Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Nov 1.
Published in final edited form as: Cancer J. 2019 Nov-Dec;25(6):394–400. doi: 10.1097/PPO.0000000000000414

PI3K Inhibitors: Present and Future

Jennifer R Brown 1
PMCID: PMC7082729  NIHMSID: NIHMS1068133  PMID: 31764120

Abstract

Inhibitors of PI3Kδ hold great potential for the therapy of CLL and B cell malignancies. After initially exciting efficacy results with idelalisib, the first in class inhibitor, the emergence of unexpected and unpredictable autoimmune toxicities, worse in less heavily treated and younger patients, has decreased the use of the currently available inhibitors. Newer drugs in development are attempting to reduce toxicity with novel schedules and/or combinations. This article reviews the clinical data on efficacy and toxicity across the class, and discusses ongoing efforts to understand and mitigate the likely on-target autoimmune toxicity.

Keywords: PI3Kδ, idelalisib, duvelisib, copanlisib


Interest in the phosphatidylinositol 3 kinase (PI3K) as a therapeutic target in B cell malignancies arose from observations that mice with either deletion of the delta isoform of PI3K1,2, or a kinase dead knock-in3, had significant B cell immunodeficiency. Specifically, these mice have significant reductions in several types of mature B cells - B1 (peritoneal); B2 (follicular); and marginal zone1,3,4 – leading to loss of germinal centers throughout lymph nodes, spleen and Peyer’s patches3, reduced immunoglobulin levels and reduced humoral response to antigens. In contrast, individual deletion of the other class 1 isoforms in mice, namely p110α4, p110β5, and p110γ6, has no obvious effect on B cells. This finding is perhaps unsurprising, as the alpha and beta isoforms have ubiquitous expression while expression of the delta isoform is largely limited to hematopoietic cells including B cells. Data from the delta deficient mice show that PI3K activation after B cell receptor (BCR) activation is primarily dependent on p110δ1,3, as is downstream signaling from cytokine/chemokine receptors and RTKs in B cells.

These observations provided the rationale for the initial development of PI3K delta inhibitors in B cell malignancies, although recent work has further evaluated the impact of PI3Kδ abrogation on CLL development in the TCL1 mouse model of CLL7. The p110δD910A/D910A kinase dead mouse was crossed with the Eμ–TCL1 mouse model of CLL, and the resulting global inactivation of p110δ profoundly inhibited the onset and severity of leukemia in these mice. The mice also resisted engraftment of TCL1 CLL cells through a T cell dependent mechanism, while a subset of those engrafted with a very high leukemia burden were able to clear the disease and resist rechallenge, suggesting an adaptive immune response contributing to disease clearance7. These data suggest that both cell autonomous and cell non autonomous mechanisms contribute to the potential potency of PI3Kδ inhibition in CLL. Also of interest, both this study and another8 demonstrated expansion of regulatory T cells (Tregs) in the setting of the TCL1 disease, with marked reduction in these Tregs with either genetic7 or pharmacologic8 inhibition of PI3Kδ. In the genetic model, the mice also developed an autoimmune colitis similar to that seen in patients treated with PI3Kδ inhibitors, suggesting that at least colitis is an on target side effect7.

Idelalisib: Efficacy

The first in class clinical PI3Kδ inhibitor was idelalisib, which demonstrated potent and specific inhibition of PI3Kδ in isoform-specific cell based assays and was able to induce apoptosis in CLL cells in vitro5,9 (Table 1). Treatment of CLL cells with idelalisib blocked AKT phosphorylation and downstream signaling from the BCR, chemokine receptors and CD405,9. The phase 1 study of idelalisib enrolled heavily pretreated patients with relapsed refractory B cell malignancies, and no formal DLTs were observed, although 25% of patients did have grade 3 or higher transaminase elevations10 (Table 2). The recommended phase 2 dose (RP2D) of 150 mg BID was ultimately chosen based on a careful assessment of this toxicity, which tended to be more frequent at higher doses, together with evidence of a plateau in both drug exposure and nodal reduction on CT, at 150 mg BID. The CLL subset of this study enrolled 54 patients with a median of 5 prior regimens, 91% with unmutated IGHV, 80% with bulky lymphadenopathy and 70% with treatment refractory disease – hence a very high risk population. In this study we were among the first to identify the phenomenon of treatment-related lymphocytosis, leading to the definition of partial response with lymphocytosis (PR-L); 81% of patients had a nodal response, with 39% meeting traditional PR criteria and an additional 32% meeting PR-L criteria. The median PFS at all dose levels was 15.8 months, but was 32 months in patients treated at the recommended phase 2 dose (RP2D) or higher.

Table 1.

Isoform Inhibition Profiles of PI3Kδ Inhibitors that are FDA Approved or in Advanced Clinical Development (IC50 values, nM)

Drug Name p110α p110β p110δ p110γ
Idelalisib9 820 565 2.5 89
Duvelisib26 1602 85 2.5 27
Copanlisib51 0.5 3.7 0.7 6.4
Umbralisib37 >10000 1116 22 1065
Parsaclisib43 >20000 >20000 1 >20000
ME-40145 22867 30 0.6 713

Table 2.

Comparison of Rates of Autoimmune Toxicity for Each Drug, Based on Lines of Prior Therapy and Duration on Inhibitor

Drug Disease / Study Median Prior Tx Median Time on Therapy Grade 3+ Neutropenia Grade 3+ Diarrhea/Colitis Grade 3+ Transaminitis Grade 3+ Rash Grade 3+ Pneumonitis
Idelalisib CLL ph 110 5 15 mos 43% 5.6% 2% 0% 5.6%
CLL / NHL combined safety analysis15 1-2 N.R. 30% 14% 14% 5% 3%
CLL frontline >65 + R17 0 22.4 mos 28% 42% 23% 13% 6%
CLL frontline > 6552 0 10.4 mos 17% 27% 22% 10% 5%
CLL frontline any age18 0 8.1 mos 33% 15% 52% 7% 7%
Duvelisib CLL, Phase 131 4 6 mos 44% 9.1%; 5.5% colitis 11% 0% 9.1%
CLL, Duo32 2 22.4 mos 30% 15%; 12% colitis 3% 2% 3%
NHL, Dynamo53 3 6 mos 23% 15% 6% 5% 4%
CLL, Ph 1 frontline cohort54 0 15.6 mos 33% 22% 17% 5.6% 11%
NHL, Contempo + R55 0 6.2 mos 10.7% 14% 25% 10.7% N.R.
NHL Contempo + G55 0 6.1 mos 22.2% 11% 26% 7.4% N.R.
Umbralisib CLL / NHL, phase 137 3 4.7 mos 13% 3% diarrhea / 2% colitis 3% 4% N.R.
CLL / NHL, integrated safety analysis42 3 6.5 mos 16% 4% diarrhea / <1% colitis 2% N.R. <0.5%
CLL/NHL Integrated Analysis, pts c > 6 mos exposure42 2 1.3 yrs 9% 8% diarrhea (includes 1 colitis) 3% 0% 0.5%
Copanlisib NHL Ph 2 registration trial34 3 5.5 mos 24% 5% diarrhea 2% 1% 1%
Parsaclisib NHL, CLL44 3 4 mos 19% 9% 3% 6% 0%
ME-401 CS: CLL, FL46 CLL 1, FL 2 9.8 mo NR (<15%) 20% 7.5% 10% 12.5%
IS: CLL, FL46 CLL 1, FL 2 4.8 mos (Ton T) NR (<15%) 9.7% 3.2% 0% 3.2%

These very promising results led to three registration trials, all in combination, due to concerns about how the FDA would interpret the persistent redistribution lymphocytosis that was observed with the single agent in the phase 1 study. The first to report was a randomized double-blind placebo-controlled trial comparing idelalisib with rituximab to rituximab with placebo11, with 110 patients per arm. The patient population included relapsed refractory CLL patients not appropriate for chemoimmunotherapy, with comorbidities (median CIRS comorbidity score 8), who had a median of three prior therapies, 84% with unmutated IGHV and 43.5% with TP53 dysfunction. The study was terminated early for efficacy, with a median time on idelalisib of 3.8 months, due to markedly superior PFS in the idelalisib arm (HR for progression or death, 0.15 (0.08–0.28) p<0.001)11. An improved OS was also seen at this early timepoint, while toxicity of idelalisib was low, likely due to the short follow-up11. This trial led to the 2014 FDA approval of idelalisib rituximab for relapsed refractory CLL patients in whom rituximab would be considered an appropriate therapy.

Recently, the final follow-up of this study has been published, demonstrating a median PFS of 20.3 months (95% CI, 17.3 to 26.3 months) among patients initially assigned to idela-R, with median OS 40.6 months (95% CI, 28.5 to 57.3 months)12. Prolonged exposure to idelalisib over a median of eighteen months increased the incidence of diarrhea (all grade, 46.4%; grade 2, 17.3%; and grade 3, 16.4%), colitis (grade 3+, 8.2%) and grade 3 or greater pneumonitis (6.4%).

The second registration trial comparing idelalisib ofatumumab to ofatumumab among 261 CLL patients with a median of 3 prior therapies demonstrated a median PFS of 16.3 mos in the idelalisib-ofatumumab group compared to 8 mos in the ofatumumab alone group (p<0.0001)13. Excess grade 3 or higher diarrhea and neutropenia were observed in the idelalisib arm, and treatment related deaths were 22 in the idelalisib-ofatumumab group compared to 6 in the ofatumumab group13. The final registration trial evaluated the addition of idelalisib to bendamustine rituximab (BR) among 416 relapsed refractory CLL patients with a median of 2 prior regimens, one-third of whom had 17p deletion14. At a median follow-up of 14 months, the median PFS was 20·8 months (95% CI 16·6–26·4) in the idelalisib-BR group compared to 11·1 months (8·9–11·1) in the placebo-BR group (HR 0·33, 95% CI 0·25–0·44; p<0·0001)14. Treatment emergent deaths were 11% on the idelalisib arm compared to 7% on the placebo arm, primarily infection14.

Idelalisib: Toxicity

These registration trials started to demonstrate a characteristic pattern of idelalisib toxicity, which included a relatively early transaminitis, followed by later events of pneumonitis, rash and diarrhea that could progress to colitis. Although all of these toxicities were seen on the phase 1 study, they became much more frequent in the registration program, possibly because the patient population was less heavily pretreated (Table 2). A pooled analysis of 760 patients with a median of 2-3 prior therapies (i.e. patients from the registration trials) demonstrated a 14% incidence of grade 3 or higher diarrhea and transaminitis, significantly higher than in phase 115. Furthermore, a combined 24 month follow-up analysis of the idelalisib-rituximab and idelalisib-ofatumumab registration trials demonstrated that 41% of patients had discontinued therapy due to adverse events, including infection as well as the above toxicities, with only 22.5% continuing on therapy at two years16.

Studies that attempted to move idelalisib into the front-line setting demonstrated even higher rates of similar toxicities. A company-sponsored frontline study of idelalisib with rituximab in patients over age 65 demonstrated a 42% incidence of grade 3+ diarrhea, 23% grade 3+ transaminitis, and 13% grade 3+ rash, leading to 45.3% of patients discontinuing therapy for toxicity17. Our group performed an investigator initiated frontline study of idelalisib with ofatumumab, among patients of any age, and found early fulminant hepatotoxicity typically at week 3-4, and requiring not just drug hold but usually immunosuppression to resolve18. Fifty-two percent of patients ultimately had grade 3-4 hepatotoxicity on this trial, which was associated with an activated CD8 T cell, perforin-positive infiltrate on liver biopsy. Risk factors for developing this toxicity included younger age and mutated IGHV18. Immune infiltrates were also found in bowel, and the colitis was also shown to be steroid-responsive1921. The biopsy data as well as the clinical response to immunosuppression suggested an autoimmune etiology for these toxicities, which could also explain why they were worse in less pretreated and younger patients, both of whom are presumably more immunologically intact.

Despite these higher rates of still poorly understood toxicities particularly in the frontline setting, Gilead embarked on a large frontline / early line phase 3 registration program in both CLL and follicular lymphoma. In March 2016, a combined safety analysis of three of these studies demonstrated worse overall survival in the idelalisib containing arms, at 7.4% compared to 3.5% on the control arms. This observation led to the summary closure of all frontline studies evaluating idelalisib. Interestingly, the deaths were primarily related to bacterial infections, sometimes with neutropenia, which had also been seen previously in the relapsed setting, but were not necessarily flagged as unusual among heavily pretreated CLL patients. Some cases of CMV and PJP were also noted. These findings demonstrate that idelalisib likely has multiple immunomodulatory effects, not only inducing autoimmune toxicity, but also increasing risk of both bacterial and opportunistic infections, as well as neutropenia.

The more unusual pattern of hepatitis and colitis strongly suggests an autoimmune process: delayed onset but rapid reoccurrence on rechallenge with drug, immune infiltrates on biopsy, responsiveness to steroids, and higher frequency among less pretreated and younger (i.e. more immunologically intact) patients. Following our initial observation that younger patients had greater risk of transaminitis in our frontline study, we looked at >750 patients across the Gilead registration program, and demonstrated that the rate of grade 3 or higher transaminitis with idelalisib was higher in untreated patients, and in younger patients, by decile of age22. Furthermore, mice with genetic inactivation of PI3K delta develop an autoimmune colitis which is similar to that seen in the patients3, and the same was seen in the TCL1 mice with the PI3KδD910A/D910A kinase dead mutation7. Interestingly, both mice and humans with mutations that disrupt regulatory T cell function also develop a syndrome of hepatitis, enteritis and pneumonitis23,24, similar to patients treated with idelalisib. We therefore investigated the impact of idelalisib on regulatory T cells among patients on our upfront study, and we demonstrated a significant decrease in Tregs by one month of therapy, particularly among patients with early hepatotoxicity18. In the TCL1 mouse model, regulatory T cells are expanded, and this expansion is abrogated in the p110δ kinase dead background, demonstrating that p110δ is needed for regulatory T cell survival7. Similar results have also been seen with pharmacological inhibition of PI3Kδ in the mouse model8.

Although idelalisib is a very effective drug, the unpredictable often delayed onset as well as severity of its autoimmune toxicity has markedly decreased its use in CLL and lymphoma. Further scientific work may allow its immunomodulatory properties to be harnessed for the therapy of CLL, lymphoma or solid tumors, particularly given that p110δ inactivation in Tregs in mouse models is both necessary and sufficient to confer resistance to a variety of solid tumors25. Meanwhile, however, given the importance of the PI3Kδ target, much ongoing drug development is continuing to target PI3Kδ in CLL and lymphoma.

Duvelisib

Duvelisib, an inhibitor of both the delta and gamma isoforms of PI3K (Table 1), was approved by the FDA in September 2018 for the therapy of relapsed CLL after at least two prior therapies, as well as follicular lymphoma. Duvelisib is about ten-fold more potent in inhibiting the delta isoform compared to gamma26, and at the 25 mg BID dose ultimately chosen for CLL therapy, maintains blood levels consistently above the IC90 for delta inhibition and the IC50 for gamma inhibition27. The importance of additionally inhibiting gamma in CLL is not definitely known, although the gamma isoform does have some expression in CLL cells. Duvelisib is cytotoxic to primary CLL cells in vitro, although this may be mediated primarily by delta isoform inhibition, and also inhibits signaling downstream of the BCR28,29. As importantly, however, PI3K gamma is thought to modulate the microenvironment, in particular gamma inhibition decreases myeloid derived suppressor cells (MDSCs) and shifts macrophages toward a more immunostimulatory, anti-cancer phenotype30.

In a phase 1 study in indolent NHL, dose escalation continued up to 100 mg BID, at which dose two DLTs occurred, one grade 3 transaminitis and one grade 3 rash, such that the MTD was determined to be 75 mg BID27. In subsequent expansion at both 25 and 75 mg BID doses, pAKT was substantially inhibited in CLL cells at 25 mg BID, with near complete inhibition of Ki67 seen by cycle 227,31. The ORR in phase 1, not including PR-L, was 56%, with 1 CR, and the median response duration was 21 months27,31. Given that response rates and durability appeared similar, and toxicity better, at the lower dose, 25 mg BID was moved forward as the RP2D27 and the phase 3 DUO trial was launched, randomizing relapsed refractory CLL patients between duvelisib and ofatumumab32. The median age of the patients was 69 and they had a median of two prior therapies. One-third had 17p deletion or TP53 mutation. DUO met its primary endpoint of improved PFS by independent review committee, at 13.3 mos compared to 9.9 mos for ofatumumab. The PFS of duvelisib as measured by investigators was 17.6 mos, which is very comparable to that seen in the idelalisib registration trials. About one-third of patients discontinued duvelisib for adverse events which included neutropenia and infections, and diarrhea and colitis, which occurred at similar frequencies to idelalisib, while rash, pneumonitis and transaminitis appeared to be less common (Table 2). This study led to the approval of duvelisib monotherapy for the therapy of CLL, currently the only monotherapy approval for a PI3K inhibitor in CLL. Many studies are ongoing, exploring duvelisib in combination, for example with venetoclax at our institution; on alternative schedules with treatment breaks skipping either days within a week, or weeks within a cycle, in an effort to reduce autoimmune toxicity; and in other disease settings, including T cell lymphomas.

Copanlisib

Copanlisib is an extremely potent pan-PI3K inhibitor with some relative selectivity for the alpha and delta isoforms, in fact often billed as an alpha / delta inhibitor, even though it is actually the most potent gamma inhibitor under investigation in CLL or lymphoma (Table 1). Copanlisib has been primarily developed in lymphoma, and received accelerated approval from the FDA in 2017 for the treatment of follicular lymphoma previously treated with alkylators and rituximab. In contrast to all other PI3Kδ inhibitors currently in the clinic, copanlisib is given intravenously, weekly for three weeks out of four. Thus copanlisib has several distinguishing features from idelalisib or duvelisib: IV administration; a punctuated schedule; and potency against the alpha isoform of PI3K. These differences are of particular interest because the toxicity profile of copanlisib is also distinct, and includes acute infusional hyperglycemia and hypertension, likely both related to its potency against PI3K alpha33. Toxicities thought to be related to delta inhibition, including transaminitis and diarrhea/colitis, are much less common, at 2% and 5% grade 3, respectively, in the lymphoma registration trial34. This trial enrolled 8 SLL patients, and in those patients, copanlisib had a 75% response rate, but data in CLL remain very limited. We are launching a study of adding copanlisib to enhance response among CLL patients with partial response on ibrutinib, in order to explore the activity and toxicity of copanlisib among CLL patients.

Investigational PI3K Inhibitors

Umbralisib

Among the as yet unapproved PI3K inhibitors, umbralisib (TGR-1202) is most advanced in clinical trials, having completed enrollment to its potential registration trial, UNITY-CLL. Umbralisib is a very specific inhibitor of the delta isoform of PI3K, but is approximately 10X less potent than idelalisib or duvelisib (Table 1). The structure of umbralisib is significantly different from idelalisib and duvelisib, and it also has in vitro activity against casein kinase 1μ35, which has been shown recently to be a therapeutic target in CLL36. The phase 1 study of umbralisib enrolled patients with advanced B cell malignancies, and required reformulation and extensive dose escalation to achieve adequate exposure and a high response rate37. Four DLTs were seen, including a grade 3 rash at the 800 mg initial formulation, a grade 3 hypokalemia at the 1800 mg original formulation, and two events of grade 3 fatigue at 1800 mg of the micronized formulation. The RP2D was ultimately chosen to be 800 mg daily of the micronized formulation in a fed state, which reportedly maintained concentrations above 5.25 μM throughout the 24 hr dosing interval. This concentration is below the EC50 of umbralisib for inhibition of casein kinase 1μ in a cell free kinase assay, which is 6.1 μM35, and below the 15 μM used to inhibit autophosphorylation in vitro13, suggesting that inhibition of CK1μ does not play a major role in vivo. To date pharmacodynamic analysis demonstrating inhibition of either potential target in patient samples treated on clinical trials has been lacking.

With a median duration of treatment and follow-up of 4.7 cycles, the ORR was 37% across all enrolled hematologic malignancies and all dose levels, with 50% ORR per iwCLL 2008 criteria plus an additional 35% PR-L among the 18 CLL patients37. The mean duration of response was 13.4 months in 16 CLL patients. Patients were eligible for intrapatient dose escalation, and, once the RP2D was determined, all patients transitioned to that dose (800 mg daily in the fed state). At the conclusion of the study, 13 of 90 patients remained on umbralisib, with a median duration of treatment and follow-up among those patients of 29.7 mos. Most patients discontinued for disease progression (56%), and 10% for adverse events37.

Clinical development of umbralisib moved rapidly into combination studies, with the novel anti-CD20 antibody ublituximab38, as well as with ibrutinib. In the phase 1 study of umbralisib with ublituximab in B cell malignancies, 22 CLL patients were enrolled, with a median of 3 prior regimens and 41% with 17p deletion39. The ORR was 62% (13/21), with 2 CRs and a median duration of response of 26 months. Their median treatment duration was 11 mos, with the majority of discontinuations due to progressive disease39. Our group led a multicenter phase 1/1b combination study of ibrutinib with umbralisib in relapsed refractory CLL and MCL, with 42 patients with a median of two prior therapies treated in parallel dose cohorts by disease40. No DLTs were observed and the MTD was not reached; umbralisib was given at its full single agent dose in combination with the standard dose of ibrutinib for each disease. ORR in CLL was 90% including 29% CRs, and two year PFS was 90%40. The MD Anderson Cancer Center led a similar phase 1 study of the triple-drug combination umbralisib, ibrutinib and ublituximab, and treated 46 patients, including 23 CLL patients41. The recommended phase 2 doses were again the full single agent doses of each drug. ORR was 84% across all evaluable patients, and was 100% among 22 CLL patients, including 36% CRs. Seven of the nine CLL patients evaluated for MRD had undetectable MRD. Median duration of response in CLL patients was 22.7 mos.

While the efficacy data with umbralisib are generally encouraging, it is the adverse event profile that has attracted the most attention. In the early studies, rates of grade 3 or higher autoimmune toxicities seen with PI3Kδ inhibitors appeared reduced: 2-3% transaminitis, 2-4% rash and 3-10% diarrhea3941. These initial results were difficult to interpret as many patients had very short follow-up on therapy, and particularly the diarrhea tends to occur later. A recent comprehensive safety analysis was therefore performed and encompassed 347 patients, with a median drug exposure of 6.5 mos, and found that grade 3 or higher transaminitis was seen in 2%, grade 3+ colitis in <1%, and grade 3+ pneumonitis in <0.5%42. Given this still relatively short follow-up, a sub-analysis focused particularly on 177 patients who remained on umbralisib for at least 6 months, and identified rates of grade 3 or higher toxicity as follows: transaminitis 3%, rash 0%, pneumonitis 0.6%, diarrhea 8%42. Only 1 of the 14 patients with diarrhea had colitis, and most of the patients were managed only with dose interruption, not requiring steroids. These data suggest that umbralisib has some similar autoimmune toxicities to idelalisib and duvelisib, but that these occur less frequently and/or are milder. We still await the results of randomized trials, larger patient populations and longer follow-up to truly clarify this.

The UNITY registration trial in CLL is comparing umbralisib given with ublituximab, to chlorambucil-obinutuzumab, in a patient population which is approximately 60% treatment naïve and 40% relapsed. The study is fully accrued and an announcement that overall response rate (ORR) would not be assessed for potential accelerated approval occurred in September 2018. TG Therapeutics is now awaiting the results of the primary PFS endpoint, for potential filing for full approval.

Parsaclisib

One of two highly potent and specific PI3Kδ inhibitors in advanced clinical development is parsaclisib (INCB050465), which has an IC50 for delta inhibition of 10 nM in whole blood and is 19000X selective for inhibition of delta compared to the other class 1 isoforms43. Parsaclisib was assessed in patients with advanced B cell malignancies in phase 1, both alone and in combination with the JAK1 inhibitor itacitinib, as well as with the chemotherapy regimen RICE44. Seventy-two patients (6 with CLL) were treated with the single agent at doses ranging from 5-45 mg daily in phase 1, and expansion cohorts were treated at 20 mg daily, 30 mg daily, and an intermittent schedule of 20 mg daily for 9 weeks, then once weekly. All doses tested led to continuous plasma exposure above the IC90, and no DLTs occurred. The primary adverse events of any grade were diarrhea/colitis in 36% (9% grade 3-4), rash in 31% (6% grade 3-4), transaminitis in 29% (albeit almost all grade 1) and grade 3-4 neutropenia in 19%. There were 3 events each of hypotension and sepsis, as well as 3 fatal AEs in 2 patients (2 respiratory failure and 1 sepsis). Dose interruptions occurred in 42% of patients. An intermittent schedule was piloted in an effort to reduce adverse events, and in fact none of 26 patients who received intermittent dosing discontinued. Furthermore, no grade 4 neutropenia or grade 4 non-heme adverse events occurred on the intermittent schedule, and only 1 case each of grade 3 rash and diarrhea, suggesting that the intermittent dosing did improve tolerability. Objective response rates were 71% in follicular lymphoma, 78% in marginal zone lymphoma, 67% in mantle cell lymphoma, 30% in DLBCL and 33% in CLL (3 of whom had had prior ibrutinib)44. Phase 2 studies are ongoing, as well as some phase 1 combination studies, in order to identify an optimal potential registration path.

ME-401

ME-401 is the other highly potent and specific novel delta inhibitor that has been investigated in both CLL and follicular lymphoma and is currently enrolling a potential registration trial in follicular lymphoma. ME-401 inhibits the PI3Kδ isoform with IC50 in the low nM range even in whole blood and in isoform-specific cell based assays (Table 1). The first in human study of this drug was an ascending dose study performed in healthy male volunteers in three groups, with each subject given two doses at least one week apart45. No significant or recurrent adverse events were seen. Exposure was dose proportional up to 60 mg, and inhibition of basophil activation, a δ isoform specific assay, was also dose proportional and reached 90% inhibition at 60 mg. Furthermore, dosing at 60 mg was predicted to result in trough plasma concentrations that would maintain 90% inhibition of basophil activation. This dose was therefore selected as the starting dose for patient studies.

ME-401 has now also been studied in patients with CLL/SLL and follicular lymphoma in phase 1, as a single agent and together with rituximab in data that have been reported, and most recently together with zanubrutinib in a cohort that is still enrolling46. ME-401 was dosed at 60, 120 and 180 mg without DLTs in a 56 day window, with high response rates and with typical adverse event (AE) rates which were as high as one-third of patients developing grade 3+ diarrhea/colitis or rash. An intermittent schedule was then implemented, with two months continuous dosing at 60 mg, followed by one week on, three weeks off. This schedule is supported by the 28 hour half-life of the drug and was intended to allow recovery of regulatory T cells47 in the second two weeks. The loss of Tregs with PI3Kδ inhibitors is believed to lead to the autoimmune adverse events of PI3Kδ inhibition. Intermittent dosing reduced the grade 3+ diarrhea rate to 9.7%, and allowed 77% of patients to remain on therapy, compared to 53% on continuous dosing, albeit with shorter follow-up. Grade 3+ rash was reduced from 10% to 0%, and pneumonia/pneumonitis from 12.5% to 3.2%. Response rates were preserved, and were 80% in 50 FL patients across all dose groups, and 100% in 14 CLL patients (83% overall)46.

A randomized phase 2 registration trial has been initiated in relapsed follicular lymphoma patients. All patients will receive two months continuous dosing lead-in, and then be randomized in a double blind fashion to continuous vs intermittent dosing. In the event of progression on intermittent dosing, patients may switch to continuous dosing, and in the event of adverse event on either arm, drug is held and patients may resume on intermittent dosing regardless of their original arm. This study should provide high quality data on the true value of the intermittent dose schedule, which currently appears to be the preferred path forward to maximize the clinical use and benefit of PI3Kδ inhibitors.

Summary: The Future

The preponderance of evidence from mouse models as well as clinical trials suggests that the autoimmune toxicities of PI3Kδ inhibitors are a class effect, seen as they are even with the highly specific, highly potent newest agents, parsaclisib and ME-401. Given this, a few options exist for optimizing the use of these drugs in order to harness their efficacy. Preliminary data from intermittent dosing with both parsaclisib and especially ME-401 suggest that these intermittent schedules may reduce toxicity while largely maintaining efficacy. The results of the ME-401 registration trial will provide substantial evidence to address this hypothesis. Correlative studies exploring the impact of these schedules on T cells are also of great interest and are ongoing in the context of clinical trials. Additionally, combination studies that use lower intermittent doses of the PI3K inhibitor as a potentiating agent are also likely to be fruitful; these studies could rely on either the direct anti-B cell effects of the delta inhibitors, or on their immunomodulatory properties, to enhance the efficacy of the combination partner. Umbralisib may see success through this mechanism, as its similar exposure relative to its lower potency likely leads to less overall drug exposure. A better scientific understanding of the immunomodulatory properties of the drugs in vivo would certainly help optimize this approach. Finally, and likely furthest away, would be combinations that prevent the effects of the PI3Kδ inhibitors on T cells. For example, preclinical data suggest that inhibition of histone deacetylases may protect regulatory T cells48,49, and in fact, a study combining romidepsin with duvelisib provided suggestive evidence for reduced toxicity of the combination50. Whether this approach or others would prove clinically viable or helpful depends on the drugs involved and remains for future study. Meanwhile, the PI3Kδ inhibitor class is a very active drug class in lymphoma and CLL, if the toxicity can be managed; while managing this toxicity has proved more challenging than initially expected, ongoing work is now pointing a way forward.

Acknowledgments

This work is funded by NCI, 1R01CA213442-01A1 to JRB.

CONFLICTS OF INTEREST: JRB has served as a consultant for Abbvie, Acerta, Astra-Zeneca, Beigene, Catapult, Dynamo Therapeutics, Genentech/Roche, Gilead, Juno/Celgene, Kite, Loxo, Novartis, Octapharma, Pfizer, Pharmacyclics, Sunesis, TG Therapeutics, Verastem; received honoraria from Janssen and Teva; received research funding from Gilead, Loxo, Sun and Verastem; and served on data safety monitoring committees for Morphosys and Invectys.

REFERENCES

  • 1.Clayton E, Bardi G, Bell SE, et al. A crucial role for the p110delta subunit of phosphatidylinositol 3-kinase in B cell development and activation. J Exp Med 2002;196:753–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Jou ST, Carpino N, Takahashi Y, et al. Essential, nonredundant role for the phosphoinositide 3-kinase p110delta in signaling by the B-cell receptor complex. Mol Cell Biol 2002;22:8580–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Okkenhaug K, Bilancio A, Farjot G, et al. Impaired B and T cell antigen receptor signaling in p110delta PI 3-kinase mutant mice. Science 2002;297:1031–4. [DOI] [PubMed] [Google Scholar]
  • 4.Ramadani F, Bolland DJ, Garcon F, et al. The PI3K isoforms p110alpha and p110delta are essential for pre-B cell receptor signaling and B cell development. Sci Signal 2010;3:ra60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Herman SE, Gordon AL, Wagner AJ, et al. Phosphatidylinositol 3-kinase-delta inhibitor CAL-101 shows promising preclinical activity in chronic lymphocytic leukemia by antagonizing intrinsic and extrinsic cellular survival signals. Blood 2010;116:2078–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Webb LM, Vigorito E, Wymann MP, Hirsch E, Turner M. Cutting edge: T cell development requires the combined activities of the p110gamma and p110delta catalytic isoforms of phosphatidylinositol 3-kinase. J Immunol 2005;175:2783–7. [DOI] [PubMed] [Google Scholar]
  • 7.Dong S, Harrington BK, Hu EY, et al. PI3K p110delta inactivation antagonizes chronic lymphocytic leukemia and reverses T cell immune suppression. J Clin Invest 2019;129:122–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hanna BS, Roessner PM, Scheffold A, et al. PI3Kdelta inhibition modulates regulatory and effector T-cell differentiation and function in chronic lymphocytic leukemia. Leukemia 2019;33:1427–38. [DOI] [PubMed] [Google Scholar]
  • 9.Lannutti BJ, Meadows SA, Herman SE, 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]
  • 10.Brown JR, Byrd JC, Coutre SE, et al. Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110delta, for relapsed/refractory chronic lymphocytic leukemia. Blood 2014;123:3390–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Furman RR, Sharman JP, Coutre SE, et al. Idelalisib and rituximab in relapsed chronic lymphocytic leukemia. N Engl J Med 2014;370:997–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Sharman JP, Coutre SE, Furman RR, et al. Final Results of a Randomized, Phase III Study of Rituximab With or Without Idelalisib Followed by Open-Label Idelalisib in Patients With Relapsed Chronic Lymphocytic Leukemia. J Clin Oncol 2019;37:1391–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Jones JA, Robak T, Brown JR, et al. Efficacy and safety of idelalisib in combination with ofatumumab for previously treated chronic lymphocytic leukaemia: an open-label, randomised phase 3 trial. Lancet Haematol 2017;4:e114–e26. [DOI] [PubMed] [Google Scholar]
  • 14.Zelenetz AD, Barrientos JC, Brown JR, et al. Idelalisib or placebo in combination with bendamustine and rituximab in patients with relapsed or refractory chronic lymphocytic leukaemia: interim results from a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Oncol 2017;18:297–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Coutre S, Barrientos J, Brown JR, et al. Safety of idelalisib in B-cell malignancies: Integrated analysis of eight clinical trials. ASCO Meeting Abstracts 2015;33:abstr e18030. [Google Scholar]
  • 16.Brown J, Ghia P, Jones J, et al. Outcomes of Patients with Relapsed and Refractory Chronic Lymphocytic Leukemia Who Discontinue Idelalisib Treatment. Haematologica 2016;101 Abstract 7531.26819059 [Google Scholar]
  • 17.O’Brien SM, Lamanna N, Kipps TJ, et al. A phase 2 study of idelalisib plus rituximab in treatment-naive older patients with chronic lymphocytic leukemia. Blood 2015;126:2686–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lampson BL, Kasar SN, Matos TR, et al. Idelalisib given front-line for treatment of chronic lymphocytic leukemia causes frequent immune-mediated hepatotoxicity. Blood 2016;128:195–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Coutre SE, Barrientos JC, Brown JR, et al. Management of adverse events associated with idelalisib treatment: expert panel opinion. Leuk Lymphoma 2015:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Louie CY, DiMaio MA, Matsukuma KE, Coutre SE, Berry GJ, Longacre TA. Idelalisib-associated Enterocolitis: Clinicopathologic Features and Distinction From Other Enterocolitides. Am J Surg Pathol 2015. [DOI] [PubMed] [Google Scholar]
  • 21.Weidner AS, Panarelli NC, Geyer JT, et al. Idelalisib-associated Colitis: Histologic Findings in 14 Patients. Am J Surg Pathol 2015. [DOI] [PubMed] [Google Scholar]
  • 22.Brown J, Zelenetz A, Furman R, et al. Risk Factors for Grade 3/4 Transaminase Elevation in Patients with Chronic Lymphocytic Leukemia Treated with Idelalisib. Blood 2017;130:3016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Godfrey VL, Wilkinson JE, Russell LB. X-linked lymphoreticular disease in the scurfy (sf) mutant mouse. The American journal of pathology 1991;138:1379–87. [PMC free article] [PubMed] [Google Scholar]
  • 24.Torgerson TR, Ochs HD. Immune dysregulation, polyendocrinopathy, enteropathy, X-linked: forkhead box protein 3 mutations and lack of regulatory T cells. J Allergy Clin Immunol 2007;120:744–50; quiz 51-2. [DOI] [PubMed] [Google Scholar]
  • 25.Ali K, Soond DR, Pineiro R, et al. Inactivation of PI(3)K p110delta breaks regulatory T-cell-mediated immune tolerance to cancer. Nature 2014;510:407–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Winkler DG, Faia KL, DiNitto JP, et al. PI3K-delta and PI3K-gamma 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]
  • 27.Flinn IW, O’Brien S, Kahl B, et al. Duvelisib, a novel oral dual inhibitor of PI3K-delta,gamma, is clinically active in advanced hematologic malignancies. Blood 2018;131:877–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Balakrishnan K, Peluso M, Fu M, et al. The phosphoinositide-3-kinase (PI3K)-delta and gamma inhibitor, IPI-145 (Duvelisib), overcomes signals from the PI3K/AKT/S6 pathway and promotes apoptosis in CLL. Leukemia 2015;29:1811–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Dong S, Guinn D, Dubovsky JA, et al. IPI-145 antagonizes intrinsic and extrinsic survival signals in chronic lymphocytic leukemia cells. Blood 2014;124:3583–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kaneda MM, Messer KS, Ralainirina N, et al. PI3Kgamma is a molecular switch that controls immune suppression. Nature 2016;539:437–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.O’Brien S, Patel M, Kahl BS, et al. Duvelisib, an oral dual PI3K-delta,gamma inhibitor, shows clinical and pharmacodynamic activity in chronic lymphocytic leukemia and small lymphocytic lymphoma in a Phase 1 study. Am J Hematol 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Flinn IW, Hillmen P, Montillo M, et al. The phase 3 DUO trial: duvelisib vs ofatumumab in relapsed and refractory CLL/SLL. Blood 2018;132:2446–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Dreyling M, Morschhauser F, Bouabdallah K, et al. Phase II study of copanlisib, a PI3K inhibitor, in relapsed or refractory, indolent or aggressive lymphoma. Annals of oncology : official journal of the European Society for Medical Oncology / ESMO 2017;28:2169–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Dreyling M, Santoro A, Mollica L, et al. Phosphatidylinositol 3-Kinase Inhibition by Copanlisib in Relapsed or Refractory Indolent Lymphoma. J Clin Oncol 2017;35:3898–905. [DOI] [PubMed] [Google Scholar]
  • 35.Deng C, Lipstein MR, Scotto L, et al. Silencing c-Myc translation as a therapeutic strategy through targeting PI3Kdelta and CK1epsilon in hematological malignancies. Blood 2017;129:88–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Janovska P, Verner J, Kohoutek J, et al. Casein kinase 1 is a therapeutic target in chronic lymphocytic leukemia. Blood 2018;131:1206–18. [DOI] [PubMed] [Google Scholar]
  • 37.Burris HA 3rd, Flinn IW, Patel MR, et al. Umbralisib, a novel PI3Kdelta and casein kinase-1epsilon inhibitor, in relapsed or refractory chronic lymphocytic leukaemia and lymphoma: an open-label, phase 1, dose-escalation, first-in-human study. Lancet Oncol 2018;19:486–96. [DOI] [PubMed] [Google Scholar]
  • 38.de Romeuf C, Dutertre CA, Le Garff-Tavernier M, et al. Chronic lymphocytic leukaemia cells are efficiently killed by an anti-CD20 monoclonal antibody selected for improved engagement of FcgammaRIIIA/CD16. Br J Haematol 2008;140:635–43. [DOI] [PubMed] [Google Scholar]
  • 39.Lunning M, Vose J, Nastoupil L, et al. Ublituximab and Umbralisib in Relapsed/ Refractory B-cell Non-Hodgkin Lymphoma and Chronic Lymphocytic Leukemia. Blood 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Davids MS, Kim HT, Nicotra A, et al. Umbralisib in combination with ibrutinib in patients with relapsed or refractory chronic lymphocytic leukaemia or mantle cell lymphoma: a multicentre phase 1–1b study. Lancet Haematol 2019;6:e38–e47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Nastoupil LJ, Lunning MA, Vose JM, et al. Tolerability and activity of ublituximab, umbralisib, and ibrutinib in patients with chronic lymphocytic leukaemia and non-Hodgkin lymphoma: a phase 1 dose escalation and expansion trial. Lancet Haematol 2019;6:e100–e9. [DOI] [PubMed] [Google Scholar]
  • 42.Davids M, Flinn I, Mato A, et al. Long Term Integrated Safety Analysis of Umbralisib (TGR-1202), a PI3K-Delta/CK1-Epsilon Inhibitor with a Differentiated Safety Profile, in Patients with Relapsed/Refractory Lymphoid Malignancies. EHA Meeting Abstracts 2018:PF444. [Google Scholar]
  • 43.Shin N, Koblish H, Covington M, et al. INCB050465, a novel PI3Kδ inhibitor, synergizes with PIM protein kinase inhibition to cause tumor regression in a model of DLBCL. Cancer Res 2015;75:Abstract 2671. [Google Scholar]
  • 44.Forero-Torres A, Ramchandren R, Yacoub A, et al. Parsaclisib, a potent and highly selective PI3Kdelta inhibitor, in patients with relapsed or refractory B-cell malignancies. Blood 2019;133:1742–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Moreno O, Butler T, Zann V, Willson A, Leung P, Connor A. Safety, Pharmacokinetics, and Pharmacodynamics of ME-401, an Oral, Potent, and Selective Inhibitor of Phosphatidylinositol 3-Kinase P110delta, Following Single Ascending Dose Administration to Healthy Volunteers. Clin Ther 2018;40:1855–67. [DOI] [PubMed] [Google Scholar]
  • 46.Zelenetz A, Jagadeesh D, Kenkre V, et al. The PI3Kδ Inhibitor ME‐401 ± Rituximab in Relapsed/Refractory (R/R) Follicular Lymphoma (FL), Chronic Lymphocytic Leukemia (CLL), and Small Lymphocytic Lymphoma (SLL). Hematological Oncology 2019;37:176.30261551 [Google Scholar]
  • 47.Mahnke K, Schonfeld K, Fondel S, et al. Depletion of CD4+CD25+ human regulatory T cells in vivo: kinetics of Treg depletion and alterations in immune functions in vivo and in vitro. International journal of cancer Journal international du cancer 2007;120:2723–33. [DOI] [PubMed] [Google Scholar]
  • 48.Choi SW, Gatza E, Hou G, et al. Histone deacetylase inhibition regulates inflammation and enhances Tregs after allogeneic hematopoietic cell transplantation in humans. Blood 2015;125:815–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lucas JL, Mirshahpanah P, Haas-Stapleton E, Asadullah K, Zollner TM, Numerof RP. Induction of Foxp3+ regulatory T cells with histone deacetylase inhibitors. Cell Immunol 2009;257:97–104. [DOI] [PubMed] [Google Scholar]
  • 50.Horwitz S, Moskowitz A, Jacobsen E, et al. The Combination of Duvelisib, a PI3K-δ,γ Inhibitor, and Romidepsin Is Highly Active in Relapsed/Refractory Peripheral T-Cell Lymphoma with Low Rates of Transaminitis: Results of Parallel Multicenter, Phase 1 Combination Studies with Expansion Cohorts. Blood 2018;132:683. [Google Scholar]
  • 51.Liu N, Rowley BR, Bull CO, et al. BAY 80-6946 is a highly selective intravenous PI3K inhibitor with potent p110alpha and p110delta activities in tumor cell lines and xenograft models. Molecular cancer therapeutics 2013;12:2319–30. [DOI] [PubMed] [Google Scholar]
  • 52.Zelenetz A, Lamanna N, Kipps T, et al. A phase 2 study of idelalisib monotherapy in previously untreated patients ≥65 years with chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL). British journal of haematology 2016;173:97. [Google Scholar]
  • 53.Flinn IW, Miller CB, Ardeshna KM, et al. DYNAMO: A Phase II Study of Duvelisib (IPI-145) in Patients With Refractory Indolent Non-Hodgkin Lymphoma. J Clin Oncol 2019;37:912–22. [DOI] [PubMed] [Google Scholar]
  • 54.O’Brien S, Faia K, White K, et al. Early Clinical Activity and Pharmacodynamic Effects of Duvelisib, a PI3K-Delta, Gamma Inhibitor, in Patients with Treatment-NA. Haematologica 2015;100:154–5. [Google Scholar]
  • 55.Casulo C, Sancho J-M, Van Eygen K, et al. Contempo: Preliminary Results in First-Line Treatment of Follicular Lymphoma with the Oral Dual PI3K-δ,γ Inhibitor, Duvelisib, in Combination with Rituximab or Obinutuzumab. Blood 2016;128:2979. [Google Scholar]

RESOURCES