Next Article in Journal
Children and Ultraviolet Radiation
Previous Article in Journal
Oral Health Preventive Program in Patients with Autism Spectrum Disorder
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Second and Third Generational Advances in Therapies of the Immune-Mediated Kidney Diseases in Children and Adolescents

Department of Nephrology, Kidney Transplantation and Hypertension, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Children 2022, 9(4), 536; https://doi.org/10.3390/children9040536
Submission received: 24 March 2022 / Revised: 6 April 2022 / Accepted: 8 April 2022 / Published: 11 April 2022
(This article belongs to the Section Pediatric Nephrology)

Abstract

:
Therapy of immune-mediated kidney diseases has evolved during recent decades from the non-specific use of corticosteroids and antiproliferative agents (like cyclophosphamide or azathioprine), towards the use of more specific drugs with measurable pharmacokinetics, like calcineurin inhibitors (cyclosporine A and tacrolimus) and mycophenolate mofetil, to the treatment with biologic drugs targeting detailed specific receptors, like rituximab, eculizumab or abatacept. Moreover, the data coming from a molecular science revealed that several drugs, which have been previously used exclusively to modify the upregulated adaptive immune system, may also exert a local effect on the kidney microstructure and ameliorate the functional instability of podocytes, reducing the leak of protein into the urinary space. The innate immune system also became a target of new therapies, as its specific role in different kidney diseases has been de novo defined. Current therapy of several immune kidney diseases may now be personalized, based on the detailed diagnostic procedures, including molecular tests. However, in most cases there is still a space for standard therapies based on variable protocols including usage of steroids with the steroid-sparing agents. They are used as a first-line treatment, while modern biologic agents are selected as further steps in cases of lack of the efficacy or toxicity of the basic therapies. In several clinical settings, the biologic drugs are effective as the add-on therapy.

1. Introduction

There are a variety of immune-mediated kidney diseases diagnosed in children from early childhood to adolescence. Most of them are presented as one of the clinical forms of isolated glomerulonephritis, while the other ones as a vasculitis-related nephritis or a systemic disease with renal involvement. Nevertheless, the clinical pattern, the innate and/or adaptive immune system is upregulated in most cases and several specific mechanisms of immune dysfunction are involved, as an underlying cause. The typical clinical characteristics of nephrotic glomerulopathies are related to the frequent relapses and the remission dependence from several drugs (as the dose reduction or drug withdrawal results in a relapse of the disease). The hypocomplementemic kidney diseases are often resistant to the standard therapies. The scientific knowledge and clinical experience regarding the relevant therapies have evolved over time. The adaptive immune system was a general target of steroids, which has been the first-line therapy for decades [1,2,3,4,5]. The steroid-related toxicity, especially important in the long-term treatment, was a reason for introducing the steroid-sparing, primarily non-selective antiproliferative drugs, including cyclophosphamide (CYC) and azathioprine (AZA) [5,6,7,8,9]. Treatment with these drugs was not routinely followed by therapeutic drug monitoring (TDM), which precisely evaluates the exposure to the native drugs or its active metabolites This monitoring possibility has appeared more recently (for CYC and AZA) [10], however, it is not frequently used in clinical practice. The next step in specific pharmacotherapy was related to the wider availability of calcineurine inhibitors—cyclosporine A (CsA) and tacrolimus (TAC) [11,12,13,14,15]—and the selective antiproliferative drug mycophenolate mofetil (MMF) [16,17]. The exposure to all of them is easily measurable with TDM. The next step of progress was associated with introduction of the biologic drugs, targeting specific receptors or narrow pathways of the innate and/or adaptive immune system, important for the variable mechanisms of a specific kidney disease [18,19,20,21]. The final issue in the therapy of immune-mediated kidney diseases was the discovery of local mechanisms of specific drugs, targeting podocytes in glomeruli, independently of the impact on the immune system [22,23]. Evolution of the management of pediatric immune-mediated kidney diseases is presented in Figure 1.
The variety of drugs allows individualization of therapy; however, from a clinical point of view, there are several significant limitations, which may decrease the efficacy and overall suitability of a certain protocol in a particular case. The summary characteristics of the classic drugs used in pediatric immune-mediated kidney diseases is presented in Table 1, and of the modern biologic drugs in Table 2.

2. Therapeutic Drug Monitoring (TDM) and Immunomonitoring in Kidney Diseases

TDM is a mandatory tool in clinical management of immune-mediated kidney diseases. The general rules of TDM are summarized in Table 3. There are two distinct ways of surveillance of the exposure to the drugs, used in the therapy of the kidney diseases: a classic TDM, based on a regular monitoring of drug concentration in the blood (or plasma), as by a single test (pre-dose trough level; C0) or by the parameter defined as the area under the curve (AUC), combining the results of multiple tests/daily [40,41,42] and immunomonitoring of the number of target cells, depleted by a specific monoclonal antibody (like B CD20) (pre-dose test) [43]. The associations between the dose and drug concentration or a number of target cells are clinically relevant in terms of systemic action of the certain drugs; however, in terms of local effect on podocytes, the associations are not clear. The clinical rules of TDM and immunomonitoring are presented in Table 3.

3. Nephrotic Syndrome—An Alternative Management beyond the Classic Protocols

Nephrotic syndrome is an “umbrella term” covering the typical “idiopathic” nephrotic syndrome in children, with minimal change and (less frequently) focal segmental glomerulosclerosis (FSGS) in a kidney biopsy, as well as symptomatic proteinuria presented in other types of glomerulonephritis (GN), especially in mesangial-proliferative GN and membranous nephropathy. The pediatric nephrotic “family” is heterogeneous in terms of underlying immune-mediated (non-genetic) mechanisms, and this was also important for selection of the secondary-step therapies. One of these mechanisms is related to the dysregulation of the interaction between B and T cells, resulting in their dysfunction. B and T cells may produce several bioactive cytokines, which are regarded as “protein permeability factors”, the heterogeneous family of cytokines binding specific targets on podocytes, impact their functionality and shape, and also provoke and maintain the clinically overt proteinuria. This family includes cardiothropin-like cytokine-1 (CLC-1)—a soluble urokinase-type plasminogen activator receptor (suPAR), IL-4, IL-10, IL-13, TNFα and anti-CD40 antibody (and probably many others, still not defined) [44]. Some of these processes may be modified by the use of rituximab, a IgG1 kappa- type monoclonal antibody, which induces lysis of B cells (pre-B, immature, mature and activated), expressing the CD20 receptor. The effect of rituximab is exerted via B cell depletion, indirect impact on T cells and local activity in podocytes. The depletion is executed by the antibody-dependent or complement-dependent cytotoxicity resulting in apoptosis, inhibition of proliferation and alteration of the cell cycle. Activation and differentiation of T cells, as well as the ability to release several podocyte-specific cytokines are (in some part) regulated by B cells, which produce specific costimulatory signals; therefore, B cell depletion may ameliorate the T-cell specific activities. Rituximab is also able to modulate the Treg and Breg cell interactions, which are involved in the mechanism of a relapsing nephrotic syndrome [24,25,45]. Rituximab also exerts a specific local effect on podocytes (independently from its effect on B CD20 receptor), related to the stabilization of the podocyte’s cytoskeleton by regulation (preservation) of sphingomyelin phosphodiesterase acid-like 3b (SMLPD-3b)—a protein involved in the podocyte cytoskeleton activity regulation [23]. Clinical relevance of this mechanism is not clear [46]; however, it is speculated that partial remissions achieved in some of the treated patients or the lack of correlation between number of peripheral CD20 cells (or CD19; used for drug effect monitoring) and clinical efficacy (seen also in some patients) are the results of the local (not systemic) effect of rituximab [26]. Whether the routine of the dosing regimen used for systemic treatment (e.g., 4 × 375 mg/m2 of BSA) is also adequate for local effect exerted by rituximab is not clear either.

4. Cyclosporine A as a Podocyte Cytoskeleton Stabilizer

The systemic immunosuppressive mechanism of cyclosporine A (CsA) is related to the inhibition of the nuclear factor of activated T cells (NFAT) signaling in the T lymphocytes. There is also another distinct mechanism of CsA (not dependent on NFAT inhibition in T cells), which is related to the stabilization of the actin cytoskeleton in the podocytes. CsA blocks the calcineurin-mediated dephosphorylation of synaptopodin in podocytes, preserving the phosphorylation-dependent synaptopodin–14-3-3β interaction. This protects synaptopodin from cathepsin L-mediated degradation [47]. In clinical translation, it may reduce the degree of proteinuria, when a nephrotic patient is constantly exposed to cyclosporine A [48]. The exact association between the value of CsA concentration in the blood and activity of the local mechanism in terms of the podocyte stabilization in unclear, however there are patients with genetic forms of nephrotic syndrome, where a major underlying cause of proteinuria is a defect of kidney microstructure, who gain a clinical profit (decrease of proteinuria during constant treatment) under the routine doses and the trough concentrations of the drug [49,50]. This suggests that a routine “clinical” dosing is relevant also for the local effect of CsA.

5. Is Targeting of a CD80 Molecule-Clinically Relevant?

The CD80 (B7-1) is a molecule expressed on the surface of T cells, dendritic cells and podocytes. The “two-hit” hypothesis suggests that an immune response to external trigger (e.g., viral infection) in the immunocompetent humans is limited to a transient expression of CD80 on podocytes, which may induce a short-term and minor proteinuria. Humans with a sustained dysfunction of the regulatory T cells (Treg) react to the exposure to the similar trigger with marked and sustained expression of CD80 in podocytes, which causes a dysregulation of cytoskeleton and in clinical consequence—a persistent heavy proteinuria (nephrotic syndrome) [51,52]. CD80 is also expressed on the T cells infiltrating the kidney tissue (at least) in some nephrotic children [53]. CTLA-4 (cytotoxic T-lymphocyte-associated protein 4), expressed on podocytes and Tregs, is an important co-factor in this mechanism [54]. High urine excretion of CD80 is being regarded as a biomarker of minimal change disease [55,56]. Translation of these mechanisms to the potentially relevant therapy involves abatacept, which is a fusion molecule including the Fc region of IgG1 and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4-Ig). The drug acts as a co-stimulatory inhibitor and targets B7-1 (CD80) and CD86, disrupting the activation of T-cells. It also blocks the disruption of binding of talin to ẞ1-integrin caused by CD80. This mechanism should ameliorate a podocyte injury and decrease proteinuria. A primary clinical report showed clinical efficacy of abatacept in 4 (of 5) cases (including one child and two adolescents), resistant to other drugs, including rituximab [57]. Further reports were more reluctant in terms of the efficacy of abatacept [27,58]. Overall, the clinical efficacy was reported in about 44% of nephrotic patients [27]. Probably, the use of abatacept should be limited to the cases, in which the renal biopsy (of native or transplanted kidney) will confirm the local expression of B7-1 (CD80) in podocytes and/or in which the baseline value of the urinary excretion of CD80 is markedly increased [59].

6. The Role of Angiopoietin-like-4 (Angptl4) in Minimal Change Nephrotic Syndrome and a Hypothetic Target-Treatment

Podocytes of patients with minimal change nephrotic syndrome (MCNS) express angiopoietin-like 4 (Angptl4). There two types of Angptl4 protein: hyposialylated Angptl4 secreted locally by podocytes, and a neutral form, present in high concentration in the plasma of nephrotic patients. The podocyte-derived hyposialylated Angptl4 mediates proteinuria in patients with MCNS and binds to a glomerular basement membrane and endothelial cells [60,61]. Converting a hyposialylated Angptl4 to the sialylated form with N-acetyl-D-mannosamine (a precursor of sialic acid), that can be taken up and stored in podocytes, may reduce proteinuria and has the potential to be used in small maintenance doses to prevent the relapses of MCNS [62]. Despite the attractive hypothesis, there are no available relevant clinical data so far.

7. Complement System Blocking Agents in Glomerular Diseases (with Defect of Complement)

The role of a complement system in the mechanism of the specific glomerular kidney diseases is very complex. In physiology, the complement is being used as a natural tool for the clearance of immune complexes; however, it may also act as a link between innate and adaptive immunity, activating B-cells and T-cells in the local renal environment. The genetic mutations of complement components and/or regulators or a presence of specific acquired autoantibodies targeting the complement components lead to the excessive activation of the alternative pathway and to the glomerular deposition of the complement fragments (debris), as may be documented in the kidney biopsies [63,64,65]. The clinical spectrum of complement-associated kidney diseases includes membranous nephropathy, lupus nephritis, C3 glomerulonephritis (C3GN), membranoproliferative glomerulopathy type I, dense deposit disease (DDD), IgAN and ANCA-associated vasculitis. Several monoclonal antibodies and other molecules targeting the pathways of complement system are currently evaluated in clinical trials associated with different types of glomerulonephritis [28]. The pediatric experience is limited to the use of eculizumab in cases of C3GN, dense deposit disease and membranoproliferative GN [29,30,31]. The most significant experience was reported in the multicentre series of children and adolescents with C3GN, treated with eculizumab, in whom the efficacy of this monoclonal antibody was confirmed in cases of crescentic rapidly progressive C3 glomerulopathy, while the clinical benefit was limited in the clinically mild cases [32].

8. Biologic Drugs in Pediatric Systemic Vasculitis

Systemic vasculitis is a family of diseases characterized by the presence of vascular wall inflammation and involving multiorgan symptoms. Despite the low incidence in children (mainly adolescents), this group of diseases, and particularly ANCA-associated vasculitis (AAV)), has gained an access to several biologic agents, targeting the underlying mechanisms. The pathogenesis of AAV is multifactorial and genetic/epigenetic factors interact with different external triggers. Dysregulation of B cells and lack of balance between T helpers and T effectors lead to the production of ANCA (anti-neutrophil cytoplasmic antibody), activation of neutrophils, and further damage of vessel walls with the late multiorgan consequences [66]. The relevant biologic agents, preliminarily described in case reports, then evaluated in several clinical trials enrolling adult and (in some projects) also pediatric and adolescent patients, include rituximab (anti-B CD20 moab), infliximab (anti-TNFα moab), etanercept (TNFα-receptor blocker), abatacept (CTLA-4 Ig Fc fusion protein), alemtuzumab (humanized anti-CD52 moab) and tocilizumab (anti-IL6 moab) [67]. The use of rituximab is aimed to block B-cell dependent T-cell activation, causing the overproduction of several interleukins (like IL-5). A pediatric trial (among several others enrolling only adult patients), in which children with granulomatosis with polyangiitis (GPA) and microscopic polyangiitis (MPA) received four doses of rituximab, has demonstrated a positive effect, as the remission (verified with the Pediatric Vasculitis Activity Score) was achieved in 56%, 92%, and 100% of patients at months 6, 12, and 18, respectively [33]. The idea of using TNFα blockers is based on the mechanism, in which inhibition of TNFα decreases the formation of granuloma, as a result of the TNFα-mediated activation of neutrophils that enhances the ability of ANCA to stimulate degranulation of neutrophils, is a which important factor in a process of the vascular wall damage [67]. The study named WGET (Wegener’s Granulomatosis Etanercept Trial), which enrolled adult patients, did not prove a clinical benefit in terms of achieving remission, as compared with the placebo group (notably, both groups also received standard care drugs, including steroid and cyclophosphamide or methotrexate [34]. Abatacept was used as a T-cell activation blocking agent in adult patients with GPA (granulomatosis with polyangiitis). Overall, 80% of enrolled patients achieved remission, and in 78% of cases a complete steroid withdrawal was possible [35]. Alemtuzumab, a depleting moab against CD52, the receptor expressed by the several cells (T lymphocytes, monocytes, macrophages), was used in adult AAV patients. The remission was achieved in two thirds of patients at six months, and was maintained to twelve months in one third of them [36].
The clinical view on the real success rate in several trials, comparing the efficacy and safety of modern biologic agents with “traditional” drugs, is complex, as in several cases the interpretation of final outcome and safety was difficult and not always in favor of the use of particular biologics, which have been used as the add-on therapy.

9. Biologic Drugs in Systemic Lupus Erythematosus with Renal Involvement

Several specific mechanisms relevant for the lupus erythematosus with renal involvement have been currently targeted by narrow biologic agents, evaluated in clinical trials. One of them is B-cell activating factor (BAFF). The agents used in this setting included blisibimod (A-623, AMG 623), a fusion protein, built of tetrameric BAFF binding domain fused to human IgG1 Fc region and belimumab (a monoclonal antibody). Clinical data on blisibimod from CHABLIS-SC1, randomized, double-blind, placebo-controlled clinical trial, conducted in adult patients, treated previously with variable drugs (including steroids, MMF, methotrexate, AZA or antimalarian agents), showed that the primary end-point (week 52 SLE Responder Index-6) was not achieved; however, the use of blisibimod was associated with decreased proteinuria, successful reduction of exposure to steroids, and a positive response of the relevant biomarkers [37]. Several clinical trials have been conducted in adults and children with SLE, evaluating the efficacy and safety of belimumab. Overall, in children, belimumab reduced the risk of severe flares by 64% (versus standard therapy), which was more pronounced than in the adult studies (23–50%, respectively) [38]. Atacicept is a recombinant fusion protein consisting of the binding portion of transmembrane activator and CAML interactor (TACI; also known as tumor necrosis factor receptor superfamily member 13B), that binds to BLyS (B lymphocyte stimulator) and APRIL (a proliferation-inducing ligand), inhibiting interactions with their specific receptors. An adult trial with extended follow-up showed a reduction of the incidence of severe SLE flares and prolongation of time to the occurrence of severe flares with a high dose of the drug, as compared to the placebo [39]. Blisibimod and atacitept have also been evaluated in adult patients with IgA nephropathy-related persistent proteinuria >1 g (NCT02062684; NCT02808429); however, (published) results are not available. In general, the role of these kind of narrow-targeted therapies may be regarded mainly as add-on treatments, ameliorating the clinical course of the disease and acting as the steroid-sparing drugs. Anyway, the direction towards the use of “therapeutic arrows” [68], drugs and agents designed based on increasing molecular knowledge on the mechanisms of immune-mediated kidney diseases is a promising clinical approach, with prospects for the future.

10. Summary

  • Upregulation of the innate and/or adaptive immune system leads to the development of a variety of immune-mediated kidney diseases in children.
  • There is an ongoing progress in pharmacotherapy of immune kidney diseases, based on scientific knowledge, which defines detailed, variable underlying disease-related mechanisms.
  • The adaptive immune system is a target of steroids, antiproliferative drugs, calcineurin inhibitors and several receptor-specific biologic agents.
  • The innate immune system is a target of specific monoclonal antibodies.
  • Surveillance of the current therapies is based on therapeutic drug monitoring and/or immunomonitoring.
  • Apart from the effect on the immune system, specific drugs (calcineurin inhibitors, rituximab, abatacept) also exert a local effect on the microstructure of the podocyte cytoskeleton, which may be clinically relevant in selected cases.

Author Contributions

Both authors contributed equally to the conceptualization, writing and editing of the review manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Coutinho, A.E.; Chapman, K.E. The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Mol. Cell. Endocrinol. 2011, 335, 2–13. [Google Scholar] [CrossRef] [PubMed]
  2. Yoshikawa, N.; Nakanishi, K.; Sako, M.; Oba, M.S.; Mori, R.; Ota, E.; Ishikura, K.; Hataya, H.; Honda, M.; Ito, S.; et al. A multicenter randomized trial indicates initial prednisolone treatment for childhood nephrotic syndrome for two months is not inferior to six-month treatment. Kidney Int. 2015, 87, 225–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lombel, R.M.; Gipson, D.S.; Hodson, E.M. Treatment of steroid-sensitive nephrotic syndrome: New guidelines from KDIGO. Pediatric Nephrol. 2012, 28, 415–426. [Google Scholar] [CrossRef] [PubMed]
  4. Haun, M.W.; Estel, S.; Rucker, G.; Friederich, H.C.; Villalobos, M.; Thomas, M.; Hartmann, M. Early palliative care for adults with advanced cancer. Cochrane Database Syst. Rev. 2017, 6, CD011129. [Google Scholar] [CrossRef]
  5. Yoshikawa, N.; Honda, M.; Iijima, K.; Awazu, M.; Hattori, S.; Nakanishi, K.; Ito, H. Steroid Treatment for Severe Childhood IgA Nephropathy: A Randomized, Controlled Trial. Clin. J. Am. Soc. Nephrol. 2006, 1, 511–517. [Google Scholar] [CrossRef] [Green Version]
  6. Moore, M.J. Clinical Pharmacokinetics of Cyclophosphamide. Clin. Pharmacokinet. 1991, 20, 194–208. [Google Scholar] [CrossRef]
  7. Tejani, A.; Phadke, K.; Nicastri, A.; Adamson, O.; Chen, C.; Trachtman, H.; Tejani, C. Efficacy of Cyclophosphamide in Steroid-Sensitive Childhood Nephrotic Syndrome with Different Morphological Lesions. Nephron Exp. Nephrol. 1985, 41, 170–173. [Google Scholar] [CrossRef]
  8. Zagury, A.; de Oliveira, A.L.; de Moraes, C.A.P.; Montalvão, J.A.D.A.; Novaes, R.H.L.L.; de Sá, V.M.; Carvalho, D.D.B.M.D.; Matuck, T. Long-term follow-up after cyclophosphamide therapy in steroid-dependent nephrotic syndrome. Pediatric Nephrol. 2011, 26, 915–920. [Google Scholar] [CrossRef]
  9. Kamei, K.; Nakanishi, K.; Ito, S.; Saito, M.; Sako, M.; Ishikura, K.; Hataya, H.; Honda, M.; Iijima, K.; Yoshikawa, N.; et al. Long-Term Results of a Randomized Controlled Trial in Childhood IgA Nephropathy. Clin. J. Am. Soc. Nephrol. 2011, 6, 1301–1307. [Google Scholar] [CrossRef] [Green Version]
  10. Mircheva, J.; Legendre, C.; Soria-Royer, C.; Thervet, E.; Beaune, P.; Kreis, H. Monitoring of azathioprine-induced immunosuppression with thiopurine methyltransferase activity in kidney transplant recipients. Transplantation 1995, 60, 639–642. [Google Scholar] [CrossRef]
  11. Plank, C.; Nephrologie, F.A.F.P.; Kalb, V.; Hinkes, B.; Hildebrandt, F.; Gefeller, O.; Rascher, W. Cyclosporin A is superior to cyclophosphamide in children with steroid-resistant nephrotic syndrome—A randomized controlled multicentre trial by the Arbeitsgemeinschaft für Pädiatrische Nephrologie. Pediatric Nephrol. 2008, 23, 1483–1493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Kemper, M.J.; Kuwertz-Broeking, E.; Bulla, M.; Mueller-Wiefel, D.E.; Neuhaus, T.J. Recurrence of severe steroid dependency in cyclosporin A-treated childhood idiopathic nephrotic syndrome. Nephrol. Dial. Transplant. 2004, 19, 1136–1141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Choudhry, S.; Bagga, A.; Hari, P.; Sharma, S.; Kalaivani, M.; Dinda, A. Efficacy and Safety of Tacrolimus Versus Cyclosporine in Children with Steroid-Resistant Nephrotic Syndrome: A Randomized Controlled Trial. Am. J. Kidney Dis. 2009, 53, 760–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Jahan, A.; Prabha, R.; Chaturvedi, S.; Mathew, B.; Fleming, D.; Agarwal, I. Clinical efficacy and pharmacokinetics of tacrolimus in children with steroid-resistant nephrotic syndrome. Pediatric Nephrol. 2015, 30, 1961–1967. [Google Scholar] [CrossRef]
  15. Yang, E.M.; Lee, S.T.; Choi, H.J.; Cho, H.Y.; Lee, J.H.; Kang, H.G.; Park, Y.S.; Cheong, H.I.; Ha, I.-S. Tacrolimus for children with refractory nephrotic syndrome: A one-year prospective, multicenter, and open-label study of Tacrobell®, a generic formula. World J. Pediatric 2015, 12, 60–65. [Google Scholar] [CrossRef]
  16. Dorresteijn, E.M.; Holthe, J.E.K.-V.; Levtchenko, E.N.; Nauta, J.; Hop, W.C.J.; van der Heijden, A.J. Mycophenolate mofetil versus cyclosporine for remission maintenance in nephrotic syndrome. Pediatric Nephrol. 2008, 23, 2013–2020. [Google Scholar] [CrossRef] [Green Version]
  17. Gellermann, J.; Weber, L.; Pape, L.; Tönshoff, B.; Hoyer, P.; Querfeld, U. Mycophenolate Mofetil versus Cyclosporin A in Children with Frequently Relapsing Nephrotic Syndrome. J. Am. Soc. Nephrol. 2013, 24, 1689–1697. [Google Scholar] [CrossRef] [Green Version]
  18. Kemper, M.J.; Valentin, L.; Van Husen, M. Difficult-to-treat idiopathic nephrotic syndrome: Established drugs, open questions and future options. Pediatric Nephrol. 2017, 33, 1641–1649. [Google Scholar] [CrossRef]
  19. Purohit, S.; Piani, F.; Ordoñez, F.A.; de Lucas-Collantes, C.; Bauer, C.; Cara-Fuentes, G. Molecular Mechanisms of Proteinuria in Minimal Change Disease. Front. Med. 2021, 8, 761600. [Google Scholar] [CrossRef]
  20. Iijima, K.; Sako, M.; Kamei, K.; Nozu, K. Rituximab in steroid-sensitive nephrotic syndrome: Lessons from clinical trials. Pediatric Nephrol. 2017, 33, 1449–1455. [Google Scholar] [CrossRef] [Green Version]
  21. Sinha, R.; Agrawal, N.; Xue, Y.; Chanchlani, R.; Pradhan, S.; Raina, R.; Marks, S.D. Use of rituximab in paediatric nephrology. Arch. Dis. Child. 2021, 106, 1058–1065. [Google Scholar] [CrossRef] [PubMed]
  22. Salvadori, M.; Tsalouchos, A. How immunosuppressive drugs may directly target podocytes in glomerular diseases. Pediatric Nephrol. 2021, 1–11. [Google Scholar] [CrossRef] [PubMed]
  23. Fornoni, A.; Sageshima, J.; Wei, C.; Merscher-Gomez, S.; Aguillon-Prada, R.; Jauregui, A.N.; Li, J.; Mattiazzi, A.; Ciancio, G.; Chen, L.; et al. Rituximab Targets Podocytes in Recurrent Focal Segmental Glomerulosclerosis. Sci. Transl. Med. 2011, 3, 85ra46. [Google Scholar] [CrossRef] [Green Version]
  24. Weiner, G.J. Rituximab: Mechanism of Action. Semin. Hematol. 2010, 47, 115–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Colucci, M.; Carsetti, R.; Serafinelli, J.; Rocca, S.; Massella, L.; Gargiulo, A.; Russo, A.L.; Capponi, C.; Cotugno, N.; Porzio, O.; et al. Prolonged Impairment of Immunological Memory After Anti-CD20 Treatment in Pediatric Idiopathic Nephrotic Syndrome. Front. Immunol. 2019, 10, 1653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Grenda, R.; Jarmużek, W.; Rubik, J.; Piątosa, B.; Prokurat, S. Rituximab is not a “magic drug” in post-transplant recurrence of nephrotic syndrome. Eur. J. Pediatric 2016, 175, 1133–1137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Hansrivijit, P.; Puthenpura, M.M.; Ghahramani, N. Efficacy of abatacept treatment for focal segmental glomerulosclerosis and minimal change disease: A systematic review of case reports, case series, and observational studies. Clin. Nephrol. 2020, 94, 117–126. [Google Scholar] [CrossRef]
  28. Bomback, A.S.; Appel, G.B.; Gipson, D.S.; Hladunewich, M.A.; Lafayette, R.; Nester, C.M.; Parikh, S.V.; Smith, R.J.; Trachtman, H.; Heeger, P.S.; et al. Improving Clinical Trials for Anticomplement Therapies in Complement-Mediated Glomerulopathies: Report of a Scientific Workshop Sponsored by the National Kidney Foundation. Am. J. Kidney Dis. 2021, 71, 570–581. [Google Scholar] [CrossRef]
  29. Muff-Luett, M.; Sanderson, K.R.; Engen, R.M.; Zahr, R.S.; Wenderfer, S.E.; Tran, C.L.; Sharma, S.; Cai, Y.; Ingraham, S.; Winnicki, E.; et al. Eculizumab exposure in children and young adults: Indications, practice patterns, and outcomes—a Pediatric Nephrology Research Consortium study. Pediatric Nephrol. 2021, 36, 2349–2360. [Google Scholar] [CrossRef]
  30. Oosterveld, M.J.; Garrelfs, M.R.; Hoppe, B.; Florquin, S.; Roelofs, J.J.; Heuvel, L.V.D.; Amann, K.; Davin, J.-C.; Bouts, A.H.; Schriemer, P.J.; et al. Eculizumab in Pediatric Dense Deposit Disease. Clin. J. Am. Soc. Nephrol. 2015, 10, 1773–1782. [Google Scholar] [CrossRef] [Green Version]
  31. Holle, J.; Berenberg-Goßler, L.; Wu, K.; Beringer, O.; Kropp, F.; Müller, D.; Thumfart, J. Outcome of membranoproliferative glomerulonephritis and C3-glomerulopathy in children and adolescents. Pediatric Nephrol. 2018, 33, 2289–2298. [Google Scholar] [CrossRef] [PubMed]
  32. Le Quintrec, M.; Lapeyraque, A.-L.; Lionet, A.; Sellier-Leclerc, A.-L.; Delmas, Y.; Baudouin, V.; Daugas, E.; Decramer, S.; Tricot, L.; Cailliez, M.; et al. Patterns of Clinical Response to Eculizumab in Patients with C3 Glomerulopathy. Am. J. Kidney Dis. 2018, 72, 84–92. [Google Scholar] [CrossRef] [PubMed]
  33. Brogan, P.; Yeung, R.S.M.; Cleary, G.; Rangaraj, S.; Kasapcopur, O.; Hersh, A.O.; Li, S.; Paripovic, D.; Schikler, K.; Zeft, A.; et al. Phase IIa Global Study Evaluating Rituximab for the Treatment of Pediatric Patients with Granulomatosis with Polyangiitis or Microscopic Polyangiitis. Arthritis Rheumatol. 2021, 74, 124–133. [Google Scholar] [CrossRef] [PubMed]
  34. Null, N. Etanercept plus Standard Therapy for Wegener’s Granulomatosis. N. Engl. J. Med. 2005, 352, 351–361. [Google Scholar] [CrossRef]
  35. Langford, C.A.; Monach, P.A.; Specks, U.; Seo, P.; Cuthbertson, D.; McAlear, C.A.; Ytterberg, S.R.; Hoffman, G.S.; Krischer, J.P.; Merkel, P.A.; et al. An open-label trial of abatacept (CTLA4-IG) in non-severe relapsing granulomatosis with polyangiitis (Wegener’s). Ann. Rheum. Dis. 2014, 73, 1376–1379. [Google Scholar] [CrossRef]
  36. Gopaluni, S.; Smith, R.; Goymer, D.; Cahill, H.; Broadhurst, E.; Wallin, E.; McClure, M.; Chaudhry, A.; Jayne, D. Alemtuzumab for refractory primary systemic vasculitis—A randomised controlled dose ranging clinical trial of efficacy and safety (ALEVIATE). Arthritis Res. Ther. 2022, 24, 81. [Google Scholar] [CrossRef]
  37. Merrill, J.T.; Shanahan, W.R.; Scheinberg, M.; Kalunian, K.C.; Wofsy, D.; Martin, R.S. Phase III trial results with blisibimod, a selective inhibitor of B-cell activating factor, in subjects with systemic lupus erythematosus (SLE): Results from a randomised, double-blind, placebo-controlled trial. Ann. Rheum. Dis. 2018, 77, 883–889. [Google Scholar] [CrossRef]
  38. Brunner, H.I.; Abud-Mendoza, C.; Mori, M.; Pilkington, C.A.; Syed, R.; Takei, S.; Viola, D.O.; Furie, R.A.; Navarra, S.; Zhang, F.; et al. Efficacy and safety of belimumab in paediatric and adult patients with systemic lupus erythematosus: An across-study comparison. RMD Open 2021, 7, e001747. [Google Scholar] [CrossRef]
  39. Wallace, D.J.; Isenberg, D.A.; Morand, E.F.; Vazquez–Mateo, C.; Kao, A.H.; Aydemir, A.; Pudota, K.; Ona, V.; Aranow, C.; Merrill, J.T. Safety and clinical activity of atacicept in the long-term extension of the phase 2b ADDRESS II study in systemic lupus erythematosus. Rheumatology 2021, 60, 5379–5389. [Google Scholar] [CrossRef]
  40. Huang, L.; Wang, J.; Yang, J.; Zhang, H.; Hu, Y.; Miao, J.; Mao, J.; Fang, L. Impact of Sampling Time Variability on Tacrolimus Dosage Regimen in Pediatric Primary Nephrotic Syndrome: Single-Center, Prospective, Observational Study. Front. Pharmacol. 2022, 12, 726667. [Google Scholar] [CrossRef]
  41. Bergan, S.; Brunet, M.; Hesselink, D.A.; Johnson-Davis, K.L.; Kunicki, P.K.; Lemaitre, F.; Marquet, P.; Molinaro, M.; Noceti, O.; Pattanaik, S.; et al. Personalized Therapy for Mycophenolate: Consensus Report by the International Association of Therapeutic Drug Monitoring and Clinical Toxicology. Ther. Drug Monit. 2021, 43, 150–200. [Google Scholar] [CrossRef] [PubMed]
  42. Zijp, T.R.; Izzah, Z.; Åberg, C.; Gan, C.T.; Bakker, S.J.L.; Touw, D.J.; van Boven, J.F.M. Clinical Value of Emerging Bioanalytical Methods for Drug Measurements: A Scoping Review of Their Applicability for Medication Adherence and Therapeutic Drug Monitoring. Drugs 2021, 81, 1983–2002. [Google Scholar] [CrossRef] [PubMed]
  43. Teisseyre, M.; Cremoni, M.; Boyer-Suavet, S.; Crepin, T.; Benzaken, S.; Zorzi, K.; Esnault, V.; Brglez, V.; Seitz-Polski, B. Rituximab Immunomonitoring Predicts Remission in Membranous Nephropathy. Front. Immunol. 2021, 12, 738788. [Google Scholar] [CrossRef] [PubMed]
  44. Davin, J.-C. The glomerular permeability factors in idiopathic nephrotic syndrome. Pediatric Nephrol. 2015, 31, 207–215. [Google Scholar] [CrossRef] [Green Version]
  45. Bhatia, D.; Sinha, A.; Hari, P.; Sopory, S.; Saini, S.; Puraswani, M.; Saini, H.; Mitra, D.K.; Bagga, A. Rituximab modulates T- and B-lymphocyte subsets and urinary CD80 excretion in patients with steroid-dependent nephrotic syndrome. Pediatric Res. 2018, 84, 520–526. [Google Scholar] [CrossRef]
  46. Kemper, M.J.; Lehnhardt, A.; Zawischa, A.; Oh, J. Is rituximab effective in childhood nephrotic syndrome? Yes and no. Pediatric Nephrol. 2013, 29, 1305–1311. [Google Scholar] [CrossRef]
  47. Faul, C.; Donnelly, M.; Merscher-Gomez, S.; Chang, Y.H.; Franz, S.; Delfgaauw, J.; Chang, J.-M.; Choi, H.Y.; Campbell, K.N.; Kim, K.; et al. The actin cytoskeleton of kidney podocytes is a direct target of the antiproteinuric effect of cyclosporine A. Nat. Med. 2008, 14, 931–938. [Google Scholar] [CrossRef] [Green Version]
  48. Zhang, B.; Shi, W. Is the Antiproteinuric Effect of Cyclosporine A Independent of Its Immunosuppressive Function in T Cells? Int. J. Nephrol. 2012, 2012, 809456. [Google Scholar] [CrossRef] [Green Version]
  49. Büscher, A.K.; Beck, B.B.; Melk, A.; Hoefele, J.; Kranz, B.; Bamborschke, D.; Baig, S.; Lange-Sperandio, B.; Jungraithmayr, T.; Weber, L.T.; et al. Rapid Response to Cyclosporin A and Favorable Renal Outcome in Nongenetic Versus Genetic Steroid– Resistant Nephrotic Syndrome. Clin. J. Am. Soc. Nephrol. 2015, 11, 245–253. [Google Scholar] [CrossRef] [Green Version]
  50. Malakasioti, G.; Iancu, D.; Tullus, K. Calcineurin inhibitors in nephrotic syndrome secondary to podocyte gene mutations: A systematic review. Pediatric Nephrol. 2021, 36, 1353–1364. [Google Scholar] [CrossRef]
  51. Shimada, M.; Araya, C.; Rivard, C.; Ishimoto, T.; Johnson, R.J.; Garin, E.H. Minimal change disease: A “two-hit” podocyte immune disorder? Pediatric Nephrol. 2011, 26, 645–649. [Google Scholar] [CrossRef] [PubMed]
  52. Ishimoto, T.; Shimada, M.; Araya, C.E.; Huskey, J.; Garin, E.H.; Johnson, R.J. Minimal Change Disease: A CD80 podocytopathy? Semin. Nephrol. 2011, 31, 320–325. [Google Scholar] [CrossRef] [PubMed]
  53. Eroglu, F.K.; Orhan, D.; Inözü, M.; Duzova, A.; Gulhan, B.; Ozaltin, F.; Topaloglu, R. CD 80 expression and infiltrating regulatory T cells in idiopathic nephrotic syndrome of childhood. Pediatric Int. 2019, 61, 1250–1256. [Google Scholar] [CrossRef] [PubMed]
  54. Cara-Fuentes, G.; Wasserfall, C.H.; Wang, H.; Johnson, R.J.; Garin, E.H. Minimal change disease: A dysregulation of the podocyte CD80– CTLA-4 axis? Pediatric Nephrol. 2014, 29, 2333–2340. [Google Scholar] [CrossRef] [Green Version]
  55. Garin, E.H.; Mu, W.; Arthur, J.M.; Rivard, C.J.; Araya, C.E.; Shimada, M.; Johnson, R.J. Urinary CD80 is elevated in minimal change disease but not in focal segmental glomerulosclerosis. Kidney Int. 2010, 78, 296–302. [Google Scholar] [CrossRef] [Green Version]
  56. Ling, C.; Liu, X.; Shen, Y.; Chen, Z.; Fan, J.; Jiang, Y.; Meng, Q. Urinary CD80 levels as a diagnostic biomarker of minimal change disease. Pediatric Nephrol. 2014, 30, 309–316. [Google Scholar] [CrossRef]
  57. Yu, C.-C.; Fornoni, A.; Weins, A.; Hakroush, S.; Maiguel, D.; Sageshima, J.; Chen, L.; Ciancio, G.; Faridi, M.H.; Behr, D.; et al. Abatacept in B7-1– Positive Proteinuric Kidney Disease. N. Engl. J. Med. 2013, 369, 2416–2423. [Google Scholar] [CrossRef] [Green Version]
  58. Jayaraman, V.K.; Thomas, M. Abatacept experience in steroid and rituximab-resistant focal segmental glomeruloscle-rosis. BMJ Case Rep. 2016, 2016, bcr2016214396. [Google Scholar] [CrossRef] [Green Version]
  59. Teh, Y.M.; Lim, S.K.; Jusoh, N.; Osman, K.; Mualif, S.A. CD80 Insights as Therapeutic Target in the Current and Future Treatment Options of Frequent-Relapse Minimal Change Disease. BioMed Res. Int. 2021, 2021, 6671552. [Google Scholar] [CrossRef]
  60. Clement, L.C.; Avila-Casado, C.; Macé, C.; Soria-Castro, E.; Bakker, W.W.; Kersten, S.; Chugh, S.S. Podocyte-secreted angiopoietin-like-4 mediates proteinuria in glucocorticoid-sensitive nephrotic syndrome. Nat. Med. 2010, 17, 117–122. [Google Scholar] [CrossRef] [Green Version]
  61. Cara-Fuentes, G.; Segarra, A.; Silva-Sanchez, C.; Wang, H.; Lanaspa, M.A.; Johnson, R.J.; Garin, E.H. Angiopoietin-like-4 and minimal change disease. PLoS ONE 2017, 12, e0176198. [Google Scholar] [CrossRef] [PubMed]
  62. Macé, C.; Chugh, S.S. Nephrotic Syndrome: Components, Connections, and Angiopoietin-Like 4–Related Therapeutics. J. Am. Soc. Nephrol. 2014, 25, 2393–2398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Thurman, J.M.; Nester, C.M. All Things Complement. Clin. J. Am. Soc. Nephrol. 2016, 11, 1856–1866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Angeletti, A.; Reyes-Bahamonde, J.; Cravedi, P.; Campbell, K.N. Complement in Non-Antibody-Mediated Kidney Diseases. Front. Med. 2017, 4, 99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Koopman, J.J.E.; van Essen, M.F.; Rennke, H.G.; de Vries, A.P.J.; van Kooten, C. Deposition of the Membrane Attack Complex in Healthy and Diseased Human Kidneys. Front. Immunol. 2021, 11, 599974. [Google Scholar] [CrossRef]
  66. Mastrangelo, A.; Serafinelli, J.; Giani, M.; Montini, G. Clinical and Pathophysiological Insights into Immunological Mediated Glomerular Diseases in Childhood. Front. Pediatric 2020, 8, 205. [Google Scholar] [CrossRef]
  67. Akamine, K.; Punaro, M. Biologics for childhood systemic vasculitis. Pediatric Nephrol. 2018, 34, 2295–2309. [Google Scholar] [CrossRef]
  68. Mundel, P.; Greka, A. Developing therapeutic ‘arrows’ with the precision of William Tell. Curr. Opin. Nephrol. Hypertens. 2015, 24, 388–392. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Evolution of therapies used in pediatric immune-mediated kidney diseases.
Figure 1. Evolution of therapies used in pediatric immune-mediated kidney diseases.
Children 09 00536 g001
Table 1. Summary characteristics of the “classic” drugs used in pediatric immune-mediated kidney diseases [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17].
Table 1. Summary characteristics of the “classic” drugs used in pediatric immune-mediated kidney diseases [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17].
DrugSystemic
Target/Mechanism
Local (Kidney)
Target/Mechanism
DiseasesGenerational
Line
of Therapy
Steroids
(including ACTH)
Induce downregulation of pro-inflammatory genes by
binding to a specific cytoplasmic glucocorticoid
receptor (GR);
regulate gene expression by binding to glucocorticoid
response elements (GRE) on DNA
Attenuation of podocyte apoptosis
Blocking of TRPC6 signal pathway
Blocking of CD80 upregulation
Nephrotic syndrome
All other types of immune-mediated glomerulonephritis
first
CyclophosphamideExerts cytotoxic effect by cross-linking of strands of DNA and RNA, and by inhibition of protein synthesisnoneNephrotic syndrome
Lupus nephritis
Vasculitis with renal involvement
IgAN
second
AzathioprineActs by the incorporation into replicating DNA; blocks de novo pathway of purine synthesis noneIgAN
AAV
second
Mycophenolate
mofetil
Mycophenolic acid (MPA), a drug derivative, acts as
a selective, noncompetitive inhibitor of inosine monophosphate dehydrogenase IMPDH); inhibits T- and B-lymphocyte proliferation
Decrease of uPAR expression
Modification of ATP depletion (by inhibiting IMPDH) in podocytes
Nephrotic syndrome
Lupus nephritis
Vasculitis with renal involvement
IgAN
second
Cyclosporine AInhibits the phosphatase activity of calcineurin, which regulates nuclear translocation and subsequent activation of NFAT transcription factors;
CsA is a specific inhibitor of T cell activation
Stabilization of actin cytoskeleton by preserving a phosphorylation-dependent synaptopodin-14-3-3-ẞ integrin interaction
Decrease of TRPC6 and ANGPTL4 upregulation
Nephrotic syndromesecond
TacrolimusInhibits T lymphocyte activation and transcription of cytokine genes, including the gene for interleukin-2Decrease of TRPC6 and ANGPTL4 upregulationNephrotic syndromethird
(off-label)
ACTH—adrenocorticotrophic hormone. NFAT—nuclear factor of activated T-cells.
Table 2. Summary characteristics of the biologic drugs used in pediatric immune-mediated kidney diseases [20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39].
Table 2. Summary characteristics of the biologic drugs used in pediatric immune-mediated kidney diseases [20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39].
DrugSystemic
Target/Mechanism
Local (Kidney)
Target/Mechanism
DiseasesGenerational
Line
of Therapy
rituximabBinds to CD20 on B cells and mediates B-cell lysis and depletionstabilizes podocytes cytoskeleton by regulation (preservation) of sphingomyelin phosphodiesterase acid-like 3b (SMLPD-3b), a protein participating in the podocyte cytoskeleton activityNephrotic syndrome
AAV
GPA
MPA
IgA vasculitis
Membranous nephropathy
third
(off-label)
Clinical trials
(adults and
adolescents)
Case reports
eculizumabBinds to complement protein C5, inhibiting its cleavage to C5a and C5b; this prevents a generation of the terminal complement complex C5b-9noneAtypical hemolytic uremic syndrome (aHUS)
Lupus nephritis
C3 nephropathy
MPGN type I
DDD
first
third (off label)
second/third
(off-label)
abataceptA fusion molecule including Fc region of IgG1 and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4-Ig)
Blocks CTLA-4 present on Tregs
Blocking of B7-1 signalling and restoration of ẞ1 integrin activationNephrotic syndrome
GPA (Granulomatosis with Polyangiitis)
Takayasu arteritis
fourth
(off-label)
Clinical trials
(adults and
adolescents)
Case reports
infliximab
etanercept
Anti-TNFα moab
Human Fc fusion protein- blocks the TNFα -receptor
noneAAV
Takayasu disease
PAN
Clinical trials
(children)
Case reports
alemtuzumabAnti-CD52 depleting moab, lysis of all cells expressing CD52noneAAVClinical trials
(adults)
Case reports
tocilizumabAnti-IL6 moabnoneKawasaki diseaseChildren (case
reports)
anakinraAnti-IL-1 moabnoneKawasaki diseaseClinical trial
(children)
blisibimodBlocks of B-cell activating factor (BAFF)noneSLEClinical trial
(adults)
belimumabAnti-BAFF moabnoneSLEClinical trials
(adults and
children)
ataciceptA recombinant human fusion protein that binds to BLyS (B lymphocyte stimulator) and APRIL (a proliferation-inducing ligand); inhibits interactions with their specific receptorsnoneSLEClinical trial
(adults)
AAV—ANCA-associated renal vasculitis. ANGPTL4—angiopoietin-like 4 molecule. APRIL—a proliferation-inducing ligand. BAFF—B-cell activating factor. BlyS—B lymphocyte stimulator. B7-1 (CD80)—a molecule expressed on the T cells, dendritic cells and podocytes surface. CTLA-4 (cytotoxic T-lymphocyte-associated protein-4)—molecular co-factor expressed on podocytes and Tregs. DDD—dense deposit disease. GPA—granulomatosis with polyangiitis. IMPDH—inosine 5′-monophosphate dehydrogenase. moab—monoclonal antibody. MPA—microscopic polyangiitis. MPGN—membrano-proliferative glomerulonephritis. PAN—polyarteritis nodosa. TRPC6—transient receptor potential channel 6. SLE—systemic lupus erythromatosus.
Table 3. Clinical background of TDM (therapeutic drug monitoring) and immunomonitoring [40,41,42,43].
Table 3. Clinical background of TDM (therapeutic drug monitoring) and immunomonitoring [40,41,42,43].
RuleClinical Practice
There is an association between the dose and drug concentration in the blood/plasma.The activity of drug-specific metabolic pathways is programmed genetically and also is age-dependent.
Pharmacokinetics and pharmacodynamics of the drug depend on function of the specific pathways and organs (routes of drug metabolism and clearance).There is an association between drug concentration and its clinical efficacy and toxicity.
Specific drugs administered simultaneously may interact, and this reaction changes its’ metabolism and pharmacokinetics.Multidrug management requires more frequent TDM.
The pharmacokinetic effect of the biologic drug is expressed as a number of targeted cells.There is an association between the number of target cells and the clinical course of the disease.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Grenda, R.; Obrycki, Ł. Second and Third Generational Advances in Therapies of the Immune-Mediated Kidney Diseases in Children and Adolescents. Children 2022, 9, 536. https://doi.org/10.3390/children9040536

AMA Style

Grenda R, Obrycki Ł. Second and Third Generational Advances in Therapies of the Immune-Mediated Kidney Diseases in Children and Adolescents. Children. 2022; 9(4):536. https://doi.org/10.3390/children9040536

Chicago/Turabian Style

Grenda, Ryszard, and Łukasz Obrycki. 2022. "Second and Third Generational Advances in Therapies of the Immune-Mediated Kidney Diseases in Children and Adolescents" Children 9, no. 4: 536. https://doi.org/10.3390/children9040536

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop