Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Jan 1.
Published in final edited form as: J Allergy Clin Immunol Pract. 2023 Sep 28;12(1):69–78. doi: 10.1016/j.jaip.2023.09.016

PI3Kδ Pathway Dysregulation and Unique Features of Its Inhibition by Leniolisib in Activated PI3Kδ Syndrome and Beyond

Andrew J Cant a, Anita Chandra b, Ewen Munro c, V Koneti Rao d, Carrie L Lucas e
PMCID: PMC10872751  NIHMSID: NIHMS1941204  PMID: 37777067

Abstract

The phosphoinositide 3-kinase (PI3K) pathway regulates diverse cellular processes, with finely tuned PI3Kδ activity being crucial for immune cell development and function. Genetic hyperactivation of PI3Kδ causes the inborn error of immunity activated PI3Kδ syndrome (APDS). Several PI3Kδ inhibitors have been investigated as treatment options for APDS, but only leniolisib has shown both efficacy and tolerability. In contrast, severe immune-mediated adverse events such as colitis, neutropenia, and hepatotoxicity have been observed with other PI3Kδ inhibitors, particularly those indicated for hematologic malignancies. We propose that leniolisib is distinguished from other PI3Kδ inhibitors due to its structure, specific inhibitory properties selectively targeting the δ isoform without overinhibition of the δ or γ isoforms, and the precise match between APDS mechanism of disease and drug mechanism of action.

Keywords: leniolisib, PI3K, PI3Kδ, PI3Kδ inhibitor, mechanism of action, APDS, activated PI3Kδ syndrome

Overview of the Phosphoinositide 3-Kinase Pathway

Cell growth, proliferation, survival, motility, differentiation, and metabolism are affected by intracellular signaling cascades often involving AKT, forkhead box O (FOXO), glycogen synthase kinase-3 (GSK3), and mechanistic (or mammalian) target of rapamycin (mTOR).1,2 These cascades can be initiated by the phosphorylation of membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2) to generate phosphatidylinositol 3,4,5-trisphosphate (PIP3).3 This reaction—and control of the associated intracellular signaling cascades—is regulated by class I phosphoinositide 3-kinases (PI3Ks).3 These PI3Ks are heterodimers comprising distinct catalytic and regulatory subunits.1 The most-studied regulatory subunit, p85α, is encoded by PIK3R1 and maintains inhibitory contacts with the catalytic subunit. These contacts regulate stability and recruitment of the catalytic subunit to membrane-associated phosphoproteins that relieve its inhibitory activity and position the catalytic subunit near PIP2.1,3 The p110 catalytic subunit has 4 isoforms, α, β, γ, and δ, encoded by PIK3CA, PIK3CB, PIK3CG, and PIK3CD, respectively.3 The γ subunit is unique among the class I PI3Ks by binding alternative regulatory subunits.1 The α and β isoforms are expressed ubiquitously; γ and δ isoforms, mainly in immune cells.1,4 The γ isoform is primarily involved with innate immunity; the δ isoform, with adaptive immunity, albeit with overlap.1 B- and T-lymphocyte development and function are regulated partly by the degree of PI3Kδ activity.3 Activity must be dynamically regulated to permit FOXO-dependent transcription and periods of mTOR signaling.3 Therefore, homeostatic control of PI3Kδ activity is vital for immune health (Figure 1A).

Figure 1. PI3Kδ pathway in lymphocytes under physiological and pathological conditions.

Figure 1.

A) Under healthy physiological conditions, PI3Kδ activity in B and T cells is dynamically regulated during development and function, with periods of enhanced activity where FOXO and GSK3 are inhibited and mTOR signaling occurs and periods of dampened activity when mTOR is not activated and FOXO/GSK3 are disinhibited. Balanced PI3Kδ activity results in proper lymphocyte development and function.13 B) In APDS, PI3Kδ is hyperactive, resulting in excessive FOXO/GSK3 inhibition and mTOR activation. As a result, lymphocytes do not develop properly, with a general excess of immature or senescent cells and a deficit of functional cells. This deficiency and dysregulation create or contribute to the constellation of clinical manifestations, namely, infections, lymphoproliferation, autoimmunity, enteropathy, bronchiectasis, and an increased risk of malignancy, particularly lymphoma. Patients with APDS may also display neurological or cognitive symptoms, liver disease, and atopy. It is unclear whether some symptoms may be due to PI3Kδ expression in non-immune cells.59 Please note that PI3Kδ activity levels depicted are for illustrative purposes only and do not represent actual data. APDS indicates activated PI3Kδ syndrome; FOXO, forkhead box O; GSK3, glycogen synthase kinase-3; PI3Kδ, phosphoinositide 3-kinase δ.

PI3Kδ is also expressed at low levels in other cell types, where its expression can be further induced by specific stimuli. PI3Kδ is expressed in the central nervous system, where it may be involved with axon regeneration.10,11 Low levels of PI3Kδ are found in fibroblast-like synoviocytes, with increased expression in synovium of patients with rheumatoid arthritis.12 Tumor necrosis factor α with glucose can induce further expression of PI3Kδ in endothelial cells, where basal levels are low.13,14 In the lungs, PI3Kδ is expressed in fibroblasts and bronchial epithelial cells.15,16

PI3Kδ Pathway Dysregulation In Activated PI3Kδ Syndrome

PI3Kδ hyperactivation from gain-of-function variants in PIK3CD or loss-of-regulatory-function variants in PIK3R1 causes the inborn error of immunity activated PI3Kδ syndrome (APDS), first characterized in 2013.5,6,17,18 Pathogenic variants in p85α or p110δ that disrupt the inhibitory contacts between the two subunits, or in p110δ that promote kinase activity or increased interaction with the plasma membrane and thus access to PIP2, drive hyperactive PI3Kδ signaling.5,6,1821 Constitutively active PI3Kδ causes reduced or absent dynamic tuning of activity, resulting in excessive GSK3/FOXO inhibition and mTOR activation (Figure 1B).8 Hyperactivity can be approximated by measuring increased downstream phosphorylation of AKT (pAKT) or S6 (pS6).5

The hallmark of APDS is recurrent sinopulmonary infections, often associated with severe bronchiectasis, due to derangement of B- and T-cell development and function, causing both immune deficiency and dysregulation.5,6,10,2225 Transitional B cell levels, often elevated, may be entry points and reservoirs for Epstein-Barr virus (EBV).5,10,23,26,27 In contrast, levels of mature naïve and memory B cells are often decreased, along with defects in class-switch recombination.5,10,23,26,27 Hyperactive PI3Kδ signaling also drives terminal differentiation: IgM+ plasma cell levels are often increased in patients; many patients with APDS display elevated IgM levels, low IgG and/or IgA levels, and poor specific antibody production.5,10,23,2628 The T-cell compartment also displays a shift away from functional cells, with an inverted CD4+/CD8+ T-cell ratio, decreased naïve T-cell levels, and increased CD8+ effector memory and effector memory cells re-expressing CD45-RA (TEMRA) at the expense of long-lived central memory cells.5,6,10,23,26,27,29 These TEMRA cells have characteristics of highly inflammatory senescent (CD57+) CD8+ T cells and generate ineffective responses to infections, particularly EBV and cytomegalovirus, allowing lifelong infections that can be severe and result in end-organ damage and malignancy.5,6,2224

Lymphadenopathy, due to follicular hyperplasia, can further be driven by B-lymphocyte proliferation associated with increased levels of follicular helper and effector T cells, the latter demonstrating an enhanced proliferative burst on encounter with antigen.5,10,26 B- and T-cell proliferation also contributes to the splenomegaly and hepatomegaly observed in APDS.9,10,23,2628,30 B-cell proliferation may underlie the profound submucosal nodular lymphoid hyperplasia seen in many patients, leading to derangement of gastrointestinal or respiratory tract anatomy and functions.7,10,26

Autoimmunity, particularly cytopenias, may be caused partly by elevated levels of plasmablasts secreting autoreactive IgM as well as elevated levels of CD21low transitional B cells, as occurs in patients with other primary immune regulatory disorders, including common variable immune deficiency and monogenic diseases such as NF-κB1 or CTLA4 haploinsufficiency.3134 Additionally, studies in a mouse model of APDS show increased plasma cell numbers and elevated levels of natural antibodies, which may also contribute to autoimmunity.35

Less well-characterized manifestations of APDS include liver disease, neurodevelopmental delay, seizures, fatigue, and atopy, some of which may be due to PI3Kδ expression in non-immune cells.9,10,26,36,37 The more ubiquitous expression of p85α may account for the higher prevalence of failure to thrive and neurodevelopmental delay in patients with PIK3R1 defects.10,26,30 Interestingly, bronchiectasis and autoimmune cytopenias have been reported as less prevalent in patients with PIK3R1 variants.10,26,30

Historically, there has been no standard of care and management of APDS has remained largely empirical. Most therapies do not directly target disease pathogenesis nor address the full complexity of APDS. Immune deficiency is treated with prophylactic antimicrobials and immunoglobulin replacement therapy (IRT), while immune dysregulation is generally treated with immunomodulatory therapies such as corticosteroids or mTOR inhibtors, and/or surgical procedures.10,26,30 Hematopoetic stem cell transplantation may be considered curative but carries the risk of serious complications including increased rates of graft rejection, and may not correct non-immune/hematopoietic manifestations.37

PI3Kδ Inhibitors in APDS

As APDS is caused by PI3Kδ overactivity, PI3Kδ inhibitors have been investigated as treatment options, albeit in studies with small sample sizes given the rarity of the disease. A 12-week, open-label, phase 2 trial of the once-daily inhaled PI3Kδ inhibitor nemiralisib for patients with APDS (n=5) demonstrated no efficacy and tolerable adverse events (AEs).38 No deaths or severe AEs were reported. The most common AEs were cough, headache, and nasopharyngitis (all n=3). There were no changes in PIP3 or downstream inflammatory markers in induced sputum, nor were there changes in blood inflammatory markers or lymphocyte subsets. The authors speculate that lack of efficacy could be due to nemiralisib not being retained in the lung long enough.

Two trials have studied oral, systemic PI3Kδ inhibition in APDS. A 12-week phase 1b open-label trial of the PI3Kδ inhibitor seletalisib (15-25 mg once daily) demonstrated moderate efficacy and AEs leading to discontinuation.39 However, of the 7 patients receiving drug, only 5 completed the study, with 2 discontinuing because of study drug–related AEs in the liver. Four patients entered the extension study, 3 completing ≥84 weeks of treatment (1 withdrew consent at week 36 for personal reasons). Across both parts of the study, aphthous ulcers and deranged liver function were the most common drug-related AEs, including a potentially drug-induced liver injury in one patient. One patient also experienced a severe AE of colitis.

The second oral, systemic PI3Kδ inhibitor trial was a 12-week, open-label, within-patient, dose-escalation study of patients with APDS (N=6) that demonstrated leniolisib treatment inhibited hyperactive PI3Kδ, as assessed by pAKT and pS6, ameliorated immune dysregulation, and increased patient well-being.40 Based on the results of the dose-finding study, a phase 3, randomized 2:1, placebo-controlled trial of 70-mg leniolisib twice daily was conducted in patients with APDS (N=31).41 Both primary end points—increase in the percentage of naïve B cells and reduction in lymph node size—were met. Compared with placebo (n=8), leniolisib (n=18) significantly reduced lymphadenopathy by day 85, as measured by the decreased log10-transformed sum of product diameters of index lymph nodes (p=0.0006), and secondary analyses revealed significant reductions in spleen size. Exploratory analyses of baseline cytopenias, which varied among patients and included patients with multiple cytopenias, showed 14 of 17 cytopenias (82%) improved or resolved in patients receiving leniolisib, while 3 of 5 (60%) improved or resolved in patients receiving placebo. However, this study was not designed to specifically assess leniolisib efficacy in cytopenias. Leniolisib (n=8) significantly increased naïve B-cell levels versus placebo (n=5) by day 85 (p=0.0002), meeting the other primary end point. Exploratory analyses revealed that leniolisib improved other key immune parameters, decreasing levels of IgM, transitional B cells, senescent T cells, and programmed cell death protein 1+ T cells that are often elevated in APDS.42 Additionally, a reduction in the annualized infection rate by 0.351 (p=0.0040) was seen in a post hoc interim analysis of an open-label extension study (N=37) that followed patients with exposures up to 5 years (median, approximately 2 years).43,44 During this extension study, immunoglobulin replacement therapy (IRT) use relative to baseline reduced in 10 of 27 patients (37%), including 6 who achieved IRT freedom, of whom 4 had been IRT-free for 1-2.5 years at the time of data cutoff in December 2021. The interim analysis also demonstrated durability of effect, with continued improvement in lymphadenopathy, splenomegaly, cytopenias, and regulation of immune cells.43,44 Overall, the APDS studies show that leniolisib treatment changed molecular biomarkers and improved clinical manifestations, as well as a reduction in an existing prescribed therapy. In March 2023, leniolisib received FDA approval for the treatment of APDS in adults and children ≥12 years of age.41

Across all 3 studies, leniolisib was well tolerated, and the severe AEs observed with other PI3Kδ inhibitors used to treat malignancy were not seen with leniolisib treatment in patients with APDS (Tables I and II). In the dose-escalating study (N=6), leniolisib was well tolerated at all doses, with no significant neutropenia, hypertriglyceridemia, hyperglycemia, gastrointestinal disturbances, skin rashes, or liver toxicity reported.40 In the phase 3 randomized trial (N=31), study drug-related AEs occurred in 8 patients across both the leniolisib (23.8%) and placebo (30.0%) arms. Leniolisib-related AEs included transient alopecia (n=2), aphthous ulcer, taste disorder, vomiting, and vertigo (all n=1); none were serious. In total, 5 patients reported a serious AE (SAE), with none judged as related to study medication. Leniolisib remained well tolerated in the open-label extension study (N=37).43,44 Study drug–related AEs, including weight increase (n=3), arthralgia, hyperglycemia, and decreased neutrophil count (all n=1), occurred in 5 patients. SAEs occurred in 6 patients; none were related to leniolisib treatment. Including non–drug-related AEs, 86.5% of patients with up to 5 years of exposure experienced an AE, 78.4% of which were grade 1 and none of which were grade 4. Across all 3 studies, no discontinuations or interruptions of leniolisib occurred due to treatment-related AE. One death occurred in a patient who had significant baseline comorbidities, including cardiomyopathy, tachycardia, recurrent pneumonia, necrotizing lymphadenitis, disseminated Mycoplasma infection, bronchiectasis, pancytopenia, liver disease, and peripheral edema. This patient experienced cardiac arrest resulting in death, determined not related to study drug, at extension day 879.

Table I.

FDA-Approved or Formerly Approved PI3Kδ Inhibitors

Agent Target Current Approvals AEs Withdrawals of Approvals or NDA
Copanlisib (BAY 80-6946)45 Pan-PI3K inhibitor, but predominantly PI3Kα and PI3Kε Relapsed FL (2017; accelerated approval based on single-arm trial) Warnings: infections, hyperglycemia, hypertension, non-infectious pneumonitis, neutropenia, and severe cutaneous reactions
Other AEs: diarrhea, decreased general strength and energy, and nausea
Laboratory abnormalities: leukopenia and thrombocytopenia
NDA for indolent NHL withdrawn Dec 2021 for additional analyses from ongoing trials46
Idelalisib (CAL-101, GS-1101)47 PI3Kδ Relapsed CLL in combination with rituximab (2014) Fatal and serious toxicities: hepatotoxicity, severe diarrhea or colitis, pneumonitis, infections, and intestinal perforations
Warnings: severe cutaneous reactions, hypersensitivity reactions, and neutropenia
Other AEs: pyrexia, fatigue, rash, cough, and nausea
Laboratory abnormalities: ALT and AST elevations
Relapsed follicular B-cell NHL and relapsed SLL indications also granted accelerated approval based on single-arm trial in 2014, but voluntarily withdrawn Feb 2022 due to inability to provide evidence to verify clinical benefit due to enrollment challenges48
Duvelisib (IPI-145)49 PI3Kγ and PI3Kδ Relapsed or refectory: CLL, SLL (2018) Fatal and serious toxicities: infections, diarrhea or colitis, cutaneous reactions, and pneumonitis
Warnings: hepatotoxicity and neutropenia
Other AEs: rash, fatigue, pyrexia, cough, nausea, musculoskeletal pain, and anemia



FDA drug safety communication Jun 2022 warning of possible increased risk of death and SAEs50
FL indication also granted accelerated approval based on single-arm trial in 2018, but voluntarily withdrawn Dec 2021 due to inability to provide evidence to verify clinical benefit due to trial feasibility issues and commercial reasons51
Umbralisib (TGR-1202)52 PI3Kδ and CK1ε None Warnings: fever, infection, neutropenia, diarrhea or non-infectious colitis, hepatotoxicity, severe cutaneous infections, allergic reactions due to inactive ingredient FD&C yellow No. 5, and embryo-fetal toxicity
Other AEs: fatigue, nausea, musculoskeletal pain, anemia, thrombocytopenia, vomiting, abdominal pain, decreased appetite, and rash
Laboratory abnormalities: increased creatine and transaminase elevation
Approval withdrawn: was granted accelerated approval based on single-arm studies for relapsed or refractory MZL or FL, both after specific prior therapies (2021). Withdrawn voluntarily by company in Apr 2022 and by FDA in Jun 2022 after FDA issued safety alert in Feb 2022 for possible increased risk of death after review of trial for CLL/SLL NDA53,54
Leniolisib (CDZ173)41 PI3Kδ APDS (2023) Other AEs: headache, sinusitis, and atopic dermatitis None

Abbreviations: AEs, adverse events; ALT, alanine aminotransferase; APDS, activated PI3Kδ syndrome; AST, aspartate aminotransferase; CK1ε, casein kinase 1 epsilon; CLL, chronic lymphocytic leukemia; FDA, Food and Drug Administration; FD&C, food, drugs, and cosmetics; FL, follicular lymphoma; MZL, marginal zone lymphoma; NDA, New Drug Application; NHL, non-Hodgkin lymphoma; PI3Kδ, phosphoinositide 3-kinase δ; SAEs, serious adverse events; SLL, small lymphocytic lymphoma.

Table II.

Summary of AEs of Approved or Formerly Approved PI3Kδ Inhibitors When Administered as Monotherapy for Malignancies and Leniolisib When Administered for APDS

Copanlisib
(N=244)46,a
Idelalisib
(N=146)46,b
Duvelisib
(N=442)46,c
Umbralisib
(N=371)46,d
Leniolisib
(N=38)e
Median exposure (range), mo 4.3 (0.2, 47.2) 6.1 (0.3, 26.4) 9.0 (0.1, 53.0) 5.9 (0.1, 75.1) 23.6 (2.8, 60.3)
Toxicity, %
Death due to drug-related AE 4 5 4 1 0
Grade ≥3 AE 85 71 84 51 29
SAE 51 50 65 26 21
AEs of special interest, %
Grade ≥3 infection 23 23 27 20 5
Grade ≥3 neutropeniaf 29 28 43 17 13
Grade ≥3 diarrhea/colitis 5 14 23 7 3
Grade ≥3 ALT/AST increasef 2 18 8 7 11g
Grade ≥3 rash 2 4 9 3 0
Any grade pneumonitis 7 5 7 1 0
Actions due to any AE, %
Discontinuation 24 23 35 15 3
Dose reduction 24 41h 23 10 0
Dose interruption 64 41h 64 45 13
a

N includes 52% FL, 11% MZL, 9% DLBCL, 5% CLL, 4% MCL, 4% SLL, 3% LPL/WM, and 12% other hematologic malignancies. Copanlisib was administered 60 mg IV on days 1, 8, and 15 of a 28-day treatment cycle.

b

N includes 60% FL, 21% SLL, 11% MZL, and 8% LPL. Idelalisib was administered orally 150 mg twice-daily in a 28-day treatment cycle.

c

N includes 64% CLL, 22% FL, 9% SLL, 4% MZL, and 1% other hematologic malignancies. Duvelisib was administered orally 25 mg twice-daily in a 28-day treatment cycle.

d

N includes 40% FL, 22% MZL, 20% DLBCL and MCL, 12% CLL or SLL, and 6% other hematologic malignancies. Umbralisib was administered orally 800 mg once daily in a 28-day treatment cycle.

e

N includes data from a dose-finding study, RCT, and open-label extension. Leniolisib was administered orally 10, 30, or 70 mg twice-daily for 4 weeks each in the dose-finding study, 70 mg twice-daily for 85 days in the RCT, and 70 mg twice-daily for ≥5 years in the open-label extension.

f

Based on analysis of laboratory data.

g

Based on investigator-reported grade 3 AEs.

h

A dose reduction or interruption due to an AE occurred in 41% of patients.

Cross-trial comparisons of safety are not intended.

Abbreviations: AEs, adverse events; ALT, alanine aminotransferase; APDS, activated PI3Kδ syndrome; AST, aspartate aminotransferase; DLBCL, diffuse large B-cell lymphoma; FL, follicular lymphoma; IV, intravenously; LPL, lymphoplasmacytic lymphoma; MZL, marginal zone lymphoma; PI3Kδ, phosphoinositide 3-kinase δ; RCT, randomized controlled trial; SAE, serious adverse event; WM, Waldenström macroglobulinemia.

Skin rash is a known class effect of PI3Kδ inhibitors used for malignancy. Notably, only one patient in the leniolisib APDS phase 3 study reported skin rash (grade 1 maculopapular rash), which was deemed fungal and not related to leniolisib by the investigator. Severe gastrointestinal AEs can occur with use of other PI3Kδ inhibitors over time. In the open-label extension study, none were reported related to study treatment, and 85.3% of gastrointestinal AEs that did occur were grades 1-2. Furthermore, 10 of the 15 patients in whom these occurred had a prior history of gastrointestinal disease. Transaminitis is another effect of PI3Kδ inhibition. While grade ≥3 AST/ALT increase ranged from 2-18% with inhibitors used for malignancy, 11% (n=4) of patients receiving leniolisib had transaminitis. For 3 of these patients, increased AST/ALT resolved; the last patient died from cardiac arrest while levels were elevated. Notably, laboratory values were used to determine grade ≥3 AST/ALT increase for the malignancy treatments,46 while investigator reports were used for leniolisib.

Severe neutropenia has also been observed with PI3Kδ inhibitor use for malignancies. Across all leniolisib APDS trials (N=38), transient neutropenia developed in 14 patients: mild (1.0-1.5 x 109/L) in 9 patients and moderate (0.5-1.0 x 109/L) in 5 patients. In all cases, neutropenia resolved without leniolisib withdrawal. No unresolved or severe infections were concurrent with the neutropenias. Notably, grade 3 neutropenia was observed in 13% of patients, whereas grade 3 or higher neutropenia has been observed in 17%-43% of patients receiving other PI3Kδ inhibitors for malignancies.46 Neutropenia has also been observed in healthy volunteers receiving leniolisib (unpublished data). In these studies (leniolisib, n=118; placebo, n=70), mean absolute neutrophil count (ANC) remained within normal limits. In the 70-mg twice-daily group, small mean reductions in ANC were observed post-dosing over a two-week period, with means ranging from 2.66-3.17 x 109/L (n=15-22). Only 3 individuals in this group had levels below normal limits, and only a single ANC was below 1 x 109/L (0.9 x 109/L on day 15; repeat assessment read 1 x 109/L).

Impact of the Properties of PI3Kδ Inhibitors on AEs

The causes of PI3Kδ inhibition–associated AEs in APDS and malignancies are poorly understood (Table I).45,47,49,52,55 These AEs may be due to on-target effects of the agents, and reductions in dosage or cessation of treatment until resolution is advised when treating malignancy.47,56,57 For instance, autoimmune AEs seen with idelalisib, such as colitis, hepatotoxicity, transaminitis, and pneumonitis, may be mediated by reduced suppressive functions of regulatory T cells leading to a cytotoxic CD8+ T-cell response, as revealed by ex vivo human cell models.58 Another ex vivo human cell model suggested neutropenia may be due to inhibition of neutrophil polarization and directional migration, although the precise mechanism leading to neutropenia is unknown.59 PI3Kδ inhibition potentially contributing to genomic instability has been noted in one study of ex vivo mouse B cells via increased expression of activation-induced cytokine deaminase (AID).60 Those experiments were performed in cells not exhibiting the hyperactive PI3Kδ pathway activation seen in APDS; thus, PI3Kδ inhibitor use did not normalize, but rather bluntly suppressed, pathway activity. In fact, B cells expressing the APDS E1021K PIK3CD variant showed decreased AID expression and chromosomal translocations, suggesting that use of PI3Kδ inhibitors in patients with APDS may not increase AID to harmful levels.

Toxicities and increased rates of death from other PI3Kδ inhibitors have led to voluntary withdrawal of New Drug Applications (NDAs) and approved malignancy indications (Table I).46,48,50,51,53,54,61 Many initial malignancy approvals were accelerated and based on single-arm studies. Post marketing randomized trials revealed SAEs, and increased rates of death led to voluntary withdrawals. Other voluntary withdrawals resulted from additional analyses of ongoing trials, enrollment challenges, or decisions not to pursue further trials.46,48,50,51,53,54 Consequently, the Food and Drug Administration (FDA) is encouraging dose-finding studies, and intermittent dosing schedules are being investigated for many agents.46 Intermittent dosing regimens in mouse models have recently shown reductions in immune-mediated AEs.62 These data suggest that excessive inhibition of PI3K activity disrupts homeostasis via PI3Kδ hypoactivation and yields on-target adverse effects.

Additionally, the level of inhibition may be influenced by the chemical structure of the inhibitors (Figure 2). Many PI3Kδ inhibitors, including idelalisib, duvelisib, and umbralisib, are propeller-shaped.56 They achieve their selectivity by wedging between Met752 and Trp760 in the active site of the catalytic domain of p110δ to open what is known as a “specificity pocket.”66,69 In contrast, multi- and pan-selective class I PI3K inhibitors with a flat conformation such as copanlisib do not bind the specificity pocket.64,69,70 Leniolisib is structurally unique relative to other approved PI3Kδ inhibitors; it is not propeller-shaped. Instead of binding to the specificity pocket, leniolisib uses a “tryptophan shelf” and stacks with Trp760 in p110δ, whereas the corresponding interaction with Trp780 in p110α is prevented, thus conferring specificity for the δ isoform.68,71

Figure 2. Leniolisib binds PI3Kδ differently than other approved inhibitors.

Figure 2.

A) Copanlisib, a flat, pan-PI3K inhibitor, bound to p110γ. Copanlisib does not wedge between Met804 and Trp812 (lime green), equivalent to Met752 and Trp760 in p110δ, which form the specificity pocket. PDB ID: 5G2N.63,64 B) Idelalisib, a propeller-shaped PI3Kδ-specific inhibitor, bound to p110δ. Idelalisib wedges between Met752 and Trp760 (lime green) to open the specificity pocket to achieve specificity for PI3Kδ. PDB ID: 4XE0.65,66 C) Leniolisib, a PI3Kδ-specific inhibitor, bound to p110δ. While leniolisib does not open the specificity pocket formed by Met752 and Trp760 (lime green), it achieves selectivity by stacking with Trp760, which forms a tryptophan shelf. PDB ID: 5O83.67,68 Blue, inhibitor; gray, p110 protein; lime green, key residues. Dashed lines indicate hydrogen bonds. PDB ID indicates Protein Data Bank identification; PI3Kδ, phosphoinositide 3-kinase δ. Images from the Research Collaborator for Structural Bioinformatics PDB (rcsb.org).

On an equimolar basis, leniolisib is less chemically potent at inhibiting PI3Kδ than approved inhibitors targeting PI3Kδ (Table III).68,7274 In cell-free assays, the half-maximal inhibitory concentration (IC50; where a lower value indicates that a lower concentration of drug is needed to inhibit 50% of the response) of leniolisib was higher than that of idelalisib, duvelisib, and copanlisib. Additionally, leniolisib appears to be much more specific to the δ isoform than the γ isoform, unlike idelalisib, which also appears to inhibit γ, and duvelisib, which is a γ/δ inhibitor. As both the γ and δ isoforms are expressed in immune cells, some side effects of idelalisib treatment may be due to dual inhibition.56,76 Although umbralisib does not appear to bind γ and binds δ less potently than leniolisib, it is also an inhibitor of CK1ε, which may contribute to AEs. Of note, although seletalisib’s IC50 for PI3Kδ is similar to that of leniolisib at 12 nM, its IC50 for the γ subunit is much lower at 282 nM (compared with leniolisib’s 2230 nM), and the corresponding δ/γ ratio is 0.0426 versus 0.0049 for leniolisib, which may contribute to the differences in outcomes between seletalisib and leniolisib in APDS.68,77

Table III.

PI3K Isoform Selectivity of Leniolisib vs Other (Previously) Approved PI3Kδ Inhibitors

Biochemical IC50 (nM)
Agent PI3Kα PI3Kβ PI3Kγ PI3Kδ δ/γ ratio
Copanlisib73 0.5 3.7 6.4 0.7 0.1094
Idelalisib72 820 565 89 2.5 0.0281
Duvelisib74 1602 85 27 2.5 0.0926
Umbralisib75 >10,000 1116 1065 22 0.0207
Leniolisib68 244 424 2230 11 0.0049

Abbreviations: IC50, half-maximal inhibitory concentration; PI3Kδ, phosphoinositide 3-kinase δ.

The overall lack of toxicity observed with leniolisib treatment in patients with APDS may highlight the benefit of fine-tuning rather than blocking PI3Kδ signaling, as both hyperactive and hypoactive signaling can result in immune deficiency and dysregulation.30,41,56,78,79 The unique pathomechanism of APDS, combined with a higher IC50 for the δ and γ isoforms than other PI3Kδ inhibitors, may enable leniolisib to generate better balance of high and low activity necessary for appropriate immune cell development and function in patients with APDS (Figure 3).

Figure 3. Tightly balanced PI3Kδ signaling is required for proper immune development and drug tolerability.

Figure 3.

A) Both overactivation of PI3Kδ (as seen in APDS) and overinhibition of PI3Kδ (as seen with inhibitor AEs and in patients with PI3Kδ loss-of-function) can disrupt the immune system and result in immune manifestations, highlighting the need for tightly balanced signaling.56,78,79 B) Leniolisib may be effective and tolerable in APDS by reducing continuous PI3Kδ hyperactivation without overinhibiting the kinase, which may result from lower affinity for the δ isoform than other FDA-approved inhibitors. These inhibitory properties and improved specificity may be due to the unique structure and binding method of leniolisib, which additionally largely spares the γ isoform to avoid off-target immune effects. Combined with precise matching of leniolisib MOA with APDS MOD, these features may explain why leniolisib demonstrated efficacy and tolerability in the APDS trials. AEs indicates adverse events; APDS, activated PI3Kδ syndrome; FDA, Food and Drug Administration; IC50, half-maximal inhibitory concentration; Ig, immunoglobulin; MOA, mechanism of action; MOD, mechanism of disease; PI3Kδ, phosphoinositide 3-kinase δ; Tfh, T follicular helper cell.

Leniolisib In Other Conditions

Although 70-mg leniolisib twice-daily appears to be safe and effective in APDS, leniolisib at that dose may not be appropriate for conditions in which hyperactive PI3Kδ is involved in, but not the cause of, disease. For instance, in a randomized, double-blind, phase 2 trial for primary Sjögren syndrome (pSS; N=30), 70-mg leniolisib twice-daily was not effective, and although it had an acceptable safety and tolerability profile, skin rashes occurred in 11/20 patients.80,81 pSS is an autoimmune condition characterized by B-cell hyperactivation, and although PI3Kδ hyperactivity is observed in patients with pSS, it is not the mechanism of disease (MOD).82,83 Dose range finding will be important in indications in which the level of PI3Kδ activity may be different from that in APDS.

Future Directions and Outstanding Questions

In APDS, leniolisib normalizes PI3Kδ activity, thereby modifying disease activity through restoring immune function. In the absence of easy ways to measure cellular PI3K activity, it is difficult to know whether PI3Kδ activity is completely normalized. A commercially accessible method to monitor pAKT or other markers of pathway activity or inhibition would be helpful.

The impact of long-term leniolisib use is being investigated. A small number of patients in the APDS open-label extension study have been receiving leniolisib for >6 years with good long-term tolerability. Patient registries such as the European Society for Immunodeficiencies Level 3 APDS registry will also be able to provide long-term data on lung function, vaccine responses, and IRT use. The effects of leniolisib on pregnant women, their fetuses, nursing children, and pediatric populations in general also require further study. A pediatric trial is recruiting (NCT05438407).

The emergence of drug resistance is not uncommon in this class of inhibitors when used to treat malignancy and remains to be studied in leniolisib; however, we suspect this concern may be specific to cancer, because in APDS, PI3Kδ hyperactivity is the result of germline variants, whereas cancers can coopt other non-PI3Kδ pathways to survive.

As PI3Kδ hyperactivity is not unique to APDS, the potential to use leniolisib, likely with different dosing, in other disease states, needs exploration. Indications to evaluate include those for which mTOR inhibitors are used, because leniolisib targets molecules further upstream in the PI3K-AKT-mTOR pathway. As discussed for pSS, the precise mechanism of PI3Kδ inhibition in these diseases needs careful consideration. The utility of leniolisib in patients with lymphoma who have increased PI3Kδ activity but no germline variants is unknown.

In conclusion, an unmet need exists for PI3Kδ inhibitors that are effective and tolerable in diseases driven by hyperactive PI3Kδ signaling, such as APDS. Leniolisib demonstrates disease-modifying properties in APDS, and by virtue of its novel structure, selectivity, and inhibitory properties, it may confer a more tolerable safety profile than other PI3Kδ inhibitors, particularly when the levels of inhibition by leniolisib are precisely matched to the level of PI3Kδ hyperactivity.

Acknowledgements

Technical editorial and medical writing assistance were provided by Carolyn Keating, PhD and Amy FitzPatrick, MS at PRECISIONscientia. Funding for this support was provided by Pharming Healthcare, Inc.

Disclosures

AJC and AC receive consulting fees, honoraria, and meeting/travel support from Pharming Group NV. AC also receives meeting/travel support from CSL Behring and Takeda, participates in advisory boards for Pharming Group NV, and is a principal investigator for the ESID level 3 APDS registry. CLL receives consulting fees from Pharming Healthcare Inc and unrelated research funding from Ono Pharma. VKR received meeting/travel support from Pharming Healthcare Inc and participates in advisory boards for Pharming Healthcare Inc. EM is currently employed by Pharming Group NV.

Abbreviations:

AE

adverse event

AID

activation-induced cytokine deaminase

ANC

absolute neutrophil count

ALT

alanine aminotransferase

APDS

activated PI3Kδ syndrome

AST

aspartate aminotransferase

CK1ε

casein kinase 1 epsilon

CLL

chronic lymphocytic leukemia

CTLA4

cytotoxic T-lymphocyte associated protein 4

DLBCL

diffuse large B-cell lymphoma

EBV

Epstein-Barr virus

FDA

Food and Drug Administration

FD&C

food, drugs, and cosmetics

FL

follicular lymphoma

FOXO

forkhead box O

GSK3

glycogen synthase kinase-3

IC50

half-maximal inhibitory concentration

Ig

immunoglobulin

IRT

immunoglobulin replacement therapy

IV

intravenously

LPL

lymphoplasmacytic lymphoma

MOA

mechanism of action

MOD

mechanism of disease

mTOR

mechanistic [or mammalian] target of rapamycin

MZL

marginal zone lymphoma

NDA

New Drug Application

NF-κB1

nuclear factor kappa-light-chain-enhancer of activated B cells-1

NHL

non-Hodgkin lymphoma

pAKT

phosphorylated AKT

PDB ID

Protein Data Bank identification

PDK1

3-phosphoinositide-dependent protein kinase-1

PIP2

phosphatidylinositol 4,5-bisphosphate

PIP3

phosphatidylinositol 3,4,5-trisphosphate

PI3K

phosphoinositide 3-kinase

PKB

protein kinase B

pSS

primary Sjögren syndrome

pS6

phosphorylated S6

RCT

randomized controlled trial

SAE

serious adverse event

SLL

small lymphocytic lymphoma

TEMRA

T effector memory cells expressing CD45RA

Tfh

T follicular helper cell

WM

Waldenström macroglobulinemia

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC, Abraham RT. The PI3K pathway in human disease. Cell. 2017;170:605–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gold MR, Scheid MP, Santos L, Dang-Lawson M, Roth RA, Matsuuchi L, et al. The B cell antigen receptor activates the Akt (protein kinase B)/glycogen synthase kinase-3 signaling pathway via phosphatidylinositol 3-kinase. J Immunol. 1999;163:1894–905. [PubMed] [Google Scholar]
  • 3.Okkenhaug K. Signaling by the phosphoinositide 3-kinase family in immune cells. Annu Rev Immunol. 2013;31:675–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Chantry D, Vojtek A, Kashishian A, Holtzman DA, Wood C, Gray PW, et al. p110delta, a novel phosphatidylinositol 3-kinase catalytic subunit that associates with p85 and is expressed predominantly in leukocytes. J Biol Chem. 1997;272:19236–41. [DOI] [PubMed] [Google Scholar]
  • 5.Lucas CL, Kuehn HS, Zhao F, Niemela JE, Deenick EK, Palendira U, et al. Dominant-activating germline mutations in the gene encoding the PI(3)K catalytic subunit p110δ result in T cell senescence and human immunodeficiency. Nat Immunol. 2014;15:88–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lucas CL, Zhang Y, Venida A, Wang Y, Hughes J, McElwee J, et al. Heterozygous splice mutation in PIK3R1 causes human immunodeficiency with lymphoproliferation due to dominant activation of PI3K. J Exp Med. 2014;211:2537–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Nunes-Santos CJ, Uzel G, Rosenzweig SD. PI3K pathway defects leading to immunodeficiency and immune dysregulation. J Allergy Clin Immunol. 2019;143:1676–87. [DOI] [PubMed] [Google Scholar]
  • 8.Preite S, Gomez-Rodriguez J, Cannons JL, Schwartzberg PL. T and B-cell signaling in activated PI3K delta syndrome: from immunodeficiency to autoimmunity. Immunol Rev. 2019;291:154–73. [DOI] [PubMed] [Google Scholar]
  • 9.Oh J, Garabedian E, Fuleihan R, Cunningham-Rundles C. Clinical manifestations and outcomes of activated phosphoinositide 3-kinase δ syndrome from the USIDNET cohort. J Allergy Clin Immunol Pract. 2021;9:4095–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Coulter TI, Chandra A, Bacon CM, Babar J, Curtis J, Screaton N, et al. Clinical spectrum and features of activated phosphoinositide 3-kinase δ syndrome: a large patient cohort study. J Allergy Clin Immunol. 2017;139:597–606.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Nieuwenhuis B, Barber AC, Evans RS, Pearson CS, Fuchs J, MacQueen AR, et al. PI 3-kinase delta enhances axonal PIP(3) to support axon regeneration in the adult CNS. EMBO Mol Med. 2020;12:e11674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bartok B, Boyle DL, Liu Y, Ren P, Ball ST, Bugbee WD, et al. PI3 kinase δ is a key regulator of synoviocyte function in rheumatoid arthritis. Am J Pathol. 2012;180:1906–16. [DOI] [PubMed] [Google Scholar]
  • 13.Whitehead MA, Bombardieri M, Pitzalis C, Vanhaesebroeck B. Isoform-selective induction of human p110δ PI3K expression by TNFα: identification of a new and inducible PIK3CD promoter. Biochem J. 2012;443:857–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wu W, Zhou G, Han H, Huang X, Jiang H, Mukai S, et al. PI3Kδ as a novel therapeutic target in pathological angiogenesis. Diabetes. 2020;69:736–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Conte E, Fruciano M, Fagone E, Gili E, Caraci F, Lemmolo M, et al. Inhibition of PI3K prevents the proliferation and differentiation of human lung fibroblasts into myofibroblasts: the role of class I P110 isoforms. PLoS One. Preprint posted online October 3, 2011. doi: 10.1371/journal.pone.0024663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fujita A, Kan OK, Tonai K, Yamamoto N, Ogawa T, Fukuyama S, et al. Inhibition of PI3Kδ enhances poly I:C-induced antiviral responses and inhibits replication of human metapneumovirus in murine lungs and human bronchial epithelial cells. Front Immunol. Preprint posted online March 11, 2020. doi: 10.3389/fimmu.2020.00432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Angulo I, Vadas O, Gargon F, Banham-Hall E, Plagnol V, Leahy TR, et al. Phosphoinositide 3-kinase δ gene mutation predisposes to respiratory infection and airway damage. Science. 2013;342:866–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Deau MC, Heurtier L, Frange P, Suarez F, Bole-Feysot C, Nitschke P, et al. A human immunodeficiency caused by mutations in the PIK3R1 gene. J Clin Invest. 2014;124:3923–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Dornan GL, Siempelkamp BD, Jenkins ML, Vadas O, Lucas CL, Burke JE. Conformational disruption of PI3Kδ regulation by immunodeficiency mutations in PIK3CD and PIK3R1. Proc Natl Acad Sci U S A. 2017;114:1982–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Takeda AJ, Zhang Y, Dornan GL, Siempelkamp BD, Jenkins ML, Matthews HF, et al. Novel PIK3CD mutations affecting N-terminal residues of p110δ cause activated PI3Kδ syndrome (APDS) in humans. J Allergy Clin Immunol. 2017;140:1152–6.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Thauland TJ, Pellerin L, Ohgami RS, Bacchetta R, Butte MJ. Case study: mechanism for increased follicular helper T cell development in activated PI3K delta syndrome. Front Immunol. Preprint posted online April 12, 2019. doi: 10.3389/fimmu.2019.00753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Cannons JL, Preite S, Kapnick SM, Uzel G, Schwartzberg PL. Genetic defects in phosphoinositide 3-kinase δ influence CD8(+) T cell survival, differentiation, and function. Front Immunol. Preprint posted online August 2, 2018. doi: 10.3389/fimmu.2018.01758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Carpier JM, Lucas CL. Epstein-Barr virus susceptibility in activated PI3Kδ syndrome (APDS) immunodeficiency. Front Immunol. Preprint posted online January 16, 2018. doi: 10.3389/fimmu.2017.02005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cohen JI. Herpesviruses in the activated phosphatidylinositol-3-Kinase-δ syndrome. Front Immunol. Preprint posted online February 23, 2018. doi: 10.3389/fimmu.2018.00237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Stark AK, Chandra A, Chakraborty K, Alam R, Carbonaro V, Clark J, et al. PI3Kδ hyper-activation promotes development of B cells that exacerbate Streptococcus pneumoniae infection in an antibody-independent manner. Nat Commun. 2018;9:3174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Elkaim E, Neven B, Bruneau J, Mitsui-Sekinaka K, Stanislas A, Heurtier L, et al. Clinical and immunologic phenotype associated with activated phosphoinositide 3-kinase δ syndrome 2: a cohort study. J Allergy Clin Immunol. 2016;138:210–8.e9. [DOI] [PubMed] [Google Scholar]
  • 27.Jamee M, Moniri S, Zaki-Dizaji M, Olbrich P, Yazdani R, Jadidi-Niaragh F, et al. Clinical, immunological, and genetic features in patients with activated PI3Kδ syndrome (APDS): a systematic review. Clin Rev Allergy Immunol. 2020;59:323–33. [DOI] [PubMed] [Google Scholar]
  • 28.Bloomfield M, Klocperk A, Zachova R, Milota T, Kanderova V, Sediva A. Natural course of activated phosphoinositide 3-kinase delta syndrome in childhood and adolescence. Front Pediatr. Preprint posted online July 19, 2021. doi: 10.3389/fped.2021.697706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Cannons JL, Villarino AV, Kapnick SM, Preite S, Shih HY, Gomez-Rodriguez J, et al. PI3Kδ coordinates transcriptional, chromatin, and metabolic changes to promote effector CD8(+) T cells at the expense of central memory. Cell Rep. Preprint posted online October 12, 2021. doi: 10.1016/j.celrep.2021.109804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Maccari ME, Abolhassani H, Aghamohammadi A, Aiuti A, Aleinikova O, Bangs C, et al. Disease evolution and response to rapamycin in activated phosphoinositide 3-Kinase δ syndrome: The European Society for Immunodeficiencies-Activated Phosphoinositide 3-Kinase δ Syndrome Registry. Front Immunol. Preprint posted online March 16, 2018. doi: 10.3389/fimmu.2018.00543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Avery DT, Kane A, Nguyen T, Lau A, Nguyen A, Lenthall H, et al. Germline-activating mutations in PIK3CD compromise B cell development and function. J Exp Med. 2018;215:2073–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lau A, Avery DT, Jackson K, Lenthall H, Volpi S, Brigden H, et al. Activated PI3Kδ breaches multiple B cell tolerance checkpoints and causes autoantibody production. J Exp Med. 2020;217:e20191336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kuehn HS, Ouyang W, Lo B, Deenick EK, Niemela JE, Avery DT, et al. Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science. 2014;345:1623–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Lougaris V, Moratto D, Baronio M, Tampella G, van der Meer JWM, Badolato R, et al. Early and late B-cell developmental impairment in nuclear factor kappa B, subunit 1-mutated common variable immunodeficiency disease. J Allergy Clin Immunol. 2017;139:349–52.e1. [DOI] [PubMed] [Google Scholar]
  • 35.Wray-Dutra MN, Al Qureshah F, Metzler G, Oukka M, James RG, Rawlings DJ. Activated PIK3CD drives innate B cell expansion yet limits B cell-intrinsic immune responses. J Exp Med. 2018;215:2485–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Rivalta B, Amodio D, Milito C, Chiriaco M, Di Cesare S, Giancotta C, et al. Case report: EBV chronic infection and lymphoproliferation in four APDS patients: the challenge of proper characterization, therapy, and follow-up. Front Pediatr. Preprint posted online August 27, 2021. doi: 10.3389/fped.2021.703853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Dimitrova D, Nademi Z, Maccari ME, Ehl S, Uzel G, Tomoda T, et al. International retrospective study of allogeneic hematopoietic cell transplantation for activated PI3K-delta syndrome. J Allergy Clin Immunol. 2022;149:410–21.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Begg M, Amour A, Jarvis E, Tang T, Franco SS, Want A, et al. An open label trial of nemiralisib, an inhaled PI3 kinase delta inhibitor for the treatment of activated PI3 kinase delta syndrome. Pulm Pharmacol Ther. 2023;79:102201. [DOI] [PubMed] [Google Scholar]
  • 39.Diaz N, Juarez M, Cancrini C, Heeg M, Soler-Palacín P, Payne A, et al. Seletalisib for activated PI3Kδ syndromes: open-label phase 1b and extension studies. J Immunol. 2020;205:2979–87. [DOI] [PubMed] [Google Scholar]
  • 40.Rao VK, Webster S, Dalm VA, Šedivá A, van Hagen PM, Holland S, et al. Effective “activated PI3Kδ syndrome”-targeted therapy with the PI3Kδ inhibitor leniolisib. Blood. 2017;130:2307–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Joenja [package insert]. Leiden, Netherlands: Pharming Technologies BV; 2023. [Google Scholar]
  • 42.Rao VK, Webster S, Šedivá A, Plebani A, Schuetz C, Shcherbina A, et al. A randomized, placebo-controlled phase 3 trial of PI3Kδ inhibitor leniolisib for activated PI3Kδ syndrome. Blood. 2023;141:971–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Rao VK, et al. Interim analysis of safety and hematological parameters of an ongoing long-term open-label extension study of investigational PI3Kδ inhibitor leniolisib for patients with activated pi3k delta syndrome (APDS) through December 2021. Poster presented at: 64th Annual American Society of Hematology Annual Meeting; 2022 Dec 10-13; New Orleans, LA. [Google Scholar]
  • 44.Rao VK, et al. Interim safety and efficacy analysis of an ongoing long-term open-label extension study of leniolisib for patients with activated PI3K delta syndrome (APDS). Poster presented at: 20th Biennial Meeting of European Society for Immunodeficiencies; 2022 Oct 12-16; Gothenburg, Sweden. [Google Scholar]
  • 45.Aliqopa [package insert]. Whippany, NJ: Bayer Healthcare Pharmaceuticals Inc; 2022. [Google Scholar]
  • 46.US Food & Drug Administration. FDA briefing document: phosphatidylinositol 3-kinase (PI3K) inhibitors in hematologic malignancies [Internet]. 2022. Apr. Available from: https://www.fda.gov/media/157762/download
  • 47.Zydelig [package insert]. Foster City, CA: Gilead Sciences Inc; 2022. [Google Scholar]
  • 48.US Food & Drug Administration. Gilead Sciences, Inc.; withdrawal of approval of indications for relapsed follicular lymphoma and relapsed small lymphocytic lymphoma for ZYDELIG (Idelalisib) tablets. Fed Regist. 2022;87:32031. [Google Scholar]
  • 49.Copiktra [package insert]. Las Vegas, NV: Verastem Inc; 2021. [Google Scholar]
  • 50.US Food & Drug Administration [Internet]. FDA warns about possible increased risk of death and serious side effects with cancer drug Copiktra (duvelisib); 2022. [cited 2022 Jul 14]. Available from: https://www.fda.gov/drugs/drug-safety-and-availability/fda-warns-about-possible-increased-risk-death-and-serious-side-effects-cancer-drug-copiktra
  • 51.US Food & Drug Administration. Secura Bio, Inc.; withdrawal of approval of relapsed or refractory follicular lymphoma indication for COPIKTRA. Fed Regist. 2022;87:21888–9. [Google Scholar]
  • 52.Ukoniq [package insert]. Edison, NJ: TG Therapeutics Inc; 2021. [Google Scholar]
  • 53.US Food & Drug Administration [Internet]. FDA approval of lymphoma medicine Ukoniq (umbralisib) is withdrawn due to safety concerns; 2022. [cited 2022 Jun 7]. Available from : https://www.fda.gov/drugs/drug-safety-and-availability/fda-approval-lymphoma-medicine-ukoniq-umbralisib-withdrawn-due-safety-concerns
  • 54.TG Therapeutics Inc. TG Therapeutics announces voluntary withdrawal of the BLA/sNDA for U2 to treat patients with CLL and SLL [Internet]; 2022. Apr. Available from: https://ir.tgtherapeutics.com/news-releases/news-release-details/tg-therapeutics-announces-voluntary-withdrawal-blasnda-u2-treat
  • 55.Piqray [package insert]. East Hanover, New Jersey: Novartis Pharmaceuticals Corp; 2022. [Google Scholar]
  • 56.Vanhaesebroeck B, Perry MWD, Brown JR, André F, Okkenhaug K. PI3K inhibitors are finally coming of age. Nat Rev Drug Discov. 2021;20:741–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Cuneo A, Barosi G, Danesi R, Fagiuoli S, Ghia P, Marzano A, et al. Management of adverse events associated with idelalisib treatment in chronic lymphocytic leukemia and follicular lymphoma: a multidisciplinary position paper. Hematol Oncol. 2019;37:3–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chellappa S, Kushekhar K, Munthe LA, Tjønnfjord GE, Aandahl EM, Okkenhaug K, et al. The PI3K p110δ isoform inhibitor idelalisib preferentially inhibits human regulatory T cell function. J Immunol. 2019;202:1397–405. [DOI] [PubMed] [Google Scholar]
  • 59.Sadhu C, Masinovsky B, Dick K, Sowell CG, Staunton DE. Essential role of phosphoinositide 3-kinase delta in neutrophil directional movement. J Immunol. 2003;170:2647–54. [DOI] [PubMed] [Google Scholar]
  • 60.Compagno M, Wang Q, Pighi C, Cheong TC, Meng FL, Poggio T, et al. Phosphatidylinositol 3-kinase δ blockade increases genomic instability in B cells. Nature. 2017;542:489–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Bou Zeid N, Yazbeck V. PI3k inhibitors in NHL and CLL: an unfulfilled promise. Blood Lymphat Cancer. 2023;13:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Eschweiler S, Ramírez-Suástegui C, Li Y, King E, Chudley L, Thomas J, et al. Intermittent PI3Kδ inhibition sustains anti-tumour immunity and curbs irAEs. Nature. 2022;605:741–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Schaefer M, Scott WJ, Hentemann MF, Rowley RB, Bull CO, Jenkins S, et al. 5G2N: X-ray structure of PI3Kinase gamma in complex with copanlisib [Internet]. doi: 10.2210/pdb5G2N/pdb. [DOI] [Google Scholar]
  • 64.Scott WJ, Hentemann MF, Rowley RB, Bull CO, Jenkins S, Bullion AM, et al. Discovery and SAR of novel 2,3-dihydroimidazo[1,2-c]quinazoline PI3K inhibitors: identification of copanlisib (BAY 80-6946). ChemMedChem. 2016;11:1517–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Somoza JR, Villasenor A. 4XE0: idelalisib bound to the p110 subunit of PI3K delta [Internet]. doi: 10.2210/pdb4XE0/pdb. [DOI] [Google Scholar]
  • 66.Somoza JR, Koditek D, Villaseñor AG, Novikov N, Wong MH, Liclican A, et al. Structural, biochemical, and biophysical characterization of idelalisib binding to phosphoinositide 3-kinase δ. J Biol Chem. 2015;290:8439–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Gutmann S, Rummel G, Shrestha B. Discovery of CDZ173 (leniolisib), representing a structurally novel class of PI3K delta-selective inhibitors [Internet]. doi: 10.2210/pdb5083/pdb. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Hoegenauer K, Soldermann N, Zécri F, Strang RS, Graveleau N, Wolf RM, et al. Discovery of CDZ173 (leniolisib), representing a structurally novel class of PI3K delta-selective inhibitors. ACS Med Chem Lett. 2017;8:975–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Berndt A, Miller S, Williams O, Le DD, Houseman BT, Pacold JI, et al. The p110 delta structure: mechanisms for selectivity and potency of new PI(3)K inhibitors. Nat Chem Biol. 2010;6:117–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Krause G, Hassenrück F, Hallek M. Copanlisib for treatment of B-cell malignancies: the development of a PI3K inhibitor with considerable differences to idelalisib. Drug Des Devel Ther. 2018;12:2577–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Miller MS, Thompson PE, Gabelli SB. Structural determinants of isoform selectivity in PI3K inhibitors. Biomolecules. 2019;9:82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Lannutti BJ, Meadows SA, Herman SE, Kashishian A, Steiner B, Johnson AJ, et al. CAL-101, a p110delta selective phosphatidylinositol-3-kinase inhibitor for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability. Blood. 2011;117:591–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Liu N, Rowley BR, Bull CO, Schneider C, Haegebarth A, Schatz CA, et al. BAY 80-6946 is a highly selective intravenous PI3K inhibitor with potent p110α and p110δ activities in tumor cell lines and xenograft models. Mol Cancer Ther. 2013;12:2319–30. [DOI] [PubMed] [Google Scholar]
  • 74.Winkler DG, Faia KL, DiNitto JP, Ali JA, White KF, Brophy EE, et al. PI3K-δ and PI3K-γ inhibition by IPI-145 abrogates immune responses and suppresses activity in autoimmune and inflammatory disease models. Chem Biol. 2013;20:1364–74. [DOI] [PubMed] [Google Scholar]
  • 75.Lampson BL, Brown JR. PI3Kδ-selective and PI3Kα/δ-combinatorial inhibitors in clinical development for B-cell non-Hodgkin lymphoma. Expert Opin Investig Drugs. 2017;26:1267–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lanahan SM, Wymann MP, Lucas CL. The role of PI3Kγ in the immune system: new insights and translational implications. Nat Rev Immunol. 2022;22:687–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Allen RA, Brookings DC, Powell MJ, Delgado J, Shuttleworth LK, Merriman M, et al. Seletalisib: characterization of a novel, potent, and selective inhibitor of PI3Kδ. J Pharmacol Exp Ther. 2017;361:429–40. [DOI] [PubMed] [Google Scholar]
  • 78.Cohen SB, Bainter W, Johnson JL, Lin TY, Wong JCY, Wallace JG, et al. Human primary immunodeficiency caused by expression of a kinase-dead p110δ mutant. J Allergy Clin Immunol. 2019;143:797–9.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Sogkas G, Fedchenko M, Dhingra A, Jablonka A, Schmidt RE, Atschekzei F. Primary immunodeficiency disorder caused by phosphoinositide 3-kinase δ deficiency. J Allergy Clin Immunol. 2018;142:1650–3.e2. [DOI] [PubMed] [Google Scholar]
  • 80.ClinicalTrials.gov. NCT02775916; c2016 [updated 2021 Jan 5; cited 2023 Mar 27].
  • 81.Dörner T, Zeher M, Laessing U, Chaperon F, De Buck S, Hasselberg A, et al. OP0250: a randomised, double-blind study to assess the safety, tolerability and preliminary efficacy of leniolisib (CDZ173) in patients with primary Sjögren’s syndrome. Ann Rheum Dis. 2018;77(suppl 2):174. [Google Scholar]
  • 82.Both T, Dalm VA, van Hagen PM, van Daele PL. Reviewing primary Sjögren’s syndrome: beyond the dryness - from pathophysiology to diagnosis and treatment. Int J Med Sci. 2017;14:191–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Nayar S, Campos J, Smith CG, Iannizzotto V, Gardner DH, Colafrancesco S, et al. Phosphatidylinositol 3-kinase delta pathway: a novel therapeutic target for Sjögren’s syndrome. Ann Rheum Dis. 2019;78:249–60. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES