Abstract
Drugs targeting the immune system such as corticosteroids, antihistamines and immunosuppressants have been widely exploited in the treatment of inflammatory, allergic and autoimmune disorders during the second half of the 20th century. The recent advances in immunopharmacological research have made available new classes of clinically relevant drugs. These comprise protein kinase inhibitors and biologics, such as monoclonal antibodies, that selectively modulate the immune response not only in cancer and autoimmunity but also in a number of other human pathologies. Likewise, more effective vaccines utilizing novel antigens and adjuvants are valuable tools for the prevention of transmissible infectious diseases and for allergen-specific immunotherapy. Consequently, immunopharmacology is presently considered as one of the expanding fields of pharmacology. Immunopharmacology addresses the selective regulation of immune responses and aims to uncover and exploit beneficial therapeutic options for typical and non-typical immune system-driven unmet clinical needs. While in the near future a number of new agents will be introduced, improving the effectiveness and safety of those currently in use is imperative for all researchers and clinicians working in the fields of immunology, pharmacology and drug discovery. The newly formed ImmuPhar (http://iuphar.us/index.php/sections-subcoms/immunopharmacology) is the Immunopharmacology Section of the International Union of Basic and Clinical Pharmacology (IUPHAR, http://iuphar.us/). ImmuPhar provides a unique international expert-lead platform that aims to dissect and promote the growing understanding of immune (patho)physiology. Moreover, it challenges the identification and validation of drug targets and lead candidates for the treatment of many forms of debilitating disorders, including, among others, cancer, allergies, autoimmune and metabolic diseases.
Tables of Links
| TARGETS | ||
|---|---|---|
| GPCRsa | Catalytic receptorsc | Enzymesd |
| Chemokine receptors | ALK | Abl |
| H1 receptor | CD52 | Akt (PKB) |
| H2 receptor | CTLA4 (CD152) | Cytochrome P450 |
| H4 receptor | EGFR | IRAK4 |
| Nuclear hormone receptorsb | Ephrin receptor family (EPH) | Janus kinase family (JAK) |
| Retinoic acid receptor-related orphan receptors (ROR) | FGFR1 | MEK1 |
| FLT3 | mTOR | |
| HER2 | PI3K | |
| HGFR | RAF | |
| IL6R | Src | |
| KIT (c-Kit) | Syk | |
| MET | ||
| NOD-like receptor family (NLR) | ||
| Pattern recognition receptor family (PRR) | ||
| PDGFRα | ||
| PDGFRβ | ||
| RET | ||
| Toll-like receptor family (TLR) | ||
| TrkB | ||
| VEGFR-1 | ||
| VEGFR-2 | ||
| VEGFR-3 |
| LIGANDS | |||
|---|---|---|---|
| Abatacept | Etanercept | Lapatinib | SCF |
| Adalimumab | Certolizumab pegol | Nilotinib | Secukinumab |
| Alemtuzumab | Gefitinib | Nintedanib | Sorafenib |
| Bosutinib | Golimumab | Ocrelizumab | Sunitinib |
| Brodalumab | Histamine | Ofatumumab | Tocilizumab |
| Cabozantinib | IFN | Omalizumab | Trametinib |
| Crizotinib | IL | PDGF | Tumor necrosis factor |
| Dabrafenib | Imatinib | Rituximab | Vandetanib |
| Dasatinib | Infliximab | Ruxolitinib | |
| Erlotinib | Ixekizumab | Sarilumab |
These tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14
Alexander et al., 2013a,b,c,d,,,).
Introduction
For more than 50 years, drugs targeting immune cell pathways and receptors have been extensively exploited in the treatment of inflammatory, allergic and autoimmune disorders, and in preventing rejection following organ transplantation. Among them, many non-steroidal anti-inflammatory drugs, antihistamines, corticosteroids and immunosuppressant agents (Figure 1) have reached blockbuster status and are even included in the list of essential medicines of the World Health Organization (WHO, 2013).
Figure 1.

Common clinically relevant drugs used in the treatment of human inflammatory, allergic and other immune system-associated disorders.
In recent years, several notable changes in our understanding and appreciation of the immune system, greater knowledge of the activity of agents that modify the immune responses and the significant biotechnological advances have made available new classes of drugs. For instance, PK inhibitors (PKIs) and biologics, such as monoclonal antibodies (mAbs) (Figure 1) are capable of selectively modulating immune cell subsets (Dollery, 2014). At the same time, there has been growing evidence connecting the majority of human pathologies to dysfunctions of the innate and adaptive immune systems (Figure 2). Thus, scientists and clinicians working in universities and industry have shown enormous interest in the interrelationship between the disciplines of pharmacology and immunology, including immunotoxicology and immunogenetics (Cohen, 2006). Despite the use of vaccines and immunomodulating agents in clinical practice for many years (Figure 1), immunopharmacology is presently considered as one of the youngest fields of pharmacology. Immunopharmacology addresses the selective up- or down-regulation of immune responses. It aims to uncover and exploit more effective and safer therapeutic options for unmet clinical needs for an expanding range of pathologies, such as cancer and inflammatory, infectious, immune and metabolic diseases (Figure 2).
Figure 2.

Examples of human pathologies linked to inflammation and to dysfunctions of the immune system.
The importance of this area of pharmacology is evidenced by the recently launched ImmuPhar (Figure 3), the Immunopharmacology Section of the International Union of Basic and Clinical Pharmacology (IUPHAR Immunopharmacology Section, 2015). The main objective of ImmuPhar is to encourage the international cooperation and dissemination of knowledge in immunopharmacology. The activities are organized by the Executive Committee, the International Advisory Board, and the subcommittees on ‘molecular targets for immunomodulatory drugs’ (molecular oriented), ‘targets in immune-related diseases’ (disease oriented) and ‘antibodies as therapeutics’.
Figure 3.

Logo of ImmuPhar, the Immunopharmacology Section of the International Union of Basic and Clinical Pharmacology (IUPHAR). ImmuPhar aims to promote the international cooperation and knowledge dissemination in the growing field of immunopharmacology.
The objectives of ImmuPhar will be achieved by (i) stimulating worldwide research in basic and clinical immunopharmacology; (ii) promoting high scientific and ethical standards in research into related medicines and therapeutics; (iii) encouraging related scientific meetings, workshops and courses in different parts of the world; (iv) improving and harmonizing the teaching of immunopharmacology; (v) supporting the utilization of immunopharmacological agents in health care delivery, particularly in developing countries; (vi) evaluating patients experiencing adverse drug reactions by utilizing clinical immunopharmacology skills; (vii) encouraging collaboration with other agencies and organizations interested in the study, development and rational use of immunopharmacological agents; (viii) exchanging and disseminating information on the safety and pharmacovigilance of related medicines and therapeutics; and (ix) fostering cooperative efforts among educational, research, clinical, industrial and governmental personnel engaged in activities relevant to translational research in immunopharmacology. Membership of the Section is open to pharmacologists, immunopharmacologists, clinical pharmacologists, pathologists, immunologists and clinicians interested in the interrelationships between pharmacology and immunology. ImmuPhar works in close collaboration with the IUPHAR Committee on Receptor Nomenclature and Drug Classification (IUPHAR/BPS Guide to PHARMACOLOGY2015; http://www.guidetopharmacology.org/). Any IUPHAR member societies and their sections are also eligible for affiliation.
This review aims to summarize the new concepts on the role of immunopharmacology in the ongoing innovations in immunomodulatory drug development, from small molecules to vaccines and other biological modifiers. Moreover, due to the increasing number of PKIs and mAbs that enter the clinic, the challenge of both academic and industrial audiences is to consider the complex pharmacological profile of these novel options during drug development, without excluding the important advances in the pharmacology of classical therapeutic approaches.
Small molecules
Despite the emergence and the clinical success of biologics, several limitations hamper the therapeutic manipulation of the inflammatory networks underlying the multifaceted aetiology of many immune disorders. For instance, agents produced by means of biological processes frequently involving recombinant DNA technology are expensive. More importantly, they lack oral availability and often show inefficient delivery to target tissues in vivo (Kopf et al., 2010). By controlling the signalling pathways involved in tissue-specific inflammation, small molecules remain an effective approach for immunomodulatory drug development and repurposing (Thomson et al., 2009; Sundberg et al., 2014). Related emerging data confer new properties to old medications, as is the case with glucocorticoids or the immunosuppressive drug rapamycin. In addition to the potent inhibition of growth factor-induced T-cell proliferation, the serine/threonine PK mammalian target of rapamycin (mTOR) has been reported to play an important role in the regulation of diverse functions of various immune cells (Thomson et al., 2009). Another example is the recently recognized rapid onset and short duration of the non-genomic glucocorticoid actions. These new discoveries should help in facilitating the development of new improved strategies for the management of inflammatory and autoimmune diseases (Alangari, 2010; Simon et al., 2013).
Furthermore, the latest advances in mast cell-derived mediator research, including histamine (Zampeli and Tiligada, 2009; Tiligada, 2012) and prostaglandins (Woodward et al., 2011), are illustrative examples of the existing challenge to identify and validate new targets and to optimize lead candidates for asthma and allergies (Schumacher et al., 2014; Chliva et al., 2015). In particular, histamine interacts with four types of GPCRs, designated as H1–H4, and it is a major component of the immune system playing a critical role in inflammation (Parsons and Ganellin, 2006). For more than 70 years, histamine has been one of the most exploited substances in medicine, providing blockbuster drugs acting on H1 and H2 receptors for the treatment of allergies and gastric ulcers respectively (Parsons and Ganellin, 2006). Yet, the continuing appreciation of the pharmacodynamic and pharmacokinetic diversity of H1 antihistamines reflects the ongoing efforts to translate preclinical drug actions into promising therapies for pathologies with a high economic and societal impact (del Cuvillo et al., 2015; Schumacher et al., 2014). Interestingly, the discovery of the high affinity histamine H4 receptor in 2000 and its constitutive activity and expression mostly on cells of the immune system (Figure 4) revealed new pathways in the extensive biological functions of histamine. Besides its role in allergy, the translational potential of this new drug target in acute and chronic inflammation, host defence and neuropathic pain provides attractive novel perspectives (Tiligada et al., 2009; Zampeli and Tiligada, 2009; Tiligada, 2012; Kyriakidis et al., 2015).
Figure 4.

The histamine H4 receptor is expressed in various cell types and mediates a variety of distinct effects depending on the endogenous complement of receptor expression and signal transduction pathways upon binding of histamine, unbiased or biased ligands.
The rapid entry of H4 receptor-targeting compounds into advanced clinical development will benefit patients with poorly treatable chronic diseases. Moreover, the pluridimensional rather than linear pharmacological efficacy of H4 receptor ligands (Figure 4) represents a paradigm of the recently described concept of ‘biased agonism’ or functional selectivity for GPCRs (Nijmeijer et al., 2013). GPCRs account for more than 65% of the medicines marketed today, highlighting their relevance in human (patho)physiology including immune responses (Rask-Andersen et al., 2011). By realizing the distinct functional outcomes of GPCR-mediated activation of complex signalling networks upon agonist binding, biased ligands represent an opportunity for the discovery of new drugs with specific on-target efficacy and fewer on-target side effects (Kenakin and Christopoulos, 2013). Taken together, the advances in these fields of research suggest that the differential expression and/or the selective modulation of receptor activity can alter pro- and anti-inflammatory signals orchestrating acute and chronic inflammation reflected by the repertoire of immune cells and mediators (Zampeli and Tiligada, 2009; Tiligada, 2012; Nijmeijer et al., 2013; Corbisier et al., 2015).
PK inhibitors
The family of PKs includes two major subfamilies: the serine/threonine kinases and the TKs. PKs are components of signal transduction pathways involved in diverse biological processes. They are now linked either directly or indirectly to more than 400 human diseases ranging from cancer to inflammatory, metabolic and cardiovascular disorders (Steinman et al., 2012; Galluzzi et al., 2014; Fabbro, 2015; Fabbro et al., 2015). There are more than 500 kinases in the human genome and as 30% of the proteome is phosphorylated, the modulators will have a vast pharmacology. Thus, PKs constitute multiple targets for anticancer treatments and potentially for the modulation of inflammation and immunity if safety can be assured (Galluzzi et al., 2014; Marfe and Di Stefano, 2014).
PKIs are usually small, cell-permeant molecules, which bind to the ATP-binding region of receptor and non-receptor kinases (Table 2011). There are currently 39 marketed drugs acting on kinases and more than 130 in phase II/III ongoing clinical trials since the approval of imatinib in 2001 (Nagar et al., 2002; Fabbro et al., 2015). PKIs have potentiated and sometimes replaced therapy with mAbs or vice versa. Whereas kinase inhibitors are validated in certain types of cancer (Table 2011), the situation is far from clear in autoimmune and other inflammatory diseases (Table 2010). PKIs are designed to have a single or limited number of primary targets. However, most of them might interact with more than one PK and exhibit significant cross-reactivity (Table 2011). In fact, among the drugs approved for clinical use, only a few, including lapatinib and imatinib, are highly selective; the majority inhibit more than 10 and up to more than 100 kinases. The non-specificity of the target together with new and still unknown molecular mechanisms may be responsible for unexpected off-target mechanisms and side effects, including drug resistance (Davies et al., 2000; Ubersax and Ferrell, 2007; Loriot et al., 2008; Chen and Fu, 2011).
Table 1.
Examples of PKIs and cancer therapy
| Drug | Molecular target | Tumour |
|---|---|---|
| Imatinib | PDGFR, PDGF, SCF, c-Kit, Bcr-Abl | Chronic myeloid leukaemia, gastrointestinal stromal tumours |
| Gefitinib | EGFR | Metastatic non-small cell lung cancer |
| Erlotinib | HER2, EGFR | Metastatic non-small cell lung cancer |
| Sorafenib | VEGFR-2,VEGFR-3, PDGFRβ, c-Kit, Fit-3 | Renal cell carcinoma |
| Sunitinib | PDGFR, VEGFR, c-Kit, Fit-3 | Renal cancer, gastrointestinal stromal tumours |
| Dasatinib | Bcr-Abl, Src, c-Kit, EPH, PDGFRβ | Imatinib-resistant chronic myeloid leukaemia |
| Nilotinib | PDGFR, c-Kit, Bcr-Abl | Chronic myeloid leukaemia |
| Lapatinib | HER2, EGFR | Breast cancer |
| Crizotinib | ALK, HGFR | ALK-positive lung cancer |
| Ruxolitinib | JAK | Myelofibrosis |
| Vandetanib | RET, VEGFR2, EGFR | Thyroid cancer |
| Cabozantinib | RET, MET, VEGFR, c-Kit, TrkB | Thyroid cancer |
| Bosutinib | Bcr-Abl, Src | Chronic myeloid leukaemia |
| Dabrafenib | Raf | Melanoma |
| Trametinib | MEK | Metastatic cutaneous melanoma |
| Nintedanib | VEGFR, FGFR, PDGFR | Idiopathic pulmonary fibrosis |
Abl, Abelson kinase; Bcr, breakpoint cluster region; c-Kit, mast/stem cell growth factor receptor; Raf, rapidly accelerated fibrosarcoma.
Table 2.
Targets that need to be validated for immune and inflammatory diseases
| Target, inhibitors | For which diseases? |
|---|---|
|
|
PKI specificity has been tested in vitro with binding affinity and activity inhibition tests have also detected allosteric binding and modulation of TK activity. In vitro assays have a number of limitations due, for example, to lack of post-translational target modifications or of other regulatory proteins and non-kinase targets that are often responsible for important off-target effects (see Fabbro et al., 2015). Recent progress in quantitative proteomics has allowed a more impartial interpretation of PKI specificity and provided models for PKI–PK interaction in the biological context. Furthermore, pharmacokinetic characteristics and the possible interaction with drugs modulating their metabolism through cytochrome P450 isoenzymes are as important as the pharmacodynamic parameters for the potential utility of PKIs (van Leeuwen et al., 2014).
The choice of therapeutic target (Table 2010) is certainly a critical issue. The broad spectrum of PKI target interactions and the off-target effects are important not only to better understand the actual mechanism of action and the molecular basis of adverse drug reactions, but also to define ‘secondary’ therapeutic approaches that will permit the use of those drugs in other diseases, in the future. Testing a novel concept in this field is extremely expensive and the idea of pan modulators working in multiple disorders is clearly incorrect. Indeed, Steinman et al. (2012) have powerfully argued that different strategies are needed for different diseases. TNF antagonists are active in rheumatoid arthritis (RA), and type 1 interferon modulators inactive, whereas in multiple sclerosis the converse is true and anti-CD20 therapies work in both (Steinman et al., 2012). Because of the ability of PKIs to bind TKs in the active or inactive conformation, sometimes they may activate rather than inhibit kinases (Moebitz and Fabbro, 2012). The block in active conformation can explain in part the effect of some drugs that stabilize the phosphorylation state. However, in some cases, following drug binding, the kinase is activated. This paradoxical effect, which may be related to kinase interacting with molecules that affect the conformational state of the kinase, can be part of the drug’s action or even constitute a mechanism of resistance (Chen and Fu, 2011; Marfe and Di Stefano, 2014; Fabbro et al., 2015).
Although PKIs share the same mechanism of action, competing with ATP for the catalytic site of the enzyme, major acute and chronic side effects involving different organs limit their clinical use and have resulted in clinical trials being suspended and drugs being withdrawn (Loriot et al., 2008). Thus, there is a real need for an expert-lead initiative to help drug discovery and development. IUPHAR has developed a database of all the kinases, with their main pharmacology (IUPHAR/BPS Guide to PHARMACOLOGY2015; http://www.guidetopharmacology.org/), and will be leading a major initiative on their role in immunopharmacology.
Monoclonal antibody therapies
Advances in basic immunology have contributed to the identification of various critical molecules involved in several immune reactions and their respective pathophysiological roles in a variety of immunological diseases. One of the most important key technical advances for promoting immunology research is the establishment of mAbs, led by the Nobel Prize laureates, Milstein and Köhler (Köhler and Milstein, 1975). Interestingly, the generation of mAbs recognizing various specific targets, such as cell surface molecules and cytokines, accompanied by flow cytometrical methodology, has enabled us to respectively distinguish an increasing number of cellular subsets. This in turn has resulted in the recent explosive progress of the immunology research field in identifying new potential drug targets (Thomas, 1989).
mAbs that neutralize and inactivate target molecules/cells have been utilized for a while for treating several human diseases (Beck et al., 2010; Chan and Carter, 2010). For example, the initial trial was done using rituximab, an anti-CD20 mAb, to treat B-cell lymphoma by depleting CD20-expressing B-lineage cells (Reff et al., 1994). In the case of rheumatic diseases, a number of mAbs targeting TNF are now frequently used for treating RA, such as infliximab, adalimumab and golimumab (Breedveld, 2000; Feldmann and Maini, 2001). The development and clinical application of mAbs, so-called ‘biological agents’, have undoubtedly caused a paradigm shift in the therapeutics of RA. Besides TNF, several targets have been utilized to date, such as IL 6 and its receptor (tocilizumab), cytotoxic T-lymphocyte antigen 4 (CTLA4), and so on. In addition to RA, several immunological disorders are now targeted by mAb therapies. These include multiple sclerosis (treated with alemtuzumab anti-CD52), inflammatory bowel diseases (anti-TNF mAbs), psoriasis (anti-IL17 mAbs) and asthma (omalizumab anti-IgE) (Scalapino and Daikh, 2008; Pelaia et al., 2012; Tanaka et al., 2012). Some representative examples are illustrated in Table 2013a.
Table 3.
Representative examples of therapeutic monoclonal antibodies
| Molecule | Name | Type | Disease target |
|---|---|---|---|
| TNF | Infliximab | Chimera | RA, Crohn’s diseases, Behçet’s disease |
| Adalimumab | Human | ||
| Golimumab | Human | ||
| Etanercept | TNFR-Ig | ||
| Certolizumab pegol | Fab-pegosyl | ||
| CD20 | Rituximab | Chimera | B-cell lymphoma (vasculitis) |
| Ocrelizumab | Humanized | ||
| Ofatumumab | Human | ||
| IL6R | Tocilizumab | Humanized | RA |
| Sarilumab | Human | ||
| CTLA4 | Abatacept | CTLA4-Ig | RA |
| IL17 | Secukinumab | Human | Psoriasis |
| Ixekizumab | Humanized | ||
| Brodalumab | Human | ||
| CD52 | Alemtuzumab | Humanized | Multiple sclerosis |
| IgE | Omalizumab | Humanized | Severe asthma, chronic urticaria |
CTLA, cytotoxic T-lymphocyte-associated protein.
One of the most significant advantages of mAb therapies is the high specificity for their targets, which would minimize off-target adverse effects. It is really surprising that depletion or inactivation of a single molecule by a mAb alters the cytokine cascade and blocks inflammatory responses in certain conditions. However, one should not disregard the challenges that therapeutic mAb therapy is raising, such as their immunogenicity, delivery only through injection and the usually extremely long half-life. Nevertheless, recent bioengineering technology has enabled us to develop less immunogenic mAbs, such as ‘chimera’ (having murine variable regions), ‘humanized’ (with murine complementary determining regions) or complete ‘human’ therapeutic mAbs. In addition to conventional mAbs, the pegylated Fab portion of IgG, for example, against TNF (certolizumab pegol) and a fusion protein of IgG Fc region with several targets, such as the extracellular domain of CTLA4 (abatacept) or TNF receptor (etanercept), also have the potential to be used clinically. These and possibly other developments will facilitate the creation of mAbs with fewer side-effects and perhaps even patient-specific drugs (Breedveld, 2000; Beck et al., 2010).
Vaccines and adjuvants
Vaccines have had an enormous impact on human health and are probably the most important tool for preventing transmissible infectious diseases. There have been many new developments in the search for vaccines. These include novel techniques for the attenuation or inactivation of microorganisms, new ways of delivering antigens, such as the use of viruses or liposomes, emulsions and low size particles and, more recently, novel adjuvants with known mechanisms of action (Leroux-Roels, 2010; Gebril et al., 2012).
The discovery of pattern recognition receptors (PRRs) and their role in innate immunity to identify pathogen or microbial-associated molecular patterns (PAMPs or MAMPs, respectively) have literally boosted interest in the field of vaccinology (Song and Lee, 2012). In fact, an expert subcommittee recently published new propositions for PRR nomenclature (Bryant et al., 2015). PAMPs, including LPS and unmethylated motifs of bacterial DNA (CpG), can activate toll-like receptors (TLRs) on the surface or in the cytosolic compartments of antigen-presenting cells (APCs) (Song and Lee, 2012). Aluminium salts are the most commonly used adjuvants in clinical practice and activate another class of PRRs, namely the nucleotide-binding oligomerization domain (NOD)-like receptors (NLR), thus leading to the activation of APCs (Eisenbarth et al., 2008). APC activation increases cytokine expression, antigen presentation and other events that cause maturation of APCs. Mature APCs modulate the activation of T- and B-cells and the commitment of CD4+ T-cells to the various subsets of Th and to regulatory (Treg) cells (Zhu et al., 2010). It is now clear that activation of different PRRs may favour preferentially specific Th subsets and various aspects of the immune response, hence favouring better protective responses against microorganisms (Medzhitov, 2007). The adjuvant component of vaccines also appears to contribute to the local and systemic side effects of vaccination (Eisenbarth et al., 2008). A list of adjuvants available for human use is shown in Table 2013b.
Table 4.
Adjuvants licensed for human prophylactic vaccines
| Adjuvants | Mechanism of action | Antigens (in vaccines) | Manufacturer | |
|---|---|---|---|---|
| DS | IS | |||
| Aluminium salts | √ | Th2 | DPT, HBV, IPV, HiB, etc. | Several |
| Emulsion o/w MF59 | √ | Th1 | H1N1 (Fluad™) | Novartis Vaccines and Diagnostics, Cambridge, MA, USA |
| H5N1 (Focetria™) | ||||
| Emulsion o/w AS03 | √ | Humoral (Th?) | H5N1 (Prepandrix™) | GSK Biologicals, Rixensart, Belgium |
| H1N1 (Arepanrix™) | ||||
| Emulsion o/w AF03 | √ | Humoral (Th?) | H1N1 (Humenza™) | Sanofi Pasteur, Lyon, France |
| MPL + Alum (AS04) | √ | Th2/Th1 | HBV (Fendrix™) | GSK Biologicals, Rixensart, Belgium |
| HPV (Cervarix™) | ||||
| RC529 + Alum | √ | Th1 | HBV (Supervax™) | Dynavax, Berkeley, CA, USA |
| Virosomes | √ | Th2/Th1 | H5N1 (Inflexal™) | Crucell, Leiden, The Netherlands |
| HAV (Epaxal™) | ||||
| CTB* | – | IgA? | Vibrio cholerae (Dukoral™) | SBL Vaccines, Stockholm, Sweden |
The only mucosal adjuvant licensed in oral cholera vaccine; CTB, cholera toxin subunit B; DPT, diphtheria, pertussis and tetanus; DS, delivery system; GSK, Glaxo Smith Kline; HAV, hepatitis A virus; HBV, hepatitis B virus; HPV, human papilloma virus; HxNx, influenza virus; IPV, inactivated polio virus; IS, preferentially activated immune response.
In the context of allergy, there are now vaccines developed from a number of allergens that are used for allergen-specific immunotherapy (Akkoc et al., 2011). Recently, a detoxified derivative of LPS from Salmonella minnesota, referred to as monophosphoryl lipid A (MPL), has been used in adjuvanted pollen allergy vaccines (Patel and Salapatek, 2006). MPL is an adjuvant that induces a Th1-skewed immune response through binding to the TLR4 on APCs. The subsequent stimulation of cytokine secretion, such as IL-12, favours the reduction of IgE and the induction of protective IgG antibodies in allergic individuals (Mothes et al., 2003). Pollinex Quattro is a vaccine for the treatment of pollen allergies that is enhanced with MPL (Rosewich et al., 2013). Therefore, the search for novel adjuvants with a known molecular mechanism of action, greater efficacy and safety profile is an area of particular interest to immunopharmacologists.
Summary and conclusions
In addition to the classical therapeutic approaches, the novel immunopharmacological concepts and tools and their relevance to human disease offer new options for unmet medical needs including, among others, cancer, inflammatory, autoimmune, metabolic and infectious diseases. The recent developments in immunology and pharmacology emphasize the necessity not only to exploit new classes of drugs, such as cytokines, PKIs and mAbs, but also to improve those that are already in use. The optimal translation of experimental data (Siebenhaar et al., 2015), the characterization of the links between genetic, epigenetic and non-genetic factors (Almouzni et al., 2014), and the application of the ‘omic’ technologies are likely to identify novel disease pathways and to repurpose a number of therapeutics (Holgate, 2013). While in the near future a number of new agents will be introduced, the common challenge for all researchers and clinicians working in the fields of immunology, pharmacology and drug development is to improve the efficacy and safety of the diverse classes of drugs discussed herein. The newly formed ImmuPhar is the Immunopharmacology Section of the IUPHAR that provides a unique international expert-lead platform to dissect and promote the growing knowledge and understanding of immune (patho)physiology and its exploitable modification by a variety of medicines.
Acknowledgments
The authors thank Dr Doriano Fabbro (PIQUR Therapeutics AG, Basel, Switzerland) and Dr Reinaldo Acevedo (Finlay Institute, Cuba) for their valuable contributions in the preparation of this review. NC-IUPHAR receives financial support from the Wellcome Trust.
Glossary
- Abl
Abelson kinase
- ALK
anaplastic lymphoma kinase
- APC
antigen-presenting cell
- CD
cluster of differentiation
- c-Kit
stem cell factor receptor
- CpG
unmethylated motifs of bacterial DNA
- CTLA
cytotoxic T-lymphocyte antigen
- EPH
ephrin kinase
- Fab
fragment antigen-binding
- HER
human epidermal growth factor receptor
- HGFR
hepatocyte growth factor receptor
- HiB
Haemofilus influenzae B antigen
- IRAK
IL-1 receptor-associated kinase
- mAbs
monoclonal antibodies
- MAMP
microbial-associated molecular pattern
- MEK
mitogen activated kinase kinase
- MET
mesenchymal epithelial transition factor or hepatocyte growth or scatter factor receptor
- MPL
monophosphoryl lipid
- mTOR
mammalian target of rapamycin
- PAMP
pathogen-associated molecular pattern
- PKI
PK inhibitor
- PRR
pattern recognition receptor
- RA
rheumatoid arthritis
- RET
receptor for GDNF-family ligands
- ROR
retinoic acid receptor-related orphan receptor
- SCF
stem cell factor
- Src
proto-oncogene tyrosine-PK Src
- Syk
spleen TK
- TLR
toll-like receptor
- Treg
regulatory T-cells
- TrkB
tropomyosin receptor kinase B
Conflict of interest
None.
References
- Akkoc T, Akdis M, Akdis CA. Update in the mechanisms of allergen-specific immunotherapy. Allergy Asthma Immunol Res. 2011;3:11–20. doi: 10.4168/aair.2011.3.1.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alangari AA. Genomic and non-genomic actions of glucocorticoids in asthma. Ann Thorac Med. 2010;5:133–139. doi: 10.4103/1817-1737.65040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The concise guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol. 2013a;170:1459–1581. doi: 10.1111/bph.12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The concise guide to PHARMACOLOGY 2013/14: nuclear hormone receptors. Br J Pharmacol. 2013b;170:1652–1675. doi: 10.1111/bph.12448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The concise guide to PHARMACOLOGY 2013/14: catalytic receptors. Br J Pharmacol. 2013c;170:1676–1705. doi: 10.1111/bph.12449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The concise guide to PHARMACOLOGY 2013/14: enzymes. Br J Pharmacol. 2013d;170:1797–1867. doi: 10.1111/bph.12451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Almouzni G, Altucci L, Amati B, Ashley N, Baulcombe D, Beaujean N, et al. Relationship between genome and epigenome – challenges and requirements for future research. BMC Genomics. 2014;15:487. doi: 10.1186/1471-2164-15-487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beck A, Wurch T, Bailly C, Corvaia N. Strategies and challenges for the next generation of therapeutic antibodies. Nat Rev Immunol. 2010;10:345–352. doi: 10.1038/nri2747. [DOI] [PubMed] [Google Scholar]
- Breedveld FC. Therapeutic monoclonal antibodies. Lancet. 2000;355:735–740. doi: 10.1016/s0140-6736(00)01034-5. [DOI] [PubMed] [Google Scholar]
- Bryant CE, Orr S, Ferguson B, Symmons MF, Boyle JP, Monie TP. International union of basic and clinical pharmacology. XCVI. Pattern recognition receptors in health and disease. Pharmacol Rev. 2015;67:462–504. doi: 10.1124/pr.114.009928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chan AC, Carter PJ. Therapeutic antibodies for autoimmunity and inflammation. Nat Rev Immunol. 2010;10:301–316. doi: 10.1038/nri2761. [DOI] [PubMed] [Google Scholar]
- Chen IF, Fu LW. Mechanisms of acquired resistance to tyrosine kinase inhibitors. Acta Pharm Sin B. 2011;1:197–207. doi: 10.1016/j.apsb.2015.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chliva C, Aggelides X, Makris M, Katoulis A, Rigopoulos D, Tiligada E. Comparable profiles of serum histamine and IgG4 levels in allergic beekeepers. Allergy. 2015;70:457–460. doi: 10.1111/all.12568. [DOI] [PubMed] [Google Scholar]
- Cohen A. From pharmacology to immunopharmacology. Br J Clin Pharmacol. 2006;62:379–382. doi: 10.1111/j.1365-2125.2006.02760.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corbisier J, Galès C, Huszagh A, Parmentier M, Springael JY. Biased signaling at chemokine receptors. J Biol Chem. 2015;290:9542–9554. doi: 10.1074/jbc.M114.596098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- del Cuvillo A, Mullol J, Bartra J, Dávila I, Jáuregui I, Montoro J, et al. Comparative pharmacology of the H1 antihistamines. J Investig Allergol Clin Immunol. 2006;16:3–12. [PubMed] [Google Scholar]
- Davies SP, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J. 2000;351:95–105. doi: 10.1042/0264-6021:3510095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dollery CT. Lost in Translation (LiT) Br J Pharmacol. 2014;171:2269–2290. doi: 10.1111/bph.12580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eisenbarth SC, Colegio OR, O’Connor W, Sutterwala FS, Flavell RA. Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nature. 2008;453:1122–1126. doi: 10.1038/nature06939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fabbro D. 25 years of small molecular weight kinase inhibitors: potentials and limitations. Mol Pharmacol. 2015;87:766–775. doi: 10.1124/mol.114.095489. [DOI] [PubMed] [Google Scholar]
- Fabbro D, Cowan-Jacob SW, Moebitz H. 10 things you should know about protein kinases’ IUPHAR Review 14. Br J Pharmacol. 2015;172:2675–2700. doi: 10.1111/bph.13096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feldmann M, Maini RN. Anti-TNF alpha therapy of rheumatoid arthritis: what have we learned? Annu Rev Immunol. 2001;19:163–196. doi: 10.1146/annurev.immunol.19.1.163. [DOI] [PubMed] [Google Scholar]
- Galluzzi L, Vacchelli E, Bravo-San Pedro JM, Buqué A, Senovilla L, Baracco EE, et al. Classification of current anticancer immunotherapies. Oncotarget. 2014;5:12472–12508. doi: 10.18632/oncotarget.2998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gebril A, Alsaadi M, Acevedo R, Mullen AB, Ferro VA. Optimising efficacy of mucosal vaccines. Expert Rev Vaccine. 2012;11:1139–1155. doi: 10.1586/erv.12.81. [DOI] [PubMed] [Google Scholar]
- Holgate ST. Immune circuits in asthma. Curr Opin Pharmacol. 2013;13:345–350. doi: 10.1016/j.coph.2013.03.008. [DOI] [PubMed] [Google Scholar]
- Kenakin T, Christopoulos A. Signaling bias in new drug discovery: detection, quantification and therapeutic impact. Nat Rev Drug Discov. 2013;12:205–216. doi: 10.1038/nrd3954. [DOI] [PubMed] [Google Scholar]
- Kopf M, Bachmann MF, Marsland BJ. Averting inflammation by targeting the cytokine environment. Nat Rev Drug Discov. 2010;9:703–718. doi: 10.1038/nrd2805. [DOI] [PubMed] [Google Scholar]
- Köhler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256:495–497. doi: 10.1038/256495a0. [DOI] [PubMed] [Google Scholar]
- Kyriakidis K, Zampeli E, Palaiologou M, Tiniakos D, Tiligada E. Histamine H3 and H4 receptor ligands modify vascular histamine levels in normal and arthritic large blood vessels in vivo. Inflammation. 2015;38:949–958. doi: 10.1007/s10753-014-0057-1. [DOI] [PubMed] [Google Scholar]
- van Leeuwen RW, van Gelder T, Mathijssen RH, Jansman FG. Drug-drug interactions with tyrosine-kinase inhibitors: a clinical perspective. Lancet Oncol. 2014;15:e315–e326. doi: 10.1016/S1470-2045(13)70579-5. [DOI] [PubMed] [Google Scholar]
- Leroux-Roels G. Unmet needs in modern vaccinology: adjuvants to improve the immune response. Vaccine. 2010;28S:25–36. doi: 10.1016/j.vaccine.2010.07.021. [DOI] [PubMed] [Google Scholar]
- Loriot Y, Perlemuter G, Malka D, Penault-Lorca F, Boige V, Deutsch E. Drug insight: gastrointestinal and hepatic adverse effects of molecular-targeted agents in cancer therapy. Nat Clin Pract Oncol. 2008;5:268–278. doi: 10.1038/ncponc1087. [DOI] [PubMed] [Google Scholar]
- Marfe G, Di Stefano C. Bypass mechanisms of resistance to tyrosine kinase inhibition in chronic myelogenous leukaemia. Curr Drug Discov Technol. 2014;11:145–153. doi: 10.2174/1570163811666140212111508. [DOI] [PubMed] [Google Scholar]
- Medzhitov R. Recognition of microorganisms and activation of the immune response. Nature. 2007;449:819–826. doi: 10.1038/nature06246. [DOI] [PubMed] [Google Scholar]
- Moebitz H, Fabbro D. Conformational bias: a key concept for protein kinase inhibition. Eur Phar Rev. 2012;17:41–51. [Google Scholar]
- Mothes N, Heinzkill M, Drachenberg KJ, Sperr WR, Krauth MT, Majlesi Y, et al. Allergen-specific immunotherapy with a monophosphoryl lipid A-adjuvanted vaccine: reduced seasonally boosted immunoglobulin E production and inhibition of basophil histamine release by therapy-induced blocking antibodies. Clin Exp Allergy. 2003;33:1198–1208. doi: 10.1046/j.1365-2222.2003.01699.x. [DOI] [PubMed] [Google Scholar]
- Nagar B, Bornmann WG, Pellicena P, Schindler T, Veach DR, Miller WT, et al. Crystal structures of the kinase domain of c-Abl in complex with the small molecule inhibitors PD173955 and Imatinib (STI-571) Cancer Res. 2002;62:4236–4243. [PubMed] [Google Scholar]
- Nijmeijer S, Vischer HF, Sirci F, Schultes S, Engelhardt H, de Graaf C, et al. Detailed analysis of biased histamine H4 receptor signalling by JNJ 7777120 analogues. Br J Pharmacol. 2013;170:78–88. doi: 10.1111/bph.12117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parsons ME, Ganellin CR. Histamine and its receptors. Br J Pharmacol. 2006;147:127–135. doi: 10.1038/sj.bjp.0706440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patel P, Salapatek AM. Pollinex Quattro: a novel and well-tolerated, ultra short-course allergy vaccine. Expert Rev Vaccines. 2006;5:617–629. doi: 10.1586/14760584.5.5.617. [DOI] [PubMed] [Google Scholar]
- Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP, Buneman OP, et al. The IUPHAR/BPS Guide to PHARMACOLOGY: an expert-driven knowledgebase of drug targets and their ligands. Nucleic Acids Res. 2014;42:D1098–D1106. doi: 10.1093/nar/gkt1143. (Database Issue): [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pelaia G, Vatrella A, Maselli R. The potential of biologics for the treatment of asthma. Nat Rev Drug Discov. 2012;11:958–972. doi: 10.1038/nrd3792. [DOI] [PubMed] [Google Scholar]
- Rask-Andersen M, Almen MS, Schioth HB. Trends in the exploitation of novel drug targets. Nat Rev Drug Discov. 2011;10:579–590. doi: 10.1038/nrd3478. [DOI] [PubMed] [Google Scholar]
- Reff ME, Carner K, Chambers KS, Chinn PC, Leonard JE, Raab R, et al. Depletion of B cells in vivo by a chimeric mouse human monoclonal antibody to CD20. Blood. 1994;83:435–445. [PubMed] [Google Scholar]
- Rosewich M, Lee D, Zielen S. Pollinex Quattro: an innovative four injections immunotherapy in allergic rhinitis. Hum Vaccin Immunother. 2013;9:1523–1531. doi: 10.4161/hv.24631. [DOI] [PubMed] [Google Scholar]
- Scalapino KJ, Daikh DI. CTLA-4: a key regulatory point in the control of autoimmune disease. Immunol Rev. 2008;223:143–155. doi: 10.1111/j.1600-065X.2008.00639.x. [DOI] [PubMed] [Google Scholar]
- Schumacher S, Kietzmann M, Stark H, Bäumer W. Unique immunomodulatory effects of azelastine on dendritic cells in vitro. Naunyn Schmiedebergs Arch Pharmacol. 2014;387:1091–1099. doi: 10.1007/s00210-014-1033-x. [DOI] [PubMed] [Google Scholar]
- Siebenhaar F, Falcone FH, Tiligada E, Hammel I, Maurer M, Sagi-Eisenberg R, et al. The search for mast cell and basophil models – are we getting closer to pathophysiological relevance? Allergy. 2015;70:1–5. doi: 10.1111/all.12517. [DOI] [PubMed] [Google Scholar]
- Simon D, Borradori L, Simon HU. Glucocorticoids in autoimmune bullous diseases: are neutrophils the key cellular target? J Invest Dermatol. 2013;133:2314–2315. doi: 10.1038/jid.2013.205. [DOI] [PubMed] [Google Scholar]
- Song DH, Lee JO. Sensing of microbial molecular patterns by Toll-like receptors. Immunol Rev. 2012;250:216–229. doi: 10.1111/j.1600-065X.2012.01167.x. [DOI] [PubMed] [Google Scholar]
- Steinman L, Merrill JT, McInnes IB, Peakman M. Optimization of current and future therapy for autoimmune diseases. Nat Med. 2012;18:59–65. doi: 10.1038/nm.2625. [DOI] [PubMed] [Google Scholar]
- Sundberg TB, Xavier RJ, Schreiber SL, Shamji AF. Small-molecule control of cytokine function: new opportunities for treating immune disorders. Curr Opin Chem Biol. 2014;23C:23–30. doi: 10.1016/j.cbpa.2014.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanaka T, Narazaki M, Kishimoto T. Therapeutic targeting of the interleukin-6 receptor. Annu Rev Pharmacol Toxicol. 2012;52:199–219. doi: 10.1146/annurev-pharmtox-010611-134715. [DOI] [PubMed] [Google Scholar]
- Thomas ML. The leukocyte common antigen family. Annu Rev Immunol. 1989;7:339–369. doi: 10.1146/annurev.iy.07.040189.002011. [DOI] [PubMed] [Google Scholar]
- Thomson AW, Turnquist HR, Raimondi G. Immunoregulatory functions of mTOR inhibition. Nat Rev Immunol. 2009;9:324–337. doi: 10.1038/nri2546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tiligada E. Editorial: is histamine the missing link in chronic inflammation? J Leukoc Biol. 2012;92:4–6. doi: 10.1189/jlb.0212093. [DOI] [PubMed] [Google Scholar]
- Tiligada E, Zampeli E, Sander K, Stark H. Histamine H3 and H4 receptors as novel drug targets. Expert Opin Investig Drugs. 2009;18:1519–1531. doi: 10.1517/14728220903188438. [DOI] [PubMed] [Google Scholar]
- Ubersax JA, Ferrell JE., Jr Mechanisms of specificity in protein phosphorylation. Nat Rev Mol Cell Biol. 2007;8:530–541. doi: 10.1038/nrm2203. [DOI] [PubMed] [Google Scholar]
- Woodward DF, Jones RL, Narumiya S. International Union of Basic and Clinical Pharmacology. LXXXIII: classification of prostanoid receptors, updating 15 years of progress. Pharmacol Rev. 2011;63:471–538. doi: 10.1124/pr.110.003517. [DOI] [PubMed] [Google Scholar]
- Zampeli E, Tiligada E. The role of histamine H4 receptor in immune and inflammatory disorders. Br J Pharmacol. 2009;157:24–33. doi: 10.1111/j.1476-5381.2009.00151.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell populations (*) Annu Rev Immunol. 2010;28:445–489. doi: 10.1146/annurev-immunol-030409-101212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- IUPHAR Immunopharmacology Section. 2015. Introduction [Online]. Available at: http://www.iuphar.us/index.php/sections-subcoms/immunopharmacology (accessed 1/11/2015)
- IUPHAR/BPS Guide to Pharmacology. 2015. Home [Online] Available at: http://www.guidetopharmacology.org/ (accessed 1/11/2015)
- WHO Model Lists of Essential Medicines. 2013. WHO Model Lists of Essential Medicines [Online]. Available at: http://www.who.int/medicines/publications/essentialmedicines/en/index.html (accessed 1/12/2015)
