 51.Drachman JG, Senter PD. Antibody-drug conjugates: the chemistry behind empowering antibodies to fight cancer. Hematology Am Soc Hematol Educ Program. 2013;2013:306–310. doi: 10.1182/asheducation-2013.1.306. [DOI] [PubMed] [Google Scholar]
  • 52.Traynor K. FDA approves pertuzumab for breast cancer. Am J Health Syst Pharm. 2012;69:1178. doi: 10.2146/news120049. [DOI] [PubMed] [Google Scholar]
  • 53.Gampenrieder SP, Rinnerthaler G, Greil R. Neoadjuvant Chemotherapy and Targeted Therapy in Breast Cancer: Past, Present, and Future. J Oncol. 2013;2013:732047. doi: 10.1155/2013/732047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Cho HS, Mason K, Ramyar KX, Stanley AM, Gabelli SB, et al. Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab. Nature. 2003;421:756–760. doi: 10.1038/nature01392. [DOI] [PubMed] [Google Scholar]
  • 55.Cai Z, Zhang G, Zhou Z, Bembas K, Drebin JA, et al. Differential binding patterns of monoclonal antibody 2C4 to the ErbB3-p185her2/neu and the EGFR-p185her2/neu complexes. Oncogene. 2008;27:3870–3874. doi: 10.1038/onc.2008.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Pinkas-Kramarski R, Soussan L, Waterman H, Levkowitz G, Alroy I, et al. Diversification of Neu differentiation factor and epidermal growth factor signaling by combinatorial receptor interactions. EMBO J. 1996;15:2452–2467. [PMC free article] [PubMed] [Google Scholar]
  • 57.Metzger-Filho O, Winer EP, Krop I. Pertuzumab: optimizing HER2 blockade. Clin Cancer Res. 2013;19:5552–5556. doi: 10.1158/1078-0432.CCR-13-0518. [DOI] [PubMed] [Google Scholar]
  • 58.Duneau JP, Vegh AP, Sturgis JN. A dimerization hierarchy in the transmembrane domains of the HER receptor family. Biochemistry. 2007;46:2010–2019. doi: 10.1021/bi061436f. [DOI] [PubMed] [Google Scholar]
  • 59.Barros FF, Powe DG, Ellis IO, Green AR. Understanding the HER family in breast cancer: interaction with ligands, dimerization and treatments. Histopathology. 2010;56:560–572. doi: 10.1111/j.1365-2559.2010.03494.x. [DOI] [PubMed] [Google Scholar]
  • 60.Badache A, Hynes NE. A new therapeutic antibody masks ErbB2 to its partners. Cancer Cell. 2004;5:299–301. doi: 10.1016/s1535-6108(04)00088-1. [DOI] [PubMed] [Google Scholar]
  • 61.Scheuer W, Friess T, Burtscher H, Bossenmaier B, Endl J, et al. Strongly enhanced antitumor activity of trastuzumab and pertuzumab combination treatment on HER2-positive human xenograft tumor models. Cancer Res. 2009;69:9330–9336. doi: 10.1158/0008-5472.CAN-08-4597. [DOI] [PubMed] [Google Scholar]
  • 62.Blumenthal GM, Scher NS, Cortazar P, Chattopadhyay S, Tang S, et al. First FDA approval of dual anti-HER2 regimen: pertuzumab in combination with trastuzumab and docetaxel for HER2-positive metastatic breast cancer. Clin Cancer Res. 2013;19:4911–4916. doi: 10.1158/1078-0432.CCR-13-1212. [DOI] [PubMed] [Google Scholar]
  • 63.Dalle S, Thieblemont C, Thomas L, Dumontet C. Monoclonal antibodies in clinical oncology. Anticancer Agents Med Chem. 2008;8:523–532. doi: 10.2174/187152008784533071. [DOI] [PubMed] [Google Scholar]
  • 64.Kute T, Stehle JR, Jr, Ornelles D, Walker N, Delbono O, et al. Understanding key assay parameters that affect measurements of trastuzumab-mediated ADCC against Her2 positive breast cancer cells. Oncoimmunology. 2012;1:810–821. doi: 10.4161/onci.20447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Karagiannis P, Singer J, Hunt J, Gan SK, Rudman SM, et al. Characterisation of an engineered trastuzumab IgE antibody and effector cell mechanisms targeting HER2/neu-positive tumour cells. Cancer Immunol Immunother. 2009;58:915–930. doi: 10.1007/s00262-008-0607-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Challacombe JM, Suhrbier A, Parsons PG, Jones B, Hampson P, et al. Neutrophils are a key component of the antitumor efficacy of topical chemotherapy with ingenol-3-angelate. J Immunol. 2006;177:8123–8132. doi: 10.4049/jimmunol.177.11.8123. [DOI] [PubMed] [Google Scholar]
  • 67.Collins SM, Bakan CE, Swartzel GD, Hofmeister CC, Efebera YA, et al. Elotuzumab directly enhances NK cell cytotoxicity against myeloma via CS1 ligation: evidence for augmented NK cell function complementing ADCC. Cancer Immunol Immunother. 2013;62:1841–1849. doi: 10.1007/s00262-013-1493-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Furlan SN, Mandraju R2, Brewer T2, Roybal K2, Troutman TD2, et al. Enhancement of anti-tumor CD8 immunity by IgG1-mediated targeting of Fc receptors. MAbs. 2014;6:108–118. doi: 10.4161/mabs.27052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Flacher V, Sparber F, Tripp CH, Romani N, Stoitzner P. Targeting of epidermal Langerhans cells with antigenic proteins: attempts to harness their properties for immunotherapy. Cancer Immunol Immunother. 2009:1137–1147. doi: 10.1007/s00262-008-0563-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Yang X, Zhang X, Mortenson ED, Radkevich-Brown O, Wang Y, et al. Cetuximab-mediated tumor regression depends on innate and adaptive immune responses. Mol Ther. 2013;21:91–100. doi: 10.1038/mt.2012.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Scott AM, Allison JP, Wolchok JD. Monoclonal antibodies in cancer therapy. Cancer Immun. 2012;12:14. [PMC free article] [PubMed] [Google Scholar]
  • 72.Agus DB, Gordon MS, Taylor C, Natale RB, Karlan B, et al. Phase I clinical study of pertuzumab, a novel HER dimerization inhibitor, in patients with advanced cancer. J Clin Oncol. 2005;23:2534–2543. doi: 10.1200/JCO.2005.03.184. [DOI] [PubMed] [Google Scholar]
  • 73.Saltz L, Easley C, Kirkpatrick P. Panitumumab. Nat Rev Drug Discov. 2006;5:987–988. doi: 10.1038/nrd2204. [DOI] [PubMed] [Google Scholar]
  • 74.Spiegelberg HL. Biological role of different antibody classes. Int Arch Allergy Appl Immunol. 1989;90(Suppl 1):22–27. doi: 10.1159/000235071. [DOI] [PubMed] [Google Scholar]
  • 75.Schroeder HW, Jr1, Cavacini L. Structure and function of immunoglobulins. J Allergy Clin Immunol. 2010;125:S41–52. doi: 10.1016/j.jaci.2009.09.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Racine R, Winslow GM. IgM in microbial infections: taken for granted? Immunol Lett. 2009;125:79–85. doi: 10.1016/j.imlet.2009.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Peng Y, Kowalewski R, Kim S, Elkon KB. The role of IgM antibodies in the recognition and clearance of apoptotic cells. Mol Immunol. 2005;42:781–787. doi: 10.1016/j.molimm.2004.07.045. [DOI] [PubMed] [Google Scholar]
  • 78.Haeckel R, Hänecke P. The application of saliva, sweat and tear fluid for diagnostic purposes. Ann Biol Clin (Paris) 1993;51:903–910. [PubMed] [Google Scholar]
  • 79.Furst DE. Serum immunoglobulins and risk of infection: how low can you go? Semin Arthritis Rheum. 2009;39:18–29. doi: 10.1016/j.semarthrit.2008.05.002. [DOI] [PubMed] [Google Scholar]
  • 80.Franklin RM. The ocular secretory immune system: a review. Curr Eye Res. 1989;8:599–606. doi: 10.3109/02713688908995760. [DOI] [PubMed] [Google Scholar]
  • 81.Berin MC. Mucosal antibodies in the regulation of tolerance and allergy to foods. Semin Immunopathol. 2012;34:633–642. doi: 10.1007/s00281-012-0325-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Iyengar SR, Walker WA. Immune factors in breast milk and the development of atopic disease. J Pediatr Gastroenterol Nutr. 2012;55:641–647. doi: 10.1097/MPG.0b013e3182617a9d. [DOI] [PubMed] [Google Scholar]
  • 83.Brandtzaeg P. The mucosal immune system and its integration with the mammary glands. J Pediatr. 2010;156:S8–15. doi: 10.1016/j.jpeds.2009.11.014. [DOI] [PubMed] [Google Scholar]
  • 84.ROWE DS, FAHEY JL. A new class of human immunoglobulins. Ii. Normal serum igd. J exp med. 1965;121:185–199. doi: 10.1084/jem.121.1.185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.ROWE DS, FAHEY JL. A new class of human immunoglobulins. I. A unique myeloma protein. J exp med. 1965;121:171–184. doi: 10.1084/jem.121.1.171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Preud’homme JL, Petit I, Barra A, Morel F, Lecron JC, et al. Structural and functional properties of membrane and secreted IgD. Mol Immunol. 2000;37:871–887. doi: 10.1016/s0161-5890(01)00006-2. [DOI] [PubMed] [Google Scholar]
  • 87.Chen K, Xu W, Wilson M, He B, Miller NW, et al. Immunoglobulin D enhances immune surveillance by activating antimicrobial, proinflammatory and B cell-stimulating programs in basophils. Nat Immunol. 2009;10:889–898. doi: 10.1038/ni.1748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ishizaka K, Ishizaka T, Hornbrook MM. Physico-chemical properties of human reaginic antibody. IV. Presence of a unique immunoglobulin as a carrier of reaginic activity. J Immunol. 1966;97:75–85. [PubMed] [Google Scholar]
  • 89.Johansson SG, Bennich H. Immunological studies of an atypical (myeloma) immunoglobulin. Immunology. 1967;13:381–394. [PMC free article] [PubMed] [Google Scholar]
  • 90.Johansson SG. Raised levels of a new immunoglobulin class (IgND) in asthma. Lancet. 1967;2:951–953. doi: 10.1016/s0140-6736(67)90792-1. [DOI] [PubMed] [Google Scholar]
  • 91.Stanworth DR. The discovery of IgE. Allergy. 1993;48:67–71. doi: 10.1111/j.1398-9995.1993.tb00687.x. [DOI] [PubMed] [Google Scholar]
  • 92.Bennich HH, Ishizaka K, Johansson SG, Rowe DS, Stanworth DR, Terry WD. Immunoglobulin E: a new class of human immunoglobulin. Immunology. 1968:323–324. [PMC free article] [PubMed] [Google Scholar]
  • 93.Butterworth AE. Cell-mediated damage to helminths. Adv Parasitol. 1984;23:143–235. doi: 10.1016/s0065-308x(08)60287-0. [DOI] [PubMed] [Google Scholar]
  • 94.Gould HJ, Sutton BJ. IgE in allergy and asthma today. Nat Rev Immunol. 2008;8:205–217. doi: 10.1038/nri2273. [DOI] [PubMed] [Google Scholar]
  • 95.Larché M, Akdis CA, Valenta R. Immunological mechanisms of allergen-specific immunotherapy. Nat Rev Immunol. 2006;6:761–771. doi: 10.1038/nri1934. [DOI] [PubMed] [Google Scholar]
  • 96.Jensen-Jarolim E, Achatz G, Turner MC, Karagiannis S, Legrand F, et al. AllergoOncology: the role of IgE-mediated allergy in cancer. Allergy. 2008;63:1255–1266. doi: 10.1111/j.1398-9995.2008.01768.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Jensen-Jarolim E, Pawelec G. The nascent field of AllergoOncology. Cancer Immunol Immunother. 2012;61:1355–1357. doi: 10.1007/s00262-012-1315-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Turner MC. Epidemiology: allergy history, IgE, and cancer. Cancer Immunol Immunother. 2012;61:1493–1510. doi: 10.1007/s00262-011-1180-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Turner MC, Chen Y, Krewski D, Ghadirian P, Thun MJ, et al. Cancer mortality among US men and women with asthma and hay fever. Am J Epidemiol. 2005;162:212–221. doi: 10.1093/aje/kwi193. [DOI] [PubMed] [Google Scholar]
  • 100.Turner MC1, Chen Y, Krewski D, Ghadirian P. An overview of the association between allergy and cancer. Int J Cancer. 2006;118:3124–3132. doi: 10.1002/ijc.21752. [DOI] [PubMed] [Google Scholar]
  • 101.Van Hemelrijck M, Garmo H, Binda E, Hayday A, Karagiannis SN, et al. Immunoglobulin E and cancer: a meta-analysis and a large Swedish cohort study. Cancer Causes Control. 2010;21:1657–1667. doi: 10.1007/s10552-010-9594-6. [DOI] [PubMed] [Google Scholar]
  • 102.Liu AH. Treatment of asthma with anti-immunoglobulin E monoclonal antibody. Allergy Asthma Proc. 2006;27:S24–28. [PubMed] [Google Scholar]
  • 103.Busse W, Buhl R, Fernandez Vidaurre C, Blogg M, Zhu J, et al. Omalizumab and the risk of malignancy: results from a pooled analysis. J Allergy Clin Immunol. 2012;129:983–989. doi: 10.1016/j.jaci.2012.01.033. [DOI] [PubMed] [Google Scholar]
  • 104.Fu SL, Pierre J, Smith-Norowitz TA, Hagler M, Bowne W, et al. Immunoglobulin E antibodies from pancreatic cancer patients mediate antibody-dependent cell-mediated cytotoxicity against pancreatic cancer cells. Clin Exp Immunol. 2008:401–409. doi: 10.1111/j.1365-2249.2008.03726.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Gould HJ, Mackay GA, Karagiannis SN, O’Toole CM, Marsh PJ, et al. Comparison of IgE and IgG antibody-dependent cytotoxicity in vitro and in a SCID mouse xenograft model of ovarian carcinoma. Eur J Immunol. 1999;29:3527–3537. doi: 10.1002/(SICI)1521-4141(199911)29:11<3527::AID-IMMU3527>3.0.CO;2-5. [DOI] [PubMed] [Google Scholar]
  • 106.O’Shannessy DJ, Somers EB, Smale R, Fu YS. Expression of folate receptor-α (FRA) in gynecologic malignancies and its relationship to the tumor type. Int J Gynecol Pathol. 2013;32:258–268. doi: 10.1097/PGP.0b013e3182774562. [DOI] [PubMed] [Google Scholar]
  • 107.Clifton GT, Sears AK, Clive KS, Holmes JP, Mittendorf EA, et al. Folate receptor α: a storied past and promising future in immunotherapy. Hum Vaccin. 2011;7:183–190. doi: 10.4161/hv.7.2.13784. [DOI] [PubMed] [Google Scholar]
  • 108.Karagiannis SN, Wang Q, East N, Burke F, Riffard S, et al. Activity of human monocytes in IgE antibody-dependent surveillance and killing of ovarian tumor cells. Eur J Immunol. 2003;33:1030–1040. doi: 10.1002/eji.200323185. [DOI] [PubMed] [Google Scholar]
  • 109.Fridman WH. Fc receptors and immunoglobulin binding factors. FASEB J. 1991;5:2684–2690. doi: 10.1096/fasebj.5.12.1916092. [DOI] [PubMed] [Google Scholar]
  • 110.St John AL, Abraham SN. Innate immunity and its regulation by mast cells. J Immunol. 2013;190:4458–4463. doi: 10.4049/jimmunol.1203420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Reilly RM, Sandhu J, Alvarez-Diez TM, Gallinger S, Kirsh J, et al. Problems of delivery of monoclonal antibodies. Pharmaceutical and pharmacokinetic solutions. Clin Pharmacokinet. 1995;28:126–142. doi: 10.2165/00003088-199528020-00004. [DOI] [PubMed] [Google Scholar]
  • 112.Jutel M, Akdis CA. Immunological mechanisms of allergen-specific immunotherapy. Allergy. 2011;66:725–732. doi: 10.1111/j.1398-9995.2011.02589.x. [DOI] [PubMed] [Google Scholar]
  • 113.Greenberger PA, Ditto AM. Chapter 24: Anaphylaxis. Allergy Asthma Proc. 2012;33(Suppl 1):S80–83. doi: 10.2500/aap.2012.33.3557. [DOI] [PubMed] [Google Scholar]
  • 114.Karagiannis SN, Josephs DH, Karagiannis P, Gilbert AE, Saul L, et al. Recombinant IgE antibodies for passive immunotherapy of solid tumours: from concept towards clinical application. Cancer Immunol Immunother. 2012;61:1547–1564. doi: 10.1007/s00262-011-1162-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Rudman SM, Josephs DH, Cambrook H, Karagiannis P, Gilbert AE, et al. Harnessing engineered antibodies of the IgE class to combat malignancy: initial assessment of FcvarepsilonRI-mediated basophil activation by a tumour-specific IgE antibody to evaluate the risk of type I hypersensitivity. Clin Exp Allergy. 2011:1400–1413. doi: 10.1111/j.1365-2222.2011.03770.x. [DOI] [PubMed] [Google Scholar]
  • 116.Theoharides TC, Conti P. Mast cells: the Jekyll and Hyde of tumor growth. Trends Immunol. 2004;25:235–241. doi: 10.1016/j.it.2004.02.013. [DOI] [PubMed] [Google Scholar]
  • 117.Legrand F, Driss V, Delbeke M, Loiseau S, Hermann E, et al. Human eosinophils exert TNF-α and granzyme A-mediated tumoricidal activity toward colon carcinoma cells. J Immunol. 2010;185:7443–7451. doi: 10.4049/jimmunol.1000446. [DOI] [PubMed] [Google Scholar]
  • 118.Blanchard C, Rothenberg ME. Biology of the eosinophil. Adv Immunol. 2009;101:81–121. doi: 10.1016/S0065-2776(08)01003-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Nimmerjahn F, Ravetch JV. Fcgamma receptors: old friends and new family members. Immunity. 2006;24:19–28. doi: 10.1016/j.immuni.2005.11.010. [DOI] [PubMed] [Google Scholar]
  • 120.Bruhns P, Iannascoli B, England P, Mancardi DA, Fernandez N, et al. Specificity and affinity of human Fcgamma receptors and their polymorphic variants for human IgG subclasses. Blood. 2009;113:3716–3725. doi: 10.1182/blood-2008-09-179754. [DOI] [PubMed] [Google Scholar]
  • 121.Karagiannis P, Gilbert AE, Josephs DH, Ali N, Dodev T, et al. IgG4 subclass antibodies impair antitumor immunity in melanoma. J Clin Invest. 2013;123:1457–1474. doi: 10.1172/JCI65579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Dombrowicz D, Lin S, Flamand V, Brini AT, Koller BH, et al. Allergy-associated FcRbeta is a molecular amplifier of IgE- and IgG-mediated in vivo responses. Immunity. 1998;8:517–529. doi: 10.1016/s1074-7613(00)80556-7. [DOI] [PubMed] [Google Scholar]
  • 123.Kinet JP. The high-affinity IgE receptor (Fc epsilon RI): from physiology to pathology. Annu Rev Immunol. 1999;17:931–972. doi: 10.1146/annurev.immunol.17.1.931. [DOI] [PubMed] [Google Scholar]
  • 124.Daniels TR, Martínez-Maza O, Penichet ML. Animal models for IgE-meditated cancer immunotherapy. Cancer Immunol Immunother. 2012;61:1535–1546. doi: 10.1007/s00262-011-1169-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Bradley OC. What is Comparative Medicine? Proc R Soc Med. 1927;21:129–134. [PMC free article] [PubMed] [Google Scholar]
  • 126.Jensen-Jarolim E. Definition of Comparative Medicine: History and New Identity. In: Jensen-Jarolim (Ed), editor. Comparative Medicine - Anatomy and Physiology. 2014. pp. 1–18. [Google Scholar]
  • 127.Reed LD. The important interface between public health and veterinary medicine for improving human health, animal health, and food safety. Public Health Rep. 2008;123:257. doi: 10.1177/003335490812300301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Rabozzi G, Bonizzi L, Crespi E, Somaruga C, Sokooti M, et al. Emerging zoonoses: the “one health approach”. Saf Health Work. 2012;3:77–83. doi: 10.5491/SHAW.2012.3.1.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Cruz-Coke R. [Hippocratic philosophy] Rev Med Chil. 1999;127:611–614. [PubMed] [Google Scholar]
  • 130.Somvanshi R. Veterinary medicine and animal keeping in ancient India. Asian Agri-History. 2006:133–146. [Google Scholar]
  • 131.Buell PD, May T, Ramey D. Greek and Chinese horse medicine: déjà vu all over again. Sudhoffs Arch. 2010;94:31–56. [PubMed] [Google Scholar]
  • 132.Smith KA. Edward jenner and the small pox vaccine. Front Immunol. 2011;2:21. doi: 10.3389/fimmu.2011.00021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Doherty M, Robertson MJ. Some early Trends in Immunology. Trends Immunol. 2004;25:623–631. doi: 10.1016/j.it.2004.10.008. [DOI] [PubMed] [Google Scholar]
  • 134.Sakula A. Robert koch: centenary of the discovery of the tubercle bacillus, 1882. Can Vet J. 1983;24:127–131. [PMC free article] [PubMed] [Google Scholar]
  • 135.Reuter G. [Veterinary medicine and preventive medicine] Berl Munch Tierarztl Wochenschr. 1997;110:431–435. [PubMed] [Google Scholar]
  • 136.Teigen PM. William osler and comparative medicine. Can Vet J. 1984;25:400–405. [PMC free article] [PubMed] [Google Scholar]
  • 137.Paoloni MC, Tandle A, Mazcko C, Hanna E, Kachala S, et al. Launching a novel preclinical infrastructure: comparative oncology trials consortium directed therapeutic targeting of TNFalpha to cancer vasculature. PLoS One. 2009:e4972. doi: 10.1371/journal.pone.0004972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Ranieri G, Gadaleta CD, Patruno R, Zizzo N, Daidone MG, et al. A model of study for human cancer: Spontaneous occurring tumors in dogs. Biological features and translation for new anticancer therapies. Crit Rev Oncol Hematol. 2013;88:187–197. doi: 10.1016/j.critrevonc.2013.03.005. [DOI] [PubMed] [Google Scholar]
  • 139.Simon R. Lost in translation: problems and pitfalls in translating laboratory observations to clinical utility. Eur J Cancer. 2008;44:2707–2713. doi: 10.1016/j.ejca.2008.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Gordon I, Paoloni M, Mazcko C, Khanna C. The Comparative Oncology Trials Consortium: using spontaneously occurring cancers in dogs to inform the cancer drug development pathway. PLoS Med. 2009;6:e1000161. doi: 10.1371/journal.pmed.1000161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kummar S, Rubinstein L, Kinders R, Parchment RE, Gutierrez ME, et al. Phase 0 clinical trials: conceptions and misconceptions. Cancer J. 2008;14:133–137. doi: 10.1097/PPO.0b013e318172d6f3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Franco NH, Olsson IA. Scientists and the 3Rs: attitudes to animal use in biomedical research and the effect of mandatory training in laboratory animal science. Lab Anim. 2014;48:50–60. doi: 10.1177/0023677213498717. [DOI] [PubMed] [Google Scholar]
  • 143.Hendriksen CF. Refinement, reduction, and replacement of animal use for regulatory testing: current best scientific practices for the evaluation of safety and potency of biologicals. ILAR J. 2002;43(Suppl):S43–48. doi: 10.1093/ilar.43.suppl_1.s43. [DOI] [PubMed] [Google Scholar]
  • 144.Hendriksen CF. Replacement, reduction and refinement alternatives to animal use in vaccine potency measurement. Expert Rev Vaccines. 2009;8:313–322. doi: 10.1586/14760584.8.3.313. [DOI] [PubMed] [Google Scholar]
  • 145.Centre for Cancer research Clinical Trials.
  • 146.Bavcar S1, Argyle DJ. Receptor tyrosine kinase inhibitors: molecularly targeted drugs for veterinary cancer therapy. Vet Comp Oncol. 2012;10:163–173. doi: 10.1111/j.1476-5829.2012.00342.x. [DOI] [PubMed] [Google Scholar]
  • 147.London CA, Malpas PB, Wood-Follis SL, Boucher JF, Rusk AW, et al. Multi-center, placebo-controlled, double-blind, randomized study of oral toceranib phosphate (SU11654), a receptor tyrosine kinase inhibitor, for the treatment of dogs with recurrent (either local or distant) mast cell tumor following surgical excision. Clin Cancer Res. 2009:3856–3865. doi: 10.1158/1078-0432.CCR-08-1860. [DOI] [PubMed] [Google Scholar]
  • 148.Bernabe LF, Portela R, Nguyen S, Kisseberth WC, Pennell M, et al. Evaluation of the adverse event profile and pharmacodynamics of toceranib phosphate administered to dogs with solid tumors at doses below the maximum tolerated dose. BMC Vet Res. 2013;9:190. doi: 10.1186/1746-6148-9-190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Bareck E, Ba-Ssalamah A, Brodowicz T, Eisterer W, Häfner M, et al. Gastrointestinal stromal tumors: diagnosis, therapy and follow-up care in Austria. Wien Med Wochenschr. 2013;163:137–152. doi: 10.1007/s10354-013-0187-3. [DOI] [PubMed] [Google Scholar]
  • 150.Figlin R, Sternberg C, Wood CG. Novel agents and approaches for advanced renal cell carcinoma. J Urol. 2012;188:707–715. doi: 10.1016/j.juro.2012.04.108. [DOI] [PubMed] [Google Scholar]
  • 151.Brazís P, De Mora F, Ferrer L, Puigdemont A. IgE enhances Fc epsilon RI expression and IgE-dependent TNF-alpha release from canine skin mast cells. Vet Immunol Immunopathol. 2002;85:205–212. doi: 10.1016/s0165-2427(01)00428-7. [DOI] [PubMed] [Google Scholar]
  • 152.Bonkobara M, Miyake F, Yagihara H, Yamada O, Azakami D, et al. Canine epidermal langerhans cells express alpha and gamma but not beta chains of high-affinity IgE receptor. Vet Res Commun. 2005;29:499–505. doi: 10.1007/s11259-005-2494-7. [DOI] [PubMed] [Google Scholar]
  • 153.Nuttall T, Uri M, Halliwell R. Canine atopic dermatitis - what have we learned? Vet Rec. 2013;172:201–207. doi: 10.1136/vr.f1134. [DOI] [PubMed] [Google Scholar]
  • 154.Gaschen FP, Merchant SR. Adverse food reactions in dogs and cats. Vet Clin North Am Small Anim Pract. 2011;41:361–379. doi: 10.1016/j.cvsm.2011.02.005. [DOI] [PubMed] [Google Scholar]
  • 155.Singer J, Weichselbaumer M, Stockner T, Mechtcheriakova D, Sobanov Y, et al. Comparative oncology: ErbB-1 and ErbB-2 homologues in canine cancer are susceptible to cetuximab and trastuzumab targeting. Mol Immunol. 2012:200–209. doi: 10.1016/j.molimm.2012.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Singer J, Fazekas J, Wang W, Weichselbaumer M, Matz M, et al. Generation of a canine anti-EGFR (ErbB-1) antibody for passive immunotherapy in dog cancer patients. Mol Cancer Ther. 2014 doi: 10.1158/1535-7163.MCT-13-0288. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES