Next Article in Journal
Antimicrobial Nano-Zinc Oxide Biocomposites for Wound Healing Applications: A Review
Next Article in Special Issue
RETRACTED: An Innovative Tool for Evidence-Based, Personalized Treatment Trials in Mucopolysaccharidosis
Previous Article in Journal
Effects of Dendrimer-microRNA Nanoformulations against Glioblastoma Stem Cells
Previous Article in Special Issue
Decreased Levels of Chaperones in Mucopolysaccharidoses and Their Elevation as a Putative Auxiliary Therapeutic Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmacologic Management of Monogenic and Very Early Onset Inflammatory Bowel Diseases

1
Division of Gastroenterology, Seattle Children’s Hospital, Seattle, WA 98105, USA
2
Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA
3
Department of Pharmacy, Seattle Children’s Hospital, Seattle, WA 98105, USA
*
Author to whom correspondence should be addressed.
Pharmaceutics 2023, 15(3), 969; https://doi.org/10.3390/pharmaceutics15030969
Submission received: 23 January 2023 / Revised: 10 March 2023 / Accepted: 14 March 2023 / Published: 17 March 2023
(This article belongs to the Special Issue Novel Therapeutic Approaches in Rare Genetic Diseases)

Abstract

:
Inflammatory bowel disease (IBD) is treated with a variety of immunomodulating and immunosuppressive therapies; however, for the majority of cases, these therapies are not targeted for specific disease phenotypes. Monogenic IBD with causative genetic defect is the exception and represents a disease cohort where precision therapeutics can be applied. With the advent of rapid genetic sequencing platforms, these monogenic immunodeficiencies that cause inflammatory bowel disease are increasingly being identified. This subpopulation of IBD called very early onset inflammatory bowel disease (VEO-IBD) is defined by an age of onset of less than six years of age. Twenty percent of VEO-IBDs have an identifiable monogenic defect. The culprit genes are often involved in pro-inflammatory immune pathways, which represent potential avenues for targeted pharmacologic treatments. This review will provide an overview of the current state of disease-specific targeted therapies, as well as empiric treatment for undifferentiated causes of VEO-IBD.

1. Introduction

Inflammatory bowel disease (IBD), including ulcerative colitis (UC), Crohn’s disease (CD), and IBD-unspecified (IBD-U), is a chronic, relapsing intestinal disease characterized by inflammation. The pathogenesis involves a complex interplay of the intestinal immune system and gut microbiome, as well as environmental factors. IBD is a heterogeneous disease with a wide variety of presentations. The majority of patients have what is considered polygenic IBD, in that their disease is a result of the interplay of many genes with environmental factors. Patients who present before the age of six years are designated as having very early onset IBD (VEO-IBD), which can be further subclassified into infantile IBD, for children diagnosed before two years of age, and neonatal IBD, for those diagnosed before 28 days of life [1]. In contrast to older children and adolescents with polygenic IBD, patients with VEO-IBD often have recalcitrant disease course and are more likely to have a causative monogenic defect underlying their disease [1,2]. Recent studies suggest that a monogenic immunodeficiency may be identified in 20–30% of patients with VEO-IBD (Table 1) [3,4]. These patients are usually identified during early life. In an increasing number of cases, these monogenic immunodeficiencies can be treated with targeted pharmacologic agents and other therapies, such as stem cell transplantation. The armamentarium of biologic and small molecule therapies continues to grow, which allows for more narrowly targeted treatments even in undifferentiated VEO-IBD. As the incidence of pediatric IBD, and with it VEO-IBD, continues to rise, identifying patients with monogenic immunodeficiencies becomes ever more urgent to target treatment and minimize medication toxicities [5,6]. This review will provide an overview of the current state of pharmacologic therapies for monogenic IBD, as well as empiric therapies for undifferentiated VEO-IBD.

2. Methods

A comprehensive search of MEDLINE limited to English language articles was performed using PubMed (http://pubmed.ncbi.nlm.gov) accessed on 4 January 2023. The following search terms were used: “very early onset inflammatory bowel disease”, “monogenic inflammatory bowel disease”, as well as gene names, syndrome names, and medication names (both brand and generic). Articles were used if they were peer-reviewed and included clinical information about monogenic or very early onset IBD cases, or if they discussed the use of a medication to target a specific immunologic pathway.

3. Monogenic Inflammatory Bowel Diseases

Currently, over 100 disease-associated genes have been identified [1,7,8]. Causative immunodeficiencies can be classified as epithelial cell defects, phagocytic defects, defects of adaptive immunity, T regulatory defects, IL-10 pathway disorders, and hyperinflammatory or autoinflammatory conditions [3,9,10,11,12] (Table 1). Monogenic defects of the epithelium often involve the gut and the skin due to shared embryologic precursors. Defects of phagocytosis lead to defective degranulation, as in chronic granulomatous disease (CGD) [13,14], or impaired migration of neutrophils into the tissue in leukocyte adhesion disorder (LAD) [15,16,17]. T or B cell defects that result in severe combined immunodeficiency (SCID) or common variable immunodeficiency (CVID) often present with an IBD-like phenotype [18,19,20,21,22]. Wiskott-Aldrich syndrome [23], or X-linked agammaglobulinemia [24], can also have an IBD-like component. IBD is a common presentation of T regulatory defects, such as immunodysregulation, polyendocrinopathy, enteropathy X-linked syndrome (IPEX), or IPEX-like syndromes [25,26,27,28,29,30]. Hyperinflammatory conditions, such as X-linked inhibitor of apoptosis (XIAP), often present with a severe perianal Crohn’s disease phenotype [31,32]. Finally, loss of function mutations in the IL-10 ligand and receptor can cause a broad spectrum of IBD-like disease [33,34,35].

4. Diagnosing Monogenic Inflammatory Bowel Disease

As more genes are identified and new therapeutics continued to be developed, a high level of suspicion for monogenic disorders should be maintained in patients with both VEO-IBD and severe refractory disease at older ages. Appropriate testing should be performed with the assistance of a multidisciplinary team, including pediatric gastroenterologists, geneticists, and immunologists [12]. Other team members may include dieticians, psychologists, and social workers to support the complex needs of these patients and their families. A thorough evaluation should be performed in these patients, including a comprehensive history with focus on family and genetic history, physical exam, blood work and stool studies, endoscopy, histology, and genetic work up. First-line laboratory evaluation should include a complete blood count (CBC) with differential, comprehensive metabolic panel, inflammatory markers with C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR), and stool evaluation to rule out infection. Several diagnostic algorithms now exist to guide immunologic work up, which may include immunoglobulin levels, vaccine response titers, and flow cytometry of B, T, and NK cell populations. Specific disorders may be diagnosed via neutrophil oxidative burst (CGD), gene panels, or targeted flow cytometry (IPEX due to FOXP3 mutation, XIAP), or cytokine panels [3,12]. Upper endoscopy and colonoscopy with histologic evaluation is a standard diagnostic evaluation in cases of suspected IBD. Findings in the VEO and monogenic IBD population are often non-specific, although the presence of certain histologic findings, including apoptosis, severe chronic architectural changes, villous blunting, and abundance of eosinophils, should increase the index of suspicion for a monogenic disorder [9,36].
Currently, approximately 20% of children with VEO-IBD have an underlying genetic disorder identified, although a recent study of 207 patients with VEO-IBD aimed at defining the role of next generation sequencing showed that a molecular diagnosis was achieved 32% of the time [4]. If patients primarily had small bowel inflammation, the yield increased to 61%. For those patients with colitis and perianal lesions, the yield was 39%, and it was 18% for colitis only [4]. Monogenic IBD has also been found with increased prevalence in older children and adolescents with severe, refractory IBD, with some cohorts finding monogenic disorders in up to 30% of older patients with refractory disease [37]. In the future, there is potential for the use of polygenic risk scores (PRS) to predict risks of disease progression and severity in these patients. Adult studies have shown that PRS can help identify patients at risk of developing stenotic disease requiring ileocecal resection or primary sclerosing cholangitis (Voskuil 2021 930) [38]. Another PRS replicated the association of two VEO-IBD genes, ADAM17 and LRBA, and showed that heterozygous carriage of a specific mutant LRBA allele is also associated with significantly decreased LRBA and CTLA-4 expression with T-cell activation [39]. Currently, the applicability of PRS is limited by low discriminative accuracy due to small population sizes, and they cannot account for environmental factors or microbiome interplay.
As monogenic disorders are inborn errors of immunity leading to dysfunction in specific immune pathways with known alterations in cytokine activity, treatment teams are increasingly choosing targeted pharmacologic agents, rather than empiric IBD therapies in these patients. In the absence of a treatment-guiding diagnosis, children with undifferentiated VEO-IBD are treated empirically with corticosteroids, 5-aminosalicylates, immunomodulators, biologics, small molecules, and antibiotics [10,40]. Severe, refractory disease is more common in VEO-IBD [3], and management often includes non-pharmacologic therapies, including nutrition and surgery. In some cases, children with underlying monogenic defects also benefit from hematopoietic stem cell transplant (HSCT) to cure their immunodeficiency [41].

5. Targeted Therapies for Monogenic IBD

5.1. Anti-TNF Antibodies

Monoclonal antibodies against tumor necrosis factor alpha (TNFα) include infliximab, adalimumab, and golimumab. Infliximab has been trialled to target TNF-driven IBD-like inflammation seen in X-linked ectodermal dysplasia and immunodeficiency, an epithelial cell defect secondary to mutations in IKBKG/NEMO, in which defective NF-κB activation impairs immune response to circulating TNF [42], as well as Hermansky-Pudlak syndrome, a hyperinflammatory disorder involving defective cellular trafficking of lysosome-related organelles [43]. Studies of infliximab in these disorders have not been performed, but case reports show promise. A patient with X-linked ectodermal dysplasia showed good response in one report [44], and another with Hermansky-Pudlak syndrome and Crohn’s-like colitis remained in long-term remission at 22 months of treatment [45]. In another case report, an adult patient with incomplete IPEX syndrome who was treated with infliximab had improvement in clinical symptoms and inflammatory markers, as well as increase in circulating FOXP3+ CD4+ T regulatory cells, suggesting a role for anti-TNF therapy in potentiating T regulatory cell expansion and activity in this disorder [46]. However, infliximab should not be used in patients with chronic granulomatous disease, as it has been linked with severe infections and death [47].

5.1.1. Pharmacologic Considerations and Therapeutic Drug Monitoring for Anti-TNFα Therapy

Anti-TNFα agents are widely used as first-line biologic therapy in undifferentiated pediatric IBD and VEO-IBD. Infliximab has a remission rate of 60% by week 10 of treatment in children, with up to 65% maintaining remission at one year [48]. The clinical response to infliximab is variable in both adults and children, and therefore the role of therapeutic drug monitoring (TDM) has become the standard of care in IBD across both populations [49]. It is expected that up to one third of patients will require dose escalation within the first year of treatment due to secondary loss of response [50]. Infliximab requires therapeutic drug level monitoring to ensure that patients maintain therapeutic levels and to monitor for development of anti-infliximab antibodies that can lead to loss of response [51]. Younger children also have increased clearance of infliximab and may require higher doses or shorter intervals to maintain therapeutic levels [51,52,53,54]. Concurrent use of an immunomodulator, such as azathioprine or methotrexate, has been shown to reduce immunogenicity, leading to higher drug levels, reduced antibody formation, and endoscopic improvement [55,56]. However, the use of azathioprine in particular as combination therapy has been linked to the development of uniformly fatal hepatosplenic T-cell lymphoma primarily in teenage males, and thus duration of therapy is often limited to two years or less [10].
There is no consensus on the role of proactive TDM as compared to reactive monitoring; however, the literature supports the role of TDM in maintaining optimal treatment effect [57]. Recent recommendations from the ECCO-ESPGHAN group suggest early proactive TDM to optimize drug dosing [49]. Trough levels are typically checked during the maintenance phase of therapy, as compared to the induction phase. A trough level of >5 mcg/mL is preferred. However, in perianal fistulizing disease, a target trough level of >12.7 mcg/mL has been correlated with better clinical response (Table 2).
Recommendations regarding the use of adalimumab in undifferentiated pediatric IBD suggest that it can also be considered as a first line-agent. It remains an effective option in both biologic naïve patients, as well as those who have had previous biologic exposure. Trough levels are checked typically during the maintenance of treatment, with a goal level of at least 7.5 mcg/mL recommended for endoscopic healing by week eight of therapy [49] (Table 2).

5.1.2. Time to Therapeutic Effect

Anti-TNFs can require up to 12 weeks to see full clinical effect, and response can be variable and patient specific; however, data in the adult population suggest that up to 81% of patients with Crohn’s disease will have evidence of clinical response after four weeks of infliximab therapy [77,78,79]. Adult response to adalimumab varies from 12 to 15 weeks in Crohn’s disease, and as early as four weeks in the UC population [79,80,81]. In pediatric UC patients, one study showed that up to 75% of patients with effective response to infliximab treatment by week eight of therapy [82]. However, loss of response is quite common, with up to one third of patients on infliximab experiencing secondary loss of response [50]. For pediatric patients treated with adalimumab, clinical remission rates range from two weeks to two years [83].

5.2. Vedolizumab

Vedolizumab is a gut-specific anti-α4β7 integrin monoclonal antibody that prevents migration of T cells into colonic tissue, thereby reducing intestinal inflammation. It has been used with varying degrees of success for certain monogenic IBD variants, including chronic granulomatous disease and CTLA4 deficiency. There is a report of sustained remission of CGD colitis and perianal disease in an adult [84]; however, the pediatric data are less promising. In a study of 11 children, more than half had symptomatic improvement and many were able to wean steroids, but none achieved mucosal healing after six months of treatment [85]. Vedolizumab has also been trialled for CTLA4 deficiency, and in a case report of a single adult patient, there was sustained remission; however, pediatric studies of this disorder are lacking, and it is not generally recommended as a first-line agent in this disorder [86]. It is, however, safe and effective in undifferentiated VEO-IBD with a 56% clinical response rate by the fourth dose [87].

5.2.1. Pharmacologic Considerations and Therapeutic Drug Monitoring for Vedolizumab

Vedolizumab has a low incidence of adverse drug reactions reported in the literature, and anti-drug antibody production is relatively rare [49,88] (Table 3). From a TDM perspective, there is not clear guidance on optimal trough levels or when to check them. In one pediatric study, average trough levels of 32.1 mcg/mL were observed at week two of therapy and 29.9 mcg/mL at week six in a cohort of 22 pediatric patients. When this population was further delineated by type of IBD, trough levels in patients with UC/IBD-U were higher than in those with CD [66] (Table 2).

5.2.2. Time to Therapeutic Effect

Pediatric studies indicate that vedolizumab may be more effective in UC compared to CD, and patients without prior biologic exposure and less severe disease may have higher rates of both mucosal healing and clinical remission [104]. One study showed that response to vedolizumab may take up to 16 weeks, with a range of therapeutic onset between eight and twelve weeks. In a pediatric retrospective study, 76% of those with UC compared to 42% of those with CD achieved clinical remission after 14 weeks of treatment [105] (Table 4).

5.3. Ustekinumab

Ustekinumab is a humanized monoclonal anti-IL-12/23 antibody used in pediatric patients who are non-responsive to anti-TNF medications. While it has been shown to be safe and efficacious in both biologic-exposed and naïve patients, its use in the VEO-IBD and monogenic IBD population is limited, and there are no large-scale trials [112,113]. A single case report in a patient with VEO-IBD due to Loeys-Dietz syndrome reported clinical remission with a trough level of 6 mcg/mL after 18 months of treatment [68]. The authors posited that the patient’s colitis could have been the result of overactivation of TH17 cells secondary to the underlying mutation in TGF-β, resulting in high levels of IL-23.

Pharmacologic Considerations and Therapeutic Drug Monitoring for Ustekinumab

Studies have shown that ustekinumab is generally well tolerated, but it still holds an increased risk of infection and hypersensitivity reactions during administration (Table 3). When evaluating the use of ustekinumab in pediatric IBD, there has been some evidence to show better response and higher rates of clinical remission in patients who are biologic naïve [113]. In a recent retrospective study conducted in the pediatric population, 64% of patients achieved biomarker remission at the 52-week mark, with 76% of patients still remaining on ustekinumab at the end of the study period. From this cohort of patients, 58% achieved steroid free clinical remission [113]. In adult trials, clinical response was seen after eight weeks of treatment, with some data to suggest a duration of up to six months needed for endoscopic remission [67] (Table 4). In the future, these patients may also be candidates for treatment with selective anti-IL-23 agents, including risankizumab, which is approved for adults with Crohn’s disease, and mirikizumab, which is approved for adults with ulcerative colitis.

5.4. Janus Kinase Inhibitors

Ruxolitinib and tofacitinib inhibit JAK1/2 and JAK1/3, respectively, and via this pathway downregulate several inflammatory cytokines, including IL-6, IL-11, IL-4, IL-7, and IL-9 [51]. There have been several case studies showing good response to these agents in patients with mutations in both JAK and STAT genes, which lead to IPEX-like syndromes [114]. A patient with a STAT3 gain-of-function mutation who was refractory to multiple previous therapies had sustained remission at one year on ruxolitinib [115]. IL2RA mutations are another target for Janus kinase inhibitors, as a key component of the IL-2R signaling pathway is encoded by the STAT5B gene [116,117]. In vitro exposure to tofacitinib of resected colonic tissue from a patient with refractory colitis due to IL2RA IPEX-like syndrome led to reduction in IL-2 and IFNγ secretion [118].

Pharmacologic Considerations and Therapeutic Drug Monitoring for JAK Inhibitors

Janus kinase inhibitors are small molecules and thus do not have the same risk of loss of response due to immunogenicity as the anti-TNFs. They are available in oral form and do not require drug level monitoring. Tofacitinib is approved for use in adults with ulcerative colitis; off-label use in pediatric UC also shows promise [119]. In a single-center, retrospective study, Tofacitinib induced clinical remission in nearly 50% of tested patients after the 12-week induction period (N = 9/21). After 52 weeks, approximately 40% of subjects achieved steroid free remission (N = 7/17) [119]. Of note, 67% of patients included in this study had ulcerative colitis, 14% had Crohn’s disease, and 19% had indeterminant IBD. More selective agents, such as upadicitinib, a JAK1 inhibitor, are coming to market, but they are not yet approved for use in children. Risks associated with the use of JAK inhibitors include anemia due to inhibition of erythropoietin signaling and venous thromboembolism [120]. From a monitoring standpoint, patients receiving JAK inhibitors should have lipids, CBC, renal function, and liver function assessed prior to initiating therapy and periodically while on maintenance (Table 3) [102,103].

5.5. IL-1 Antagonists

There are two recombinant IL-1 inhibitors available, canakinumab and anakinra, both of which act by competitive inhibition of the IL-1 receptor. IL-1 release is triggered by activation of the inflammasome, and it leads to a proinflammatory cascade [121,122]. Several monogenic disorders involve activation of the inflammasome and subsequent IL-1 secretion, including CGD and mevalonate kinase deficiency (MKD). Patients with chronic granulomatous disease have defective autophagy, which has been linked to dysregulated activation of the inflammasome [123]. A small number of case studies using anakinra in patients with CGD enterocolitis have shown a modest response, though with low rates of long-term remission [123,124]. Successful use of anakinra ranging from partial symptomatic improvement to remission has been reported in several case series of infants with MKD, an autoinflammatory disorder that often presents in infancy with IBD-like enterocolitis, and it also involves inflammasome-driven IL-1β secretion [125,126]. In a recent study, a cohort of nineteen children with VEO-IBD (approximately 50% with infantile onset, 42% with Crohn’s disease, and 58% with unclassified IBD) received canakinumab with some success. These patients had an autoinflammatory phenotype without monogenic disease and were treated with canakinumab therapy for at least six months. At baseline, close to 40% of patients were biologic naïve; in 74% of the studied population, canakinumab was used in dual therapy with either anti-TNF alpha blockers, vedolizumab, ruxolitinib, ustekinumab, or rapamycin. Clinical remission was achieved in 32% of patients after six months of treatment, and clinical response was achieved in 89% of patients (17/19) [127,128]. Additionally, in a 2020 study, which included nine pediatric patients with Crohn’s disease, one patient received dual therapy with infliximab and anakinra. However, the outcome in this patient was difficult to assess based on reported results (Goyal 2020 S122) [129].
There is also preliminary data to suggest that IL-1 antagonists may be useful in defects of the IL-10 pathway. IL-10 is an immunoregulatory cytokine, which mediates several anti-inflammatory pathways via its receptor. Mutations in the receptor encoding genes IL10RA and IL10RB can lead to the development of severe, refractory infantile VEO-IBD [34,130]. Anakinra has been used as a steroid-sparing agent to bridge a small number of patients with IL10RA mutations to stem cell transplant, based on work showing that IL-10 deficient macrophages produce higher levels of IL-1b and lead to downstream inflammasome activation [131].

Pharmacologic Considerations and Therapeutic Drug Monitoring for IL-1 Inhibitors

Anakinra and canakinumab both require baseline screening for TB and hepatitis B in addition to baseline and periodic hematologic, renal, and hepatic monitoring. There is no drug level monitoring required (Table 3).

5.6. Abatacept

Abatacept is a cytotoxic T lymphocyte antigen-4 (CTLA4) immunoglobulin fusion drug. CTLA4 is an inhibitory immune checkpoint protein that is expressed on FOXP3+ T-regulatory cells. Abatacept is used as a targeted therapy for LRBA deficiency, which is marked by very low CTLA4 expression and presents with enteropathy and other systemic manifestations [3,11,12,132]. In a study of 22 children with LRBA deficiency who were treated with abatacept, 11 achieved complete remission, and three had partial remission. The majority were also able to stop steroids and other immunosuppressive agents [133]. Abatacept was also used with some success in the case of a child who developed post-heart transplant autoimmune enteropathy consistent with Omenn syndrome, marked by the presence of autoreactive oligoclonal T cells [134]. Overall, the use of abatacept in VEO-IBD has been quite limited and overall remission rates are not reported.

Pharmacologic Considerations and Therapeutic Drug Monitoring for Abatacept

Patients who are initiated on abatacept therapy require baseline TB and hepatitis B screening, in addition to assessment for increased risk of infection and possible hypersensitivity reactions. No drug level monitoring is required (Table 3) [101].

5.7. IL-18 Antagonists

IL-18 is another downstream product of the proinflammatory cascade produced by inflammasome activation. IL-18 production is a hallmark of several monogenic immunodeficiencies that produce IBD symptoms, including macrophage activation syndrome (MAS) secondary to NLRC4 mutation, as well as mutations in X-linked inhibitor of apoptosis (XIAP). NLRC4 encodes a key component of the inflammasome complex. Patients with this mutation develop macrophage activation syndrome and severe neonatal-onset enterocolitis [135,136]. This disease is marked by very high levels of IL-18. There are not currently any IL-18 antagonists approved for use in humans; however, there are ongoing clinical trials of several recombinant IL-18 binding proteins (rhIL-18BP). These have occasionally been made available by compassionate use protocols for patients with IL-18-related disorders [137]. There is a pediatric phase II trial of MAS825, an anti-IL-1b/IL-18 monoclonal antibody, for patients with NLRC4 gain-of-function mutations (US NLoM NCT04641442). XIAP also involves inflammasome activation and significant elevation in IL-18 [10]. There is an ongoing phase III trial at Cincinnati Children’s Hospital studying the rhIL-18BP drug tadekinig alfa in patients with both XIAP and NCLR4 mutations (US NloM NCT03113760). As these medications are still experimental, there is no information available about side effects, screening, or drug level monitoring.

6. Empiric Therapies

Therapy for inflammatory bowel diseases historically involved targeting immune dysregulation through systemic immunosuppression using glucocorticoids, non-specific immunomodulators, such as azathioprine and methotrexate, and calcineurin inhibitors. In recent years, the biologic therapies and small molecules that target specific immune pathways previously discussed have reduced reliance on these medications.

6.1. Immunomodulators

The immunomodulators azathioprine (AZA) and methotrexate can be used as monotherapy or in combination with biologics to treat IBD. AZA blocks de novo purine synthesis and thus arrests the development of immune cells. Methotrexate is a folate antagonist, which exerts its immunomodulatory action via several pathways: inhibition of purine and pyrimidine synthesis, production of reactive oxygen species, and modulation of cytokine production [138]. In one case report, an infant who presented with neonatal liver failure and refractory IBD-like pancolitis due to a pathogenic cytosolic isoleucyl-tRNA synthetase mutation showed improvement after initiation of subcutaneous methotrexate. CD4+ memory T-cell secretion of proinflammatory cytokines, including IL-2, IL-5, IL-9, and IL-13 were elevated in this patient [139]. Methotrexate has been shown to be an inhibitor of cytokine production in activated T cells in in vitro studies of patients with rheumatoid arthritis, which suggests a possible mechanism for the noted improvement [140]. One meta-analysis showed that, for patients with Crohn’s disease, 37% of the studied pediatric population was able to achieve 12 month remission with methotrexate monotherapy [56]. In a retrospective study conducted in Canada, investigators evaluated the use of methotrexate monotherapy in pediatric patients with Crohn’s disease, ulcerative colitis, and indeterminate colitis. Clinical remission was achieved in 16% of the Crohn’s disease group compared to 7% in patients with UC or IBD-U. Overall, long-term remission rates with methotrexate in this population was low [141]. When evaluating data supporting the use of azathioprine, the use of genetic testing becomes important due to the role of thiopurine methyltransferase, which is required for conversion of AZA into its active metabolites. Reduced activity of this enzyme can result in increased risk of myelosuppression and hepatotoxicity. Out of 41 pediatric patients evaluated in one retrospective study conducted in England, 12 patients required dose increases to achieve clinical remission. A total of 28 patients did not require AZA dose adjustments and achieved [142].

6.1.1. Pharmacologic Considerations and Therapeutic Drug Monitoring for Immunomodulators

Young children with VEO-IBD may require higher doses of AZA to achieve therapeutic levels, which leads to increased risk of hepatoxicity and bone marrow failure [10,143]. As previously mentioned, azathioprine therapy has also been linked with hepatosplenic T-cell lymphoma, and its use for longer than two years is not recommended [10].

6.1.2. Time to Therapeutic Effect

When evaluating time to clinical response, adult data have shown that in Crohn’s disease it may take between two weeks and nine months to achieve clinical remission with azathioprine [79,106]. In pediatric IBD, maximum effectiveness in Crohn’s disease has been reported to take between eight and sixteen weeks [49] (Table 4).

6.2. Calcineurin Inhibitors

Calcineurin inhibitors (cyclosporine A and tacrolimus) work by suppressing transcription of IL-2, TNFα, and interferon-c in T cells, and have been used for induction of remission in steroid-refractory pediatric IBD [144,145]. While pediatric studies have shown response rates of 60–80% in older children, their use in the VEO-IBD population has had mixed results [146]. One review of XIAP patients treated with tacrolimus found that 92% of patients were refractory to treatment with combination corticosteroids and tacrolimus, as well as cyclosporine and AZA [147].

6.2.1. Pharmacologic Considerations and Therapeutic Drug Monitoring for Calcineurin Inhibitors

Tacrolimus and cyclosporine have been used to treat severe colitis in children and guidelines suggest their use as an alternative second line agent after failure to respond to steroids [71]. These medications are typically used to bridge to agents with a longer time to effect, such as vedolizumab, ustekinumab, or thiopurines. Tacrolimus has been dosed as 0.1 mg/kg orally every 12 h as part of induction therapy, targeting a goal trough level of 10–15 ng/mL [71]. One study reported decreasing the goal trough goal to 5–7 ng/dL once therapeutic remission was achieved [71]. Cyclosporine can be given as a continuous infusion, with one study comparing 4 mg/kg/day with goal trough range of 250–350 ng/mL to 2 mg/kg/day with goal levels between 150–250 in adult patients. Authors found that higher dosing was not shown to have any additional clinical benefit [73,74]. In the pediatric literature, the goal trough level for induction when using 2 mg/kg/day via continuous infusion is recommended to be between 150–300 ng/mL, and, once remission is achieved, this can be decreased to 100–200 ng/mL [71]. For oral dosing, one retrospective study with 14 children who received calcineurin inhibitors therapy (six of which were given cyclosporine) evaluated 4–8 mg/kg/day of cyclosporine and found that a goal trough range of 150–300 ng/mL was effective [75]. A second study with 28 children evaluated 5 mg/kg/day oral dosing while targeting trough levels between 150–250 ng/mL [76](Table 2). There are several different preparations of oral cyclosporine on the market, including Sandimmune and Neoral. Caution should be used when selecting an agent as these are not interchangeable products; Neoral is a microemulsion with better bioavailability [148]. Both tacrolimus and cyclosporine possess the risk of causing nephrotoxicity and serum electrolyte derangements, for which close monitoring of renal function in addition to potassium, magnesium, and phosphate are recommended (Table 3).

6.2.2. Time to Therapeutic Effect

The reported time to see clinical effect from calcineurin inhibitors is relatively quicker than other classes of medications used to treat steroid refractory ulcerative colitis in the pediatric population. With enteral tacrolimus response can be seen in just 14 days [72]. Oral cyclosporine showed clinical response within seven to fifteen days when using the enteral dosing scheme of 5 mg/kg/day [76,111] (Table 4).

7. Non-Pharmacologic Therapies

Several non-pharmacologic therapies are employed in treating monogenic and VEO-IBD, including nutrition, surgery, and hematopoietic stem cell transplantation (HSCT). HSCT is the only non-pharmacologic therapy that can be targeted at specific monogenic defects; however, it is important to note that it may worsen intestinal disease in some monogenic disorders, and thus accurate genetic diagnosis is critical prior to initiating therapy or considering HSCT. HSCT is curative in SCID, CGD, DOCK8 deficiency, IPEX syndrome, Wiskott-Aldrich syndrome, LRBA deficiency, and LAD1, and it improves intestinal disease in STAT-1 and STAT-3 GOF syndromes, IL-10 deficiency, and IL-10R deficiency [3,41]. However, it should be used with caution in patients with epithelial barrier defects, such as X-linked recessive ectodermal dysplasia with immunodeficiency. While HSCT cures the defect in immune cells, it does not affect the defective intestinal epithelial cells, and thus it may not improve intestinal disease. In some cases, HSCT can worsen or lead to de novo IBD symptoms in these patients after transplant [149]. The risks and benefits of HSCT should be carefully weighed in all patients with monogenic IBD, as HSCT can have severe consequences, including sepsis, graft versus host disease, secondary malignancy, and death [3].
Nutritional therapy with exclusive enteral nutrition (EEN) is used as a steroid-sparing strategy for induction of remission in pediatric Crohn’s disease and has been shown to be as effective as corticosteroids [150,151]. The mechanism of its action remains unclear, though is thought to involve modulation of the gut microbiome. It can be used safely in VEO-IBD, and in one small study, its use led to clinical remission in two infants [152]. Surgery can improve symptoms and quality of life in treatment-refractory monogenic and VEO-IBD, but it does not target underlying genetic defects. There is some evidence to suggest that VEO and monogenic IBD patients are more likely than older patients to require surgery due to the refractory nature of their disease [10,153], and thus referral to a center with experienced pediatric surgeons is needed.

8. Conclusions

The recent development of new medications that target specific immune pathways has revolutionized the treatment of inflammatory immune diseases, and it is poised to include monogenic IBD. Targeted therapy may allow for decreased reliance on systemic immunosuppressants, thereby reducing the likelihood of developing side effects or drug toxicity. When considering pharmacologic options for managing the VEO-IBD and monogenic IBD populations specifically, there is currently limited published data to support the use of these medications. Current practice relies on observational case studies, retrospective reviews, and ongoing clinical trials, or extrapolation from the polygenic IBD population. However, navigating the unknown in pediatric medicine is not foreign to those caring for these patients.
Large-scale randomized control trials may never be feasible in this population due to the rarity of these disorders. However, the field of oncology has lately been revolutionized by the advent of functional precision medicine. This allows for a single individual’s tumor cells to be directly inoculated with drugs and allows for instant, personalized profiling of response to therapy [154]. While such strategies do not yet exist for IBD, there are currently some promising research protocols aimed at predicting response to therapies, including cytokines and gene, microRNA, and microbial signatures [155]. When evaluating the role of precision medicine in the diagnosis of these patients, a high index of suspicion leading to early genetic diagnosis is of paramount importance in monogenic IBD, as patients with these disorders require complex care delivered by multidisciplinary teams in order to have the best possible outcomes. Early recognition and diagnosis allow children maximal opportunity for normal growth and development with minimal impairment due to treatment effects, and in some cases, a cure via stem cell transplantation. As more disease-causing genes are identified and new medications are developed, children with monogenic IBD will benefit from their position at the forefront of precision medicine.

Funding

This research received no external funding.

Conflicts of Interest

Suskind and Zheng consult for Pharming LLC. No other conflict of interest exists.

References

  1. Uhlig, H.H.; Schwerd, T.; Koletzko, S.; Shah, N.; Kammermeier, J.; Elkadri, A.; Ouahed, J.; Wilson, D.C.; Travis, S.P.; Turner, D.; et al. The diagnostic approach to monogenic very early onset inflammatory bowel disease. Gastroenterology 2014, 147, 990–1007.e3. [Google Scholar] [CrossRef] [Green Version]
  2. Worthey, E.A.; Mayer, A.N.; Syverson, G.D.; Helbling, D.; Bonacci, B.B.; Decker, B.; Serpe, J.M.; Dasu, T.; Tschannen, M.R.; Veith, R.L.; et al. Making a definitive diagnosis: Successful clinical application of whole exome sequencing in a child with intractable inflammatory bowel disease. Genet. Med. 2011, 13, 255–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Ouahed, J.; Spencer, E.; Kotlarz, D.; Shouval, D.S.; Kowalik, M.; Peng, K.; Field, M.; Grushkin-Lerner, L.; Pai, S.Y.; Bousvaros, A.; et al. Very Early Onset Inflammatory Bowel Disease: A Clinical Approach With a Focus on the Role of Genetics and Underlying Immune Deficiencies. Inflamm. Bowel Dis. 2020, 26, 820–842. [Google Scholar] [CrossRef] [PubMed]
  4. Charbit-Henrion, F.; Parlato, M.; Hanein, S.; Duclaux-Loras, R.; Nowak, J.; Begue, B.; Rakotobe, S.; Bruneau, J.; Fourrage, C.; Alibeu, O.; et al. Diagnostic Yield of Next-generation Sequencing in Very Early-onset Inflammatory Bowel Diseases: A Multicentre Study. J. Crohns Colitis 2018, 12, 1104–1112. [Google Scholar] [CrossRef] [PubMed]
  5. Benchimol, E.I.; Fortinsky, K.J.; Gozdyra, P.; Van den Heuvel, M.; Van Limbergen, J.; Griffiths, A.M. Epidemiology of pediatric inflammatory bowel disease: A systematic review of international trends. Inflamm. Bowel Dis. 2011, 17, 423–439. [Google Scholar] [CrossRef]
  6. Sykora, J.; Pomahacova, R.; Kreslova, M.; Cvalinova, D.; Stych, P.; Schwarz, J. Current global trends in the incidence of pediatric-onset inflammatory bowel disease. World J. Gastroenterol. 2018, 24, 2741–2763. [Google Scholar] [CrossRef]
  7. Azabdaftari, A.; Jones, K.D.J.; Kammermeier, J.; Uhlig, H.H. Monogenic inflammatory bowel disease-genetic variants, functional mechanisms and personalised medicine in clinical practice. Hum. Genet. 2022. [Google Scholar] [CrossRef]
  8. Bolton, C.; Smillie, C.S.; Pandey, S.; Elmentaite, R.; Wei, G.; Argmann, C.; Aschenbrenner, D.; James, K.R.; McGovern, D.P.B.; Macchi, M.; et al. An Integrated Taxonomy for Monogenic Inflammatory Bowel Disease. Gastroenterology 2022, 162, 859–876. [Google Scholar] [CrossRef]
  9. Conrad, M.A.; Kelsen, J.R. Genomic and Immunologic Drivers of Very Early-Onset Inflammatory Bowel Disease. Pediatr. Dev. Pathol. 2019, 22, 183–193. [Google Scholar] [CrossRef]
  10. Kelsen, J.R.; Sullivan, K.E.; Rabizadeh, S.; Singh, N.; Snapper, S.; Elkadri, A.; Grossman, A.B. North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition Position Paper on the Evaluation and Management for Patients With Very Early-onset Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2020, 70, 389–403. [Google Scholar] [CrossRef]
  11. Levine, A.E.; Zheng, H.B.; Suskind, D.L. Linking Genetic Diagnosis to Therapeutic Approach in Very Early Onset Inflammatory Bowel Disease: Pharmacologic Considerations. Paediatr. Drugs 2022, 24, 207–216. [Google Scholar] [CrossRef] [PubMed]
  12. Zheng, H.B.; de la Morena, M.T.; Suskind, D.L. The Growing Need to Understand Very Early Onset Inflammatory Bowel Disease. Front. Immunol. 2021, 12, 675186. [Google Scholar] [CrossRef] [PubMed]
  13. Schappi, M.G.; Smith, V.V.; Goldblatt, D.; Lindley, K.J.; Milla, P.J. Colitis in chronic granulomatous disease. Arch. Dis. Child. 2001, 84, 147–151. [Google Scholar] [CrossRef] [PubMed]
  14. Alimchandani, M.; Lai, J.P.; Aung, P.P.; Khangura, S.; Kamal, N.; Gallin, J.I.; Holland, S.M.; Malech, H.L.; Heller, T.; Miettinen, M.; et al. Gastrointestinal histopathology in chronic granulomatous disease: A study of 87 patients. Am. J. Surg. Pathol. 2013, 37, 1365–1372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. D’Agata, I.D.; Paradis, K.; Chad, Z.; Bonny, Y.; Seidman, E. Leucocyte adhesion deficiency presenting as a chronic ileocolitis. Gut 1996, 39, 605–608. [Google Scholar] [CrossRef] [PubMed]
  16. Uzel, G.; Kleiner, D.E.; Kuhns, D.B.; Holland, S.M. Dysfunctional LAD-1 neutrophils and colitis. Gastroenterology 2001, 121, 958–964. [Google Scholar] [CrossRef]
  17. van de Vijver, E.; Maddalena, A.; Sanal, O.; Holland, S.M.; Uzel, G.; Madkaikar, M.; de Boer, M.; van Leeuwen, K.; Koker, M.Y.; Parvaneh, N.; et al. Hematologically important mutations: Leukocyte adhesion deficiency (first update). Blood Cells Mol. Dis. 2012, 48, 53–61. [Google Scholar] [CrossRef] [Green Version]
  18. Roifman, C.M.; Zhang, J.; Atkinson, A.; Grunebaum, E.; Mandel, K. Adenosine deaminase deficiency can present with features of Omenn syndrome. J. Allergy Clin. Immunol. 2008, 121, 1056–1058. [Google Scholar] [CrossRef]
  19. Rohr, J.; Pannicke, U.; Doring, M.; Schmitt-Graeff, A.; Wiech, E.; Busch, A.; Speckmann, C.; Muller, I.; Lang, P.; Handgretinger, R.; et al. Chronic inflammatory bowel disease as key manifestation of atypical ARTEMIS deficiency. J. Clin. Immunol. 2010, 30, 314–320. [Google Scholar] [CrossRef]
  20. Grunebaum, E.; Bates, A.; Roifman, C.M. Omenn syndrome is associated with mutations in DNA ligase IV. J. Allergy Clin. Immunol. 2008, 122, 1219–1220. [Google Scholar] [CrossRef]
  21. Chan, A.Y.; Punwani, D.; Kadlecek, T.A.; Cowan, M.J.; Olson, J.L.; Mathes, E.F.; Sunderam, U.; Fu, S.M.; Srinivasan, R.; Kuriyan, J.; et al. A novel human autoimmune syndrome caused by combined hypomorphic and activating mutations in ZAP-70. J. Exp. Med. 2016, 213, 155–165. [Google Scholar] [CrossRef] [PubMed]
  22. Takahashi, N.; Matsumoto, K.; Saito, H.; Nanki, T.; Miyasaka, N.; Kobata, T.; Azuma, M.; Lee, S.K.; Mizutani, S.; Morio, T. Impaired CD4 and CD8 effector function and decreased memory T cell populations in ICOS-deficient patients. J. Immunol. 2009, 182, 5515–5527. [Google Scholar] [CrossRef] [Green Version]
  23. Catucci, M.; Castiello, M.C.; Pala, F.; Bosticardo, M.; Villa, A. Autoimmunity in wiskott-Aldrich syndrome: An unsolved enigma. Front. Immunol. 2012, 3, 209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Maekawa, K.; Yamada, M.; Okura, Y.; Sato, Y.; Yamada, Y.; Kawamura, N.; Ariga, T. X-linked agammaglobulinemia in a 10-year-old boy with a novel non-invariant splice-site mutation in Btk gene. Blood Cells Mol. Dis. 2010, 44, 300–304. [Google Scholar] [CrossRef] [PubMed]
  25. Bennett, C.L.; Christie, J.; Ramsdell, F.; Brunkow, M.E.; Ferguson, P.J.; Whitesell, L.; Kelly, T.E.; Saulsbury, F.T.; Chance, P.F.; Ochs, H.D. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat. Genet. 2001, 27, 20–21. [Google Scholar] [CrossRef]
  26. Barzaghi, F.; Passerini, L.; Bacchetta, R. Immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome: A paradigm of immunodeficiency with autoimmunity. Front. Immunol. 2012, 3, 211. [Google Scholar] [CrossRef] [Green Version]
  27. Zeissig, S.; Petersen, B.S.; Tomczak, M.; Melum, E.; Huc-Claustre, E.; Dougan, S.K.; Laerdahl, J.K.; Stade, B.; Forster, M.; Schreiber, S.; et al. Early-onset Crohn’s disease and autoimmunity associated with a variant in CTLA-4. Gut 2015, 64, 1889–1897. [Google Scholar] [CrossRef]
  28. Lopez-Herrera, G.; Tampella, G.; Pan-Hammarstrom, Q.; Herholz, P.; Trujillo-Vargas, C.M.; Phadwal, K.; Simon, A.K.; Moutschen, M.; Etzioni, A.; Mory, A.; et al. Deleterious mutations in LRBA are associated with a syndrome of immune deficiency and autoimmunity. Am. J. Hum. Genet. 2012, 90, 986–1001. [Google Scholar] [CrossRef] [Green Version]
  29. Gambineri, E.; Ciullini Mannurita, S.; Hagin, D.; Vignoli, M.; Anover-Sombke, S.; DeBoer, S.; Segundo, G.R.S.; Allenspach, E.J.; Favre, C.; Ochs, H.D.; et al. Clinical, Immunological, and Molecular Heterogeneity of 173 Patients With the Phenotype of Immune Dysregulation, Polyendocrinopathy, Enteropathy, X-Linked (IPEX) Syndrome. Front. Immunol. 2018, 9, 2411. [Google Scholar] [CrossRef]
  30. Caudy, A.A.; Reddy, S.T.; Chatila, T.; Atkinson, J.P.; Verbsky, J.W. CD25 deficiency causes an immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like syndrome, and defective IL-10 expression from CD4 lymphocytes. J. Allergy Clin. Immunol. 2007, 119, 482–487. [Google Scholar] [CrossRef] [PubMed]
  31. Latour, S.; Aguilar, C. XIAP deficiency syndrome in humans. Semin. Cell Dev. Biol. 2015, 39, 115–123. [Google Scholar] [CrossRef] [PubMed]
  32. Aguilar, C.; Latour, S. X-linked inhibitor of apoptosis protein deficiency: More than an X-linked lymphoproliferative syndrome. J. Clin. Immunol. 2015, 35, 331–338. [Google Scholar] [CrossRef] [PubMed]
  33. Glocker, E.O.; Frede, N.; Perro, M.; Sebire, N.; Elawad, M.; Shah, N.; Grimbacher, B. Infant colitis--it’s in the genes. Lancet 2010, 376, 1272. [Google Scholar] [CrossRef] [PubMed]
  34. Glocker, E.O.; Kotlarz, D.; Boztug, K.; Gertz, E.M.; Schaffer, A.A.; Noyan, F.; Perro, M.; Diestelhorst, J.; Allroth, A.; Murugan, D.; et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N. Engl. J. Med. 2009, 361, 2033–2045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Neven, B.; Mamessier, E.; Bruneau, J.; Kaltenbach, S.; Kotlarz, D.; Suarez, F.; Masliah-Planchon, J.; Billot, K.; Canioni, D.; Frange, P.; et al. A Mendelian predisposition to B-cell lymphoma caused by IL-10R deficiency. Blood 2013, 122, 3713–3722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Conrad, M.A.; Carreon, C.K.; Dawany, N.; Russo, P.; Kelsen, J.R. Distinct Histopathological Features at Diagnosis of Very Early Onset Inflammatory Bowel Disease. J. Crohns Colitis 2019, 13, 615–625. [Google Scholar] [CrossRef]
  37. Nambu, R.; Muise, A.M. Advanced Understanding of Monogenic Inflammatory Bowel Disease. Front. Pediatr. 2020, 8, 618918. [Google Scholar] [CrossRef]
  38. Voskuil, M.D.; Spekhorst, L.M.; van der Sloot, K.W.J.; Jansen, B.H.; Dijkstra, G.; van der Woude, C.J.; Hoentjen, F.; Pierik, M.J.; van der Meulen, A.E.; de Boer, N.K.H.; et al. Genetic Risk Scores Identify Genetic Aetiology of Inflammatory Bowel Disease Phenotypes. J. Crohns Colitis 2021, 15, 930–937. [Google Scholar] [CrossRef]
  39. Gettler, K.; Levantovsky, R.; Moscati, A.; Giri, M.; Wu, Y.; Hsu, N.Y.; Chuang, L.S.; Sazonovs, A.; Venkateswaran, S.; Korie, U.; et al. Common and Rare Variant Prediction and Penetrance of IBD in a Large, Multi-ethnic, Health System-based Biobank Cohort. Gastroenterology 2021, 160, 1546–1557. [Google Scholar] [CrossRef]
  40. Kelsen, J.R.; Conrad, M.A.; Dawany, N.; Patel, T.; Shraim, R.; Merz, A.; Maurer, K.; Sullivan, K.E.; Devoto, M. The Unique Disease Course of Children with Very Early onset-Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2020, 26, 909–918. [Google Scholar] [CrossRef]
  41. Castagnoli, R.; Delmonte, O.M.; Calzoni, E.; Notarangelo, L.D. Hematopoietic Stem Cell Transplantation in Primary Immunodeficiency Diseases: Current Status and Future Perspectives. Front. Pediatr. 2019, 7, 295. [Google Scholar] [CrossRef] [Green Version]
  42. Fusco, F.; Pescatore, A.; Conte, M.I.; Mirabelli, P.; Paciolla, M.; Esposito, E.; Lioi, M.B.; Ursini, M.V. EDA-ID and IP, two faces of the same coin: How the same IKBKG/NEMO mutation affecting the NF-kappaB pathway can cause immunodeficiency and/or inflammation. Int. Rev. Immunol. 2015, 34, 445–459. [Google Scholar] [CrossRef] [PubMed]
  43. O’Brien, K.J.; Parisi, X.; Shelman, N.R.; Merideth, M.A.; Introne, W.J.; Heller, T.; Gahl, W.A.; Malicdan, M.C.V.; Gochuico, B.R. Inflammatory bowel disease in Hermansky-Pudlak syndrome: A retrospective single-centre cohort study. J. Intern. Med. 2021, 290, 129–140. [Google Scholar] [CrossRef]
  44. Mizukami, T.; Obara, M.; Nishikomori, R.; Kawai, T.; Tahara, Y.; Sameshima, N.; Marutsuka, K.; Nakase, H.; Kimura, N.; Heike, T.; et al. Successful treatment with infliximab for inflammatory colitis in a patient with X-linked anhidrotic ectodermal dysplasia with immunodeficiency. J. Clin. Immunol. 2012, 32, 39–49. [Google Scholar] [CrossRef] [PubMed]
  45. Ishihara, J.; Mizuochi, T.; Uchida, T.; Takaki, Y.; Konishi, K.I.; Joo, M.; Takahashi, Y.; Yoshioka, S.; Kusano, H.; Sasahara, Y.; et al. Infantile-onset inflammatory bowel disease in a patient with Hermansky-Pudlak syndrome: A case report. BMC Gastroenterol. 2019, 19, 9. [Google Scholar] [CrossRef] [Green Version]
  46. Boschetti, G.; Sarfati, M.; Fabien, N.; Flourie, B.; Lachaux, A.; Nancey, S.; Coury, F. Infliximab induces clinical resolution of sacroiliitis that coincides with increased circulating FOXP3(+) T cells in a patient with IPEX syndrome. Jt. Bone Spine 2020, 87, 483–486. [Google Scholar] [CrossRef]
  47. Uzel, G.; Orange, J.S.; Poliak, N.; Marciano, B.E.; Heller, T.; Holland, S.M. Complications of tumor necrosis factor-alpha blockade in chronic granulomatous disease-related colitis. Clin. Infect. Dis. 2010, 51, 1429–1434. [Google Scholar] [CrossRef]
  48. Hyams, J.; Crandall, W.; Kugathasan, S.; Griffiths, A.; Olson, A.; Johanns, J.; Liu, G.; Travers, S.; Heuschkel, R.; Markowitz, J.; et al. Induction and maintenance infliximab therapy for the treatment of moderate-to-severe Crohn’s disease in children. Gastroenterology 2007, 132, 863–873, quiz 1165–1166. [Google Scholar] [CrossRef]
  49. van Rheenen, P.F.; Aloi, M.; Assa, A.; Bronsky, J.; Escher, J.C.; Fagerberg, U.L.; Gasparetto, M.; Gerasimidis, K.; Griffiths, A.; Henderson, P.; et al. The Medical Management of Paediatric Crohn’s Disease: An ECCO-ESPGHAN Guideline Update. J. Crohns Colitis 2020, 15, 171–194. [Google Scholar] [CrossRef]
  50. Bolia, R.; Rosenbaum, J.; Schildkraut, V.; Hardikar, W.; Oliver, M.; Cameron, D.; Alex, G. Secondary Loss of Response to Infliximab in Pediatric Crohn Disease: Does It Matter How and When We Start? J. Pediatr. Gastroenterol. Nutr. 2018, 66, 637–640. [Google Scholar] [CrossRef] [PubMed]
  51. Legeret, C.; Furlano, R.; Kohler, H. Therapy Strategies for Children Suffering from Inflammatory Bowel Disease (IBD)-A Narrative Review. Children 2022, 9, 617. [Google Scholar] [CrossRef] [PubMed]
  52. Kelsen, J.R.; Grossman, A.B.; Pauly-Hubbard, H.; Gupta, K.; Baldassano, R.N.; Mamula, P. Infliximab therapy in pediatric patients 7 years of age and younger. J. Pediatr. Gastroenterol. Nutr. 2014, 59, 758–762. [Google Scholar] [CrossRef] [Green Version]
  53. Bramuzzo, M.; Arrigo, S.; Romano, C.; Filardi, M.C.; Lionetti, P.; Agrusti, A.; Dipasquale, V.; Paci, M.; Zuin, G.; Aloi, M.; et al. Efficacy and safety of infliximab in very early onset inflammatory bowel disease: A national comparative retrospective study. United Eur. Gastroenterol. J. 2019, 7, 759–766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Assa, A.; Dorfman, L.; Shouval, D.S.; Shamir, R.; Cohen, S. Therapeutic Drug Monitoring-guided High-dose Infliximab for Infantile-onset Inflammatory Bowel Disease: A Case Series. J. Pediatr. Gastroenterol. Nutr. 2020, 71, 516–520. [Google Scholar] [CrossRef]
  55. Chi, L.Y.; Zitomersky, N.L.; Liu, E.; Tollefson, S.; Bender-Stern, J.; Naik, S.; Snapper, S.; Bousvaros, A. The Impact of Combination Therapy on Infliximab Levels and Antibodies in Children and Young Adults With Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2018, 24, 1344–1351. [Google Scholar] [CrossRef] [PubMed]
  56. Colman, R.J.; Lawton, R.C.; Dubinsky, M.C.; Rubin, D.T. Methotrexate for the Treatment of Pediatric Crohn’s Disease: A Systematic Review and Meta-analysis. Inflamm. Bowel Dis. 2018, 24, 2135–2141. [Google Scholar] [CrossRef]
  57. Aardoom, M.A.; Veereman, G.; de Ridder, L. A Review on the Use of Anti-TNF in Children and Adolescents with Inflammatory Bowel Disease. Int. J. Mol. Sci. 2019, 20, 2529. [Google Scholar] [CrossRef] [Green Version]
  58. Kolho, K.L. Therapeutic Drug Monitoring and Outcome of Infliximab Therapy in Pediatric Onset Inflammatory Bowel Disease. Front. Pediatr. 2020, 8, 623689. [Google Scholar] [CrossRef]
  59. Clarkston, K.; Tsai, Y.T.; Jackson, K.; Rosen, M.J.; Denson, L.A.; Minar, P. Development of Infliximab Target Concentrations During Induction in Pediatric Crohn Disease Patients. J. Pediatr. Gastroenterol. Nutr. 2019, 69, 68–74. [Google Scholar] [CrossRef]
  60. deBruyn, J.C.C.; Jacobson, K.; El-Matary, W.; Wine, E.; Carroll, M.W.; Goedhart, C.; Panaccione, R.; Wrobel, I.T.; Huynh, H.Q. Early Serum Infliximab Levels in Pediatric Ulcerative Colitis. Front. Pediatr. 2021, 9, 668978. [Google Scholar] [CrossRef]
  61. Adedokun, O.J.; Xu, Z.; Padgett, L.; Blank, M.; Johanns, J.; Griffiths, A.; Ford, J.; Zhou, H.; Guzzo, C.; Davis, H.M.; et al. Pharmacokinetics of infliximab in children with moderate-to-severe ulcerative colitis: Results from a randomized, multicenter, open-label, phase 3 study. Inflamm. Bowel Dis. 2013, 19, 2753–2762. [Google Scholar] [CrossRef] [PubMed]
  62. Colman, R.J.; Dhaliwal, J.; Rosen, M.J. Predicting Therapeutic Response in Pediatric Ulcerative Colitis-A Journey Towards Precision Medicine. Front. Pediatr. 2021, 9, 634739. [Google Scholar] [CrossRef] [PubMed]
  63. Rinawi, F.; Ricciuto, A.; Church, P.C.; Frost, K.; Crowley, E.; Walters, T.D.; Griffiths, A.M. Association of Early Postinduction Adalimumab Exposure With Subsequent Clinical and Biomarker Remission in Children with Crohn’s Disease. Inflamm. Bowel Dis. 2021, 27, 1079–1087. [Google Scholar] [CrossRef] [PubMed]
  64. Vedolizumab Package Insert. 2022. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/125476Orig1s046lbl.pdf (accessed on 10 February 2023).
  65. Vaughn, B.P.; Yarur, A.J.; Graziano, E.; Campbell, J.P.; Bhattacharya, A.; Lee, J.Y.; Gheysens, K.; Papamichael, K.; Osterman, M.T.; Cheifetz, A.S.; et al. Vedolizumab Serum Trough Concentrations and Response to Dose Escalation in Inflammatory Bowel Disease. J. Clin. Med. 2020, 9, 3142. [Google Scholar] [CrossRef]
  66. Aardoom, M.A.; Jongsma, M.M.E.; de Vries, A.; Wolthoorn, J.; de Ridder, L.; Escher, J.C. Vedolizumab Trough Levels in Children With Anti-Tumor Necrosis Factor Refractory Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2020, 71, 501–507. [Google Scholar] [CrossRef]
  67. Restellini, S.; Afif, W. Update on TDM (Therapeutic Drug Monitoring) with Ustekinumab, Vedolizumab and Tofacitinib in Inflammatory Bowel Disease. J. Clin. Med. 2021, 10, 1242. [Google Scholar] [CrossRef]
  68. Opréa, A.; Collardeau-Frachon, S.; Heissat, S.; Peretti, N.; Lachaux, A.; Duclaux-Loras, R. Therapeutic approach of very early-onset inflammatory bowel disease in a Loeys-Dietz Syndrome Child. JPGN Rep. 2021, 3, e139. [Google Scholar] [CrossRef]
  69. Soufflet, N.; Boschetti, G.; Roblin, X.; Cuercq, C.; Williet, N.; Charlois, A.L.; Duclaux-Loras, R.; Danion, P.; Mialon, A.; Faure, M.; et al. Concentrations of Ustekinumab During Induction Therapy Associate With Remission in Patients With Crohn’s Disease. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2019, 17, 2610–2612. [Google Scholar] [CrossRef]
  70. Adedokun, O.J.; Xu, Z.; Gasink, C.; Jacobstein, D.; Szapary, P.; Johanns, J.; Gao, L.L.; Davis, H.M.; Hanauer, S.B.; Feagan, B.G.; et al. Pharmacokinetics and Exposure Response Relationships of Ustekinumab in Patients With Crohn’s Disease. Gastroenterology 2018, 154, 1660–1671. [Google Scholar] [CrossRef] [Green Version]
  71. Turner, D.; Ruemmele, F.M.; Orlanski-Meyer, E.; Griffiths, A.M.; de Carpi, J.M.; Bronsky, J.; Veres, G.; Aloi, M.; Strisciuglio, C.; Braegger, C.P.; et al. Management of Paediatric Ulcerative Colitis, Part 2: Acute Severe Colitis-An Evidence-based Consensus Guideline From the European Crohn’s and Colitis Organization and the European Society of Paediatric Gastroenterology, Hepatology and Nutrition. J. Pediatr. Gastroenterol. Nutr. 2018, 67, 292–310. [Google Scholar] [CrossRef]
  72. Bousvaros, A.; Kirschner, B.S.; Werlin, S.L.; Parker-Hartigan, L.; Daum, F.; Freeman, K.B.; Balint, J.P.; Day, A.S.; Griffiths, A.M.; Zurakowski, D.; et al. Oral tacrolimus treatment of severe colitis in children. J. Pediatr. 2000, 137, 794–799. [Google Scholar] [CrossRef] [PubMed]
  73. Van Assche, G.; D’Haens, G.; Noman, M.; Vermeire, S.; Hiele, M.; Asnong, K.; Arts, J.; D’Hoore, A.; Penninckx, F.; Rutgeerts, P. Randomized, double-blind comparison of 4 mg/kg versus 2 mg/kg intravenous cyclosporine in severe ulcerative colitis. Gastroenterology 2003, 125, 1025–1031. [Google Scholar] [CrossRef] [PubMed]
  74. Nakase, H.; Yoshino, T.; Matsuura, M. Role in calcineurin inhibitors for inflammatory bowel disease in the biologics era: When and how to use. Inflamm. Bowel Dis. 2014, 20, 2151–2156. [Google Scholar] [CrossRef]
  75. Hait, E.J.; Bousvaros, A.; Schuman, M.; Shamberger, R.C.; Lillehei, C.W. Pouch outcomes among children with ulcerative colitis treated with calcineurin inhibitors before ileal pouch anal anastomosis surgery. J. Pediatr. Surg. 2007, 42, 31–34, discussion 34–35. [Google Scholar] [CrossRef] [PubMed]
  76. Bradley, G.M.; Oliva-Hemker, M. Pediatric ulcerative colitis: Current treatment approaches including role of infliximab. Biologics 2012, 6, 125–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Targan, S.R.; Hanauer, S.B.; van Deventer, S.J.; Mayer, L.; Present, D.H.; Braakman, T.; DeWoody, K.L.; Schaible, T.F.; Rutgeerts, P.J. A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn’s disease. Crohn’s Disease cA2 Study Group. N. Engl. J. Med. 1997, 337, 1029–1035. [Google Scholar] [CrossRef] [Green Version]
  78. Hanauer, S.B.; Feagan, B.G.; Lichtenstein, G.R.; Mayer, L.F.; Schreiber, S.; Colombel, J.F.; Rachmilewitz, D.; Wolf, D.C.; Olson, A.; Bao, W.; et al. Maintenance infliximab for Crohn’s disease: The ACCENT I randomised trial. Lancet 2002, 359, 1541–1549. [Google Scholar] [CrossRef]
  79. Vasudevan, A.; Gibson, P.R.; van Langenberg, D.R. Time to clinical response and remission for therapeutics in inflammatory bowel diseases: What should the clinician expect, what should patients be told? World J. Gastroenterol. 2017, 23, 6385–6402. [Google Scholar] [CrossRef]
  80. Rutgeerts, P.; Van Assche, G.; Sandborn, W.J.; Wolf, D.C.; Geboes, K.; Colombel, J.F.; Reinisch, W.; Investigators, E.; Kumar, A.; Lazar, A.; et al. Adalimumab induces and maintains mucosal healing in patients with Crohn’s disease: Data from the EXTEND trial. Gastroenterology 2012, 142, 1102–1111.e2. [Google Scholar] [CrossRef]
  81. Reinisch, W.; Sandborn, W.J.; Hommes, D.W.; D’Haens, G.; Hanauer, S.; Schreiber, S.; Panaccione, R.; Fedorak, R.N.; Tighe, M.B.; Huang, B.; et al. Adalimumab for induction of clinical remission in moderately to severely active ulcerative colitis: Results of a randomised controlled trial. Gut 2011, 60, 780–787. [Google Scholar] [CrossRef]
  82. Hyams, J.; Damaraju, L.; Blank, M.; Johanns, J.; Guzzo, C.; Winter, H.S.; Kugathasan, S.; Cohen, S.; Markowitz, J.; Escher, J.C.; et al. Induction and maintenance therapy with infliximab for children with moderate to severe ulcerative colitis. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2012, 10, 391–399.e391. [Google Scholar] [CrossRef]
  83. Choi, S.Y.; Kang, B. Adalimumab in Pediatric Inflammatory Bowel Disease. Front. Pediatr. 2022, 10, 852580. [Google Scholar] [CrossRef]
  84. Campbell, N.; Chapdelaine, H. Treatment of chronic granulomatous disease-associated fistulising colitis with vedolizumab. J. Allergy Clin. Immunol. Pract. 2017, 5, 1748–1749. [Google Scholar] [CrossRef] [PubMed]
  85. Kamal, N.; Marciano, B.; Curtin, B.; Strongin, A.; DeRavin, S.S.; Bousvaros, A.; Koh, C.; Malech, H.L.; Holland, S.M.; Zerbe, C.; et al. The response to vedolizumab in chronic granulomatous disease-related inflammatory bowel disease. Gastroenterol. Rep. 2020, 8, 404–406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Navarini, A.A.; Hruz, P.; Berger, C.T.; Hou, T.Z.; Schwab, C.; Gabrysch, A.; Higgins, R.; Frede, N.; Padberg Sgier, B.C.; Kampe, O.; et al. Vedolizumab as a successful treatment of CTLA-4-associated autoimmune enterocolitis. J. Allergy Clin. Immunol. 2017, 139, 1043–1046.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Fabiszewska, S.; Derda, E.; Szymanska, E.; Osiecki, M.; Kierkus, J. Safety and Effectiveness of Vedolizumab for the Treatment of Pediatric Patients with Very Early Onset Inflammatory Bowel Diseases. J. Clin. Med. 2021, 10, 2997. [Google Scholar] [CrossRef]
  88. Nassar, I.O.; Cheesbrough, J.; Quraishi, M.N.; Sharma, N. Proposed pathway for therapeutic drug monitoring and dose escalation of vedolizumab. Front. Gastroenterol. 2022, 13, 430–435. [Google Scholar] [CrossRef]
  89. Jagt, J.Z.; Pothof, C.D.; Buiter, H.J.C.; van Limbergen, J.E.; van Wijk, M.P.; Benninga, M.A.; de Boer, N.K.H.; de Meij, T.G.J. Adverse Events of Thiopurine Therapy in Pediatric Inflammatory Bowel Disease and Correlations with Metabolites: A Cohort Study. Dig. Dis. Sci. 2022, 67, 241–251. [Google Scholar] [CrossRef]
  90. Ooi, C.Y.; Bohane, T.D.; Lee, D.; Naidoo, D.; Day, A.S. Thiopurine metabolite monitoring in paediatric inflammatory bowel disease. Aliment. Pharmacol. Ther. 2007, 25, 941–947. [Google Scholar] [CrossRef]
  91. Relling, M.V.; Schwab, M.; Whirl-Carrillo, M.; Suarez-Kurtz, G.; Pui, C.H.; Stein, C.M.; Moyer, A.M.; Evans, W.E.; Klein, T.E.; Antillon-Klussmann, F.G.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline for Thiopurine Dosing Based on TPMT and NUDT15 Genotypes: 2018 Update. Clin. Pharmacol. Ther. 2019, 105, 1095–1105. [Google Scholar] [CrossRef] [Green Version]
  92. Ruemmele, F.M.; Veres, G.; Kolho, K.L.; Griffiths, A.; Levine, A.; Escher, J.C.; Amil Dias, J.; Barabino, A.; Braegger, C.P.; Bronsky, J.; et al. Consensus guidelines of ECCO/ESPGHAN on the medical management of pediatric Crohn’s disease. J. Crohns Colitis 2014, 8, 1179–1207. [Google Scholar] [CrossRef] [Green Version]
  93. Azathioprine. Glaxo Smith Kline (GSK): Prescribing Information/Insert for Azathioprine. 2022. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/016324s034s035lbl.pdf (accessed on 10 February 2023).
  94. Zvuloni, M.; Matar, M.; Levi, R.; Shouval, D.S.; Shamir, R.; Assa, A. High anti-TNFalpha Concentrations Are Not Associated With More Adverse Events in Pediatric Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2021, 73, 717–721. [Google Scholar] [CrossRef] [PubMed]
  95. Chebli, J.M.; Gaburri, P.D.; Chebli, L.A.; da Rocha Ribeiro, T.C.; Pinto, A.L.; Ambrogini Junior, O.; Damiao, A.O. A guide to prepare patients with inflammatory bowel diseases for anti-TNF-alpha therapy. Med. Sci. Monit. 2014, 20, 487–498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Ustekinumab Package Insert. 2022. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/125261s161lbl.pdf (accessed on 10 February 2023).
  97. Prograf (Tacrolimus) Package Insert. 2022. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/050708s054,050709s047,204096s010lbl.pdf (accessed on 10 February 2023).
  98. Cyclosporine (Neoral) Package Insert. 2022. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/021083s069s070,021110s087s088lbl.pdf (accessed on 10 February 2023).
  99. Anakinra Package Insert; Sobi Orphan Biovitrum: Stockholm, Sweden, 2012.
  100. Canakinumab Package Insert; Novartis Pharmaceuticals Corporation: East Hanover, NJ, USA, 2016.
  101. Abetacept. 2023. Available online: https://online.lexi.com/lco/action/doc/retrieve/docid/pdh_f/521853?cesid=5N3h7bKhudl&searchUrl=%2Flco%2Faction%2Fsearch%3Fq%3Dabatacept%26t%3Dname%26acs%3Dfalse%26acq%3Dabatacept#parentdoc-tab-content (accessed on 10 February 2023).
  102. Tofacitinib. 2023. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/203214s018lbl.pdf (accessed on 10 February 2023).
  103. Ruxolitinib Package Insert. 2023. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/202192s023lbl.pdf (accessed on 10 February 2023).
  104. Meserve, J.; Dulai, P. Predicting Response to Vedolizumab in Inflammatory Bowel Disease. Front. Med. 2020, 7, 76. [Google Scholar] [CrossRef] [PubMed]
  105. Shah, P.; McDonald, D. Vedolizumab: An Emerging Treatment Option for Pediatric Inflammatory Bowel Disease. J Pediatr Pharmacol. Ther. 2021, 26, 795–801. [Google Scholar] [CrossRef]
  106. Present, D.H.; Korelitz, B.I.; Wisch, N.; Glass, J.L.; Sachar, D.B.; Pasternack, B.S. Treatment of Crohn’s disease with 6-mercaptopurine. A long-term, randomized, double-blind study. N. Engl. J. Med. 1980, 302, 981–987. [Google Scholar] [CrossRef]
  107. Kirk, A.P.; Lennard-Jones, J.E. Controlled trial of azathioprine in chronic ulcerative colitis. Br. Med. J. (Clin. Res. Ed.) 1982, 284, 1291–1292. [Google Scholar] [CrossRef] [Green Version]
  108. Gurram, B.; Patel, A.S. Recent advances in understanding and managing pediatric inflammatory bowel disease. F1000Res 2019, 8. [Google Scholar] [CrossRef] [Green Version]
  109. Cohen, R.D.; Tsang, J.F.; Hanauer, S.B. Infliximab in Crohn’s disease: First anniversary clinical experience. Am. J. Gastroenterol. 2000, 95, 3469–3477. [Google Scholar] [CrossRef]
  110. Laharie, D.; Bourreille, A.; Branche, J.; Allez, M.; Bouhnik, Y.; Filippi, J.; Zerbib, F.; Savoye, G.; Nachury, M.; Moreau, J.; et al. Ciclosporin versus infliximab in patients with severe ulcerative colitis refractory to intravenous steroids: A parallel, open-label randomised controlled trial. Lancet 2012, 380, 1909–1915. [Google Scholar] [CrossRef] [Green Version]
  111. Castro, M.; Papadatou, B.; Ceriati, E.; Knafelz, D.; De Angelis, P.; Ferretti, F.; Gambarara, M.; Diamanti, A.; De Peppo, F.; Rivosecchi, M. Role of cyclosporin in preventing or delaying colectomy in children with severe ulcerative colitis. Langenbecks Arch. Surg. 2007, 392, 161–164. [Google Scholar] [CrossRef]
  112. Conrad, M.A.; Kelsen, J.R. The Treatment of Pediatric Inflammatory Bowel Disease with Biologic Therapies. Curr. Gastroenterol. Rep. 2020, 22, 36. [Google Scholar] [CrossRef]
  113. Dayan, J.R.; Dolinger, M.; Benkov, K.; Dunkin, D.; Jossen, J.; Lai, J.; Phan, B.L.; Pittman, N.; Dubinsky, M.C. Real World Experience With Ustekinumab in Children and Young Adults at a Tertiary Care Pediatric Inflammatory Bowel Disease Center. J. Pediatr. Gastroenterol. Nutr. 2019, 69, 61–67. [Google Scholar] [CrossRef] [PubMed]
  114. Rudra, S.; Shaul, E.; Conrad, M.; Patel, T.; Moore, A.; Dawany, N.; Canavan, M.C.; Sullivan, K.E.; Behrens, E.; Kelsen, J.R. Ruxolitinib: Targeted Approach for Treatment of Autoinflammatory Very Early Onset Inflammatory Bowel Disease. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2022, 20, 1408–1410.e2. [Google Scholar] [CrossRef]
  115. Parlato, M.; Charbit-Henrion, F.; Abi Nader, E.; Begue, B.; Guegan, N.; Bruneau, J.; Khater, S.; Macintyre, E.; Picard, C.; Frederic, R.L.; et al. Efficacy of Ruxolitinib Therapy in a Patient With Severe Enterocolitis Associated With a STAT3 Gain-of-Function Mutation. Gastroenterology 2019, 156, 1206–1210.e1. [Google Scholar] [CrossRef] [Green Version]
  116. Casanova, J.L.; Holland, S.M.; Notarangelo, L.D. Inborn errors of human JAKs and STATs. Immunity 2012, 36, 515–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Kofoed, E.M.; Hwa, V.; Little, B.; Woods, K.A.; Buckway, C.K.; Tsubaki, J.; Pratt, K.L.; Bezrodnik, L.; Jasper, H.; Tepper, A.; et al. Growth hormone insensitivity associated with a STAT5b mutation. N. Engl. J. Med. 2003, 349, 1139–1147. [Google Scholar] [CrossRef] [PubMed]
  118. Joosse, M.E.; Charbit-Henrion, F.; Boisgard, R.; Raatgeep, R.H.C.; Lindenbergh-Kortleve, D.J.; Costes, L.M.M.; Nugteren, S.; Guegan, N.; Parlato, M.; Veenbergen, S.; et al. Duplication of the IL2RA locus causes excessive IL-2 signaling and may predispose to very early onset colitis. Mucosal Immunol. 2021, 14, 1172–1182. [Google Scholar] [CrossRef] [PubMed]
  119. Moore, H.; Dubes, L.; Fusillo, S.; Baldassano, R.; Stein, R. Tofacitinib Therapy in Children and Young Adults With Pediatric-onset Medically Refractory Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2021, 73, e57–e62. [Google Scholar] [CrossRef]
  120. O’Shea, J.J.; Gadina, M. Selective Janus kinase inhibitors come of age. Nat. Rev. Rheumatol. 2019, 15, 74–75. [Google Scholar] [CrossRef]
  121. Magnani, A.; Mahlaoui, N. Managing Inflammatory Manifestations in Patients with Chronic Granulomatous Disease. Paediatr. Drugs 2016, 18, 335–345. [Google Scholar] [CrossRef] [PubMed]
  122. Pariano, M.; Pieroni, S.; De Luca, A.; Iannitti, R.G.; Borghi, M.; Puccetti, M.; Giovagnoli, S.; Renga, G.; D’Onofrio, F.; Bellet, M.M.; et al. Anakinra Activates Superoxide Dismutase 2 to Mitigate Inflammasome Activity. Int. J. Mol. Sci. 2021, 22, 6531. [Google Scholar] [CrossRef]
  123. de Luca, A.; Smeekens, S.P.; Casagrande, A.; Iannitti, R.; Conway, K.L.; Gresnigt, M.S.; Begun, J.; Plantinga, T.S.; Joosten, L.A.; van der Meer, J.W.; et al. IL-1 receptor blockade restores autophagy and reduces inflammation in chronic granulomatous disease in mice and in humans. Proc. Natl. Acad. Sci. USA 2014, 111, 3526–3531. [Google Scholar] [CrossRef] [Green Version]
  124. Hahn, K.J.; Ho, N.; Yockey, L.; Kreuzberg, S.; Daub, J.; Rump, A.; Marciano, B.E.; Quezado, M.; Malech, H.L.; Holland, S.M.; et al. Treatment With Anakinra, a Recombinant IL-1 Receptor Antagonist, Unlikely to Induce Lasting Remission in Patients With CGD Colitis. Am. J. Gastroenterol. 2015, 110, 938–939. [Google Scholar] [CrossRef]
  125. Peciuliene, S.; Burnyte, B.; Gudaitiene, R.; Rusoniene, S.; Drazdiene, N.; Liubsys, A.; Utkus, A. Perinatal manifestation of mevalonate kinase deficiency and efficacy of anakinra. Pediatr. Rheumatol. Online J. 2016, 14, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Campanilho-Marques, R.; Brogan, P.A. Mevalonate kinase deficiency in two sisters with therapeutic response to anakinra: Case report and review of the literature. Clin. Rheumatol. 2014, 33, 1681–1684. [Google Scholar] [CrossRef]
  127. Shaul, E.; Conrad, M.A.; Dawany, N.; Patel, T.; Canavan, M.C.; Baccarella, A.; Weinbrom, S.; Aleynick, D.; Sullivan, K.E.; Kelsen, J.R. Canakinumab for the treatment of autoinflammatory very early onset- inflammatory bowel disease. Front. Immunol. 2022, 13, 972114. [Google Scholar] [CrossRef] [PubMed]
  128. Wlazlo, M.; Kierkus, J. Dual Biologic Therapy for the Treatment of Pediatric Inflammatory Bowel Disease: A Review of the Literature. J. Clin. Med. 2022, 11, 2004. [Google Scholar] [CrossRef]
  129. Goyal, A.; Bass, J. Safety and Efficacy of Combining Biologicals in Children with Inflammatory Bowel Disease. Gastroenterology 2020, 158, s122. [Google Scholar] [CrossRef]
  130. Kotlarz, D.; Beier, R.; Murugan, D.; Diestelhorst, J.; Jensen, O.; Boztug, K.; Pfeifer, D.; Kreipe, H.; Pfister, E.D.; Baumann, U.; et al. Loss of interleukin-10 signaling and infantile inflammatory bowel disease: Implications for diagnosis and therapy. Gastroenterology 2012, 143, 347–355. [Google Scholar] [CrossRef] [Green Version]
  131. Shouval, D.S.; Biswas, A.; Kang, Y.H.; Griffith, A.E.; Konnikova, L.; Mascanfroni, I.D.; Redhu, N.S.; Frei, S.M.; Field, M.; Doty, A.L.; et al. Interleukin 1beta Mediates Intestinal Inflammation in Mice and Patients With Interleukin 10 Receptor Deficiency. Gastroenterology 2016, 151, 1100–1104. [Google Scholar] [CrossRef] [Green Version]
  132. Lo, B.; Zhang, K.; Lu, W.; Zheng, L.; Zhang, Q.; Kanellopoulou, C.; Zhang, Y.; Liu, Z.; Fritz, J.M.; Marsh, R.; et al. AUTOIMMUNE DISEASE. Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy. Science 2015, 349, 436–440. [Google Scholar] [CrossRef] [Green Version]
  133. Kiykim, A.; Ogulur, I.; Dursun, E.; Charbonnier, L.M.; Nain, E.; Cekic, S.; Dogruel, D.; Karaca, N.E.; Cogurlu, M.T.; Bilir, O.A.; et al. Abatacept as a Long-Term Targeted Therapy for LRBA Deficiency. J. Allergy Clin. Immunol. Pract. 2019, 7, 2790–2800.e15. [Google Scholar] [CrossRef]
  134. Kalaidina, E.; Utterson, E.C.; Mokshagundam, D.; He, M.; Shenoy, S.; Cooper, M.A. Case Report: “Primary Immunodeficiency”-Severe Autoimmune Enteropathy in a Pediatric Heart Transplant Recipient Treated With Abatacept and Alemtuzumab. Front. Immunol. 2022, 13, 863218. [Google Scholar] [CrossRef] [PubMed]
  135. Romberg, N.; Al Moussawi, K.; Nelson-Williams, C.; Stiegler, A.L.; Loring, E.; Choi, M.; Overton, J.; Meffre, E.; Khokha, M.K.; Huttner, A.J.; et al. Mutation of NLRC4 causes a syndrome of enterocolitis and autoinflammation. Nat. Genet. 2014, 46, 1135–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Canna, S.W.; de Jesus, A.A.; Gouni, S.; Brooks, S.R.; Marrero, B.; Liu, Y.; DiMattia, M.A.; Zaal, K.J.; Sanchez, G.A.; Kim, H.; et al. An activating NLRC4 inflammasome mutation causes autoinflammation with recurrent macrophage activation syndrome. Nat. Genet. 2014, 46, 1140–1146. [Google Scholar] [CrossRef] [Green Version]
  137. Canna, S.W.; Girard, C.; Malle, L.; de Jesus, A.; Romberg, N.; Kelsen, J.; Surrey, L.F.; Russo, P.; Sleight, A.; Schiffrin, E.; et al. Life-threatening NLRC4-associated hyperinflammation successfully treated with IL-18 inhibition. J. Allergy Clin. Immunol. 2017, 139, 1698–1701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Bedoui, Y.; Guillot, X.; Selambarom, J.; Guiraud, P.; Giry, C.; Jaffar-Bandjee, M.C.; Ralandison, S.; Gasque, P. Methotrexate an Old Drug with New Tricks. Int. J. Mol. Sci. 2019, 20, 5023. [Google Scholar] [CrossRef] [Green Version]
  139. Fagbemi, A.; Newman, W.G.; Tangye, S.G.; Hughes, S.M.; Cheesman, E.; Arkwright, P.D. Refractory very early-onset inflammatory bowel disease associated with cytosolic isoleucyl-tRNA synthetase deficiency: A case report. World J. Gastroenterol. 2020, 26, 1841–1846. [Google Scholar] [CrossRef]
  140. Gerards, A.H.; de Lathouder, S.; de Groot, E.R.; Dijkmans, B.A.; Aarden, L.A. Inhibition of cytokine production by methotrexate. Studies in healthy volunteers and patients with rheumatoid arthritis. Rheumatology 2003, 42, 1189–1196. [Google Scholar] [CrossRef] [Green Version]
  141. Willot, S.; Noble, A.; Deslandres, C. Methotrexate in the treatment of inflammatory bowel disease: An 8-year retrospective study in a Canadian pediatric IBD center. Inflamm. Bowel Dis. 2011, 17, 2521–2526. [Google Scholar] [CrossRef] [PubMed]
  142. Walker, R.; Kammermeier, J.; Vora, R.; Mutalib, M. Azathioprine dosing and metabolite measurement in pediatric inflammatory bowel disease: Does one size fit all? Ann. Gastroenterol. 2019, 32, 387–391. [Google Scholar] [CrossRef]
  143. Grossman, A.B.; Noble, A.J.; Mamula, P.; Baldassano, R.N. Increased dosing requirements for 6-mercaptopurine and azathioprine in inflammatory bowel disease patients six years and younger. Inflamm. Bowel Dis. 2008, 14, 750–755. [Google Scholar] [CrossRef] [PubMed]
  144. Treem, W.R.; Cohen, J.; Davis, P.M.; Justinich, C.J.; Hyams, J.S. Cyclosporine for the treatment of fulminant ulcerative colitis in children. Immediate response, long-term results, and impact on surgery. Dis. Colon Rectum 1995, 38, 474–479. [Google Scholar] [CrossRef]
  145. Kino, T.; Hatanaka, H.; Miyata, S.; Inamura, N.; Nishiyama, M.; Yajima, T.; Goto, T.; Okuhara, M.; Kohsaka, M.; Aoki, H.; et al. FK-506, a novel immunosuppressant isolated from a Streptomyces. II. Immunosuppressive effect of FK-506 in vitro. J. Antibiot. 1987, 40, 1256–1265. [Google Scholar] [CrossRef] [PubMed]
  146. Turner, D. Severe acute ulcerative colitis: The pediatric perspective. Dig. Dis. 2009, 27, 322–326. [Google Scholar] [CrossRef]
  147. Lekbua, A.; Ouahed, J.; O’Connell, A.E.; Kahn, S.A.; Goldsmith, J.D.; Imamura, T.; Duncan, C.N.; Kelsen, J.R.; Worthey, E.; Snapper, S.B.; et al. Risk-factors Associated With Poor Outcomes in VEO-IBD Secondary to XIAP Deficiency: A Case Report and Literature Review. J. Pediatr. Gastroenterol. Nutr. 2019, 69, e13–e18. [Google Scholar] [CrossRef]
  148. Hricik, D.E.; Dixit, A.; Knauss, T.C.; Donley, V.; Bartucci, M.R.; Schulak, J.A. Benefits of pre-emptive dose reduction for Sandimmune to Neoral conversion in stable renal transplant recipients. Clin. Transplant. 1998, 12, 575–578. [Google Scholar] [PubMed]
  149. Miot, C.; Imai, K.; Imai, C.; Mancini, A.J.; Kucuk, Z.Y.; Kawai, T.; Nishikomori, R.; Ito, E.; Pellier, I.; Dupuis Girod, S.; et al. Hematopoietic stem cell transplantation in 29 patients hemizygous for hypomorphic IKBKG/NEMO mutations. Blood 2017, 130, 1456–1467. [Google Scholar] [CrossRef] [Green Version]
  150. Critch, J.; Day, A.S.; Otley, A.; King-Moore, C.; Teitelbaum, J.E.; Shashidhar, H.; Committee, N.I. Use of enteral nutrition for the control of intestinal inflammation in pediatric Crohn disease. J. Pediatr. Gastroenterol. Nutr. 2012, 54, 298–305. [Google Scholar] [CrossRef] [Green Version]
  151. Heuschkel, R.B.; Menache, C.C.; Megerian, J.T.; Baird, A.E. Enteral nutrition and corticosteroids in the treatment of acute Crohn’s disease in children. J. Pediatr. Gastroenterol. Nutr. 2000, 31, 8–15. [Google Scholar] [CrossRef] [PubMed]
  152. Miller, T.L.; Lee, D.; Giefer, M.; Wahbeh, G.; Suskind, D.L. Nutritional Therapy in Very Early-Onset Inflammatory Bowel Disease: A Case Report. Dig. Dis. Sci. 2017, 62, 2196–2200. [Google Scholar] [CrossRef]
  153. Aloi, M.; Lionetti, P.; Barabino, A.; Guariso, G.; Costa, S.; Fontana, M.; Romano, C.; Lombardi, G.; Miele, E.; Alvisi, P.; et al. Phenotype and disease course of early-onset pediatric inflammatory bowel disease. Inflamm. Bowel Dis. 2014, 20, 597–605. [Google Scholar] [CrossRef] [PubMed]
  154. Letai, A.; Bhola, P.; Welm, A.L. Functional precision oncology: Testing tumors with drugs to identify vulnerabilities and novel combinations. Cancer Cell 2022, 40, 26–35. [Google Scholar] [CrossRef] [PubMed]
  155. Borg-Bartolo, S.P.; Boyapati, R.K.; Satsangi, J.; Kalla, R. Precision medicine in inflammatory bowel disease: Concept, progress and challenges. F1000Res 2020, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Summary of monogenic etiologies of VEO-IBD.
Table 1. Summary of monogenic etiologies of VEO-IBD.
Type of DisorderRepresentative SyndromesGenes
Epithelial cell defectsTTC7A deficiency
NEMO deficiency
Dystrophic epidermolysis bullosa
ADAM-17 deficiency
TTC7A
IKBKG
COL7A1
ADAM17
Phagocytic defectsChronic granulomatous diseaseLeukocyte adhesion deficiencyCYBA, CYBB, NCF1, NCF2, NCF4
ITGB2
Defects of adaptive immunity (T and B cells)Wiskott-Aldrich syndrome
Bruton agammaglobulinemia
Loeys-Dietz syndrome
Severe Combined Immunodeficiency (SCID)
WAS
BTK
TGFBR1, TGFBR2
ZAP70, RAG2, 1L-2RG, LIG4, ADA
T regulatory defectsIPEX
IPEX-like syndrome
CTLA4 deficiency
LRBA deficiency
FOXP3
STAT1, STAT3, JAK1, IL-2RA
CTLA4
LRBA
IL-10 pathway defectsIL-10
IL-10R
IL10
IL10RA, IL10RB
Hyperinflammatory or autoinflammatory defectsX-linked lymphoproliferative syndrome
Hermansky-Pudlak syndrome
Mevalonate kinase deficiency
XIAP
HPS1, HPS4, HPS6
MVK
Table 2. Therapeutic Drug Monitoring (TDM) for Monoclonal Antibodies and Calcineurin Inhibitors in IBD.
Table 2. Therapeutic Drug Monitoring (TDM) for Monoclonal Antibodies and Calcineurin Inhibitors in IBD.
DrugTiming of Trough LevelsGoal Trough Level (mcg/mL)
InfliximabInduction Phase (with 2nd and 3rd infusions)
Maintenance Phase (prior to 4th infusion)
Pediatric General (Maintenance): >5 mcg/mL [49]
Pediatric Perianal Fistulizing Disease (Maintenance): >12.7 mcg/mL [58]
Pediatric Luminal Crohn’s Disease (Induction): Infusion two and Infusion three: trough level of ≥29 mcg/mL at the 2nd infusion and ≥18 mcg/mL at 3rd infusion were strongly associated with improve outcomes early on in therapy and higher levels during maintenance phase [59]
Pediatric Luminal Crohn’s Disease (Induction/Maintenance): ≥25 mcg/mL at infusion two (week 2) and ≥15 mcg/mL at infusion three (week six) were associated with better outcomes [49]
Pediatric Ulcerative Colitis (Induction): trough level of ≥33 mcg/mL prior to the second dose was associated with clinical remission at eight weeks of therapy [60]. PK analysis showed that a trough level of ≥41.1 mcg/mL at week eight was associated with higher clinical remission, mucosal healing, and clinical response based on Mayo scoring [61,62]
AdalimumabMaintenance Phase (prior to 3rd injection)Pediatric General (Maintenance): >7.5 mcg/mL for endoscopic healing at week 8 [49]
Pediatric Crohn’s Disease (Maintenance): levels of >22.5 mcg/mL at week four and trough levels > 12.5 mcg at week eight were associated with prediction of clinical remission at week 24 [63]
Vedolizumab [64]Lack of data to suggest optimal timingLack of data to suggest optimal goal trough levels [49]
Vedolizumab trough concentrations are associated with clinical response and dose escalation may be required to maintain this response [65]
Pediatric IBD: average trough levels of 32.1 mcg/mL at week 2 of therapy and 29.9 mcg/mL at week six were observed in a cohort of 22 pediatric patients.; when delineated by type of IBD, trough levels in patients with UC/IBD-U were higher than in those with CD [66]
Dose escalation in adult IBD: vedolizumab trough level of <7.4 mcg/mL at eight weeks of therapy indicated probable response to dose escalation (dosing frequency of up to every four weeks) [65]
Adult study (maintenance): trough level of 33–37 mcg/mL at week 6,15–20 mcg/mL after induction (week 14) and 10–15 mcg during maintenance phase was associated with improved clinical outcomes [67]
UstekinumabLack of data to suggest optimal timingSingle case report in which a VEO-IBD patient achieved clinical remission with a trough level of 6 mcg/mL after 18 months of treatment (therapy started at age seven yrs) [68]
Adult studies: In one CD study: trough level of at least 2 mcg/mL at week eight was associated with clinical response to induction by week 16 of therapy [69]
  • Another study indicated a trough of at least 4.5 mcg/mL by week 26 of therapy to be associated with better endoscopic response in CD
  • Trough level of 0.8–1.4 mcg/mL was associated with clinical remission during the maintenance phase of the STARDUST trial (assessing CD patients) [67,70]
  • Trough level of 3–7 mcg/mL at week eight and 1–3 mcg/mL during maintenance for UC and CD [67]
Tacrolimus
Cyclosporine
Tacrolimus (pediatric): for severe colitis in children: 0.1 mg/kg/dose q12 with goal trough of 10–15 ng/ ml for induction therapy; one study reported decreasing the goal trough to 8–10 ng/dL once frank blood was absent; initial response rate has been shown to be similar to that of cyclosporine treated patients. Some guidelines recommend decreasing to 5–7 ng/mL once remission achieved [71,72]
CSA (adult): for UC in an RCT comparing 4 mg/kg/day via continuous infusion (goal trough range of 250–350 ng/mL) to 2 mg/kg/day of IV therapy (goal 150–250 ng/mL): high dose CSA was not shown to have additional clinical benefit compared to lower dosing [73,74]
CSA IV (pediatric): goal trough during induction with 2 mg/kg/day continuous infusion: 150–300 ng/mL and once remission achieved, may decrease to 100–200 ng/mL [71]
CSA PO (pediatric): goal trough of 150–300 ng/mL for 4–8 mg/kg/day of oral dosing in a retrospective study with 14 children, of which six were treated with CSA [75]; Second study with 28 patients started on 5 mg/kg/day of oral CSA while targeting goal trough levels of 150–250 ng/mL [76]
Abbreviations: CSA: cyclosporine; PK: pharmacokinetics; IBD: inflammatory bowel disease; CD: Crohn’s Disease; UC: Ulcerative Colitis: PO: by mouth; IV: intravenous.
Table 3. Baseline Screening and Monitoring for Immune-Targeted Therapies.
Table 3. Baseline Screening and Monitoring for Immune-Targeted Therapies.
MedicationMechanism of ActionScreening/Baseline LabsBlack Box Warnings/Common ADRs/Monitoring
Azathioprine
6-MP [89,90,91,92,93]
Purine analog, blocks DNA replication and proliferation of T-cells; possible inhibition of CD28 T-cell co-stimulationTPMT, NUDT15 prior to starting treatment
LFTs, CBC + diff: check at baseline, then every one to two weeks during the first month, then every three months
BBW: Malignancy (hepatosplenic T cell lymphoma)
Hematologic (leukopenia, thrombocytopenia, anemia)
Pancreatitis (would warrant discontinuation of the drug)
Gastrointestinal symptoms (nausea/vomiting), hepatotoxicity
Infliximab
Adalimumab [49,94,95]
Anti- TNFαTB status, hepatitis B, varicella, vaccination status; creatinine, fecal calprotectin, CRP, LFTs prior to starting treatmentBBW: Malignancy, TB, infection
Infliximab: infusion related reactions (cutaneous, psoriatic rash, elevated transaminases, infection
Adalimumab: injection site reaction, infection
Vedolizumab [49,64,88]Anti-α4β7 integrin; blockade of MAdCAM-1 directed lymphocyte traffic to intestinal Peyer’s patchesTB status prior to therapy initiation, LFTs prior to starting treatmentBBW: N/A
Low percentage of ADRs reported which have led to therapy discontinuation (5–10%)
ADA production is uncommon
Not associated with increased risk of malignancy or opportunistic infections
Hypersensitivity reactions may occur during infusion
Ustekinumab [96]Anti-IL12 and IL-23TB, hepatitis B, hepatitis C, HIV screening prior to starting therapy, CBC with differential, CMP, reversible posterior leuko-encephalopathy syndrome, ADAsBBW: N/A
Hypersensitivity reactions may occur during infusion
Increased risk of infection
Tacrolimus
Cyclosporine [71,72,76,97,98]
Suppression of IL-2, TNFα, and interferon-c production in T cellsSerum electrolytes (K, Mag, Phos), LFTs, renal function, blood pressure, glucose: should be checked three timesper week upon therapy initiation.
Serum cholesterol recommended before starting treatment with CSA. Frequency of lab checks may be spaced once stability has been shown
BBW for Tacrolimus: increased risk of infection and malignancy
BBW for CSA: increased infection, development of neoplasia
Tacrolimus: infection, malignancy (lymphoma, skin related cancers)
CSA: increased risk of infection, nephrotoxicity, and hypertension. Dosage forms may affect drug concentrations and bioavailability
Tacrolimus and CSA: Nephrotoxicity, serum electrolyte derangements (K, Mag, Phos), immune suppression, azotemia, gingival hyperplasia, hirsutism, tremor
DDIs: tacrolimus and cyclosporine metabolized by CYP3A enzymes (thorough drug interaction checking recommended when initiating or discontinuing medications for patients on calcineurin inhibitors)
Anakinra [99]
Canakinumab [100]
IL-1 antagonistTB and hepatitis B status prior to starting therapy, CBC with differential, LFTsBBW: N/A
Increased risk of infection
Signs of hypersensitivity reactions
CBC with differential q3 months up to one year, renal function
Abatacept [101]Cytotoxic T lymphocyte antigen-4 (CTLA4) immunoglobulin fusion moleculeTB and hepatitis status prior to starting therapyBBW: N/A
Hypersensitivity reactions
Increased risk of infection
Tofactinib [102]
Ruxolitinib [103]
JAK 1/2 and 1/3 inhibitorTofacitinib:
-
TB status prior to starting therapy
-
CBC with differential (hemoglobin, neutrophil count, lymphocyte count, platelets)
-
Heart rate and blood pressure
-
Renal function and LFTs
Ruxolitinib: CBC, renal function, hepatic function
BBW: malignancy (including solid tumors and lymphoma), higher infection risk, increased thrombotic risk
Tofacitinib: lipid abnormalities (dose- dependent), renal and hepatic impairment.
-
Lymphocyte count q3 months
-
CBC with differential (hemoglobin, neutrophil count, platelet count) every four to eight weeks, then every three months
-
Lipids: four to eight weeks after starting treatment
Ruxolitinib: CBC every two to four weeks until on stable dose, lipid count eight to twelve weeks after starting therapy, hepatic and renal function every two to four weeks until on stable dose (extrapolated from treatment of GVHD)
DDIs: Tofacitinib and Ruxolitinib are major CYP3A4 substrates (thorough drug interaction checking recommended when initiating or discontinuing medications for patients on JAK inhibitors)
TPMT: thiopurine methyltransferase; BBW: black box warning; ADR: N/A: not applicable; adverse drug reaction. TB: tuberculosis; ADA: anti-drug antibody; CRP: C-reactive protein; HIV: human immunodeficiency virus; CBC: complete blood count; CMP: complete metabolic panel; K: potassium; mag: magnesium; phos: phosphate; LFTs: liver function testes; DDI: drug–drug interaction; IV: intravenous; PO: by mouth; RCT: randomized controlled trial; CSA: cyclosporine; SrCr: serum creatinine.
Table 4. Time to Therapeutic Effect for Immune-Targeted Therapies.
Table 4. Time to Therapeutic Effect for Immune-Targeted Therapies.
MedicationTime to Full Therapeutic Effect/Clinical Remission
Azathioprine
6-MP
CD (adult): May take a minimum of eight weeks to achieve clinical remission; wide range reported ranging from two weeks to nine months [79,106]
UC (adult): typical range of three to six months to see clinical and endoscopic response [79,107]
UC and CD (pediatric): pediatric IBD network showed remission within 180 days of starting thiopurine therapy, with approximately 50% of patients reporting sustained steroid free remission at six months [108]. Range of eight to sixteen weeks has been reported to reach maximum effectiveness in CD [49]
Infliximab
Adalimumab
Infliximab: response is variable and patient dependent; requires monitoring of trough levels and ADAs. Many patients may require dose escalation within the first year of treatment [77,78,79]
CD (adult): clinical response and remission for infliximab reported in one study to take eight to nine days, with up to 81% of patients with clinical response rates after four weeks of therapy [77,78,79,109]
  • UC (adult): could be dose dependent; for acute severe colitis could expect response to infliximab to be within seven days [79,110]
  • CD (pediatric): 1/3 of patients are reported to lose response within one year (secondary loss of response); immunomodulator use has made a clinical difference in this loss of clinical response [50]
  • UC (pediatric): time to clinical response varies; infliximab was found to be safe and effective with a response by week eight of therapy in nearly 75% of pediatric patients with moderate to severe UC [82]
Adalimumab: Significant patient variability: clinical remission rates have ranged from two to twenty-four weeks in the literature in adults
  • CD (adult): initial response and remission for adalimumab reported within a few weeks; endoscopic remission would be longer, with response time ranging from 12–52 weeks [79,80]
  • UC (adult): clinical remission with adalimumab was seen as early as four weeks [79,81]
  • CD and UC (pediatric): wide range of clinical response times reported in the literature; clinical response seen ranging from two weeks to two years [83]
VedolizumabVedolizumab may be more effective in UC vs. CD. Lower percent of patients with mucosal healing shown in patients with CD compared to UC [49]
  • Response may take up to 16 weeks (some studies reported therapeutic onset to be between eight and twelve weeks); may require a bridging agent such a steroids until clinical response is seen [49]
  • 76% of UC patients and 42% of CD in a retrospective study who had failed anti-TNF therapy achieved clinical remission at week 14 [105]
Overall lower immunogenicity when compared to adalimumab or infliximab and lower percentage of patients with ADAs compared to anti-TNFs [104]
Patients without prior biologic exposure, less severe disease, and early response to vedolizumab may have higher rates of endoscopic and clinical remission [104]
UstekinumabSome evidence to show better response and higher rate of clinical remission in patients who are biologic non-naïve
In adult trials, clinical response was seen after eight weeks of treatment. Some studies suggest a timeframe of up to 14 weeks for clinical response, and up to six months for endoscopic remission
Addition of immunomodulator therapy does not appear to affect immunogenicity
Tacrolimus
Cyclosporine
Tacrolimus:
  • Generally used in steroid refractory UC
  • Response may be seen within the first 14 days after tacrolimus initiation (enteral dosage form).
  • May be used as a bridge while waiting for biologic therapy such as vedolizumab to take effect (this is post steroid and anti-TNF therapy failure); may take between eight and twelve weeks to see effects of vedolizumab
  • ECCO Guidelines suggested role in adult UC and proctitis: tacrolimus in patients with refractory UC extending beyond the rectum. Topical tacrolimus: clinical response may be seen after four weeks of therapy.
  • Limited evidence for use of tacrolimus enemas in pouchitis
CSA IV (adult): would have a predicted response in at least 50% of patients after four to five days of treatment, with full response seen by 14 days. Would typically begin with continuous infusion and then transition to enteral dosing [73]
CSA PO (pediatric): in a study of 28 children, clinical response was seen within seven to fifteen days after starting an enteral dose of 5 mg/kg/day with a goal target of 150–250 ng/ml [76,111]
Abbreviations: CSA: cyclosporine; IBD: inflammatory bowel disease; CD: Crohn’s disease; UC: ulcerative colitis; ADA: anti-drug antibodies; PO: by mouth; IV: intravenous.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Levine, A.E.; Mark, D.; Smith, L.; Zheng, H.B.; Suskind, D.L. Pharmacologic Management of Monogenic and Very Early Onset Inflammatory Bowel Diseases. Pharmaceutics 2023, 15, 969. https://doi.org/10.3390/pharmaceutics15030969

AMA Style

Levine AE, Mark D, Smith L, Zheng HB, Suskind DL. Pharmacologic Management of Monogenic and Very Early Onset Inflammatory Bowel Diseases. Pharmaceutics. 2023; 15(3):969. https://doi.org/10.3390/pharmaceutics15030969

Chicago/Turabian Style

Levine, Anne E., Dominique Mark, Laila Smith, Hengqi B. Zheng, and David L. Suskind. 2023. "Pharmacologic Management of Monogenic and Very Early Onset Inflammatory Bowel Diseases" Pharmaceutics 15, no. 3: 969. https://doi.org/10.3390/pharmaceutics15030969

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