Mechanism of Action and Pharmacokinetics of Biologics

 

Infliximab

Adalimumab

Certolizumab

Golimumab

CT-P13

ABP501

sTNF binding

Y

Y

Y

Y

Y

Y

mTNF binding

Y

Y

Y

Y

Y

Y

mTNF reverse signaling

Y

Y

?

Y

Y

?

Inhibits cytokine production

Y

Y

Y

?

Y

Y

Fc-mediat ed ADCC/CDC

Y

Y

N

Y

Y

Y



The net consequences of binding of anti-TNF antibodies to mTNF and sTNF are to limit their ongoing effects on immune responses on patients. Treatment with infliximab, for example, leads to a decrease in neutrophil growth factors (GM-CSF), lamina propria polymorphonuclear cells, and the pro-inflammatory cytokines IL-1 beta, IFN-γ, IL-13, IL-17A, IL-6, and MMP9 [11]. Anti-TNF treatment also alters the balance of pro- to anti-inflammatory cell phenotypes of the immune system. Infliximab has been shown to restore functional deficits in regulatory T-cells (Tregs), reflected in an increased expression of FoxP3 and in an increase in the suppressive activity of CD4+/CD25+ T-cells [12, 13]. Beyond T-cells, a range of beneficial effects have been reported in epithelial cells, regulator macrophages, and myofibroblasts in response to anti-TNF exposure [11].

When anti-TNF antibodies bind to membrane TNF (mTNF), they can also trigger “reverse signaling” via mTNF, which shuts down intracellular signaling pathways and induces apoptosis [14, 15]. Both infliximab and adalimumab induce apoptosis in peripheral blood cells, but etanercept and certolizumab do not [16]. Interestingly, infliximab and adalimumab have also been shown to induce cell cycle arrest, as a separate mechanism for suppression of immune cells [17]. The induction of apoptosis of T lymphocytes and CD14+ macrophages in patients with IBD occurs via TNFR2 [18]. A related potentia l mechanism of action is the induction of antibody-dependent cell-mediated cytotoxicity (ADCC) by anti-TNFs that can engage with IgG Fc receptors (FcR). Lysis of mTNF-expressing cells and PBMCs could be induced by infliximab and adalimumab more potently than etanercept, whereas certolizumab pegol did not show any effect (it lacks the Fc domain) [6]. Complement-dependent cytotoxicity (CDC) of cell lines in vitro is a third mechanism though which anti-TNFs could disrupt pro-inflammatory cell populations in vivo [19]. It is unclear at this time if this pathway is relevant in their mechanism of action in patients with IBD [9]. Both currently approved biosimilars show similar ability to induce both ADCC and CDC in cell lines assays [7].

Despite these well-documented alterations in cytokines, cell survival, and phenotypes in response to anti-TNF treatment, their association with the typical measures of clinical response in patients has been lacking. This reflects the gaps between the artificial scenario of cell lines and transfected cells in vitro, the complex cellular matrix of the lamina propria in patients, and the disconnect between symptoms and objective indices of mucosal inflammation. Associations between baseline biomarkers and subsequent clinical outcomes of anti-TNF therapy have yet to be validated in prospective cohorts [20]. One promising approach requires quantification of mucosal mTNF-positive cells using a confocal laser endomicroscope but reported a 70% differential in clinical response rates based on baseline mTNF levels [18].



Pharmacokinetics of Anti-TNFs


Pharmacokinetics (pK) describes the effects of the body’s physiological processes on an administered drug. For monoclonal antibodies (IgGs), adequate concentrations of the drug need to be achieved in the circulation for it to obtain its intended effects on circulating and intestinal mucosal cells. Individuals’ differences in bioavailability and pK have been associated in IBD with lack of clinical response and mucosal healing. Intravenous administration of anti-TNFs, such as infliximab, allows for administration of large volumes, rapid central distribution, and low variability in bioavailability; peak serum concentrations are attained almost immediately post-infusion [21]. In contrast, subcutaneous anti-TNFs can only be given in low-volume doses and are taken up by lymphatic drainage and paracellular movement, leading to slower absorption into the vascular compartment. For adalimumab, peak serum concentrations are reached approximately 5 days after a single 40 mg dose, with average bioavailability around 65% [21]. Once in the circulation, extravasation of anti-TNFs occurs primarily via receptor-mediated endocytosis into vascular endothelial cells. The volume of distribution of anti-TNFs is ~0.1 L/kg, suggesting these drugs are mainly distributed within the extracellular fluid [22]. Preliminary data with biosimilar infliximab (CT-P13) and adalimumab (ABP501) also report comparable pK profiles to their reference products in rheumatological diseases [7].

Elimination of monoclonal antibodies occurs mostly via proteolytic catabolism by phagocytic cells of the reticuloendothelial system [23]. The reported serum half-life of infliximab ranges from 7 to 12 days in patients with Crohn’s disease, in both those in remission and those with active disease [24, 25]. There is also the phenomenon of the “antigen sink” whereby internalization of anti-TNFs by their binding to mTNF can lead to their clearance from the extracellular space. This may explain the variability in clearance associated with inflammatory burden in patients with ulcerative colitis [26]. Balancing this process is the recycling of intact monoclonal antibodies back into the circulation, leading to the long serum half-life of IgGs (~23 days) and the slow systemic clearance of about 11–15 mL/h [25]. This system is disrupted by the presence of anti-drug antibodies (ADAs); ADAs congregate anti-TNFs into multimeric antibody complexes that are retained and degraded, but not recycled, by reticuloendothelial cells [27]. As an example of the impact of these ADAs on clearance, the clearance of infliximab increases threefold in patients with ADAs as compared with patients without ADAs [28]. The development of ADAs in patients with IBD is influenced by many factors, including genotype, trough drug levels, and concomitant medications [29]. Finally, fecal loss of anti-TNFs has been described as a particular problem to patients with active IBD. In patients with severe IBD, infliximab was noted in a greater proportion of patients failing therapy, compared to those with a clinical response [30]. It is unclear whether the drug leakage caused the loss of response or whether ongoing mucosal inflammation led to drug leakage.

Much study has been undertaken in recent years on the association between pK and clinical response to anti-TNFs and will be covered in detail in another chapter of this book. For many drugs, response is dependent on drug concentrations or drug exposure (the AUC), and therefore drug concentration-guided individualized therapy can be important [24]. For infliximab, for example, a meta-analysis concluded that patients who achieved an infliximab level >2 μg/mL were more three times more likely to be in clinical remission or achieve endoscopic remission than patients with levels <2 μg/mL [31]. This concentration-effect relationship has also been described for adal imumab, certolizumab, and golimumab [21].



Anti-integrins


Two anti-integrins are currently FDA approved for use in IBD: natalizumab and vedolizumab. Natalizumab is a humanized monoclonal antibody against the cell adhesion molecule α4-integrin. Although approved to treat Crohn’s disease, its association with progressive multifocal leukoencephalopathy (PML) has limited its use in IBD, particularly since the approval of vedolizumab. Vedolizumab is a humanized monoclonal antibody which acts against α4β7 integrin heterodimer and blocks the interaction of α4β7 integrin with MAdCAM-1. Other anti-integrins remain in clinical development, such as etrolizumab and the anti-MAdCAM antibody PF-00547659. Since vedolizumab is the only currently approved and widely used anti-integrin, this section will primarily discuss this agent.


Pharmacodynamics of Vedolizumab


Infiltration of the intestinal lamina propria by T lymphocytes is an established component of the pathogenic process in IBD, through molecular mechanisms unique to the intestinal tract [32]. Adhesion and signaling molecules on the surface of T lymphocytes (selectins, integrins, chemokine receptors) interact with ligands on the endothelium to instigate the migration process [33]. T lymphocytes utilize the α4β7 integrin to bind to mucosal addressin cell adhesion molecule 1 (MAdCAM-1) on endothelial cells [34]. Vedolizumab binds to the α4β7 integrin on peripheral blood lymphocytes and inhibits adhesion of the lymphocyte to MAdCAM-1. In addition to circulating mononuclear cells, vedolizumab also binds to mononuclear cells in the lymphoid tissues, intestinal tract, and bladder [35]. The highest level of binding by vedolizumab was observed on the α4β7+ population of memory CD45RO+ CD4+ T lymphocytes but also to B lymphocytes, naive CD8 T lymphocytes, Th17 cells, natural killer cells, and basophils. After administration of vedolizumab, almost 100% of MAdCAM-1-Fc receptors are saturated immediately, and this effect wears off around 20 weeks after the last dose [36, 37]. These data suggest potent inhibition of trafficking of a number of pro-inflammatory immune cells to the intestinal tract after vedolizumab is administered.

In addition to its effects on effector (pro-inflammatory) T-cells (Teff), β7 integrin is a component of migration of regulatory T-cells. Mice lacking β7 integrin exhibit depleted colonic regulatory T (Treg) cells and excessive macrophage infiltration in the colon, thereby exacerbating DSS-induced colitis [38]. Additionally, in patients with UC, Treg homing to the gut was suppressed significantly by vedolizumab, and this led to a decrease in the ratio between Teff and Treg cells in the peripheral circulation [39]. It is unclear whether this has implications for the protective role of Tregs and CD4+ cells in immune surveillance. Clinical trial data reported a greater risk of serious infections in patients treated with vedolizumab (6% vs. 3%), and a recent case series reported a significantly higher rate of surgical site infections with vedolizumab than in patients receiving anti-TNF agents [40, 41]. Further analysis of tissue T-cells will be required to determine the mucosal impact of limiting T-cell migration.


Pharmacokinetics of Vedolizumab


Like the anti-TNFs, vedolizumab is a humani zed immunoglobulin G1 (IgG1) monoclonal antibody, and therefore it shares many pK properties with them. In patients with UC, serum concentrations increased linearly with increasing doses of vedolizumab and declined linearly after the last dose [36]. A population pharmacokinetic analysis that included data from phase II studies suggested that disease type (UC or CD) had no impact on the pharmacokinetics of vedolizumab [37]. Linear clearance was 0.15 L/day for patients with UC and CD, and the terminal elimination half-life (t1/2) was 26 days. Extreme low albumin concentrations (<3.2 g/dL) and extreme high weight values (>120 kg) were both associated with higher drug clearance of vedolizumab in these studies. In contrast, fecal calprotectin, CDAI score, disease activity scores, age, prior anti-TNF exposure, ADA status, and concomitant therapy use had no clinically relevant effects on vedolizumab clearance [37]. In this pK model, patients with an endoscopic subscore of 3 after induction therapy had on average 25% higher clearance than patients with an endoscopic subscore of 0, highlighting the importance of the “tissue sink” noted with anti-TNFs. Eleven (28%) vedolizumab-treated participants were persistently positive for ADAs, and clearance of vedolizumab was 12% greater than in participants in the same dose group who were not persistently ADA positive [42]. Surprisingly, α4β7 receptor saturation was maintained at vedolizumab concentrations considered subtherapeutic (1 μg/mL), raising the question of whether receptor saturation alone is sufficient for clinical efficacy (vedolizumab concentrations abo ve 15 μg/mL) [37].


Anti-IL-12/23


The cytokines IL-12 and IL-23 are secreted heterodimeric cytokines, which both contain a p40 protein subunit. IL-12 is primarily produced by phagocytic and dendritic cells in response to microbial stimulation and drives cell-mediated immunity by inducing lymphokine-activated killer cells and activation of natural killer (NK) cells and T lymphocytes, particularly Th1 populations [43]. IL-23 drives a population of T-cells (Th17) that produce IL-17, IL-6, and TNF [44]. In IBD, genome-wide association studies revealed that variants of the gene encoding the IL-23 receptor, and the p40 chain, conferred genetic risk for developing IBD. IL-17 mRNA expression is increased in the colon of patients with active UC and CD, correlating with the density of CD4+ T-cells [45]. IL-17 production by isolated lamina propria CD4+ T-cells from patients with UC is significantly increased by IL-23 [46]. IL-17 appears to play a role in IBD pathogenesis, as it can stimulate innate immune cells and epithelial cells to produce IL-1, IL-6, and IL-8, which induce increased neutrophil recruitment and other pro-inflammatory signals [47]. However, it should be noted that there is also evidence that IL-17 plays a role in mucosal homeostasis, with protective effects on the intestinal epithelium, and generation of antimicrobial peptides [13].


Pharmacodynamics of Ustekinumab


Ustekinumab is a human IgG1 monoclonal antibody developed to bind to IL-12 and later discovered to bind specifically to the p40 protein subunit of this cytokine [48]. After ustekinumab was developed, it was subsequently established that the cytokine IL-23 contains a p40 subunit, to which ustekinumab also binds. This dual specificity was unique in approved biologics but provides challenges by engaging an unintended pathway (IL-17). Ustekinumab binding to the p40 subunits of these cytokines prevents IL-12 and IL-23 from binding to the IL-12Rβ1 receptor and IL-23 (IL-12Rβ1/23R) receptor complexes on the surface of NK and T-cells [49]. It can only bind to free cytokines, not receptor-bound complexes, and is thus unlikely to mediate Fc effector functions, such as ADCC or CDC (see anti-TNFs). Binding to ustekinumab neutralizes IL-12/23-mediated responses, including production of IFNγ, IL-17A, IL-17F, and IL-22. It is important to note that while ustekinumab will effectively neutralize IL-12- and IL-23-mediated functional responses, it will not affect immune responses stimulated through other cytokines or cellular activities, e.g., Th2 cytokines.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Feb 6, 2018 | Posted by in GASTROENTEROLOGY | Comments Off on Mechanism of Action and Pharmacokinetics of Biologics

Full access? Get Clinical Tree

Get Clinical Tree app for offline access